1
|
Zhao Z, Zhou J, Li X, Zhang T, Tian Z, Sun T, Jiang C. Manganese-based virus-mimicking nanomedicine with triple immunomodulatory functions inhibits breast cancer brain metastasis. Biomaterials 2025; 320:123262. [PMID: 40138963 DOI: 10.1016/j.biomaterials.2025.123262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
Hindered by the challenges of blood-brain barrier (BBB) hindrance, tumor heterogeneity and immunosuppressive microenvironment, patients with breast cancer brain metastasis have yet to benefit from current clinical treatments, experiencing instead a decline in quality of life due to radiochemotherapy. While virus-mimicking nanosystems (VMN) mimicking viral infection processes show promise in treating peripheral tumors, the inability to modulate the immunosuppressive microenvironment limits the efficacy against brain metastasis. Accordingly, a VMN-based triple immunomodulatory strategy is initially proposed, aiming to activate innate and adaptive immune responses and reverse the immunosuppressive microenvironment. Here, manganese-based virus-mimicking nanomedicine (Vir-HD@HM) with intratumoral drug enrichment is engineered. Vir-HD@HM can induce the immune response through the activation of cGAS-STING by mimicking the in vivo infection process of herpesviruses. Meanwhile, DNAzyme mimicking the genome can rescue the epigenetic silencing of PTEN with the assistance of Mn2+, thus ameliorating the immunosuppressive metastatic microenvironment and achieving synergistic sensitizing therapeutic efficacy. In vivo experiments substantiate the efficacy of Vir-HD@HM in recruiting NK cells and CD8+ T cells to metastatic foci, inhibiting Treg cells infiltration, and prolonging murine survival without adjunctive radiochemotherapy. This study demonstrates that Vir-HD@HM with triple immunomodulation offers an encouraging therapeutic option for patients with brain metastasis.
Collapse
Affiliation(s)
- Zhenhao Zhao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Jingyi Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tongyu Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Zonghua Tian
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
2
|
Ye C, Liu H, Wang S, Zhang M, Zhang C, Yang F, Shen F, Wang L. Cascade-amplification-based electrochemical detection of Akashiwo sanguinea at pre-outbreak stage. Talanta 2025; 287:127671. [PMID: 39919474 DOI: 10.1016/j.talanta.2025.127671] [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: 12/03/2024] [Revised: 01/10/2025] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
Red tide events caused by Akashiwo sanguinea (A. sanguinea) pose a significant threat to ecosystems. However, studies that offer promising approaches for portable and onsite detection with precise identification of A. sanguinea remain insufficient. In this study, we developed an electrochemical biosensor (E-biosensor) for detecting A. sanguinea combined with cascade amplification strategies, termed TDW-E-biosensor. A predictive relationship was also established to predict algal cell density based on electrochemical signals. The experiment results showed that the TDW-E-biosensor was successfully applied for detecting A. sanguinea at the pre-outbreak stage and demonstrated excellent analytical performance, showing a low limit of detection (LOD) of 0.0676 fM and quantitation (LOQ) of 0.102 fM for the three-electrode system, and a low LOD of 6.873 fg μL-1 and LOQ of 20.460 fg μL-1 for the portable system. The accuracy of the TDW-E-biosensor was validated through comparison with droplet digital PCR (ddPCR) and Bland-Altman analysis, demonstrating a high level of agreement (a mean difference of 0.132 and a standard deviation of 0.184). The reliability of the predictive relationship was evidenced by controlled laboratory experiments and Bland-Altman analysis. The developed TDW-E-biosensor provides an innovative and promising tool for early warning efforts regarding harmful algae.
Collapse
Affiliation(s)
- Changrui Ye
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China
| | - Hongjie Liu
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China.
| | - Shaopeng Wang
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China; Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, PR China
| | - Man Zhang
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, PR China
| | - Chaoxin Zhang
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China
| | - Fulin Yang
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China
| | - Fang Shen
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, Guangxi University, Nanning, 530004, PR China.
| | - Liwei Wang
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, PR China
| |
Collapse
|
3
|
He H, Xie B, Li S, Gao H, Zhang J, Fu H, Liao Y. Modular Engineering of the DNAzyme-Based Electrochemiluminescence Biosensor with the Nitrogen and Sulfur Codoped Carbon Dots as Near-Infrared Emitters for MicroRNA detection. Anal Chem 2025. [PMID: 40298082 DOI: 10.1021/acs.analchem.5c00522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
DNAzyme-based biosensors remain at the forefront of microRNA (miRNA) analysis efforts. In this work, an electrochemiluminescence (ECL) biosensor technology integrating miRNA-initiated DNAzyme with a near-infrared ECL emission system was designed to detect miRNA. The DNAzyme was designed through the rational reconstitution of Mg2+-specific 8-17 DNAzyme. We engineered the DNAzyme strand with incorporation of a blocker strand to inhibit the DNAzyme activity. The blocker strand formed a duplex structure that prohibited the hybridization of the DNAzyme strand with the one arm of the substrate strand, leading to the formation of an inactive DNAzyme structure. In the presence of target miRNA, the hybridization between the target miRNA and the blocker strand would induce the breakage of the duplex, followed by the formation of the active DNAzyme. Thus, the activity of the DNAzyme biosensors could be switched from the OFF state into the ON state. Moreover, the nitrogen and sulfur codoped near-infrared emission carbon dots (NIR NS-CDs) were used as neoteric ECL emitters, which not only possessed an NIR ECL emission to reduce the photochemical damage but also had a low excitation potential range to avoid the side reaction. Therefore, by combining the DNAzyme modular engineering strategy and NIR NS-CDs as neoteric ECL emitters, the constructed ECL biosensor could realize the ultrasensitive analysis of miRNA-155 with an identification limit of 58.5 aM. This DNAzyme-guided NIR ECL emission of the neoteric CD strategy provides a sensitive and reliable method for miRNA detection, and it has great potential in clinical diagnosis.
Collapse
Affiliation(s)
- Haonan He
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Benting Xie
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Shuang Li
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Hejun Gao
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Juan Zhang
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Hongquan Fu
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| | - Yunwen Liao
- College of Chemistry and Chemical Engineering, Precise Synthesis and Function Development Key Laboratory of Sichuan Province, Institute of Applied Chemistry, China West Normal University, Nanchong, Sichuan637000, China
| |
Collapse
|
4
|
Muñoz-González M, Aguilar R, Moreno AA, Cepeda-Plaza M. Influence of LNA modifications on the activity of the 10-23 DNAzyme. RSC Adv 2025; 15:13031-13040. [PMID: 40271416 PMCID: PMC12016023 DOI: 10.1039/d5ra00161g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
The 10-23 DNAzyme is a catalytic DNA molecule that efficiently cleaves RNA in the presence of divalent cations such as Mg2+ or Ca2+. Following their discovery, the 10-23 DNAzymes demonstrated numerous advantages that quickly led them to be considered powerful molecular tools for the development of gene-silencing tools. In this study, we evaluate the efficiency of the 10-23 DNAzyme and an LNA-modified analog in cleaving human MALAT1, an RNA overexpressed in cancer cells. First, we perform in vitro assays using a 20 nt RNA fragment from the MALAT1 sequence, with 2 mM and 10 mM Mg2+ and Ca2+ as cofactors, to evaluate how LNA modifications influence catalytic activity. We found that the activity is increased in the LNA-modified DNAzyme compared to the unmodified version with both cofactors, in a concentration-dependent manner. Finally, the RNA-cleaving activity of the LNA-modified, catalytically active 10-23 DNAzyme was tested in MCF7 human breast cancer cells. We found that the DNAzyme persists for up to 72 h in cells and effectively silences MALAT1 RNA in a concentration-dependent manner as early as 12 h post-transfection.
Collapse
Affiliation(s)
- Marcelo Muñoz-González
- Chemical Sciences Department, Universidad Andres Bello Santiago Chile
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello Chile
- Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andres Bello Santiago Chile
| | - Rodrigo Aguilar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello Chile
| | - Adrian A Moreno
- Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andres Bello Santiago Chile
| | | |
Collapse
|
5
|
Muñoz-González M, Silva-Galleguillos V, Parra-Meneses V, Aguilar R, Cepeda-Plaza M. Catalytic mechanisms of the 8-17 and 10-23 DNAzymes: shared mechanistic strategies. Org Biomol Chem 2025. [PMID: 40243498 DOI: 10.1039/d5ob00387c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
RNA-cleaving DNAzymes have emerged as promising catalytic nucleic acids with potential applications in biotechnology and therapeutics. Among them, the 10-23 and 8-17 DNAzymes, both derived from the same in vitro selection experiment, have emerged as the most widely studied RNA-cleaving DNAzymes due to their high catalytic efficiency and broad metal ion dependence. Despite their apparent structural differences, recent structural, functional, and computational studies have revealed convergent catalytic strategies in their mechanisms. This review examines the commonalities between the 8-17 and 10-23 DNAzymes, offering a comparative mechanistic perspective on the catalytic strategies underlying their RNA cleavage activity, following the nomenclature proposed by Breaker et al. We discuss recent evidence from functional, crystallographic, NMR-based, and molecular dynamics studies that highlight how conserved guanine residues act as general bases, while hydrated metal ions contribute as general acids in both DNAzymes. By summarizing the latest advancements in the field, this review aims to provide a comprehensive perspective on the fundamental catalytic strategies employed by the 8-17 and the 10-23 DNAzymes and their relevance for future applications in nucleic acid-based catalysis.
Collapse
Affiliation(s)
- Marcelo Muñoz-González
- Chemical Sciences Department, Universidad Andres Bello, Santiago, Chile.
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile
| | | | | | - Rodrigo Aguilar
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile
| | | |
Collapse
|
6
|
Hilber S, Attionu SK, Dayie TK, Kreutz C. Advances in Isotope Labeling for Solution Nucleic Acid Nuclear Magnetic Resonance Spectroscopy. Chempluschem 2025:e2400752. [PMID: 40202339 DOI: 10.1002/cplu.202400752] [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: 12/02/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/10/2025]
Abstract
The availability of structural biology methods for nucleic acid still lags behind that of proteins, as evidenced by the smaller number of structures (DNA: 2513, RNA: 1899, nucleic acid-protein complexes: 13 842, protein: 196 887) deposited in the protein database. The skewed ratio of nucleic acid structures, relative to proteins (≈1:50), is inverted with respect to the cellular output of RNA and proteins in higher organisms (≈50:1). While nuclear magnetic resonance (NMR) is an attractive biophysical tool capable of bridging this gap at the molecular level, the conformational flexibility, line broadening, and low chemical shift dispersion of nucleic acids have made the NMR method challenging, especially for structures larger than 35 nucleotides. The incorporation of NMR-active isotopes is a f strategy to combat these problems. Significant strides made to push the size limits of nucleic acid structures solved by NMR using chemoenzymatic 13C- methyl and aromatic 15N- and 19F-13C-labeling are reviewed and challenges and opportunities are evaluated. Combining these isotopic labeling patterns with superior NMR spectroscopic properties, and new DNA/RNA synthesis methods (palindrome-nicking-dependent amplification and segmental labeling and site-specific modifications by template-directed tension), may stimulate advances in NMR studies of large DNA/RNA and their complexes with important biological functions.
Collapse
Affiliation(s)
- Stefan Hilber
- Institute of Organic Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain, 80/82, 6020, Innsbruck, Austria
| | - Solomon Kojo Attionu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20782, USA
| | - Theodore Kwaku Dayie
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20782, USA
| | - Christoph Kreutz
- Institute of Organic Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain, 80/82, 6020, Innsbruck, Austria
| |
Collapse
|
7
|
Kolpashchikov DM, Gerasimova YV. Cleavage of Structured RNAs Is Accelerated by High Affinity DNAzyme Agents. Chembiochem 2025; 26:e202400950. [PMID: 39901000 DOI: 10.1002/cbic.202400950] [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: 11/21/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/05/2025]
Abstract
DNAzymes (Dz) have been suggested as sequence-specific agents for cleaving RNA for therapeutic purposes. This concept paper discusses the challenges of Dz 10-23 design to effciently cleave folded RNA substrates. Dz with traditionally designed RNA binding arms (Tm~37 °C) have low affinity to the folded RNA substrates, which limits the overall cleavage rate. The RNA cleavage can be facilitated using Dz with high-affinity arms. However, this strategy is efficient only for cleaving RNA into folded RNA products. The unfolded products inhibit multiple substrate turnover. In a more general approach, Dz should be equipped with additional RNA binding arms to achieve tight RNA binding. This can be accomplished by bivalent and multivalent Dz constructs that have multiple catalytic cores. In all cases, high selectivity toward single nucleotide variations can be achieved in addition to multiple turnovers. The presence of RNase H, which plays a role in the antisense effect of oligonucleotide gene therapy agents, stabilizes the Dz:RNA complex and reduces its selectivity but significantly increases RNA cleavage efficiency. This work proposes changes in the algorithms of Dz design, which can help in constructing potent Dz agents for RNA inhibition both in cell cultures and in vivo. The concept article is supplemented with a quiz, which tests knowledge of the main concepts discussed in this work.
Collapse
Affiliation(s)
- Dmitry M Kolpashchikov
- Chemistry Department, University of Central Florida, Orlando, FL, 32816-2366, USA
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32816, USA
| | - Yulia V Gerasimova
- Chemistry Department, University of Central Florida, Orlando, FL, 32816-2366, USA
| |
Collapse
|
8
|
Zhou D, Li Z, Bao L, Zhao X, Hao J, Xu C, Sun F, He D, Jiang C, Zeng T, Li D. DNAzyme hydrogels specifically inhibit the NLRP3 pathway to prevent radiation-induced skin injury in mice. J Nanobiotechnology 2025; 23:238. [PMID: 40119386 PMCID: PMC11929335 DOI: 10.1186/s12951-025-03147-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/22/2025] [Indexed: 03/24/2025] Open
Abstract
Radiation-induced skin injury (RISI) is a frequent complication of radiotherapy, yet current preventive strategies exhibit suboptimal efficacy. Our previous publications have consistently demonstrated the effectiveness of biomaterials and hydrogels in preventing RISI. Based on comprehensive literature reviews, we speculate that NLRP3 overexpression plays a central role in the development of RISI. Therefore, designing DNAzyme (DZ)-hydrogels with targeted inhibition of NLRP3 overexpression is crucial for preventing RISI.To achieve this, we designed and screened the optimal NLRP3-DZ using bioinformatics, molecular dynamics, and gel electrophoresis methods. We encapsulated the NLRP3-DZ within ZIF-8 to enhance its stability, controlled release, and safety. To enhance the material's transdermal penetration and practicality, we attached the TAT transmembrane peptide. The final preparation and characterization of NLRP3-DZ@ZIF-8/TAT was achieved.In vitro cell models revealed that DZ-hydrogels exhibit high biosafety, effectively inhibit NLRP3 expression, promote cell migration, inhibit cell apoptosis, and possess antibacterial properties. Genomics analysis suggested that DZ-hydrogels may exert these functions by regulating changes in relevant mRNA pathways.Furthermore, we established a mouse model of RISI and found that the material can promote wound healing by regulating proteins associated with apoptosis, oxidative stress, and the inflammatory response. These research findings provide valuable insights for the prevention of RISI using DZ-hydrogels.
Collapse
Affiliation(s)
- Daijun Zhou
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Zhihui Li
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Linbo Bao
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Xiang Zhao
- Department of Oncology, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jie Hao
- Department of Oncology, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Chuan Xu
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Feifan Sun
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Dan He
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Chaoyang Jiang
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China.
| | - Tian Zeng
- Department of Oncology, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Dong Li
- Department of Oncology, General Hospital of Western Theater Command, Chengdu, 610083, China.
| |
Collapse
|
9
|
Wei M, Zhu Z, Wan L, Li Y. Nonspecific metal-coordination-driven control over higher-order DNA self-assembly. NANOSCALE 2025; 17:6676-6684. [PMID: 39950942 DOI: 10.1039/d4nr03516j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The interactions between chemicals and DNA molecules provide effective regulation tools for dynamically controlling the self-assembly of higher-order DNA nanostructures, which mostly rely on non-covalent π-π stacking, hydrogen bonding and electrostatic interactions. If strong covalent interactions could be introduced as a new regulation strategy, the current control toolbox in DNA nanotechnology would be greatly enriched. Herein, we adopt the silver ion (Ag+) to demonstrate a general, versatile coordination-driven regulation strategy for higher-order DNA self-assembly and systematically explore the impacts of Ag+ on the assembly and stability of DNA origami and tile-based nanostructures. The kilobase single-stranded scaffold DNA is condensed into uniform nanoparticles by Ag+, therefore inhibiting the formation of DNA origami during thermal annealing. Switchable disassembly and re-assembly of DNA tile-based architectures through Ag+ and cysteine have been proved. The coordination-driven regulation strategy in this work could in principle be expanded to other metal ions, which might bring unique functions and controls to higher-order DNA self-assembly through metal coordination chemistry.
Collapse
Affiliation(s)
- Mengzhou Wei
- Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| | - Zhiyuan Zhu
- Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| | - Lingjun Wan
- Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| | - Yulin Li
- Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| |
Collapse
|
10
|
Stockert JC, Horobin RW. Prebiotic RNA self-assembling and the origin of life: Mechanistic and molecular modeling rationale for explaining the prebiotic origin and replication of RNA. Acta Histochem 2025; 127:152226. [PMID: 39788859 DOI: 10.1016/j.acthis.2024.152226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
In recent years, a great interest has been focused on the prebiotic origin of nucleic acids and life on Earth. An attractive idea is that life was initially based on an autocatalytic and autoreplicative RNA (the RNA-world). RNA duplexes are right-handed helical chains with antiparallel orientation, but the rationale for these features is not yet known. An antiparallel (inverted) stacking of purine nucleosides was reported in crystallographic studies. Molecular modeling also supports the inverted orientation of nucleosides. This preferential stacking can also appear when nucleosides are included in a montmorillonite clay matrix. Free-energy values and geometrical parameters show that D-ribose chirality is preferred for the formation of right-handed RNA molecules. Thus, a "zipper" model with antiparallel and auto-intercalated nucleosides linked by phosphate groups can be proposed to form single RNA chains. Unstacking with strand separation and base pairing by H-bonding, results in shortening and inclination of ribose-phosphate chains, leading to right-handed helicity and antiparallel duplexes. Incorporation of complementary precursors on the major groove template by a self-assembly mechanism provides a prebiotic (non-enzymatic) "tetris" replication model by formation of a transient RNA tetrad and tetraplex. Original hairpin motifs appear as simple building units that form typical RNA structures such as hammerheads, cloverleaves and dumbbells. They occur today in the circular viroids and virusoids, as well as in highly branched and complex rRNA molecules.
Collapse
Affiliation(s)
- Juan C Stockert
- Institute of Health and Environmental Sciences, Prosama Foundation, Paysandú 752, Buenos Aires, CABA CP1405, Argentina; Integrative Center of Biology and Applied Chemistry, University Bernardo O'Higgins, General Gana 1702, Santiago 8370854, Chile.
| | | |
Collapse
|
11
|
Liu Y, Zhang S, Zhang M, Liu X, Wu Y, Wu Q, Chaput J, Wang Y. Chemical evolution of ASO-like DNAzymes for effective and extended gene silencing in cells. Nucleic Acids Res 2025; 53:gkaf144. [PMID: 40037707 PMCID: PMC11879453 DOI: 10.1093/nar/gkaf144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Antisense oligonucleotides (ASOs) and small interfering RNA (siRNA) therapeutics highlight the power of oligonucleotides in silencing disease-causing messenger RNAs (mRNAs). Another promising class of gene-silencing oligonucleotides is RNA-cleaving nucleic acid enzymes, which offer the potential for allele-specific RNA inhibition with greater precision than ASOs and siRNAs. Herein, we chemically evolved the nucleolytic DNA enzyme (DNAzyme) 10-23, by incorporating the modifications that are essential to the success of ASO drugs, including 2'-fluoro, 2'-O-methyl, and 2'-O-methoxyethyl RNA analogues, and backbone phosphorothioate, to enhance catalytic efficiency by promoting RNA substrate binding and preventing dimerization of 10-23. These ASO-like DNAzymes cleaved structured RNA targets in long transcripts, showed prolonged intracellular stability, and downregulated mRNA and protein levels of both exogenously transfected eGFP and endogenously elevated oncogenic c-MYC. In colon cancer HCT116 cells, the downregulation of oncogenic c-MYC RNA resulted in cell cycle arrest, reduced proliferation, and increased apoptosis. RACE (rapid amplification of cDNA ends) polymerase chain reaction and Sanger sequencing confirmed precise, site-specific mRNA transcript cleavage with minimal RNase H activation in cells. By merging ASO structural and pharmacokinetic advantages with DNAzyme catalytic versatility, these ASO-like 10-23 variants offer a promising new class of potent gene-silencing agents, representing a significant step toward therapeutic DNAzyme development.
Collapse
Affiliation(s)
- Yingyu Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Sheyu Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Meiqi Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Xin Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yashu Wu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Qin Wu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Zhejiang Cancer Hospital, The University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - John C Chaput
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697-3958, United States
- Department of Chemistry, University of California, Irvine, CA 92697-3958, United States
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3958, United States
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697-3958, United States
| | - Yajun Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Zhejiang Cancer Hospital, The University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
12
|
Li C, Xu T, Hou G, Wang Y, Fu Q. DNA nanotechnology-based strategies for gastric cancer diagnosis and therapy. Mater Today Bio 2025; 30:101459. [PMID: 39866794 PMCID: PMC11762204 DOI: 10.1016/j.mtbio.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Gastric cancer (GC) is a formidable adversary in the field of oncology. The low early diagnosis rate of GC results in a low overall survival rate. Therefore, early accurate diagnosis and effective treatment are the key to reduce the mortality of GC. With the advent of nanotechnology, researchers continue to explore new possibilities for accurate diagnosis and effective treatment. One such breakthrough is the application of DNA nanotechnology. In this paper, the application of exciting DNA nanomaterials in the diagnosis and treatment of GC is discussed in depth. Firstly, the biomarkers related to GC and the diagnostic strategies related to DNA nanotechnology are summarized. Second, the latest research progress of DNA nanomaterials in the GC targeted therapy are summarized. Finally, the challenges and opportunities of DNA nanomaterials in the research and clinical application of GC are prospected.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Tongyang Xu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Guopeng Hou
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Yin Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Qinrui Fu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| |
Collapse
|
13
|
Xiao L, Zhu H, Gao Y, Xu L. Host-guest recognition-mediated reversible and orthogonal regulation of DNAzyme activity. Chem Commun (Camb) 2025; 61:1890-1893. [PMID: 39775627 DOI: 10.1039/d4cc05330c] [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: 01/11/2025]
Abstract
The host-guest recognition system is incorporated into the core region of the 10-23 DNAzyme for precise regulation of its functionality. Biochemical experiments demonstrate reversible and orthogonal control of the DNAzyme function using cucurbit(7)uril and its competitive guests. Furthermore, cellular experiments indicate the gene expression can be effectively manipulated through this ligand-controllable DNAzyme.
Collapse
Affiliation(s)
- Lin Xiao
- School of Laboratory Medicine, Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College & Translational Medicine Research Center, North Sichuan Medical College, Nanchong 637000, China
| | - Haitao Zhu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Yanmei Gao
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China.
| | - Liang Xu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
14
|
Aguilar R, Mardones C, Moreno AA, Cepeda-Plaza M. A guide to RNA structure analysis and RNA-targeting methods. FEBS J 2024. [PMID: 39718192 DOI: 10.1111/febs.17368] [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: 08/10/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
RNAs are increasingly recognized as promising therapeutic targets, susceptible to modulation by strategies that include targeting with small molecules, antisense oligonucleotides, deoxyribozymes (DNAzymes), or CRISPR/Cas13. However, while drug development for proteins follows well-established paths for rational design based on the accurate knowledge of their three-dimensional structure, RNA-targeting strategies are challenging since comprehensive RNA structures are yet scarce and challenging to acquire. Numerous methods have been developed to elucidate the secondary and three-dimensional structure of RNAs, including X-ray crystallography, cryo-electron microscopy, nuclear magnetic resonance, SHAPE, DMS, and bioinformatic methods, yet they have often revealed flexible transcripts and co-existing populations rather than single-defined structures. Thus, researchers aiming to target RNAs face a critical decision: whether to acquire the detailed structure of transcripts in advance or to adopt phenotypic screens or sequence-based approaches that are independent of the structure. Still, even in strategies that seem to rely only on the nucleotide sequence (like the design of antisense oligonucleotides), researchers may need information about the accessibility of the compounds to the folded RNA molecule. In this concise guide, we provide an overview for researchers interested in targeting RNAs: We start by revisiting current methodologies for defining secondary or three-dimensional RNA structure and then we explore RNA-targeting strategies that may or may not require an in-depth knowledge of RNA structure. We envision that complementary approaches may expedite the development of RNA-targeting molecules to combat disease.
Collapse
Affiliation(s)
- Rodrigo Aguilar
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Constanza Mardones
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Adrian A Moreno
- Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | |
Collapse
|
15
|
Xie X, Nan H, Peng J, Zeng K, Wang HH, Huang Y, Nie Z. Hydrogen Sulfide-Triggered Artificial DNAzyme Switches for Precise Manipulation of Cellular Functions. Angew Chem Int Ed Engl 2024; 63:e202410380. [PMID: 39327234 DOI: 10.1002/anie.202410380] [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: 06/02/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
The development of synthetic molecular tools responsive to biological cues is crucial for advancing targeted cellular regulation. A significant challenge is the regulation of cellular processes in response to gaseous signaling molecules such as hydrogen sulfide (H2S). To address this, we present the design of Gas signaling molecule-Responsive Artificial DNAzyme-based Switches (GRAS) to manipulate cellular functions via H2S-sensitive synthetic DNAzymes. By incorporating stimuli-responsive moieties to the phosphorothioate backbone, DNAzymes are strategically designed with H2S-responsive azide groups at cofactor binding locations within the catalytic core region. These modifications enable their activation through H2S-reducing decaging, thereby initiating substrate cleavage activity. Our approach allows for the flexible customization of various DNAzymes to regulate distinct cellular processes in diverse scenarios. Intracellularly, the enzymatic activity of GRAS promotes H2S-induced cleavage of specific mRNA sequences, enabling targeted gene silencing and inducing apoptosis in cancer cells. Moreover, integrating GRAS with dynamic DNA assembly allows for grafting these functional switches onto cell surface receptors, facilitating H2S-triggered receptor dimerization. This extracellular activation transmits signals intracellularly to regulate cellular behaviors such as migration and proliferation. Collectively, synthetic switches are capable of rewiring cellular functions in response to gaseous cues, offering a promising avenue for advanced targeted cellular engineering.
Collapse
Affiliation(s)
- Xuan Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Hexin Nan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Jialong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Kaiqiang Zeng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Yan Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| |
Collapse
|
16
|
Zhao L, Zhao M, Zhou X, Yuan R, Zhong X, Zhuo Y. A close-looped DNAzyme walker with an available catalytic domain for electrochemiluminescent detection of acetamiprid. Food Chem 2024; 458:140202. [PMID: 38954955 DOI: 10.1016/j.foodchem.2024.140202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/02/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
Traditional DNA walkers face enormous challenges due to limited biostability and reaction kinetics. Herein, we designed a self-driven close-looped DNAzyme walker (cl-DW) with high structural biostability and catalytic activity that enabled rapid electrochemiluminescence (ECL) detection of pesticide residue acetamiprid. Specifically, cl-DW exhibited increasing ability to resist nuclease degradation with a 570-fold longer half-degradation time than that of the single-stranded DNAzyme walker (ss-DW) due to the protected DNA terminal. Furthermore, cl-DW achieved high catalytic activity with a 4.3-fold faster reaction kinetic than that of ss-DW due to the circularized nanostructure of an available catalytic domain. Consequently, we utilized cl-DW as a signal amplifier and tin-based sulfide (SnS2) nanoflowers as ECL emitters to construct an ECL aptasensor, which realized the sensitive detection of acetamiprid with a limit of detection of 0.85 nM. This work provides a reliable approach to exploring DNA walkers with high catalytic activity and better biostability for molecular monitoring.
Collapse
Affiliation(s)
- Lidan Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Meiling Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Xuemei Zhou
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Xia Zhong
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China.
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
17
|
Das PK, Silverman SK. Sequence-Dependent Acylation of Peptide Lysine Residues by DNAzymes. Chembiochem 2024; 25:e202400578. [PMID: 39239825 PMCID: PMC11543514 DOI: 10.1002/cbic.202400578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/07/2024]
Abstract
Methods for modifying intact peptides are useful but can be unselective with regard to amino acid position and sequence context. In this work, we used in vitro selection to identify DNAzymes that acylate a Lys residue of a short peptide in sequence-dependent fashion. The DNAzymes do not acylate Lys when placed at other residues in the peptide, and the acylation activity depends on the Lys sequence context. A high acylation yield is observed on the preparative nanomole scale. These findings are promising for further development of DNAzymes for broader application to top-down Lys acylation of peptide and protein substrates.
Collapse
Affiliation(s)
- Prakriti K Das
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, 61801, United States
| | - Scott K Silverman
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, 61801, United States
| |
Collapse
|
18
|
Stratton RL, Pokhrel B, Smith B, Adeyemi A, Dhakal A, Shen H. DNA Catalysis: Design, Function, and Optimization. Molecules 2024; 29:5011. [PMID: 39519652 PMCID: PMC11547689 DOI: 10.3390/molecules29215011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Catalytic DNA has gained significant attention in recent decades as a highly efficient and tunable catalyst, thanks to its flexible structures, exceptional specificity, and ease of optimization. Despite being composed of just four monomers, DNA's complex conformational intricacies enable a wide range of nuanced functions, including scaffolding, electrocatalysis, enantioselectivity, and mechano-electro spin coupling. DNA catalysts, ranging from traditional DNAzymes to innovative DNAzyme hybrids, highlight the remarkable potential of DNA in catalysis. Recent advancements in spectroscopic techniques have deepened our mechanistic understanding of catalytic DNA, paving the way for rational structural optimization. This review will summarize the latest studies on the performance and optimization of traditional DNAzymes and provide an in-depth analysis of DNAzyme hybrid catalysts and their unique and promising properties.
Collapse
Affiliation(s)
- Rebecca L. Stratton
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
| | - Bishal Pokhrel
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
| | - Bryce Smith
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
| | - Adeola Adeyemi
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
| | - Ananta Dhakal
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
| | - Hao Shen
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA; (R.L.S.); (B.P.); (B.S.); (A.A.)
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
19
|
Nurmi C, Gu J, Mathai A, Brennan J, Li Y. Making target sites in large structured RNAs accessible to RNA-cleaving DNAzymes through hybridization with synthetic DNA oligonucleotides. Nucleic Acids Res 2024; 52:11177-11187. [PMID: 39248110 PMCID: PMC11472044 DOI: 10.1093/nar/gkae778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
The 10-23 DNAzyme is one of the most active DNA-based enzymes, and in theory, can be designed to target any purine-pyrimidine junction within an RNA sequence for cleavage. However, purine-pyrimidine junctions within a large, structured RNA (lsRNA) molecule of biological origin are not always accessible to 10-23, negating its general utility as an RNA-cutting molecular scissor. Herein, we report a generalizable strategy that allows 10-23 to access any purine-pyrimidine junction within an lsRNA. Using three large SARS-CoV-2 mRNA sequences of 566, 584 and 831 nucleotides in length as model systems, we show that the use of antisense DNA oligonucleotides (ASOs) that target the upstream and downstream regions flanking the cleavage site can restore the activity (kobs) of previously poorly active 10-23 DNAzyme systems by up to 2000-fold. We corroborated these findings mechanistically using in-line probing to demonstrate that ASOs reduced 10-23 DNAzyme target site structure within the lsRNA substrates. This approach represents a simple, efficient, cost-effective, and generalizable way to improve the accessibility of 10-23 to a chosen target site within an lsRNA molecule, especially where direct access to the genomic RNA target is necessary.
Collapse
MESH Headings
- DNA, Catalytic/chemistry
- DNA, Catalytic/metabolism
- SARS-CoV-2/genetics
- RNA, Viral/chemistry
- RNA, Viral/metabolism
- RNA, Viral/genetics
- Nucleic Acid Hybridization
- Oligonucleotides, Antisense/chemistry
- Nucleic Acid Conformation
- RNA Cleavage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/chemistry
- COVID-19/virology
- RNA/chemistry
- RNA/metabolism
- DNA, Single-Stranded
Collapse
Affiliation(s)
- Connor Nurmi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario L8S 4L8, Canada
- Biointerfaces Institute, McMaster University, Ontario L8S 4L8, Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario L8S 4L8, Canada
| | - Amal Mathai
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario L8S 4L8, Canada
- Biointerfaces Institute, McMaster University, Ontario L8S 4L8, Canada
| | - John D Brennan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario L8S 4L8, Canada
- Biointerfaces Institute, McMaster University, Ontario L8S 4L8, Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario L8S 4L8, Canada
| |
Collapse
|
20
|
Chen K, Zhu L, Li J, Zhang Y, Yu Y, Wang X, Wei W, Huang K, Xu W. High-content tailoring strategy to improve the multifunctionality of functional nucleic acids. Biosens Bioelectron 2024; 261:116494. [PMID: 38901394 DOI: 10.1016/j.bios.2024.116494] [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: 05/08/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Functional nucleic acids (FNAs) have attracted increasing attention in recent years due to their diverse physiological functions. The understanding of their conformational recognition mechanisms has advanced through nucleic acid tailoring strategies and sequence optimization. With the development of the FNA tailoring techniques, they have become a methodological guide for nucleic acid repurposing. Therefore, it is necessary to systematize the relationship between FNA tailoring strategies and the development of nucleic acid multifunctionality. This review systematically categorizes eight types of FNA multifunctionality, and introduces the traditional FNA tailoring strategy from five aspects, including deletion, substitution, splitting, fusion and elongation. Based on the current state of FNA modification, a new generation of FNA tailoring strategy, called the high-content tailoring strategy, was unprecedentedly proposed to improve FNA multifunctionality. In addition, the multiple applications of rational tailoring-driven FNA performance enhancement in various fields were comprehensively summarized. The limitations and potential of FNA tailoring and repurposing in the future are also explored in this review. In summary, this review introduces a novel tailoring theory, systematically summarizes eight FNA performance enhancements, and provides a systematic overview of tailoring applications across all categories of FNAs. The high-content tailoring strategy is expected to expand the application scenarios of FNAs in biosensing, biomedicine and materials science, thus promoting the synergistic development of various fields.
Collapse
Affiliation(s)
- Keren Chen
- Food Laboratory of Zhongyuan, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Longjiao Zhu
- Food Laboratory of Zhongyuan, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Jie Li
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yangzi Zhang
- Food Laboratory of Zhongyuan, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Yongxia Yu
- Food Laboratory of Zhongyuan, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Xiaofu Wang
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Wei Wei
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Kunlun Huang
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Wentao Xu
- Food Laboratory of Zhongyuan, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
21
|
He Y, Wang Q, Zhang Q, Wang Y, Jiang Y, Zhao Q, Liu X, Wang F. A Methyl-Engineered DNAzyme for Endogenous Alkyltransferase Monitoring and Self-Sufficient Gene Regulation. SMALL METHODS 2024:e2401160. [PMID: 39295467 DOI: 10.1002/smtd.202401160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/30/2024] [Indexed: 09/21/2024]
Abstract
The on-demand gene regulation is crucial for extensively exploring specific gene functions and developing personalized gene therapeutics, which shows great promise in precision medicines. Although some nucleic acid-based gene regulatory tools (antisense oligonucleotides and small interfering RNAs) are devised for achieving on-demand activation, the introduction of chemical modifications may cause undesired side effects, thereby impairing the gene regulatory efficacy. Herein, a methyl-engineered DNAzyme (MeDz) is developed for the visualization of endogenous alkyltransferase (AGT) and the simultaneous self-sufficiently on-demand gene regulation. The catalytic activity of DNAzyme can be efficiently blocked by O6-methylguanine (O6MeG) modification and specifically restored via the AGT-mediated DNA-repairing pathway. This simply designed MeDz is demonstrated to reveal AGT of varying expression levels in different cells, opening the possibility to explore the AGT-related biological processes. Moreover, the AGT-guided MeDz exhibits cell-selective regulation on the human early growth response-1 (EGR-1) gene, with efficient gene repression in breast cancer cells and low effectiveness in normal cells. The proposed MeDz offers an attractive strategy for on-demand gene regulation, displaying great potential in biomedical applications.
Collapse
Affiliation(s)
- Yuqiu He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qing Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qingqing Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yifei Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yuqian Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| |
Collapse
|
22
|
Wang J, Chang Y, Liu M. Proximity-Dependent Activation of Split DNAzyme Kinase. Chembiochem 2024; 25:e202400368. [PMID: 38954271 DOI: 10.1002/cbic.202400368] [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: 04/21/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
Binary (also known as split) nucleic acid enzymes have emerged as novel tools in biosensors. We report a new split strategy to split the DNAzyme kinase into two independent and non-functional fragments, denoted Dk1sub and Dk1enz. In the presence of the specific target, their free ends are brought sufficiently close to interact with each other without the formation of Watson-Crick base pairings between Dk1sub and Dk1enz, thus allowing the DNA phosphorylation reaction. We term this approach proximity-dependent activation of split DNAzyme kinase (ProxSDK). The utility of ProxSDK is demonstrated by engineering a biosensing system that is capable of measuring specific DNA-protein interactions. We envision that the approach described herein will find useful applications in biosensing, imaging, and clinical diagnosis.
Collapse
Affiliation(s)
- Jiayi Wang
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian POCT Laboratory, Dalian University of Technology, Dalian, 116024, China
| | - Yangyang Chang
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian POCT Laboratory, Dalian University of Technology, Dalian, 116024, China
| | - Meng Liu
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian POCT Laboratory, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
23
|
Boyd R, Kennebeck M, Miranda A, Liu Z, Silverman S. Site-specific N-alkylation of DNA oligonucleotide nucleobases by DNAzyme-catalyzed reductive amination. Nucleic Acids Res 2024; 52:8702-8716. [PMID: 39051544 PMCID: PMC11347174 DOI: 10.1093/nar/gkae639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
DNA and RNA nucleobase modifications are biologically relevant and valuable in fundamental biochemical and biophysical investigations of nucleic acids. However, directly introducing site-specific nucleobase modifications into long unprotected oligonucleotides is a substantial challenge. In this study, we used in vitro selection to identify DNAzymes that site-specifically N-alkylate the exocyclic nucleobase amines of particular cytidine, guanosine, and adenosine (C, G and A) nucleotides in DNA substrates, by reductive amination using a 5'-benzaldehyde oligonucleotide as the reaction partner. The new DNAzymes each require one or more of Mg2+, Mn2+, and Zn2+ as metal ion cofactors and have kobs from 0.04 to 0.3 h-1, with rate enhancement as high as ∼104 above the splinted background reaction. Several of the new DNAzymes are catalytically active when an RNA substrate is provided in place of DNA. Similarly, several new DNAzymes function when a small-molecule benzaldehyde compound replaces the 5'-benzaldehyde oligonucleotide. These findings expand the scope of DNAzyme catalysis to include nucleobase N-alkylation by reductive amination. Further development of this new class of DNAzymes is anticipated to facilitate practical covalent modification and labeling of DNA and RNA substrates.
Collapse
Affiliation(s)
- Robert D Boyd
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| | - Morgan M Kennebeck
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| | - Aurora A Miranda
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| | - Zehui Liu
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| | - Scott K Silverman
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL 61801, USA
| |
Collapse
|
24
|
Parra-Meneses V, Silva-Galleguillos V, Cepeda-Plaza M. Exploring the catalytic mechanism of the 10-23 DNAzyme: insights from pH-rate profiles. Org Biomol Chem 2024; 22:6833-6840. [PMID: 39115293 DOI: 10.1039/d4ob01125b] [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: 08/23/2024]
Abstract
The 10-23 DNAzyme, a catalytic DNA molecule with RNA-cleaving activity, has garnered significant interest for its potential therapeutic applications as a gene-silencing agent. However, the lack of a detailed understanding about its mechanism has hampered progress. A recent structural analysis has revealed a highly organized conformation thanks to the stabilization of specific interactions within the catalytic core of the 10-23 DNAzyme, which facilitate the cleavage of RNA. In this configuration, it has been shown that G14 is in good proximity to the cleavage site which suggests its role as a general base, by activating the 2'-OH nucleophile, in the catalysis of the 10-23 DNAzyme. Also, the possibility of a hydrated metal acting as a general acid has been proposed. In this study, through activity assays, we offer evidence of the involvement of general acid-base catalysis in the mechanism of the 10-23 DNAzyme by analyzing its pH-rate profiles and the role of G14, and metal cofactors like Mg2+ and Pb2+. By substituting G14 with its analogue 2-aminopurine and examining the resultant pH-rate profiles, we propose the participation of G14 in a catalytically relevant proton transfer event, acting as a general base. Further analysis, using Pb2+ as a cofactor, suggests the capability of the hydrated metal ion to act as a general acid. These functional results provide critical insights into the catalytic strategies of RNA-cleaving DNAzymes, revealing common mechanisms among nucleic acid enzymes that cleave RNA.
Collapse
|
25
|
He A, Wan L, Zhang Y, Yan Z, Guo P, Han D, Tan W. Structure-based investigation of a DNA aptamer targeting PTK7 reveals an intricate 3D fold guiding functional optimization. Proc Natl Acad Sci U S A 2024; 121:e2404060121. [PMID: 38985770 PMCID: PMC11260122 DOI: 10.1073/pnas.2404060121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
DNA aptamers have emerged as novel molecular tools in disease theranostics owing to their high binding affinity and specificity for protein targets, which rely on their ability to fold into distinctive three-dimensional (3D) structures. However, delicate atomic interactions that shape the 3D structures are often ignored when designing and modeling aptamers, leading to inefficient functional optimization. Challenges persist in determining high-resolution aptamer-protein complex structures. Moreover, the experimentally determined 3D structures of DNA molecules with exquisite functions remain scarce. These factors impede our comprehension and optimization of some important DNA aptamers. Here, we performed a streamlined solution NMR-based structural investigation on the 41-nt sgc8c, a prominent DNA aptamer used to target membrane protein tyrosine kinase 7, for cancer theranostics. We show that sgc8c prefolds into an intricate three-way junction (3WJ) structure stabilized by long-range tertiary interactions and extensive base-base stackings. Delineated by NMR chemical shift perturbations, site-directed mutagenesis, and 3D structural information, we identified essential nucleotides constituting the key functional elements of sgc8c that are centralized at the core of 3WJ. Leveraging the well-established structure-function relationship, we efficiently engineered two sgc8c variants by modifying the apical loop and introducing L-DNA base pairs to simultaneously enhance thermostability, biostability, and binding affinity for both protein and cell targets, a feat not previously attained despite extensive efforts. This work showcases a simplified NMR-based approach to comprehend and optimize sgc8c without acquiring the complex structure, and offers principles for the sophisticated structure-function organization of DNA molecules.
Collapse
Affiliation(s)
- Axin He
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Liqi Wan
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Yuchao Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Zhenzhen Yan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Pei Guo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Da Han
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| | - Weihong Tan
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang310022, China
| |
Collapse
|
26
|
Xia L, Du L, Hou X, Zhou R, Cheng N, Chen J. Protein-Controlled Split DNAzyme to Enhance Catalytic Activity: Design and Performance. Anal Chem 2024. [PMID: 39010288 DOI: 10.1021/acs.analchem.3c05909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
In this study, we utilized proteins to control the assembly of split DNAzyme to establish protein-controlled split DNAzymes (Pc SD), with the aim of enhancing its catalytic activity. To achieve this, simultaneous recognition of protein by affinity ligands at both ends of split DNAzyme fragments induced an increased local concentration of each split fragment, leading to reassembly of the split catalytic core with a rigid conformation and higher affinity to its cofactor. As a result, under protein control, Pc SD exhibits unexpected cleavage efficiency compared to free split DNAzyme. To further explore the catalytic features, we then systematically positioned split sites within the catalytic core of three popular DNAzyme-based Pc SDs, thus revealing the important nucleic acids that influence Pc SDs activity. Based on the excellent analytical performance of Pc SD for streptavidin (with a LOD of 0.1 pM in buffer),we equipped Pc SD with antibodies as rapid diagnostic tools for inpatient care (AFP as biomarker) with a minimized workflow (with a LOD of 2 pM in 5% human serum). The results of this study offer fundamental insights into external factors for boosting DNAzyme catalysis and will be promising for applications that utilize split DNAzymes.
Collapse
Affiliation(s)
- Lingying Xia
- Analytical & Testing Center, Sichuan University, Sichuan, Chengdu 610064, PR China
- Biliary Surgical Department of West China Hospital, Sichuan University, Sichuan, Chengdu 610064, PR China
| | - Lijie Du
- Analytical & Testing Center, Sichuan University, Sichuan, Chengdu 610064, PR China
| | - Xiandeng Hou
- Analytical & Testing Center, Sichuan University, Sichuan, Chengdu 610064, PR China
| | - Rongxing Zhou
- Biliary Surgical Department of West China Hospital, Sichuan University, Sichuan, Chengdu 610064, PR China
| | - Nansheng Cheng
- Biliary Surgical Department of West China Hospital, Sichuan University, Sichuan, Chengdu 610064, PR China
| | - Junbo Chen
- Analytical & Testing Center, Sichuan University, Sichuan, Chengdu 610064, PR China
| |
Collapse
|
27
|
Schmuck JF, Borggräfe J, Etzkorn M. The dynamic world of the 8-17 DNAzyme. Nat Commun 2024; 15:5145. [PMID: 38886339 PMCID: PMC11183194 DOI: 10.1038/s41467-024-49500-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Affiliation(s)
- Jessica Felice Schmuck
- Heinrich Heine University, Faculty of Mathematics and Natural Sciences, Institute of Physical Biology, Düsseldorf, Germany
| | - Jan Borggräfe
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764, München, Germany.
- Bavarian NMR Center, School of Natural Sciences, Technical University of Munich Garching, 85748, München, Germany.
| | - Manuel Etzkorn
- Heinrich Heine University, Faculty of Mathematics and Natural Sciences, Institute of Physical Biology, Düsseldorf, Germany.
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
28
|
Wieruszewska J, Pawłowicz A, Połomska E, Pasternak K, Gdaniec Z, Andrałojć W. The 8-17 DNAzyme can operate in a single active structure regardless of metal ion cofactor. Nat Commun 2024; 15:4218. [PMID: 38760331 PMCID: PMC11101458 DOI: 10.1038/s41467-024-48638-x] [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: 04/13/2023] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
DNAzymes - synthetic enzymes made of DNA - have long attracted attention as RNA-targeting therapeutic agents. Yet, as of now, no DNAzyme-based drug has been approved, partially due to our lacking understanding of their molecular mode of action. In this work we report the solution structure of 8-17 DNAzyme bound to a Zn2+ ion solved through NMR spectroscopy. Surprisingly, it turned out to be very similar to the previously solved Pb2+-bound form (catalytic domain RMSD = 1.28 Å), despite a long-standing literature consensus that Pb2+ recruits a different DNAzyme fold than other metal ion cofactors. Our follow-up NMR investigations in the presence of other ions - Mg2+, Na+, and Pb2+ - suggest that at DNAzyme concentrations used in NMR all these ions induce a similar tertiary fold. Based on these findings, we propose a model for 8-17 DNAzyme interactions with metal ions postulating the existence of only a single catalytically-active structure, yet populated to a different extent depending on the metal ion cofactor. Our results provide structural information on the 8-17 DNAzyme in presence of non-Pb2+ cofactors, including the biologically relevant Mg2+ ion.
Collapse
Affiliation(s)
- Julia Wieruszewska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland
| | - Aleksandra Pawłowicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland
| | - Ewa Połomska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland
| | - Karol Pasternak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland
| | - Zofia Gdaniec
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland
| | - Witold Andrałojć
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704, Poznań, Noskowskiego, 12/14, Poland.
| |
Collapse
|
29
|
Beriashvili D, Zhou J, Liu Y, Folkers GE, Baldus M. Cellular Applications of DNP Solid-State NMR - State of the Art and a Look to the Future. Chemistry 2024; 30:e202400323. [PMID: 38451060 DOI: 10.1002/chem.202400323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Sensitivity enhanced dynamic nuclear polarization solid-state NMR is emerging as a powerful technique for probing the structural properties of conformationally homogenous and heterogenous biomolecular species irrespective of size at atomic resolution within their native environments. Herein we detail advancements that have made acquiring such data, specifically within the confines of intact bacterial and eukaryotic cell a reality and further discuss the type of structural information that can presently be garnered by the technique's exploitation. Subsequently, we discuss bottlenecks that have thus far curbed cellular DNP-ssNMR's broader adoption namely due a lack of sensitivity and spectral resolution. We also explore possible solutions ranging from utilization of new pulse sequences, design of better performing polarizing agents, and application of additional biochemical/ cell biological methodologies.
Collapse
Affiliation(s)
- David Beriashvili
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padaulaan 8, 3584 CH, Utrecht, The Netherlands
| | - Jiaxin Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics, Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics, Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Gert E Folkers
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padaulaan 8, 3584 CH, Utrecht, The Netherlands
| | - Marc Baldus
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padaulaan 8, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
30
|
Yan J, Bhadane R, Ran M, Ma X, Li Y, Zheng D, Salo-Ahen OMH, Zhang H. Development of Aptamer-DNAzyme based metal-nucleic acid frameworks for gastric cancer therapy. Nat Commun 2024; 15:3684. [PMID: 38693181 PMCID: PMC11063048 DOI: 10.1038/s41467-024-48149-9] [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: 08/22/2023] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
The metal-nucleic acid nanocomposites, first termed metal-nucleic acid frameworks (MNFs) in this work, show extraordinary potential as functional nanomaterials. However, thus far, realized MNFs face limitations including harsh synthesis conditions, instability, and non-targeting. Herein, we discover that longer oligonucleotides can enhance the synthesis efficiency and stability of MNFs by increasing oligonucleotide folding and entanglement probabilities during the reaction. Besides, longer oligonucleotides provide upgraded metal ions binding conditions, facilitating MNFs to load macromolecular protein drugs at room temperature. Furthermore, longer oligonucleotides facilitate functional expansion of nucleotide sequences, enabling disease-targeted MNFs. As a proof-of-concept, we build an interferon regulatory factor-1(IRF-1) loaded Ca2+/(aptamer-deoxyribozyme) MNF to target regulate glucose transporter (GLUT-1) expression in human epidermal growth factor receptor-2 (HER-2) positive gastric cancer cells. This MNF nanodevice disrupts GSH/ROS homeostasis, suppresses DNA repair, and augments ROS-mediated DNA damage therapy, with tumor inhibition rate up to 90%. Our work signifies a significant advancement towards an era of universal MNF application.
Collapse
Affiliation(s)
- Jiaqi Yan
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Rajendra Bhadane
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520, Turku, Finland
| | - Meixin Ran
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Xiaodong Ma
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Yuanqiang Li
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Dongdong Zheng
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Outi M H Salo-Ahen
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520, Turku, Finland
| | - Hongbo Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
31
|
Dantu SC, Khalil M, Bria M, Saint-Pierre C, Orio M, Gasparutto D, Sicoli G. Cleaving DNA with DNA: Cooperative Tuning of Structure and Reactivity Driven by Copper Ions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306710. [PMID: 38419268 DOI: 10.1002/advs.202306710] [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: 09/18/2023] [Revised: 01/15/2024] [Indexed: 03/02/2024]
Abstract
A copper-dependent self-cleaving DNA (DNAzyme or deoyxyribozyme) previously isolated by in vitro selection has been analyzed by a combination of Molecular Dynamics (MD) simulations and advanced Electron Paramagnetic Resonance (Electron Spin Resonance) EPR/ESR spectroscopy, providing insights on the structural and mechanistic features of the cleavage reaction. The modeled 46-nucleotide deoxyribozyme in MD simulations forms duplex and triplex sub-structures that flank a highly conserved catalytic core. The DNA self-cleaving construct can also form a bimolecular complex that has a distinct substrate and enzyme domains. The highly dynamic structure combined with an oxidative site-specific cleavage of the substrate are two key-aspects to elucidate. By combining EPR/ESR spectroscopy with selectively isotopically labeled nucleotides it has been possible to overcome the major drawback related to the "metal-soup" scenario, also known as "super-stoichiometric" ratios of cofactors versus substrate, conventionally required for the DNA cleavage reaction within those nucleic acids-based enzymes. The focus on the endogenous paramagnetic center (Cu2+) here described paves the way for analysis on mixtures where several different cofactors are involved. Furthermore, the insertion of cleavage reaction within more complex architectures is now a realistic perspective towards the applicability of EPR/ESR spectroscopic studies.
Collapse
Affiliation(s)
- Sarath Chandra Dantu
- Department of Computer Science, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Mahdi Khalil
- LASIRE, CNRS UMR 8516, University of Lille, C4 building, Avenue Paul Langevin, Villeneuve d'Ascq, F-59655, France
| | - Marc Bria
- Michle-Eugène Chevreul Institute, FR 2638, Avenue Paul Langevin, Villeneuve d'Ascq, F-59655, France
| | - Christine Saint-Pierre
- Université Grenoble Alpes, CEA, CNRS, UMR 5819 SyMMES-CREAB, Avenue des Martyrs, Grenoble, F-38000, France
| | - Maylis Orio
- Aix Marseille Université, CNRS, Centrale Marseille, iSm2, UMR CNRS 7313, Marseille, 13397, France
| | - Didier Gasparutto
- Université Grenoble Alpes, CEA, CNRS, UMR 5819 SyMMES-CREAB, Avenue des Martyrs, Grenoble, F-38000, France
| | - Giuseppe Sicoli
- LASIRE, CNRS UMR 8516, University of Lille, C4 building, Avenue Paul Langevin, Villeneuve d'Ascq, F-59655, France
| |
Collapse
|
32
|
Shi C, Yang D, Ma X, Pan L, Shao Y, Arya G, Ke Y, Zhang C, Wang F, Zuo X, Li M, Wang P. A Programmable DNAzyme for the Sensitive Detection of Nucleic Acids. Angew Chem Int Ed Engl 2024; 63:e202320179. [PMID: 38288561 DOI: 10.1002/anie.202320179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Indexed: 02/17/2024]
Abstract
Nucleic acids in biofluids are emerging biomarkers for the molecular diagnostics of diseases, but their clinical use has been hindered by the lack of sensitive detection assays. Herein, we report the development of a sensitive nucleic acid detection assay named SPOT (sensitive loop-initiated DNAzyme biosensor for nucleic acid detection) by rationally designing a catalytic DNAzyme of endonuclease capability into a unified one-stranded allosteric biosensor. SPOT is activated once a nucleic acid target of a specific sequence binds to its allosteric module to enable continuous cleavage of molecular reporters. SPOT provides a highly robust platform for sensitive, convenient and cost-effective detection of low-abundance nucleic acids. For clinical validation, we demonstrated that SPOT could detect serum miRNAs for the diagnostics of breast cancer, gastric cancer and prostate cancer. Furthermore, SPOT exhibits potent detection performance over SARS-CoV-2 RNA from clinical swabs with high sensitivity and specificity. Finally, SPOT is compatible with point-of-care testing modalities such as lateral flow assays. Hence, we envision that SPOT may serve as a robust assay for the sensitive detection of a variety of nucleic acid targets enabling molecular diagnostics in clinics.
Collapse
Affiliation(s)
- Chenzhi Shi
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Donglei Yang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaowei Ma
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Li Pan
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuanchuan Shao
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708, USA
| | - Gaurav Arya
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, 27708, USA
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, 30322, USA
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Li
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Pengfei Wang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Center for DNA Information Storage, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
33
|
Zhang Y, Zhang J, Wan H, Wu Z, Xu H, Zhang Z, Wang Y, Wang J. New Insights into the Dependence of CPEB3 Ribozyme Cleavage on Mn 2+ and Mg 2. J Phys Chem Lett 2024; 15:2708-2714. [PMID: 38427973 DOI: 10.1021/acs.jpclett.3c03221] [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: 03/03/2024]
Abstract
CPEB3 ribozyme is a self-cleaving RNA that occurs naturally in mammals and requires divalent metal ions for efficient activity. Ribozymes exhibit preferences for specific metal ions, but the exact differences in the catalytic mechanisms of various metal ions on the CPEB3 ribozyme remain unclear. Our findings reveal that Mn2+ functions as a more effective cofactor for CPEB3 ribozyme catalysis compared to Mg2+, as confirmed by its stronger binding affinity to CPEB3 by EPR. Cleavage assays of CPEB3 mutants and molecular docking analyses further showed that excessive Mn2+ ions can bind to a second binding site near the catalytic site, hindering CPEB3 catalytic efficiency and contributing to the Mn2+ bell-shaped curve. These results implicate a pivotal role for the local nucleobase-Mn2+ interactions in facilitating RNA folding and modulating the directed attack of nucleophilic reagents. Our study provides new insights and experimental evidence for exploring the divalent cation dependent cleavage mechanism of the CPEB3 ribozyme.
Collapse
Affiliation(s)
- Yaoyao Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jing Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hengjia Wan
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ziwei Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Huangtao Xu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Zhe Zhang
- Institute for Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Yujuan Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- International Magnetobiology Frontier Research Center (iMFRC), Science Island, Hefei 230031, China
| | - Junfeng Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
- International Magnetobiology Frontier Research Center (iMFRC), Science Island, Hefei 230031, China
| |
Collapse
|
34
|
Zhang Z, Wei W, Chen S, Yang J, Song D, Chen Y, Zhao Z, Chen J, Wang F, Wang J, Li Z, Liang Y, Yu H. Chemoenzymatic Installation of Site-Specific Chemical Groups on DNA Enhances the Catalytic Activity. J Am Chem Soc 2024; 146:7052-7062. [PMID: 38427585 DOI: 10.1021/jacs.4c00484] [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: 03/03/2024]
Abstract
Functional DNAs are valuable molecular tools in chemical biology and analytical chemistry but suffer from low activities due to their limited chemical functionalities. Here, we present a chemoenzymatic method for site-specific installation of diverse functional groups on DNA, and showcase the application of this method to enhance the catalytic activity of a DNA catalyst. Through chemoenzymatic introduction of distinct chemical groups, such as hydroxyl, carboxyl, and benzyl, at specific positions, we achieve significant enhancements in the catalytic activity of the RNA-cleaving deoxyribozyme 10-23. A single carboxyl modification results in a 100-fold increase, while dual modifications (carboxyl and benzyl) yield an approximately 700-fold increase in activity when an RNA cleavage reaction is catalyzed on a DNA-RNA chimeric substrate. The resulting dually modified DNA catalyst, CaBn, exhibits a kobs of 3.76 min-1 in the presence of 1 mM Mg2+ and can be employed for fluorescent imaging of intracellular magnesium ions. Molecular dynamics simulations reveal the superior capability of CaBn to recruit magnesium ions to metal-ion-binding site 2 and adopt a catalytically competent conformation. Our work provides a broadly accessible strategy for DNA functionalization with diverse chemical modifications, and CaBn offers a highly active DNA catalyst with immense potential in chemistry and biotechnology.
Collapse
Affiliation(s)
- Ze Zhang
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Wanqing Wei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Siqi Chen
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jintao Yang
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yinghan Chen
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zerun Zhao
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Jiawen Chen
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Fulong Wang
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jiahuan Wang
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Zhe Li
- State Key Laboratory of Analytical Chemistry for Life Science, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Hanyang Yu
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| |
Collapse
|
35
|
Li X, Chang Y, Wu Y, Liu M. A DNAzymes-in-droplets assay for Burkholderia gladioli pathovar cocovenenans with single-bacterium sensitivity. Chem Sci 2024; 15:2996-3002. [PMID: 38404397 PMCID: PMC10882462 DOI: 10.1039/d3sc05874c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024] Open
Abstract
Foodborne pathogens pose a serious risk to human health, and the simple and rapid detection of such bacteria in complex food matrices remains challenging. Herein, we present the selection and characterization of a novel RNA-cleaving fluorogenic DNAzyme, named RFD-BC1, with exceptional specificity for Burkholderia gladioli pv. cocovenenans (B. cocovenenans), a pathogen strongly associated with fatal food poisoning cases. RFD-BC1 was activated by a protein secreted specifically by whole viable B. cocovenenans and displayed an optimum pH distinct from the selection pH, with a rate constant of approximately 0.01 min-1 at pH 5.0. Leveraging this newly discovered DNAzyme, we developed a novel system, termed DNAzymes-in-droplets (DID), that integrates droplet microfluidics to achieve the rapid and selective detection of live B. cocovenenans with single-cell sensitivity. We believe that the approach described herein holds promise for combating specific bacterial pathogens in food samples, offering significant potential for broader applications in food safety and public health.
Collapse
Affiliation(s)
- Xiaoqian Li
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian POCT Laboratory Dalian 116024 China
| | - Yangyang Chang
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian POCT Laboratory Dalian 116024 China
| | - Yunping Wu
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian POCT Laboratory Dalian 116024 China
| | - Meng Liu
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian POCT Laboratory Dalian 116024 China
| |
Collapse
|
36
|
Cieślak M, Karwowski BT. The Effect of 8,5'-Cyclo 2'-deoxyadenosine on the Activity of 10-23 DNAzyme: Experimental and Theoretical Study. Int J Mol Sci 2024; 25:2519. [PMID: 38473767 DOI: 10.3390/ijms25052519] [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: 01/24/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The in vivo effectiveness of DNAzymes 10-23 (Dz10-23) is limited due to the low concentration of divalent cations. Modifications of the catalytic loop are being sought to increase the activity of Dz10-23 in physiological conditions. We investigated the effect of 5'S or 5'R 5',8-cyclo-2'deoxyadenosine (cdA) on the activity of Dz10-23. The activity of Dz10-23 was measured in a cleavage assay using radiolabeled RNA. The Density Functional Tight Binding methodology with the self-consistent redistribution of Mulliken charge modification was used to explain different activities of DNAzymes. The substitution of 2'-deoxyadenosine with cdA in the catalytic loop decreased the activity of DNAzymes. Inhibition was dependent on the position of cdA and its absolute configuration. The order of activity of DNAzymes was as follows: wt-Dz > ScdA5-Dz ≈ RcdA15-Dz ≈ ScdA15-Dz > RcdA5-Dz. Theoretical studies revealed that the distance between phosphate groups at position 5 in RcdA5-Dz was significantly increased compared to wt-Dz, while the distance between O4 of dT4 and nonbonding oxygen of PO2 attached to 3'O of dG2 was much shorter. The strong inhibitory effect of RcdA5 may result from hampering the flexibility of the catalytic loop (increased rigidity), which is required for the proper positioning of Me2+ and optimal activity.
Collapse
Affiliation(s)
- Marcin Cieślak
- Food Science Department, Faculty of Pharmacy, Medical University of Lodz, ul. Muszynskiego 1, 90-151 Lodz, Poland
| | - Bolesław T Karwowski
- Food Science Department, Faculty of Pharmacy, Medical University of Lodz, ul. Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
37
|
Kennebeck MM, Kaminsky CK, Massa MA, Das PK, Boyd RD, Bishka M, Tricarico JT, Silverman SK. DNAzyme-Catalyzed Site-Specific N-Acylation of DNA Oligonucleotide Nucleobases. Angew Chem Int Ed Engl 2024; 63:e202317565. [PMID: 38157448 PMCID: PMC10873475 DOI: 10.1002/anie.202317565] [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: 11/17/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
We used in vitro selection to identify DNAzymes that acylate the exocyclic nucleobase amines of cytidine, guanosine, and adenosine in DNA oligonucleotides. The acyl donor was the 2,3,5,6-tetrafluorophenyl ester (TFPE) of a 5'-carboxyl oligonucleotide. Yields are as high as >95 % in 6 h. Several of the N-acylation DNAzymes are catalytically active with RNA rather than DNA oligonucleotide substrates, and eight of nine DNAzymes for modifying C are site-specific (>95 %) for one particular substrate nucleotide. These findings expand the catalytic ability of DNA to include site-specific N-acylation of oligonucleotide nucleobases. Future efforts will investigate the DNA and RNA substrate sequence generality of DNAzymes for oligonucleotide nucleobase N-acylation, toward a universal approach for site-specific oligonucleotide modification.
Collapse
Affiliation(s)
- Morgan M Kennebeck
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Caroline K Kaminsky
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Maria A Massa
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Prakriti K Das
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Robert D Boyd
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Michelle Bishka
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - J Tomas Tricarico
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| | - Scott K Silverman
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL-61801, USA
| |
Collapse
|
38
|
Wang X, Yu B, Sakurabayashi S, Paz-Villatoro JM, Iwahara J. Robust Enzymatic Production of DNA G-Quadruplex, Aptamer, DNAzyme, and Other Oligonucleotides: Applications for NMR. J Am Chem Soc 2024; 146:1748-1752. [PMID: 38191993 PMCID: PMC10926321 DOI: 10.1021/jacs.3c11219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Single-stranded DNA (ssDNA) oligonucleotides are widely used in biological research, therapeutics, biotechnology, and nanomachines. Large-scale enzymatic production of ssDNA oligonucleotides forming noncanonical structures has been difficult. Here, we present a simple and robust method named "palindrome-nicking-dependent amplification" (PaNDA) for enzymatic production of a large amount of ssDNA oligonucleotides. It utilizes a strand-displacing DNA polymerase and a nicking enzyme together with input DNA and deoxynucleotide triphosphates at 55 °C. Scaling up of PaNDA is straightforward due to its isothermal nature. The ssDNA products can easily be isolated through anion-exchange chromatography under nondenaturing conditions. We demonstrate applications of PaNDA to 13C/15N-labeling of various DNA strands, including a 22-nt telomere repeat G-quadruplex, a 26-nt therapeutic aptamer, and a 33-nt DNAzyme. The 13C/15N-labeling by PaNDA greatly facilitates the characterization of noncanonical DNA by nuclear magnetic resonance (NMR) spectroscopy. For example, the behavior of therapeutic DNA aptamers in human serum can be investigated.
Collapse
Affiliation(s)
- Xi Wang
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, United States
| | - Binhan Yu
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, United States
| | - Shuhei Sakurabayashi
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, United States
| | - Jonathan M Paz-Villatoro
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, United States
| | - Junji Iwahara
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, United States
| |
Collapse
|
39
|
Liu J, Cui L, Shi X, Yan J, Wang Y, Ni Y, He J, Wang X. Generation of DNAzyme in Bacterial Cells by a Bacterial Retron System. ACS Synth Biol 2024; 13:300-309. [PMID: 38171507 DOI: 10.1021/acssynbio.3c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
DNAzymes are catalytically active single-stranded DNAs in which DNAzyme 10-23 (Dz 10-23) consists of a catalytic core and a substrate-binding arm that reduces gene expression through sequence-specific mRNA cleavage. However, the in vivo application of Dz 10-23 depends on exogenous delivery, which leads to its inability to be synthesized and stabilized in vivo, thus limiting its application. As a unique reverse transcription system, the bacterial retron system can synthesize single-stranded DNA in vivo using ncRNA msr/msd as a template. The objective of this work is to reduce target gene expression using Dz 10-23 generated in vivo by the retron system. In this regard, we successfully generated Dz 10-23 by cloning the Dz 10-23 coding sequence into the retron msd gene and tested its ability to reduce specific gene expression by examining the mRNA levels of cfp encoding cyan fluorescence protein and other functional genes such as mreB and ftsZ. We found that Dz had different repressive effects when targeting different mRNA regions, and in general, the repressive effect was stronger when targeting downstream of mRNAs. Our results also suggested that the reduction effect was due to cleavage of the substrate mRNA by Dz 10-23 rather than the antisense effect of the substrate-binding arm. Therefore, this study not only provided a retron-based method for the intracellular generation of Dz 10-23 but also demonstrated that Dz 10-23 could reduce gene expression by cleaving target mRNAs in cells. We believe that this new strategy would have great potential in the regulation of gene expression.
Collapse
Affiliation(s)
- Jie Liu
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Lina Cui
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xinyu Shi
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jiahao Yan
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yifei Wang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yuyang Ni
- College of Life Sciences, Shangrao Normal University, Shangrao 334001, PR China
| | - Jin He
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xun Wang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| |
Collapse
|
40
|
Nedorezova DD, Dubovichenko MV, Kalnin AJ, Nour MAY, Eldeeb AA, Ashmarova AI, Kurbanov GF, Kolpashchikov DM. Cleaving Folded RNA with DNAzyme Agents. Chembiochem 2024; 25:e202300637. [PMID: 37870555 DOI: 10.1002/cbic.202300637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/24/2023]
Abstract
Cleavage of biological mRNA by DNAzymes (Dz) has been proposed as a variation of oligonucleotide gene therapy (OGT). The design of Dz-based OGT agents includes computational prediction of two RNA-binding arms with low affinity (melting temperatures (Tm ) close to the reaction temperature of 37 °C) to avoid product inhibition and maintain high specificity. However, RNA cleavage might be limited by the RNA binding step especially if the RNA is folded in secondary structures. This calls for the need for two high-affinity RNA-binding arms. In this study, we optimized 10-23 Dz-based OGT agents for cleavage of three RNA targets with different folding energies under multiple turnover conditions in 2 mM Mg2+ at 37 °C. Unexpectedly, one optimized Dz had each RNA-binding arm with a Tm ≥60 °C, without suffering from product inhibition or low selectivity. This phenomenon was explained by the folding of the RNA cleavage products into stable secondary structures. This result suggests that Dz with long (high affinity) RNA-binding arms should not be excluded from the candidate pool for OGT agents. Rather, analysis of the cleavage products' folding should be included in Dz selection algorithms. The Dz optimization workflow should include testing with folded rather than linear RNA substrates.
Collapse
Affiliation(s)
- Daria D Nedorezova
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Mikhail V Dubovichenko
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Arseniy J Kalnin
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Moustapha A Y Nour
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Ahmed A Eldeeb
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Anna I Ashmarova
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Gabdulla F Kurbanov
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
| | - Dmitry M Kolpashchikov
- Laboratory of molecular robotics and biosensor systems, Laboratory of Frontier nucleic acid technologies in gene therapy of cancer, SCAMT Institute, ITMO University, St. Petersburg, 191002, Russian Federation
- Chemistry Department, University of Central Florida, Orlando, FL 32816-2366, USA
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
41
|
Lu Y, Yu Y, Wang Y, Zhou W, Cheng Z, Yu L, Zheng S, Gao R. A micro-nano interface integrated SERS-based microfluidic sensor for miRNA detection using DNAzyme walker amplification. Anal Chim Acta 2023; 1283:341957. [PMID: 37977782 DOI: 10.1016/j.aca.2023.341957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Precise and specific miRNA detection plays a vital role in exploring development mechanisms of cancer disease, thereby it can significantly improve relevant prevention and treatment strategies. RESULTS In this work, a surface-enhanced Raman spectroscopy (SERS)-based microfluidic chip has been devised with a microcone array SERS substrate (MCASS) for the miR-141 detection. This substrate excels in unique SERS activity and large surface area for DNA oligonucleotide modification. As the presence of miR-141, the DNAzyme walker induced cleavage reaction took place on the finely designed and prepared dual DNA conjugated SERS nanoprobes. The SERS nanoprobes can anchor on MCASS by the DNA hybridization that achieved an impressive detection limit in the femtomolar level. SIGNIFICANCE With this integrated SERS-based microfluidic chip, we provided a miRNA detection strategy using DNAzyme walker amplification technology. It is believed that this strategy could be a powerful tool for miRNA detection and related cancer screening test.
Collapse
Affiliation(s)
- Yang Lu
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yiyue Yu
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yeru Wang
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Wenbo Zhou
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Ziyi Cheng
- Hainan Cancer Medical Center of The First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, 571199, China
| | - Liandong Yu
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China.
| | - Shaojiang Zheng
- Hainan Cancer Medical Center of The First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, 571199, China.
| | - Rongke Gao
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao, 266580, China.
| |
Collapse
|
42
|
Yan J, Ma X, Liang D, Ran M, Zheng D, Chen X, Zhou S, Sun W, Shen X, Zhang H. An autocatalytic multicomponent DNAzyme nanomachine for tumor-specific photothermal therapy sensitization in pancreatic cancer. Nat Commun 2023; 14:6905. [PMID: 37903795 PMCID: PMC10616286 DOI: 10.1038/s41467-023-42740-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/20/2023] [Indexed: 11/01/2023] Open
Abstract
Multicomponent deoxyribozymes (MNAzymes) have great potential in gene therapy, but their ability to recognize disease tissue and further achieve synergistic gene regulation has rarely been studied. Herein, Arginylglycylaspartic acid (RGD)-modified Distearyl acylphosphatidyl ethanolamine (DSPE)-polyethylene glycol (PEG) (DSPE-PEG-RGD) micelle is prepared with a DSPE hydrophobic core to load the photothermal therapy (PTT) dye IR780 and the calcium efflux pump inhibitor curcumin. Then, the MNAzyme is distributed into the hydrophilic PEG layer and sealed with calcium phosphate through biomineralization. Moreover, RGD is attached to the outer tail of PEG for tumor targeting. The constructed nanomachine can release MNAzyme and the cofactor Ca2+ under acidic conditions and self-assemble into an active mode to cleave heat shock protein (HSP) mRNA by consuming the oncogene miRNA-21. Silencing miRNA-21 enhances the expression of the tumor suppressor gene PTEN, leading to PTT sensitization. Meanwhile, curcumin maintains high intracellular Ca2+ to further suppress HSP-chaperone ATP by disrupting mitochondrial Ca2+ homeostasis. Therefore, pancreatic cancer is triple-sensitized to IR780-mediated PTT. The in vitro and in vivo results show that the MNAzyme-based nanomachine can strongly regulate HSP and PTEN expression and lead to significant pancreatic tumor inhibition under laser irradiation.
Collapse
Affiliation(s)
- Jiaqi Yan
- Joint Centre of Translational Medicine, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Xiaodong Ma
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Danna Liang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Meixin Ran
- Joint Centre of Translational Medicine, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Dongdong Zheng
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Xiaodong Chen
- Joint Centre of Translational Medicine, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shichong Zhou
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Xian Shen
- Joint Centre of Translational Medicine, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Hongbo Zhang
- Joint Centre of Translational Medicine, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
43
|
Zhu X, Xu J, Ling G, Zhang P. Tunable metal-organic frameworks assist in catalyzing DNAzymes with amplification platforms for biomedical applications. Chem Soc Rev 2023; 52:7549-7578. [PMID: 37817667 DOI: 10.1039/d3cs00386h] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Various binding modes of tunable metal organic frameworks (MOFs) and functional DNAzymes (Dzs) synergistically catalyze the emergence of abundant functional nanoplatforms. Given their serial variability in formation, structural designability, and functional controllability, Dzs@MOFs tend to be excellent building blocks for the precise "intelligent" manufacture of functional materials. To present a clear outline of this new field, this review systematically summarizes the progress of Dz integration into MOFs (MOFs@Dzs) through different methods, including various surface infiltration, pore encapsulation, covalent binding, and biomimetic mineralization methods. Atomic-level and time-resolved catalytic mechanisms for biosensing and imaging are made possible by the complex interplay of the distinct molecular structure of Dzs@MOF, conformational flexibility, and dynamic regulation of metal ions. Exploiting the precision of DNAzymes, MOFs@Dzs constructed a combined nanotherapy platform to guide intracellular drug synthesis, photodynamic therapy, catalytic therapy, and immunotherapy to enhance gene therapy in different ways, solving the problems of intracellular delivery inefficiency and insufficient supply of cofactors. MOFs@Dzs nanostructures have become excellent candidates for biosensing, bioimaging, amplification delivery, and targeted cancer gene therapy while emphasizing major advancements and seminal endeavors in the fields of biosensing (nucleic acid, protein, enzyme activity, small molecules, and cancer cells), biological imaging, and targeted cancer gene delivery and gene therapy. Overall, based on the results demonstrated to date, we discuss the challenges that the emerging MOFs@Dzs might encounter in practical future applications and briefly look forward to their bright prospects in other fields.
Collapse
Affiliation(s)
- Xiaoguang Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Jiaqi Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
44
|
Chang T, Li G, Chang D, Amini R, Zhu X, Zhao T, Gu J, Li Z, Li Y. An RNA-Cleaving DNAzyme That Requires an Organic Solvent to Function. Angew Chem Int Ed Engl 2023; 62:e202310941. [PMID: 37648674 DOI: 10.1002/anie.202310941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/01/2023]
Abstract
Engineering functional nucleic acids that are active under unusual conditions will not only reveal their hidden abilities but also lay the groundwork for pursuing them for unique applications. Although many DNAzymes have been derived to catalyze diverse chemical reactions in aqueous solutions, no prior study has been set up to purposely derive DNAzymes that require an organic solvent to function. Herein, we utilized in vitro selection to isolate RNA-cleaving DNAzymes from a random-sequence DNA pool that were "compelled" to accept 35 % dimethyl sulfoxide (DMSO) as a cosolvent, via counter selection in a purely aqueous solution followed by positive selection in the same solution containing 35 % DMSO. This experiment led to the discovery of a new DNAzyme that requires 35 % DMSO for its catalytic activity and exhibits drastically reduced activity without DMSO. This DNAzyme also requires divalent metal ions for catalysis, and its activity is enhanced by monovalent ions. A minimized, more efficient DNAzyme was also derived. This work demonstrates that highly functional, organic solvent-dependent DNAzymes can be isolated from random-sequence DNA libraries via forced in vitro selection, thus expanding the capability and potential utility of catalytic DNA.
Collapse
Affiliation(s)
- Tianjun Chang
- School of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Guangping Li
- School of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Dingran Chang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | - Ryan Amini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | - Xiaoni Zhu
- School of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Tongqian Zhao
- School of Resources and Environment, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | - Zhongping Li
- Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| |
Collapse
|
45
|
Zhou Q, Zhang G, Wu Y, Zhang Q, Liu Y, Chang Y, Liu M. In Vitro Selection of M 2+-Independent, Fast-Responding Acidic Deoxyribozymes for Bacterial Detection. J Am Chem Soc 2023; 145:21370-21377. [PMID: 37683187 DOI: 10.1021/jacs.3c06155] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
We report on the first efforts to isolate acidic RNA-cleaving DNAzymes (aRCDs) from a random-sequence DNA pool by in vitro selection that are activated by a microbe Escherichia coli (E. coli), at pH 5.3. Importantly, these E. coli-responsive aRCDs only require monovalent metal ions as cofactors for cleaving a fluorogenic chimeric DNA/RNA substrate. Such characteristics can be used to efficiently protect RCDs from both intrinsic chemical instability and external enzymatic degradation. One remarkable DNAzyme, aRCD-EC1, is specific for E. coli, and its target is likely a protein. Furthermore, truncated aRCD-EC1 had significantly improved catalytic activity with an observed rate constant (kobs) of 1.18 min-1, making it the fastest bacteria-responding RCD reported to date. Clinical evaluation of this aRCD-based fluorescent assay using 40 patient urine samples demonstrated a diagnostic sensitivity of 100% and a specificity of 100% at a total analysis time of 50 min without a bacterial culture. This work can expand the repertoire of DNAzymes that are active under nonphysiological conditions, thus facilitating the development of diverse DNAzyme-based biosensors in clinical diagnosis.
Collapse
Affiliation(s)
- Qinbin Zhou
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian 116024, China
- Dalian POCT Laboratory, Dalian 116024, China
| | - Guangxiao Zhang
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian 116024, China
- Dalian POCT Laboratory, Dalian 116024, China
| | - Yunping Wu
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian 116024, China
- Dalian POCT Laboratory, Dalian 116024, China
| | - Qiang Zhang
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Yi Liu
- Department of Neurology, Dalian Municipal Central Hospital Affiliated Hospital of Dalian Medical University, Dalian 116033, China
| | - Yangyang Chang
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian 116024, China
- Dalian POCT Laboratory, Dalian 116024, China
| | - Meng Liu
- School of Environmental Science and Technology, Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Dalian 116024, China
- Dalian POCT Laboratory, Dalian 116024, China
| |
Collapse
|
46
|
Ali MM, Mukherjee M, Radford K, Patel Z, Capretta A, Nair P, Brennan JD. A Rapid Sputum-based Lateral Flow Assay for Airway Eosinophilia using an RNA-cleaving DNAzyme Selected for Eosinophil Peroxidase. Angew Chem Int Ed Engl 2023; 62:e202307451. [PMID: 37477970 DOI: 10.1002/anie.202307451] [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: 05/26/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
The first protein-binding allosteric RNA-cleaving DNAzyme (RCD) obtained by direct in vitro selection against eosinophil peroxidase (EPX), a validated marker for airway eosinophilia, is described. The RCD has nanomolar affinity for EPX, shows high selectivity against related peroxidases and other eosinophil proteins, and is resistant to degradation by mammalian nucleases. An optimized RCD was used to develop both fluorescence and lateral flow assays, which were evaluated using 38 minimally processed patient sputum samples (23 non-eosinophilic, 15 eosinophilic), producing a clinical sensitivity of 100 % and specificity of 96 %. This RCD-based lateral flow assay should allow for rapid evaluation of airway eosinophilia as an aid for guiding asthma therapy.
Collapse
Affiliation(s)
- M Monsur Ali
- Biointerfaces Institute, McMaster University, 1280 Main Street West, L8S 4K1, Hamilton, ON, Canada
| | - Manali Mukherjee
- Division of Respirology, McMaster University, Firestone Institute of Respiratory Health at St. Joseph's Health Care, L8N 4A6, Hamilton, ON, Canada
| | - Katherine Radford
- Division of Respirology, McMaster University, Firestone Institute of Respiratory Health at St. Joseph's Health Care, L8N 4A6, Hamilton, ON, Canada
| | - Zil Patel
- Division of Respirology, McMaster University, Firestone Institute of Respiratory Health at St. Joseph's Health Care, L8N 4A6, Hamilton, ON, Canada
| | - Alfredo Capretta
- Biointerfaces Institute, McMaster University, 1280 Main Street West, L8S 4K1, Hamilton, ON, Canada
| | - Parameswaran Nair
- Division of Respirology, McMaster University, Firestone Institute of Respiratory Health at St. Joseph's Health Care, L8N 4A6, Hamilton, ON, Canada
| | - John D Brennan
- Biointerfaces Institute, McMaster University, 1280 Main Street West, L8S 4K1, Hamilton, ON, Canada
| |
Collapse
|
47
|
Montserrat Pagès A, Hertog M, Nicolaï B, Spasic D, Lammertyn J. Unraveling the Kinetics of the 10-23 RNA-Cleaving DNAzyme. Int J Mol Sci 2023; 24:13686. [PMID: 37761982 PMCID: PMC10531344 DOI: 10.3390/ijms241813686] [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: 06/12/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
DNA-based enzymes, or DNAzymes, are single-stranded DNA sequences with the ability to catalyze various chemical reactions, including the cleavage of the bond between two RNA nucleotides. Lately, an increasing interest has been observed in these RNA-cleaving DNAzymes in the biosensing and therapeutic fields for signal generation and the modulation of gene expression, respectively. Additionally, multiple efforts have been made to study the effects of the reaction environment and the sequence of the catalytic core on the conversion of the substrate into product. However, most of these studies have only reported alterations of the general reaction course, but only a few have focused on how each individual reaction step is affected. In this work, we present for the first time a mathematical model that describes and predicts the reaction of the 10-23 RNA-cleaving DNAzyme. Furthermore, the model has been employed to study the effect of temperature, magnesium cations and shorter substrate-binding arms of the DNAzyme on the different kinetic rate constants, broadening the range of conditions in which the model can be exploited. In conclusion, this work depicts the prospects of such mathematical models to study and anticipate the course of a reaction given a particular environment.
Collapse
Affiliation(s)
- Aida Montserrat Pagès
- Department of Biosystems, Biosensors Group, KU Leuven—University of Leuven, 3001 Leuven, Belgium
| | - Maarten Hertog
- Department of Biosystems, Postharvest Group, KU Leuven—University of Leuven, 3001 Leuven, Belgium
| | - Bart Nicolaï
- Department of Biosystems, Postharvest Group, KU Leuven—University of Leuven, 3001 Leuven, Belgium
| | - Dragana Spasic
- Department of Biosystems, Biosensors Group, KU Leuven—University of Leuven, 3001 Leuven, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven—University of Leuven, 3001 Leuven, Belgium
| |
Collapse
|
48
|
Yao Y, Cheng W, Li T, Chen Y, Duan C, Wang Z, Xiang Y. Triple Amplification Strategy of CHA-PER-DNAzyme for Ultrasensitive Detection of circRNA Associated with Hepatocellular Carcinoma. Anal Chem 2023; 95:13149-13155. [PMID: 37607407 DOI: 10.1021/acs.analchem.3c01951] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Biological and clinical studies have indicated that aberrant expression of circMTO1 served as a crucial biomarker for the diagnosis and prognosis of hepatocellular carcinoma (HCC) patients as well as a potential therapeutic target. However, the detection of circRNAs currently faces challenges such as homologous linear RNA interference and low-expression abundance of certain circRNAs. Therefore, we developed a triple amplification method based on catalytic hairpin assembly (CHA) activation by back-splice junction (BSJ), resulting in CHA products that triggered primer exchange reaction to generate DNAzyme. Subsequently, DNAzyme cleaved the fluorescent reporter chain, enabling ultrasensitive detection of hepatocellular carcinoma-associated circMTO1 through the output fluorescence signal. The catalytic hairpin opening sequence within CHA specifically targeted the BSJ sequence in circRNA, thereby avoiding false positive signals observed in circRNA assays due to the recognition of homologous linear RNA molecules. Moreover, this triple amplification method facilitated sensitive detection of circRNA and addressed the issue of low-abundance expression levels associated with circMTO1 in HCC samples. Notably, our newly designed assay for detecting circRNA exhibited a linear range from 1 fM to 100 nM with a detection limit of 0.265 fM. Furthermore, it demonstrated excellent and consistent performance even within complex systems. Our proposed method enabled the specific and sensitive detection of circMTO1 in various cancer cells and blood samples from HCC patients, providing an innovative approach for investigating the role of circRNA in tumorigenesis and development while promoting its clinical application.
Collapse
Affiliation(s)
- Yanheng Yao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Wenting Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Tingting Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Chengjie Duan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Zhongyun Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Yang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
49
|
Wang R, He W, Yi X, Wu Z, Chu X, Jiang JH. Site-Specific Bioorthogonal Activation of DNAzymes for On-Demand Gene Therapy. J Am Chem Soc 2023; 145:17926-17935. [PMID: 37535859 DOI: 10.1021/jacs.3c05413] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
RNA-cleaving DNAzymes hold great promise as gene silencers, and spatiotemporal control of their activity through site-specific reactions is crucial but challenging for on-demand therapy. We herein report a novel design of a bioorthogonally inducible DNAzyme that is deactivated by site-specific installation of bioorthogonal caging groups on the designated backbone sites but restores the activity via a phosphine-triggered Staudinger reduction. We perform a systematical screening for installing the caging groups on each backbone site in the catalytic core of 10-23 DNAzyme and identify an inducible DNAzyme with very low leakage activity. This design is demonstrated to achieve bioorthogonally controlled cleavage of exogenous and endogenous mRNA in live cells. It is further extended to photoactivation and endogenous stimuli activation for spatiotemporal or targeted control of gene silencing. The bioorthogonally inducible DNAzyme is applied to a triple-negative breast cancer mouse model using a lipid nanoparticle delivery system, demonstrating high efficiency in knockdown of Lcn2 oncogenes and substantial suppression of tumor growth, thus highlighting the potential of precisely controlling the DNAzyme functions for on-demand gene therapy.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Wenhan He
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Xin Yi
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Xia Chu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biomedical Sciences, Hunan University, Changsha 410082, China
| |
Collapse
|
50
|
Lu B, Ohayon YP, Woloszyn K, Yang CF, Yoder JB, Rothschild LJ, Wind SJ, Hendrickson WA, Mao C, Seeman NC, Canary JW, Sha R, Vecchioni S. Heterobimetallic Base Pair Programming in Designer 3D DNA Crystals. J Am Chem Soc 2023; 145:17945-17953. [PMID: 37530628 DOI: 10.1021/jacs.3c05478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Metal-mediated DNA (mmDNA) presents a pathway toward engineering bioinorganic and electronic behavior into DNA devices. Many chemical and biophysical forces drive the programmable chelation of metals between pyrimidine base pairs. Here, we developed a crystallographic method using the three-dimensional (3D) DNA tensegrity triangle motif to capture single- and multi-metal binding modes across granular changes to environmental pH using anomalous scattering. Leveraging this programmable crystal, we determined 28 biomolecular structures to capture mmDNA reactions. We found that silver(I) binds with increasing occupancy in T-T and U-U pairs at elevated pH levels, and we exploited this to capture silver(I) and mercury(II) within the same base pair and to isolate the titration points for homo- and heterometal base pair modes. We additionally determined the structure of a C-C pair with both silver(I) and mercury(II). Finally, we extend our paradigm to capture cadmium(II) in T-T pairs together with mercury(II) at high pH. The precision self-assembly of heterobimetallic DNA chemistry at the sub-nanometer scale will enable atomistic design frameworks for more elaborate mmDNA-based nanodevices and nanotechnologies.
Collapse
Affiliation(s)
- Brandon Lu
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Yoel P Ohayon
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Karol Woloszyn
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Chu-Fan Yang
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Jesse B Yoder
- IMCA-CAT, Argonne National Lab, Argonne, Illinois 60439, United States
| | - Lynn J Rothschild
- NASA Ames Research Center, Planetary Sciences Branch, Moffett Field, California 94035, United States
| | - Shalom J Wind
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, United States
| | - Wayne A Hendrickson
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, United States
| | - Chengde Mao
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nadrian C Seeman
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - James W Canary
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Ruojie Sha
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Simon Vecchioni
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|