1
|
Ngo KH, Distler ME, Evangelopoulos M, Ocampo TA, Ma Y, Minorik AJ, Mirkin CA. DNA Dendron Tagging as a Universal Way to Deliver Proteins to Cells. J Am Chem Soc 2025; 147:2129-2136. [PMID: 39812088 PMCID: PMC11755410 DOI: 10.1021/jacs.4c16205] [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: 01/30/2025]
Abstract
The use of proteins as intracellular probes and therapeutic tools is often limited by poor intracellular delivery. One approach to enabling intracellular protein delivery is to transform proteins into spherical nucleic acid (proSNA) nanoconstructs, with surfaces chemically modified with a dense shell of radially oriented DNA that can engage with cell-surface receptors that facilitate endocytosis. However, proteins often have a limited number of available reactive surface residues for DNA conjugation such that the extent of DNA loading and cellular uptake is restricted. Indeed, DNA surface density and sequence have been correlated with scavenger-receptor engagement, the first step of cellular internalization. Here, we report how branched DNA dendrons with dibenzocyclooctyne groups and proteins genetically engineered to include the noncanonical amino acid azido-phenylalanine for click chemistry can be used to synthesize hybrid DNA dendron-protein architectures that exhibit outstanding cellular internalization properties, without the need for extensive surface modification. In a head-to-head comparison, protein-DNA dendron structures (where DNA is concentrated in a local area) are taken up by cells more rapidly and to a greater extent than proSNAs (where the DNA is evenly distributed). Also, protein-G-rich dendron structures show enhanced uptake compared to protein-T-rich dendron structures, highlighting the importance of oligonucleotide sequence on nanoconjugate uptake. Finally, a generalizable method for chemically tagging proteins with dendrons that does not require mutagenesis is described. When a range of proteins, spanning 42 to 464 kDa, were modified through surface lysines with this method, a significant increase in their cellular uptake (up to 17-fold) compared to proteins that are not coupled to a DNA dendron was observed.
Collapse
Affiliation(s)
- Kathleen H. Ngo
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Max E. Distler
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Michael Evangelopoulos
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Tonatiuh A. Ocampo
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Yinglun Ma
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Andrew J. Minorik
- Department of Neurobiology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| |
Collapse
|
2
|
Drachuk I, Ramani N, Harbaugh S, Mirkin CA, Chávez JL. Implantable Fluorogenic DNA Biosensor for Stress Detection. ACS APPLIED MATERIALS & INTERFACES 2025; 17:130-139. [PMID: 39417681 DOI: 10.1021/acsami.4c08940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Implantable sensors that can monitor analytes related to cognitive and physiological status have gained significant focus in recent years. We have developed an implantable biosensor to detect dehydroepiandrosterone sulfate (DHEA-S), a biomarker related to stress. The biosensor strategy was based on the principle of forced intercalation (FIT) aptamers designed to detect subtle intramolecular changes during aptamer-target binding events. By incorporating a steroid-specific fluorogenic aptamer into a hydrogel, the sensitivity and biostability of the FIT biosensor fiber were improved, which were essential for designing implantable sensors to monitor biomarker levels in the living body. The polyethylenimine-based hydrogel chosen for this study produced an optically transparent cross-linked network with optimal microstructure, physicochemical, and mechanical properties, making it suitable for optical biosensors. The in vitro studies showed that the biosensor fiber was successfully activated in human serum and skin analogue, providing a linear response to physiological concentrations of the steroid. We believe that this type of implantable platform can be effective in monitoring more complex biomarkers associated with physiological or psychological health.
Collapse
Affiliation(s)
- Irina Drachuk
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
- UES, a BlueHalo Company, 4401 Dayton-Xenia Rd., Dayton, Ohio 45432, United States
| | - Namrata Ramani
- Department of Materials Science and Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Svetlana Harbaugh
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
| | - Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jorge L Chávez
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
3
|
Chan AM, Ebrahimi SB, Samanta D, Leshchev D, Nijhawan AK, Hsu DJ, Ho MB, Ramani N, Kosheleva I, Henning R, Mirkin CA, Kohlstedt KL, Chen LX. Early Folding Dynamics of i-Motif DNA Revealed by pH-Jump Time-Resolved X-ray Solution Scattering. J Am Chem Soc 2024; 146:33743-33752. [PMID: 39607431 DOI: 10.1021/jacs.4c11768] [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: 11/29/2024]
Abstract
The i-motif is a pH-responsive cytosine-rich oligonucleotide sequence that forms, under acidic conditions, a quadruplex structure. This tunable structural switching has made the i-motif a useful platform for designing pH-responsive nanomaterials. Despite the widespread application of i-motif DNA constructs as biomolecular switches, the mechanism of i-motif folding on the atomic scale has yet to be established. We investigate the early folding structural dynamics of i-motif oligonucleotides with laser-pulse-induced pH-jump time-resolved X-ray solution scattering. Following the pH-jump, we observe that the initial random coil ensemble converts into a contracted intermediate state within 113 ns followed by further folding on the 10 ms time scale. We reveal the representative structures of these transient species, hitherto unknown, with molecular dynamics simulations and ensemble fitting. These results pave the way for understanding metastable conformations of i-motif folding and for benchmarking emerging theoretical models for simulating noncanonical nucleic acid structures.
Collapse
Affiliation(s)
- Arnold M Chan
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Sasha B Ebrahimi
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemical and Biomolecular Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Devleena Samanta
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Denis Leshchev
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Adam K Nijhawan
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Darren J Hsu
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Madeline B Ho
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Namrata Ramani
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Material Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Irina Kosheleva
- Center for Advanced Radiation Sources, The University of Chicago, Chicago, Illinois 60637, United States
| | - Robert Henning
- Center for Advanced Radiation Sources, The University of Chicago, Chicago, Illinois 60637, United States
| | - Chad A Mirkin
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemical and Biomolecular Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Material Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Kevin L Kohlstedt
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Lin X Chen
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Chemical Sciences and Engineering Division, Argonne National Laboratory, Argonne 60439, Illinois, United States
| |
Collapse
|
4
|
Wang G, Han S, Lu Y. From Structure to Application: The Evolutionary Trajectory of Spherical Nucleic Acids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310026. [PMID: 38860348 DOI: 10.1002/smll.202310026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/09/2024] [Indexed: 06/12/2024]
Abstract
Since the proposal of the concept of spherical nucleic acids (SNAs) in 1996, numerous studies have focused on this topic and have achieved great advances. As a new delivery system for nucleic acids, SNAs have advantages over conventional deoxyribonucleic acid (DNA) nanostructures, including independence from transfection reagents, tolerance to nucleases, and lower immune reactions. The flexible structure of SNAs proves that various inorganic or organic materials can be used as the core, and different types of nucleic acids can be conjugated to realize diverse functions and achieve surprising and exciting outcomes. The special DNA nanostructures have been employed for immunomodulation, gene regulation, drug delivery, biosensing, and bioimaging. Despite the lack of rational design strategies, potential cytotoxicity, and structural defects of this technology, various successful examples demonstrate the bright and convincing future of SNAs in fields such as new materials, clinical practice, and pharmacy.
Collapse
Affiliation(s)
- Guijia Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
5
|
Zhao Y, Hou J, Guo L, Zhu S, Hou X, Cao S, Zhou M, Shi J, Li J, Liu K, Zhang H, Wang L, Fan C, Zhu Y. DNA-Engineered Degradable Invisibility Cloaking for Tumor-Targeting Nanoparticles. J Am Chem Soc 2024; 146:25253-25262. [PMID: 39196310 DOI: 10.1021/jacs.4c09479] [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: 08/29/2024]
Abstract
Nanoparticle (NP) delivery systems have been actively exploited for cancer therapy and vaccine development. Nevertheless, the major obstacle to targeted delivery lies in the substantial liver sequestration of NPs. Here we report a DNA-engineered approach to circumvent liver phagocytosis for enhanced tumor-targeted delivery of nanoagents in vivo. We find that a monolayer of DNA molecules on the NP can preferentially adsorb a dysopsonin protein in the serum to induce functionally invisibility to livers; whereas the tumor-specific uptake is triggered by the subsequent degradation of the DNA shell in vivo. The degradation rate of DNA shells is readily tunable by the length of coated DNA molecules. This DNA-engineered invisibility cloaking (DEIC) is potentially generic as manifested in both Ag2S quantum dot- and nanoliposome-based tumor-targeted delivery in mice. Near-infrared-II imaging reveals a high tumor-to-liver ratio of up to ∼5.1, approximately 18-fold higher than those with conventional nanomaterials. This approach may provide a universal strategy for high-efficiency targeted delivery of theranostic agents in vivo.
Collapse
Affiliation(s)
- Yan Zhao
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Junjun Hou
- Zhangjiang Laboratory, 100 Haike Rd, Shanghai 201210, China
| | - Linjie Guo
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Shitai Zhu
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201800, China
| | - Xiaoling Hou
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201800, China
| | | | - Mo Zhou
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiye Shi
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiang Li
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Kai Liu
- Department of Chemistry, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Hongjie Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Lihua Wang
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China
- Zhangjiang Laboratory, 100 Haike Rd, Shanghai 201210, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Corner Stone Science Laboratory, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Zhu
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
6
|
Valatabar N, Oroojalian F, Kazemzadeh M, Mokhtarzadeh AA, Safaralizadeh R, Sahebkar A. Recent advances in gene delivery nanoplatforms based on spherical nucleic acids. J Nanobiotechnology 2024; 22:386. [PMID: 38951806 PMCID: PMC11218236 DOI: 10.1186/s12951-024-02648-5] [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: 04/08/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Gene therapy is a therapeutic option for mitigating diseases that do not respond well to pharmacological therapy. This type of therapy allows for correcting altered and defective genes by transferring nucleic acids to target cells. Notably, achieving a desirable outcome is possible by successfully delivering genetic materials into the cell. In-vivo gene transfer strategies use two major classes of vectors, namely viral and nonviral. Both of these systems have distinct pros and cons, and the choice of a delivery system depends on therapeutic objectives and other considerations. Safe and efficient gene transfer is the main feature of any delivery system. Spherical nucleic acids (SNAs) are nanotechnology-based gene delivery systems (i.e., non-viral vectors). They are three-dimensional structures consisting of a hollow or solid spherical core nanoparticle that is functionalized with a dense and highly organized layer of oligonucleotides. The unique structural features of SNAs confer them a high potency in internalization into various types of tissue and cells, a high stability against nucleases, and efficay in penetrating through various biological barriers (such as the skin, blood-brain barrier, and blood-tumor barrier). SNAs also show negligible toxicity and trigger minimal immune response reactions. During the last two decades, all these favorable physicochemical and biological attributes have made them attractive vehicles for drug and nucleic acid delivery. This article discusses the unique structural properties, types of SNAs, and also optimization mechanisms of SNAs. We also focus on recent advances in the synthesis of gene delivery nanoplatforms based on the SNAs.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mina Kazemzadeh
- Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | | | - Reza Safaralizadeh
- Department of Animal Biology Faculty of Natural Science, University of Tabriz, Tabriz, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Huang Y, Liu X, Zhu J, Chen Z, Yu L, Huang X, Dong C, Li J, Zhou H, Yang Y, Tan W. Enzyme Core Spherical Nucleic Acid That Enables Enhanced Cuproptosis and Antitumor Immune Response through Alleviating Tumor Hypoxia. J Am Chem Soc 2024; 146:13805-13816. [PMID: 38552185 DOI: 10.1021/jacs.3c14247] [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: 05/23/2024]
Abstract
Cuproptosis, a copper-dependent cell death process, has been confirmed to further activate the immune response and mediate the immune resistance. However, hypoxic tumor microenvironment hampers cuproptosis sensitivity and suppresses the body's antitumor immune response. Herein, we have successfully immobilized and functionalized catalase (CAT) with long single-stranded DNA containing polyvalent CpG sequences through rolling circle amplification (RCA) techniques, obtaining an enzyme-cored spherical nucleic acid nanoplatform (CAT-ecSNA-Cu) to deliver copper ions for cuproptosis. The presence of long-stranded DNA-protected CAT enhances mitochondrial respiration by catalyzing the conversion of H2O2 to O2, thereby sensitizing cuproptosis. Meanwhile, increased tumor oxygenation suppresses the expression of the hypoxia-inducible factor-1 (HIF-1) protein, resulting in the alleviation of the immunosuppressive tumor microenvironment. Of note, cuproptosis induces immunogenic cell death (ICD), which facilitates dendritic cell (DC) maturation and enhances antigen presentation through polyCpG-supported Toll-like receptor 9 (TLR9) activation. Furthermore, cuproptosis-induced PD-L1 upregulation in tumor cells complements checkpoint blockers (αPD-L1), enhancing antitumor immunity. The strategy of enhancing cuproptosis-mediated antitumor immune responses by alleviating hypoxia effectively promotes the activation and proliferation of effector T cells, ultimately leading to long-term immunity against cancer.
Collapse
Affiliation(s)
- Yuting Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Jiawei Zhu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Zhejie Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Lu Yu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Xin Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Chuhuang Dong
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Jiabei Li
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Huayuan Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
8
|
Miller MA, Medina S. Life at the interface: Engineering bio-nanomaterials through interfacial molecular self-assembly. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1966. [PMID: 38725255 PMCID: PMC11090466 DOI: 10.1002/wnan.1966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/15/2024]
Abstract
Interfacial self-assembly describes the directed organization of molecules and colloids at phase boundaries. Believed to be fundamental to the inception of primordial life, interfacial assembly is exploited by a myriad of eukaryotic and prokaryotic organisms to execute physiologic activities and maintain homeostasis. Inspired by these natural systems, chemists, engineers, and materials scientists have sought to harness the thermodynamic equilibria at phase boundaries to create multi-dimensional, highly ordered, and functional nanomaterials. Recent advances in our understanding of the biophysical principles guiding molecular assembly at gas-solid, gas-liquid, solid-liquid, and liquid-liquid interphases have enhanced the rational design of functional bio-nanomaterials, particularly in the fields of biosensing, bioimaging and biotherapy. Continued development of non-canonical building blocks, paired with deeper mechanistic insights into interphase self-assembly, holds promise to yield next generation interfacial bio-nanomaterials with unique, and perhaps yet unrealized, properties. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Michael A Miller
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Scott Medina
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
9
|
Park J, Evangelopoulos M, Vasher MK, Kudruk S, Ramani N, Mayer V, Solivan AC, Lee A, Mirkin CA. Enhancing Endosomal Escape and Gene Regulation Activity for Spherical Nucleic Acids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306902. [PMID: 37932003 PMCID: PMC10947971 DOI: 10.1002/smll.202306902] [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: 08/11/2023] [Revised: 10/19/2023] [Indexed: 11/08/2023]
Abstract
The therapeutic potential of small interfering RNAs (siRNAs) is limited by their poor stability and low cellular uptake. When formulated as spherical nucleic acids (SNAs), siRNAs are resistant to nuclease degradation and enter cells without transfection agents with enhanced activity compared to their linear counterparts; however, the gene silencing activity of SNAs is limited by endosomal entrapment, a problem that impacts many siRNA-based nanoparticle constructs. To increase cytosolic delivery, SNAs are formulated using calcium chloride (CaCl2 ) instead of the conventionally used sodium chloride (NaCl). The divalent calcium (Ca2+ ) ions remain associated with the multivalent SNA and have a higher affinity for SNAs compared to their linear counterparts. Importantly, confocal microscopy studies show a 22% decrease in the accumulation of CaCl2 -salted SNAs within the late endosomes compared to NaCl-salted SNAs, indicating increased cytosolic delivery. Consistent with this finding, CaCl2 -salted SNAs comprised of siRNA and antisense DNA all exhibit enhanced gene silencing activity (up to 20-fold), compared to NaCl-salted SNAs regardless of sequence or cell line (U87-MG and SK-OV-3) studied. Moreover, CaCl2 -salted SNA-based forced intercalation probes show improved cytosolic mRNA detection.
Collapse
Affiliation(s)
- Jungsoo Park
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Michael Evangelopoulos
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Matthew K. Vasher
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Sergej Kudruk
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Namrata Ramani
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Material Sciences and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Vinzenz Mayer
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Alexander C. Solivan
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Andrew Lee
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208
| | - Chad A. Mirkin
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Material Sciences and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208
| |
Collapse
|
10
|
Mariconti M, Dechamboux L, Heckmann M, Gros J, Morel M, Escriou V, Baigl D, Hoffmann C, Rudiuk S. Intracellular Delivery of Functional Proteins with DNA-Protein Nanogels-Lipids Complex. J Am Chem Soc 2024; 146:5118-5127. [PMID: 38363821 PMCID: PMC10910493 DOI: 10.1021/jacs.3c08000] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
Using functional proteins for therapeutic purposes due to their high selectivity and/or catalytic properties can enable the control of various cellular processes; however, the transport of active proteins inside living cells remains a major challenge. In contrast, intracellular delivery of nucleic acids has become a routine method for a number of applications in gene therapy, genome editing, or immunization. Here we report a functionalizable platform constituting of DNA-protein nanogel carriers cross-linked through streptavidin-biotin or streptactin-biotin interactions and demonstrate its applicability for intracellular delivery of active proteins. We show that the nanogels can be loaded with proteins bearing either biotin, streptavidin, or strep-tag, and the resulting functionalized nanogels can be delivered into living cells after complexation with cationic lipid vectors. We use this approach for delivery of alkaline phosphatase enzyme, which is shown to keep its catalytic activity after internalization by mouse melanoma B16 cells, as demonstrated by the DDAO-phosphate assay. The resulting functionalized nanogels have dimensions on the order of 100 nm, contain around 100 enzyme molecules, and are shown to be transfectable at low lipid concentrations (charge ratio R± = 0.75). This ensures the low toxicity of our system, which in combination with high local enzyme concentration (∼100 μM) underlines potential interest of this nanoplatform for biomedical applications.
Collapse
Affiliation(s)
- Marina Mariconti
- PASTEUR,
UMR8640, Department of Chemistry, PSL University,
Sorbonne Université, CNRS, Ecole Normale Supérieure, Paris 75005 France
| | | | - Marion Heckmann
- Université
Paris Cité, CNRS, INSERM, UTCBS, Paris 75006, France
| | - Julien Gros
- PASTEUR,
UMR8640, Department of Chemistry, PSL University,
Sorbonne Université, CNRS, Ecole Normale Supérieure, Paris 75005 France
| | - Mathieu Morel
- PASTEUR,
UMR8640, Department of Chemistry, PSL University,
Sorbonne Université, CNRS, Ecole Normale Supérieure, Paris 75005 France
| | | | - Damien Baigl
- PASTEUR,
UMR8640, Department of Chemistry, PSL University,
Sorbonne Université, CNRS, Ecole Normale Supérieure, Paris 75005 France
| | - Céline Hoffmann
- Université
Paris Cité, CNRS, INSERM, UTCBS, Paris 75006, France
| | - Sergii Rudiuk
- PASTEUR,
UMR8640, Department of Chemistry, PSL University,
Sorbonne Université, CNRS, Ecole Normale Supérieure, Paris 75005 France
| |
Collapse
|
11
|
Confederat S, Lee S, Vang D, Soulias D, Marcuccio F, Peace TI, Edwards MA, Strobbia P, Samanta D, Wälti C, Actis P. Next-Generation Nanopore Sensors Based on Conductive Pulse Sensing for Enhanced Detection of Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305186. [PMID: 37649152 PMCID: PMC11475450 DOI: 10.1002/smll.202305186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Nanopore sensing has been successfully used to characterize biological molecules with single-molecule resolution based on the resistive pulse sensing approach. However, its use in nanoparticle characterization has been constrained by the need to tailor the nanopore aperture size to the size of the analyte, precluding the analysis of heterogeneous samples. Additionally, nanopore sensors often require the use of high salt concentrations to improve the signal-to-noise ratio, which further limits their ability to study a wide range of nanoparticles that are unstable at high ionic strength. Here, a new paradigm in nanopore research that takes advantage of a polymer electrolyte system to comprise a conductive pulse sensing approach is presented. A finite element model is developed to explain the conductive pulse signals observed and compare these results with experiments. This system enables the analytical characterization of heterogeneous nanoparticle mixtures at low ionic strength . Furthermore, the wide applicability of the method is demonstrated by characterizing metallic nanospheres of varied sizes, plasmonic nanostars with various degrees of branching, and protein-based spherical nucleic acids with different oligonucleotide loadings. This system will complement the toolbox of nanomaterials characterization techniques to enable real-time optimization workflow for engineering a wide range of nanomaterials.
Collapse
Affiliation(s)
- Samuel Confederat
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
| | - Seungheon Lee
- Department of ChemistryThe University of Texas at AustinAustinTX78712USA
| | - Der Vang
- Department of ChemistryUniversity of CincinnatiCincinnatiOH45221USA
| | - Dimitrios Soulias
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
- Physical and Theoretical Chemistry LaboratoryDepartment of ChemistryUniversity of OxfordOX1 3QZOxfordUK
| | - Fabio Marcuccio
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
- Faculty of MedicineImperial College LondonSW7 2AZLondonUK
| | - Timotheus I. Peace
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLS2 9JTLeedsUK
| | - Martin Andrew Edwards
- Department of Chemistry and BiochemistryUniversity of ArkansasFayettevilleAR72701USA
| | - Pietro Strobbia
- Department of ChemistryUniversity of CincinnatiCincinnatiOH45221USA
| | - Devleena Samanta
- Department of ChemistryThe University of Texas at AustinAustinTX78712USA
| | - Christoph Wälti
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
| | - Paolo Actis
- Bragg Centre for Materials ResearchUniversity of LeedsLS2 9JTLeedsUK
- School of Electronic and Electrical Engineering and Pollard InstituteUniversity of LeedsLS2 9JTLeedsUK
| |
Collapse
|
12
|
Yuan Q, Mao D, Tang X, Liu C, Zhang R, Deng J, Zhu X, Li W, Man Q, Sun F. Biological effect abundance analysis of hemolytic pathogens based on engineered biomimetic sensor. Biosens Bioelectron 2023; 237:115502. [PMID: 37423067 DOI: 10.1016/j.bios.2023.115502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/11/2023]
Abstract
Conventional pathogen detection strategies based on the molecular structure or chemical characteristics of biomarkers can only provide the "physical abundance" of microorganisms, but cannot reflect the "biological effect abundance" in the true sense. To address this issue, we report an erythrocyte membrane-encapsulated biomimetic sensor cascaded with CRISPR-Cas12a (EMSCC). Taking hemolytic pathogens as the target model, we first constructed an erythrocyte membrane-encapsulated biomimetic sensor (EMS). Only hemolytic pathogens with biological effects can disrupt the erythrocyte membrane (EM), resulting in signal generation. Then the signal was amplified by cascading CRISPR-Cas12a, and more than 6.67 × 104-fold improvement in detection sensitivity compared to traditional erythrocyte hemolysis assay was achieved. Notably, compared with polymerase chain reaction (PCR) or enzyme linked immunosorbent assay (ELISA)-based quantification methods, EMSCC can sensitively respond to the pathogenicity change of pathogens. For the detection of simulated clinical samples based on EMSCC, we obtained an accuracy of 95% in 40 samples, demonstrating its potential clinical value.
Collapse
Affiliation(s)
- Qianqin Yuan
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Dongsheng Mao
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xiaochen Tang
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, PR China
| | - Chenbin Liu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Runchi Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Jie Deng
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xiaoli Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Wenxing Li
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| | - Qiuhong Man
- Department of Clinical Laboratory Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200080, PR China.
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| |
Collapse
|
13
|
Liu S, Yu CY, Wei H. Spherical nucleic acids-based nanoplatforms for tumor precision medicine and immunotherapy. Mater Today Bio 2023; 22:100750. [PMID: 37545568 PMCID: PMC10400933 DOI: 10.1016/j.mtbio.2023.100750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
Precise diagnosis and treatment of tumors currently still face considerable challenges due to the development of highly degreed heterogeneity in the dynamic evolution of tumors. With the rapid development of genomics, personalized diagnosis and treatment using specific genes may be a robust strategy to break through the bottleneck of traditional tumor treatment. Nevertheless, efficient in vivo gene delivery has been frequently hampered by the inherent defects of vectors and various biological barriers. Encouragingly, spherical nucleic acids (SNAs) with good modularity and programmability are excellent candidates capable of addressing traditional gene transfer-associated issues, which enables SNAs a precision nanoplatform with great potential for diverse biomedical applications. In this regard, there have been detailed reviews of SNA in drug delivery, gene regulation, and dermatology treatment. Still, to the best of our knowledge, there is no published systematic review summarizing the use of SNAs in oncology precision medicine and immunotherapy, which are considered new guidelines for oncology treatment. To this end, we summarized the notable advances in SNAs-based precision therapy and immunotherapy for tumors following a classification standard of different types of precise spatiotemporal control on active species by SNAs. Specifically, we focus on the structural diversity and programmability of SNAs. Finally, the challenges and possible solutions were discussed in the concluding remarks. This review will promote the rational design and development of SNAs for tumor-precise medicine and immunotherapy.
Collapse
Affiliation(s)
- Songbin Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
14
|
Mirkin CA, Petrosko SH. Inspired Beyond Nature: Three Decades of Spherical Nucleic Acids and Colloidal Crystal Engineering with DNA. ACS NANO 2023; 17:16291-16307. [PMID: 37584399 DOI: 10.1021/acsnano.3c06564] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The conception, synthesis, and invention of a nanostructure, now known as the spherical nucleic acid, or SNA, in 1996 marked the advent of a new field of chemistry. Over the past three decades, the SNA and its analogous anisotropic equivalents have provided an avenue for us to think about some of the most fundamental concepts in chemistry in new ways and led to technologies that are significantly impacting fields from medicine to materials science. A prime example is colloidal crystal engineering with DNA, the framework for using SNAs and related structures to synthesize programmable matter. Herein, we document the evolution of this framework, which was initially inspired by nature, and describe how it now allows researchers to chart paths to move beyond it, as programmable matter with real-world significance is envisioned and created.
Collapse
Affiliation(s)
- Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Sarah Hurst Petrosko
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
15
|
Nixon SR, Phukan IK, Armijo BJ, Ebrahimi SB, Samanta D. Proximity-Driven DNA Nanosensors. ECS SENSORS PLUS 2023; 2:030601. [PMID: 37424706 PMCID: PMC10323711 DOI: 10.1149/2754-2726/ace068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/12/2023] [Indexed: 07/11/2023]
Abstract
In proximity-driven sensing, interactions between a probe and an analyte produce a detectable signal by causing a change in distance of two probe components or signaling moieties. By interfacing such systems with DNA-based nanostructures, platforms that are highly sensitive, specific, and programmable can be designed. In this Perspective, we delineate the advantages of using DNA building blocks in proximity-driven nanosensors and provide an overview of recent progress in the field, from sensors that rapidly detect pesticides in food to probes that identify rare cancer cells in blood. We also discuss current challenges and identify key areas that need further development.
Collapse
Affiliation(s)
- Sara R. Nixon
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Imon Kanta Phukan
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Brian J. Armijo
- Department of Chemistry, Southwestern University, Georgetown, TX 78626, United States of America
| | - Sasha B. Ebrahimi
- Drug Product Development—Steriles, GlaxoSmithKline, Collegeville, PA 19426, United States of America
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, United States of America
| |
Collapse
|
16
|
Fang L, Shi C, Wang Y, Xiong Z, Wang Y. Exploring the diverse biomedical applications of programmable and multifunctional DNA nanomaterials. J Nanobiotechnology 2023; 21:290. [PMID: 37612757 PMCID: PMC10464147 DOI: 10.1186/s12951-023-02071-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023] Open
Abstract
DNA nanoparticles hold great promise for a range of biological applications, including the development of cutting-edge treatments and diagnostic tests. Their subnanometer-level addressability enables precise, specific modifications with a variety of chemical and biological entities, making them ideal as diagnostic instruments and carriers for targeted delivery. This paper focuses on the potential of DNA nanomaterials, which offer scalability, programmability, and functionality. For example, they can be engineered to provide highly specific biosensing and bioimaging capabilities and show promise as a platform for disease diagnosis and treatment. Successful operation of various biomedical nanomaterials has been demonstrated both in vitro and in vivo. However, there are still significant challenges to overcome, including the need to improve the scalability and reliability of the technology, and to ensure safety in clinical applications. We discuss these challenges and opportunities in detail and highlight the progress and prospects of DNA nanotechnology for biomedical applications.
Collapse
Affiliation(s)
- Liuru Fang
- Hubei Province Key Laboratory of Systems Science in Metallurgical Process, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Yuhua Wang
- Hubei Province Key Laboratory of Systems Science in Metallurgical Process, Wuhan University of Science and Technology, Wuhan, 430081, China.
| | - Zuzhao Xiong
- Hubei Province Key Laboratory of Systems Science in Metallurgical Process, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Yumei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
17
|
Kaviani S, Fakih HH, Asohan J, Katolik A, Damha MJ, Sleiman HF. Sequence-Controlled Spherical Nucleic Acids: Gene Silencing, Encapsulation, and Cellular Uptake. Nucleic Acid Ther 2023; 33:265-276. [PMID: 37196168 DOI: 10.1089/nat.2022.0062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
Abstract
Antisense oligonucleotides (ASOs) can predictably alter RNA processing and control protein expression; however, challenges in the delivery of these therapeutics to specific tissues, poor cellular uptake, and endosomal escape have impeded progress in translating these agents into the clinic. Spherical nucleic acids (SNAs) are nanoparticles with a DNA external shell and a hydrophobic core that arise from the self-assembly of ASO strands conjugated to hydrophobic polymers. SNAs have recently shown significant promise as vehicles for improving the efficacy of ASO cellular uptake and gene silencing. However, to date, no studies have investigated the effect of the hydrophobic polymer sequence on the biological properties of SNAs. In this study, we created a library of ASO conjugates by covalently attaching polymers with linear or branched [dodecanediol phosphate] units and systematically varying polymer sequence and composition. We show that these parameters can significantly impact encapsulation efficiency, gene silencing activity, SNA stability, and cellular uptake, thus outlining optimized polymer architectures for gene silencing.
Collapse
Affiliation(s)
- Sepideh Kaviani
- Department of Chemistry, McGill University, Montreal, Canada
| | - Hassan H Fakih
- Department of Chemistry, McGill University, Montreal, Canada
| | - Jathavan Asohan
- Department of Chemistry, McGill University, Montreal, Canada
| | - Adam Katolik
- Department of Chemistry, McGill University, Montreal, Canada
| | - Masad J Damha
- Department of Chemistry, McGill University, Montreal, Canada
| | | |
Collapse
|
18
|
Feng H, Zhao Y, Li Y, Qi X, Shen S, Zhou S. Multi-Armed Anti-CD40-Mediated Dual Drug Delivery System Based on Mesoporous Silica/Au Nanorod Nanocomposites for Multimodality Imaging and Combination Therapy. ACS APPLIED NANO MATERIALS 2023; 6:13001-13012. [DOI: 10.1021/acsanm.3c01722] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Affiliation(s)
- Honghong Feng
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Yangjing Zhao
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Yeping Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Song Shen
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Shengwang Zhou
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| |
Collapse
|
19
|
Hincapie R, Bhattacharya S, Keshavarz-Joud P, Chapman AP, Crooke SN, Finn MG. Preparation and Biological Properties of Oligonucleotide-Functionalized Virus-like Particles. Biomacromolecules 2023. [PMID: 37257068 DOI: 10.1021/acs.biomac.3c00178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Oligonucleotides are powerful molecules for programming function and assembly. When arrayed on nanoparticle scaffolds in high density, the resulting molecules, spherical nucleic acids (SNAs), become imbued with unique properties. We used the copper-catalyzed azide-alkyne cycloaddition to graft oligonucleotides on Qβ virus-like particles to see if such structures also gain SNA-like behavior. Copper-binding ligands were shown to promote the click reaction without degrading oligonucleotide substrates. Reactions were first optimized with a small-molecule fluorogenic reporter and were then applied to the more challenging synthesis of polyvalent protein nanoparticle-oligonucleotide conjugates. The resulting particles exhibited the enhanced cellular uptake and protection from nuclease-mediated oligonucleotide cleavage characteristic of SNAs, had similar residence time in the liver relative to unmodified particles, and were somewhat shielded from immune recognition, resulting in nearly 10-fold lower antibody titers relative to unmodified particles. Oligonucleotide-functionalized virus-like particles thus provide an interesting option for protein nanoparticle-mediated delivery of functional molecules.
Collapse
|
20
|
Ebrahimi SB, Samanta D. Engineering protein-based therapeutics through structural and chemical design. Nat Commun 2023; 14:2411. [PMID: 37105998 PMCID: PMC10132957 DOI: 10.1038/s41467-023-38039-x] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Protein-based therapeutics have led to new paradigms in disease treatment. Projected to be half of the top ten selling drugs in 2023, proteins have emerged as rivaling and, in some cases, superior alternatives to historically used small molecule-based medicines. This review chronicles both well-established and emerging design strategies that have enabled this paradigm shift by transforming protein-based structures that are often prone to denaturation, degradation, and aggregation in vitro and in vivo into highly effective therapeutics. In particular, we discuss strategies for creating structures with increased affinity and targetability, enhanced in vivo stability and pharmacokinetics, improved cell permeability, and reduced amounts of undesired immunogenicity.
Collapse
Affiliation(s)
- Sasha B Ebrahimi
- Drug Product Development-Steriles, GlaxoSmithKline, Collegeville, PA, 19426, USA.
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
21
|
Pathak N, Patino CA, Ramani N, Mukherjee P, Samanta D, Ebrahimi SB, Mirkin CA, Espinosa HD. Cellular Delivery of Large Functional Proteins and Protein-Nucleic Acid Constructs via Localized Electroporation. NANO LETTERS 2023; 23:3653-3660. [PMID: 36848135 PMCID: PMC10433461 DOI: 10.1021/acs.nanolett.2c04374] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Delivery of proteins and protein-nucleic acid constructs into live cells enables a wide range of applications from gene editing to cell-based therapies and intracellular sensing. However, electroporation-based protein delivery remains challenging due to the large sizes of proteins, their low surface charge, and susceptibility to conformational changes that result in loss of function. Here, we use a nanochannel-based localized electroporation platform with multiplexing capabilities to optimize the intracellular delivery of large proteins (β-galactosidase, 472 kDa, 75.38% efficiency), protein-nucleic acid conjugates (protein spherical nucleic acids (ProSNA), 668 kDa, 80.25% efficiency), and Cas9-ribonucleoprotein complex (160 kDa, ∼60% knock-out and ∼24% knock-in) while retaining functionality post-delivery. Importantly, we delivered the largest protein to date using a localized electroporation platform and showed a nearly 2-fold improvement in gene editing efficiencies compared to previous reports. Furthermore, using confocal microscopy, we observed enhanced cytosolic delivery of ProSNAs, which may expand opportunities for detection and therapy.
Collapse
Affiliation(s)
- Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Namrata Ramani
- Department of Materials Science and Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Devleena Samanta
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Sasha B Ebrahimi
- Department of Chemical and Biological Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
22
|
Liu Q, Huang Y, Li L, Li Z, Li M. Endogenous Enzyme-Operated Spherical Nucleic Acids for Cell-Selective Protein Capture and Localization Regulation. Angew Chem Int Ed Engl 2023; 62:e202214958. [PMID: 36788111 DOI: 10.1002/anie.202214958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/16/2023]
Abstract
Precise regulation of protein activity and localization in cancer cells is crucial to dissect the function of the protein-involved cellular network in tumorigenesis, but there is a lack of suitable methodology. Here we report the design of enzyme-operated spherical nucleic acids (E-SNAs) for manipulation of the nucleocytoplasmic translocation of proteins with cancer-cell selectivity. The E-SNAs are constructed by programmable engineering of aptamer-based modules bearing enzyme-responsive units in predesigned sites and further combination with SNA nanotechnology. We demonstrate that E-SNAs are able to regulate cytoplasmic-to-nuclear shuttling of RelA protein efficiently and specifically in tumor cells, while they remain inactive in normal cells due to insufficient enzyme expression. We further confirmed the generality of this strategy by investigating the enzyme-modulated inhibition/activation of thrombin activity by varying the aptamer-based design.
Collapse
Affiliation(s)
- Qing Liu
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhengping Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Mengyuan Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| |
Collapse
|
23
|
Jin X, Wang Q, Pan J, Wang J, He Y, Shang J, Chen M, He X, Zhang Y, Wang B, Wang Y, Gong G, Guo J. A biologically stable, self-catalytic DNAzyme machine encapsulated by metal-phenolic nanoshells for multiple microRNA imaging. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
24
|
Schöllkopf S, Knoll A, Homer A, Seitz O. Double FIT hybridization probes – towards enhancing brightness, turn-on and specificity of RNA detection. Chem Sci 2023; 14:4166-4173. [PMID: 37063796 PMCID: PMC10094420 DOI: 10.1039/d3sc00363a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Efficient fluorogenic hybridization probes combine high brightness and specificity of fluorescence signaling with large turn-on of fluorescence.
Collapse
Affiliation(s)
- Sophie Schöllkopf
- Institut für Chemie, Humboldt-Universität zu Berlin 12489 Berlin Germany
| | - Andrea Knoll
- Institut für Chemie, Humboldt-Universität zu Berlin 12489 Berlin Germany
| | - Amal Homer
- Institut für Chemie, Humboldt-Universität zu Berlin 12489 Berlin Germany
| | - Oliver Seitz
- Institut für Chemie, Humboldt-Universität zu Berlin 12489 Berlin Germany
| |
Collapse
|
25
|
Snider DM, Pandit S, Coffin ML, Ebrahimi SB, Samanta D. DNA-Mediated Control of Protein Function in Semi-Synthetic Systems. Chembiochem 2022; 23:e202200464. [PMID: 36058885 DOI: 10.1002/cbic.202200464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Indexed: 01/25/2023]
Abstract
The development of strategies for controlling protein function in a precise and predictable manner has the potential to revolutionize catalysis, diagnostics, and medicine. In this regard, the use of DNA has emerged as a powerful approach for modulating protein activity. The programmable nature of DNA allows for constructing sophisticated architectures wherein proteins can be placed with control over position, orientation, and stoichiometry. This ability is especially useful considering that the properties of proteins can be influenced by their local environment or their proximity to other functional molecules. Here, we chronicle the different strategies that have been developed to interface DNA with proteins in semi-synthetic systems. We further delineate the unique applications unlocked by the unprecedented level of structural control that DNA affords. We end by outlining outstanding challenges in the area and discuss future research directions towards potential solutions.
Collapse
Affiliation(s)
- Dylan M Snider
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Subrata Pandit
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Mackenzie L Coffin
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| | - Sasha B Ebrahimi
- Drug Product Development - Steriles, GlaxoSmithKline 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, 105 E 24th St, Austin, TX, 78712, USA
| |
Collapse
|
26
|
Spherical nucleic acids-based biosensors for cancer biomarkers detection. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Kusmierz CD, Callmann CE, Kudruk S, Distler ME, Mirkin CA. Transferrin Aptamers Increase the In Vivo Blood-Brain Barrier Targeting of Protein Spherical Nucleic Acids. Bioconjug Chem 2022; 33:1803-1810. [PMID: 36194889 PMCID: PMC10424462 DOI: 10.1021/acs.bioconjchem.2c00389] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The systemic delivery of exogenous proteins to cells within the brain and central nervous system (CNS) is challenging due to the selective impermeability of the blood-brain barrier (BBB). Herein, we hypothesized that protein delivery to the brain could be improved via functionalization with DNA aptamers designed to bind transferrin (TfR) receptors present on the endothelial cells that line the BBB. Using β-galactosidase (β-Gal) as a model protein, we synthesized protein spherical nucleic acids (ProSNAs) comprised of β-Gal decorated with TfR aptamers (Transferrin-ProSNAs). The TfR aptamer motif significantly increases the accumulation of β-Gal in brain tissue in vivo following intravenous injection over both the native protein and ProSNAs containing nontargeting DNA sequences. Furthermore, the widespread distribution of β-Gal throughout the brain is only observed for Transferrin-ProSNAs. Together, this work shows that the SNA architecture can be used to selectively deliver protein cargo to the brain and CNS if the appropriate aptamer sequence is employed as the DNA shell. Moreover, this highlights the importance of DNA sequence design and provides a potential new avenue for designing highly targeted protein delivery systems by combining the power of DNA aptamers together with the SNA platform.
Collapse
Affiliation(s)
- Caroline D. Kusmierz
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Cassandra E. Callmann
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Sergej Kudruk
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Max E. Distler
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
28
|
Abstract
The use of CRISPR/Cas9 systems in genome editing has been limited by the inability to efficiently deliver the key editing components to and across tissues and cell membranes, respectively. Spherical nucleic acids (SNAs) are nanostructures that provide privileged access to both but have yet to be explored as a means of facilitating gene editing. Herein, a new class of CRISPR SNAs are designed and evaluated in the context of genome editing. Specifically, Cas9 ProSNAs comprised of Cas9 cores densely modified with DNA on their exteriors and preloaded with single-guide RNA were synthesized and evaluated for their genome editing capabilities in the context of multiple cell lines. The radial orientation of the DNA on the Cas9 protein surface enhances cellular uptake, without the need for electroporation or transfection agents. In addition, the Cas9 proteins defining the cores of the ProSNAs were fused with GALA peptides on their N-termini and nuclear localization signals on their C-termini to facilitate endosomal escape and maximize nuclear localization and editing efficiency, respectively. These constructs were stable against protease digestion under conditions that fully degrade the Cas9 protein, when not transformed into an SNA, and used to achieve genome editing efficiency between 32 and 47%. Taken together, these novel constructs and advances point toward a way of significantly broadening the scope of use and impact of CRISPR-Cas9 genome editing systems.
Collapse
Affiliation(s)
- Chi Huang
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208-3113, United States
| | - Zhenyu Han
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208-3113, United States
| | - Michael Evangelopoulos
- Department of Biomedical Engineering and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208-3113, United States
| | - Chad A. Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208-3113, United States
- Department of Biomedical Engineering and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208-3113, United States
| |
Collapse
|
29
|
Theodorou A, Gounaris D, Voutyritsa E, Andrikopoulos N, Baltzaki CIM, Anastasaki A, Velonia K. Rapid Oxygen-Tolerant Synthesis of Protein-Polymer Bioconjugates via Aqueous Copper-Mediated Polymerization. Biomacromolecules 2022; 23:4241-4253. [PMID: 36067415 DOI: 10.1021/acs.biomac.2c00726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synthesis of protein-polymer conjugates usually requires extensive and costly deoxygenation procedures, thus limiting their availability and potential applications. In this work, we report the ultrafast synthesis of polymer-protein bioconjugates in the absence of any external deoxygenation via an aqueous copper-mediated methodology. Within 10 min and in the absence of any external stimulus such as light (which may limit the monomer scope and/or disrupt the secondary structure of the protein), a range of hydrophobic and hydrophilic monomers could be successfully grafted from a BSA macroinitiator, yielding well-defined polymer-protein bioconjugates at quantitative yields. Our approach is compatible with a wide range of monomer classes such as (meth) acrylates, styrene, and acrylamides as well as multiple macroinitiators including BSA, BSA nanoparticles, and beta-galactosidase from Aspergillus oryzae. Notably, the synthesis of challenging protein-polymer-polymer triblock copolymers was also demonstrated, thus significantly expanding the scope of our strategy. Importantly, both lower and higher scale polymerizations (from 0.2 to 35 mL) were possible without compromising the overall efficiency and the final yields. This simple methodology paves the way for a plethora of applications in aqueous solutions without the need of external stimuli or tedious deoxygenation.
Collapse
Affiliation(s)
- Alexis Theodorou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Dimitris Gounaris
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Errika Voutyritsa
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nicholas Andrikopoulos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | | | | | - Kelly Velonia
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| |
Collapse
|
30
|
Han Q, Zhang X, Jia Y, Guo S, Zhu J, Luo S, Na N, Ouyang J. Synthesis and Characteristics of Self‐Assembled Multifunctional Ln
3+
‐DNA Hybrid Coordination Polymers. Chemistry 2022; 28:e202200281. [DOI: 10.1002/chem.202200281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Qingzhi Han
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Xinlian Zhang
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Yijing Jia
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Shaoshi Guo
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Jiale Zhu
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Shirui Luo
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Na Na
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| | - Jin Ouyang
- Key Laboratory of Theoretical and Computational Photochemistry College of Chemistry Beijing Normal University Beijing 100875 China
| |
Collapse
|
31
|
Lee S, Godhulayyagari S, Nguyen ST, Lu JK, Ebrahimi SB, Samanta D. Signal Transduction Strategies for Analyte Detection Using DNA-Based Nanostructures. Angew Chem Int Ed Engl 2022; 61:e202202211. [PMID: 35307938 DOI: 10.1002/anie.202202211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Indexed: 12/14/2022]
Abstract
The use of DNA-based nanostructures as probes has led to significant advances in chemical and biological sensing, allowing the detection of analytes in complex media, the understanding of fundamental biological processes, and the ability to diagnose diseases based on molecular signatures. The utility of these structures arises both from DNA's inherent ability to selectively recognize and bind a variety of chemical species and from the unique properties observed when DNA is restructured at the nanoscale. In this Minireview, we chronicle the most commonly used signal transduction strategies that have been interfaced with various DNA-based nanostructures. We discuss the types of analytes and the detection scenarios that are sought after, delineate the advantages and disadvantages of each signaling strategy, and outline the key considerations that guide the selection of each signaling method.
Collapse
Affiliation(s)
- Seungheon Lee
- Department of Chemistry, The University of Texas at Austin, 105 E 24th Street, Austin, TX 78712, USA
| | - Shivudu Godhulayyagari
- Department of Chemistry, The University of Texas at Austin, 105 E 24th Street, Austin, TX 78712, USA
| | - Shadler T Nguyen
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Jasmine K Lu
- Department of Chemistry, The University of Texas at Austin, 105 E 24th Street, Austin, TX 78712, USA
| | - Sasha B Ebrahimi
- Biopharmaceutical Product Sciences, GlaxoSmithKline, 1250 S Collegeville Road, Collegeville, PA 19426, USA
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, 105 E 24th Street, Austin, TX 78712, USA
| |
Collapse
|
32
|
Yu Y, Wang Z, Wu S, Zhu C, Meng X, Li C, Cheng S, Tao W, Wang F. Glutathione-Sensitive Nanoglue Platform with Effective Nucleic Acids Gluing onto Liposomes for Photo-Gene Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:25126-25134. [PMID: 35608168 DOI: 10.1021/acsami.2c04022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Liposomal spherical nucleic acids possess a high density of nucleic acids, e.g., DNA, on a liposomal core. There are two approaches to conjugate DNA onto the zwitterionic liposomes, i.e., covalent and noncovalent conjugation, otherwise using cationic liposomes. However, complex and expensive DNA chemical modification methods need to seek a novel and easy-operating approach to decorating DNA onto liposomes. Inspired by the nanoparticle solution as nanoglues for gels and biological tissues, we use MnO2 nanosheets to "glue" DNA onto liposomes without DNA modification. In tumor cells with a high glutathione concentration, MnO2-based nanoglues are degraded, generating water-soluble Mn2+ ions, further "unglue" DNA (i.e., DNAzyme), and liposomes. Using the intelligent liposomal nanoglue (DNAzyme/MnO2/Lip) combining glutathione-sensitive MnO2 nanosheets, gene silencing agent DNAzyme, and photosensitizer Chlorin e6 (Ce6) in liposomes, effective photo-gene therapy was demonstrated.
Collapse
Affiliation(s)
- Yue Yu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Zhenfeng Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Sichen Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Chunmeng Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Xianshe Meng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Chao Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Sheng Cheng
- Instrumental Analysis Center, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Wei Tao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Feng Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| |
Collapse
|
33
|
Lee S, Godhulayyagari S, Nguyen ST, Lu JK, Ebrahimi SB, Samanta D. Signal Transduction Strategies for Analyte Detection Using DNA‐Based Nanostructures. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Seungheon Lee
- Department of Chemistry The University of Texas at Austin 105 E 24th Street Austin TX 78712 USA
| | - Shivudu Godhulayyagari
- Department of Chemistry The University of Texas at Austin 105 E 24th Street Austin TX 78712 USA
| | - Shadler T. Nguyen
- Department of Molecular Biosciences The University of Texas at Austin 2500 Speedway Austin TX 78712 USA
| | - Jasmine K. Lu
- Department of Chemistry The University of Texas at Austin 105 E 24th Street Austin TX 78712 USA
| | - Sasha B. Ebrahimi
- Biopharmaceutical Product Sciences GlaxoSmithKline 1250 S Collegeville Road Collegeville PA 19426 USA
| | - Devleena Samanta
- Department of Chemistry The University of Texas at Austin 105 E 24th Street Austin TX 78712 USA
| |
Collapse
|
34
|
Mahajan AS, Stegh AH. Spherical Nucleic Acids as Precision Therapeutics for the Treatment of Cancer-From Bench to Bedside. Cancers (Basel) 2022; 14:cancers14071615. [PMID: 35406387 PMCID: PMC8996871 DOI: 10.3390/cancers14071615] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Spherical Nucleic Acids (SNAs) emerged as a new class of nanotherapeutics consisting of a nanoparticle core densely functionalized with a shell of radially oriented synthetic oligonucleotides. The unique three-dimensional architecture of SNAs protects the oligonucleotides from nuclease-mediated degradation, increases oligonucleotide bioavailability, and in the absence of auxiliary transfection agents, enables robust uptake into tumor and immune cells through polyvalent association with cell surface pattern recognition receptors. When composed of gene-regulatory small interfering (si)RNA or immunostimulatory DNA or RNA oligonucleotides, SNAs silence gene expression and induce immune responses superior to those raised by the oligonucleotides in their "free" form. Early phase clinical trials of gene-regulatory siRNA-based SNAs in glioblastoma (NCT03020017) and immunostimulatory Toll-like receptor 9 (TLR9)-agonistic SNAs carrying unmethylated CpG-rich oligonucleotides in solid tumors (NCT03086278) have shown that SNAs represent a safe, brain-penetrant therapy for inhibiting oncogene expression and stimulating immune responses against tumors. This review focuses on the application of SNAs as precision cancer therapeutics, summarizes the findings from first-in-human clinical trials of SNAs in solid tumors, describes the most recent preclinical efforts to rationally design next-generation multimodal SNA architectures, and provides an outlook on future efforts to maximize the anti-neoplastic activity of the SNA platform.
Collapse
Affiliation(s)
- Akanksha S. Mahajan
- Ken and Ruth Davee Department of Neurology, The International Institute for Nanotechnology, The Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA;
| | - Alexander H. Stegh
- Ken and Ruth Davee Department of Neurology, The International Institute for Nanotechnology, The Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA;
- Department of Neurological Surgery, The Brain Tumor Center, Washington University School of Medicine, Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
- Correspondence:
| |
Collapse
|
35
|
Samanta D, Zhou W, Ebrahimi SB, Petrosko SH, Mirkin CA. Programmable Matter: The Nanoparticle Atom and DNA Bond. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107875. [PMID: 34870875 DOI: 10.1002/adma.202107875] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/22/2021] [Indexed: 05/21/2023]
Abstract
Colloidal crystal engineering with DNA has led to significant advances in bottom-up materials synthesis and a new way of thinking about fundamental concepts in chemistry. Here, programmable atom equivalents (PAEs), comprised of nanoparticles (the "atoms") functionalized with DNA (the "bonding elements"), are assembled through DNA hybridization into crystalline lattices. Unlike atomic systems, the "atom" (e.g., the nanoparticle shape, size, and composition) and the "bond" (e.g., the DNA length and sequence) can be tuned independently, yielding designer materials with unique catalytic, optical, and biological properties. In this review, nearly three decades of work that have contributed to the evolution of this class of programmable matter is chronicled, starting from the earliest examples based on gold-core PAEs, and then delineating how advances in synthetic capabilities, DNA design, and fundamental understanding of PAE-PAE interactions have led to new classes of functional materials that, in several cases, have no natural equivalent.
Collapse
Affiliation(s)
- Devleena Samanta
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Wenjie Zhou
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Sasha B Ebrahimi
- Department of Chemical Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Sarah Hurst Petrosko
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
- Department of Chemical Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| |
Collapse
|
36
|
Halloy F, Biscans A, Bujold KE, Debacker A, Hill AC, Lacroix A, Luige O, Strömberg R, Sundstrom L, Vogel J, Ghidini A. Innovative developments and emerging technologies in RNA therapeutics. RNA Biol 2022; 19:313-332. [PMID: 35188077 PMCID: PMC8865321 DOI: 10.1080/15476286.2022.2027150] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RNA-based therapeutics are emerging as a powerful platform for the treatment of multiple diseases. Currently, the two main categories of nucleic acid therapeutics, antisense oligonucleotides and small interfering RNAs (siRNAs), achieve their therapeutic effect through either gene silencing, splicing modulation or microRNA binding, giving rise to versatile options to target pathogenic gene expression patterns. Moreover, ongoing research seeks to expand the scope of RNA-based drugs to include more complex nucleic acid templates, such as messenger RNA, as exemplified by the first approved mRNA-based vaccine in 2020. The increasing number of approved sequences and ongoing clinical trials has attracted considerable interest in the chemical development of oligonucleotides and nucleic acids as drugs, especially since the FDA approval of the first siRNA drug in 2018. As a result, a variety of innovative approaches is emerging, highlighting the potential of RNA as one of the most prominent therapeutic tools in the drug design and development pipeline. This review seeks to provide a comprehensive summary of current efforts in academia and industry aimed at fully realizing the potential of RNA-based therapeutics. Towards this, we introduce established and emerging RNA-based technologies, with a focus on their potential as biosensors and therapeutics. We then describe their mechanisms of action and their application in different disease contexts, along with the strengths and limitations of each strategy. Since the nucleic acid toolbox is rapidly expanding, we also introduce RNA minimal architectures, RNA/protein cleavers and viral RNA as promising modalities for new therapeutics and discuss future directions for the field.
Collapse
Affiliation(s)
- François Halloy
- Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Annabelle Biscans
- Oligonucleotide Chemistry, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| | - Katherine E. Bujold
- Department of Chemistry & Chemical Biology, McMaster University, (Ontario), Canada
| | | | - Alyssa C. Hill
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eth Zürich, Zürich, Switzerland
| | - Aurélie Lacroix
- Sixfold Bioscience, Translation & Innovation Hub, London, UK
| | - Olivia Luige
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Roger Strömberg
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Linda Sundstrom
- Mechanistic and Structural Biology, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| | - Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research (Hiri), Helmholtz Center for Infection Research (Hzi), Würzburg, Germany
- RNA Biology Group, Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Alice Ghidini
- Mechanistic and Structural Biology, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
37
|
Two-dimensional coordination polymer-based nanosensor for sensitive and reliable nucleic acids detection in living cells. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.07.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Ebrahimi SB, Samanta D, Kusmierz CD, Mirkin CA. Protein transfection via spherical nucleic acids. Nat Protoc 2022; 17:327-357. [PMID: 35039669 DOI: 10.1038/s41596-021-00642-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
The efficient transfection of functional proteins into cells can serve as a means for regulating cellular processes toward solving fundamental challenges in biology and medicine. However, the use of proteins as effective intracellular agents is hindered by their low cellular uptake and susceptibility to degradation. Over the past 15 years, our group has been developing spherical nucleic acids (SNAs), nanoparticles functionalized with a dense radially oriented shell of nucleic acids. These structures actively enter cells and have opened new frontiers in chemical sensing, biodiagnostics and therapeutics. Recently, we have shown that proteins can be used as structurally precise and homogeneous nanoparticle cores in SNAs. The resultant protein SNAs (ProSNAs) allow previously cell-impermeable proteins to actively enter cells, exhibit high degrees of stability and activity both in cell culture and in vivo, and show enhanced pharmacokinetics. Consequently, these modular structures constitute a plug-and-play platform in which the protein core and nucleic acid shell can be independently varied to achieve a desired function. Here, we describe the synthesis of ProSNAs through the chemical modification of solvent-accessible surface residues (3-5 d). We also discuss design considerations, strategies for characterization, and applications of ProSNAs in cellular transfection, biological sensing and functional enzyme delivery in vivo.
Collapse
Affiliation(s)
- Sasha B Ebrahimi
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
| | - Devleena Samanta
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Caroline D Kusmierz
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Chad A Mirkin
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
39
|
Song Y, Song W, Lan X, Cai W, Jiang D. Spherical nucleic acids: Organized nucleotide aggregates as versatile nanomedicine. AGGREGATE (HOBOKEN, N.J.) 2022; 3:e120. [PMID: 35386748 PMCID: PMC8982904 DOI: 10.1002/agt2.120] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Spherical nucleic acids (SNAs) are composed of a nanoparticle core and a layer of densely arranged oligonucleotide shells. After the first report of SNA by Mirkin and coworkers in 1996, it has created a significant interest by offering new possibilities in the field of gene and drug delivery. The controlled aggregation of oligonucleotides on the surface of organic/inorganic nanoparticles improves the delivery of genes and nucleic acid-based drugs and alters and regulates the biological profiles of the nanoparticle core within living organisms. Here in this review, we present an overview of the recent progress of SNAs that has speeded up their biomedical application and their potential transition to clinical use. We start with introducing the concept and characteristics of SNAs as drug/gene delivery systems and highlight recent efforts of bioengineering SNA by imaging and treatmenting various diseases. Finally, we discuss potential challenges and opportunities of SNAs, their ongoing clinical trials, and future translation, and how they may affect the current landscape of clinical practices. We hope that this review will update our current understanding of SNA, organized oligonucleotide aggregates, for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Wenyu Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
40
|
Kyriazi ME, El-Sagheer AH, Medintz IL, Brown T, Kanaras AG. An Investigation into the Resistance of Spherical Nucleic Acids against DNA Enzymatic Degradation. Bioconjug Chem 2022; 33:219-225. [PMID: 35001632 DOI: 10.1021/acs.bioconjchem.1c00540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nanoparticles coated with oligonucleotides, also termed spherical nucleic acids (SNAs), are at the forefront of scientific research and have been applied in vitro and in vivo for sensing, gene regulation, and drug delivery. They demonstrate unique properties stemming from the three-dimensional shell of oligonucleotides and present high cellular uptake. However, their resistance to enzymatic degradation is highly dependent on their physicochemical characteristics. In particular, the oligonucleotide loading of SNAs has been determined to be a critical parameter in SNA design. In order to ensure the successful function of SNAs, the degree of oligonucleotide loading has to be quantitatively determined to confirm that a dense oligonucleotide shell has been achieved. However, this can be time-consuming and may lead to multiple syntheses being required to achieve the necessary degree of surface functionalization. In this work we show how this limitation can be overcome by introducing an oligonucleotide modification. By replacing the phosphodiester bond on the oligonucleotide backbone with a phosphorothioate bond, SNAs even with a low DNA loading showed remarkable stability in the presence of nucleases. Furthermore, these chemically modified SNAs exhibited high selectivity and specificity toward the detection of mRNA in cellulo.
Collapse
Affiliation(s)
- Maria-Eleni Kyriazi
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton SO171BJ, United Kingdom
- College of Engineering and Technology, American University of the Middle East, Kuwait City, 15453, Kuwait
| | - Afaf H El-Sagheer
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
- Chemistry Branch, Department of Science and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez 43721, Egypt
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - Tom Brown
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Antonios G Kanaras
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton SO171BJ, United Kingdom
- Institute for Life Science, University of Southampton, Southampton, SO171BJ, United Kingdom
| |
Collapse
|
41
|
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H, Yue J. Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater 2022; 7:292-323. [PMID: 34466734 PMCID: PMC8379367 DOI: 10.1016/j.bioactmat.2021.05.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/28/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic oligonucleotides (TOs) represent one of the most promising drug candidates in the targeted cancer treatment due to their high specificity and capability of modulating cellular pathways that are not readily druggable. However, efficiently delivering of TOs to cancer cellular targets is still the biggest challenge in promoting their clinical translations. Emerging as a significant drug delivery vector, nanoparticles (NPs) can not only protect TOs from nuclease degradation and enhance their tumor accumulation, but also can improve the cell uptake efficiency of TOs as well as the following endosomal escape to increase the therapeutic index. Furthermore, targeted and on-demand drug release of TOs can also be approached to minimize the risk of toxicity towards normal tissues using stimuli-responsive NPs. In the past decades, remarkable progresses have been made on the TOs delivery based on various NPs with specific purposes. In this review, we will first give a brief introduction on the basis of TOs as well as the action mechanisms of several typical TOs, and then describe the obstacles that prevent the clinical translation of TOs, followed by a comprehensive overview of the recent progresses on TOs delivery based on several various types of nanocarriers containing lipid-based nanoparticles, polymeric nanoparticles, gold nanoparticles, porous nanoparticles, DNA/RNA nanoassembly, extracellular vesicles, and imaging-guided drug delivery nanoparticles.
Collapse
Affiliation(s)
- Lei Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Lihua Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Anhong Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Jing Feng
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Xiangmeng Qu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
| | - Jun Yue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
42
|
Zhang B, Bai S, Chao X, Wu T, Chen Z, Cheng Z, Xiao Y, Zhang K, Bai Y. Molecularly pure miktoarm spherical nucleic acids: preparation and usage as a scaffold for abiotic intracellular catalysis. Chem Sci 2021; 12:15843-15848. [PMID: 35024108 PMCID: PMC8672723 DOI: 10.1039/d1sc04833c] [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: 09/01/2021] [Accepted: 10/31/2021] [Indexed: 11/26/2022] Open
Abstract
We present a fullerene-based strategy that allows the synthesis of molecularly pure miktoarm spherical nucleic acids (SNAs) with diverse structures, which, with post-functionalization, could serve as efficient scaffolds for intracellular catalysis. The SNA structure promotes cell permeability, nucleic acid stability, and catalytic efficiency, making the platform ideal for in cellulo reactions. Consequently, the tris(triazole)-bearing miktoarm SNA was able to effectively mediate intracellular copper-catalyzed alkyne-azide cycloaddition at nanomolar level of copper, and facilitate the same reaction in live zebrafish.
Collapse
Affiliation(s)
- Bohan Zhang
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Silei Bai
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Xiangyu Chao
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Tong Wu
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Zhiyong Chen
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Zehong Cheng
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Yue Xiao
- School of Chemistry and Chemical Engineering, Zhengzhou University Zhengzhou Henan 450001 China
| | - Ke Zhang
- School of Chemistry and Chemical Engineering, Zhengzhou University Zhengzhou Henan 450001 China
- Department of Chemistry and Chemical Biology, Northeastern University Boston MA 02115 USA
| | - Yugang Bai
- State Key Laboratory of Chem-/Bio-Sensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| |
Collapse
|
43
|
Liu J, Yan L, He S, Hu J. Engineering DNA quadruplexes in DNA nanostructures for biosensor construction. NANO RESEARCH 2021; 15:3504-3513. [PMID: 35401944 PMCID: PMC8983328 DOI: 10.1007/s12274-021-3869-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Accepted: 09/04/2021] [Indexed: 06/14/2023]
Abstract
DNA quadruplexes are nucleic acid conformations comprised of four strands. They are prevalent in human genomes and increasing efforts are being directed toward their engineering. Taking advantage of the programmability of Watson-Crick base-pairing and conjugation methodology of DNA with other molecules, DNA nanostructures of increasing complexity and diversified geometries have been artificially constructed since 1980s. In this review, we investigate the interweaving of natural DNA quadruplexes and artificial DNA nanostructures in the development of the ever-prosperous field of biosensing, highlighting their specific roles in the construction of biosensor, including recognition probe, signal probe, signal amplifier and support platform. Their implementation in various sensing scenes was surveyed. And finally, general conclusion and future perspective are discussed for further developments.
Collapse
Affiliation(s)
- Jingxin Liu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118 China
| | - Li Yan
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118 China
| | - Shiliang He
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118 China
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118 China
- Shenzhen Bey Laboratory, Shenzhen, 518132 China
| |
Collapse
|
44
|
Ebrahimi SB, Samanta D, Partridge BE, Kusmierz CD, Cheng HF, Grigorescu AA, Chávez JL, Mirau PA, Mirkin CA. Programming Fluorogenic DNA Probes for Rapid Detection of Steroids. Angew Chem Int Ed Engl 2021; 60:15260-15265. [PMID: 33878237 DOI: 10.1002/anie.202103440] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/13/2021] [Indexed: 12/24/2022]
Abstract
The ability of aptamers to recognize a variety of different molecules has fueled their emergence as recognition agents to probe complex media and cells. Many detection strategies require aptamer binding to its target to result in a dramatic change in structure, typically from an unfolded to a folded state. Here, we report a strategy based on forced intercalation (FIT) that increases the scope of aptamer recognition by transducing subtle changes in aptamer structures into fluorescent readouts. By screening a library of green-fluorescent FIT-aptamers whose design is guided by computational modeling, we could identify hits that sense steroids like dehydroepiandrosterone sulfate (DHEAS) down to 1.3 μM with no loss in binding affinity compared to the unmodified aptamer. This enabled us to study DHEAS in clinical serum samples with several advantages over gold standard methods, including rapid readout (<30 min), simple instrumentation (plate-reader), and low sample volumes (10 μL).
Collapse
Affiliation(s)
- Sasha B Ebrahimi
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Devleena Samanta
- Department of Chemistry and International Institute for, Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Benjamin E Partridge
- Department of Chemistry and International Institute for, Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Caroline D Kusmierz
- Department of Chemistry and International Institute for, Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Ho Fung Cheng
- Department of Chemistry and International Institute for, Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | - Arabela A Grigorescu
- Keck Biophysics Facility, Northwestern University, 2220 Campus Drive, Evanston, IL, 60208, USA
| | - Jorge L Chávez
- Airman Systems Directorate and 711th Human Performance Wing, Air Force Research Laboratory, 2510 Fifth St, Area B Bldg 840, WPAFB, OH, 45433, USA
| | - Peter A Mirau
- Materials and Manufacturing Directorate, Air Force Research Laboratory, 2941 Hobson Way, WPAFB, OH, 45433, USA
| | - Chad A Mirkin
- Department of Chemistry and International Institute for, Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| |
Collapse
|
45
|
Ebrahimi SB, Samanta D, Partridge BE, Kusmierz CD, Cheng HF, Grigorescu AA, Chávez JL, Mirau PA, Mirkin CA. Programming Fluorogenic DNA Probes for Rapid Detection of Steroids. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sasha B. Ebrahimi
- Department of Chemical and Biological Engineering Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Devleena Samanta
- Department of Chemistry and International Institute for, Nanotechnology Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Benjamin E. Partridge
- Department of Chemistry and International Institute for, Nanotechnology Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Caroline D. Kusmierz
- Department of Chemistry and International Institute for, Nanotechnology Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Ho Fung Cheng
- Department of Chemistry and International Institute for, Nanotechnology Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Arabela A. Grigorescu
- Keck Biophysics Facility Northwestern University 2220 Campus Drive Evanston IL 60208 USA
| | - Jorge L. Chávez
- Airman Systems Directorate and 711th Human Performance Wing Air Force Research Laboratory 2510 Fifth St, Area B Bldg 840 WPAFB OH 45433 USA
| | - Peter A. Mirau
- Materials and Manufacturing Directorate Air Force Research Laboratory 2941 Hobson Way WPAFB OH 45433 USA
| | - Chad A. Mirkin
- Department of Chemistry and International Institute for, Nanotechnology Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| |
Collapse
|
46
|
Patil TV, Patel DK, Dutta SD, Ganguly K, Lim KT. Graphene Oxide-Based Stimuli-Responsive Platforms for Biomedical Applications. Molecules 2021; 26:2797. [PMID: 34068529 PMCID: PMC8126026 DOI: 10.3390/molecules26092797] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 01/13/2023] Open
Abstract
Graphene is a two-dimensional sp2 hybridized carbon material that has attracted tremendous attention for its stimuli-responsive applications, owing to its high surface area and excellent electrical, optical, thermal, and mechanical properties. The physicochemical properties of graphene can be tuned by surface functionalization. The biomedical field pays special attention to stimuli-responsive materials due to their responsive abilities under different conditions. Stimuli-responsive materials exhibit great potential in changing their behavior upon exposure to external or internal factors, such as pH, light, electric field, magnetic field, and temperature. Graphene-based materials, particularly graphene oxide (GO), have been widely used in stimuli-responsive applications due to their superior biocompatibility compared to other forms of graphene. GO has been commonly utilized in tissue engineering, bioimaging, biosensing, cancer therapy, and drug delivery. GO-based stimuli-responsive platforms for wound healing applications have not yet been fully explored. This review describes the effects of different stimuli-responsive factors, such as pH, light, temperature, and magnetic and electric fields on GO-based materials and their applications. The wound healing applications of GO-based materials is extensively discussed with cancer therapy and drug delivery.
Collapse
Affiliation(s)
- Tejal V. Patil
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (T.V.P.); (D.K.P.); (S.D.D.); (K.G.)
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Korea
| | - Dinesh K. Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (T.V.P.); (D.K.P.); (S.D.D.); (K.G.)
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (T.V.P.); (D.K.P.); (S.D.D.); (K.G.)
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (T.V.P.); (D.K.P.); (S.D.D.); (K.G.)
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (T.V.P.); (D.K.P.); (S.D.D.); (K.G.)
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
47
|
Fukumoto S, Kawade M, Kimura K, Akiyama Y, Kikuchi A. Preparation of Spherical Nucleic Acid Nanoparticles Containing a Self-immolative Poly(carbamate) Core. ANAL SCI 2021; 37:781-784. [PMID: 33487596 DOI: 10.2116/analsci.20scn06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We prepared a novel spherical nucleic acid, containing a core structure of self-immolative poly(carbamate) (PC), with aminobenzyl alcohol as a repeating unit, by conjugating an end-activated PC derivative with an amine-terminated oligoDNA on a solid support for PC-oligoDNA. Dynamic light-scattering measurements revealed a hydrodynamic diameter of 107 nm with a narrow size distribution. A fluorescent monomer with aminobenzyl alcohol is available for PC-oligoDNA synthesis to enhance the fluorescence emission by a domino-like disassembly of PC in response to various external stimuli.
Collapse
Affiliation(s)
- Shione Fukumoto
- Department of Materials Science and Technology, Tokyo University of Science
| | - Mami Kawade
- Department of Materials Science and Technology, Tokyo University of Science
| | - Kazunori Kimura
- Department of Materials Science and Technology, Tokyo University of Science
| | - Yoshitsugu Akiyama
- Faculty of Industrial Science and Technology, Tokyo University of Science
| | - Akihiko Kikuchi
- Department of Materials Science and Technology, Tokyo University of Science
| |
Collapse
|
48
|
Walter EH, Ge Y, Mason JC, Boyle JJ, Long NJ. A Coumarin-Porphyrin FRET Break-Apart Probe for Heme Oxygenase-1. J Am Chem Soc 2021; 143:6460-6469. [PMID: 33845576 PMCID: PMC8154531 DOI: 10.1021/jacs.0c12864] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Heme oxygenase-1 (HO-1) is a vital enzyme in humans that primarily regulates free heme concentrations. The overexpression of HO-1 is commonly associated with cardiovascular and neurodegenerative diseases including atherosclerosis and ischemic stroke. Currently, there are no known chemical probes to detect HO-1 activity, limiting its potential as an early diagnostic/prognostic marker in these serious diseases. Reported here are the design, synthesis, and photophysical and biological characterization of a coumarin-porphyrin FRET break-apart probe to detect HO-1 activity, Fe-L1. We designed Fe-L1 to "break-apart" upon HO-1-catalyzed porphyrin degradation, perturbing the efficient FRET mechanism from a coumarin donor to a porphyrin acceptor fluorophore. Analysis of HO-1 activity using Escherichia coli lysates overexpressing hHO-1 found that a 6-fold increase in emission intensity at 383 nm was observed following incubation with NADPH. The identities of the degradation products following catabolism were confirmed by MALDI-MS and LC-MS, showing that porphyrin catabolism was regioselective at the α-position. Finally, through the analysis of Fe-L2, we have shown that close structural analogues of heme are required to maintain HO-1 activity. It is anticipated that this work will act as a foundation to design and develop new probes for HO-1 activity in the future, moving toward applications of live fluorescent imaging.
Collapse
Affiliation(s)
- Edward
R. H. Walter
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, White City Campus, Wood Lane, London W12 0BZ, U.K.
- National
Lung and Heart Institute, Imperial College London, Du Cane Road, London W12 0NN, U.K.
| | - Ying Ge
- National
Lung and Heart Institute, Imperial College London, Du Cane Road, London W12 0NN, U.K.
| | - Justin C. Mason
- National
Lung and Heart Institute, Imperial College London, Du Cane Road, London W12 0NN, U.K.
| | - Joseph J. Boyle
- National
Lung and Heart Institute, Imperial College London, Du Cane Road, London W12 0NN, U.K.
| | - Nicholas J. Long
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, White City Campus, Wood Lane, London W12 0BZ, U.K.
| |
Collapse
|
49
|
de la Fuente IF, Sawant SS, Tolentino MQ, Corrigan PM, Rouge JL. Viral Mimicry as a Design Template for Nucleic Acid Nanocarriers. Front Chem 2021; 9:613209. [PMID: 33777893 PMCID: PMC7987652 DOI: 10.3389/fchem.2021.613209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Therapeutic nucleic acids hold immense potential in combating undruggable, gene-based diseases owing to their high programmability and relative ease of synthesis. While the delivery of this class of therapeutics has successfully entered the clinical setting, extrahepatic targeting, endosomal escape efficiency, and subcellular localization. On the other hand, viruses serve as natural carriers of nucleic acids and have acquired a plethora of structures and mechanisms that confer remarkable transfection efficiency. Thus, understanding the structure and mechanism of viruses can guide the design of synthetic nucleic acid vectors. This review revisits relevant structural and mechanistic features of viruses as design considerations for efficient nucleic acid delivery systems. This article explores how viral ligand display and a metastable structure are central to the molecular mechanisms of attachment, entry, and viral genome release. For comparison, accounted for are details on the design and intracellular fate of existing nucleic acid carriers and nanostructures that share similar and essential features to viruses. The review, thus, highlights unifying themes of viruses and nucleic acid delivery systems such as genome protection, target specificity, and controlled release. Sophisticated viral mechanisms that are yet to be exploited in oligonucleotide delivery are also identified as they could further the development of next-generation nonviral nucleic acid vectors.
Collapse
Affiliation(s)
| | | | | | | | - Jessica L. Rouge
- Department of Chemistry, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
50
|
Shao Y, Zhao J, Yuan J, Zhao Y, Li L. Organelle‐Specific Photoactivation of DNA Nanosensors for Precise Profiling of Subcellular Enzymatic Activity. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202016738] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Yulei Shao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100149 China
- Department of Chemistry Tsinghua University Beijing 100084 China
| | - Jian Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100149 China
| | - Jinying Yuan
- Department of Chemistry Tsinghua University Beijing 100084 China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100149 China
| |
Collapse
|