51
|
Lointier M, Dussouillez C, Glattard E, Kichler A, Bechinger B. Different Biological Activities of Histidine-Rich Peptides Are Favored by Variations in Their Design. Toxins (Basel) 2021; 13:363. [PMID: 34065185 PMCID: PMC8160934 DOI: 10.3390/toxins13050363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
The protein transduction and antimicrobial activities of histidine-rich designer peptides were investigated as a function of their sequence and compared to gene transfection, lentivirus transduction and calcein release activities. In membrane environments, the peptides adopt helical conformations where the positioning of the histidine side chains defines a hydrophilic angle when viewed as helical wheel. The transfection of DNA correlates with calcein release in biophysical experiments, being best for small hydrophilic angles supporting a model where lysis of the endosomal membrane is the limiting factor. In contrast, antimicrobial activities show an inverse correlation suggesting that other interactions and mechanisms dominate within the bacterial system. Furthermore, other derivatives control the lentiviral transduction enhancement or the transport of proteins into the cells. Here, we tested the transport into human cell lines of luciferase (63 kDa) and the ribosome-inactivating toxin saporin (30 kDa). Notably, depending on the protein, different peptide sequences are required for the best results, suggesting that the interactions are manifold and complex. As such, designed LAH4 peptides assure a large panel of biological and biophysical activities whereby the optimal result can be tuned by the physico-chemical properties of the sequences.
Collapse
Affiliation(s)
- Morane Lointier
- Université de Strasbourg, CNRS, UMR7177, Institut de Chimie, 4, Rue Blaise Pascal, 67070 Strasbourg, France; (M.L.); (E.G.)
| | - Candice Dussouillez
- Laboratoire de Conception et Application de Molécules Bioactives, UMR7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France;
| | - Elise Glattard
- Université de Strasbourg, CNRS, UMR7177, Institut de Chimie, 4, Rue Blaise Pascal, 67070 Strasbourg, France; (M.L.); (E.G.)
| | - Antoine Kichler
- Laboratoire de Conception et Application de Molécules Bioactives, UMR7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France;
| | - Burkhard Bechinger
- Université de Strasbourg, CNRS, UMR7177, Institut de Chimie, 4, Rue Blaise Pascal, 67070 Strasbourg, France; (M.L.); (E.G.)
- Institut Universitaire de France, 75005 Paris, France
| |
Collapse
|
52
|
Abeyratne-Perera HK, Basu S, Chandran PL. Shells of compacted DNA as nanocontainers transporting proteins in multiplexed delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112184. [PMID: 34225845 DOI: 10.1016/j.msec.2021.112184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022]
Abstract
Polyethyleneimine (PEI) polymers are known to compact DNA strands into spheroid, toroid, or rod structures. A formulation with mannose-grafted PEI (PEIm), however, was reported to compact DNA into ~100 nm spheroids that indented like thin-walled pressurized shells. The goal of the study is to understand why mannose bristles divert the traditional pathway of PEI-DNA compaction to produce shell-like structures, and to manipulate the process so that proteins can be packed into the core of the assembling shells for co-delivering DNA and proteins into cells. DLS, AFM, and TEM imaging provide a consistent picture that BSA proteins can be packed into the shells without altering the shell architecture, as long as the proteins were added during the time course of shell assembly. Force spectroscopy studies reveal that DNA shells that buckle also have a rich surface-coating of mannose, indicating that a micelle-like partitioning of hydrophobic and hydrophilic layers governs shell assembly. When HEK293T cells are spiked with BSA-laden DNA shells, co-transfection of DNA and BSA is observed at higher levels than control formulations. Distinct micron-sized features appear having both green fluorescence from BSA-FITC and blue fluorescence from NucBlue DNA stain, suggesting BSA release in nucleus and secretory granules. With DNA nanocontainers, proteins can take advantage of the efficiency of PEI-based DNA transfection for hitchhiking into cells while being shielded from the challenges of the intracellular route. DNA nanocontainers are rapid to assemble, not dependent on the DNA sequence, and can be adapted for different protein types; thereby having potential to serve as a high-throughput platform in scenarios where DNA and protein have to be released at the same site and time within cells (e.g., theranostics, multiplexed co-delivery, gene editing).
Collapse
Affiliation(s)
- Hashanthi K Abeyratne-Perera
- Biochemistry and Molecular Biology Department, College of Medicine, Howard University, Washington, DC, United States of America
| | - Saswati Basu
- Chemical Engineering Department, College of Engineering and Architecture, Howard University, Washington, DC, United States of America
| | - Preethi L Chandran
- Biochemistry and Molecular Biology Department, College of Medicine, Howard University, Washington, DC, United States of America; Chemical Engineering Department, College of Engineering and Architecture, Howard University, Washington, DC, United States of America.
| |
Collapse
|
53
|
Luther DC, Jeon T, Goswami R, Nagaraj H, Kim D, Lee YW, Rotello VM. Protein Delivery: If Your GFP (or Other Small Protein) Is in the Cytosol, It Will Also Be in the Nucleus. Bioconjug Chem 2021; 32:891-896. [PMID: 33872490 PMCID: PMC8508718 DOI: 10.1021/acs.bioconjchem.1c00103] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intracellular protein delivery is a transformative tool for biologics research and medicine. Delivery into the cytosol allows proteins to diffuse throughout the cell and access subcellular organelles. Inefficient delivery caused by endosomal entrapment is often misidentified as cytosolic delivery. This inaccuracy muddles what should be a key checkpoint in assessing delivery efficiency. Green fluorescent protein (GFP) is a robust cargo small enough to passively diffuse from the cytosol into the nucleus. Fluorescence of GFP in the nucleus is a direct readout for cytosolic access and effective delivery. Here, we highlight recent examples from the literature for the accurate assessment of cytosolic protein delivery using GFP fluorescence in the cytosol and nucleus.
Collapse
Affiliation(s)
- David C. Luther
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
- These authors contributed equally
| | - Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, MA 01003, USA
- These authors contributed equally
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Dongkap Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Chemistry, Hanyang University, Seoul 04763, Republic of Korea
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
54
|
Zhu T, Shi L, Ma C, Xu L, Yang J, Zhou G, Zhu X, Shen L. Fluorinated chitosan-mediated intracellular catalase delivery for enhanced photodynamic therapy of oral cancer. Biomater Sci 2021; 9:658-662. [PMID: 33463639 DOI: 10.1039/d0bm01898h] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A pH-responsive fluorinated chitosan-chlorin e6 (FC-Ce6) was employed here for the intracellular delivery of catalase to relieve the hypoxic micro-environment. Upon simple mixing, FC-Ce6 and catalase co-assemble to form stable nanoparticles, which show a greatly improved cross-membrane penetration capacity compared with catalase alone or nonfluorinated CS-Ce6/catalase nanoparticles. Under catalase catalysis, a high concentration of intracellular H2O2 can be transformed into O2. Upon irradiation, due to the continuous formation of cytotoxic singlet oxygen (1O2), our nanoparticles showed superior anti-cancer activity in contrast to free Ce6 and nonfluorinated CS-Ce6/catalase nanoparticles. Our study proposes an effective intracellular catalase delivery system to overcome hypoxia for enhanced PDT against oral cancer.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| | - Leilei Shi
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chuan Ma
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Li Xu
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiapei Yang
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guoyu Zhou
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lingyue Shen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| |
Collapse
|
55
|
Kretzmann JA, Luther DC, Evans CW, Jeon T, Jerome W, Gopalakrishnan S, Lee YW, Norret M, Iyer KS, Rotello VM. Regulation of Proteins to the Cytosol Using Delivery Systems with Engineered Polymer Architecture. J Am Chem Soc 2021; 143:4758-4765. [PMID: 33705125 PMCID: PMC10613456 DOI: 10.1021/jacs.1c00258] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Intracellular protein delivery enables selective regulation of cellular metabolism, signaling, and development through introduction of defined protein quantities into the cell. Most applications require that the delivered protein has access to the cytosol, either for protein activity or as a gateway to other organelles such as the nucleus. The vast majority of delivery vehicles employ an endosomal pathway however, and efficient release of entrapped protein cargo from the endosome remains a challenge. Recent research has made significant advances toward efficient cytosolic delivery of proteins using polymers, but the influence of polymer architecture on protein delivery is yet to be investigated. Here, we developed a family of dendronized polymers that enable systematic alterations of charge density and structure. We demonstrate that while modulation of surface functionality has a significant effect on overall delivery efficiency, the endosomal release rate can be highly regulated by manipulating polymer architecture. Notably, we show that large, multivalent structures cause slower sustained release, while rigid spherical structures result in rapid burst release.
Collapse
Affiliation(s)
- Jessica A. Kretzmann
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - David C. Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Cameron W. Evans
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road., Amherst, MA 01003, USA
| | - William Jerome
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Marck Norret
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - K. Swaminathan Iyer
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| |
Collapse
|
56
|
Bonadio A, Shifman JM. Computational design and experimental optimization of protein binders with prospects for biomedical applications. Protein Eng Des Sel 2021; 34:gzab020. [PMID: 34436606 PMCID: PMC8388154 DOI: 10.1093/protein/gzab020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/11/2021] [Accepted: 07/11/2021] [Indexed: 11/12/2022] Open
Abstract
Protein-based binders have become increasingly more attractive candidates for drug and imaging agent development. Such binders could be evolved from a number of different scaffolds, including antibodies, natural protein effectors and unrelated small protein domains of different geometries. While both computational and experimental approaches could be utilized for protein binder engineering, in this review we focus on various computational approaches for protein binder design and demonstrate how experimental selection could be applied to subsequently optimize computationally-designed molecules. Recent studies report a number of designed protein binders with pM affinities and high specificities for their targets. These binders usually characterized with high stability, solubility, and low production cost. Such attractive molecules are bound to become more common in various biotechnological and biomedical applications in the near future.
Collapse
Affiliation(s)
- Alessandro Bonadio
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
57
|
Kim GC, Cheon DH, Lee Y. Challenge to overcome current limitations of cell-penetrating peptides. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140604. [PMID: 33453413 DOI: 10.1016/j.bbapap.2021.140604] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022]
Abstract
The penetration of biological membranes is a prime obstacle for the delivery of pharmaceutical drugs. Cell-penetrating peptide (CPP) is an efficient vehicle that can deliver various cargos across the biological membranes. Since the discovery, CPPs have been rigorously studied to unveil the underlying penetrating mechanism as well as to exploit CPPs for various biomedical applications. This review will focus on the various strategies to overcome current limitations regarding stability, selectivity, and efficacy of CPPs.
Collapse
Affiliation(s)
- Gyu Chan Kim
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
58
|
Andrian T, Riera R, Pujals S, Albertazzi L. Nanoscopy for endosomal escape quantification. NANOSCALE ADVANCES 2021; 3:10-23. [PMID: 36131870 PMCID: PMC9419860 DOI: 10.1039/d0na00454e] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/26/2020] [Indexed: 05/04/2023]
Abstract
The successful cytosolic delivery of nanoparticles is hampered by their endosomal entrapment and degradation. To push forward the smart development of nanoparticles we must reliably detect and quantify their endosomal escape process. However, the current methods employed are not quantitative enough at the nanoscale to achieve this. Nanoscopy is a rapidly evolving field that has developed a diverse set of powerful techniques in the last two decades, opening the door to explore nanomedicine with an unprecedented resolution and specificity. The understanding of key steps in the drug delivery process - such as endosomal escape - would benefit greatly from the implementation of the most recent advances in microscopy. In this review, we provide the latest insights into endosomal escape of nanoparticles obtained by nanoscopy, and we discuss the features that would allow these techniques to make a great impact in the field.
Collapse
Affiliation(s)
- Teodora Andrian
- Nanoscopy for Nanomedicine, Institute for Bioengineering of Catalonia Barcelona Spain
| | - Roger Riera
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology Eindhoven Netherlands
| | - Silvia Pujals
- Nanoscopy for Nanomedicine, Institute for Bioengineering of Catalonia Barcelona Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, Universitat de Barcelona Av. Diagonal 647 08028 Barcelona Spain
| | - Lorenzo Albertazzi
- Nanoscopy for Nanomedicine, Institute for Bioengineering of Catalonia Barcelona Spain
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology Eindhoven Netherlands
| |
Collapse
|
59
|
Freitag F, Wagner E. Optimizing synthetic nucleic acid and protein nanocarriers: The chemical evolution approach. Adv Drug Deliv Rev 2021; 168:30-54. [PMID: 32246984 DOI: 10.1016/j.addr.2020.03.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
Optimizing synthetic nanocarriers is like searching for a needle in a haystack. How to find the most suitable carrier for intracellular delivery of a specified macromolecular nanoagent for a given disease target location? Here, we review different synthetic 'chemical evolution' strategies that have been pursued. Libraries of nanocarriers have been generated either by unbiased combinatorial chemistry or by variation and novel combination of known functional delivery elements. As in natural evolution, definition of nanocarriers as sequences, as barcode or design principle, may fuel chemical evolution. Screening in appropriate test system may not only provide delivery candidates, but also a refined understanding of cellular delivery including novel, unpredictable mechanisms. Combined with rational design and computational algorithms, candidates can be further optimized in subsequent evolution cycles into nanocarriers with improved safety and efficacy. Optimization of nanocarriers differs for various cargos, as illustrated for plasmid DNA, siRNA, mRNA, proteins, or genome-editing nucleases.
Collapse
|
60
|
Zhang S, Shen J, Li D, Cheng Y. Strategies in the delivery of Cas9 ribonucleoprotein for CRISPR/Cas9 genome editing. Theranostics 2021; 11:614-648. [PMID: 33391496 PMCID: PMC7738854 DOI: 10.7150/thno.47007] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/31/2020] [Indexed: 12/26/2022] Open
Abstract
CRISPR/Cas9 genome editing has gained rapidly increasing attentions in recent years, however, the translation of this biotechnology into therapy has been hindered by efficient delivery of CRISPR/Cas9 materials into target cells. Direct delivery of CRISPR/Cas9 system as a ribonucleoprotein (RNP) complex consisting of Cas9 protein and single guide RNA (sgRNA) has emerged as a powerful and widespread method for genome editing due to its advantages of transient genome editing and reduced off-target effects. In this review, we summarized the current Cas9 RNP delivery systems including physical approaches and synthetic carriers. The mechanisms and beneficial roles of these strategies in intracellular Cas9 RNP delivery were reviewed. Examples in the development of stimuli-responsive and targeted carriers for RNP delivery are highlighted. Finally, the challenges of current Cas9 RNP delivery systems and perspectives in rational design of next generation materials for this promising field will be discussed.
Collapse
Affiliation(s)
- Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jiangtao Shen
- The Second People's Hospital of Taizhou affiliated to Yangzhou University, Taizhou, 225500, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| |
Collapse
|
61
|
Lv J, Wang C, Li H, Li Z, Fan Q, Zhang Y, Li Y, Wang H, Cheng Y. Bifunctional and Bioreducible Dendrimer Bearing a Fluoroalkyl Tail for Efficient Protein Delivery Both In Vitro and In Vivo. NANO LETTERS 2020; 20:8600-8607. [PMID: 33155820 DOI: 10.1021/acs.nanolett.0c03287] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Rational design of stimuli-responsive polymers for cytosolic protein delivery with robust efficiency is of great importance but remains a challenging task. Here, we reported a bioreducible and amphiphilic dendrimer bearing a fluoroalkyl tail for this purpose. The fluorolipid was conjugated to the focal point of a cysteamine-cored polyamidoamine dendrimer via disulfide bond, while phenylboronic acid moieties were decorated on dendrimer surface for efficient protein binding. The yielding polymer showed high protein binding capability and complex stability and could efficiently release the cargo proteins in a glutathione-responsive manner. The designed polymer was effective in the delivery of various membrane-impermeable proteins into living cells with reserved bioactivities. In addition, the polymer efficiently delivered a toxin protein saporin into 4T1 breast cancer cells and inhibited the tumor growth in vivo after intravenous injection. The developed polymer in this study is a promising vector for the delivery of membrane-impermeable proteins to treat various diseases.
Collapse
Affiliation(s)
- Jia Lv
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Changping Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Hongru Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhan Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qianqian Fan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
62
|
Zhang P, Zhang Y, Ding X, Shen W, Li M, Wagner E, Xiao C, Chen X. A Multistage Cooperative Nanoplatform Enables Intracellular Co-Delivery of Proteins and Chemotherapeutics for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000013. [PMID: 33035385 DOI: 10.1002/adma.202000013] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/18/2020] [Indexed: 06/11/2023]
Abstract
Combining intracellularly active proteins with chemotherapeutics represents a promising strategy for synergistic cancer therapy. However, the lack of nanocarrier systems for delivery into cancer cells and controlled intracellular release of both physicochemically very distinct cargos significantly impedes the biomedical translation of this combination strategy in cancer therapy. Here, a well-designed triblock copolymer, mPEG-b-PGCA-b-PGTA, is reported for application in a multistage cooperative drug delivery nanoplatform that accomplishes effective intracellular co-delivery of hydrophilic ribonuclease A (RNase A) and hydrophobic doxorubicin (DOX). RNase A bioreversibly modified with phenylboronic acid groups via a ROS-cleavable carbamate linker is incorporated into the triblock copolymer nanoparticles with high efficiency through a pH-reversible phenylboronic acid-catechol linkage. The reversible covalent conjugations between RNase A and the triblock copolymer endow the nanoparticles with high stability under normal physiological conditions. Upon cellular internalization, the cooperative release of DOX and RNase A from the triblock copolymer nanoparticles is triggered at multiple stages by endosomal acidic environment and subsequent DOX-enhanced intracellular ROS environment. This leads to enhanced synergistic anticancer effects as demonstrated both in vitro and in vivo. Given the versatility of dynamic covalent conjugations, this work provides a universal and stable platform for intracellular co-delivery of various combinations of proteins and chemotherapeutics.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xiaoya Ding
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Wei Shen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University, Munich, 81377, Germany
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
63
|
Goswami R, Jeon T, Nagaraj H, Zhai S, Rotello VM. Accessing Intracellular Targets through Nanocarrier-Mediated Cytosolic Protein Delivery. Trends Pharmacol Sci 2020; 41:743-754. [PMID: 32891429 PMCID: PMC7502523 DOI: 10.1016/j.tips.2020.08.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022]
Abstract
Protein-based therapeutics have unique therapeutic potential due to their specificity, potency, and low toxicity. The vast majority of intracellular applications of proteins require access to the cytosol. Direct entry to the cytosol is challenging due to the impermeability of the cell membrane to proteins. As a result, multiple strategies have focused on endocytic uptake of proteins. Endosomally entrapped cargo, however, can have very low escape efficiency, with protein degradation occurring in acidic endolysosomal compartments. In this review, we briefly discuss endosomal escape strategies and review the strategy of cell membrane fusion, a recent strategy for direct delivery of proteins into the cell cytoplasm.
Collapse
Affiliation(s)
- Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
64
|
Nathamgari SSP, Pathak N, Lemaitre V, Mukherjee P, Muldoon JJ, Peng CY, McGuire T, Leonard JN, Kessler JA, Espinosa HD. Nanofountain Probe Electroporation Enables Versatile Single-Cell Intracellular Delivery and Investigation of Postpulse Electropore Dynamics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002616. [PMID: 33006271 PMCID: PMC7646188 DOI: 10.1002/smll.202002616] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/27/2020] [Indexed: 05/13/2023]
Abstract
Introducing exogenous molecules into cells with high efficiency and dosage control is a crucial step in basic research as well as clinical applications. Here, the capability of the nanofountain probe electroporation (NFP-E) system to deliver proteins and plasmids in a variety of continuous and primary cell types with appropriate dosage control is reported. It is shown that the NFP-E can achieve fine control over the relative expression of two cotransfected plasmids. Finally, the dynamics of electropore closure after the pulsing ends with the NFP-E is investigated. Localized electroporation has recently been utilized to demonstrate the converse process of delivery (sampling), in which a small volume of the cytosol is retrieved during electroporation without causing cell lysis. Single-cell temporal sampling confers the benefit of monitoring the same cell over time and can provide valuable insights into the mechanisms underlying processes such as stem cell differentiation and disease progression. NFP-E parameters that maximize the membrane resealing time, which is essential for increasing the sampled volume and in meeting the challenge of monitoring low copy number biomarkers, are identified. Its application in CRISPR/Cas9 gene editing, stem cell reprogramming, and single-cell sampling studies is envisioned.
Collapse
Affiliation(s)
- Samba Shiva Prasad Nathamgari
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | | | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Joseph J Muldoon
- Department of Chemical and Biological Engineering and Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA
| | - Chian-Yu Peng
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Tammy McGuire
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering and Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - John A Kessler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Horacio Dante Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
65
|
Ranjan K, Waghela BN, Vaidya FU, Pathak C. Cell-Penetrable Peptide-Conjugated FADD Induces Apoptosis and Regulates Inflammatory Signaling in Cancer Cells. Int J Mol Sci 2020; 21:ijms21186890. [PMID: 32961826 PMCID: PMC7555701 DOI: 10.3390/ijms21186890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Dysregulated expression of Fas-associated death domain (FADD) is associated with the impediment of various cellular pathways, including apoptosis and inflammation. The adequate cytosolic expression of FADD is critical to the regulation of cancer cell proliferation. Importantly, cancer cells devise mechanisms to suppress FADD expression and, in turn, escape from apoptosis signaling. Formulating strategies, for direct delivery of FADD proteins into cancer cells in a controlled manner, may represent a promising therapeutic approach in cancer therapy. We chemically conjugated purified FADD protein with cell permeable TAT (transactivator of transcription) peptide, to deliver in cancer cells. TAT-conjugated FADD protein internalized through the caveolar pathway of endocytosis and retained in the cytosol to augment cell death. Inside cancer cells, TAT-FADD rapidly constituted DISC (death inducing signaling complex) assembly, which in turn, instigate apoptosis signaling. The apoptotic competency of TAT-FADD showed comparable outcomes with the conventional apoptosis inducers. Notably, TAT-FADD mitigates constitutive NF-κB activation and associated downstream anti-apoptotic genes Bcl2, cFLIPL, RIP1, and cIAP2, independent of pro-cancerous TNF-α priming. In cancer cells, TAT-FADD suppresses the canonical NLRP3 inflammasome priming and restricts the processing and secretion of proinflammatory IL-1β. Our results demonstrate that TAT-mediated intracellular delivery of FADD protein can potentially recite apoptosis signaling with simultaneous regulation of anti-apoptotic and proinflammatory NF-κB signaling activation in cancer cells.
Collapse
|
66
|
Chen K, Pei D. Engineering Cell-Permeable Proteins through Insertion of Cell-Penetrating Motifs into Surface Loops. ACS Chem Biol 2020; 15:2568-2576. [PMID: 32786266 DOI: 10.1021/acschembio.0c00593] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Effective delivery of proteins into the cytosol of mammalian cells would open the door to a wide range of applications. However, despite great efforts from numerous investigators, effective protein delivery in a clinical setting is yet to be accomplished. Herein we report a potentially general approach to engineering cell-permeable proteins by genetically grafting a short cell-penetrating peptide (CPP) to an exposed loop of a protein of interest. The grafted peptide is conformationally constrained, exhibiting enhanced proteolytic stability and cellular entry efficiency. Applying this technique to enhanced green fluorescent protein (EGFP), protein-tyrosine phosphatase 1B (PTP1B), and purine nucleoside phosphorylase (PNP) rendered all three proteins cell-permeable and biologically active in cellular assays. When added into growth medium at 0.5-5 μM concentrations, the engineered PTP1B dose-dependently reduced the phosphotyrosine levels of intracellular proteins, while the modified PNP corrected the metabolic deficiency of PNP-deficient mouse T lymphocytes, providing a potential enzyme replacement therapy for a rare genetic disease.
Collapse
Affiliation(s)
- Kuangyu Chen
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
67
|
He W, Xing X, Wang X, Wu D, Wu W, Guo J, Mitragotri S. Nanocarrier‐Mediated Cytosolic Delivery of Biopharmaceuticals. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910566] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/16/2020] [Indexed: 01/04/2025]
Abstract
AbstractBiopharmaceuticals have emerged to play a vital role in disease treatment and have shown promise in the rapidly expanding pharmaceutical market due to their high specificity and potency. However, the delivery of these biologics is hindered by various physiological barriers, owing primarily to the poor cell membrane permeability, low stability, and increased size of biologic agents. Since many biological drugs are intended to function by interacting with intracellular targets, their delivery to intracellular targets is of high relevance. In this review, the authors summarize and discuss the use of nanocarriers for intracellular delivery of biopharmaceuticals via endosomal escape and, especially, the routes of direct cytosolic delivery by means including the caveolae‐mediated pathway, contact release, intermembrane transfer, membrane fusion, direct translocation, and membrane disruption. Strategies with high potential for translation are highlighted. Finally, the authors conclude with the clinical translation of promising carriers and future perspectives.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xuyang Xing
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xiaoling Wang
- School of Biomass Science and Engineering Sichuan University Chengdu 610065 China
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of Ministry of Education of China School of Pharmacy Fudan University Shanghai 201203 China
| | - Junling Guo
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
68
|
Niamsuphap S, Fercher C, Kumble S, Huda P, Mahler SM, Howard CB. Targeting the undruggable: emerging technologies in antibody delivery against intracellular targets. Expert Opin Drug Deliv 2020; 17:1189-1211. [DOI: 10.1080/17425247.2020.1781088] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Suchada Niamsuphap
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
| | - Christian Fercher
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
- ARC Centre of Excellence in Convergent BioNano Science and Technology, AIBN, University of Queensland, Brisbane, Australia
| | - Sumukh Kumble
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
| | - Pie Huda
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
- Centre for Advanced Imaging (CAI), University of Queensland, Brisbane, Australia
| | - Stephen M Mahler
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
- Centre for Personalised Nanomedicine, AIBN, University of Queensland, Brisbane, Australia
| |
Collapse
|
69
|
Honda Y, Nomoto T, Matsui M, Takemoto H, Kaihara Y, Miura Y, Nishiyama N. Sequential Self-Assembly Using Tannic Acid and Phenylboronic Acid-Modified Copolymers for Potential Protein Delivery. Biomacromolecules 2020; 21:3826-3835. [PMID: 32786730 DOI: 10.1021/acs.biomac.0c00903] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tannic acid (TA) can form stable complexes with proteins, attracting significant attention as protein delivery systems. However, its systemic application has been limited due to nonspecific interaction. Here, we report a simple technique to prepare systemically applicable protein delivery systems using sequential self-assembly of a protein, TA, and phenylboronic acid-conjugated PEG-poly(amino acid) block copolymers in aqueous solution. Mixing the protein and TA in aqueous solution led to covering of the protein with TA, and subsequent addition of the copolymer resulted in the formation of boronate esters between TA and copolymers, constructing the core-shell-type ternary complex. The ternary complex covered with PEG exhibited a small hydrodynamic diameter of ∼10-20 nm and prevented an unfavorable interaction with serum components, thereby accomplishing significantly prolonged blood circulation and enhanced tumor accumulation in a subcutaneous tumor model. The technique utilizing supramolecular self-assembly may serve as a novel approach for designing protein delivery systems.
Collapse
Affiliation(s)
- Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yuka Kaihara
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
70
|
Luther DC, Huang R, Jeon T, Zhang X, Lee YW, Nagaraj H, Rotello VM. Delivery of drugs, proteins, and nucleic acids using inorganic nanoparticles. Adv Drug Deliv Rev 2020; 156:188-213. [PMID: 32610061 PMCID: PMC8559718 DOI: 10.1016/j.addr.2020.06.020] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 01/03/2023]
Abstract
Inorganic nanoparticles provide multipurpose platforms for a broad range of delivery applications. Intrinsic nanoscopic properties provide access to unique magnetic and optical properties. Equally importantly, the structural and functional diversity of gold, silica, iron oxide, and lanthanide-based nanocarriers provide unrivalled control of nanostructural properties for effective transport of therapeutic cargos, overcoming biobarriers on the cellular and organismal level. Taken together, inorganic nanoparticles provide a key addition to the arsenal of delivery vectors for fighting disease and improving human health.
Collapse
Affiliation(s)
- David C Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA.
| |
Collapse
|
71
|
Guo L, Zhang Y, Wei R, Zhang X, Wang C, Feng M. Proinflammatory macrophage-derived microvesicles exhibit tumor tropism dependent on CCL2/CCR2 signaling axis and promote drug delivery via SNARE-mediated membrane fusion. Theranostics 2020; 10:6581-6598. [PMID: 32550891 PMCID: PMC7295053 DOI: 10.7150/thno.45528] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Exosome (Exo)-based chemotherapeutic drug delivery systems have been extensively investigated; however, the therapeutic potential of other subtypes of extracellular vesicles (EVs), in particular microvesicles (MiV), seem to be overlooked. Moreover, despite a general agreement on organ tropism of EVs, few studies have clearly demonstrated that EVs specifically target tumor tissue. Methods: Proinflammatory macrophage-derived EV subpopulations comprising apoptotic bodies (ApB), MiV and Exo were isolated under differential ultracentrifugation, and further analyzed using comparative proteomic and lipid approach. Results: On the basis of EV biogenesis pathways, our data demonstrated that MiV acquire the tumor-targeting capacity probably through inheritance of CCR2-enriched cell membrane which also drives the recruitment of donor cells to tumor sites. Further, our data validate MiV utilize SNARE-mediated membrane fusion to directly discharge doxorubicin to nucleus and bypass endocytic degradation. Conclusions: Compared with other EV subtypes, MiV loaded with doxorubicin gain significant benefits in chemotherapeutic outcomes including survival rate improvements in metastatic ovarian cancer. Therefore, MiV represent a potent alterative to Exo and synthetic liposomes (Lipo) for tumor-targeting drug delivery.
Collapse
Affiliation(s)
- Ling Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Ye Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Runxiu Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Xiaochen Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| |
Collapse
|
72
|
Harguindey A, Culver HR, Sinha J, Bowman CN, Cha JN. Efficient cellular uptake of click nucleic acid modified proteins. Chem Commun (Camb) 2020; 56:4820-4823. [PMID: 32236172 DOI: 10.1039/c9cc09401f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Efficient intracellular delivery of biomacromolecules such as proteins continues to remain a challenge despite its potential for medicine. In this work, we show that mScarlet, a non cytotoxic red fluorescent protein (RFP) conjugated to Click Nucleic Acid (CNA), a synthetic analog of DNA, undergo cell uptake significantly more than either native proteins or proteins conjugated with similar amounts of DNA in MDA-MB-468 cells. We further demonstrate that the process of cell uptake is metabolically driven and that scavenger receptors and caveolae mediated endocytosis play a significant role. Co-localization studies using anti-scavenger receptor antibodies suggest that scavenger receptors are implicated in the mechanism of uptake of CNA modified proteins.
Collapse
Affiliation(s)
- Albert Harguindey
- Department of Chemical and Biological University of Colorado, Boulder, USA.
| | | | | | | | | |
Collapse
|
73
|
Methods for protein delivery into cells: from current approaches to future perspectives. Biochem Soc Trans 2020; 48:357-365. [DOI: 10.1042/bst20190039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
The manipulation of cultured mammalian cells by the delivery of exogenous macromolecules is one of the cornerstones of experimental cell biology. Although the transfection of cells with DNA expressions constructs that encode proteins is routine and simple to perform, the direct delivery of proteins into cells has many advantages. For example, proteins can be chemically modified, assembled into defined complexes and subject to biophysical analyses prior to their delivery into cells. Here, we review new approaches to the injection and electroporation of proteins into cultured cells. In particular, we focus on how recent developments in nanoscale injection probes and localized electroporation devices enable proteins to be delivered whilst minimizing cellular damage. Moreover, we discuss how nanopore sensing may ultimately enable the quantification of protein delivery at single-molecule resolution.
Collapse
|
74
|
Pan S, Jeon T, Luther DC, Duan X, Rotello VM. Cytosolic Delivery of Functional Proteins In Vitro through Tunable Gigahertz Acoustics. ACS APPLIED MATERIALS & INTERFACES 2020; 12:15823-15829. [PMID: 32150373 PMCID: PMC7392053 DOI: 10.1021/acsami.9b21131] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Intracellular delivery is essential to therapeutic applications such as genome engineering and disease diagnosis. Current methods lack simple, noninvasive strategies and are often hindered by long incubation time or high toxicity. Hydrodynamic approaches offer rapid and controllable delivery of small molecules, but thus far have not been demonstrated for delivering functional proteins. In this work, we developed a robust hydrodynamic approach based on gigahertz (GHz) acoustics to achieve rapid and noninvasive cytosolic delivery of biologically active proteins. With this method, GHz-based acoustic devices trigger oscillations through a liquid medium (acoustic streaming), generating shear stress on the cell membrane and inducing transient nanoporation. This mechanical effect enhances membrane permeability and enables cytosolic access to cationic proteins without disturbing their bioactivity. We evaluated the versatility of this approach through the delivery of cationic fluorescent proteins to a range of cell lines, all of which displayed equally efficient delivery speed (≤20 min). Delivery of multiple enzymatically active proteins with functionality related to apoptosis or genetic recombination further demonstrated the relevance of this method.
Collapse
Affiliation(s)
- Shuting Pan
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- State Key Laboratory of Precision Measuring Technology & Instruments, College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, United States
| | - David C. Luther
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology & Instruments, College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Corresponding Author, . Tel./Fax: +86 2227401002 (X.D.)
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Corresponding Author, . Tel./Fax: +86 2227401002 (X.D.)
| |
Collapse
|
75
|
Lee YW, Luther DC, Goswami R, Jeon T, Clark V, Elia J, Gopalakrishnan S, Rotello VM. Direct Cytosolic Delivery of Proteins through Coengineering of Proteins and Polymeric Delivery Vehicles. J Am Chem Soc 2020; 142:4349-4355. [PMID: 32049533 PMCID: PMC7392052 DOI: 10.1021/jacs.9b12759] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanocarrier-mediated protein delivery is a promising strategy for fundamental research and therapeutic applications. However, the efficacy of the current platforms for delivery into cells is limited by endosomal entrapment of delivered protein cargo with concomitantly inefficient access to the cytosol and other organelles, including the nucleus. We report here a robust, versatile polymeric-protein nanocomposite (PPNC) platform capable of efficient (≥90%) delivery of proteins to the cytosol. We synthesized a library of guanidinium-functionalized poly(oxanorborneneimide) (PONI) homopolymers with varying molecular weights to stabilize and deliver engineered proteins featuring terminal oligoglutamate "E-tags". The polymers were screened for cytosolic delivery efficiency using imaging flow cytometry with cytosolic delivery validated using confocal microscopy and activity of the delivered proteins demonstrated through functional assays. These studies indicate that the PPNC platform provides highly effective and tunable cytosolic delivery over a wide range of formulations, making them robust agents for therapeutic protein delivery.
Collapse
Affiliation(s)
- Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - David C. Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent Clark
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - James Elia
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
76
|
Ren L, Lv J, Wang H, Cheng Y. A Coordinative Dendrimer Achieves Excellent Efficiency in Cytosolic Protein and Peptide Delivery. Angew Chem Int Ed Engl 2020; 59:4711-4719. [DOI: 10.1002/anie.201914970] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Lanfang Ren
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| |
Collapse
|
77
|
Ren L, Lv J, Wang H, Cheng Y. A Coordinative Dendrimer Achieves Excellent Efficiency in Cytosolic Protein and Peptide Delivery. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lanfang Ren
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| |
Collapse
|
78
|
Recent advances in physiologically based pharmacokinetic and pharmacodynamic models for anticancer nanomedicines. Arch Pharm Res 2020; 43:80-99. [PMID: 31975317 DOI: 10.1007/s12272-020-01209-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
Nanoparticles (NPs) have distinct pharmacokinetic (PK) properties and can potentially improve the absorption, distribution, metabolism, and elimination (ADME) of small-molecule drugs loaded therein. Owing to the unwanted toxicities of anticancer agents in healthy organs and tissues, their precise delivery to the tumor is an essential requirement. There have been numerous advancements in the development of nanomedicines for cancer therapy. Physiologically based PK (PBPK) models serve as excellent tools for describing and predicting the ADME properties and the efficacy and toxicity of drugs, in combination with pharmacodynamic (PD) models. The recent preliminary application of these modeling approaches to NPs demonstrated their potential benefits in research and development processes relevant to the ADME and pharmacodynamics of NPs and nanomedicines. Here, we comprehensively review the pharmacokinetics of NPs, the developed PBPK models for anticancer NPs, and the developed PD model for anticancer agents.
Collapse
|
79
|
Zhang S, Cheng Y. Boronic acid-engineered gold nanoparticles for cytosolic protein delivery. Biomater Sci 2020; 8:3741-3750. [DOI: 10.1039/d0bm00679c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Boronic acid-engineered gold nanoparticles for effective cytosolic protein delivery with the help of hypertonicity.
Collapse
Affiliation(s)
- Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology
- School of Molecular Science and Engineering
- South China University of Technology
- Guangzhou 510640
- China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology
- School of Molecular Science and Engineering
- South China University of Technology
- Guangzhou 510640
- China
| |
Collapse
|
80
|
Wen R, Zhang AH, Liu D, Feng J, Yang J, Xia D, Wang J, Li C, Zhang T, Hu N, Hang T, He G, Xie X. Intracellular Delivery and Sensing System Based on Electroplated Conductive Nanostraw Arrays. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43936-43948. [PMID: 31696695 DOI: 10.1021/acsami.9b15619] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One-dimensional nanoneedle-like arrays have emerged as an attractive tool for penetrating the cell membrane to achieve intracellular applications including drug delivery, electrical recording, and biochemical detection. Hollow nanoneedles, also called nanostraws (NSs), combined with nanoelectroporation have been demonstrated as a powerful platform for intracellular drug delivery and extraction of intracellular contents. However, the fabrication technique of nanostraws still requires complicated and expensive atomic layer deposition and etching processes and fails to produce conductive nanostraws. Herein, we developed a commonly accessible and versatile electrodeposition approach to controllably fabricate conductive nanostraw arrays based on various types of metal or conductive polymer materials. Representatively, Pt nanostraws (Pt NSs) with 400 nm diameter were further integrated with a low-voltage nanoelectroporation system to achieve cell detection, intracellular drug delivery, and sensing of intracellular enzymes. Both theoretical simulations and experimental results revealed that the conductive nanostraws in direct contact with cells could induce high-efficiency cell electroporation at relatively low voltage (∼5 V). Efficient delivery of reagents into live cells with spatial control and repeated extraction of intracellular enzymes (e.g., caspase-3) for temporal monitoring from the same set of cells were demonstrated. This work not only pioneers a new avenue for universal production of conductive nanostraws on a large scale but also presents great potential for developing nanodevices to achieve a variety of biomedical applications including cell re-engineering, cell-based therapy, and signaling pathway monitoring.
Collapse
Affiliation(s)
- Rui Wen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Ai-Hua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Di Liu
- Pritzker School of Medicine , University of Chicago , Chicago , Illinois 60637 , United States
| | - Jianming Feng
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Jiang Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-sen University Cancer Center , Guangzhou 510060 , China
| | - Dehua Xia
- School of Environmental Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Ji Wang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Chunwei Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Tao Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Tian Hang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| |
Collapse
|
81
|
Chen X, Zheng G, Cheng J, Yang YY. Supramolecular Nanotheranostics. Am J Cancer Res 2019; 9:3014-3016. [PMID: 31244939 PMCID: PMC6567977 DOI: 10.7150/thno.36788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/09/2023] Open
Abstract
This supramolecular nanotheranostics special issue collected a total of 17 review articles and 3 research articles broadly covering the current and emerging supramolecular nanotheranostics.
Collapse
|