1
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
2
|
Lee M, Hwang J, Song Y, Kim S, Park N. Anti-miR21-conjugated DNA nanohydrogel for enhanced cancer therapy. BIOMATERIALS ADVANCES 2024; 169:214160. [PMID: 39731836 DOI: 10.1016/j.bioadv.2024.214160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
MicroRNAs (miRNAs) are non-coding, endogenous small single-stranded RNA molecules involved in post-transcriptional regulation of gene expression. It has been demonstrated that dysregulation of miRNA plays a major role in tumor formation, proliferation, and metastasis. Therefore, the delivery of anti-miRNA oligonucleotides to block the activity of these oncogenic miRNAs is a high-potential anti-cancer therapy approach. In particular, miRNA-21 (miR-21) can be an excellent target as it is an oncogenic miRNA that is upregulated in various cancers including glioblastoma, breast cancer, and colon cancer. However, anti-miRNAs are unstable in the physiological environment and have low cell membrane permeability, making it difficult to accumulate at certain concentrations to have anti-cancer effects within cancer cells. To overcome these difficulties, we developed anti-miR-21 functionalized DNA hydrogel (amiR-21 Dgel). We confirmed the improved physiological stability of amiR-21 Dgel in vitro, and observed that it blocked up to 96.6 % of miR-21 in HeLa cells, and reduced cell viability down to 77.9 % for 72 h. In particular, RT-qPCR analysis demonstrated that blocking of miR-21 induces increased mRNA expression of the tumor suppressor genes, PTEN and PDCD4 by 6.23- and 6.87-fold, respectively. In addition, the Dgel can act as a drug delivery vehicle, intercalating anticancer drugs such as doxorubicin (Dox) to be delivered into cells. DOX, an anticancer drug, showed a synergistic anticancer effect with amiR-21, which was delivered together. We expect that this approach will be a convenient to optimization and highly effective strategy for anticancer therapy employing antisense miRNAs.
Collapse
Affiliation(s)
- Minhyuk Lee
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jimin Hwang
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Youngseo Song
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| | - Nokyoung Park
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea.
| |
Collapse
|
3
|
Ye J, Cui H, Liu E, Pei X, Chai M, Sun L, Wang D, Yang VC, Yu F. Temperature switchable linkers suitable for triggered drug release in cancer thermo-chemotherapy. Int J Pharm 2024; 666:124757. [PMID: 39332459 DOI: 10.1016/j.ijpharm.2024.124757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/11/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
In drug delivery systems, a stimuli-responsive linker that attaches a targeting carrier and a cytotoxic payload can be dissociated to release the payload on the target over the action of a stimuli, thereby it would harden the selectivity, specificity and potency of the cytotoxic agent against targeted tissues whilst sparing the drug-induced toxicity on normal cells. Oligonucleotide duplexes can unwind and be separated into single-stranded random coils under a defined temperature, and this property makes the oligonucleotide an appealing thermo-responsive linker. In this work, we studied the melting temperatures of different DNA linkers with various lengths and mismatches inserted in the double helix with either different numbers or positions. We further chose the DNA linkers that can unwind at the hyperthermia temperature and used them in the construction of four different drug delivery systems both in vitro and in vivo. Results showed that the chosen DNA linkers in all of the constructed delivery systems can successfully unwind and release cargos or drugs after application of heat compared to control groups. This research demonstrated the potential applications of DNA duplexes as temperature-sensitive linkers of drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Junxiao Ye
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; School of Pharmaceutical Sciences , Tsinghua University, Beijing 100084, China
| | - Hui Cui
- YUGEN MEDCH (Tianjin) Co., Ltd, Tianjin 300450, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xing Pei
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Meihong Chai
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Lu Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Dongmei Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Victor C Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, Ann Arbor, MI 48109-1065, USA
| | - Fei Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
4
|
Farasatkia A, Maeso L, Gharibi H, Dolatshahi-Pirouz A, Stojanovic GM, Edmundo Antezana P, Jeong JH, Federico Desimone M, Orive G, Kharaziha M. Design of nanosystems for melanoma treatment. Int J Pharm 2024; 665:124701. [PMID: 39278291 DOI: 10.1016/j.ijpharm.2024.124701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Melanoma is a prevalent and concerning form of skin cancer affecting millions of individuals worldwide. Unfortunately, traditional treatments can be invasive and painful, prompting the need for alternative therapies with improved efficacy and patient outcomes. Nanosystems offer a promising solution to these obstacles through the rational design of nanoparticles (NPs) which are structured into nanocomposite forms, offering efficient approaches to cancer treatment procedures. A range of NPs consisting of polymeric, metallic and metal oxide, carbon-based, and virus-like NPs have been studied for their potential in treating skin cancer. This review summarizes the latest developments in functional nanosystems aimed at enhancing melanoma treatment. The fundamentals of these nanosystems, including NPs and the creation of various functional nanosystem types, facilitating melanoma treatment are introduced. Then, the advances in the applications of functional nanosystems for melanoma treatment are summarized, outlining both their benefits and the challenges encountered in implementing nanosystem therapies.
Collapse
Affiliation(s)
- Asal Farasatkia
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Lidia Maeso
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Hamidreza Gharibi
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | | | - Goran M Stojanovic
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000, Novi Sad, Serbia
| | - Pablo Edmundo Antezana
- Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA, CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Jee-Heon Jeong
- Laboratory of Drug Delivery and Cell Therapy (LDDCT). Department of Precision Medicine. School of Medicine, Sungkyunkwan University. South Korea
| | - Martin Federico Desimone
- Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA, CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina; Instituto de Ciências Biológicas (ICB), Universidade Federal do Rio Grande - FURG, Rio Grande, RS, Brazil
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria 01007, Spain.
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| |
Collapse
|
5
|
Liu H, Li X, Yan R, Yang J, Lu Q, Wang L. DNA tetrahedron nanoparticles service as a help carrier and adjvant of mRNA vaccine. J Transl Med 2024; 22:1024. [PMID: 39543727 PMCID: PMC11566622 DOI: 10.1186/s12967-024-05837-w] [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: 07/02/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
AIM OF THE STUDY To investigate the potential of DNA nanoparticles (DNPs) as carriers and adjuvants for mRNA vaccines. MATERIALS AND METHODS Customized oligonucleotides were assembled into DNA tetrahedra (DNA-TH), which were subsequently complexed with streptavidin and mRNA encoding green fluorescent protein (GFP). Various assays were conducted to evaluat the stability of the DNPs, their cellular uptake, immune activation potential, and GFP mRNA transcription efficiency. P53-mutant HSC-3 cells were used to establish a subcutaneous xenograft tumor model to explore the effects of DNPs as carriers and adjuvants in a disease model. RESULTS The DNPs were remained stable extracellularly and rapidly taken up by antigen-presenting cells. Compared to naked GFP mRNA, DNPs statistically significantly activated immune responses and facilitated GFP mRNA transcription and protein expression both in vitro and in vivo. Immunization with DNP-GFP mRNA complexes induced higher antibody titers compared to naked mRNA. The DNPs demonstrated good biocompatibility. DNP-p53 inhibited the growth of subcutaneous xenograft tumors in mice with p53-mutant HSC-3 cells, outperforming both the naked p53 mRNA and blank control groups, with a statistically significant difference (P < 0.05). CONCLUSION DNA nanoparticles show promise for improving mRNA vaccine delivery and efficacy. Further optimization of these nanoparticles could lead to highly effective mRNA vaccine carriers with broad applications.
Collapse
Affiliation(s)
- Henglang Liu
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xianxian Li
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruike Yan
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qun Lu
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lili Wang
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Hu Y, Chen F, Lu H, Tan S, Ke Y, Loh WW, Soh EJH, Taniya A, Tabaglio T, Wee DKB, Ying JY. A splice-switch oligonucleotide loaded self-cleavable DNA nanogel. Chem Commun (Camb) 2024; 60:11516-11519. [PMID: 39308402 DOI: 10.1039/d4cc01942c] [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: 10/09/2024]
Abstract
A self-cleavable DNA nanogel loaded with splice-switch oligonucleotide (SSO) has been developed. Under acidic conditions (pH 5.0), cleavage of the acid-labile chemical linker and generation of the i-motif structure led to the disintegration of the DNA nanogel and efficient release of SSO in its unaltered native state.
Collapse
Affiliation(s)
- Yuwei Hu
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- NanoBio Lab, A*STAR, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Feng Chen
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- NanoBio Lab, A*STAR, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), A*STAR, 1 Pesek Road Jurong Island, Singapore 627833, Republic of Singapore
| | - Hongfang Lu
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- NanoBio Lab, A*STAR, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, The Proteos, Singapore 138673, Republic of Singapore
| | - Susi Tan
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- NanoBio Lab, A*STAR, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, The Proteos, Singapore 138673, Republic of Singapore
| | - Yujie Ke
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- School of Interdisciplinary Studies, Lingnan University, Tuen Mun, Hong Kong SAR, China
| | - Wei Wei Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
| | - Eugene Jia Hao Soh
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
| | - Agarwal Taniya
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, The Proteos, Singapore 138673, Republic of Singapore
| | - Tommaso Tabaglio
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, The Proteos, Singapore 138673, Republic of Singapore
| | - Dave Keng Boon Wee
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, The Proteos, Singapore 138673, Republic of Singapore
| | - Jackie Y Ying
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore.
- NanoBio Lab, A*STAR, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- Bioengineering and Nanomedicine Department, King Faisal Specialist Hospital & Research Centre, PO Box 3354, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
7
|
Han S, Zou J, Xiao F, Xian J, Liu Z, Li M, Luo W, Feng C, Kong N. Nanobiotechnology boosts ferroptosis: opportunities and challenges. J Nanobiotechnology 2024; 22:606. [PMID: 39379969 PMCID: PMC11460037 DOI: 10.1186/s12951-024-02842-5] [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: 06/14/2024] [Accepted: 09/07/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, and autophagy, is a unique type of cell death driven by iron-dependent phospholipid peroxidation. Since ferroptosis was defined in 2012, it has received widespread attention from researchers worldwide. From a biochemical perspective, the regulation of ferroptosis is strongly associated with cellular metabolism, primarily including iron metabolism, lipid metabolism, and redox metabolism. The distinctive regulatory mechanism of ferroptosis holds great potential for overcoming drug resistance-a major challenge in treating cancer. The considerable role of nanobiotechnology in disease treatment has been widely reported, but further and more systematic discussion on how nanobiotechnology enhances the therapeutic efficacy on ferroptosis-associated diseases still needs to be improved. Moreover, while the exciting therapeutic potential of ferroptosis in cancer has been relatively well summarized, its applications in other diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular diseases, and kidney disease, remain underreported. Consequently, it is necessary to fill these gaps to further complete the applications of nanobiotechnology in ferroptosis. In this review, we provide an extensive introduction to the background of ferroptosis and elaborate its regulatory network. Subsequently, we discuss the various advantages of combining nanobiotechnology with ferroptosis to enhance therapeutic efficacy and reduce the side effects of ferroptosis-associated diseases. Finally, we analyze and discuss the feasibility of nanobiotechnology and ferroptosis in improving clinical treatment outcomes based on clinical needs, as well as the current limitations and future directions of nanobiotechnology in the applications of ferroptosis, which will not only provide significant guidance for the clinical applications of ferroptosis and nanobiotechnology but also accelerate their clinical translations.
Collapse
Affiliation(s)
- Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Jianhua Zou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jing Xian
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Meng Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Chan Feng
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
8
|
Lee M, Lee M, Song Y, Kim S, Park N. Recent Advances and Prospects of Nucleic Acid Therapeutics for Anti-Cancer Therapy. Molecules 2024; 29:4737. [PMID: 39407665 PMCID: PMC11477775 DOI: 10.3390/molecules29194737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Nucleic acid therapeutics are promising alternatives to conventional anti-cancer therapy, such as chemotherapy and radiation therapy. While conventional therapies have limitations, such as high side effects, low specificity, and drug resistance, nucleic acid therapeutics work at the gene level to eliminate the cause of the disease. Nucleic acid therapeutics treat diseases in various forms and using different mechanisms, including plasmid DNA (pDNA), small interfering RNA (siRNA), anti-microRNA (anti-miR), microRNA mimics (miRNA mimic), messenger RNA (mRNA), aptamer, catalytic nucleic acid (CNA), and CRISPR cas9 guide RNA (gRNA). In addition, nucleic acids have many advantages as nanomaterials, such as high biocompatibility, design flexibility, low immunogenicity, small size, relatively low price, and easy functionalization. Nucleic acid therapeutics can have a high therapeutic effect by being used in combination with various nucleic acid nanostructures, inorganic nanoparticles, lipid nanoparticles (LNPs), etc. to overcome low physiological stability and cell internalization efficiency. The field of nucleic acid therapeutics has advanced remarkably in recent decades, and as more and more nucleic acid therapeutics have been approved, they have already demonstrated their potential to treat diseases, including cancer. This review paper introduces the current status and recent advances in nucleic acid therapy for anti-cancer treatment and discusses the tasks and prospects ahead.
Collapse
Affiliation(s)
- Minhyuk Lee
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Minjae Lee
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Youngseo Song
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Nokyoung Park
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, Republic of Korea
| |
Collapse
|
9
|
Feng H, Ang K, Guan P, Li J, Meng H, Yang J, Fan L, Sun Y. Application of adhesives in the treatment of cartilage repair. INTERDISCIPLINARY MEDICINE 2024; 2. [DOI: 10.1002/inmd.20240015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/08/2024] [Indexed: 01/04/2025]
Abstract
AbstractFrom degeneration causing intervertebral disc issues to trauma‐induced meniscus tears, diverse factors can injure the different types of cartilage. This review highlights adhesives as a promising and rapidly implemented repair strategy. Compared to traditional techniques such as sutures and wires, adhesives offer several advantages. Importantly, they seamlessly connect with the injured tissue, deliver bioactive substances directly to the repair site, and potentially alleviate secondary problems like inflammation or degeneration. This review delves into the cutting‐edge advancements in adhesive technology, specifically focusing on their effectiveness in cartilage injury treatment and their underlying mechanisms. We begin by exploring the material characteristics of adhesives used in cartilage tissue, focusing on essential aspects like adhesion, biocompatibility, and degradability. Subsequently, we investigate the various types of adhesives currently employed in this context. Our discussion then moves to the unique role adhesives play in addressing different cartilage injuries. Finally, we acknowledge the challenges currently faced by this promising technology.
Collapse
Affiliation(s)
- Haoyang Feng
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Kai Ang
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Pengfei Guan
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Junji Li
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| | - Huan Meng
- Postdoc Cartilage Biology AO Research Institute Davos Davos Platz Wellington Switzerland
| | - Jian Yang
- Biomedical Engineering Program School of Engineering Westlake University Hangzhou China
| | - Lei Fan
- Department of Orthopedic Surgery Nanfang Hospital Southern Medical University Guangzhou China
| | - Yongjian Sun
- Department of Pediatric Orthopedics The Third Affiliated Hospital of Southern Medical University Guangzhou China
| |
Collapse
|
10
|
Miao Y, Fu C, Yu Z, Yu L, Tang Y, Wei M. Current status and trends in small nucleic acid drug development: Leading the future. Acta Pharm Sin B 2024; 14:3802-3817. [PMID: 39309508 PMCID: PMC11413693 DOI: 10.1016/j.apsb.2024.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/15/2024] [Accepted: 04/12/2024] [Indexed: 09/25/2024] Open
Abstract
Small nucleic acid drugs, composed of nucleotides, represent a novel class of pharmaceuticals that differ significantly from conventional small molecule and antibody-based therapeutics. These agents function by selectively targeting specific genes or their corresponding messenger RNAs (mRNAs), further modulating gene expression and regulating translation-related processes. Prominent examples within this category include antisense oligonucleotides (ASO), small interfering RNAs (siRNAs), microRNAs (miRNAs), and aptamers. The emergence of small nucleic acid drugs as a focal point in contemporary biopharmaceutical research is attributed to their remarkable specificity, facile design, abbreviated development cycles, expansive target spectrum, and prolonged activity. Overcoming challenges such as poor stability, immunogenicity, and permeability issues have been addressed through the integration of chemical modifications and the development of drug delivery systems. This review provides an overview of the current status and prospective trends in small nucleic acid drug development. Commencing with a historical context, we introduce the primary classifications and mechanisms of small nucleic acid drugs. Subsequently, we delve into the advantages of the U.S. Food and Drug Administration (FDA) approved drugs and mainly discuss the challenges encountered during their development. Apart from researching chemical modification and delivery system that efficiently deliver and enrich small nucleic acid drugs to target tissues, promoting endosomal escape is a critical scientific question and important research direction in siRNA drug development. Future directions in this field will prioritize addressing these challenges to facilitate the clinical transformation of small nucleic acid drugs.
Collapse
Affiliation(s)
- Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
- Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| | - Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Tang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
- Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| |
Collapse
|
11
|
Wu R, Li W, Yang P, Shen N, Yang A, Liu X, Ju Y, Lei L, Fang B. DNA hydrogels and their derivatives in biomedical engineering applications. J Nanobiotechnology 2024; 22:518. [PMID: 39210464 PMCID: PMC11360341 DOI: 10.1186/s12951-024-02791-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Deoxyribonucleotide (DNA) is uniquely programmable and biocompatible, and exhibits unique appeal as a biomaterial as it can be precisely designed and programmed to construct arbitrary shapes. DNA hydrogels are polymer networks comprising cross-linked DNA strands. As DNA hydrogels present programmability, biocompatibility, and stimulus responsiveness, they are extensively explored in the field of biomedicine. In this study, we provide an overview of recent advancements in DNA hydrogel technology. We outline the different design philosophies and methods of DNA hydrogel preparation, discuss its special physicochemical characteristics, and highlight the various uses of DNA hydrogels in biomedical domains, such as drug delivery, biosensing, tissue engineering, and cell culture. Finally, we discuss the current difficulties facing DNA hydrogels and their potential future development.
Collapse
Affiliation(s)
- Rui Wu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Wenting Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine, Peking Union Medical College, Beijing, 100000, China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Naisi Shen
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Anqi Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xiangjun Liu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
12
|
Li C, Wang M, Li PF, Sheng J, Fu Q. Construction of Smart DNA-Based Drug Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306257. [PMID: 38377302 DOI: 10.1002/smll.202306257] [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: 07/24/2023] [Revised: 02/10/2024] [Indexed: 02/22/2024]
Abstract
Due to the disadvantages of poor targeting, slow action, and low effectiveness of current commonly used cancer treatments, including surgery, chemotherapy, and radiotherapy, researchers have turned to DNA as a biomaterial for constructing drug delivery nanocarriers. DNA is favored for its biocompatibility and programmability. In order to overcome the limitations associated with traditional drug delivery systems (DDSs), researchers have developed smart-responsive DNA DDSs that can control drug release in response to specific physical or chemical stimuli at targeted sites. In this review, a summary of multiple targeted ligand structures is provided, various shapes of stable DNA nanomaterials, and different stimuli-responsive drug release strategies in DNA DDSs. Specifically, targeted cell recognition, in vivo stable transport, and controlled drug release of smart DDSs are focused. Finally, the further development prospects and challenges of clinical application of DNA nanomaterials in the field of smart drug delivery are discussed. The objective of this review is to enhance researchers' comprehension regarding the potential application of DNA nanomaterials in precision drug delivery, with the aim of expediting the clinical implementation of intelligent DDSs.
Collapse
Affiliation(s)
- Congcong Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Junyue Sheng
- Qingdao No.58 High School of Shandong Province, 20 Jiushui Road, Qingdao, 266100, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
13
|
Hu Y, Liu J, Ke Y, Wang B, Lim JYC, Dong Z, Long Y, Willner I. Oligo-Adenine and Cyanuric Acid Supramolecular DNA-Based Hydrogels Exhibiting Acid-Resistance and Physiological pH-Responsiveness. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29235-29247. [PMID: 38769743 DOI: 10.1021/acsami.4c03834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Expanding the functions and applications of DNA by integrating noncanonical bases and structures into biopolymers is a continuous scientific effort. An adenine-rich strand (A-strand) is introduced as functional scaffold revealing, in the presence of the low-molecular-weight cofactor cyanuric acid (CA, pKa 6.9), supramolecular hydrogel-forming efficacies demonstrating multiple pH-responsiveness. At pH 1.2, the A-strand transforms into a parallel A-motif duplex hydrogel cross-linked by AH+-H+A units due to the protonation of adenine (pKa 3.5). At pH 5.2, and in the presence of coadded CA, a helicene-like configuration is formed between adenine and protonated CA, generating a parallel A-CA triplex cross-linked hydrogel. At pH 8.0, the hydrogel undergoes transition into a liquid state by deprotonation of CA cofactor units and disassembly of A-CA triplex into its constituent components. Density functional theory calculations and molecular dynamics simulations, supporting the structural reconfigurations of A-strand in the presence of CA, are performed. The sequential pH-stimulated hydrogel states are rheometrically characterized. The hydrogel framework is loaded with fluorescein-labeled insulin, and the pH-stimulated release of insulin from the hydrogel across the pH barriers present in the gastrointestinal tract is demonstrated. The results provide principles for future application of the hydrogel for oral insulin administration for diabetes.
Collapse
Affiliation(s)
- Yuwei Hu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jia Liu
- State Key Laboratory Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Yujie Ke
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Binju Wang
- State Key Laboratory Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Jason Y C Lim
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Zhaogang Dong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Yi Long
- Electronic Engineering Department, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Itamar Willner
- Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
14
|
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
|
15
|
Shi S, Kan A, Lu L, Zhao W, Jiang W. An acid-responsive DNA hydrogel-mediated cascaded enzymatic nucleic acid amplification system for the sensitive imaging of alkaline phosphatase in living cells. Analyst 2024; 149:3026-3033. [PMID: 38618891 DOI: 10.1039/d4an00258j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Alkaline phosphatase (ALP) is a class of hydrolase that catalyzes the dephosphorylation of phosphorylated species in biological tissues, playing an important role in many physiological and pathological processes. Sensitive imaging of ALP activity in living cells is contributory to the research on these processes. Herein, we propose an acid-responsive DNA hydrogel to deliver a cascaded enzymatic nucleic acid amplification system into cells for the sensitive imaging of intracellular ALP activity. The DNA hydrogel is formed by two kinds of Y-shaped DNA monomers and acid-responsive cytosine-rich linkers. The amplification system contained Bst DNA polymerase (Bst DP), Nt.BbvCI endonuclease, a Recognition Probe (RP, containing a DNAzyme sequence, a Nt.BbvCI recognition sequence, and a phosphate group at the 3'-end), and a Signal Probe (SP, containing a cleavage site for DNAzyme, Cy3 and BHQ2 at the two ends). The amplification system was trapped into the DNA hydrogel and taken up by cells, and the cytosine-rich linkers folded into a quadruplex i-motif in the acidic lysosomes, leading to the collapse of the hydrogel and releasing the amplification system. The phosphate groups on RPs were recognized and removed by the target ALP, triggering a polymerization-nicking cycle to produce large numbers of DNAzyme sequences, which then cleaved multiple SPs, restoring Cy3 fluorescence to indicate the ALP activity. This strategy achieved sensitive imaging of ALP in living HeLa, MCF-7, and NCM460 cells, and realized the sensitive detection of ALP in vitro with a detection limit of 2.0 × 10-5 U mL-1, providing a potential tool for the research of ALP-related physiological and pathological processes.
Collapse
Affiliation(s)
- Shaochuan Shi
- School of Chemistry and Chemical Engineering, Shandong University, 250100 Jinan, P. R. China.
| | - Ailing Kan
- School of Chemistry and Chemical Engineering, Shandong University, 250100 Jinan, P. R. China.
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, P. R. China.
| | - Lu Lu
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, P. R. China.
| | - Weichong Zhao
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, P. R. China.
| | - Wei Jiang
- School of Chemistry and Chemical Engineering, Shandong University, 250100 Jinan, P. R. China.
| |
Collapse
|
16
|
Lee SR, Ong CYJ, Wong JY, Ke Y, Lim JYC, Dong Z, Long Y, Hu Y. Programming the Assembly of Oligo-Adenine with Coralyne into a pH-Responsive DNA Hydrogel. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38489480 DOI: 10.1021/acsami.4c01678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
External stimuli-responsive DNA hydrogels present interesting platforms for drug loading and triggered release. Typically, drug molecules are encapsulated within three-dimensionally hybridized DNA networks. However, the utilization of drug molecules as cofactors to facilitate the directed assembly of DNA strands into hydrogel frameworks and their subsequent controlled release remains to be explored. Herein, we introduce the guided assembly of oligo-adenine (A-strand) into an acidic pH-responsive DNA hydrogel using an anticancer drug, coralyne (COR), as a low-molecular-weight cofactor. At pH 7, COR orchestrates the assembly of A-strand into an antiparallel duplex configuration cross-linked by A-COR-A units at a stoichiometric ratio of one COR cofactor per four adenine bases, resulting in a DNA hydrogel characterized by A-COR-A duplex bridges. At pH 4-5, the instability of A-COR-A units results in the disintegration of the duplex into its constituent components, leading to the release of COR and simultaneous dissociation of the DNA hydrogel matrix. This study introduces a method by which drug molecules, exemplified here by COR, facilitate the direct formation of a supramolecular cofactor-DNA complex, subsequently leading to the creation of a stimuli-responsive DNA hydrogel. This approach may inspire future investigations into DNA hydrogels tailored for controlled drug encapsulation and release applications.
Collapse
Affiliation(s)
- Shu Rui Lee
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Division of Chemistry and Biological Chemistry, School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Republic of Singapore
| | - Clemen Yu Jie Ong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Republic of Singapore
| | - Jing Yi Wong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117543, Republic of Singapore
| | - Yujie Ke
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jason Y C Lim
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Zhaogang Dong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Yi Long
- Electronic Engineering Department, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Yuwei Hu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| |
Collapse
|
17
|
Singh D. Macromolecular Polymer Based Complexes: A Diverse Strategy for the Delivery of Nucleotides. Protein Pept Lett 2024; 31:586-601. [PMID: 39177133 DOI: 10.2174/0109298665310091240809103048] [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: 03/11/2024] [Revised: 06/06/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
This review explores the burgeoning field of macromolecular polymer-based complexes, highlighting their revolutionary potential for the delivery of nucleotides for therapeutic applications. These complexes, ingeniously crafted from a variety of polymers, offer a unique solution to the challenges of nucleotide delivery, including protection from degradation, targeted delivery, and controlled release. The focus of this report is primarily on the design principles, encapsulation strategies, and biological interactions of these complexes, with an emphasis on their biocompatibility, biodegradability, and ability to form diverse structures, such as nanoparticles and micelles. Significant attention is paid to the latest advancements in polymer science that enable the precise tailoring of these complexes for specific nucleotides, such as DNA, RNA, and siRNA. The review discusses the critical role of surface modifications and the incorporation of targeting ligands in enhancing cellular uptake and ensuring delivery to specific tissues or cells, thereby reducing off-target effects and improving therapeutic efficacy. Clinical applications of these polymer-based delivery systems are thoroughly examined with a focus on their use in treating genetic disorders, cancer, and infectious diseases. The review also addresses the challenges and limitations currently faced in this field, such as scalability, manufacturing complexities, and regulatory hurdles. Overall, this review provides a comprehensive overview of the current state and future prospects of macromolecular polymer-based complexes in nucleotide delivery. It underscores the significance of these systems in advancing the field of targeted therapeutics and their potential to reshape the landscape of medical treatment for a wide range of diseases.
Collapse
Affiliation(s)
- Dilpreet Singh
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali, 140413, India
| |
Collapse
|
18
|
Yang X, Xiong M, Fu X, Sun X. Bioactive materials for in vivo sweat gland regeneration. Bioact Mater 2024; 31:247-271. [PMID: 37637080 PMCID: PMC10457517 DOI: 10.1016/j.bioactmat.2023.07.025] [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: 04/20/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 08/29/2023] Open
Abstract
Loss of sweat glands (SwGs) commonly associated with extensive skin defects is a leading cause of hyperthermia and heat stroke. In vivo tissue engineering possesses the potential to take use of the body natural ability to regenerate SwGs, making it more conducive to clinical translation. Despite recent advances in regenerative medicine, reconstructing SwG tissue with the same structure and function as native tissue remains challenging. Elucidating the SwG generation mechanism and developing biomaterials for in vivo tissue engineering is essential for understanding and developing in vivo SwG regenerative strategies. Here, we outline the cell biology associated with functional wound healing and the characteristics of bioactive materials. We critically summarize the recent progress in bioactive material-based cell modulation approaches for in vivo SwG regeneration, including the recruitment of endogenous cells to the skin lesion for SwG regeneration and in vivo cellular reprogramming for SwG regeneration. We discussed the re-establishment of microenvironment via bioactive material-mediated regulators. Besides, we offer promising perspectives for directing in situ SwG regeneration via bioactive material-based cell-free strategy, which is a simple and effective approach to regenerate SwG tissue with both fidelity of structure and function. Finally, we discuss the opportunities and challenges of in vivo SwG regeneration in detail. The molecular mechanisms and cell fate modulation of in vivo SwG regeneration will provide further insights into the regeneration of patient-specific SwGs and the development of potential intervention strategies for gland-derived diseases.
Collapse
Affiliation(s)
- Xinling Yang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
19
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Zhang P, Wang N, Ren D, Jing Z, Sekhar KPC, Hao J, Cui J. Cascade Reaction of Thiol-Disulfide Exchange Potentiates Rapid Fabrication of Polymer Hydrogels. ACS Macro Lett 2023; 12:1437-1442. [PMID: 37819638 DOI: 10.1021/acsmacrolett.3c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
We report a rapid cross-linking strategy for the fabrication of polymer hydrogels based on a thiol-disulfide cascade reaction. Specifically, thiolated polymers (e.g., poly(ethylene glycol), hyaluronic acid, sodium alginate, poly(acrylic acid), and poly(methylacrylic acid)) can be cross-linked via the trigger of Ellman's reagent, resulting in the rapid formation of hydrogels over 20-fold faster than that via the oxidation in air. The gelation kinetics of hydrogels can be tuned by varying the polymer concentration and the molar ratio of Ellman's reagent and free thiols. The obtained hydrogels can be further functionalized with functional moieties (e.g., targeting ligands) for the selective adhesion of cells. This approach is applicable to various natural and synthetic polymers for the assembly of hydrogels with a minimized gelation time, which is promising for various biological applications.
Collapse
Affiliation(s)
- Peiyu Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Dandan Ren
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Zongyu Jing
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Kanaparedu P C Sekhar
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
21
|
Hu Y, Gao S, Lu H, Tan S, Chen F, Ke Y, Ying JY. A Self-Immolative DNA Nanogel Vaccine toward Cancer Immunotherapy. NANO LETTERS 2023; 23:9778-9787. [PMID: 37877690 DOI: 10.1021/acs.nanolett.3c02449] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The development of precisely engineered vehicles for intracellular delivery and the controlled release of payloads remains a challenge. DNA-based nanomaterials offer a promising solution based on the A-T-G-C alphabet-dictated predictable assembly and high programmability. Herein, we present a self-immolative DNA nanogel vaccine, which can be tracelessly released in the intracellular compartments and activate the immune response. Three building blocks with cytosine-rich overhang domains are designed to self-assemble into a DNA nanogel framework with a controlled size. Two oligo agonists and one antigen peptide are conjugated to the building blocks via an acid-labile chemical linker. Upon internalization into acidic endosomes, the formation of i-motif configurations leads to dissociation of the DNA nanogel vaccine. The acid-labile chemical linker is cleaved, releasing the agonists and antigen in their traceless original form to activate antigen-presenting cells and an immune response. This study presents a novel strategy for constructing delivery platforms for intracellularly stimuli-triggered traceless release of therapeutics.
Collapse
Affiliation(s)
- Yuwei Hu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Shujun Gao
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Hongfang Lu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Susi Tan
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Feng Chen
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
| | - Yujie Ke
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jackie Y Ying
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- A*STAR Infectious Diseases Laboratories, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Republic of Singapore
- Bioengineering Department, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
22
|
Huang P, Deng H, Wang C, Zhou Y, Chen X. Cellular Trafficking of Nanotechnology-Mediated mRNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2307822. [PMID: 37929780 DOI: 10.1002/adma.202307822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA)-based therapy has emerged as a powerful, safe, and rapidly scalable therapeutic approach that involves technologies for both mRNA itself and the delivery vehicle. Although there are some unique challenges for different applications of mRNA therapy, a common challenge for all mRNA therapeutics is the transport of mRNA into the target cell cytoplasm for sufficient protein expression. This review is focused on the behaviors at the cellular level of nanotechnology-mediated mRNA delivery systems, which have not been comprehensively reviewed yet. First, the four main therapeutic applications of mRNA are introduced, including immunotherapy, protein replacement therapy, genome editing, and cellular reprogramming. Second, common types of mRNA cargos and mRNA delivery systems are summarized. Third, strategies to enhance mRNA delivery efficiency during the cellular trafficking process are highlighted, including accumulation to the cell, internalization into the cell, endosomal escape, release of mRNA from the nanocarrier, and translation of mRNA into protein. Finally, the challenges and opportunities for the development of nanotechnology-mediated mRNA delivery systems are presented. This review can provide new insights into the future fabrication of mRNA nanocarriers with desirable cellular trafficking performance.
Collapse
Affiliation(s)
- Pei Huang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongzhang Deng
- School of Life Science and Technology and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Changrong Wang
- School of Life Science and Technology and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Yongfeng Zhou
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| |
Collapse
|
23
|
Bai C, Wang C, Lu Y. Novel Vectors and Administrations for mRNA Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303713. [PMID: 37475520 DOI: 10.1002/smll.202303713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/28/2023] [Indexed: 07/22/2023]
Abstract
mRNA therapy has shown great potential in infectious disease vaccines, cancer immunotherapy, protein replacement therapy, gene editing, and other fields due to its central role in all life processes. However, mRNA is challenging to pass through the cell membrane due to its significant negative charges and degradation from RNase, so the key to mRNA therapy is efficient packaging and delivery of it with appropriate vectors. Presently researchers have developed various vectors such as viruses and liposomes, but these conventional vectors are now difficult to meet the growing requirement like safety, efficiency, and targeting, so many novel delivery vectors with unique advantages have emerged recently. This review mainly introduces two categories of novel vectors: biomacromolecules and inorganic nanoparticles, as well as two novel methods of control and administration based on these novel vectors: controlled-release administration and non-invasive administration. These novel delivery strategies have the advantages of high safety, biocompatibility, versatility, intelligence, and targeting. This paper analyzes the challenges faced by the field of mRNA delivery in depth, and discusses how to use the characteristics of novel vectors and administrations to solve these problems.
Collapse
Affiliation(s)
- Chenghai Bai
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Chen Wang
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yuan Lu
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
24
|
Yuan M, Han Z, Liang Y, Sun Y, He B, Chen W, Li F. mRNA nanodelivery systems: targeting strategies and administration routes. Biomater Res 2023; 27:90. [PMID: 37740246 PMCID: PMC10517595 DOI: 10.1186/s40824-023-00425-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/26/2023] [Indexed: 09/24/2023] Open
Abstract
With the great success of coronavirus disease (COVID-19) messenger ribonucleic acid (mRNA) vaccines, mRNA therapeutics have gained significant momentum for the prevention and treatment of various refractory diseases. To function efficiently in vivo and overcome clinical limitations, mRNA demands safe and stable vectors and a reasonable administration route, bypassing multiple biological barriers and achieving organ-specific targeted delivery of mRNA. Nanoparticle (NP)-based delivery systems representing leading vector approaches ensure the successful intracellular delivery of mRNA to the target organ. In this review, chemical modifications of mRNA and various types of advanced mRNA NPs, including lipid NPs and polymers are summarized. The importance of passive targeting, especially endogenous targeting, and active targeting in mRNA nano-delivery is emphasized, and different cellular endocytic mechanisms are discussed. Most importantly, based on the above content and the physiological structure characteristics of various organs in vivo, the design strategies of mRNA NPs targeting different organs and cells are classified and discussed. Furthermore, the influence of administration routes on targeting design is highlighted. Finally, an outlook on the remaining challenges and future development toward mRNA targeted therapies and precision medicine is provided.
Collapse
Affiliation(s)
- Mujie Yuan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zeyu Han
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fan Li
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
25
|
Sun B, Wu W, Narasipura EA, Ma Y, Yu C, Fenton OS, Song H. Engineering nanoparticle toolkits for mRNA delivery. Adv Drug Deliv Rev 2023; 200:115042. [PMID: 37536506 DOI: 10.1016/j.addr.2023.115042] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
The concept of using mRNA to produce its own medicine in situ in the body makes it an ideal drug candidate, holding great potential to revolutionize the way we approach medicine. The unique characteristics of mRNA, as well as its customizable biomedical functions, call for the rational design of delivery systems to protect and transport mRNA molecules. In this review, a nanoparticle toolkit is presented for the development of mRNA-based therapeutics from a drug delivery perspective. Nano-delivery systems derived from either natural systems or chemical synthesis, in the nature of organic or inorganic materials, are summarised. Delivery strategies in controlling the tissue targeting and mRNA release, as well as the role of nanoparticles in building and boosting the activity of mRNA drugs, have also been introduced. In the end, our insights into the clinical and translational development of mRNA nano-drugs are presented.
Collapse
Affiliation(s)
- Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
26
|
Lv Z, Huang M, Yang J, Li P, Chang L, Tang Q, Chen X, Wang S, Yao C, Liu P, Yang D. A Smart DNA-Based Nanosystem Containing Ribosome-Regulating siRNA for Enhanced mRNA Transfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300823. [PMID: 37461803 DOI: 10.1002/adma.202300823] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
Messenger RNA (mRNA) transfection is the prerequisite for the application of mRNA-based therapeutics. In hard-to-transfect cells, such as macrophages, the effective transfection of mRNA remains a long-standing challenge. Herein, a smart DNA-based nanosystem is reported containing ribosome biogenesis-promoting siRNA, realizing efficient mRNA transfection in macrophages. Four monomers are copolymerized to form a nanoframework (NF), including N-isopropylacrylamide (NIPAM) as the skeleton and acrydite-DNA as the initiator to trigger the cascade assembly of DNA hairpins (H1-polyT and H2-siRNA). By virtue of the phase transition characteristic of polymeric NIPAM, below the lower critical solution temperature (LCST, ≈34 °C), the NF swells to expose polyT sequences to hybridize with the polyA tail of mRNA. Above the LCST, the NF deswells to encapsulate mRNA. The disulfide bond in the NF responds to glutathione, triggering the disassembly of the nanosystem; the siRNA and mRNA are released in response to triphosadenine and RNase H. The siRNA down-regulates the expression of heat shock protein 27, which up-regulates the expression of phosphorylated ribosomal protein S6. The nanosystem shows satisfactory mRNA transfection and translation efficiency in a mouse model. It is envisioned that the DNA-based nanosystem will provide a promising carrier to deliver mRNA in hard-to-transfect cells and promote the development of mRNA-based therapeutics.
Collapse
Affiliation(s)
- Zhaoyue Lv
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Mengxue Huang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Jing Yang
- Beijing Institute of Microbiology and Epidemiology, Beijing, 100850, P. R. China
| | - Peiran Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Lele Chang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Qianyun Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Xiaojing Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Shengqi Wang
- Beijing Institute of Microbiology and Epidemiology, Beijing, 100850, P. R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Peifeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| |
Collapse
|
27
|
Chen Z, Wang X, Zhao N, Chen H, Guo G. Advancements in pH-responsive nanocarriers: enhancing drug delivery for tumor therapy. Expert Opin Drug Deliv 2023; 20:1623-1642. [PMID: 38059646 DOI: 10.1080/17425247.2023.2292678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Tumors pose a significant global economic and health burden, with conventional cancer treatments lacking tumor specificity, leading to limited efficiency and undesirable side effects. Targeted tumor therapy is imminent. Tumor cells produce lactate and hydrogen ions (H+) by Warburg effect, forming an acidic tumor microenvironment (TME), which can be employed to design targeted tumor therapy. Recently, progress in nanotechnology has led to the development of pH-responsive nanocarriers, which have gathered significant attention. Under acidic tumor conditions, they exhibit targeted accumulation within tumor sites and controlled release profiles of therapeutic reagents, enabling precise tumor therapy. AREAS COVERED This review comprehensively summarize the principles underlying pH-responsive features, discussing various types of pH-responsive nanocarriers, their advantages, and limitations. Innovative therapeutic drugs are also examined, followed by an exploration of recent advancements in applying various pH-responsive nanocarriers as delivery systems for enhanced tumor therapy. EXPERT OPINIONS pH-responsive nanocarriers have garnered significant attention for their capability to achieve targeted accumulation of therapeutic agents at tumor sites and controlled drug delivery profiles, ultimately increasing the efficiency of tumor eradication. It is anticipated that the employment of pH-responsive nanocarriers will elevate the effectiveness and safety of tumor therapy, contributing to improved overall outcomes.
Collapse
Affiliation(s)
- Zhouyun Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxiao Wang
- West China School of Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Zhao
- School of Pharmacy, Shihezi University, Shihezi, China
| | - Haifeng Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Guo
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Zare I, Taheri-Ledari R, Esmailzadeh F, Salehi MM, Mohammadi A, Maleki A, Mostafavi E. DNA hydrogels and nanogels for diagnostics, therapeutics, and theragnostics of various cancers. NANOSCALE 2023. [PMID: 37337663 DOI: 10.1039/d3nr00425b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
As an efficient class of hydrogel-based therapeutic drug delivery systems, deoxyribonucleic acid (DNA) hydrogels (particularly DNA nanogels) have attracted massive attention in the last five years. The main contributor to this is the programmability of these 3-dimensional (3D) scaffolds that creates fundamental effects, especially in treating cancer diseases. Like other active biological ingredients (ABIs), DNA hydrogels can be functionalized with other active agents that play a role in targeting drug delivery and modifying the half-life of the therapeutic cargoes in the body's internal environment. Considering the brilliant advantages of DNA hydrogels, in this survey, we intend to submit an informative collection of feasible methods for the design and preparation of DNA hydrogels and nanogels, and the responsivity of the immune system to these therapeutic cargoes. Moreover, the interactions of DNA hydrogels with cancer biomarkers are discussed in this account. Theragnostic DNA nanogels as an advanced species for both detection and therapeutic purposes are also briefly reviewed.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | - Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Farhad Esmailzadeh
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Mohammad Mehdi Salehi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Adibeh Mohammadi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Duskunovic N, Im SH, Lee J, Chung HJ. Effective mRNA Delivery by Condensation with Cationic Nanogels Incorporated into Liposomes. Mol Pharm 2023; 20:3088-3099. [PMID: 37184833 DOI: 10.1021/acs.molpharmaceut.3c00089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The challenge in effective delivery of mRNA has been a major hurdle in their development as therapeutics. Herein, we present that the incorporation of cationic nanogels as the condensing material for mRNA into liposomes enables stable and enhanced mRNA delivery to cells in vitro. We prepared dextran-based nanogel particles, which were surface functionalized with oligoarginine peptide (DNPR9) and complexed with mRNA for incorporation into liposomes (LipoDNPR9). The use of DNPR9 with the liposomes resulted in enhanced internalization, as well as a 4-fold increase in transfection of luciferase mRNA when treated with A549 cells in vitro, compared to control liposomes. The enhancement in transfection efficiency was also observed in various cell lines while causing low cytotoxicity. The versatility of the strategy was also investigated by applying DNPR9 for mRNA condensation to ionizable lipid particles, which resulted in an ∼55% increase in transfection. The current development based on nanogel-incorporated liposomes introduces an effective platform for mRNA delivery, while the condensation strategy using DNPR9 can be widely applied for various lipid-based formulations to enhance their efficacy.
Collapse
Affiliation(s)
- Nevena Duskunovic
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - San Hae Im
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Juhee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
30
|
Kurniawan D, Mathew J, Rahardja MR, Pham HP, Wong PC, Rao NV, Ostrikov KK, Chiang WH. Plasma-Enabled Graphene Quantum Dot Hydrogels as Smart Anticancer Drug Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206813. [PMID: 36732883 DOI: 10.1002/smll.202206813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/10/2023] [Indexed: 05/18/2023]
Abstract
One of the major challenges on the way to low-cost, simple, and effective cancer treatments is the lack of smart anticancer drug delivery materials with the requisite of site-specific and microenvironment-responsive properties. This work reports the development of plasma-engineered smart drug nanocarriers (SDNCs) containing chitosan and nitrogen-doped graphene quantum dots (NGQDs) for drug delivery in a pH-responsive manner. Through a customized microplasma processing, a highly cross-linked SDNC with only 4.5% of NGQD ratio can exhibit enhanced toughness up to threefold higher than the control chitosan group, avoiding the commonly used high temperatures and toxic chemical cross-linking agents. The SDNCs demonstrate improved loading capability for doxorubicin (DOX) via π-π interactions and stable solid-state photoluminescence to monitor the DOX loading and release through the Förster resonance energy transfer (FRET) mechanism. Moreover, the DOX loaded SDNC exhibits anticancer effects against cancer cells during cytotoxicity tests at minimum concentration. Cellular uptake studies confirm that the DOX loaded SDNC can be successfully internalized into the nucleus after 12 h incubation period. This work provides new insights into the development of smart, environmental-friendly, and biocompatible nanographene hydrogels for the next-generation biomedical applications.
Collapse
Affiliation(s)
- Darwin Kurniawan
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Jacob Mathew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Michael Ryan Rahardja
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Hoang-Phuc Pham
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Pei-Chun Wong
- Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei Medical University, Taipei, 110, Taiwan
| | - Neralla Vijayakameswara Rao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and QUT Centre for Materials Science, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| |
Collapse
|
31
|
Li X, Guo X, Hu M, Cai R, Chen C. Optimal delivery strategies for nanoparticle-mediated mRNA delivery. J Mater Chem B 2023; 11:2063-2077. [PMID: 36794598 DOI: 10.1039/d2tb02455a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Messenger RNA (mRNA) has emerged as a new and efficient agent for the treatment of various diseases. The success of lipid nanoparticle-mRNA against the novel coronavirus (SARS-CoV-2) pneumonia epidemic has proved the clinical potential of nanoparticle-mRNA formulations. However, the deficiency in the effective biological distribution, high transfection efficiency and good biosafety are still the major challenges in clinical translation of nanomedicine for mRNA delivery. To date, a variety of promising nanoparticles have been constructed and then gradually optimized to facilitate the effective biodistribution of carriers and efficient mRNA delivery. In this review, we describe the design of nanoparticles with an emphasis on lipid nanoparticles, and discuss the manipulation strategies for nanoparticle-biology (nano-bio) interactions for mRNA delivery to overcome the biological barriers and improve the delivery efficiency, because the specific nano-bio interaction of nanoparticles usually remoulds the biomedical and physiological properties of the nanoparticles especially the biodistribution, mechanism of cellular internalization and immune response. Finally, we give a perspective for the future applications of this promising technology. We believe that the regulation of nano-bio interactions would be a significant breakthrough to improve the mRNA delivery efficiency and cross biological barriers. This review may provide a new direction for the design of nanoparticle-mediated mRNA delivery systems.
Collapse
Affiliation(s)
- Xiaoyan Li
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China.,CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Xiaocui Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China.,The GBA National Institute for Nanotechnology Innovation, Guangzhou 510700, China
| |
Collapse
|
32
|
Zhao M, Wang R, Yang K, Jiang Y, Peng Y, Li Y, Zhang Z, Ding J, Shi S. Nucleic acid nanoassembly-enhanced RNA therapeutics and diagnosis. Acta Pharm Sin B 2023; 13:916-941. [PMID: 36970219 PMCID: PMC10031267 DOI: 10.1016/j.apsb.2022.10.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
RNAs are involved in the crucial processes of disease progression and have emerged as powerful therapeutic targets and diagnostic biomarkers. However, efficient delivery of therapeutic RNA to the targeted location and precise detection of RNA markers remains challenging. Recently, more and more attention has been paid to applying nucleic acid nanoassemblies in diagnosing and treating. Due to the flexibility and deformability of nucleic acids, the nanoassemblies could be fabricated with different shapes and structures. With hybridization, nucleic acid nanoassemblies, including DNA and RNA nanostructures, can be applied to enhance RNA therapeutics and diagnosis. This review briefly introduces the construction and properties of different nucleic acid nanoassemblies and their applications for RNA therapy and diagnosis and makes further prospects for their development.
Collapse
Affiliation(s)
- Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kunmeng Yang
- The First Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130061, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yachen Peng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
33
|
Wang H, Wang X, Lai K, Yan J. Stimulus-Responsive DNA Hydrogel Biosensors for Food Safety Detection. BIOSENSORS 2023; 13:320. [PMID: 36979532 PMCID: PMC10046603 DOI: 10.3390/bios13030320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
Food safety has always been a major global challenge to human health and the effective detection of harmful substances in food can reduce the risk to human health. However, the food industry has been plagued by a lack of effective and sensitive safety monitoring methods due to the tension between the cost and effectiveness of monitoring. DNA-based hydrogels combine the advantages of biocompatibility, programmability, the molecular recognition of DNA molecules, and the hydrophilicity of hydrogels, making them a hotspot in the research field of new nanomaterials. The stimulus response property greatly broadens the function and application range of DNA hydrogel. In recent years, DNA hydrogels based on stimulus-responsive mechanisms have been widely applied in the field of biosensing for the detection of a variety of target substances, including various food contaminants. In this review, we describe the recent advances in the preparation of stimuli-responsive DNA hydrogels, highlighting the progress of its application in food safety detection. Finally, we also discuss the challenges and future application of stimulus-responsive DNA hydrogels.
Collapse
|
34
|
Wang D, Duan J, Liu J, Yi H, Zhang Z, Song H, Li Y, Zhang K. Stimuli-Responsive Self-Degradable DNA Hydrogels: Design, Synthesis, and Applications. Adv Healthc Mater 2023:e2203031. [PMID: 36708144 DOI: 10.1002/adhm.202203031] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/11/2023] [Indexed: 01/29/2023]
Abstract
DNA hydrogels play an increasingly important role in biomedicine and bioanalysis applications. Due to their high programmability, multifunctionality and biocompatibility, they are often used as effective carriers for packing drugs, cells, or other bioactive cargoes in vitro and in vivo. However, the stability of the DNA hydrogels prevents their in-demand rapid release of cargoes to achieve a full therapeutic effect in time. For bioanalysis, the generation of signals sometimes needs the DNA hydrogel to be rapidly degraded when sensing target molecules. To meet these requirements, stimulus-responsive DNA hydrogels are designed. By responding to different stimuli, self-degradable DNA hydrogels can switch from gel to solution for quantitative bioanalysis and precision cargo delivery. This review summarizes the recently developed innovative methods for designing stimuli-responsive self-degradable DNA hydrogels and showed their applications in the bioanalysis and biomedicines fields. Challenges, as well as prospects, are also discussed.
Collapse
Affiliation(s)
- Danyu Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Jie Duan
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingwen Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Hua Yi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Yinchao Li
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
35
|
Matrices Activated with Messenger RNA. J Funct Biomater 2023; 14:jfb14010048. [PMID: 36662095 PMCID: PMC9864744 DOI: 10.3390/jfb14010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Over two decades of preclinical and clinical experience have confirmed that gene therapy-activated matrices are potent tools for sustained gene modulation at the implantation area. Matrices activated with messenger RNA (mRNA) are the latest development in the area, and they promise an ideal combination of efficiency and safety. Indeed, implanted mRNA-activated matrices allow a sustained delivery of mRNA and the continuous production of therapeutic proteins in situ. In addition, they are particularly interesting to generate proteins acting on intracellular targets, as the translated protein can directly exert its therapeutic function. Still, mRNA-activated matrices are incipient technologies with a limited number of published records, and much is still to be understood before their successful implementation. Indeed, the design parameters of mRNA-activated matrices are crucial for their performance, as they affect mRNA stability, device immunogenicity, translation efficiency, and the duration of the therapy. Critical design factors include matrix composition and its mesh size, mRNA chemical modification and sequence, and the characteristics of the nanocarriers used for mRNA delivery. This review aims to provide some background relevant to these technologies and to summarize both the design space for mRNA-activated matrices and the current knowledge regarding their pharmaceutical performance. Furthermore, we will discuss potential applications of mRNA-activated matrices, mainly focusing on tissue engineering and immunomodulation.
Collapse
|
36
|
Yang C, Wu X, Liu J, Ding B. Stimuli-responsive nucleic acid nanostructures for efficient drug delivery. NANOSCALE 2022; 14:17862-17870. [PMID: 36458678 DOI: 10.1039/d2nr05316k] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Based on complementary base pairing, nucleic acid molecules have acted as engineerable building blocks to prepare versatile nanostructures with unique shapes and sizes. Benefiting from excellent programmability and biocompatibility, rationally designed nucleic acid nanostructures have been widely employed in biomedical applications. With the development of the chemical biology of nucleic acids, various stimuli-responsive nucleic acid nanostructures have been constructed by tailored chemical modification with multifunctional components. In this minireview, we summarize the representative and latest research about the employment of stimuli-responsive nucleic acid nanostructures for drug delivery in response to endogenous and exogenous stimuli (redox gradient, pH, nuclease, biomacromolecule, and light). We also discuss the broad prospects and remaining challenges of nucleic acid nanotechnology in biomedical applications.
Collapse
Affiliation(s)
- Changping Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- School of Materials Science and Engineering, Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaohui Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Materials Science and Engineering, Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
37
|
Wang Z, Chen R, Yang S, Li S, Gao Z. Design and application of stimuli-responsive DNA hydrogels: A review. Mater Today Bio 2022; 16:100430. [PMID: 36157049 PMCID: PMC9493390 DOI: 10.1016/j.mtbio.2022.100430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/25/2022] Open
Abstract
Deoxyribonucleic acid (DNA) hydrogels combine the properties of DNAs and hydrogels, and adding functionalized DNAs is key to the wide application of DNA hydrogels. In stimuli-responsive DNA hydrogels, the DNA transcends its application in genetics and bridges the gap between different fields. Specifically, the DNA acts as both an information carrier and a bridge in constructing DNA hydrogels. The programmability and biocompatibility of DNA hydrogel make it change macroscopically in response to a variety of stimuli. In order to meet the needs of different scenarios, DNA hydrogels were also designed into microcapsules, beads, membranes, microneedle patches, and other forms. In this study, the stimuli were classified into single biological and non-biological stimuli and composite stimuli. Stimuli-responsive DNA hydrogels from the past five years were summarized, including but not limited to their design and application, in particular logic gate pathways and signal amplification mechanisms. Stimuli-responsive DNA hydrogels have been applied to fields such as sensing, nanorobots, information carriers, controlled drug release, and disease treatment. Different potential applications and the developmental pro-spects of stimuli-responsive DNA hydrogels were discussed.
Collapse
Affiliation(s)
- Zhiguang Wang
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Ruipeng Chen
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Shiping Yang
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Shuang Li
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Zhixian Gao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| |
Collapse
|
38
|
Yang T, Xu Q, Chen J, Jia PK, Xie BB, Wang D, Zhou X, Shao Y. Selectively Identifying Exposed-over-Unexposed C-C + Pairs in Human Telomeric i-Motif Structures with Length-Dependent Polymorphism. Anal Chem 2022; 94:14994-15001. [PMID: 36263663 DOI: 10.1021/acs.analchem.2c02875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The i-motif structure (iM) has attracted much attention, because of its in vivo bioactivity and wide in vitro applications such as DNA-based switches. Herein, the length-dependent folding of cytosine-rich repeats of the human telomeric 5'-(CCCTAA)n-1CCC-3' (iM-n, where n = 2-8) was fully explored. We found that iM-4, iM-5, and iM-8 mainly form the intramolecular monomer iM structures, while a tetramolecular structure populates only for iM-3. However, iM-6 and iM-7 have the potential to fold as well into the dimeric iM structures besides the monomer ones. The natural hypericin (Hyp) was used as the polymorphism-selective probe to recognize the iM structures. Interestingly, only iM-3, iM-6, and iM-7 can efficiently switch on the Hyp fluorescence by specifically binding with the outmost C-C+ base pairs that are exposed directly to solution. However, other iM structures that fold in a way with a coverage of the outmost C-C+ pairs by loop sequences are totally unavailable for the Hyp binding. Theoretical modeling indicates that adaptive π-π and cation-π interactions contribute to the Hyp recognition toward the exposed C-C+ pairs. This specific iM recognition can be boosted by a photocatalytic DNAzyme construct. Our work provides a reliable fluorescence method to selectively explore the polymorphism of iM structures.
Collapse
Affiliation(s)
- Tong Yang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Qiuda Xu
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Jiahui Chen
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Pei-Ke Jia
- Hangzhou Institute of Advanced Studies, Zhejiang Normal University, 1108 Gengwen Road, Hangzhou 311231, Zhejiang, People's Republic of China
| | - Bin-Bin Xie
- Hangzhou Institute of Advanced Studies, Zhejiang Normal University, 1108 Gengwen Road, Hangzhou 311231, Zhejiang, People's Republic of China
| | - Dandan Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Xiaoshun Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| | - Yong Shao
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People's Republic of China
| |
Collapse
|
39
|
Tian Y, Deng Z, Yang P. mRNA vaccines: A novel weapon to control infectious diseases. Front Microbiol 2022; 13:1008684. [PMID: 36267192 PMCID: PMC9576954 DOI: 10.3389/fmicb.2022.1008684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 12/01/2022] Open
Abstract
Infectious diseases have always threatened human life, but with the development of vaccines, effective strategies for preventing and controlling these diseases have become available. The global outbreak of COVID-19 ushered in the advent of mRNA vaccine technologies, which quickly led to the introduction of mRNA vaccines effective against SARS-CoV-2. The success of this approach has stimulated research into the use of mRNA vaccines in the fight against other emerging as well as remerging infectious diseases. This review examines the constructive strategies and delivery systems used in mRNA vaccines and provides an overview of current clinical trials of those vaccines in the prevention of infectious diseases. The underlying mechanisms of mRNA vaccines are also discussed, including the double-edged sword of the innate immune response. Finally, the challenges but also the potential of mRNA vaccines are considered.
Collapse
Affiliation(s)
- Yuying Tian
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Inner Mongolia Medical University, Hohhot, China
| | - Zhuoya Deng
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Penghui Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Inner Mongolia Medical University, Hohhot, China
- *Correspondence: Penghui Yang,
| |
Collapse
|
40
|
Gong Y, Chen W, Chen X, He Y, Jiang H, Zhang X, Pan L, Ni B, Yang F, Xu Y, Zhang Q, Zhou L, Cheng Y. An Injectable Epigenetic Autophagic Modulatory Hydrogel for Boosting Umbilical Cord Blood NK Cell Therapy Prevents Postsurgical Relapse of Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201271. [PMID: 35712750 PMCID: PMC9376812 DOI: 10.1002/advs.202201271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Indexed: 06/15/2023]
Abstract
Triple-negative breast cancer (TNBC) exhibits resistance to conventional treatments due to the presence of cancer stem cells (CSCs), causing postsurgical relapse and a dismal prognosis. Umbilical cord blood natural killer (UCB-NK) cell-based immunotherapy represents a promising strategy for cancer treatment. However, its therapeutic efficacy is greatly restrained by downregulation of the NK cell activation ligand MHC class I-related chain A/B (MICA/B) and autophagy-mediated degradation of NK cell-derived granzyme B (GZMB) in CSCs. Herein, it is demonstrated that suberoylanilide hydroxamic acid (SAHA) epigenetically downregulates let-7e-5p and miR-615-3p to increase MICA/B expression and that 3-methyl adenine (3MA) inhibits autophagy-mediated GZMB degradation, thereby sensitizing breast CSCs to UCB-NK cells. Then, an injectable hydrogel is designed to codeliver SAHA and 3MA to enhance UCB-NK cell infusion efficacy in TNBC. The hydrogel precursors can be smoothly injected into the tumor resection bed and form a stable gel in situ, allowing for a pH-sensitive sustained release of SAHA and 3MA. Moreover, UCB-NK cell infusion in combination with the hydrogel efficiently controls postsurgical relapse of TNBC. In addition, the hydrogel exhibits good hemostasis and wound-healing functions. Therefore, the work provides proof of concept that an injectable epigenetic autophagic modulatory hydrogel augments UCB-NK cell therapy to combat postsurgical relapse of TNBC.
Collapse
Affiliation(s)
- Yihang Gong
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Wenjie Chen
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Xiuxing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Medical OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Yizhan He
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Hua Jiang
- Department of Breast & Thyroid SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Xijian Zhang
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Lijie Pan
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Beibei Ni
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Fan Yang
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yan Xu
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Qi Zhang
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and TreatmentDepartment of Spine SurgeryThe Third Affiliated HospitalGuangzhou Medical UniversityGuangzhou510150China
| | - Yusheng Cheng
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
41
|
Xu X, Jiang Y, Lu C. Self-Assembled ATP-Responsive DNA Nanohydrogel for Specifically Activated Fluorescence Imaging and Chemotherapy in Cancer Cells. Anal Chem 2022; 94:10221-10226. [PMID: 35796567 DOI: 10.1021/acs.analchem.2c01760] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor marker-responsive drug delivery systems have been developed for cancer imaging and chemotherapy. However, improving their ability of controlled drug release remains a challenge. In this study, we have developed an adenosine triphosphate (ATP)-responsive DNA nanohydrogel for specifically activated fluorescence imaging and chemotherapy in cancer cells. Acrylamide and acrydite-modified DNAs were polymerized to obtain DNA-grafted polyacrylamide copolymers. Then, the copolymers acted as the backbone of the nanohydrogel and were assembled by base complementation with ATP aptamer linkers to construct an ATP-responsive nanohydrogel. Meanwhile, the chemotherapeutic drug doxorubicin (DOX) was added and loaded into the ATP-responsive nanohydrogel during the assembly process. After endocytosis by cancer cells and response to a high intracellular ATP level, the DOX-loaded nanohydrogel disassembled due to the formation of aptamer/ATP complexes. Subsequently, the released DOX played a role in fluorescence imaging and chemotherapy of cancer cells. Through the ATP-responsive property and satisfying drug delivery capability, this nanohydrogel realized fluorescence imaging and specific cancer cell killing capabilities due to different intracellular ATP levels in normal and cancer cell lines. In summary, this study has provided a novel strategy of constructing a tumor microenvironment-responsive drug delivery system triggered by the tumor markers for tumor intracellular imaging and chemotherapy.
Collapse
Affiliation(s)
- Xin Xu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Yifan Jiang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
42
|
Deng Z, Tian Y, Song J, An G, Yang P. mRNA Vaccines: The Dawn of a New Era of Cancer Immunotherapy. Front Immunol 2022; 13:887125. [PMID: 35720301 PMCID: PMC9201022 DOI: 10.3389/fimmu.2022.887125] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
mRNA therapy is a novel anticancer strategy based on in vitro transcription (IVT), which has potential for the treatment of malignant tumors. The outbreak of the COVID-19 pandemic in the early 21st century has promoted the application of mRNA technologies in SARS-CoV-2 vaccines, and there has been a great deal of interest in the research and development of mRNA cancer vaccines. There has been progress in a number of key technologies, including mRNA production strategies, delivery systems, antitumor immune strategies, etc. These technologies have accelerated the progress and clinical applications of mRNA therapy, overcoming problems encountered in the past, such as instability, inefficient delivery, and weak immunogenicity of mRNA vaccines. This review provides a detailed overview of the production, delivery systems, immunological mechanisms, and antitumor immune response strategies for mRNA cancer vaccines. We list some mRNA cancer vaccines that are candidates for cancer treatment and discuss clinical trials in the field of tumor immunotherapy. In addition, we discuss the immunological mechanism of action by which mRNA vaccines destroy tumors as well as challenges and prospects for the future.
Collapse
Affiliation(s)
- Zhuoya Deng
- Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yuying Tian
- Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Guangwen An
- Department of Pharmacy, No. 984 Hospital of the PLA, Beijing, China
| | - Penghui Yang
- Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
43
|
Zhang H, Zhang M, Zhang X, Gao Y, Ma Y, Chen H, Wan J, Li C, Wang F, Sun X. Enhanced postoperative cancer therapy by iron-based hydrogels. Biomater Res 2022; 26:19. [PMID: 35606838 PMCID: PMC9125885 DOI: 10.1186/s40824-022-00268-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022] Open
Abstract
AbstractSurgical resection is a widely used method for the treatment of solid tumor cancers. However, the inhibition of tumor recurrence and metastasis are the main challenges of postoperative tumor therapy. Traditional intravenous or oral administration have poor chemotherapeutics bioavailability and undesirable systemic toxicity. Polymeric hydrogels with a three-dimensional network structure enable on-site delivery and controlled release of therapeutic drugs with reduced systemic toxicity and have been widely developed for postoperative adjuvant tumor therapy. Among them, because of the simple synthesis, good biocompatibility, biodegradability, injectability, and multifunctionality, iron-based hydrogels have received extensive attention. This review has summarized the general synthesis methods and construction principles of iron-based hydrogels, highlighted the latest progress of iron-based hydrogels in postoperative tumor therapy, including chemotherapy, photothermal therapy, photodynamic therapy, chemo-dynamic therapy, and magnetothermal-chemical combined therapy, etc. In addition, the challenges towards clinical application of iron-based hydrogels have also been discussed. This review is expected to show researchers broad perspectives of novel postoperative tumor therapy strategy and provide new ideas in the design and application of novel iron-based hydrogels to advance this sub field in cancer nanomedicine.
Collapse
|
44
|
Liu B, Sun L, Lu X, Yang Y, Peng H, Sun Z, Xu J, Chu H. Real-time drug release monitoring from pH-responsive CuS-encapsulated metal-organic frameworks. RSC Adv 2022; 12:11119-11127. [PMID: 35425048 PMCID: PMC8992360 DOI: 10.1039/d1ra09320g] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/04/2022] [Indexed: 11/21/2022] Open
Abstract
Real-time monitoring of drug release behaviors over extended periods of time is critical in understanding the dynamics of drug progression for personalized chemotherapeutic treatment. In this work, we report a metal–organic framework (MOF)-based nanotheranostic system encapsulated with photothermal agents (CuS) and therapeutic drug (DOX) to achieve the capabilities of real-time drug release monitoring and combined chemo-photothermal therapy. Meanwhile, folic acid-conjugated polyethylene glycol (FA-PEG) antennas were connected to the MOF through coordination interactions, endowing the MOF with an enhanced active targeting effect toward cancer cells. It is anticipated that such a theranostic agent, simultaneously possessing tumor-targeting, real-time drug monitoring and effective treatment, will potentially enhance the performance in cancer therapy. A metal–organic framework-based nanotheranostic system was fabricated to achieve the capabilities of tumor-targeting, real-time monitoring of pH-responsive drug release and combined chemo-photothermal therapy.![]()
Collapse
Affiliation(s)
- Bei Liu
- College of Science, Minzu University of China Beijing 100081 China
| | - Lirong Sun
- College of Science, Minzu University of China Beijing 100081 China
| | - Xijian Lu
- College of Science, Minzu University of China Beijing 100081 China
| | - Yuping Yang
- College of Science, Minzu University of China Beijing 100081 China
| | - Hongshang Peng
- College of Science, Minzu University of China Beijing 100081 China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 China
| | - Juan Xu
- National Research Institute for Family Planning Beijing 100005 China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 China
| |
Collapse
|
45
|
Yao S, Xiang L, Wang L, Gong H, Chen F, Cai C. pH-responsive DNA hydrogels with ratiometric fluorescence for accurate detection of miRNA-21. Anal Chim Acta 2022; 1207:339795. [DOI: 10.1016/j.aca.2022.339795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022]
|
46
|
Hu Y, Gao S, Lu H, Ying JY. Acid-Resistant and Physiological pH-Responsive DNA Hydrogel Composed of A-Motif and i-Motif toward Oral Insulin Delivery. J Am Chem Soc 2022; 144:5461-5470. [PMID: 35312303 DOI: 10.1021/jacs.1c13426] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An acid-resistant DNA hydrogel that is stable in an extremely acidic environment with pH as low as 1.2 has not been reported before, largely due to the instability of DNA-hybridized structures. To achieve this, adenine (A)-rich and cytosine (C)-rich oligonucleotides are rationally designed and integrated to form copolymers with acrylamide monomers via free-radical polymerization. In an acidic environment (pH 1.2-6.0), the generated copolymers form a hydrogel state, which is cross-linked by parallel A-motif duplex configurations (pH 1.2-3.0) and quadruplex i-motif structures (pH 4.0-6.0) due to the protonation of A and C bases, respectively. Specifically, the protonated A-rich sequences under pH 1.2-3.0 form a stable parallel A-motif duplex cross-linking unit through reverse Hoogsteen interaction and electrostatic attraction. Hemi-protonated C bases under mildly acidic pH (4.0-6.0) form quadruplex i-motif cross-linking configuration via Hoogsteen interaction. Under physiological pH, both A and C bases deprotonated, resulting in the separation of A-motif and i-motif to A-rich and C-rich single strands, respectively, and thereby the dissociation of the DNA hydrogel into the solution state. The acid-resistant and physiological pH-responsive DNA hydrogel was further developed for oral drug delivery to the hostile acidic environment in the stomach (pH 1.2), duodenum (pH 5.0), and small intestine (pH 7.2), where the drug would be released and absorbed. As a proof of concept, insulin was encapsulated in the DNA hydrogel and orally administered to diabetic rats. In vitro and in vivo studies demonstrated the potential usage of the DNA hydrogel for oral drug delivery.
Collapse
Affiliation(s)
- Yuwei Hu
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Shujun Gao
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Hongfang Lu
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Jackie Y Ying
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore.,NanoBio Lab, A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| |
Collapse
|
47
|
Rajendran AK, Amirthalingam S, Hwang NS. A brief review of mRNA therapeutics and delivery for bone tissue engineering. RSC Adv 2022; 12:8889-8900. [PMID: 35424872 PMCID: PMC8985089 DOI: 10.1039/d2ra00713d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/15/2022] [Indexed: 11/21/2022] Open
Abstract
The therapeutics for bone tissue regeneration requires constant advancements owing to the steady increase in the number of patients suffering from bone-related disorders, and also to find efficient and cost-effective treatment modalities. One of the major advancements in the field of therapeutics is the development of mRNAs. mRNAs, which have been extensively tested for the vaccines, could be very well utilized as a potential inducer for bone regeneration. The ability of mRNAs to enter the cells and instruct the cellular machinery to produce the required native proteins such as BMP or VEGF is a great way to avoid the issues faced with growth factor deliveries such as the production cost, loss of biological function etc. However, there have been a few hurdles for using mRNAs as an effective therapeutic agent, such as proper dosing, tolerating the degradation by RNases, improving the half-life, controlling the spatio-temporal release and reducing the off-target effects. This brief review discusses the various developments in the field of mRNA therapeutics especially for bone tissue engineering, how nano-formulations are being developed to effectively deliver the mRNAs into the cells by evading the immune responses, how researchers have developed certain strategies to increase the half-life, to successfully deliver the mRNAs to specific bone defect area and bring about effective bone regeneration.
Collapse
Affiliation(s)
- Arun Kumar Rajendran
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
- Institute for Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
48
|
Hou X, Zhang Y, Li Y, Chen J, Yu Z, Xu L, Liu H. Frame-guided assembly of DNA nanohydrogels via clamped hybridization chain reactions. POLYMER 2022. [DOI: 10.1016/j.polymer.2022.124659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Aye SL, Sato Y. Therapeutic Applications of Programmable DNA Nanostructures. MICROMACHINES 2022; 13:315. [PMID: 35208439 PMCID: PMC8876680 DOI: 10.3390/mi13020315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022]
Abstract
Deoxyribonucleic acid (DNA) nanotechnology, a frontier in biomedical engineering, is an emerging field that has enabled the engineering of molecular-scale DNA materials with applications in biomedicine such as bioimaging, biodetection, and drug delivery over the past decades. The programmability of DNA nanostructures allows the precise engineering of DNA nanocarriers with controllable shapes, sizes, surface chemistries, and functions to deliver therapeutic and functional payloads to target cells with higher efficiency and enhanced specificity. Programmability and control over design also allow the creation of dynamic devices, such as DNA nanorobots, that can react to external stimuli and execute programmed tasks. This review focuses on the current findings and progress in the field, mainly on the employment of DNA nanostructures such as DNA origami nanorobots, DNA nanotubes, DNA tetrahedra, DNA boxes, and DNA nanoflowers in the biomedical field for therapeutic purposes. We will also discuss the fate of DNA nanostructures in living cells, the major obstacles to overcome, that is, the stability of DNA nanostructures in biomedical applications, and the opportunities for DNA nanostructure-based drug delivery in the future.
Collapse
Affiliation(s)
| | - Yusuke Sato
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan;
| |
Collapse
|
50
|
Abstract
Stimuli-responsive DNA-based hydrogels are attracting growing interest because of their smart responsiveness, excellent biocompatibility, regulated biodegradability, and programmable design properties. Integration of reconfigurable DNA architectures and switchable supramolecular moieties (as cross-linkers) in hydrogels by responding to external stimuli provides an ideal approach for the reversible tuning structural and mechanical properties of the hydrogels, which can be exploited in the development of intelligent DNA-based materials. This review highlights recent advances in the design of responsive pure DNA hydrogels, DNA-polymer hybrid hydrogels, and autonomous DNA-based hydrogels with transient behaviors. A variety of chemically and physically triggered DNA-based stimuli-responsive hydrogels and their versatile applications in biosensing, biocatalysis, cell culture and separation, drug delivery, shape memory, self-healing, and robotic actuators are summarized. Finally, we address the key challenges that the field will face in the coming years, and future prospects are identified.
Collapse
Affiliation(s)
- Chen Wang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Junji Zhang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science & Technology, No. 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|