1
|
Zhang W, Yang X, Qu Z, Ding P, Kong X, Wang X, Liu Q, Zhang X, Lu Y, Wang J, Chen Z, Fang Y. DNA tetrahedral nanoparticles: Co-delivery of siOTUD6B/DOX against triple-negative breast cancer. J Control Release 2025; 377:197-211. [PMID: 39549731 DOI: 10.1016/j.jconrel.2024.11.025] [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: 04/14/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited targeted therapeutic options. Recently, the deubiquitinizing enzyme ovarian tumor domain-containing 6B (OTUD6B) has been reported to play a potential role in TNBC progression. Therefore, this study investigates the role and underlying molecular mechanisms of OTUD6B in vitro and xenograft models of TNBC. Specifically, we examined the therapeutic effects of siOTUD6B and doxorubicin (DOX) co-delivery using synthesized tetrahedral DNA nanoparticles (Tds) on tumor growth and progression. Additionally, the uptake and efficacy of the siOTUD6B/DOX@Td in TNBC cells were evaluated. Notably, the siOTUD6B/DOX@Td nanoparticle demonstrated efficient cellular uptake by TNBC cells, resulting in OTUD6B knockdown and controlled release of DOX. Additionally, siOTUD6B/DOX@Td treatment enhanced apoptosis rates increased DOX sensitivity, and inhibited TNBC cell growth, migration, and metastasis. Moreover, in vivo experiments confirmed that siOTUD6B/DOX@Td treatment inhibited tumor growth and metastasis without damaging the primary organs. Mechanistically, OTUD6B regulates TNBC progression by stabilizing murine double minute 2 (MDM2) and degrading forkhead box O3a (FOXO3a). Conclusively, this study demonstrates the potential applicability of DNA nanoparticles loaded with DOX and siOTUD6B for TNBC treatment.
Collapse
Affiliation(s)
- Wenxiang Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xue Yang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zheng Qu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peikai Ding
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xingsong Zhang
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China
| | - Ye Lu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China.
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
2
|
Chen Q, Liu Y, Chen Q, Li M, Xu L, Lin B, Tan Y, Liu Z. DNA Nanostructures: Advancing Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405231. [PMID: 39308253 DOI: 10.1002/smll.202405231] [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: 06/26/2024] [Revised: 08/28/2024] [Indexed: 12/06/2024]
Abstract
Cancer immunotherapy is a groundbreaking medical revolution and a paradigm shift from traditional cancer treatments, harnessing the power of the immune system to target and destroy cancer cells. In recent years, DNA nanostructures have emerged as prominent players in cancer immunotherapy, exhibiting immense potential due to their controllable structure, surface addressability, and biocompatibility. This review provides an overview of the various applications of DNA nanostructures, including scaffolded DNA, DNA hydrogels, tetrahedral DNA nanostructures, DNA origami, spherical nucleic acids, and other DNA-based nanostructures in cancer immunotherapy. These applications explore their roles in vaccine development, immune checkpoint blockade therapies, adoptive cellular therapies, and immune-combination therapies. Through rational design and optimization, DNA nanostructures significantly bolster the immunogenicity of the tumor microenvironment by facilitating antigen presentation, T-cell activation, tumor infiltration, and precise immune-mediated tumor killing. The integration of DNA nanostructures with cancer therapies ushers in a new era of cancer immunotherapy, offering renewed hope and strength in the battle against this formidable foe of human health.
Collapse
Affiliation(s)
- Qianqian Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Bingyu Lin
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| |
Collapse
|
3
|
Ye T, Luo Z, Che Y, Yuan M, Cao H, Hao L, Zhang Q, Xie Y, Zhang K, Xu F. An inverted tetrahedron-mediated DNA walker for sulfadimethoxine detection. Mikrochim Acta 2024; 191:724. [PMID: 39496845 DOI: 10.1007/s00604-024-06810-6] [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: 06/26/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024]
Abstract
An inverted DNA tetrahedron-mediated modular DNA walker was developed for the determination of sulfadimethoxine. The inverted DNA tetrahedron scaffold raises several advantages of recognition module including appropriate lateral space, multiple recognition domains, and cost-effectiveness. The proposed inverted DNA tetrahedron-based recognition module exhibited better binding affinity and kinetics toward target antibiotic than that of other DNA tetrahedron counterparts. Upon specific binding with target, the released bipedal DNA walking strand hops to the signal amplification module and moves stochastically with assistant of nicking enzyme. By coupling these two modules, a good linear relationship between the fluorescence intensity of supernatant and the concentration of sulfadimethoxine was achieved in the range 0.1-100 nM, and the limit of detection was 64.7 pM. Furthermore, this modular DNA walker had also successfully applied to spiked honey and milk samples with satisfactory recoveries from 91.5 to 108.8%, demonstrating its practical sensing capability.
Collapse
Affiliation(s)
- Tai Ye
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zheng Luo
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yueyue Che
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Min Yuan
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Hui Cao
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Liling Hao
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qian Zhang
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yongxin Xie
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Kaisen Zhang
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Fei Xu
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
4
|
Mohammadi F, Zahraee H, Zibadi F, Khoshbin Z, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Progressive cancer targeting by programmable aptamer-tethered nanostructures. MedComm (Beijing) 2024; 5:e775. [PMID: 39434968 PMCID: PMC11491555 DOI: 10.1002/mco2.775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
Scientific research in recent decades has affirmed an increase in cancer incidence as a cause of death globally. Cancer can be considered a plurality of various diseases rather than a single disease, which can be a multifaceted problem. Hence, cancer therapy techniques acquired more accelerated and urgent approvals compared to other therapeutic approaches. Radiotherapy, chemotherapy, immunotherapy, and surgery have been widely adopted as routine cancer treatment strategies to suppress disease progression and metastasis. These therapeutic approaches have lengthened the longevity of countless cancer patients. Nonetheless, some inherent limitations have restricted their application, including insignificant therapeutic efficacy, toxicity, negligible targeting, non-specific distribution, and multidrug resistance. The development of therapeutic oligomer nanoconstructs with the advantages of chemical solid-phase synthesis, programmable design, and precise adjustment is crucial for advancing smart targeted drug nanocarriers. This review focuses on the significance of the different aptamer-assembled nanoconstructs as multifunctional nucleic acid oligomeric nanoskeletons in efficient drug delivery. We discuss recent advancements in the design and utilization of aptamer-tethered nanostructures to enhance the efficacy of cancer treatment. Valuably, this comprehensive review highlights self-assembled aptamers as the exceptionally intelligent nano-biomaterials for targeted drug delivery based on their superior stability, high specificity, excellent recoverability, inherent biocompatibility, and versatile functions.
Collapse
Affiliation(s)
- Fatemeh Mohammadi
- Targeted Drug Delivery Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Pharmaceutical BiotechnologySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Hamed Zahraee
- Targeted Drug Delivery Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Pharmaceutical BiotechnologySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Farkhonde Zibadi
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Zahra Khoshbin
- Targeted Drug Delivery Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Medicinal ChemistrySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Mohammad Ramezani
- Pharmaceutical Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
| | - Mona Alibolandi
- Pharmaceutical Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
| | - Khalil Abnous
- Pharmaceutical Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Medicinal ChemistrySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Pharmaceutical BiotechnologySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
5
|
Huang J, Ullah I, Chen L, Peng K, Ou Y, Gong Z, Wu A, He Z, Yin W, Li H. H 2O 2 Self-Supplied MoS 2/CaO 2 Nanozyme Hydrogel with Near-Infrared Photothermal Synergetic Cascade Peroxidase-Like Activity for Wound Disinfection. Adv Healthc Mater 2024:e2402393. [PMID: 39370586 DOI: 10.1002/adhm.202402393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/25/2024] [Indexed: 10/08/2024]
Abstract
In wound healing and clinical anti-infection therapy, the current feasibility of nanocatalysts is extremely limited because of inadequate reactive oxygen species (ROS) generation. Herein, a novel H2O2 self-supplying nanocomposite (M/C/AEK) consists of molybdenum disulfide (MoS2) decorated with calcium peroxide (CaO2) prepared at ambient temperature and encapsulated in AEK hydrogel. In the presence of H2O2 and poly(vinyl pyrrolidone) (PVP), CaO2 nanoclusters, ≈30 nm, are anchored on the MoS2 surface. MoS2/CaO2 can induce both a cascaded peroxidase (POD)-like and catalase (CAT)-like catalytic activity to produce toxic hydroxyl radicals through self-supplied H2O2 and O2 responsive to the faintly acidic environment of acute wounds. The POD-like activity is increased under acidic compared with neutral conditions, allowing selective treatment of acute, slightly acidic wounds while avoiding the side effects of high-concentration antibacterial agents on normal tissues. The high near-infrared photothermal effect synergistically with POD-like/CAT-like activity of MoS2/CaO2 boosts the production of more ROS to eradicate Staphylococcus aureus and Escherichia coli bacteria (98.6% and 98.9%) effectively and selectively stimulate wound healing. The porous M/C/AEK hydrogel in the wound microenvironment can efficiently capture bacteria, and its Ca2+ ions and keratin stimulate healing, revealing excellent potential in advanced wound care and infection control therapies.
Collapse
Affiliation(s)
- Jinyi Huang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
| | - Ihsan Ullah
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P.R. China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Ke Peng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
| | - Yujie Ou
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
| | - Zehua Gong
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Zhimin He
- Faculty of Health, Education and Society, University of Northampton, Northampton, NN1 5PH, UK
| | - Wenyan Yin
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huaqiong Li
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, P. R. China
| |
Collapse
|
6
|
Shi X, Chen H, Yang H, Xue S, Li Y, Fang X, Ding C, Zhu Z. Aptamer-Modified Tetrahedral Framework Nucleic Acid Synergized with TGF-β3 to Promote Cartilage Protection in Osteoarthritis by Enhancing Chondrogenic Differentiation of MSCs. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50484-50496. [PMID: 39282962 DOI: 10.1021/acsami.4c12159] [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: 09/28/2024]
Abstract
Characterized by progressive and irreversible degeneration of the articular cartilage (AC), osteoarthritis (OA) is the most common chronic joint disease, and there is no cure for OA at present. Recent studies suggest that enhancing the recruitment of endogenous mesenchymal stem cells (MSCs) to damaged cartilage is a promising therapeutic strategy for cartilage repair. Tetrahedral framework nucleic acid (tFNA) is a novel DNA nanomaterial and has shown great potential in the field of biomedical science. Transforming growth factor-beta 3 (TGF-β3), a vital member of the highly conserved TGF-β superfamily, is considered to induce chondrogenesis. A 66-base DNA aptamer named HM69 is reported to identify and recruit MSCs. In this study, aptamer HM69-modified tFNAs were successfully self-assembled and used to load TGF-β3 when the disulfide bonds combined. We confirmed the successful synthesis of the final composition, HM69-tFNA@TGF-β3 (HTT), by PAGE, dynamic light scattering, and atomic force microscopy. The results of in vitro experiments showed that HTT effectively induced MSC proliferation, migration, and chondrogenic differentiation. In addition, HTT-treated MSCs were shown to protect the OA chondrocytes. In DMM mice, the injection of HTT improved the therapeutic outcome of mouse pain symptoms and AC degeneration. In conclusion, this study innovatively used the disulfide bonds combined with TGF-β3 and tFNA, and an additional sequence HM69 was loaded on tFNA for the better-targeted recruitment of MSCs. HTT demonstrated its role in promoting the chondrogenesis of MSCs and cartilage protection, indicating that it might be promising for OA therapy.
Collapse
Affiliation(s)
- Xiaorui Shi
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haowei Chen
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hao Yang
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Song Xue
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yang Li
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaofeng Fang
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Changhai Ding
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart 7000, Australia
| | - Zhaohua Zhu
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, Sydney 2065, Australia
| |
Collapse
|
7
|
Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M, Li Z. Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci 2024; 11:1472492. [PMID: 39329090 PMCID: PMC11425083 DOI: 10.3389/fmolb.2024.1472492] [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: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis, an iron-ion-dependent process of lipid peroxidation, damages the plasma membrane, leading to non-programmed cell death. Osteoarthritis (OA), a prevalent chronic degenerative joint disease among middle-aged and older adults, is characterized by chondrocyte damage or loss. Emerging evidence indicates that chondrocyte ferroptosis plays a role in OA development. However, most research has concentrated on ferroptosis regulation involving typical iron ions, potentially neglecting the significance of elevated copper ions in both serum and joint fluid of patients with OA. This review aims to fill this gap by systematically examining the interplay between copper metabolism, oxidative stress, ferroptosis, and copper-associated cell death in OA. It will provide a comprehensive overview of copper ions' role in regulating ferroptosis and their dual role in OA. This approach seeks to offer new insights for further research, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Qingyuan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yanan Xiao
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mengqi Guan
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xianshuai Zhang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jianan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mingze Han
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
8
|
Zhang Y, Wu B, Liu D, Chen Y, Xu Y, Fu L, Lin Z, Wu G, Huang F. Targeting HIF-1α with Specific DNA Yokes for Effective Anticancer Therapy. Adv Healthc Mater 2024; 13:e2401087. [PMID: 38696899 DOI: 10.1002/adhm.202401087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Indexed: 05/04/2024]
Abstract
Hypoxia, a ubiquitous hallmark in cancer, underscores the significance of targeting HIF-1α, the principal transcriptional factor of hypoxic responses, for effective cancer therapy. Herein, DNA yokes, a novel class of DNA nanomaterials harboring specific HIF-1α binding sequences (hypoxia response elements, HREs), are introduced as nanopharmaceuticals for cancer treatment. Comprising a basal tetrahedral DNA nanostructure and four HRE-bearing overhanging chains, DNA yokes exhibit exceptional stability and prolonged intracellular retention. The investigation reveals their capacity to bind HIF-1α, thereby disrupting its interaction with the downstream genomic DNAs and impeding transcriptional activity. Moreover, DNA yokes facilitate HIF-1α degradation via the ubiquitination pathway, thereby sequestering it from downstream targets and ultimately promoting its degradation. In addition, DNA yokes attenuate cancer cell proliferation, migration, and invasion under hypoxic conditions, while also displaying preferential accumulation within tumors, thereby inhibiting tumor growth and metastasis in vivo. This study pioneers a novel approach to cancer therapy through the development of DNA-based drugs characterized by high stability and low toxicity to normal cells, positioning DNA yokes as promising candidates for cancer treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Bing Wu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Danqing Liu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Yue Chen
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Yanfang Xu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Lengxi Fu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Gui Wu
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Department of Orthopaedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| | - Fei Huang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Central Laboratory, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
| |
Collapse
|
9
|
Singh R, Kansara K, Yadav P, Mandal S, Varshney R, Gupta S, Kumar A, Maiti PK, Bhatia D. DNA tetrahedral nanocages as a promising nanocarrier for dopamine delivery in neurological disorders. NANOSCALE 2024; 16:15158-15169. [PMID: 39091152 DOI: 10.1039/d4nr00612g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Dopamine is a neurotransmitter in the central nervous system that is essential for many bodily and mental processes, and a lack of it can cause Parkinson's disease. DNA tetrahedral (TD) nanocages are promising in bio-nanotechnology, especially as a nanocarrier. TD is highly programmable, biocompatible, and capable of cell differentiation and proliferation. It also has tissue and blood-brain barrier permeability, making it a powerful tool that could overcome potential barriers in treating neurological disorders. In this study, we used DNA TD as a carrier for dopamine to cells and zebrafish embryos. We investigated the mechanism of complexation between TD and dopamine hydrochloride using gel electrophoresis, fluorescence and circular dichroism (CD) spectroscopy, atomic force microscopy (AFM), and molecular dynamic (MD) simulation tools. Further, we demonstrate that these dopamine-loaded DNA TD nanostructures enhanced cellular uptake and differentiation ability in SH-SY5Y neuroblastoma cells. Furthermore, we extended the study to zebrafish embryos as a model organism to examine survival and uptake. The research provides valuable insights into the complexation mechanism and cellular uptake of dopamine-loaded DNA tetrahedral nanostructures, paving the way for further advancements in nanomedicine for Parkinson's disease and other neurological disorders.
Collapse
Affiliation(s)
- Ramesh Singh
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| | - Krupa Kansara
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| | - Pankaj Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| | - Sandip Mandal
- Department of Physics, Indian Institute of Science, Bangalore, India
| | - Ritu Varshney
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, India
| | - Prabal K Maiti
- Department of Physics, Indian Institute of Science, Bangalore, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujrat, India.
| |
Collapse
|
10
|
Lee M, Kim M, Lee M, Kim S, Park N. Nanosized DNA Hydrogel Functionalized with a DNAzyme Tetrahedron for Highly Efficient Gene Silencing. Biomacromolecules 2024; 25:4913-4924. [PMID: 38963792 DOI: 10.1021/acs.biomac.4c00356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
DNAzymes are DNA oligonucleotides that have catalytic activity without the assistance of protein enzymes. In particular, RNA-cleaving DNAzymes were considered as ideal candidates for gene therapy due to their unique characteristics. Nevertheless, efforts to use DNAzyme as a gene therapeutic agent are limited by issues such as their low physiological stability in serum and intracellular delivery efficiency. In this study, we developed a nanosized synthetic DNA hydrogel functionalized with a DNAzyme tetrahedron (TDz Dgel) to overcome these limitations. We observed remarkable improvement in the gene-silencing effect as well as intracellular uptake without the support of gene transfection reagents using TDz Dgel. The improved catalytic activity of the DNAzyme resulted from the combination of the cell-penetrating DNA tetrahedron structure and high stability of DNA hydrogel. We envision that this approach will become a convenient and efficient strategy for gene-silencing therapy using DNAzyme in the future.
Collapse
Affiliation(s)
- Minhyuk Lee
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Minchul Kim
- Department of Chemistry and the Natural Science Research Institute, Myongji University, 116 Myongji-ro, Yongin-si 17058, 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
| | - 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
|
11
|
Yadav K, Gnanakani SPE, Sahu KK, Veni Chikkula CK, Vaddi PS, Srilakshmi S, Yadav R, Sucheta, Dubey A, Minz S, Pradhan M. Nano revolution of DNA nanostructures redefining cancer therapeutics-A comprehensive review. Int J Biol Macromol 2024; 274:133244. [PMID: 38901506 DOI: 10.1016/j.ijbiomac.2024.133244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
DNA nanostructures are a promising tool in cancer treatment, offering an innovative way to improve the effectiveness of therapies. These nanostructures can be made solely from DNA or combined with other materials to overcome the limitations of traditional single-drug treatments. There is growing interest in developing nanosystems capable of delivering multiple drugs simultaneously, addressing challenges such as drug resistance. Engineered DNA nanostructures are designed to precisely deliver different drugs to specific locations, enhancing therapeutic effects. By attaching targeting molecules, these nanostructures can recognize and bind to cancer cells, increasing treatment precision. This approach offers tailored solutions for targeted drug delivery, enabling the delivery of multiple drugs in a coordinated manner. This review explores the advancements and applications of DNA nanostructures in cancer treatment, with a focus on targeted drug delivery and multi-drug therapy. It discusses the benefits and current limitations of nanoscale formulations in cancer therapy, categorizing DNA nanostructures into pure forms and hybrid versions optimized for drug delivery. Furthermore, the review examines ongoing research efforts and translational possibilities, along with challenges in clinical integration. By highlighting the advancements in DNA nanostructures, this review aims to underscore their potential in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai 490024, India
| | - S Princely E Gnanakani
- Department of Pharmaceutical Biotechnology, Parul Institute of Pharmacy, Parul University, Post Limda, Ta.Waghodia - 391760, Dist. Vadodara, Gujarat, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - C Krishna Veni Chikkula
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - Poorna Sai Vaddi
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - S Srilakshmi
- Gitam School of Pharmacy, Department of Pharmaceutical Chemistry, Gitams University, Vishakhapatnam, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak (M.P.), India
| | | |
Collapse
|
12
|
Sharma A, Vaswani P, Bhatia D. Revolutionizing cancer therapy using tetrahedral DNA nanostructures as intelligent drug delivery systems. NANOSCALE ADVANCES 2024; 6:3714-3732. [PMID: 39050960 PMCID: PMC11265600 DOI: 10.1039/d4na00145a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/24/2024] [Indexed: 07/27/2024]
Abstract
DNA nanostructures have surfaced as intriguing entities with vast potential in biomedicine, notably in the drug delivery area. Tetrahedral DNA nanostructures (TDNs) have received worldwide attention from among an array of different DNA nanostructures due to their extraordinary stability, great biocompatibility, and ease of functionalization. TDNs could be readily synthesized, making them attractive carriers for chemotherapeutic medicines, nucleic acid therapeutics, and imaging probes. Their varied uses encompass medication delivery, molecular diagnostics, biological imaging, and theranostics. This review extensively highlights the mechanisms of functional modification of TDNs and their applications in cancer therapy. Additionally, it discusses critical concerns and unanswered problems that require attention to increase the future application of TDNs in developing cancer treatment.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University Mathura Uttar Pradesh-281406 India
| | - Payal Vaswani
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj 382355 Gandhinagar India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj 382355 Gandhinagar India
| |
Collapse
|
13
|
Safarkhani M, Ahmadi S, Ipakchi H, Saeb MR, Makvandi P, Ebrahimi Warkiani M, Rabiee N, Huh Y. Advancements in Aptamer-Driven DNA Nanostructures for Precision Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401617. [PMID: 38713753 PMCID: PMC11234471 DOI: 10.1002/advs.202401617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Indexed: 05/09/2024]
Abstract
DNA nanostructures exhibit versatile geometries and possess sophisticated capabilities not found in other nanomaterials. They serve as customizable nanoplatforms for orchestrating the spatial arrangement of molecular components, such as biomolecules, antibodies, or synthetic nanomaterials. This is achieved by incorporating oligonucleotides into the design of the nanostructure. In the realm of drug delivery to cancer cells, there is a growing interest in active targeting assays to enhance efficacy and selectivity. The active targeting approach involves a "key-lock" mechanism where the carrier, through its ligand, recognizes specific receptors on tumor cells, facilitating the release of drugs. Various DNA nanostructures, including DNA origami, Tetrahedral, nanoflower, cruciform, nanostar, nanocentipede, and nanococklebur, can traverse the lipid layer of the cell membrane, allowing cargo delivery to the nucleus. Aptamers, easily formed in vitro, are recognized for their targeted delivery capabilities due to their high selectivity for specific targets and low immunogenicity. This review provides a comprehensive overview of recent advancements in the formation and modification of aptamer-modified DNA nanostructures within drug delivery systems.
Collapse
Affiliation(s)
- Moein Safarkhani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon, 22212, Republic of Korea
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran
| | - Sepideh Ahmadi
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon, 22212, Republic of Korea
| | - Hossein Ipakchi
- Department of Chemical Engineering, McMaster University, Hamilton, L8S 4L8, Canada
| | - Mohammad Reza Saeb
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, Gdańsk, 80-416, Poland
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 324000 Quzhou, Zhejiang, China
- Centre of Research Impact and Outreach, Chitkara University, Rajpura, Punjab, 140417, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, India
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices (IBMD), University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, India
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia
| | - YunSuk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon, 22212, Republic of Korea
| |
Collapse
|
14
|
Li F, Yan J, Wei C, Zhao Y, Tang X, Xu L, He B, Sun Y, Chang J, Liang Y. "Cicada Out of the Shell" Deep Penetration and Blockage of the HSP90 Pathway by ROS-Responsive Supramolecular Gels to Augment Trimodal Synergistic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401214. [PMID: 38647420 PMCID: PMC11220648 DOI: 10.1002/advs.202401214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/18/2024] [Indexed: 04/25/2024]
Abstract
Deep penetration and downregulation of heat shock protein (HSP) expression in multimodal synergistic therapy are promising approaches for curing cancer in clinical trials. However, free small-molecule drugs and most drug vehicles have a low delivery efficiency deep into the tumor owing to poor drug penetration and hypoxic conditions at the tumor site. In this study, the objective is to use reactive oxygen species (ROS)-responsive supramolecular gels co-loaded with the photosensitizer Zn(II) phthalocyanine tetrasulfonic acid (ZnPCS4) and functionalized tetrahedral DNA (TGSAs) (G@P/TGSAs) to enhance deep tissue and cell penetration and block the HSP90 pathway for chemo- photodynamic therapy (PDT) - photothermal therapy (PTT) trimodal synergistic therapy. The (G@P/TGSAs) are injected in situ into the tumor to release ZnPCS4 and TGSAs under high ROS concentrations originating from both the tumor and PDT. TGSAs penetrate deeply into tumor tissues and augment photothermal therapy by inhibiting the HSP90 pathway. Proteomics show that HSP-related proteins and molecular chaperones are inhibited/activated, inhibiting the HSP90 pathway. Simultaneously, the TGSA-regulated apoptotic pathway is activated. In vivo study demonstrates efficient tumor penetration and excellent trimodal synergistic therapy (45% tumor growth inhibition).
Collapse
Affiliation(s)
- Fashun Li
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Jianqin Yan
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Chen Wei
- Department of PharmacyQingdao Women and Children's HospitalQingdao266034China
| | - Yi Zhao
- Department of Recuperation MedicineQingdao Special Service Sanatorium of PLA NavyQingdao266071China
| | - Xiaowen Tang
- Department of Medicinal ChemistrySchool of PharmacyQingdao UniversityQingdao266073China
| | - Long Xu
- School of Materials Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Bin He
- National Engineering Research Center for BiomaterialsSichuan UniversityChengdu610064China
| | - Yong Sun
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Jing Chang
- College of Marine Life ScienceOcean University of ChinaQingdao266003China
| | - Yan Liang
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| |
Collapse
|
15
|
Hatami H, Rahiman N, Mohammadi M. Oligonucleotide based nanogels for cancer therapeutics. Int J Biol Macromol 2024; 267:131401. [PMID: 38582467 DOI: 10.1016/j.ijbiomac.2024.131401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/17/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Oligonucleotide-based nanogels, as nascent biomaterials, possess several unique functional, structural, and physicochemical features with excellent drug-loading capacity and high potential for cancer gene therapy. Ongoing studies utilizing oligonucleotide-based nanogels hold great promise, as these cutting-edge nanoplatforms can be elegantly developed with predesigned oligonucleotide sequences and complementary strands which are self-assembled or chemically crosslinked leading to the development of nanogels with predictable shape and tunable size with the desired functional properties. Current paper provides a summary of the properties, preparation methods, and applications of oligonucleotide-based nanogels in cancer therapy. The review is focused on both conventional and modified forms of oligonucleotide-based nanogels, including targeted nanogels, smart release nanogels (responsive to stimuli such as pH, temperature, and enzymes), as well as nanogels used for gene delivery. Their application in cancer immunotherapy and vaccination, photodynamic therapy, and diagnostic applications when combined with other nanoparticles is further discussed. Despite emerging designs in the development of oligonucleotide based nanogels, this field of study is still in its infancy, and clinical translation of these versatile nano-vehicles might face challenges. Hence, extensive research must be performed on in vivo behavior of such platforms determining their biodistribution, biological fate, and acute/subacute toxicity.
Collapse
Affiliation(s)
- Hooman Hatami
- Department of pharmaceutics, School of pharmacy, Mashhad University of Medical sciences, Mashhad, Iran
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of pharmaceutics, School of pharmacy, Mashhad University of Medical sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Li P, Huang Z, Duan X, Wang T, Yang S, Jiang D, Li J. PET image-guided kidney injury theranostics enabled by a bipyramidal DNA framework. Biomater Sci 2024; 12:2086-2095. [PMID: 38439626 DOI: 10.1039/d3bm01575k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Understanding the pharmacokinetic profiles of nanomaterials in living organisms is essential for their application in disease treatment. Bipyramidal DNA frameworks (BDFs) are a type of DNA nanomaterial that have shown prospects in the fields of molecular imaging and therapy. To serve as a reference for disease-related studies involving the BDF, we constructed a 68Ga-BDF and employed positron emission tomography (PET) imaging to establish its pharmacokinetic model in healthy mice. Our investigation revealed that the BDF was primarily eliminated from the body via the urinary system. Ureteral obstruction could significantly alter the metabolism of the urinary system. By utilizing the established pharmacokinetic model, we sensitively observed distinct imaging indicators in unilateral ureteral obstruction and acute kidney injury (a complication of ureteral obstruction) mouse models. Furthermore, we observed that the BDF showed therapeutic effects in an AKI model. We believe that the established pharmacokinetic model and unique renal excretion characteristics of the BDF will provide researchers with more information for studying kidney diseases.
Collapse
Affiliation(s)
- Pinghui Li
- Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhidie Huang
- Inner Mongolia Medical University, Hohhot 010050, China
| | - Xiaoyan Duan
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| | - Tao Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| | - Shaowen Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jianbo Li
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| |
Collapse
|
17
|
Lee ES, Woo J, Shin J, Cha BS, Kim S, Park KS. Tetrahedral DNA nanostructures enhance transcription isothermal amplification for multiplex detection of non-coding RNAs. Biosens Bioelectron 2024; 250:116055. [PMID: 38266617 DOI: 10.1016/j.bios.2024.116055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
This study introduces an innovative detection system for multiple cancer biomarkers, employing transcription isothermal amplification methods in conjunction with a tetrahedral DNA nanostructure (TDN). We demonstrate that TDN enhances various transcription isothermal amplification methods by placing DNA probes in proximity. Notably, the TDN-enhanced split T7 promoter-based isothermal transcription amplification with light-up RNA aptamer (STAR) system stands out for its optimal performance and operational simplicity, especially in identifying non-coding RNAs such as microRNAs and long non-coding RNAs (lncRNAs). Multiplex detection of lncRNAs was also achieved by generating distinct light-up RNA aptamers, each emitting unique fluorescence signals. The system effectively identified the target lncRNAs, demonstrating high sensitivity and selectivity in both cell lines and clinical samples. The system, utilizing the single enzyme T7 RNA polymerase, can be easily tailored for alternative targets by substituting target-specific sequences in DNA probes and seamlessly integrated with other isothermal amplification methods for greater sensitivity and accuracy in the detection of multiple cancer biomarkers.
Collapse
Affiliation(s)
- Eun Sung Lee
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jisu Woo
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jiye Shin
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Byung Seok Cha
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Seokjoon Kim
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ki Soo Park
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
18
|
Jang SJ, Kim TH. Triple multivalent aptamers within DNA tetrahedron on reduced graphene oxide electrode: Unlocking enhanced sensitivity and accelerated reactions in electrochemical sensing. Biosens Bioelectron 2024; 249:116039. [PMID: 38241797 DOI: 10.1016/j.bios.2024.116039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
DNA nanostructures are emerging as promising biosensing platforms due to their programmability, predictable assembly, and compatibility with aptamers for enhanced selectivity. This study focuses on a triple-multivalent aptamer (tApt) complex immobilized on a tetrahedral DNA nanostructure (TDN) and integrated with an electrochemically reduced graphene oxide (ERGO) electrode for highly sensitive mercury ion (Hg2+) detection. Compared to a linear multivalent aptamer-modified electrode (S2/ERGO-GCE), the 3D tApt/ERGO-GCE aptasensor exhibits superior sensitivity, signal amplification, and reaction kinetics. The tApt/ERGO-GCE sensor achieves an exceptional limit of detection (LOD) of 4.1 zM, surpassing the LOD of 0.71 fM for S2/ERGO-GCE. Additionally, the tApt/ERGO-GCE sensor demonstrates faster response times, with a half-saturation time (T1/2) of 6 minutes compared to 17 minutes for S2/ERGO/GCE. The 3D tApt aptamer's superior performance is attributed to its tetrahedral DNA structure integrated on ERGO, providing multiple aptamer binding sites, facilitating oriented immobilization on the electrode surface, and enhancing analyte capture and concentration. In contrast, the linear S2 aptamers lack rigidity, resulting in a disordered orientation on the electrode surface, hindering efficient Hg2+ binding and reducing target molecule binding efficiency. This study underscores the potential of triple-multivalent aptamer-based nanostructures for ultrasensitive and rapid biosensing applications. The tApt/ERGO-GCE aptasensor's exceptional sensitivity, signal amplification, and reaction kinetics make it a promising tool for Hg2+ detection and other biosensing applications.
Collapse
Affiliation(s)
- Seung Joo Jang
- Department of Chemistry, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Tae Hyun Kim
- Department of Chemistry, Soonchunhyang University, Asan 31538, Republic of Korea.
| |
Collapse
|
19
|
Zou W, Lu J, Zhang L, Sun D. Tetrahedral framework nucleic acids for improving wound healing. J Nanobiotechnology 2024; 22:113. [PMID: 38491372 PMCID: PMC10943864 DOI: 10.1186/s12951-024-02365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/21/2024] [Indexed: 03/18/2024] Open
Abstract
Wounds are one of the most common health issues, and the cost of wound care and healing has continued to increase over the past decade. In recent years, there has been growing interest in developing innovative strategies to enhance the efficacy of wound healing. Tetrahedral framework nucleic acids (tFNAs) have emerged as a promising tool for wound healing applications due to their unique structural and functional properties. Therefore, it is of great significance to summarize the applications of tFNAs for wound healing. This review article provides a comprehensive overview of the potential of tFNAs as a novel therapeutic approach for wound healing. In this review, we discuss the possible mechanisms of tFNAs in wound healing and highlight the role of tFNAs in modulating key processes involved in wound healing, such as cell proliferation and migration, angiogenesis, and tissue regeneration. The targeted delivery and controlled release capabilities of tFNAs offer advantages in terms of localized and sustained delivery of therapeutic agents to the wound site. In addition, the latest research progress on tFNAs in wound healing is systematically introduced. We also discuss the biocompatibility and biosafety of tFNAs, along with their potential applications and future directions for research. Finally, the current challenges and prospects of tFNAs are briefly discussed to promote wider applications.
Collapse
Affiliation(s)
- Wanqing Zou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, Guangdong, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China.
| | - Luyong Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| | - Duanping Sun
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, Guangdong, China.
| |
Collapse
|
20
|
Jayakumar R, Dash MK, Gulati S, Pandey A, Trigun SK, Joshi N. Preliminary data on cytotoxicity and functional group assessment of a herb-mineral combination against colorectal carcinoma cell line. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024; 21:61-70. [PMID: 38016708 DOI: 10.1515/jcim-2023-0221] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/01/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVES The invasive screening methods and the late stage diagnosis of colorectal carcinoma (CRC) are contributing for the devastative prognosis. The gradual shift of the disease pattern among younger generations requires the implementation of phytochemicals and traditional medicines. Arkeshwara rasa (AR) is a herb-mineral combination of Tamra bhasma/incinerated copper ashes and Dwigun Kajjali/mercury sulphide levigated with Calotropis procera leaf juice, Plumbago zeylanica root decoction and the decoction of three myrobalans (Terminalia chebula, Terminalia bellerica, Emblica Officinalis decoction)/Triphala decoction. METHODS The SW-480 cell line was checked for the cytotoxicity and the cell viability criteria with MTT(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay. The acridine orange/ethidium bromide (AO/EtBr) assay revealed the depth of apoptosis affected cells in the fluorescent images. The FTIR analysis exhibited the graphical spectrum of functional groups within the compound AR. RESULTS The IC50 from the 10-7 to 10-3 concentrations against SW-480 cells was 40.4 μg/mL. The staining of AO/EtBr was performed to visualize live and dead cells and it is evident from the result that number of apoptotic cells increases at increasing concentration of AR. The single bond with stretch vibrations of O-H and N-H are more concentrated in the 2,500-3,200 cm-1 and 3,700-4,000 cm-1 of the spectra whereas, the finger print region carries the O-H and S=O type peaks. CONCLUSIONS The AR shows strong cyto-toxicity against the SW-480 cells by inducing apoptosis. It also modulates cellular metabolism with the involvement of functional groups which antagonizes the strong acids. Moreover, these effects need to be analyzed further based in the in vivo and various in vitro models.
Collapse
Affiliation(s)
- Remya Jayakumar
- Department of Rasa Shastra and Bhaishajya Kalpana, Banaras Hindu University, Varanasi, India
| | - Manoj Kumar Dash
- Department of Rasa Shastra and Bhaishajya Kalpana, Government Ayurveda College, Raipur, Chhattisgarh, India
| | - Saumya Gulati
- Department of Rasa Shastra and Bhaishajya Kalpana, Babu Yugraj Singh Ayurvedic Medical College and Hospital, Lucknow, Uttar Pradesh, India
| | - Akanksha Pandey
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Surendra Kumar Trigun
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Namrata Joshi
- Department of Rasa Shastra and Bhaishajya Kalpana, Banaras Hindu University, Varanasi, India
| |
Collapse
|
21
|
Yan X, Wang F, Du H, Huo X, Zhang R, Zhou T, Wang X, Zhang G, Zhang Z. The switch of the DNA tetrahedral tweezers controlled by mercury ions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 306:123614. [PMID: 37939581 DOI: 10.1016/j.saa.2023.123614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
In this paper, the one-pot method is used to make the four DNA strands complement each other to construct the basic framework for DNA tetrahedral tweezers. To regulate the opening and closing of DNA tetrahedral tweezers, DNA strands with a high amount of T-base sequences is partially complementary to the tetrahedral framework. Hg2+ can form T-Hg-T hairpin structures with T-base. When DNA tetrahedral tweezers encounter Hg2+, the T-Hg-T structure is formed to shorten the connecting chain, and the tightening force causes the DNA tweezers to change from an open state to a closed state. Conversely, changes in fluorescence intensity due to the structure change can be used to detect the presence of Hg2+.
Collapse
Affiliation(s)
- Xiaoyan Yan
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Fang Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Huan Du
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Xiaobing Huo
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Ruyan Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Ting Zhou
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Xiufeng Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Guodong Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Zhiqing Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| |
Collapse
|
22
|
Liu B, Duan H, Liu Z, Liu Y, Chu H. DNA-functionalized metal or metal-containing nanoparticles for biological applications. Dalton Trans 2024; 53:839-850. [PMID: 38108230 DOI: 10.1039/d3dt03614f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The conjugation of DNA molecules with metal or metal-containing nanoparticles (M/MC NPs) has resulted in a number of new hybrid materials, enabling a diverse range of novel biological applications in nanomaterial assembly, biosensor development, and drug/gene delivery. In such materials, the molecular recognition, gene therapeutic, and structure-directing functions of DNA molecules are coupled with M/MC NPs. In turn, the M/MC NPs have optical, catalytic, pore structure, or photodynamic/photothermal properties, which are beneficial for sensing, theranostic, and drug loading applications. This review focuses on the different DNA functionalization protocols available for M/MC NPs, including gold NPs, upconversion NPs, metal-organic frameworks, metal oxide NPs and quantum dots. The biological applications of DNA-functionalized M/MC NPs in the treatment or diagnosis of cancers are discussed in detail.
Collapse
Affiliation(s)
- Bei Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| | - Zechao Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Yuechen Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| |
Collapse
|
23
|
Lim DY, Hwang BH. Aptamer-modified tetrahedral DNA nanostructure-immobilized liposome for specific gene delivery and potential cancer theragnostic. Biotechnol J 2024; 19:e2300156. [PMID: 37837335 DOI: 10.1002/biot.202300156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/19/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
Targeted delivery of therapeutic agents to cancer cells is crucial for effective cancer treatment without adverse effects. In this study, we developed a novel delivery carrier, Aptamer-modified tetrahedral DNA nanostructure (TDN) immobilized Liposome (ApTL), for specific delivery to nucleolin-overexpressing cancer cells. We demonstrated that targeted ApTL was highly effective in delivering plasmid and mRNA to nucleolin-overexpressing cancer cells compared to non-targeted ApTL with a non-specific aptamer. ApTL, which is highly negative and nano-sized, specifically delivered nucleic acids to MDA-MB-231 and HeLa cancer cells, primarily via lipid-raft-mediated endocytosis. Furthermore, the co-delivery of mRNA and doxorubicin resulted in increased apoptosis and reduced cancer cell viability. Interestingly, co-delivery of mRNA and Dox did not show a significant difference in EGFP expression at 24 h but dramatically increased EGFP expression at 48 h, making ApTL/mEGFP/Dox a promising candidate for detecting live cancer cells after targeted cancer drug treatment. Our results suggest that ApTL can be a promising tool for the targeted delivery of therapeutic agents to nucleolin-overexpressing cancer cells, providing a new strategy for cancer theragnostic.
Collapse
Affiliation(s)
- Doo Young Lim
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon, South Korea
| | - Byeong Hee Hwang
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon, South Korea
- Division of Bioengineering, Incheon National University, Incheon, South Korea
- Research Center for Bio Material & Process Development, Incheon National University, Incheon, Republic of Korea
- Institute for New Drug Development, Incheon National University, Incheon, South Korea
| |
Collapse
|
24
|
Jabbari A, Sameiyan E, Yaghoobi E, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Aptamer-based targeted delivery systems for cancer treatment using DNA origami and DNA nanostructures. Int J Pharm 2023; 646:123448. [PMID: 37757957 DOI: 10.1016/j.ijpharm.2023.123448] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/14/2023] [Accepted: 09/24/2023] [Indexed: 09/29/2023]
Abstract
Due to the limitations of conventional cancer treatment methods, nanomedicine has appeared as a promising alternative, allowing improved drug targeting and decreased drug toxicity. In the development of cancer nanomedicines, among various nanoparticles (NPs), DNA nanostructures are more attractive because of their precisely controllable size, shape, excellent biocompatibility, programmability, biodegradability, and facile functionalization. Aptamers are introduced as single-stranded RNA or DNA molecules with recognize their corresponding targets. So, incorporating aptamers into DNA nanostructures led to influential vehicles for bioimaging and biosensing as well as targeted cancer therapy. In this review, the recent developments in the application of aptamer-based DNA origami and DNA nanostructures in advanced cancer treatment have been highlighted. Some of the main methods of cancer treatment are classified as chemo-, gene-, photodynamic- and combined therapy. Finally, the opportunities and problems for targeted DNA aptamer-based nanocarriers for medicinal applications have also been discussed.
Collapse
Affiliation(s)
- Atena Jabbari
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Sameiyan
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Yaghoobi
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie-Curie, Ottawa, ON K1N 6N5, Canada
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
25
|
Cheng Y, Xue G, Lan L, Xu H, Cheng R, Song Q, Li C, Zhang J, Huang G, Shen Z, Xue C. Construction of a 3D rigidified DNA nanodevice for anti-interference and reinforced biosensing by turning nuclease into a catalyst. Biosens Bioelectron 2023; 237:115501. [PMID: 37392492 DOI: 10.1016/j.bios.2023.115501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/13/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
The practical application of DNA biosensors is impeded by numerous limitations in complicated physiological environments, particularly the susceptibility of common DNA components to nuclease degradation, which has been recognized as a major barrier in DNA nanotechnology. In contrast, the present study presents an anti-interference and reinforced biosensing strategy based on a 3D DNA-rigidified nanodevice (3D RND) by converting a nuclease into a catalyst. 3D RND is a well-known tetrahedral DNA scaffold containing four faces, four vertices, and six double-stranded edges. The scaffold was rebuilt to serve as a biosensor by embedding a recognition region and two palindromic tails on one edge. In the absence of a target, the rigidified nanodevice exhibited enhanced nuclease resistance, resulting in a low false-positive signal. 3D RNDs have been proven to be compatible with 10% serum for at least 8 h. Once exposed to the target miRNA, the system can be unlocked and converted into common DNAs from a high-defense state, followed by polymerase- and nuclease-co-driven conformational downgrading to achieve amplified and reinforced biosensing. The signal response can be improved by approximately 700% within 2 h at room temperature, and the limit of detection (LOD) is approximately 10-fold lower under biomimetic conditions. The final application to serum miRNA-mediated clinical diagnosis of colorectal cancer (CRC) patients revealed that 3D RND is a reliable approach to collecting clinical information for differentiating patients from healthy individuals. This study provides novel insights into the development of anti-interference and reinforced DNA biosensors.
Collapse
Affiliation(s)
- Yinghao Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China; College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, PR China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, Jiujiang, Jiangxi, 332000, PR China
| | - Linwen Lan
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, PR China
| | - Ruize Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Qiufeng Song
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Chan Li
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Jing Zhang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Guoqiao Huang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Zhifa Shen
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China.
| | - Chang Xue
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China.
| |
Collapse
|
26
|
Meng X, O'Hare D, Ladame S. Surface immobilization strategies for the development of electrochemical nucleic acid sensors. Biosens Bioelectron 2023; 237:115440. [PMID: 37406480 DOI: 10.1016/j.bios.2023.115440] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/20/2023] [Accepted: 05/27/2023] [Indexed: 07/07/2023]
Abstract
Following the recent pandemic and with the emergence of cell-free nucleic acids in liquid biopsies as promising biomarkers for a broad range of pathologies, there is an increasing demand for a new generation of nucleic acid tests, with a particular focus on cost-effective, highly sensitive and specific biosensors. Easily miniaturized electrochemical sensors show the greatest promise and most typically rely on the chemical functionalization of conductive materials or electrodes with sequence-specific hybridization probes made of standard oligonucleotides (DNA or RNA) or synthetic analogues (e.g. Peptide Nucleic Acids or PNAs). The robustness of such sensors is mostly influenced by the ability to control the density and orientation of the probe at the surface of the electrode, making the chemistry used for this immobilization a key parameter. This exhaustive review will cover the various strategies to immobilize nucleic acid probes onto different solid electrode materials. Both physical and chemical immobilization techniques will be presented. Their applicability to specific electrode materials and surfaces will also be discussed as well as strategies for passivation of the electrode surface as a way of preventing electrode fouling and reducing nonspecific binding.
Collapse
Affiliation(s)
- Xiaotong Meng
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK. https://in.linkedin.com/https://www.linkedin.com/profile/view?id=xiaotong-meng-888IC
| | - Danny O'Hare
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| | - Sylvain Ladame
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
27
|
Lu W, Chen T, Xiao D, Qin X, Chen Y, Shi S. Application and prospects of nucleic acid nanomaterials in tumor therapy. RSC Adv 2023; 13:26288-26301. [PMID: 37670995 PMCID: PMC10476027 DOI: 10.1039/d3ra04081j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023] Open
Abstract
Cancer poses a great threat to human life, and current cancer treatments, such as radiotherapy, chemotherapy, and surgery, have significant side effects and limitations that hinder their application. Nucleic acid nanomaterials have specific spatial configurations and can be used as nanocarriers to deliver different therapeutic drugs, thereby enabling various biomedical applications, such as biosensors and cancer therapy. In recent decades, a variety of DNA nanostructures have been synthesized, and they have demonstrated remarkable potential in cancer therapy related applications, such as DNA origami structures, tetrahedral framework nucleic acids, and dynamic DNA nanostructures. Importantly, more attention is also being paid to RNA nanostructures, which play an important role in gene therapy. Therefore, this review introduces the developmental history of nucleic acid nanotechnology, summarizes the applications of DNA and RNA nanostructures for tumor treatment, and discusses the development opportunities for nucleic acid nanomaterials in the future.
Collapse
Affiliation(s)
- Weitong Lu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Tianyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Xin Qin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Yang Chen
- Department of Pediatric Surgery, Department of Liver Surgery & Liver Transplantation Center, West China Hospital of Sichuan University Chengdu Sichuan 610041 China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| |
Collapse
|
28
|
Kasyanenko N, Baryshev A, Artamonova D, Sokolov P. Packaging of DNA Integrated with Metal Nanoparticles in Solution. ENTROPY (BASEL, SWITZERLAND) 2023; 25:1052. [PMID: 37509999 PMCID: PMC10378076 DOI: 10.3390/e25071052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
The transformation of high-molecular DNA from a random swollen coil in a solution to a discrete nanosized particle with the ordered packaging of a rigid and highly charged double-stranded molecule is one of the amazing phenomena of polymer physics. DNA condensation is a well-known phenomenon in biological systems, yet its molecular mechanism is not clear. Understanding the processes occurring in vivo is necessary for the usage of DNA in the fabrication of new biologically significant nanostructures. Entropy plays a very important role in DNA condensation. DNA conjugates with metal nanoparticles are useful in various fields of nanotechnology. In particular, they can serve as a basis for creating multicomponent nanoplatforms for theranostics. DNA must be in a compact state in such constructions. In this paper, we tested the methods of DNA integration with silver, gold and palladium nanoparticles and analyzed the properties of DNA conjugates with metal nanoparticles using the methods of atomic force microscopy, spectroscopy, viscometry and dynamic light scattering. DNA size, stability and rigidity (persistence length), as well as plasmon resonance peaks in the absorption spectra of systems were studied. The methods for DNA condensation with metal nanoparticles were analyzed.
Collapse
Affiliation(s)
- Nina Kasyanenko
- Faculty of Physics, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Andrei Baryshev
- Faculty of Physics, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Daria Artamonova
- Faculty of Physics, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Petr Sokolov
- Faculty of Physics, Saint Petersburg State University, Saint Petersburg 199034, Russia
| |
Collapse
|
29
|
Gabriele F, Palerma M, Ippoliti R, Angelucci F, Pitari G, Ardini M. Recent Advances on Affibody- and DARPin-Conjugated Nanomaterials in Cancer Therapy. Int J Mol Sci 2023; 24:ijms24108680. [PMID: 37240041 DOI: 10.3390/ijms24108680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Affibodies and designed ankyrin repeat proteins (DARPins) are synthetic proteins originally derived from the Staphylococcus aureus virulence factor protein A and the human ankyrin repeat proteins, respectively. The use of these molecules in healthcare has been recently proposed as they are endowed with biochemical and biophysical features heavily demanded to target and fight diseases, as they have a strong binding affinity, solubility, small size, multiple functionalization sites, biocompatibility, and are easy to produce; furthermore, impressive chemical and thermal stability can be achieved. especially when using affibodies. In this sense, several examples reporting on affibodies and DARPins conjugated to nanomaterials have been published, demonstrating their suitability and feasibility in nanomedicine for cancer therapy. This minireview provides a survey of the most recent studies describing affibody- and DARPin-conjugated zero-dimensional nanomaterials, including inorganic, organic, and biological nanoparticles, nanorods, quantum dots, liposomes, and protein- and DNA-based assemblies for targeted cancer therapy in vitro and in vivo.
Collapse
Affiliation(s)
- Federica Gabriele
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Marta Palerma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
30
|
Shishparenok AN, Furman VV, Zhdanov DD. DNA-Based Nanomaterials as Drug Delivery Platforms for Increasing the Effect of Drugs in Tumors. Cancers (Basel) 2023; 15:2151. [PMID: 37046816 PMCID: PMC10093432 DOI: 10.3390/cancers15072151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
DNA nanotechnology has significantly advanced and might be used in biomedical applications, drug delivery, and cancer treatment during the past few decades. DNA nanomaterials are widely used in biomedical research involving biosensing, bioimaging, and drug delivery since they are remarkably addressable and biocompatible. Gradually, modified nucleic acids have begun to be employed to construct multifunctional DNA nanostructures with a variety of architectural designs. Aptamers are single-stranded nucleic acids (both DNAs and RNAs) capable of self-pairing to acquire secondary structure and of specifically binding with the target. Diagnosis and tumor therapy are prospective fields in which aptamers can be applied. Many DNA nanomaterials with three-dimensional structures have been studied as drug delivery systems for different anticancer medications or gene therapy agents. Different chemical alterations can be employed to construct a wide range of modified DNA nanostructures. Chemically altered DNA-based nanomaterials are useful for drug delivery because of their improved stability and inclusion of functional groups. In this work, the most common oligonucleotide nanomaterials were reviewed as modern drug delivery systems in tumor cells.
Collapse
Affiliation(s)
- Anastasiya N. Shishparenok
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
| | - Vitalina V. Furman
- Center of Chemical Engineering, ITMO University, Kronverkskiy Prospekt 49A, 197101 St. Petersburg, Russia
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| |
Collapse
|
31
|
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
|
32
|
Zhang L, Wang Y, Karges J, Tang D, Zhang H, Zou K, Song J, Xiao H. Tetrahedral DNA Nanostructure with Interferon Stimulatory DNA Delivers Highly Potent Toxins and Activates the cGAS-STING Pathway for Robust Chemotherapy and Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210267. [PMID: 36484099 DOI: 10.1002/adma.202210267] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Indexed: 06/17/2023]
Abstract
Tumor metastases and reoccurrences are considered the leading cause of cancer-associated deaths. While highly efficient treatments for the eradication of primary tumors have been developed, the treatment of secondary or metastatic tumors remains poorly accessible. Over the past years, compounds that intervene through the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway against tumor metastases have emerged with potential for clinical development. While interferon stimulatory DNAs have demonstrated activation of this pathway, these compounds are associated with poor bioavailability, poor stability, and poor cancer selectivity, hindering their use for therapeutic applications. Herein, the encapsulation of a highly potent chemotherapeutic platinum(II) complex and the incorporation of interferon stimulatory DNA strands for activation of the cGAS-STING pathway into multimodal tetrahedral DNA nanostructures (84bp-TDNISD/56MESS ) for combined chemotherapy and immunotherapy is reported. It is found that 84bp-TDNISD/56MESS can work as not only a drug delivery carrier for highly potent toxins, but also an immunostimulant agent that can activate the STING pathway for antitumor immune responses. In a mouse breast cancer model, the DNA nanostructure is found to nearly fully eradicate primary as well as secondary/metastatic tumors, hence demonstrating its potential clinical translational value.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, P. R. China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yuqi Wang
- Institute of Basic Medicine and Cancer (IBMC), Cancer Hospital of the University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, P. R. China
| | - Johannes Karges
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Science, Beijing, 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Science, Beijing, 100049, P. R. China
| | - Kexuan Zou
- Institute of Basic Medicine and Cancer (IBMC), Cancer Hospital of the University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, P. R. China
| | - Jie Song
- Institute of Basic Medicine and Cancer (IBMC), Cancer Hospital of the University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Science, Beijing, 100049, P. R. China
| |
Collapse
|
33
|
Zhang P, Zhuo Y, Chai YQ, Yuan R. Structural DNA tetrahedra and its electrochemical-related surface sensing. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
34
|
Singh R, Yadav P, Naveena A H, Bhatia D. Cationic lipid modification of DNA tetrahedral nanocages enhances their cellular uptake. NANOSCALE 2023; 15:1099-1108. [PMID: 36562521 DOI: 10.1039/d2nr05749b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Self-assembled DNA nanocages are among the most promising candidates for bioimaging and payload delivery into cells. DNA nanocages have great potential to efficiently address drug resistance and nucleic acid delivery problems due to precise control of their shape and size, and excellent biocompatibility. Although DNA nanostructures demonstrate some cellular uptake, because they bear a highly negative charge, the uptake of tetrahedral nanostructures is hindered by electrostatic repulsion. In this study, we describe a method to enhance the cellular uptake of DNA nanostructures using a binary system containing DNA and a positively charged head group with a hydrophobic lipid chain containing lipids for cellular internalization. Here we represent the functionalization of a model cage, DNA tetrahedron (TD) with a cationic lipid, N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA). Atomic force microscopy (AFM) and other standard characterization techniques were used to explore the co-assembly of the DNA tetrahedron and DOTMA. We revealed a simple confocal microscopy-based approach to show the enhancement in the cellular uptake of DNA nanocages. This new method will find multiple applications in delivery applications such as gene transfection, drug delivery and targeted bioimaging.
Collapse
Affiliation(s)
- Ramesh Singh
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Pankaj Yadav
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Hema Naveena A
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
35
|
Yu H, Yan J, Li Z, Song T, Ning F, Tan J, Sun Y. Enhanced photothermal-ferroptosis effects based on RBCm-coated PDA nanoparticles for effective cancer therapy. J Mater Chem B 2023; 11:415-429. [PMID: 36512437 DOI: 10.1039/d2tb02329f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ferroptosis, a type of programmed cell death induced by the iron-dependent lipid hydroperoxide pathway, has attracted widespread attention. However, Fenton response-dependent ferroptosis has many limitations, such as insufficient reaction conditions in the tumor micro-environment. Here, we propose an all-in-one phototherapy nanoplatform consisting of iron-polydopamine (Fe-PDA), a folic acid-modified red blood cell membrane (FA-RBCm), and epirubicin (EPI), namely, Fe-PDA-EPI@FA-RBCm NPs, to achieve enhanced photothermal-ferroptosis effects via overcoming the limitations of the Fenton-like reaction. The results showed that the synthesized biomimetic nanoparticles could decompose hydrogen peroxide (H2O2) to generate hydroxyl radicals (˙OH), and further induce the non-apoptotic ferroptosis pathway. After irradiation with near-infrared (NIR) light, the uptake of Fe-PDA-EPI@FA-RBCm NPs by cells could be effectively promoted, and it presented impressive in vitro and in vivo photothermal properties. In vitro and in vivo results showed that laser irradiation could enhance ferroptosis by promoting the production of reactive oxygen species (ROS) and lipid peroxides, down-regulating the expression of glutathione peroxidase 4 (GPX4), and reducing the mitochondrial membrane potential. Furthermore, the photothermal-promoted ferroptosis and apoptosis pathways (photothermal therapy and chemotherapy) exhibited outstanding synergistic antitumor efficacy in vitro and in vivo, with an in vivo tumor inhibition rate as high as 76.95%. In conclusion, the construction of tumor-targeted biomimetic nanocarriers utilizing the advantageous properties of RBCm has been investigated as a potential anticancer strategy.
Collapse
Affiliation(s)
- Hongli Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Tingting Song
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Fang Ning
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Jinshan Tan
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
36
|
Snider DM, Pandit S, Coffin ML, Ebrahimi SB, Samanta D. DNA-Mediated Control of Protein Function in Semi-Synthetic Systems. Chembiochem 2022; 23:e202200464. [PMID: 36058885 DOI: 10.1002/cbic.202200464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Indexed: 01/25/2023]
Abstract
The development of strategies for controlling protein function in a precise and predictable manner has the potential to revolutionize catalysis, diagnostics, and medicine. In this regard, the use of DNA has emerged as a powerful approach for modulating protein activity. The programmable nature of DNA allows for constructing sophisticated architectures wherein proteins can be placed with control over position, orientation, and stoichiometry. This ability is especially useful considering that the properties of proteins can be influenced by their local environment or their proximity to other functional molecules. Here, we chronicle the different strategies that have been developed to interface DNA with proteins in semi-synthetic systems. We further delineate the unique applications unlocked by the unprecedented level of structural control that DNA affords. We end by outlining outstanding challenges in the area and discuss future research directions towards potential solutions.
Collapse
Affiliation(s)
- Dylan M Snider
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Subrata Pandit
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Mackenzie L Coffin
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Sasha B Ebrahimi
- Drug Product Development - Steriles, GlaxoSmithKline 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| |
Collapse
|
37
|
Fischer A, Zhang P, Ouyang Y, Sohn YS, Karmi O, Nechushtai R, Pikarsky E, Willner I. DNA-Tetrahedra Corona-Modified Hydrogel Microcapsules: "Smart" ATP- or microRNA-Responsive Drug Carriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204108. [PMID: 36351764 DOI: 10.1002/smll.202204108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/18/2022] [Indexed: 06/16/2023]
Abstract
The assembly of adenosine triphosphate (ATP)-responsive and miRNA-responsive DNA tetrahedra-functionalized carboxymethyl cellulose hydrogel microcapsules is presented. The microcapsules are loaded with the doxorubicin-dextran drug or with CdSe/ZnS quantum dots as a drug model. Selective unlocking of the respective microcapsules and the release of the loads in the presence of ATP or miRNA-141 are demonstrated. Functionalization of the hydrogel microcapsules a with corona of DNA tetrahedra nanostructures yields microcarriers that revealed superior permeation into cells. This is demonstrated by the effective permeation of the DNA tetrahedra-functionalized microcapsules into MDA-MB-231 breast cancer cells, as compared to epithelial MCF-10A nonmalignant breast cells. The superior permeation of the tetrahedra-functionalized microcapsules into MDA-MB-231 breast cancer cells, as compared to analog control hydrogel microcapsules modified with a corona of nucleic acid duplexes. The effective permeation of the stimuli-responsive, drug-loaded, DNA tetrahedra-modified microcapsules yields drug carriers of superior and selective cytotoxicity toward cancer cells.
Collapse
Affiliation(s)
- Amit Fischer
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Pu Zhang
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Yu Ouyang
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Yang Sung Sohn
- Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Ola Karmi
- Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Rachel Nechushtai
- Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, The Hebrew University, of Jerusalem, Jerusalem, 91904, Israel
| | - Itamar Willner
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
38
|
Combination of polythyleneimine regulating autophagy prodrug and Mdr1 siRNA for tumor multidrug resistance. J Nanobiotechnology 2022; 20:476. [DOI: 10.1186/s12951-022-01689-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
AbstractMultidrug resistance (MDR) has been restricting the efficacy of chemotherapy, which mainly include pump resistance and non-pump resistance. In order to fight overall MDR, a novel targeted gene/drug co-deliver nano system is developed, which can suppress the drug efflux pumps and modulate autophagy to overcoming both pump and non-pump resistance. Here, small interfere RNA (siRNA) is incorporated into polymer-drug conjugates (PEI-PTX, PP) which are composed of polyethyleneimine (PEI) and paclitaxel (PTX) via covalent bonds, and hyaluronic acid (HA) is coated on the surface of PP/siRNA to achieve long blood cycle and CD44-targeted delivery. The RNA interference to mdr1 gene is combined with autophagy inhibition by PP, which efficiently facilitate apoptosis of Taxol-resistant lung cancer cells (A549/T). Further study indicates that PEI in PP may play a significant role to block the autophagosome–lysosome fusion process by means of alkalizing lysosomes. Both in vitro and in vivo studies confirm that the nanoassemblies can successfully deliver PTX and siRNA into tumor cells and significantly inhibited A549/T tumor growth. In summary, the polymeric nanoassemblies provide a potential strategy for combating both pump and non-pump resistance via the synergism of RNAi and autophagy modulation.
Collapse
|
39
|
Zhang Y, Chen Y, Wu B, Liu D, Fu L, Huang F. Synthesis of tetrahedron DNA nanostructures for detecting the activation of cell signal transduction via their specific binding to transcriptional factors. NANOSCALE 2022; 14:15101-15110. [PMID: 36205195 DOI: 10.1039/d2nr01954j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A technique for detecting the activation of cell signal transduction is particularly important for disease diagnosis and therapy. Transcriptional factor (TF) activities that could indicate the status of cell signal transduction are a favorable target for cell signal detection. Tetrahedron DNA nanostructures (TDNs) which contain specific binding sequences of TFs were designed and synthesized in this research, and their effects on detecting cell signal transduction were evaluated. We found that FAM-labeled TDNs with the indicated TF binding sequences could specifically bind to activated TFs of hypoxia signaling or TGF-β signaling. Signaling pathway activities detected via TDNs could be exhibited by various methods including fluorescence imaging, flow cytometry and fluorescence spectrometer analysis. The reliability of this new technique is in line with the classical dual luciferase reporter assay system. This work develops a novel and effective tool to examine the activation of intracellular signaling pathways via nanotechnology. In addition, good stability and programmability of TDNs ensure their widespread application in various signaling pathways.
Collapse
Affiliation(s)
- Ying Zhang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Yue Chen
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Bing Wu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Danqing Liu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Lengxi Fu
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| | - Fei Huang
- Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
| |
Collapse
|
40
|
|
41
|
Patel A, Valsangkar V, Halvorsen K, Chandrasekaran AR. Purification of Self-Assembled DNA Tetrahedra Using Gel Electrophoresis. Curr Protoc 2022; 2:e560. [PMID: 36111849 PMCID: PMC9494925 DOI: 10.1002/cpz1.560] [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] [Indexed: 06/15/2023]
Abstract
DNA nanostructures have found applications in a variety of fields such as biosensing, drug delivery, cellular imaging, and computation. Several of these applications require purification of the DNA nanostructures once they are assembled. Gel electrophoresis-based purification of DNA nanostructures is one of the methods used for this purpose. Here, we describe a step-by-step protocol for a gel-based method to purify self-assembled DNA tetrahedra. With further optimization, this method could also be adapted for other DNA nanostructures. © 2022 Wiley Periodicals LLC. Basic Protocol: Purification of self-assembled DNA tetrahedra.
Collapse
Affiliation(s)
- Akul Patel
- The RNA Institute, University at Albany, State University of New York, New York
| | - Vibhav Valsangkar
- The RNA Institute, University at Albany, State University of New York, New York
| | - Ken Halvorsen
- The RNA Institute, University at Albany, State University of New York, New York
| | | |
Collapse
|
42
|
Zhong X, Dai X, Wang Y, Wang H, Qian H, Wang X. Copper-based nanomaterials for cancer theranostics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1797. [PMID: 35419993 DOI: 10.1002/wnan.1797] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/30/2022]
Abstract
Copper-based nanomaterials (Cu-based NMs) with favorable biocompatibility and unique properties have attracted the attention of many biomedical researchers. Cu-based NMs are one of the most widely studied materials in cancer treatment. In recent years, great progress has been made in the field of biomedicine, especially in the treatment and diagnosis of tumors. This review begins with the classification of Cu-based NMs and the recent synthetic strategies of Cu-based NMs. Then, according to the abundant and special properties of Cu-based NMs, their application in biomedicine is summarized in detail. For biomedical imaging, such as photoacoustic imaging, positron emission tomography imaging, and multimodal imaging based on Cu-based NMs are summarized, as well as strategies to improve the diagnostic effectiveness. Moreover, a series of unique structures and functions as well as the underlying property activity relationship of Cu-based NMs were shown to highlight their promising therapeutic performance. Cu-based NMs have been widely used in monotherapies, such as photothermal therapy (PTT) and chemodynamic therapy (CDT). Moreover, the sophisticated design in composition, structure, and surface fabrication of Cu-based NMs can endow these NMs with more modalities in cancer diagnosis and therapy. To further improve the efficiency of cancer treatment, combined therapy based on Cu-based NMs was introduced in detail. Finally, the challenges, critical factors, and future prospects for the clinical translation of Cu-based NMs as multifunctional theranostic agents were also considered and discussed. The aim of this review is to provide a better understanding and key consideration for the rational design of this increasingly important new paradigm of Cu-based NMs as theranostic agents. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Xiaoyan Zhong
- School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xingliang Dai
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| |
Collapse
|
43
|
Zhu C, Han S, Zeng X, Zhu C, Pu Y, Sun Y. Multifunctional thermo-sensitive hydrogel for modulating the microenvironment in Osteoarthritis by polarizing macrophages and scavenging RONS. J Nanobiotechnology 2022; 20:221. [PMID: 35526013 PMCID: PMC9077879 DOI: 10.1186/s12951-022-01422-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that can lead to disability. Blocking the complex malignant feedback loop system dominated by oxidative stress and pro-inflammatory factors is the key to treating OA. Here, we develop a multifunctional composite thermo-sensitive hydrogel (HPP@Cu gel), which is utilized by Poloxamer 407 (P407) and hyaluronic acid (HA) mixture as the gel matrix, then physically mixed with copper nanodots (Cu NDs) and platelet-rich plasma (PRP). Cu NDs is a novel nano-scavenger of reactive oxygen and nitrogen species (RONS) with efficient free radical scavenging activity. HPP@Cu gel is injected into the articular cavity, where it form an in situ gel that slowly released Cu NDs, HA, and PRP, prolonging the duration of drug action. Our results indicate that HPP@Cu gel could efficiently remove RONS from inflammatory sites and promote repolarization of macrophages to an anti-inflammatory phenotype. The HPP@Cu gel therapy dramatically reduces cartilage degradation and inflammatory factor production in OA rats. This study provides a reliable reference for the application of injectable hydrogels in inflammatory diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Chunrong Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Chunxiao Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
44
|
Cao C, Zhang T, Yang N, Niu X, Zhou Z, Wang J, Yang D, Chen P, Zhong L, Dong X, Zhao Y. POD Nanozyme optimized by charge separation engineering for light/pH activated bacteria catalytic/photodynamic therapy. Signal Transduct Target Ther 2022; 7:86. [PMID: 35342192 PMCID: PMC8958166 DOI: 10.1038/s41392-022-00900-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 01/18/2023] Open
Abstract
The current feasibility of nanocatalysts in clinical anti-infection therapy, especially for drug-resistant bacteria infection is extremely restrained because of the insufficient reactive oxygen generation. Herein, a novel Ag/Bi2MoO6 (Ag/BMO) nanozyme optimized by charge separation engineering with photoactivated sustainable peroxidase-mimicking activities and NIR-II photodynamic performance was synthesized by solvothermal reaction and photoreduction. The Ag/BMO nanozyme held satisfactory bactericidal performance against methicillin-resistant Staphylococcus aureus (MRSA) (~99.9%). The excellent antibacterial performance of Ag/BMO NPs was ascribed to the corporation of peroxidase-like activity, NIR-II photodynamic behavior, and acidity-enhanced release of Ag+. As revealed by theoretical calculations, the introduction of Ag to BMO made it easier to separate photo-triggered electron-hole pairs for ROS production. And the conduction and valence band potentials of Ag/BMO NPs were favorable for the reduction of O2 to ·O2−. Under 1064 nm laser irradiation, the electron transfer to BMO was beneficial to the reversible change of Mo5+/Mo6+, further improving the peroxidase-like catalytic activity and NIR-II photodynamic performance based on the Russell mechanism. In vivo, the Ag/BMO NPs exhibited promising therapeutic effects towards MRSA-infected wounds. This study enriches the nanozyme research and proves that nanozymes can be rationally optimized by charge separation engineering strategy.
Collapse
Affiliation(s)
- Changyu Cao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Tingbo Zhang
- School of Physics, Southeast University, Nanjing, 211189, China
| | - Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xianghong Niu
- School of Physics, Southeast University, Nanjing, 211189, China
| | - Zhaobo Zhou
- School of Physics, Southeast University, Nanjing, 211189, China
| | - Jinlan Wang
- School of Physics, Southeast University, Nanjing, 211189, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China.
| | - Peng Chen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Liping Zhong
- National Center for International Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China.
| | - Yongxiang Zhao
- National Center for International Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
45
|
Dou Y, Cui W, Yang X, Lin Y, Ma X, Cai X. Applications of tetrahedral DNA nanostructures in wound repair and tissue regeneration. BURNS & TRAUMA 2022; 10:tkac006. [PMID: 35280457 PMCID: PMC8912983 DOI: 10.1093/burnst/tkac006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/25/2022] [Indexed: 02/05/2023]
Abstract
Tetrahedral DNA nanostructures (TDNs) are molecules with a pyramidal structure formed by folding four single strands of DNA based on the principle of base pairing. Although DNA has polyanionic properties, the special spatial structure of TDNs allows them to penetrate the cell membrane without the aid of transfection agents in a caveolin-dependent manner and enables them to participate in the regulation of cellular processes without obvious toxic side effects. Because of their stable spatial structure, TDNs resist the limitations imposed by nuclease activity and innate immune responses to DNA. In addition, TDNs have good editability and biocompatibility, giving them great advantages for biomedical applications. Previous studies have found that TDNs have a variety of biological properties, including promoting cell migration, proliferation and differentiation, as well as having anti-inflammatory, antioxidant, anti-infective and immune regulation capabilities. Moreover, we confirmed that TDNs can promote the regeneration and repair of skin, blood vessels, muscles and bone tissues. Based on these findings, we believe that TDNs have broad prospects for application in wound repair and regeneration. This article reviews recent progress in TDN research and its applications.
Collapse
Affiliation(s)
- Yikai Dou
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610064, China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiao Yang
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610064, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
46
|
Wang M, Zhang Z, Huo Q, Wang M, Sun Y, Liu H, Chang J, He B, Liang Y. Targeted Polymeric Nanoparticles Based on Mangiferin for Enhanced Protection of Pancreatic β-Cells and Type 1 Diabetes Mellitus Efficacy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:11092-11103. [PMID: 35199981 DOI: 10.1021/acsami.1c22964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mangiferin (MGF) is found in many natural plants, such as Rhizoma Anemarrhenae, and has anti-diabetes effects. However, its clinical applications and development are limited by poor solubility and low-concentration enrichment in pancreatic islets. In this paper, targeted polymeric nanoparticles were constructed for MGF delivery with the desired drug loading content (6.86 ± 0.60%), excellent blood circulation, and missile-like delivery to the pancreas. Briefly, Glucagon-like peptide 1 (GLP-1) as an active targeting agent to the pancreas was immobilized on the block copolymer polyethyleneglycol-polycaprolactone (PEG-PCL) to obtain final GLP-1-PEG-PCL amphiphiles. Spherical MGF-loaded polymeric nanoparticles were acquired from the self-assembly of the targeted GDPP nanoparticles and MGF with a homogeneous size of 158.9 ± 1.7 nm and a negative potential for a good steady state in circulation. In this drug vehicle, GLP-1 acts as the missile vanguard via the GLP-1 receptor on the surface of the pancreas for improving the accumulation and efficiency of MGF in the pancreas, the hypoglycemic effect of MGF, and the restorative effect on pancreatic islets, which were investigated. As compared to free MGF, MGF/GDPP nanoparticles appeared to be more concentrated in the pancreas, with better blood glucose and glucose tolerance, enhanced insulin levels, increased β-cell proliferation, reduced β-cell apoptosis, and islet repair in vivo. This targeted drug delivery system provided a novel strategy and hope for enhancing MGF delivery and anti-diabetes efficacy.
Collapse
Affiliation(s)
- Mengdi Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Zhuoran Zhang
- Department of Dentistry, Qingdao Special Service Sanatorium of PLA Navy, Qingdao 266021, China
| | - Qingqing Huo
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Maolong Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Hongling Liu
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jing Chang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| |
Collapse
|
47
|
Targeting delivery of synergistic dual drugs with elastic PEG-modified multi-functional nanoparticles for hepatocellular carcinoma therapy. Int J Pharm 2022; 616:121567. [DOI: 10.1016/j.ijpharm.2022.121567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/09/2022] [Accepted: 02/07/2022] [Indexed: 11/19/2022]
|