1
|
Kansara K, Laha A, Kumar A, Bhatia D. Programmable DNA-Based Nanodevices for Next-Generation Clinical and Healthcare Applications. Adv Biol (Weinh) 2025; 9:e2400593. [PMID: 39913110 DOI: 10.1002/adbi.202400593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/17/2024] [Indexed: 02/07/2025]
Abstract
Deoxyribonucleic acid (DNA) nanotechnology has brought an unparalleled set of possibilities for self-assembled structures emerging as an independent branch of synthetic biology. The field of science uses the molecular properties of DNA to build nanoparticles and nanodevices that have the potential to bring breakthroughs in medical science. On the one hand, their biocompatibility, precision, synthetic ease, and programmability make them an ideal choice in drug delivery and healthcare. On the other, the lack of proper biodistribution profiles, stability inside the system, enzymatic cleavage, immune recognition, and translational barriers are some of the hurdles it faces. Many recent technological advancements are in progress to tackle these challenges, while some already have been used. These tools and technologies need to be understood and studied for the successful transition of these intelligent DNA nanostructures (DNs) to healthcare applications. This review thus, highlights some of the challenges being faced by the DNs in healthcare. Additionally, it provides an overview of the recent trends in using these devices in disease detection and remission and finally talks about the future scope and opportunities for an effective transition from bench to bedside.
Collapse
Affiliation(s)
- Krupa Kansara
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat, 382055, India
| | - Anwesha Laha
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat, 382055, India
| |
Collapse
|
2
|
Wu C, Yang X, Yang K, Yu Q, Huang C, Li F, Zhang L, Zhu D. Compensatory effect-based oxidative stress management microneedle for psoriasis treatment. Bioact Mater 2025; 46:229-241. [PMID: 39811463 PMCID: PMC11732109 DOI: 10.1016/j.bioactmat.2024.12.015] [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: 09/17/2024] [Revised: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Reactive oxygen species (ROS) at elevated levels trigger oxidative DNA damage, which is a significant factor in psoriasis exacerbation. However, normal ROS levels are essential for cell signaling, cell growth regulation, differentiation, and immune responses. To address this, we developed ROS control strategies inspired by compensatory effects. DNA nanostructures with the advantage of being more stable than linear nucleic acid molecules in physiological environments were exquisitely fabricated and incorporated into microneedles (MN). These nanostructures regulate ROS levels and facilitate the delivery of IL-17A siRNA to psoriatic lesions. Our findings demonstrate that this transdermal drug delivery system effectively manages ROS levels in the psoriatic microenvironment, inhibiting pyroptosis and abnormal immune activation. Moreover, modulating ROS levels enhances the therapeutic impact of IL-17A siRNA, offering a promising in situ treatment approach for psoriasis.
Collapse
Affiliation(s)
- Chaoxiong Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Xinyu Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Kaiyue Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Qingyu Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Chenlu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Fangzhou Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Linhua Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Dunwan Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
3
|
Hong S, Cui Y, He D, Wu H, Jiang W, Cao J, Wang X. GelMA Hydrogels Integrated With aptamer CH6-Functionalized Tetrahedral DNA Nanostructures for Osteoporotic Mandibular Regeneration. Macromol Biosci 2025; 25:e2400471. [PMID: 39838729 DOI: 10.1002/mabi.202400471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/22/2024] [Indexed: 01/23/2025]
Abstract
Osteoporotic bone regeneration is challenging due to impaired bone formation. Tetrahedral DNA nanostructures (TDN), promising nucleic acid nanomaterials, have garnered attention for their potential in osteoporotic mandibular regeneration owing to their ability to enhance cellular activity and promote osteogenic differentiation. Osteoblasts play a critical role in bone regeneration; however, intracellular delivery of TDN into osteoblasts remains difficult. In this study, a novel osteoblast-targeted CH6 aptamer-functionalized TDN (TDN-CH6) is aimed to develop for osteoporotic mandibular regeneration. This results demonstrated that TDN-CH6 exhibits superior osteoblast specificity and efficient recruitment to bone fracture sites. Furthermore, TDN-CH6 significantly enhances cellular activity and osteogenic differentiation compared to TDN alone. Notably, Gelatin Methacryloyl (GelMA) hydrogels incorporating TDN and TDN-CH6 shows improved biological performance and are favorable for osteoporotic mandibular regeneration, suggesting that this platform represents a promising strategy for addressing complex bone defects.
Collapse
Affiliation(s)
- Shebin Hong
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Ya Cui
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Dongming He
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Hao Wu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jian Cao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xudong Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| |
Collapse
|
4
|
Qiu Y, Qiu Y, Zhou W, Lu D, Wang H, Li B, Liu B, Wang W. Advancements in functional tetrahedral DNA nanostructures for multi-biomarker biosensing: Applications in disease diagnosis, food safety, and environmental monitoring. Mater Today Bio 2025; 31:101486. [PMID: 39935897 PMCID: PMC11810847 DOI: 10.1016/j.mtbio.2025.101486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/29/2024] [Accepted: 01/11/2025] [Indexed: 02/13/2025] Open
Abstract
Deoxyribonucleic acid (DNA) offers the fundamental building blocks for the precisely controlled assemblies due to its inherent self-assembly and programmability. The tetrahedral DNA nanostructure (TDN) stands out as a widely utilized nanostructure, attracting attention for its high biostability, excellent biocompatibility, and versatile modification sites. The capability of DNA tetrahedron to interact with various signal outputs makes it ideal for developing functional DNA nanostructures in biosensing platforms. This review highlights recent advancements in functional tetrahedral DNA nanostructures (FTDN) for various biomarkers monitoring, including nucleic acid, protein, mycotoxin, agent, and metal ion. Additionally, it discusses the potential of FTDN in the fields of disease diagnosis, food safety, and environmental monitoring. The review also introduces the application of FTDN-based biosensors for simultaneous identification of multiple biomarkers. Finally, challenges and prospects are addressed to provide guidance for the continued development of FTDN-based biosensing platforms.
Collapse
Affiliation(s)
- Yun Qiu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yixing Qiu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Wenchao Zhou
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Dai Lu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Huizhen Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Academician Atta-ur-Rahman Belt and Road Traditional Medicine Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
5
|
Huang P, Tang X, Zhao S, Luo J, Tang Y, Wang B, Xie Z, Wu X, Xie S, Chen M, Chang K. DNA nanorobot for mitochondria-targeted microRNAs detection and tailored regulation. Theranostics 2025; 15:4638-4653. [PMID: 40225575 PMCID: PMC11984390 DOI: 10.7150/thno.105762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Mitochondrial miRNAs (mitomiRs) are crucial regulators of mitochondrial functions and play pivotal roles in tumorigenesis and cancer progression. Nevertheless, direct monitoring mitomiRs and regulating mitochondrial function at the subcellular level remains challenging. Methods: In this study, we present a versatile DNA framework-based nanorobot for synchronous ultrasensitive detection of mitochondrial miRNAs (mitomiRs) and modulation of the mitochondria-associated apoptosis process. The DNA nanorobot features a tetrahedral nucleic acid framework as its structural body, two DNA hairpins (H1 and H2) as functional arms, and a mitochondria-targeting triphenylphosphine (TPP) group as the command center. The DNA nanorobot was comprehensively characterized for its morphological properties, mitochondria-targeting capacity, mitomiRs detection performance, DOX-loading and release behaviors, and antineoplastic effects both in vitro and in vivo. Results: Upon recognizing mitomiRs, the arms of the DNA nanorobot activate and trigger spatially restricted catalytic hairpin assembly (CHA) reactions with accelerated kinetics to generate amplified fluorescence signals. Additionally, the lipophilic anticancer drug doxorubicin (DOX) encapsulated within the DNA nanorobot induces reactive oxygen species (ROS) production, leading to mitochondria damage and promoting mitochondria-associated apoptosis in tumor cells. Conclusion: This newly developed DNA nanorobot provides a multifunctional platform for precise mitochondria-targeted diagnosis and enhanced therapeutic efficacy, advancing innovative strategies for mitochondria-focused tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Ping Huang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Shuang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Jie Luo
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Yu Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Binpan Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Zuowei Xie
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Xianlan Wu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Shuang Xie
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
- College of Pharmacy and Laboratory Medicine, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| |
Collapse
|
6
|
Qian C, Chen S, Chen L, Zhang C, Yang L, Li Q, Kang B, Chen X, Mei P, Gu H, Liu Y, Liu Y. Tetrahedral DNA Nanostructure-Modified Nanocoating for Improved Bioaffinity and Osseointegration of Titanium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2412747. [PMID: 40103513 DOI: 10.1002/smll.202412747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/20/2025] [Indexed: 03/20/2025]
Abstract
Titanium (Ti) is extensively used in the medical field because of its excellent biomechanical properties; however, how to precisely fabricate Ti surfaces at a nanoscale remains challenging. In this study, a DNA nanocoating system to functionalize Ti surfaces via a series of sequential reactions involving hydroxylation, silanization, and click chemistry is developed. Tetrahedral DNA nanostructures (TDNs) of two different sizes (≈7 and 30 nm) are assembled and characterized for subsequent surface attachment. In vitro and in vivo assays demonstrated significantly enhanced cell adhesion, spreading, proliferation, osteogenesis, and osseointegration on Ti surfaces modified with 30-nm TDNs, compared to slightly improved effects with 7-nm TDNs. Mechanistic studies showed that the focal adhesion pathway contributed to the enhanced bioaffinity of the 30-nm TDNs, as evidenced by the upregulated expression of vinculin and activation of the Akt signaling pathway. Moreover, under inflammatory or hypoxic conditions, Ti surfaces modified with 30-nm TDNs maintained excellent cellular performance comparable to that under normal conditions, suggesting a broader adaptability for DNA nanoparticles. Thus, better performance is achieved following modification with 30-nm TDNs. In summary, the proposed DNA-guided nanocoating system provides a novel and efficient strategy for the surface nanofabrication of Ti.
Collapse
Affiliation(s)
- Chenghui Qian
- Department of Multidisciplinary Consultant Center, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| | - Si Chen
- Department of Multidisciplinary Consultant Center, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| | - Liman Chen
- Fudan University Shanghai Cancer Center, the Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200433, China
| | - Chenyang Zhang
- Department of Oral Implantology, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| | - Lingyi Yang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Qiaowei Li
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Binbin Kang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Xiaohong Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Peter Mei
- Discipline of Orthodontics, Department of Oral Science, Faculty of Dentistry, University of Otago, Dunedin, 9016, New Zealand
| | - Hongzhou Gu
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| |
Collapse
|
7
|
Wang D, Wang Y, Zhang S, Yang X, Yang Y, Han T, Luo Y, Shui C, Yang M, Lin Y, Li C. Tetrahedral-DNA-Nanostructure-Modified Engineered Extracellular Vesicles Enhance Oral Squamous Cell Carcinomas Therapy by Targeting GPX4. ACS NANO 2025; 19:9351-9366. [PMID: 40014396 DOI: 10.1021/acsnano.5c00674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Oral squamous cell carcinoma (OSCC) represents a heterogeneous group of malignancies originating from the mucosal lining of the oral cavity. Current treatment modalities primarily involve surgery, chemotherapy, and radiotherapy. Despite the use of multimodal therapy, the 5 year overall survival rate for OSCC remains around 50%, underscoring the need for the development of nontoxic agents with potent antitumor activity. Extracellular vesicles (EVs) are nanoscale, membrane-bound structures that can selectively deliver small molecules, nucleic acids, and proteins to target cells, making them a promising platform for drug delivery in cancer therapy. Strategies to improve the uptake of EVs and enhance the delivery of therapeutic molecules to target cells are critical for advancing precision medicine. Tetrahedral DNA nanostructures (TDNs) have shown significant potential in facilitating drug endocytosis and delivery, as well as improving tissue penetration. In this study, TDN@EVs were conducted by modifying the membrane surface of M1-EVs with TDNs, which demonstrated improved biological stability and drug delivery efficiency compared to unmodified EVs. In vitro and in vivo experiments showed that TDN@EVs significantly inhibited OSCC cell proliferation and migration while promoting apoptosis. TDN@EVs exhibited superior drug penetration properties, further amplifying their antitumor effects. Proteomic analysis identified Hsc70 as the key protein responsible for the antitumor activity of the TDN@EVs. The efficient delivery of Hsc70 into tumor cells by TDN@EVs led to the degradation of GPX4, inducing ferroptosis, mitochondrial stress, and DNA damage in tumor cells. These findings highlight the potential of TDN@EVs as an effective and safe approach for cancer therapy. In conclusion, TDN@EVs present as a promising effective strategy for the targeted delivery of therapeutic agents in OSCC treatment, offering enhanced biological stability, efficient drug delivery, and significant antitumor effects.
Collapse
Affiliation(s)
- Dianri Wang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Yun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Sicheng Zhang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Xueting Yang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Yan Yang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Teng Han
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Yi Luo
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Chunyan Shui
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
- The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Mu Yang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chao Li
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center University of Electronic Science and Technology of China, Chengdu, Sichuan 610040, PR China
| |
Collapse
|
8
|
Wang R, Liu Y, Zhang Y, Yi Q, Xiao W, Wang T, Chen Q, Xiang J, Song L, Li C, Li F, Liu L, Li Q, Fan C, Mao X, Zuo X. DNA Framework-Enabled Ocular Barrier Penetration for Microinvasive Antiangiogenic Therapy. J Am Chem Soc 2025; 147:7545-7554. [PMID: 39979822 DOI: 10.1021/jacs.4c16529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Therapeutic aptamers targeting vascular endothelial growth factor A (VEGFA) have advanced the development of antiangiogenic drugs for treating choroidal neovascularization (CNV) diseases. However, despite FDA approval for use in neovascular age-related macular degeneration (nAMD), the effective in vivo delivery of therapeutic aptamers is hindered by ocular barriers and rapid degradation in biofluids. Here, we demonstrated a microinvasive delivery of VEGFA-targeted aptamers to the ocular fundus using tetrahedral framework nucleic acids (tFNAs). Upon incorporating anti-VEGFA aptamers to the tFNAs (apt-tFNA), we interrogated their penetration across the outer blood-retinal barrier (oBRB) to the innermost retinal in the eyeball, while maintaining their structural integrity. In addition, the apt-tFNA showed superior efficacy in inhibiting vascular proliferation and migration by neutralizing VEGFA. Furthermore, in a laser-induced CNV mouse model, subconjunctival injection of apt-tFNA exhibited comparable antiangiogenic efficacy to intravitreal ranibizumab, a monoclonal antibody fragment. These findings suggest that FNAs can effectively deliver therapeutic aptamers to the ocular fundus without compromising their antiangiogenic properties, highlighting their potential for microinvasive and feasible periocular administration in treating neovascular ophthalmic diseases.
Collapse
Affiliation(s)
- Ruobing Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yanhan Liu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuelu Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Qiuxue Yi
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wenjuan Xiao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tianqin Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qi Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jiayang Xiang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Song
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chunhong Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lin Liu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiuhai Mao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, and Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
9
|
Liu Y, Meng F, Feng W, Chen Z, Xing H, Zheng A. Oral DNA Vaccine Utilizing the Yeast Cell Wall for Dectin-1 Receptor-Mediated Enhancement of Mucosal Immunity. Mol Pharm 2025; 22:1241-1252. [PMID: 39960883 DOI: 10.1021/acs.molpharmaceut.4c00943] [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] [Indexed: 03/04/2025]
Abstract
Mucosal vaccines can generate localized mucosal immunity, effectively preventing initial pathogen infection and providing more effective protection. Oral vaccines are an attractive option for inducing mucosal immunity. The yeast cell wall, primarily composed of natural β-1,3-d glucan, can be recognized by the apical membrane receptor, dectin-1, which has a high expression on macrophages and intestinal M cells. In this study, by using vortexing methods to break yeast cell walls into nanometer-sized fragments, which retain the negatively charged β-glucan components on their surface and employing electrostatic adsorption/coextrusion techniques, these fragments were attached onto the surface of PS-DNA NPs, as verified by a scanning electron microscope (SEM), a transmission electron microscope (TEM), and dynamic light scattering (DLS) data. YCW-coated NPs (YNPs) showed greater drug stability compared to NPs in a simulated gastrointestinal environment. In vitro cell evaluation further demonstrated that YNPs were rapidly and efficiently taken up by antigen-presenting cells via receptor dectin-1-mediated endocytosis. In vivo experiments revealed that the oral vaccine elicited high levels of RBD-specific antibodies and triggered extensive cellular immunity in the intestinal mucosa. This study provides new insights into mucosal vaccine research.
Collapse
Affiliation(s)
- Yingqi Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fan Meng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wanting Feng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zehong Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Haonan Xing
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
10
|
Zhang Y, Li Y, Fang B, Du Y, Peng P. Framework Nucleic Acids: Innovative Tools for Cellular Sensing and Therapeutics. Chembiochem 2025; 26:e202400810. [PMID: 39653648 DOI: 10.1002/cbic.202400810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/07/2024] [Indexed: 12/18/2024]
Abstract
As emerging biomaterials, framework nucleic acids (FNAs) have recently demonstrated great potential in the biomedical field due to their high programmability, biocompatibility, unique structural diversity, and precise molecular design capabilities. This review focuses on the applications of FNAs in cellular sensing and disease treatment. First, we systematically introduce the applications of FNAs in cellular sensing, including their precise recognition and response to the extracellular tumor microenvironment, cell membrane proteins, and intracellular biomarkers. Subsequently, we review the potential of FNAs in disease treatment, covering their applications and development in drug delivery, regulation of cell behavior, and immunomodulation. We also discuss the limitations and potential role of FNAs in personalized medicine, precision diagnostics, and advanced therapies. The broad application of FNAs is expected to drive significant breakthroughs in future biomedical technological innovations and clinical translation.
Collapse
Affiliation(s)
- Yihan Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yuting Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Bowen Fang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yi Du
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Pai Peng
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| |
Collapse
|
11
|
Li S, Tian T, Zhang T, Lin Y, Cai X. A bioswitchable delivery system for microRNA therapeutics based on a tetrahedral DNA nanostructure. Nat Protoc 2025; 20:336-362. [PMID: 39215132 DOI: 10.1038/s41596-024-01050-7] [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: 12/07/2023] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
As microRNAs (miRNA) regulate almost all physiopathological activities in the human body, miRNA therapeutics that deliver miRNA regulators have attracted considerable attention in the field of nucleic acid drug development. The use of tetrahedral DNA nanostructures to deliver miRNA regulators is promising because of their simple fabrication, enhanced cell entry, effective tissue penetration, biocompatibility and functional editability. This protocol extension builds on our previous protocol for the use of tetrahedral DNA nanostructures and was designed to establish an updated bioswitchable delivery system (BDS) for achieving controlled cargo loading and release. A ribonuclease H-sensitive sequence is designed as a bioswitchable apparatus for the targeted release of the miRNA regulator. The functional sequence of the miRNA regulator and minimal secondary structure formation tendency during annealing are two key points in cargo design. We provide two BDS design strategies; BDS-A comprises an intact DNA tetrahedron with the RNA cargo hanging outside, offering the merits of lower cost, simplicity, and more direct structural design. In the BDS-B design, the RNA regulators are embedded into the DNA tetrahedron, which is beneficial for dermal tissue permeation applications. Following sequence design in Oligo 7 and Tiamat, the BDS assembly is completed and then ribonuclease H achieves controlled release of the miRNA regulator by triggering the bioswitchable apparatus. This is verified via polyacrylamide and agarose gel electrophoresis or fluorophore modifications. Both BDSs show promising cellular membrane permeability, tissue permeability and target inhibition in vitro and in vivo. The assembly and characterization of the BDS can be completed in 4 d, and the validation time for biostability and biological applications will depend on the specific use.
Collapse
Affiliation(s)
- Songhang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China.
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China.
| |
Collapse
|
12
|
Chen Y, Huang Y, Yang YR. DNA Nanotags for Multiplexed Single-Particle Electron Microscopy and In Situ Electron Cryotomography. JACS AU 2025; 5:17-27. [PMID: 39886579 PMCID: PMC11775714 DOI: 10.1021/jacsau.4c00986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025]
Abstract
DNA nanostructures present new opportunities as Nanotags for electron microscopy (EM) imaging, leveraging their high programmability, unique shapes, biomolecule conjugation capability, and stability compatible with standard cryogenic sample preparation protocols. This perspective highlights the potential of DNA Nanotags to enable high-throughput multiplexed EM analysis and facilitate in situ particle identification for cryogenic electron tomography (cryo-ET). Meanwhile, applying Nanotags in live-cell environments requires the efficient cellular uptake of intact structures and successful cytosolic migration. Promising strategies such as employing direct cytosolic delivery platforms and expressing RNA-based Nanotags in situ are discussed, while more systematic studies are needed to fully understand the intracellular trafficking and achieve precise localization of DNA Nanotags.
Collapse
Affiliation(s)
- Yuanfang Chen
- CAS Key Laboratory
of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence
in Nanoscience, National Center for Nanoscience
and Technology of China, CAS, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiqian Huang
- CAS Key Laboratory
of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence
in Nanoscience, National Center for Nanoscience
and Technology of China, CAS, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhe R. Yang
- CAS Key Laboratory
of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence
in Nanoscience, National Center for Nanoscience
and Technology of China, CAS, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Guo Y, Xiong T, Yan H, Zhang RX. Correlation of precisely fabricated geometric characteristics of DNA-origami nanostructures with their cellular entry in human lens epithelial cells. DISCOVER NANO 2025; 20:13. [PMID: 39841331 PMCID: PMC11754578 DOI: 10.1186/s11671-025-04188-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
Human lens epithelial cells (hLECs) are critical for lens transparency, and their aberrant metabolic activity and gene expression can lead to cataract. Intracellular delivery to hLECs, especially to sub-cellular organelles (e.g., mitochondrion and nucleus), is a key step in engineering cells for cell- and gene- based therapies. Despite a broad variety of nano- and microparticles can enter cells, their spatial characteristics relevant to cellular uptake and localization remains elusive. To investigate cellular internalization of nanostructures in hLECs, herein, DNA nanotechnology was exploited to precisely fabricate four distinct, mass-controlled DNA-origami nanostructures (DONs) through computer-aided design. Ensembled DONs included the rods, ring, triangle, and octahedron with defined geometric parameters of accessible surface area, effective volume, compactness, aspect ratio, size and vertex number. Atomic force microscopy and agarose gel electrophoresis showed that four DONs self-assembled within 3.5h with up to 59% yield and exhibited structural intactness in cell culture medium for 4 h. Flow cytometry analysis of four Cy5-labelled DONs in hLECs HLE-B3 found time-dependent cellular uptake over 2 h, among which the octahedron and triangle had higher cellular accumulation than the rod and ring. More importantly, the vertex number among other geometric parameters was positively correlated with cellular entry. Confocal images further revealed that four DONs had preferential localization at mitochondria to nucleus at 2 h in HLE-B3 cells, and the degree of their biodistribution varied among DONs as evidenced by Manders' correlation coefficient. This study demonstrates the DONs dependent cellular uptake and intracellular compartment localization in hLECs, heralding the future design of structure-modulating delivery of nanomedicine for ocular therapy.
Collapse
Affiliation(s)
- Yexuan Guo
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China
| | - Tianze Xiong
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China
| | - Hong Yan
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China.
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, 21 Jiefang Road, Xi'an, 710004, Shaanxi, China.
| | - Rui Xue Zhang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China.
| |
Collapse
|
14
|
Li Z, Ma Y, Li C, Xiao S, Liang H. Photo-Cross-linked DNA Structures Greatly Improves Their Serum Nuclease Resistances and Gene Knock-In Efficiencies. SMALL METHODS 2024:e2401346. [PMID: 39713911 DOI: 10.1002/smtd.202401346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/10/2024] [Indexed: 12/24/2024]
Abstract
The stabilization and structural integrity of DNA architectures remain significant challenges in their biomedical applications, particularly when inserting functional units into the genome using long single-stranded DNA (lssDNA). To address these challenges, a site-specific photo-cross-linking method is employed. Single-stranded oligonucleotides, containing one or two photosensitive cyanovinylcarbazole nucleoside (CNVK) molecules, are precisely incorporated and cross-linked at the specific sites of ssDNA through base-pairing, followed by rapid UV irradiation at 365 nm. This interstrand photo-cross-linking improves the thermal stability of DNA duplexes and allows this study to afford a tetrahedral DNA nanostructure in a yield of >94%. Most importantly, the photo-cross-linked DNA architectures exhibit high resistances against serum degradation, especially prevent digestion of exonuclease III (exo III), which is common in conventional lambda-processing method. Meanwhile, this photo-cross-linking treatment can significantly improve the knock-in (KI) efficiencies of lssDNA in different cells including 293T, K562, and HepG2, approximately three to eightfold those of the uncross-linked lssDNA, and remain a low cytotoxicity. Given the significantly enhanced nuclease resistance in serum and improved KI efficiencies, this study anticipates that this photo-cross-linking method will become a valuable tool in technologically advanced biomedical applications, such as nanotechnology and nucleic acid therapy.
Collapse
Affiliation(s)
- Zhigang Li
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Youwei Ma
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Chengxu Li
- Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shiyan Xiao
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Haojun Liang
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
15
|
Liang C, Huang Q, Zheng H, Duan M, Cheng X, Chen J, Li Q, Zhang Z, Fan C, Tian H, Shen J. Probing Cell Membrane Tension Using DNA Framework-Encoded Vibration-Induced Emission Molecular Assemblies. J Am Chem Soc 2024; 146:34341-34351. [PMID: 39641755 DOI: 10.1021/jacs.4c08271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Mechanosensitive fluorescent probes are valuable tools for detecting changes in cellular mechanics and viscosity. While numerous mechanosensitive probes have been developed, the construction of molecular assemblies for probing cellular mechanics remains largely unexplored, possibly due to the challenges of designing assemblies with synergistic and integrated functionalities. Here, we report the design and synthesis of mechanosensitive molecular assemblies by integrating DNA frameworks with vibration-induced emission (VIE) probes to enable live-cell membrane tension imaging. The molecular assemblies consist of a rigid tetrahedral DNA framework anchored with prescribed numbers of VIE probes. We find that VIE probes on the DNA framework retain their ratiometric fluorescence response characteristics in aqueous systems and on lipidic model membranes. Importantly, VIE assemblies exhibit distinct cell membrane targeting behaviors depending on the number of contact points between the molecular assemblies and the cell membrane. Especially, trivalent molecular assemblies can inhibit the internalization of the probes by the cells, a property absent in free VIE and mono/divalent molecular assemblies, thereby achieving targeted and prolonged retention on the cell membrane. Using the trivalent molecular assemblies, we successfully achieve ratiometric fluorescence imaging of cell membrane tension using confocal laser scanning microscopy, revealing the potential of such multifunctional mechanical-sensitive probes for live-cell applications.
Collapse
Affiliation(s)
- Chengpin Liang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiuling Huang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Haoran Zheng
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mulin Duan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinyi Cheng
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jielin Chen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiyun Zhang
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Joint International Research Laboratory for Precision Chemistry and Molecular Engineering, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - He Tian
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Joint International Research Laboratory for Precision Chemistry and Molecular Engineering, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Jianlei Shen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
16
|
Yue Z, Yang Y, Nie L, Sun Y, Wang Q, Lin Y, Gao Y, Cai X. A Binary siRNA-Loaded Tetrahedral DNA Nanobox for Synergetic Anti-Aging Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2408323. [PMID: 39690794 DOI: 10.1002/smll.202408323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Indexed: 12/19/2024]
Abstract
Extensive accumulation of senescent cells contributes to organismal aging, and slowing down the process of cellular senescence may ameliorate age-related pathologies. Targeted inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) is found to suppress the conversion of cells to senescence. The regulatory-associated protein of mTOR (Raptor), a key component of mTORC1, has been implicated as important in the aging process, and its druggability deserves to be investigated. Due to high efficiency and high convenience in drug construction, siRNA shows great potential in silencing Raptor expression via RNA interfering therapy. Here, we developed a functionalized anti-aging nanoplatform based on tetrahedral DNA nanostructures (TDNs) encapsulating siRNA targeting Raptor for synergistic anti-aging therapy, named siR-TDNbox. Anti-inflammatory and antioxidant properties of TDN beneficially attenuate age-associated inflammation while serving as siRNA nanocarrier, and thus play a binary role. The results suggest that the siR-TDNbox binary therapeutic nanoplatform has demonstrated an excellent ability to delay aging, inhibit mTORC1 signaling, and extend lifespan. This anti-aging nanoplatform may provide a medium for the combined application of traditional senotherapeutic drugs and promote the upgrading of nanomaterials with anti-aging effects.
Collapse
Affiliation(s)
- Ziqi Yue
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichen Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuezhang Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
17
|
Qi L, Hong S, Zhao T, Yan J, Ge W, Wang J, Fang X, Jiang W, Shen SG, Zhang L. DNA Tetrahedron Delivering miR-21-5p Promotes Senescent Bone Defects Repair through Synergistic Regulation of Osteogenesis and Angiogenesis. Adv Healthc Mater 2024; 13:e2401275. [PMID: 38979868 DOI: 10.1002/adhm.202401275] [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: 04/07/2024] [Revised: 06/08/2024] [Indexed: 07/10/2024]
Abstract
Compromised osteogenesis and angiogenesis is the character of stem cell senescence, which brought difficulties for bone defects repairing in senescent microenvironment. As the most abundant bone-related miRNA, miRNA-21-5p plays a crucial role in inducing osteogenic and angiogenic differentiation. However, highly efficient miR-21-5p delivery still confronts challenges including poor cellular uptake and easy degradation. Herein, TDN-miR-21-5p nanocomplex is constructed based on DNA tetrahedral (TDN) and has great potential in promoting osteogenesis and alleviating senescence of senescent bone marrow stem cells (O-BMSCs), simultaneously enhancing angiogenic capacity of senescent endothelial progenitor cells (O-EPCs). Of note, the activation of AKT and Erk signaling pathway may direct regulatory mechanism of TDN-miR-21-5p mediated osteogenesis and senescence of O-BMSCs. Also, TDN-miR-21-5p can indirectly mediate osteogenesis and senescence of O-BMSCs through pro-angiogenic growth factors secreted from O-EPCs. In addition, gelatin methacryloyl (GelMA) hydrogels are mixed with TDN and TDN-miR-21-5p to fabricate delivery scaffolds. TDN-miR-21-5p@GelMA scaffold exhibits greater bone repair with increased expression of osteogenic- and angiogenic-related markers in senescent critical-size cranial defects in vivo. Collectively, TDN-miR-21-5p can alleviate senescence and induce osteogenesis and angiogenesis in senescent microenvironment, which provides a novel candidate strategy for senescent bone repair and widen clinical application of TDNs-based gene therapy.
Collapse
Affiliation(s)
- Lei Qi
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Shebin Hong
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Tong Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jinge Yan
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weiwen Ge
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jing Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Xin Fang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Steve Gf Shen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Lei Zhang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| |
Collapse
|
18
|
Chen X, Xu Z, Gao Y, Chen Y, Yin W, Liu Z, Cui W, Li Y, Sun J, Yang Y, Ma W, Zhang T, Tian T, Lin Y. Framework Nucleic Acid-Based Selective Cell Catcher for Endogenous Stem Cell Recruitment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406118. [PMID: 39543443 DOI: 10.1002/adma.202406118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/23/2024] [Indexed: 11/17/2024]
Abstract
Cell-surface engineering holds great promise in boosting endogenous stem cell attraction for tissue regeneration. However, challenges such as cellular internalization of ligand and the dynamic nature of cell membranes often complicate ligand-receptor interactions. The aim of this study is to harness the innovative potential of programmable tetrahedral framework nucleic acid (tFNA) to enable precise, tunable ligand-receptor interactions, thereby improving stem cell recruitment efficiency. This approach involves experimental screening and theoretical analysis using dissipative particle dynamics. The results demonstrate that altering the flexibility and topology of ligands on tFNA changes their cellular internalization and membrane binding efficiency. Furthermore, optimizing the distribution of the mesenchymal stem cell (MSC)-binding aptamer 19S (Apt19S) on the tFNA enhances the stem cell capture efficiency. Following successful in vitro MSC capture, Apt19S-modified tFNA is chemically linked to a hyaluronic acid hydrogel, forming an efficient "stem cell catcher" system. Subsequent in vivo experiments demonstrate that this system effectively promotes early stem cell recruitment and accelerates bone regeneration in different bone healing scenarios, including cranial and maxillary defects.
Collapse
Affiliation(s)
- Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziang Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ye Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiafei Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
19
|
Wang Z, Wang X, He Y, Wu H, Mao R, Wang H, Qiu L. Exploring Framework Nucleic Acids: A Perspective on Their Cellular Applications. JACS AU 2024; 4:4110-4128. [PMID: 39610738 PMCID: PMC11600171 DOI: 10.1021/jacsau.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/30/2024]
Abstract
Cells are fundamental units of life. The coordination of cellular functions and behaviors relies on a cascade of molecular networks. Technologies that enable exploration and manipulation of specific molecular events in living cells with high spatiotemporal precision would be critical for pathological study, disease diagnosis, and treatment. Framework nucleic acids (FNAs) represent a novel class of nucleic acid materials characterized by their monodisperse and rigid nanostructure. Leveraging their exceptional programmability, convenient modification property, and predictable atomic-level architecture, FNAs have attracted significant attention in diverse cellular applications such as cell recognition, imaging, manipulation, and therapeutic interventions. In this perspective, we will discuss the utilization of FNAs in living cell systems while critically assessing the opportunities and challenges presented in this burgeoning field.
Collapse
Affiliation(s)
- Zhaoyang Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Xin Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yao He
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Rui Mao
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Liping Qiu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
20
|
Xie X, Liu Z, Xiang X, Wang S, Gao Z, Xu L, Ding F, Li Q. Mapping Endocytic Vesicular Acidification with a pH-Responsive DNA Nanomachine. Chembiochem 2024; 25:e202400363. [PMID: 39166897 DOI: 10.1002/cbic.202400363] [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: 04/20/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
Mapping the endocytic vesicular acidification process is of prior importance to better understand the health and pathological processes of cells. Herein, by integrating a pH-sensitive i-motif and a pair of fluorescence resonance energy transfer (FRET) into a tetrahedral DNA framework (TDF), we develop a pH-responsive DNA nanomachine, allowing for efficient sensing of pH from 7.0 to 5.5 via the pH-triggered spatial proximity modulation of FRET. The inheriting endo-lysosome-targeting ability of TDF enables spatiotemporal tracking of endocytic vesicle acidification during the endosomal maturation process. Analysis of pH-dependent FRET response at single fluorescent spot level reveals the significant difference of endocytic vesicular acidification between normal and cancer cells. The performance of pH-responsive DNA nanomachine underlines its potential for studies on vesicle acidification-related pathologies as a universal platform.
Collapse
Affiliation(s)
- Xiaodong Xie
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 20024, China
| | - Zhiyuan Liu
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 20024, China
| | - Xuelin Xiang
- Department of Liver Surgery, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Institute of Transplantation, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shaopeng Wang
- Department of Liver Surgery, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Institute of Transplantation, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhaoshuai Gao
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 20024, China
| | - Lifeng Xu
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 20024, China
| | - Fei Ding
- Department of Liver Surgery, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Institute of Transplantation, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 20024, China
- WLA Laboratories, World Laureates Association, Shanghai, 201203, China
| |
Collapse
|
21
|
Chen D, Xu Y, Wang Y, Li X, Yin D, Yan L. Diradicaloid-Loaded Polypeptide Nanoparticles for Two-Photon NIR Phototheranostics. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59907-59920. [PMID: 39441126 DOI: 10.1021/acsami.4c13338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Stable organic radicals, with unique electronic transitions from the ground state (D0) to the doublet excited state (D1), show promise as high-fluorescence quantum yield dyes. While organic small-molecule photosensitizers (PSs) have advanced for tumor photodynamic therapy (PDT), opportunities exist to enhance their performance and functionality. Herein, we synthesized Thiele's fluorocarbon derivative diradicaloid TFC-I with nearly 100% PLQY and integrated it into amphiphilic polypeptide nanoparticles, P-TI, using a precursor-doping approach. P-TI demonstrated notable features including high photostability, aggregation-induced emission, bright near-infrared fluorescence, substantial quantum yield (37% PLQY), robust near-infrared two-photon absorption (∼400 GM cross section), and superior ROS generation compared to commercial PSs. In vitro and in vivo experiments confirmed that P-TI performed well in mitochondria-targeted PDT, two-photon fluorescence imaging, and biosafety. This work highlights the use of organic stable radicals with precursor-doping for efficient PDT and deep tumor tissue imaging.
Collapse
Affiliation(s)
- Dejia Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| | - Yixuan Xu
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| | - Yating Wang
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| | - Xin Li
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| | - Dalong Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| | - Lifeng Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China, Jinzai Road 96, 230026 Hefei, Anhui, P. R. China
| |
Collapse
|
22
|
Huang K, Yang Q, Bao M, Wang S, Zhao L, Shi Q, Yang Y. Modulated Cell Internalization Behavior of Icosahedral DNA Framework with Programmable Surface Modification. J Am Chem Soc 2024; 146:21442-21452. [PMID: 39038211 DOI: 10.1021/jacs.4c04106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Surface modification could enhance the cell internalization efficiency of nanovehicles for targeted gene or drug delivery. However, the influence of surface modification parameters, including recognition manners, valences, and patterns, is often clouded, especially for the endocytosis of DNA nanostructures in customized shapes. Focusing on an icosahedral DNA framework, we systematically programmed three distinct types of ligands with diverse valence and spatial distribution on their outer surface to study the internalization efficiency, endocytic pathways, and postinternalization fate. The comparison in different aspects of parameters deepens our understanding of the intricate relationship between surface modification and cell entry behavior, offering insights crucial for designing and optimizing DNA framework nanostructures for potent cell-targeted purposes.
Collapse
Affiliation(s)
- Kui Huang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qiulan Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Min Bao
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shengwen Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200234, China
| | - Luming Zhao
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qian Shi
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yang Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
23
|
Zhu S, Peng H, Kong H, Yan Q, Xia K, Wang L, Zhu Y, Luo S. Visualization of the hepatic and renal cell uptake and trafficking of tetrahedral DNA origami in tumour. Cell Prolif 2024; 57:e13643. [PMID: 38572799 PMCID: PMC11294413 DOI: 10.1111/cpr.13643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
DNA nanostructures, known for their programmability, ease of modification, and favourable biocompatibility, have gained widespread application in the biomedical field. Among them, Tetrahedral DNA Origami (TDOs), as a novel DNA nanostructure, possesses well-defined structures, multiple modification sites, and large cavities, making it a promising drug carrier. However, current understanding of TDOs' interactions with biological systems, particularly with target cells and organs, remains unexplored, limiting its further applications in biomedicine. In this work, we prepared TDOs with an average particle size of 40 nm and labelled them with Cy5 fluorescent molecules. Following intravenous injection in mice, the uptake of TDOs by different types of liver and kidney cells was observed. Results indicated that TDOs accumulate in renal tubules and are metabolized by Kupffer cells, epithelial cells, and hepatocytes in the liver. Additionally, in a tumour-bearing mouse model, TDOs passively targeted tumour tissues and exhibited excellent tumour penetration and retention after rapid metabolism in hepatocytes. Our findings provide crucial insights for the development of TDO-based drug delivery systems.
Collapse
Affiliation(s)
- Shitai Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and TechnologyShanghai Institute of Applied Physics, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hongzhen Peng
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
- Institute of Materiobiology, College of Sciences, Shanghai UniversityShanghaiChina
| | - Huating Kong
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Qinglong Yan
- Institute of Materiobiology, College of Sciences, Shanghai UniversityShanghaiChina
- Xiangfu LaboratoryJiashanChina
| | - Kai Xia
- Xiangfu LaboratoryJiashanChina
- Shanghai Frontier Innovation Research InstituteShanghaiChina
| | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and TechnologyShanghai Institute of Applied Physics, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Materiobiology, College of Sciences, Shanghai UniversityShanghaiChina
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and TechnologyShanghai Institute of Applied Physics, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Materiobiology, College of Sciences, Shanghai UniversityShanghaiChina
| | - Shihua Luo
- Department of TraumatologyRui Jin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
24
|
Martins ASG, Reis SD, Benson E, Domingues MM, Cortinhas J, Vidal Silva JA, Santos SD, Santos NC, Pêgo AP, Moreno PMD. Enhancing Neuronal Cell Uptake of Therapeutic Nucleic Acids with Tetrahedral DNA Nanostructures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309140. [PMID: 38342712 DOI: 10.1002/smll.202309140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
The successful translation of therapeutic nucleic acids (NAs) for the treatment of neurological disorders depends on their safe and efficient delivery to neural cells, in particular neurons. DNA nanostructures can be a promising NAs delivery vehicle. Nonetheless, the potential of DNA nanostructures for neuronal cell delivery of therapeutic NAs is unexplored. Here, tetrahedral DNA nanostructures (TDN) as siRNA delivery scaffolds to neuronal cells, exploring the influence of functionalization with two different reported neuronal targeting ligands: C4-3 RNA aptamer and Tet1 peptide are investigated. Nanostructures are characterized in vitro, as well as in silico using molecular dynamic simulations to better understand the overall TDN structural stability. Enhancement of neuronal cell uptake of TDN functionalized with the C4-3 Aptamer (TDN-Apt), not only in neuronal cell lines but also in primary neuronal cell cultures is demonstrated. Additionally, TDN and TDN-Apt nanostructures carrying siRNA are shown to promote silencing in a process aided by chloroquine-induced endosomal disruption. This work presents a thorough workflow for the structural and functional characterization of the proposed TDN as a nano-scaffold for neuronal delivery of therapeutic NAs and for targeting ligands evaluation, contributing to the future development of new neuronal drug delivery systems based on DNA nanostructures.
Collapse
Affiliation(s)
- Ana S G Martins
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, s/n, Porto, 4200-465, Portugal
| | - Sara D Reis
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Erik Benson
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Tomtebodavägen 23, Solna, 171 65, Sweden
| | - Marco M Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - João Cortinhas
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Joana A Vidal Silva
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Sofia D Santos
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Ana P Pêgo
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal
| | - Pedro M D Moreno
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, INEB (Instituto Nacional de Engenharia Biomédica), Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| |
Collapse
|
25
|
Li Y, Wang S, Dong Y, Jin X, Wang J, Zhang H. Tetrahedral DNA-Based Functional MicroRNA-21 Delivery System: Application to Corneal Epithelial Wound Healing. Adv Healthc Mater 2024; 13:e2304381. [PMID: 38549217 DOI: 10.1002/adhm.202304381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/26/2024] [Indexed: 05/12/2024]
Abstract
Corneal injury occurs frequently which may lead to serious visual impairment. Rapid and efficient re-epithelialization after corneal epithelial injury is the key issue for maintaining corneal homeostasis. Among various treatment strategies, microRNA (miR)-based therapy shows great potential. However, structural limitations of miRNAs hinder its biomedical functionality. Nucleic acid nanotechnology is an appealing candidate for gene delivery because of its flexible modification and excellent biocompatibility. Herein, modified 3D tetrahedral framework nucleic acids (tFNAs) utilized as gene carriers for miR-21 delivery are constructed. TFNAs-miR-21 (T-21) shows great enzymatic resistance in extracellular environment and payload delivery into human corneal epithelial cells (HCECs) via clathrin-mediated endocytosis. T-21 facilitates proliferation and migration in HCECs via activating PI3K/AKT and ERK1/2 signaling pathways in vitro. In vivo studies, T-21 can be internalized by corneal epithelium in mice. In the mice corneal scratch model, T-21 ophthalmic solutions used as eye drops show no apparent side effects on the ocular surface histologically and exert great potential in accelerating corneal wound healing. These findings demonstrate that modified tFNAs are promising candidates for miRNA delivery for corneal wound healing. The convenient administration and great biocompatibility of tetrahedral DNA nanoparticles highlight its potential as gene transporter in solving ocular problems.
Collapse
Affiliation(s)
- Yulin Li
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| | - Shu Wang
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| | - Yueyan Dong
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| | - Xin Jin
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| | - Jingrao Wang
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| | - Hong Zhang
- Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Eye Hospital, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, China
| |
Collapse
|
26
|
Song L, Zuo X, Li M. Concept and Development of Algebraic Topological Framework Nucleic Acids. Chempluschem 2024; 89:e202300760. [PMID: 38529703 DOI: 10.1002/cplu.202300760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
Nucleic acids are considered as promising materials for developing exquisite nanostructures from one to three dimensions. The advances of DNA nanotechnology facilitate ingenious design of DNA nanostructures with diverse shapes and sizes. Especially, the algebraic topological framework nucleic acids (ATFNAs) are functional DNA nanostructures that engineer guest molecules (e. g., nucleic acids, proteins, small molecules, and nanoparticles) stoichiometrically and spatially. The intrinsic precise properties and tailorable functionalities of ATFNAs hold great promise for biological applications, such as cell recognition and immunotherapy. This Perspective highlights the concept and development of precisely assembled ATFNAs, and outlines the new frontiers and opportunities for exploiting the structural advantages of ATFNAs for biological applications.
Collapse
Affiliation(s)
- Lu Song
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Min Li
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| |
Collapse
|
27
|
Chen H, Li F, Ge Y, Liu J, Xing X, Li M, Ge Z, Zuo X, Fan C, Wang S, Wang F. DNA Framework-Enabled 3D Organization of Antiarrhythmic Drugs for Radiofrequency Catheter Ablation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401960. [PMID: 38843807 DOI: 10.1002/adma.202401960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/06/2024] [Indexed: 06/13/2024]
Abstract
Preorganizing molecular drugs within a microenvironment is crucial for the development of efficient and controllable therapeutic systems. Here, the use of tetrahedral DNA framework (TDF) is reported to preorganize antiarrhythmic drugs (herein doxorubicin, Dox) in 3D for catheter ablation, a minimally invasive treatment for fast heartbeats, aiming to address potential complications linked to collateral tissue damage and the post-ablation atrial fibrillation (AF) recurrence resulting from incomplete ablation. Dox preorganization within TDF transforms its random distribution into a confined, regular spatial arrangement governed by DNA. This, combined with the high affinity between Dox and DNA, significantly increases local Dox concentration. The exceptional capacity of TDF for cellular internalization leads to a 5.5-fold increase in intracellular Dox amount within cardiomyocytes, effectively promoting cellular apoptosis. In vivo investigations demonstrate that administering TDF-Dox reduces the recurrence rate of electrical conduction after radiofrequency catheter ablation (RFCA) to 37.5%, compared with the 77.8% recurrence rate in the free Dox-treated group. Notably, the employed Dox dosage exhibits negligible adverse effects in vivo. This study presents a promising treatment paradigm that strengthens the efficacy of catheter ablation and opens a new avenue for reconciling the paradox of ablation efficacy and collateral damage.
Collapse
Affiliation(s)
- Hangwei Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200800, China
| | - Fan Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yulong Ge
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200800, China
| | - Junyi Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200800, China
| | - Xing Xing
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200800, China
| | - Min Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhilei Ge
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shaopeng Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200800, China
| |
Collapse
|
28
|
Wu Y, Luo L, Hao Z, Liu D. DNA-based nanostructures for RNA delivery. MEDICAL REVIEW (2021) 2024; 4:207-224. [PMID: 38919398 PMCID: PMC11195427 DOI: 10.1515/mr-2023-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/28/2024] [Indexed: 06/27/2024]
Abstract
RNA-based therapeutics have emerged as a promising approach for the treatment of various diseases, including cancer, genetic disorders, and infectious diseases. However, the delivery of RNA molecules into target cells has been a major challenge due to their susceptibility to degradation and inefficient cellular uptake. To overcome these hurdles, DNA-based nano technology offers an unprecedented opportunity as a potential delivery platform for RNA therapeutics. Due to its excellent characteristics such as programmability and biocompatibility, these DNA-based nanostructures, composed of DNA molecules assembled into precise and programmable structures, have garnered significant attention as ideal building materials for protecting and delivering RNA payloads to the desired cellular destinations. In this review, we highlight the current progress in the design and application of three DNA-based nanostructures: DNA origami, lipid-nanoparticle (LNP) technology related to frame guided assembly (FGA), and DNA hydrogel for the delivery of RNA molecules. Their biomedical applications are briefly discussed and the challenges and future perspectives in this field are also highlighted.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Beijing SupraCirc Biotechnology Co., Ltd, Beijing, China
| | - Liangzhi Luo
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ziyang Hao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Dongsheng Liu
- Department of Chemistry, Engineering Research Center of Advanced Rare Earth Materials, (Ministry of Education), Tsinghua University, Beijing, China
| |
Collapse
|
29
|
Xu K, Du Y, Xu B, Huang Y, Feng W, Yu D, Chen Y, Wang X. Gelatin-Encapsulated Tetrahedral DNA Nanostructure Enhances Cellular Internalization for Treating Noise-Induced Hearing Loss. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310604. [PMID: 38329190 DOI: 10.1002/smll.202310604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/23/2023] [Indexed: 02/09/2024]
Abstract
Nanoparticle-based drug delivery strategies have emerged as a crucial avenue for comprehensive sensorineural hearing loss treatment. Nevertheless, developing therapy vectors crossing both biological and cellular barriers has encountered significant challenges deriving from various external factors. Herein, the rational integration of gelatin nanoparticles (GNPs) with tetrahedral DNA nanostructures (TDNs) to engineer a distinct drug-delivery nanosystem (designed as TDN@GNP) efficiently enhances the biological permeability and cellular internalization, further resolving the dilemma of noise-induced hearing loss via loading epigallocatechin gallate (EGCG) with anti-lipid peroxidation property. Rationally engineering of TDN@GNP demonstrates dramatic alterations in the physicochemical key parameters of TDNs that are pivotal in cell-particle interactions and promote cellular uptake through multiple endocytic pathways. Furthermore, the EGCG-loaded nanosystem (TDN-EGCG@GNP) facilitates efficient inner ear drug delivery by superior permeability through the biological barrier (round window membrane), maintaining high drug concentration within the inner ear. The TDN-EGCG@GNP actively overcomes the cell membrane, exhibiting hearing protection from noise insults via reduced lipid peroxidation in outer hair cells and spiral ganglion neurons. This work exemplifies how integrating diverse vector functionalities can overcome biological and cellular barriers in the inner ear, offering promising applications for inner ear disorders.
Collapse
Affiliation(s)
- Ke Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200100, P. R. China
| | - Yiwei Du
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200100, P. R. China
| | - Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yuqi Huang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200100, P. R. China
| |
Collapse
|
30
|
Wang Y, Xiong Y, Shi K, Effah CY, Song L, He L, Liu J. DNA nanostructures for exploring cell-cell communication. Chem Soc Rev 2024; 53:4020-4044. [PMID: 38444346 DOI: 10.1039/d3cs00944k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The process of coordinating between the same or multiple types of cells to jointly execute various instructions in a controlled and carefully regulated environment is a very appealing field. In order to provide clearer insight into the role of cell-cell interactions and the cellular communication of this process in their local communities, several interdisciplinary approaches have been employed to enhance the core understanding of this phenomenon. DNA nanostructures have emerged in recent years as one of the most promising tools in exploring cell-cell communication and interactions due to their programmability and addressability. Herein, this review is dedicated to offering a new perspective on using DNA nanostructures to explore the progress of cell-cell communication. After briefly outlining the anchoring strategy of DNA nanostructures on cell membranes and the subsequent dynamic regulation of DNA nanostructures, this paper highlights the significant contribution of DNA nanostructures in monitoring cell-cell communication and regulating its interactions. Finally, we provide a quick overview of the current challenges and potential directions for the application of DNA nanostructures in cellular communication and interactions.
Collapse
Affiliation(s)
- Ya Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Yamin Xiong
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kangqi Shi
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Clement Yaw Effah
- The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou 450003, China
| | - Lulu Song
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Jianbo Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, China.
| |
Collapse
|
31
|
Wang Z, Ren X, Li Y, Qiu L, Wang D, Liu A, Liang H, Li L, Yang B, Whittaker AK, Liu Z, Jin S, Lin Q, Wang T. Reactive Oxygen Species Amplifier for Apoptosis-Ferroptosis Mediated High-Efficiency Radiosensitization of Tumors. ACS NANO 2024; 18:10288-10301. [PMID: 38556985 DOI: 10.1021/acsnano.4c01625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Insufficient reactive oxygen species (ROS) production and radioresistance have consistently contributed to the failure of radiotherapy (RT). The development of a biomaterial capable of activating ROS-induced apoptosis and ferroptosis is a potential strategy to enhance RT sensitivity. To achieve precision and high-efficiency RT, the theranostic nanoplatform Au/Cu nanodots (Au/CuNDs) were designed for dual-mode imaging, amplifying ROS generation, and inducing apoptosis-ferroptosis to sensitize RT. A large amount of ROS is derived from three aspects: (1) When exposed to ionizing radiation, Au/CuNDs effectively absorb photons and emit various electrons, which can interact with water to produce ROS. (2) Au/CuNDs act as a catalase-like to produce abundant ROS through Fenton reaction with hydrogen peroxide overexpressed of tumor cells. (3) Au/CuNDs deplete overexpressed glutathione, which causes the accumulation of ROS. Large amounts of ROS and ionizing radiation further lead to apoptosis by increasing DNA damage, and ferroptosis by enhancing lipid peroxidation, significantly improving the therapeutic efficiency of RT. Furthermore, Au/CuNDs serve as an excellent nanoprobe for high-resolution near-infrared fluorescence imaging and computed tomography of tumors. The promising dual-mode imaging performance shows their potential application in clinical cancer detection and imaging-guided precision RT, minimizing damage to adjacent normal tissues during RT. In summary, our developed theranostic nanoplatform integrates dual-mode imaging and sensitizes RT via ROS-activated apoptosis-ferroptosis, offering a promising prospect for clinical cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ze Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xiaojun Ren
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Yunfeng Li
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Ling Qiu
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Dongzhou Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Liang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zhongshan Liu
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| |
Collapse
|
32
|
Wang W, Chopra B, Walawalkar V, Liang Z, Adams R, Deserno M, Ren X, Taylor RE. Cell-Surface Binding of DNA Nanostructures for Enhanced Intracellular and Intranuclear Delivery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15783-15797. [PMID: 38497300 PMCID: PMC10995898 DOI: 10.1021/acsami.3c18068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
DNA nanostructures (DNs) have found increasing use in biosensing, drug delivery, and therapeutics because of their customizable assembly, size and shape control, and facile functionalization. However, their limited cellular uptake and nuclear delivery have hindered their effectiveness in these applications. Here, we demonstrate the potential of applying cell-surface binding as a general strategy to enable rapid enhancement of intracellular and intranuclear delivery of DNs. By targeting the plasma membrane via cholesterol anchors or the cell-surface glycocalyx using click chemistry, we observe a significant 2 to 8-fold increase in the cellular uptake of three distinct types of DNs that include nanospheres, nanorods, and nanotiles, within a short time frame of half an hour. Several factors are found to play a critical role in modulating the uptake of DNs, including their geometries, the valency, positioning and spacing of binding moieties. Briefly, nanospheres are universally preferable for cell surface attachment and internalization. However, edge-decorated nanotiles compensate for their geometry deficiency and outperform nanospheres in both categories. In addition, we confirm the short-term structural stability of DNs by incubating them with cell medium and cell lysate. Further, we investigate the endocytic pathway of cell-surface bound DNs and reveal that it is an interdependent process involving multiple pathways, similar to those of unmodified DNs. Finally, we demonstrate that cell-surface attached DNs exhibit a substantial enhancement in the intranuclear delivery. Our findings present an application that leverages cell-surface binding to potentially overcome the limitations of low cellular uptake, which may strengthen and expand the toolbox for effective cellular and nuclear delivery of DNA nanostructure systems.
Collapse
Affiliation(s)
- Weitao Wang
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Bhavya Chopra
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Vismaya Walawalkar
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Zijuan Liang
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Rebekah Adams
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Markus Deserno
- Department
of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Xi Ren
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Rebecca E. Taylor
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
33
|
Han Y, Xi L, Leng F, Xu C, Zheng Y. Topical Delivery of microRNA-125b by Framework Nucleic Acids for Psoriasis Treatment. Int J Nanomedicine 2024; 19:2625-2638. [PMID: 38505169 PMCID: PMC10950082 DOI: 10.2147/ijn.s441353] [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: 10/16/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024] Open
Abstract
Purpose Psoriasis is a chronic and recurrent inflammatory dermatitis characterized by T cell imbalance and abnormal keratinocyte proliferation. MicroRNAs (miRNAs) hold promise as therapeutic agents for this disease; however, their clinical application is hindered by poor stability and limited skin penetration. This study demonstrates the utilization of Framework Nucleic Acid (FNA) for the topical delivery of miRNAs in psoriasis treatment. Methods By utilizing miRNA-125b as the model drug, FNA-miR-125b was synthesized via self-assembly. The successful synthesis and stability of FNA-miR-125b in bovine fetal serum (FBS) were verified through gel electrophoresis. Subsequently, flow cytometry was employed to investigate the cell internalization on HaCaT cells, while qPCR determined the effects of FNA-miR-125b on cellular functions. Additionally, the skin penetration ability of FNA-miR-125b was assessed. Finally, a topical administration study involving FNA-miR-125b cream on imiquimod (IMQ)-induced psoriasis mice was conducted to evaluate its therapeutic efficacy. Results The FNA-miR-125b exhibited excellent stability, efficient cellular internalization, and potent inhibition of keratinocyte proliferation. In the psoriasis mouse model, FNA-miR-125b effectively penetrated the skin tissue, resulting in reduced epidermal thickness and PASI score, as well as decreased levels of inflammatory cytokines.
Collapse
Affiliation(s)
- Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Fang Leng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| |
Collapse
|
34
|
Xu Y, Yan ZS, Ma YQ, Ding HM. Topology- and size-dependent binding of DNA nanostructures to the DNase I. Int J Biol Macromol 2024; 257:128703. [PMID: 38072351 DOI: 10.1016/j.ijbiomac.2023.128703] [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: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
The susceptibility of DNA nanomaterials to enzymatic degradation in biological environments is a significant obstacle limiting their broad applications in biomedicine. While DNA nanostructures exhibit some resistance to nuclease degradation, the underlying mechanism of this resistance remains elusive. In this study, the interaction of tetrahedral DNA nanostructures (TDNs) and double-stranded DNA (dsDNA) with DNase I is investigated using all-atom molecular dynamics simulations. Our results indicate that DNase I can effectively bind to all dsDNA molecules, and certain key residues strongly interact with the nucleic bases of DNA. However, the binding of DNase I to TDNs exhibits a non-monotonic behavior based on size; TDN15 and TDN26 interact weakly with DNase I (∼ - 75 kcal/mol), whereas TDN21 forms a strong binding with DNase I (∼ - 110 kcal/mol). Furthermore, the topological properties of the DNA nanostructures are analyzed, and an under-twisting (∼32°) of the DNA helix is observed in TDN15 and TDN26. Importantly, this under-twisting results in an increased width of the minor groove in TDN15 and TDN26, which primarily explains their reduced binding affinity to DNase I comparing to the dsDNA. Overall, this study demonstrated a novel mechanism for local structural control of DNA at the nanoscale by adjusting the twisting induced by length.
Collapse
Affiliation(s)
- Yao Xu
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Zeng-Shuai Yan
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu-Qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China.
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| |
Collapse
|
35
|
Zhao HY, Chen YQ, Luo XY, Cai MJ, Li JY, Lin XY, Zhang H, Ding HM, Jiang GL, Hu Y. Ligand Phase Separation-Promoted, "Squeezing-Out" Mode Explaining the Mechanism and Implications of Neutral Nanoparticles That Escaped from Lysosomes. ACS NANO 2024; 18:2162-2183. [PMID: 38198577 DOI: 10.1021/acsnano.3c09452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Neutral nanomaterials functionalized with PEG or similar molecules have been popularly employed as nanomedicines. Compared to positive counterparts that are capable of harnessing the well-known proton sponge effect to facilitate their escape from lysosomes, it is yet unclear how neutral substances got their entry into the cytosol. In this study, by taking PEGylated, neutral Au nanospheres as an example, we systematically investigated their time-dependent translocation postuptake. Specifically, we harnessed dissipative particle dynamics simulations to uncover how nanospheres bypass lysosomal entrapment, wherein a mechanism termed as "squeezing-out" mode was discovered. We next conducted a comprehensive investigation on how nanomaterials implicate lysosomes in terms of integrity and functionality. By using single-molecule imaging, specific preservation of PEG-terminated with targeting moieties in lysosomes supports the "squeezing-out" mode as the mechanism underlying the lysosomal escape of nanomaterials. All evidence points out that such a process is benign to lysosomes, wherein the escape of nanomaterials proceeds at the expense of targeting moieties loss. Furthermore, we proved that by fine-tuning of the efficacy of nanomaterials escaping from lysosomes, modulation of distinct pathways and metabolic machinery can be achieved readily, thereby offering us a simple and robust tool to implicate cells.
Collapse
Affiliation(s)
- Hui-Yue Zhao
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| | - Yuan-Qiang Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou, 215031, China
| | - Xing-Yu Luo
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| | - Ming-Jie Cai
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| | - Jia-Yi Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xin-Yu Lin
- School of Stomatology, Nanjing Medical University, Nanjing, 211166, China
| | - Hao Zhang
- Department of Oncology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou, 215031, China
| | - Guang-Liang Jiang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yong Hu
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| |
Collapse
|
36
|
Wang M, Ni SD, Yin YW, Ma YQ, Ding HM. Molecular Modeling of the Fluorination Effect on the Penetration of Nanoparticles across Lipid Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1295-1304. [PMID: 38173387 DOI: 10.1021/acs.langmuir.3c02817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The fluorinated decorations have recently been widely used in many biomedical applications. However, the potential mechanism of the fluorination effect on the cellular delivery of nanoparticles (NPs) still remains elusive. In this work, we systemically explore the penetration of a perfluoro-octanethiol-coated gold NP (PF-Au NP) and, for comparison, an octanethiol-coated gold NP (OT-Au NP) across lipid bilayers. We also investigated the effect of these two types of NPs on the properties of lipid bilayers. Our findings indicate that the lipid type and the surface tension of the lipid bilayer significantly impact the penetration capabilities of the fluorinated gold NP. By examining the distribution of ligands on the surface of the two types of NPs in water and during the penetration process, we unveil their distinct penetration characteristics. Specifically, the PF-Au NP exhibits amphiphobic behavior (both hydrophobic and lipophobic), while the OT-Au NP exhibits solely hydrophobic characteristics. Finally, we observe that the penetration capabilities can be increased by adjusting the degree of fluorination of the ligands on the NP surface. Overall, this study provides useful physical insights into the unique properties of the fluorinated decorations in NP permeation.
Collapse
Affiliation(s)
- Min Wang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Song-Di Ni
- Shanghai Marine Electronic Equipment Research Institute, Shanghai 201100, China
| | - Yue-Wen Yin
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Yu-Qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| |
Collapse
|
37
|
Kosara S, Singh R, Bhatia D. Structural DNA nanotechnology at the nexus of next-generation bio-applications: challenges and perspectives. NANOSCALE ADVANCES 2024; 6:386-401. [PMID: 38235105 PMCID: PMC10790967 DOI: 10.1039/d3na00692a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
DNA nanotechnology has significantly progressed in the last four decades, creating nucleic acid structures widely used in various biological applications. The structural flexibility, programmability, and multiform customization of DNA-based nanostructures make them ideal for creating structures of all sizes and shapes and multivalent drug delivery systems. Since then, DNA nanotechnology has advanced significantly, and numerous DNA nanostructures have been used in biology and other scientific disciplines. Despite the progress made in DNA nanotechnology, challenges still need to be addressed before DNA nanostructures can be widely used in biological interfaces. We can open the door for upcoming uses of DNA nanoparticles by tackling these issues and looking into new avenues. The historical development of various DNA nanomaterials has been thoroughly examined in this review, along with the underlying theoretical underpinnings, a summary of their applications in various fields, and an examination of the current roadblocks and potential future directions.
Collapse
Affiliation(s)
- Sanjay Kosara
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj Gujarat 382355 India
| | - Ramesh Singh
- Department of Mechanical Engineering, Colorado State University Fort Collins CO USA
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj Gujarat 382355 India
| |
Collapse
|
38
|
Zhu S, Yan Q, Wang L, Zhu Y, Luo S. Noninvasive Framework Nucleic Acid Eye Drops for Retinal Administration. ACS APPLIED BIO MATERIALS 2023; 6:5078-5085. [PMID: 37861694 DOI: 10.1021/acsabm.3c00760] [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] [Indexed: 10/21/2023]
Abstract
Intravitreal injection is widely employed for the treatment of retinal diseases. However, it suffers from various drawbacks, including ocular trauma, risk of infection, and poor patient compliance due to frequent administrations. Due to the presence of barriers such as the cornea, it has been a challenge to develop efficient noninvasive ophthalmic eye drops that can reach the retina. Framework nucleic acids (FNAs), known for their excellent biocompatibility and precise, controllable shape and size, have been extensively utilized in drug delivery application. Here, we report the development of size- and shape-resolved fluorescent DNA frameworks for noninvasive retinal administration. Results show that tetrahedral DNA nanostructures (TDNs) with an edge length of 20 bp can reach the retina within 6 h with the highest efficiency. Moreover, this delivery method exhibits excellent biocompatibility. Our findings provide an approach for the development of localized treatment strategies for retinal diseases using FNA-based nanocarriers.
Collapse
Affiliation(s)
- Shitai Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Institute of Materiobiology, Shanghai University, Shanghai 200444, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Institute of Materiobiology, Shanghai University, Shanghai 200444, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shihua Luo
- Department of Traumatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
39
|
Wang Z, Wang D, Ren X, Liu Z, Liu A, Li X, Guan L, Shen Y, Jin S, Zvyagin AV, Yang B, Wang T, Lin Q. One Stone, Three Birds: Multifunctional Nanodots as "Pilot Light" for Guiding Surgery, Enhanced Radiotherapy, and Brachytherapy of Tumors. ACS CENTRAL SCIENCE 2023; 9:1976-1988. [PMID: 37901175 PMCID: PMC10604975 DOI: 10.1021/acscentsci.3c00994] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 10/31/2023]
Abstract
Surgery, radiotherapy (RT), and brachytherapy are crucial treatments for localized deep tumors. However, imprecise tumor location often leads to issues such as positive surgical margins, extended radiotherapy target volumes, and radiation damage to healthy tissues. Reducing side effects in healthy tissue and enhancing RT efficacy are critical challenges. To address these issues, we developed a multifunctional theranostic platform using Au/Ag nanodots (Au/AgNDs) that act as a "pilot light" for real-time guided surgery, high-efficiency RT, and brachytherapy, achieving a strategy of killing three birds with one stone. First, dual-mode imaging of Au/AgNDs enabled precision RT, minimizing damage to adjacent normal tissue during X-ray irradiation. Au/AgNDs enhanced ionizing radiation energy deposition, increased intracellular reactive oxygen species (ROS) generation, regulated the cell cycle, promoted DNA damage formation, and inhibited DNA repair in tumor cells, significantly improving RT efficacy. Second, in brachytherapy, precise guidance provided by dual-mode imaging addressed challenges related to non-visualization of existing interstitial brachytherapy and multiple adjustments of insertion needle positions. Meanwhile, the effect of brachytherapy was improved. Third, the excellent fluorescence imaging of Au/AgNDs accurately distinguished tumors from normal tissue, facilitating their use as a powerful tool for assisting surgeons during tumor resection. Taken together, our multifunctional theranostic platform offers real-time guidance for surgery and high-efficiency RT, and improves brachytherapy precision, providing a novel strategy and vision for the clinical diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Ze Wang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Dongzhou Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Xiaojun Ren
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Zhongshan Liu
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Annan Liu
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xingchen Li
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lin Guan
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yannan Shen
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Shunzi Jin
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Andrei V. Zvyagin
- Australian
Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute
of Biology and Biomedicine, Lobachevsky
Nizhny Novgorod State University, 603105 Nizhny Novgorod, Russia
| | - Bai Yang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Tiejun Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Quan Lin
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
40
|
Fischer A, Ehrlich A, Plotkin Y, Ouyang Y, Asulin K, Konstantinos I, Fan C, Nahmias Y, Willner I. Stimuli-Responsive Hydrogel Microcapsules Harnessing the COVID-19 Immune Response for Cancer Therapeutics. Angew Chem Int Ed Engl 2023; 62:e202311590. [PMID: 37675854 DOI: 10.1002/anie.202311590] [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: 08/09/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
The combination of gene therapy and immunotherapy concepts, along recent advances in DNA nanotechnology, have the potential to provide important tools for cancer therapies. We present the development of stimuli-responsive microcapsules, loaded with a viral immunogenetic agent, harnessing the immune response against the Coronavirus Disease 2019, COVID-19, to selectively attack liver cancer cells (hepatoma) or recognize breast cancer or hepatoma, by expression of green fluorescence protein, GFP. The pH-responsive microcapsules, modified with DNA-tetrahedra nanostructures, increased hepatoma permeation by 50 %. Incorporation of a GFP-encoding lentivirus vector inside the tumor-targeting pH-stimulated miRNA-triggered and Alpha-fetoprotein-dictated microcapsules enables the demonstration of neoplasm selectivity, with approximately 5,000-, 8,000- and 50,000-fold more expression in the cancerous cells, respectively. The incorporation of the SARS-CoV-2 spike protein in the gene vector promotes specific recognition of the immune-evading hepatoma by the COVID-19-analogous immune response, which leads to cytotoxic and inflammatory activity, mediated by serum components taken from vaccinated or recovered COVID-19 patients, resulting in effective elimination of the hepatoma (>85 % yield).
Collapse
Affiliation(s)
- Amit Fischer
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Avner Ehrlich
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Yevgeni Plotkin
- The Department of Anesthesiology, Critical Care and Pain Medicine, Hadassah University Hospital, Jerusalem, 9112001, Israel
- Faculty of Medicine, Hebrew University of Jerusalem Jerusalem, 9112001, (Israel)
| | - Yu Ouyang
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Klil Asulin
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Ioannidis Konstantinos
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yaakov Nahmias
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Itamar Willner
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
41
|
Wang X, Xing X, Lu S, Du G, Zhang Y, Ren Y, Sun Y, Sun J, Fan Q, Liu K, Wang F, Ye F. Collective cell behaviors manipulated by synthetic DNA nanostructures. FUNDAMENTAL RESEARCH 2023; 3:809-812. [PMID: 38933288 PMCID: PMC11197778 DOI: 10.1016/j.fmre.2022.01.031] [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: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 12/01/2022] Open
Abstract
Cellular collective motion in confluent epithelial monolayers is involved in many processes such as embryo development, carcinoma invasion, and wound healing. The development of new chemical strategies to achieve large-scale control of cells' collective motion is essential for biomedical applications. Here a series of DNA nanostructures with different dimensions were synthesized and their influences on cells' collective migration and packing behaviors in epithelial monolayers were investigated. We found that the framed DNA nanoassemblies effectively reduced the cells' speed by increasing the rigidity of cells, while the lipid-DNA micelles had a more pronounced effect on cells' projection area and shape factor. These DNA nanostructures all significantly enhanced the dependence of cells' speed on their shape factor. Our results indicate that cells' mobility in monolayers can be manipulated by chemical intercellular interactions without any genetic intervention. This may provide a new chemical strategy for tissue engineering and tumor therapy.
Collapse
Affiliation(s)
- Xiaochen Wang
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325001, China
| | - Xiwen Xing
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shuang Lu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Guangle Du
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325001, China
| | - Yi Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yubin Ren
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yulong Sun
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325001, China
| | - Jing Sun
- Institute of Organic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Qihui Fan
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Kai Liu
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Fan Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325001, China
- Songshan Lake Materials Laboratory, Dongguan 523808, China
| |
Collapse
|
42
|
Yuwen L, Zhang S, Chao J. Recent Advances in DNA Nanotechnology-Enabled Biosensors for Virus Detection. BIOSENSORS 2023; 13:822. [PMID: 37622908 PMCID: PMC10452139 DOI: 10.3390/bios13080822] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/05/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
Virus-related infectious diseases are serious threats to humans, which makes virus detection of great importance. Traditional virus-detection methods usually suffer from low sensitivity and specificity, are time-consuming, have a high cost, etc. Recently, DNA biosensors based on DNA nanotechnology have shown great potential in virus detection. DNA nanotechnology, specifically DNA tiles and DNA aptamers, has achieved atomic precision in nanostructure construction. Exploiting the programmable nature of DNA nanostructures, researchers have developed DNA nanobiosensors that outperform traditional virus-detection methods. This paper reviews the history of DNA tiles and DNA aptamers, and it briefly describes the Baltimore classification of virology. Moreover, the advance of virus detection by using DNA nanobiosensors is discussed in detail and compared with traditional virus-detection methods. Finally, challenges faced by DNA nanobiosensors in virus detection are summarized, and a perspective on the future development of DNA nanobiosensors in virus detection is also provided.
Collapse
Affiliation(s)
- Lihui Yuwen
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (L.Y.); (S.Z.)
| | - Shifeng Zhang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (L.Y.); (S.Z.)
| | - Jie Chao
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
43
|
Wang W, Hayes PR, Ren X, Taylor RE. Synthetic Cell Armor Made of DNA Origami. NANO LETTERS 2023; 23:7076-7085. [PMID: 37463308 PMCID: PMC10416349 DOI: 10.1021/acs.nanolett.3c01878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/11/2023] [Indexed: 07/20/2023]
Abstract
The bioengineering applications of cells, such as cell printing and multicellular assembly, are directly limited by cell damage and death due to a harsh environment. Improved cellular robustness thus motivates investigations into cell encapsulation, which provides essential protection. Here we target the cell-surface glycocalyx and cross-link two layers of DNA nanorods on the cellular plasma membrane to form a modular and programmable nanoshell. We show that the DNA origami nanoshell modulates the biophysical properties of cell membranes by enhancing the membrane stiffness and lowering the lipid fluidity. The nanoshell also serves as armor to protect cells and improve their viability against mechanical stress from osmotic imbalance, centrifugal forces, and fluid shear stress. Moreover, it enables mediated cell-cell interactions for effective and robust multicellular assembly. Our results demonstrate the potential of the nanoshell, not only as a cellular protection strategy but also as a platform for cell and cell membrane manipulation.
Collapse
Affiliation(s)
- Weitao Wang
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Peter R. Hayes
- Department
of Chemical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Xi Ren
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Rebecca E. Taylor
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
44
|
Chen H, Dong X, Ou L, Ma C, Yuan B, Yang K. Thermal-controlled cellular uptake of "hot" nanoparticles. NANOSCALE 2023; 15:12718-12727. [PMID: 37470374 DOI: 10.1039/d3nr02449k] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Nanoparticles (NPs) have shown immense potential in the field of biomedical applications, particularly in NP-based photothermal therapy, which offers a remote-controlled approach to achieve precise temperature control for site-specific heating and sub-cellular tumor treatment. However, the molecular mechanisms underlying related cellular activities, such as the cellular uptake behavior of irradiated NPs in photothermal effects, remain elusive. In this study, we conducted a thorough investigation of the interaction between an irradiated NP with elevated temperature (ranging from 270 to 360 K) and a model bilayer membrane composed of DPPC or DOPC using nonequilibrium coarse-grained molecular dynamics simulations with the implicit-solvent Dry Martini force field. We observe that the interaction between a "hot" NP and a membrane is thermally regulated. In addition, the wrapping of membranes around NPs exhibits a strong dependence on the temperature of the irradiated NP, demonstrating a step-like change in behavior. This membrane wrapping effect is attributed to the heat conduction between NPs and membrane lipids, which occurs almost simultaneously with the membrane deformation and wrapping of NPs during the NP-membrane interaction process. Especially, during the process of heat conduction, a gel-to-fluid phase transition of the membrane may occur, which plays a crucial role in determining the deformation behavior of the membrane. Moreover, it is found that the membrane lipids in the two leaflets exhibit obvious and asymmetric molecular-level responses to heat flux, characterized by significant changes in packing states (e.g., the order parameter of lipid tails and area per lipid) and possible interdigitation between lipids. Furthermore, the thermal-controlled wrapping effect is tightly linked to the properties of NPs (e.g., size, NP-lipid affinity) and lipid species. Our findings are valuable for comprehending the thermal-regulated cellular internalization of NPs and offer insights into devising strategies to precisely modulate NP endocytosis by exploiting the interplay between heating and NP properties.
Collapse
Affiliation(s)
- Haibo Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Xuewei Dong
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Luping Ou
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Chiyun Ma
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| |
Collapse
|
45
|
Hong S, Jiang W, Ding Q, Lin K, Zhao C, Wang X. The Current Progress of Tetrahedral DNA Nanostructure for Antibacterial Application and Bone Tissue Regeneration. Int J Nanomedicine 2023; 18:3761-3780. [PMID: 37457798 PMCID: PMC10348378 DOI: 10.2147/ijn.s403882] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Recently, programmable assembly technologies have enabled the application of DNA in the creation of new nanomaterials with unprecedented functionality. One of the most common DNA nanostructures is the tetrahedral DNA nanostructure (TDN), which has attracted great interest worldwide due to its high stability, simple assembly procedure, high predictability, perfect programmability, and excellent biocompatibility. The unique spatial structure of TDN allows it to penetrate cell membranes in abundance and regulate cellular biological properties as a natural genetic material. Previous studies have demonstrated that TDNs can regulate various cellular biological properties, including promoting cells proliferation, migration and differentiation, inhibiting cells apoptosis, as well as possessing anti-inflammation and immunomodulatory capabilities. Furthermore, functional molecules can be easily modified at the vertices of DNA tetrahedron, DNA double helix structure, DNA tetrahedral arms or DNA tetrahedral cage structure, enabling TDN to be used as a nanocarrier for a variety of biological applications, including targeted therapies, molecular diagnosis, biosensing, antibacterial treatment, antitumor strategies, and tissue regeneration. In this review, we mainly focus on the current progress of TDN-based nanomaterials for antimicrobial applications, bone and cartilage tissue repair and regeneration. The synthesis and characterization of TDN, as well as the biological merits are introduced. In addition, the challenges and prospects of TDN-based nanomaterials are also discussed.
Collapse
Affiliation(s)
- Shebin Hong
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| | - Qinfeng Ding
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| | - Kaili Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| | - Cancan Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| | - Xudong Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
| |
Collapse
|
46
|
Baral B, Nial PS, Subudhi U. Enhanced enzymatic activity and conformational stability of catalase in presence of tetrahedral DNA nanostructures: A biophysical and kinetic study. Int J Biol Macromol 2023; 242:124677. [PMID: 37141969 DOI: 10.1016/j.ijbiomac.2023.124677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/07/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
The emergence of DNA nanotechnology has shown enormous potential in a vast array of applications, particularly in the medicinal and theranostics fields. Nevertheless, the knowledge of the compatibility between DNA nanostructures and cellular proteins is largely unknown. Herein, we report the biophysical interaction between proteins (circulatory protein bovine serum albumin, BSA, and the cellular enzyme bovine liver catalase, BLC) and tetrahedral DNA (tDNAs), which are well-known nanocarriers for therapeutics. Interestingly, the secondary conformation of BSA or BLC was unaltered in the presence of tDNAs which supports the biocompatible property of tDNA. In addition, thermodynamic studies showed that the binding of tDNAs with BLC has a stable non-covalent interaction via hydrogen bond and van der Waals contact, which is indicative of a spontaneous reaction. Furthermore, the catalytic activity of BLC was increased in the presence of tDNAs during 24 h of incubation. These findings indicate that the presence of tDNA nanostructures not only ensures a steady secondary conformation of proteins, but also stabilize the intracellular proteins like BLC. Surprisingly, our investigation discovered that tDNAs have no effect on albumin proteins, either by interfering or by adhering to the extracellular proteins. These findings will aid in the design of future DNA nanostructures for biomedical applications by increasing the knowledge on the biocompatible interaction of tDNAs with biomacromolecules.
Collapse
Affiliation(s)
- Bineeth Baral
- DNA Nanotechnology & Application Laboratory, CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751013, Odisha, India; School of Biological Sciences, Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Partha S Nial
- DNA Nanotechnology & Application Laboratory, CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751013, Odisha, India; School of Biological Sciences, Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Umakanta Subudhi
- DNA Nanotechnology & Application Laboratory, CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751013, Odisha, India; School of Biological Sciences, Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
47
|
Tian R, Shang Y, Wang Y, Jiang Q, Ding B. DNA Nanomaterials-Based Platforms for Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201518. [PMID: 36651129 DOI: 10.1002/smtd.202201518] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/29/2022] [Indexed: 05/17/2023]
Abstract
The past few decades have witnessed the evolving paradigm for cancer therapy from nonspecific cytotoxic agents to selective, mechanism-based therapeutics, especially immunotherapy. In particular, the integration of nanomaterials with immunotherapy is proven to improve the therapeutic outcome and minimize off-target toxicity in the treatment. As a novel nanomaterial, DNA-based self-assemblies featuring uniform geometries, feasible modifications, programmability, surface addressability, versatility, and intrinsic biocompatibility, are extensively exploited for innovative and effective cancer immunotherapy. In this review, the successful employment of DNA nanoplatforms for cancer immunotherapy, including the delivery of immunogenic cell death inducers, adjuvants and vaccines, immune checkpoint blockers as well as the application in immune cell engineering and adoptive cell therapy is summarized. The remaining challenges and future perspectives regarding the pharmacokinetics/pharmacodynamics, in vivo fate and immunogenicity of DNA materials, and the design of intelligent DNA nanomedicine for individualized cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Run Tian
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingxu Shang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing, 100190, China
| | - Yiming Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing, 100190, China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
48
|
Wang Z, Liu A, Li X, Guan L, Xing H, He L, Fang L, Zvyagin AV, Yang X, Yang B, Lin Q. Multifunctional nanoprobe for multi-mode imaging and diagnosis of metastatic prostate cancer. Talanta 2023; 256:124255. [PMID: 36652761 DOI: 10.1016/j.talanta.2023.124255] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
The high incidence and complex subtypes of prostate cancer put forward higher requirements for accurate diagnosis. Furthermore, advanced prostate cancer is prone to metastasis. Single biological imaging mode faces a challenge of sensitive and fast bioimaging of metastasic prostate cancer. Thus, exploring a nanoprobe with multi-mode imaging function has an important impact on preoperative imaging and intraoperative visualization guide of metastatic prostate cancer. Herein, based on the optical properties and X-ray attenuation capability of Au nanodots as well as the slow electronic relaxation of Gd3+, we designed and fabricated the multifunctional nanoprobe Au/Gd nanodots for multi-mode imaging and accurate diagnosis of bone metastatic prostate cancer. The results showed that multiple imaging modes complement each other to achieve high-precision of metastasic prostate cancer detection and accurately guide treatment. In addition, in vitro/vivo experiments showed that Au/Gd nanodots had good biocompatibility and biosafety. Therefore, the prepared multifunctional nanoprobe may provide new strategies and insights for precise diagnosis of metastatic prostate cancer in clinical practice.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Xingchen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Huiyuan Xing
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Liang He
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, Jilin, PR China.
| | - Linan Fang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130000, PR China.
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW, 2109, Australia; Australia and Institute of Biology and Biomedicine, Lobachevsky Nizhny Novgorod State University, 603105, Nizhny Novgorod, Russia
| | - Xiaoyu Yang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
49
|
Ding F, Zhang S, Chen Q, Feng H, Ge Z, Zuo X, Fan C, Li Q, Xia Q. Immunomodulation with Nucleic Acid Nanodevices. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206228. [PMID: 36599642 DOI: 10.1002/smll.202206228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/09/2022] [Indexed: 06/17/2023]
Abstract
The precise regulation of interactions of specific immunological components is crucial for controllable immunomodulation, yet it remains a great challenge. With the assistance of advanced computer design, programmable nucleic acid nanotechnology enables the customization of synthetic nucleic acid nanodevices with unprecedented geometrical and functional precision, which have shown promising potential for precise immunoengineering. Notably, the inherently immunologic functions of nucleic acids endow these nucleic acid-based assemblies with innate advantages in immunomodulatory engagement. In this review, the roles of nucleic acids in innate immunity are discussed, focusing on the definition, immunologic modularity, and enhanced bioavailability of structural nucleic acid nanodevices. In light of this, molecular programming and precise organization of functional modules with nucleic acid nanodevices for immunomodulation are emphatically reviewed. At last, the present challenges and future perspectives of nucleic acid nanodevices for immunomodulation are discussed.
Collapse
Affiliation(s)
- Fei Ding
- Shanghai Institute of Transplantation, Department of Liver Surgery, Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Shuangye Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Qian Chen
- Shanghai Institute of Transplantation, Department of Liver Surgery, Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Hao Feng
- Shanghai Institute of Transplantation, Department of Liver Surgery, Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Zhilei Ge
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xiaolei Zuo
- Shanghai Institute of Transplantation, Department of Liver Surgery, Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
- WLA Laboratories, World Laureates Association, Shanghai, 201203, P. R. China
| | - Qiang Xia
- Shanghai Institute of Transplantation, Department of Liver Surgery, Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| |
Collapse
|
50
|
Xu K, Li S, Zhou Y, Gao X, Mei J, Liu Y. Application of Computing as a High-Practicability and -Efficiency Auxiliary Tool in Nanodrugs Discovery. Pharmaceutics 2023; 15:1064. [PMID: 37111551 PMCID: PMC10144056 DOI: 10.3390/pharmaceutics15041064] [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: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/28/2023] Open
Abstract
Research and development (R&D) of nanodrugs is a long, complex and uncertain process. Since the 1960s, computing has been used as an auxiliary tool in the field of drug discovery. Many cases have proven the practicability and efficiency of computing in drug discovery. Over the past decade, computing, especially model prediction and molecular simulation, has been gradually applied to nanodrug R&D, providing substantive solutions to many problems. Computing has made important contributions to promoting data-driven decision-making and reducing failure rates and time costs in discovery and development of nanodrugs. However, there are still a few articles to examine, and it is necessary to summarize the development of the research direction. In the review, we summarize application of computing in various stages of nanodrug R&D, including physicochemical properties and biological activities prediction, pharmacokinetics analysis, toxicological assessment and other related applications. Moreover, current challenges and future perspectives of the computing methods are also discussed, with a view to help computing become a high-practicability and -efficiency auxiliary tool in nanodrugs discovery and development.
Collapse
Affiliation(s)
- Ke Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangkai Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou 510700, China
| |
Collapse
|