1
|
Zhang J, Deng M, Xu C, Li D, Yan X, Gu Y, Zhong M, Gao H, Liu Y, Zhang J, Qu X, Zhang J. Dual-Prodrug-Based Hyaluronic Acid Nanoplatform Provides Cascade-Boosted Drug Delivery for Oxidative Stress-Enhanced Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50459-50473. [PMID: 39258403 DOI: 10.1021/acsami.4c11662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Insufficient drug accumulation in tumors severely limits the antitumor efficiency of hyaluronic acid (HA) nanomedicine in solid tumors due to superficial penetration depth, low cell uptake, and nonspecific drug release. Hence, we constructed a dual NO prodrug (alkynyl-JSK) and doxorubicin prodrug (cis-DOX)-conjugated HA nanoparticle (HA-DOX-JSK NPs), which achieved cascade-boosted drug delivery efficiency based on a relay strategy of NO-mediated deep tumor penetration─HA target CD44 tumor cell uptake─tumor microenvironment (TME)-responsive drug release. The nanoparticle demonstrated sustained and locoregionally GSH/GST-triggered NO release and GSH/pH-responsive DOX release in the tumor. The released NO first mediated collagen degradation, causing deep tumor penetration of nanoparticles in the dense extracellular matrix. Immediately, HA was relayed to enhance CD44-targeted tumor cell uptake, and then, the nanoparticles were finally triggered by specific TME to release DOX and NO in the deep tumor. Relying on the relayed delivery strategy, a significant improvement of DOX accumulation in tumors was realized. Moreover, NO depleted GSH-induced intracellular reactive oxygen species, enhancing DOX chemotherapy. Based on this strategy, the tumor inhibition rate in breast cancer was up to 87.8% in vivo. The relay drug-delivery HA system would greatly cascade-boost drug accumulation in deep tumors for efficient solid tumor therapy.
Collapse
Affiliation(s)
- Junxian Zhang
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Meigui Deng
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Chang Xu
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Danting Li
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Xiaozhe Yan
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Yuxuan Gu
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Meihui Zhong
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Hui Gao
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Yingchun Liu
- Jinghua Plastics Industry Company Limited, Langfang 065800, P. R. China
| | - Jiqing Zhang
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250000, China
| | - Xiongwei Qu
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Jimin Zhang
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| |
Collapse
|
2
|
Lee JS, Park E, Oh H, Choi WI, Koo H. Levan nanoparticles with intrinsic CD44-targeting ability for tumor-targeted drug delivery. Int J Biol Macromol 2023; 234:123634. [PMID: 36773871 DOI: 10.1016/j.ijbiomac.2023.123634] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
Existing anticancer therapeutics exhibit short half-lives, non-specificity, and severe side effects. To address this, active-targeting nanoparticles have been developed; however, the complex fabrication procedures, scale-up, and low reproducibility delay FDA approval, particularly for functionalized nanoparticles. We developed levan nanoparticles via simple one-pot nanoprecipitation for specific anticancer drug delivery. Levan is a plant polysaccharide which has a binding affinity to CD44 receptors and amphiphilicity. The nanoparticles are self-assembled and enable active-targeting without chemical modifications. The paclitaxel-loaded levan nanoparticles (PTX@LevNP) demonstrated a sustained PTX release and long-term stability. The LevNP can bind CD44 receptors on cancer cells, and PTX@LevNP showed enhanced anticancer activity in CD44-positive cells (SCC7 cells). In SCC7 tumor-bearing mice, the accumulation of LevNP in tumor tissue was 3.7 times higher than that of the free-dye, resulting in improved anticancer efficacy of PTX@LevNP. This new strategy using levan can produce nanoparticles for effective cancer treatment without complex fabrication procedures.
Collapse
Affiliation(s)
- Jin Sil Lee
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123, Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Eunyoung Park
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Hyeryeon Oh
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123, Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
3
|
He J, Pang W, Gu B, Lin X, Ye J. The stiffness-dependent tumor cell internalization of liquid metal nanoparticles. NANOSCALE 2022; 14:16902-16917. [PMID: 36342434 DOI: 10.1039/d2nr04293b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The properties of nanoparticle (NP) carriers, such as size, shape and surface state, have been proven to dramatically affect their uptake by tumor cells, thereby influencing and determining the effect of nanomedicine on tumor theranostics. However, the effect of the stiffness of NPs on their cellular internalization remains unclear, especially for circumstances involving active or passive NP targeting. In this work, we constructed eutectic gallium indium liquid metal NPs with the same particle size, shape and surface charge properties but distinct stiffness via tailoring the surface oxidation and silica coating. It has been found that the softer NPs would be endocytosed much slower than their stiffer counterparts in the presence of specific ligand-receptor interaction. Interestingly, once the interaction is eliminated, softer NPs are internalized faster than the stiffer ones. Based on experimental observations and theoretical verification, we demonstrate that this phenomenon is mainly caused by varying degrees of deformation of soft NPs induced by ligand-receptor interactions. Such a finding of the stiffness effect of NPs implies great potential for fundamental biomedical applications, such as the rational design of nanomedicines.
Collapse
Affiliation(s)
- Jing He
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China.
| | - Wen Pang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China.
| | - Bobo Gu
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China.
| | - Xubo Lin
- Institute of Single Cell Engineering, Key Laboratory of Ministry of Education for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, P. R. China
| | - Jian Ye
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China.
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| |
Collapse
|
4
|
Liang Y, Ding R, Wang H, Liu L, He J, Tao Y, Zhao Z, Zhang J, Wang A, Sun K, Li Y, Shi Y. Orally administered intelligent self-ablating nanoparticles: a new approach to improve drug cellular uptake and intestinal absorption. Drug Deliv 2022; 29:305-315. [PMID: 35037529 PMCID: PMC8765251 DOI: 10.1080/10717544.2021.2023704] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Oral drug delivery to treat diabetes is being increasingly researched. The mucus and the epithelial cell layers hinder drug delivery. We designed a self-ablating nanoparticle to achieve smart oral delivery to overcome the gastrointestinal barrier. We used the zwitterionic dilauroyl phosphatidylcholine, which exhibits a high affinity toward Oligopeptide transporter 1, to modify poly(lactic-co-glycolic acid) nanoparticles and load hemagglutinin-2 peptide to facilitate its escape from lysosomes. Nanoparticles exhibit a core–shell structure, the lipid layer is degraded by the lysosomes when the nanoparticles are captured by lysosomes, then the inner core of the nanoparticles gets exposed. The results revealed that the self-ablating nanoparticles exhibited higher encapsulation ability than the self-assembled nanoparticles (77% vs 64%) and with better stability. Quantitative cellular uptake, cellular uptake mechanisms, and trans-monolayer cellular were studied, and the results revealed that the cellular uptake achieved using the self-ablating nanoparticles was higher than self-assembling nanoparticles, and the number of uptake pathways via which the self-ablating nanoparticles functioned were higher than the self-assembling nanoparticles. Intestinal mucus permeation, in vivo intestinal circulation, was studied, and the results revealed that the small self-assembling nanoparticles exhibit a good extent of intestinal uptake in the presence of mucus. In vitro flip-flop, intestinal circulation revealed that the uptake of the self-ablating nanoparticles was 1.20 times higher than the self-assembled nanoparticles. Pharmacokinetic study and the pharmacodynamic study showed that the bioavailability and hypoglycemic effect of self-ablating nanoparticles were better than self-assembled nanoparticles.
Collapse
Affiliation(s)
- Yanzi Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Ruihuan Ding
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Huihui Wang
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Lanze Liu
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Jibiao He
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Yuping Tao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Zhenyu Zhao
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Jie Zhang
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Aiping Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, P. R. China
| | - Yanan Shi
- School of Life Science, Yantai University, Yantai, P. R. China
| |
Collapse
|
5
|
Yan K, Feng Y, Gao K, Shi X, Zhao X. Fabrication of hyaluronic acid-based micelles with glutathione-responsiveness for targeted anticancer drug delivery. J Colloid Interface Sci 2022; 606:1586-1596. [PMID: 34500160 DOI: 10.1016/j.jcis.2021.08.129] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022]
Abstract
Hyaluronic acid (HA), a natural polymer, has gained much attention recently because of its good biocompatibility and extensive availability. Herein, a novel drug delivery system based on hyaluronic acid-tetraphenyl ethylene conjugate (HA-SS-TPE) with glutathione (GSH)-responsiveness for targeted drug delivery is designed. During the self-assembly of HA-SS-TPE, doxorubicin (DOX) is loaded to form DOX-loaded polymeric micelles. These as-prepared DOX-loaded polymeric micelles not only exhibit fluorescent emission, but also fast glutathione-triggered dissociation to unload DOX by responding to tumor microenvironments. In-vitro investigations showed that the DOX-loaded polymeric micelles presented a higher intracellular release ratio in CD44-positive cells (ES2 and Hela) than in CD44-negative cells (MCF-7 and L929). Notably, in vivo investigations showed that DOX@HA-SS-TPE significantly suppressed tumor growth. As a result, such a GSH-responsive drug delivery system with fluorescent feature provides a potential treatment for CD44-overexpressing cancers.
Collapse
Affiliation(s)
- Ke Yan
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Yecheng Feng
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Ke Gao
- Laboratory Animal Center, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of medical science, Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Xiaojing Shi
- Laboratory Animal Center, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of medical science, Zhengzhou University, Zhengzhou 450052, P. R. China.
| | - Xubo Zhao
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, P. R. China.
| |
Collapse
|
6
|
Moradi P, Hasanzadeh A, Radmanesh F, Rajai Daryasarei S, Hosseini ES, Kiani J, Shahbazi A, Nourizadeh H, Eslami M, Dorgalaleh A, Sahlolbei M, Hamblin MR, Karimi M. Smart arginine-equipped polycationic nanoparticles for p/CRISPR delivery into cells. NANOTECHNOLOGY 2021; 33:075104. [PMID: 34727527 DOI: 10.1088/1361-6528/ac357a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/02/2021] [Indexed: 06/13/2023]
Abstract
An efficient and safe delivery system for the transfection of CRISPR plasmid (p/CRISPR) into target cells can open new avenues for the treatment of various diseases. Herein, we design a novel nonvehicle by integrating an arginine-disulfide linker with low-molecular-weight PEI (PEI1.8k) for the delivery of p/CRISPR. These PEI1.8k-Arg nanoparticles facilitate the plasmid release and improve both membrane permeability and nuclear localization, thereby exhibiting higher transfection efficiency compared to native PEI1.8kin the delivery of nanocomplexes composed of PEI1.8k-Arg and p/CRISPR into conventional cells (HEK 293T). This nanovehicle is also able to transfect p/CRISPR in a wide variety of cells, including hard-to-transfect primary cells (HUVECs), cancer cells (HeLa), and neuronal cells (PC-12) with nearly 5-10 times higher efficiency compared to the polymeric gold standard transfection agent. Furthermore, the PEI1.8k-Arg nanoparticles can edit the GFP gene in the HEK 293T-GFP reporter cell line by delivering all possible forms of CRISPR/Cas9 system (e.g. plasmid encoding Cas9 and sgRNA targeting GFP, and Cas9/sgRNA ribonucleoproteins (RNPs) as well as Cas9 expression plasmid andin vitro-prepared sgRNA) into HEK 293T-GFP cells. The successful delivery of p/CRISPR into local brain tissue is also another remarkable capability of these nanoparticles. In view of all the exceptional benefits of this safe nanocarrier, it is expected to break new ground in the field of gene editing, particularly for therapeutic purposes.
Collapse
Affiliation(s)
- Pardis Moradi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Akbar Hasanzadeh
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Radmanesh
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saideh Rajai Daryasarei
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Sadat Hosseini
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Helena Nourizadeh
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Eslami
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Akbar Dorgalaleh
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sahlolbei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| |
Collapse
|
7
|
Hashemzadeh I, Hasanzadeh A, Radmanesh F, Khodadadi Chegeni B, Hosseini ES, Kiani J, Shahbazi A, Naseri M, Fatahi Y, Nourizadeh H, Kheiri Yeghaneh Azar B, Aref AR, Liu Y, Hamblin MR, Karimi M. Polyethylenimine-Functionalized Carbon Dots for Delivery of CRISPR/Cas9 Complexes. ACS APPLIED BIO MATERIALS 2021; 4:7979-7992. [DOI: 10.1021/acsabm.1c00890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Iman Hashemzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Fatemeh Radmanesh
- Uro-oncology Research Center, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Beheshteh Khodadadi Chegeni
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Elaheh Sadat Hosseini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, 1417613151, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Helena Nourizadeh
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Student Research Committee, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Amir R. Aref
- Belfer Center for Applied Cancer Science, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yong Liu
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Oujiang Laboratory, Wenzhou, Zhejiang 325000, China
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| |
Collapse
|
8
|
A core-shell nanoplatform as a nonviral vector for targeted delivery of genes to the retina. Acta Biomater 2021; 134:605-620. [PMID: 34329781 DOI: 10.1016/j.actbio.2021.07.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 01/05/2023]
Abstract
Retinal diseases, including age-related macular degeneration (AMD), are a major cause of blindness. Efficient delivery of therapeutic genes to retinal cells to treat retinal disease is a formidable challenge. In this study, we developed a core-shell nanoplatform composed of a core and two external layers for targeted delivery of the gene to the retina. The inner core was composed of amino acid-functionalized dendrimers and a nuclear localization signal (NLS) for DNA complexation, nuclear transport and efficient transfection. The inner core was coated in a lipid bilayer that comprised pH-sensitive lipids as the inner shell layer. Hyaluronic acid (HA)-1,2-dioleoylphosphatidylethanolamine (DOPE) as the outermost shell layer was used for retinal cell targeting. This core-shell nanoplatform was developed so that the mobility in the vitreous body of these negatively charged carriers would not be affected by their surface charge, allowing diffusion into the retina, uptake into the retinal cells via CD44-mediated internalization, and finally transport into the nucleus by the NLS. The designed nanoparticles showed safety both in vitro and in vivo and inhibited the expression of VEGF under hypoxia-mimicking conditions. In vitro angiogenesis assays exhibited significant inhibitory effects on cell migration and tube formation. The in vivo assays indicated that this nanoplatform could be delivered to the retina. Taken together, this nanoplatform has the potential to transfer gene material into the retina for the treatment of retinal diseases, including AMD. STATEMENT OF SIGNIFICANCE: It remains a challenge to develop an efficient nonviral vector for gene therapy, especially retinal gene therapy. Various barriers exist in gene delivery and the unique ocular environment, making gene delivery to the retina difficult. In this study, we designed a negatively charged core-shell nanoplatform (HD-NPPND) for the targeted delivery of gene to the retina. The developed nanoplatform possessed excellent transfection efficiency and safety both in vitro and in vivo. It efficiently delivered a gene to the retina. The results of this study suggested that this core-shell nanoplatform has the potential to deliver genes to the retina to treat retinal diseases, including age-related macular degeneration (AMD).
Collapse
|
9
|
Zhou J, Ma S, Zhang Y, He Y, Mao H, Yang J, Zhang H, Luo K, Gong Q, Gu Z. Bacterium-mimicking sequentially targeted therapeutic nanocomplexes based on O-carboxymethyl chitosan and their cooperative therapy by dual-modality light manipulation. Carbohydr Polym 2021; 264:118030. [PMID: 33910720 DOI: 10.1016/j.carbpol.2021.118030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
An integrated gene nanovector capable of overcoming complicated physiological barriers in one vector is desirable to circumvent the challenges imposed by the intricate tumor microenvironment. Herein, a nuclear localization signals (NLS)-decorated element and an iRGD-functionalized element based on O-carboxymethyl chitosan were synthesized, mixed, and coated onto PEI/DNA to fabricate bacterium-mimicking sequentially targeted therapeutic nanocomplexes (STNPs) which were internalized through receptor-mediated endocytosis and other pathways and achieved nuclear translocation of DNA. The endo/lysosomal membrane disruption triggered by reactive oxygen species (ROS) after short-time illumination, together with the DNA nuclear translocation, evoked an enhanced gene expression. Alternatively, the excessive ROS from long-time irradiation induced apoptosis in tumor cells, bringing about greater anti-tumor efficacy owing to the integration of gene and photodynamic therapy. Overall, these results demonstrated bacterium-mimicking STNPs could be a potential candidate for tumor treatments.
Collapse
Affiliation(s)
- Jie Zhou
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Shengnan Ma
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yuxin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, PR China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China; Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| |
Collapse
|
10
|
Karakoçak B, Laradji A, Primeau T, Berezin MY, Li S, Ravi N. Hyaluronan-Conjugated Carbon Quantum Dots for Bioimaging Use. ACS APPLIED MATERIALS & INTERFACES 2021; 13:277-286. [PMID: 33355448 PMCID: PMC8243741 DOI: 10.1021/acsami.0c20088] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 05/28/2023]
Abstract
This work demonstrates the application of hyaluronan-conjugated nitrogen-doped carbon quantum dots (HA-nCQDs) for bioimaging of tumor cells and illustrates their potential use as carriers in targeted drug delivery. Quantum dots are challenging to deliver with specificity, which hinders their application. To facilitate targeted internalization by cancer cells, hyaluronic acid, a natural ligand of CD44 receptors, was covalently grafted on nCQDs. The HA-nCQD conjugate was synthesized by carbodiimide coupling of the amine moieties on nCQDs and the carboxylic acids on HA chains. Conjugated HA-nCQD retained sufficient fluorescence, although with 30% lower quantum efficiency than the original nCQDs. Confocal microscopy showed enhanced internalization of HA-nCQDs, facilitated by CD44 receptors. To demonstrate the specificity of HA-nCQDs toward human tumor cells, patient-derived breast cancer tissue with high-CD44 expression was implanted in adult mice. The tumors were allowed to grow up to 200-250 mm3 prior to the injection of HA-nCQDs. With either local or systemic injection, we achieved a high level of tumor specificity judged by a strong signal-to-noise ratio between the tumor and the surrounding tissue in vivo. Overall, the results show that HA-nCQDs can be used for imaging of CD44-specific tumors in preclinical models of human cancer and potentially used as carriers for targeted drug delivery into CD44-rich cells.
Collapse
Affiliation(s)
- Bedia
Begüm Karakoçak
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| | - Amine Laradji
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| | - Tina Primeau
- Department
of Medicine, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Mikhail Y. Berezin
- Department
of Radiology, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Shunqiang Li
- Department
of Medicine, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Nathan Ravi
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| |
Collapse
|
11
|
Lee E, Park J, Youn YS, Oh KT, Kim D, Lee ES. Alendronate/cRGD-Decorated Ultrafine Hyaluronate Dot Targeting Bone Metastasis. Biomedicines 2020; 8:E492. [PMID: 33187133 PMCID: PMC7696888 DOI: 10.3390/biomedicines8110492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022] Open
Abstract
In this study, we report the hyaluronate dot (dHA) with multiligand targeting ability and a photosensitizing antitumor model drug for treating metastatic bone tumors. Here, the dHA was chemically conjugated with alendronate (ALN, as a specific ligand to bone), cyclic arginine-glycine-aspartic acid (cRGD, as a specific ligand to tumor integrin αvβ3), and photosensitizing chlorin e6 (Ce6, for photodynamic tumor therapy), denoted as (ALN/cRGD)@dHA-Ce6. These dots thus prepared (≈10 nm in diameter) enabled extensive cellular interactions such as hyaluronate (HA)-mediated CD44 receptor binding, ALN-mediated bone targeting, and cRGD-mediated tumor integrin αvβ3 binding, thus improving their tumor targeting efficiency, especially for metastasized MDA-MB-231 tumors. As a result, these dots improved the tumor targeting efficiency and tumor cell permeability in a metastatic in vivo tumor model. Indeed, we demonstrated that (ALN/cRGD)@dHA-Ce6 considerably increased photodynamic tumor ablation, the extent of which is superior to that of the tumor ablation of dot systems with single or double ligands. These results indicate that dHA with multiligand can provide an effective treatment strategy for metastatic bone tumors.
Collapse
Affiliation(s)
- Eunsol Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea; (E.L.); (J.P.)
| | - Jaeduk Park
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea; (E.L.); (J.P.)
| | - Yu Seok Youn
- School of Pharmacy, SungKyunKwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea;
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 221 Heukseok dong, Dongjak-gu, Seoul 06974, Korea;
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N Stonewall Ave, Oklahoma City, OK 73117, USA;
| | - Eun Seong Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea; (E.L.); (J.P.)
- Department of Biomedical Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea
| |
Collapse
|
12
|
Tooyserkani R, Lipiński W, Willemsen B, Löwik DWPM. Activation of cell-penetrating peptide fragments by disulfide formation. Amino Acids 2020; 52:1161-1168. [PMID: 32737661 PMCID: PMC7497323 DOI: 10.1007/s00726-020-02880-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/26/2020] [Indexed: 11/30/2022]
Abstract
Three cell-penetrating peptides (CPPs), Tat, Pep-3 and penetratin, were split into two parts and each fragment was terminated with a cysteine residue, to allow disulfide bridge formation, as well as a fluorescent label, for visualization and quantitative analysis. After disulfide formation between two complementary CPP fragments, cellular uptake of the resulting conjugates was observed. As confirmed by in vitro experiments, the conjugated peptides showed uptake activity comparable to the native CPP sequences, while the truncated peptides were hardly active. Until now, this split CPP strategy has only been demonstrated for oligo-arginine CPPs, but here we demonstrate that it is also applicable to other cell-penetrating peptides. This wider applicability may help in the design of new activatable cell-penetrating peptides for, e.g., targeted drug delivery.
Collapse
Affiliation(s)
- Raheleh Tooyserkani
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Wojciech Lipiński
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Bob Willemsen
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Dennis W P M Löwik
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.
| |
Collapse
|
13
|
Yamada Y, Somiya K, Miyauchi A, Osaka H, Harashima H. Validation of a mitochondrial RNA therapeutic strategy using fibroblasts from a Leigh syndrome patient with a mutation in the mitochondrial ND3 gene. Sci Rep 2020; 10:7511. [PMID: 32371897 PMCID: PMC7200808 DOI: 10.1038/s41598-020-64322-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
We report on the validation of a mitochondrial gene therapeutic strategy using fibroblasts from a Leigh syndrome patient by the mitochondrial delivery of therapeutic mRNA. The treatment involves delivering normal ND3 protein-encoding mRNA as a therapeutic RNA to mitochondria of the fibroblasts from a patient with a T10158C mutation in the mtDNA coding the ND3 protein, a component of the mitochondrial respiratory chain complex I. The treatment involved the use of a liposome-based carrier (a MITO-Porter) for delivering therapeutic RNA to mitochondria via membrane fusion. The results confirmed that the mitochondrial transfection of therapeutic RNA by the MITO-Porter system resulted in a decrease in the levels of mutant RNA in mitochondria of diseased cells based on reverse transcription quantitative PCR. An evaluation of mitochondrial respiratory activity by respirometry also showed that transfection using the MITO-Porter resulted in an increase in maximal mitochondrial respiratory activity in the diseased cells.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, 060-0812, Japan.
| | - Kana Somiya
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Akihiko Miyauchi
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo, 060-0812, Japan.
| |
Collapse
|
14
|
Bhatt H, Ghosh B, Biswas S. Cell-Penetrating Peptide and α-Tocopherol-Conjugated Poly(amidoamine) Dendrimers for Improved Delivery and Anticancer Activity of Loaded Paclitaxel. ACS APPLIED BIO MATERIALS 2020; 3:3157-3169. [DOI: 10.1021/acsabm.0c00179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Himanshu Bhatt
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| |
Collapse
|
15
|
Yamazaki M, Iijima K. Fabrication and Characterization of Polysaccharide Composite Films from Polyion Complex Particles. Polymers (Basel) 2020; 12:polym12020435. [PMID: 32069798 PMCID: PMC7077694 DOI: 10.3390/polym12020435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/11/2020] [Indexed: 01/21/2023] Open
Abstract
Biomaterials made of natural polysaccharides have attracted much attention due to the fact of their excellent properties, such as high biocompatibility and biodegradability, and their specific biological functions based on their chemical structures. This study demonstrates that polysaccharide composite films can be fabricated from polyion complexes (PICs) with their particles used as building components. Dispersion of PIC particles prepared by mixing, centrifugation, and re-dispersion of dilute solutions of cationic and anionic polysaccharides were cast, dried, and formed into films several micrometers thick. These films were homogenous and water insoluble. It was revealed that the component anionic polysaccharides affected the film’s properties such as the swelling behavior and mechanical characteristics. Adhesion of NIH3T3 cells (integrin: high, CD44: lack or weak) and A549 cells (integrin: high, CD44: high) to the composite films were examined. Both NIH3T3 and A549 cells adhered to heparin/chitosan (HEP/CHI) film because HEP has an affinity for integrin through fibronectin. However, A549 cells adhered to chondroitin sulfate (CS)/CHI and hyaluronic acid (HYA)/CHI films, whereas NIH3T3 cells did not, because both CS and HYA have affinity for CD44. These results indicated that the biological functions of anionic polysaccharides were maintained on the surface of the composite films. It was also possible to fabricate films composed of three kinds of polysaccharides: one cationic polysaccharide and two kinds of anionic polysaccharides. These results show that the properties of films composed of three kinds of polysaccharides may be controllable depending on the anionic polysaccharide composition rates.
Collapse
Affiliation(s)
- Makoto Yamazaki
- Department of Chemistry, Chemical Engineering and Life Science, College of Engineering Science, Yokohama National University, Tokiwadai 79-5, Hodogaya-ku, Yokohama 240-8501, Japan;
| | - Kazutoshi Iijima
- Department of Chemistry, Chemical Engineering and Life Science, College of Engineering Science, Yokohama National University, Tokiwadai 79-5, Hodogaya-ku, Yokohama 240-8501, Japan;
- Faculty of Engineering, Yokohama National University, Tokiwadai 79-5, Hodogaya-ku, Yokohama 240-8501, Japan
- Correspondence: ; Tel.: +81-45-339-3997
| |
Collapse
|
16
|
Yoo J, Rejinold NS, Lee D, Noh I, Koh WG, Jon S, Kim YC. CD44-Mediated Methotrexate Delivery by Hyaluronan-Coated Nanoparticles Composed of a Branched Cell-Penetrating Peptide. ACS Biomater Sci Eng 2019; 6:494-504. [DOI: 10.1021/acsbiomaterials.9b01724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | | | | |
Collapse
|
17
|
Nakamura T, Yamada Y, Sato Y, Khalil IA, Harashima H. Innovative nanotechnologies for enhancing nucleic acids/gene therapy: Controlling intracellular trafficking to targeted biodistribution. Biomaterials 2019; 218:119329. [DOI: 10.1016/j.biomaterials.2019.119329] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/13/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022]
|
18
|
Zhang Y, Zhang M, Fan X, Mao D, Lv S, Chen P. Effect of STAT3 decoy oligodeoxynucleotides mediated by ultrasound-targeted microbubbles combined with ultrasound on the growth of squamous cell carcinoma of the esophagus. Oncol Lett 2019; 17:2151-2158. [PMID: 30675281 PMCID: PMC6341804 DOI: 10.3892/ol.2018.9814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/12/2018] [Indexed: 01/01/2023] Open
Abstract
Effect of STAT3 decoy oligodeoxynucleotides (ODN) transduced by ultrasound microbubbles combined with ultrasound on the growth of esophageal squamous cell carcinoma and its mechanism were analyzed. EC9706 cells were cultured in vitro and divided into four groups: group E (ultrasound microbubble + ultrasound irradiation), group P (liposome + ultrasound irradiation), group C (ultrasound), and group CC (ultrasound microbubbles). Mutant ODNs were used in groups E and P and the control group was group EC and PC, respectively. Immunofluorescence assay and flow cytometry were used to detect the transfection efficiency of each group. MTT colorimetric assay was performed to analyze the inhibition rate in each group. The effect of STAT3 decoy ODN on the proliferation of esophageal squamous carcinoma cells was calculated. Revese transcription-quantitative PCR (RT-qPCR) and western blotting were performed to detect the expression of the STAT signaling pathway downstream of gene expression levels. The model of subcutaneous transplantation of nude mice was used to show the effect of different transfections and STAT3 decoy ODN on the weight and volume of the transplanted tumor in mice. The cell inhibition rate was higher in group E than in groups P (F=8.382, P<0.001) and CC (F=6.469, P<0.001). Compared with groups EC, PC and C, respectively, the mRNA expression of STAT3, bcl-xL and Cyclin D1 decreased in groups E, P and CC (F=5.328, P<0.001). The weight and volume of nude mice in groups E, P and CC exhibited an inhibitory effect on the weight and volume of nude mice. Ultrasound irradiation combined with ultrasound microbubbles is an effective transfection method. The transfection of STAT3 decoy ODN can significantly inhibit the activity of esophageal squamous cell carcinoma cells and enhance apoptosis of cells, which has potential clinical value.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Interventional Therapy, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Meiwu Zhang
- Department of Interventional Therapy, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaoxiang Fan
- Department of Interventional Therapy, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Dafeng Mao
- Department of Interventional Therapy, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Shuyi Lv
- Department of Interventional Therapy, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Ping Chen
- Department of Gastrointestinal and Minimally Invasive Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
19
|
Wang P, Shi Y, Zhang S, Huang X, Zhang J, Zhang Y, Si W, Dong X. Hydrogen Peroxide Responsive Iron-Based Nanoplatform for Multimodal Imaging-Guided Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1803791. [PMID: 30569479 DOI: 10.1002/smll.201803791] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/01/2018] [Indexed: 05/20/2023]
Abstract
Cancer multimodal phototherapy triggered by hydrogen peroxide has attracted widespread attention as a dominating strategy to increase phototherapeutic efficiency. Herein, a hydrogen peroxide responsive iron oxide nanoplatform, with the diameter of about 50 nm, is fabricated to intracellularly trigger the Fenton reaction and achieve synergistic photodynamic therapy and photothermal therapy. The nanoplatform based on iron oxide nanoparticles is decorated with indocyanine green (ICG, photosensitizer) and hyaluronic acid (HA, targeting molecular) through electrostatic interaction, thus the as-prepared nanoplatform (IONPs-ICG-HA) exhibits excellent active targeting ability and biocompatibility. More importantly, it can effectively utilize the intratumoral overproduced hydrogen peroxide to generate reactive oxygen species for cancer cell killing via intracellular Fenton reactions. In vitro and in vivo experiments reveal that the IONPs-ICG-HA nanocomposites realize effective photoacoustic/photothermal/fluorescence imaging-guided phototherapy, leading to promising hydrogen peroxide responsive cancer theranostics.
Collapse
Affiliation(s)
- Peng Wang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Yunhao Shi
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Shichao Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Xiaoyu Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Jiaojiao Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Yewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| |
Collapse
|
20
|
Gennari A, Rios de la Rosa JM, Hohn E, Pelliccia M, Lallana E, Donno R, Tirella A, Tirelli N. The different ways to chitosan/hyaluronic acid nanoparticles: templated vs direct complexation. Influence of particle preparation on morphology, cell uptake and silencing efficiency. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2019; 10:2594-2608. [PMID: 31976191 PMCID: PMC6964650 DOI: 10.3762/bjnano.10.250] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/02/2019] [Indexed: 05/02/2023]
Abstract
This study is about linking preparative processes of nanoparticles with the morphology of the nanoparticles and with their efficiency in delivering payloads intracellularly. The nanoparticles are composed of hyaluronic acid (HA) and chitosan; the former can address a nanoparticle to cell surface receptors such as CD44, the second allows both for entrapment of nucleic acids and for an endosomolytic activity that facilitates their liberation in the cytoplasm. Here, we have systematically compared nanoparticles prepared either A) through a two-step process based on intermediate (template) particles produced via ionotropic gelation of chitosan with triphosphate (TPP), which are then incubated with HA, or B) through direct polyelectrolyte complexation of chitosan and HA. Here we demonstrate that HA is capable to quantitatively replace TPP in the template process and significant aggregation takes place during the TPP-HA exchange. The templated chitosan/HA nanoparticles therefore have a mildly larger size (measured by dynamic light scattering alone or by field flow fractionation coupled to static or dynamic light scattering), and above all a higher aspect ratio (R g/R H) and a lower fractal dimension. We then compared the kinetics of uptake and the (antiluciferase) siRNA delivery performance in murine RAW 264.7 macrophages and in human HCT-116 colorectal tumor cells. The preparative method (and therefore the internal particle morphology) had little effect on the uptake kinetics and no statistically relevant influence on silencing (templated particles often showing a lower silencing). Cell-specific factors, on the contrary, overwhelmingly determined the efficacy of the carriers, with, e.g., those containing low-MW chitosan performing better in macrophages and those with high-MW chitosan in HCT-116.
Collapse
Affiliation(s)
- Arianna Gennari
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Julio M Rios de la Rosa
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester and Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
- current address: Cambridge Enterprise Limited, University of Cambridge, Hauser Forum, 3 Charles Babbage Rd, Cambridge, CB3 0GT, United Kingdom
| | - Erwin Hohn
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester and Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
- current address: Novartis EBEWE Pharma Ges.m.b.H. Nfg.KG, Lehenau 10a, 5325 Plainfield, Austria
| | - Maria Pelliccia
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
- current address: Orchard Therapeutics plc., 108 Cannon Street, EC4N 6EU London, United Kingdom
| | - Enrique Lallana
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester and Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Roberto Donno
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Annalisa Tirella
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester and Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Nicola Tirelli
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
21
|
Yamada Y, Daikuhara S, Tamura A, Nishida K, Yui N, Harashima H. Enhanced autophagy induction via the mitochondrial delivery of methylated β-cyclodextrin-threaded polyrotaxanes using a MITO-Porter. Chem Commun (Camb) 2019; 55:7203-7206. [DOI: 10.1039/c9cc03272j] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Failure of autophagy induction results in the accumulation of abnormal mitochondria to cause neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo 060-0812
- Japan
| | | | - Atsushi Tamura
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
| | - Kei Nishida
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
| | - Nobuhiko Yui
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
| | | |
Collapse
|
22
|
Lee H, Park H, Noh GJ, Lee ES. pH-responsive hyaluronate-anchored extracellular vesicles to promote tumor-targeted drug delivery. Carbohydr Polym 2018; 202:323-333. [DOI: 10.1016/j.carbpol.2018.08.141] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
|
23
|
Nisakar D, Vij M, Pandey T, Natarajan P, Sharma R, Mishra S, Ganguli M. Deciphering the Role of Chondroitin Sulfate in Increasing the Transfection Efficiency of Amphipathic Peptide-Based Nanocomplexes. ACS Biomater Sci Eng 2018; 5:45-55. [PMID: 33405865 DOI: 10.1021/acsbiomaterials.8b00069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glycosaminoglycans, both cell-surface and exogenous, can interfere with DNA delivery efficiency of nonviral carrier systems. In this work, we report an extensive comparative study to explore the effect of exogenously added chondroitin sulfate on biophysical characteristics, cellular uptake, transfection efficiency, and intracellular trafficking of nanocomplexes formed using primary and secondary amphipathic peptides developed in our laboratory. Our results indicate that the presence of exogenous chondroitin sulfate exhibits differential enhancement in transfection efficiency of the amphipathic peptides depending upon their chemical nature. The enhancement was more pronounced in primary amphipathic peptide-based nanocomplexes as compared to the secondary counterpart. This difference can be attributed to possible alteration of the intracellular entry pathway in addition to increased extracellular stability, less cellular toxicity, and assistance in nuclear accumulation. These results imply potential use of glycosaminoglycans such as chondroitin sulfate to improve the transfection efficiency of primary amphipathic peptides for possible in vivo applications.
Collapse
Affiliation(s)
- Daniel Nisakar
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Manika Vij
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Tanuja Pandey
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Poornemaa Natarajan
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Rajpal Sharma
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| |
Collapse
|
24
|
Rompicharla SVK, Kumari P, Ghosh B, Biswas S. Octa-arginine modified poly(amidoamine) dendrimers for improved delivery and cytotoxic effect of paclitaxel in cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:847-859. [PMID: 29790795 DOI: 10.1080/21691401.2018.1470527] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cell penetrating peptides (CPP) have the ability to penetrate the cell membrane and have been associated with various cargos for their facile intracellular translocation. The current study involves the synthesis of a CPP, octa-arginine (R8)-modified poly(amidoamine) dendrimer of generation 4 (G4), which has additionally been PEGylated and conjugated to the poorly soluble anticancer drug, paclitaxel (PTX). The synthesized dendrimer conjugates were characterized by proton nuclear magnetic resonance (1H-NMR) Spectroscopy and zeta potential measurements and evaluated in vitro in cell monolayers and 3D spheroids. Cellular uptake study in human cervical cancer cell line (HeLa) revealed that R8 modification significantly improved the cell association of conjugates. G4-PTX- polyethylene glycol (PEG)-R8 conjugate demonstrated enhanced cytotoxic potential and higher induction of apoptosis compared to free PTX and G4-PTX-PEG. Further, the penetrability of fluorescently labeled F-G4-PTX-PEG-R8 was evaluated in 3D spheroids of HeLa at various depths by using confocal microscopy. G4-PTX-PEG-R8 induced cell death and inhibited the growth in 3D spheroids as competently as in monolayers. The enhanced intracellular translocation of R8-modified dendrimers resulted in improved anticancer efficacy of PTX. Therefore, the newly developed dendrimer system is efficient for the intracellular delivery of PTX in cancer cells and has a strong potential to be utilized as an effective chemotherapeutic agent for cancer.
Collapse
Affiliation(s)
- Sri Vishnu Kiran Rompicharla
- a Department of Pharmacy , Birla Institute of Technology & Science-Pilani - Hyderabad Campus , Hyderabad , India
| | - Preeti Kumari
- a Department of Pharmacy , Birla Institute of Technology & Science-Pilani - Hyderabad Campus , Hyderabad , India
| | - Balaram Ghosh
- a Department of Pharmacy , Birla Institute of Technology & Science-Pilani - Hyderabad Campus , Hyderabad , India
| | - Swati Biswas
- a Department of Pharmacy , Birla Institute of Technology & Science-Pilani - Hyderabad Campus , Hyderabad , India
| |
Collapse
|
25
|
Zhou J, Mohamed Wali AR, Ma S, He Y, Yue D, Tang JZ, Gu Z. Tailoring the Supramolecular Structure of Guanidinylated Pullulan toward Enhanced Genetic Photodynamic Therapy. Biomacromolecules 2018; 19:2214-2226. [PMID: 29689167 DOI: 10.1021/acs.biomac.8b00273] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the progress of designing a gene carrier system, what is urgently needed is a balance of excellent safety and satisfactory efficiency. Herein, a straightforward and versatile synthesis of a cationic guanidine-decorated dendronized pullulan (OGG3P) for efficient genetic photodynamic therapy was proposed. OGG3P was able to block the mobility of DNA from a weight ratio of 2. However, G3P lacking guanidine residues could not block DNA migration until at a weight ratio of 15, revealing guanidination could facilitate DNA condensation via specific guanidinium-phosphate interactions. A zeta potential plateau (∼+23 mV) of OGG3P complexes indicated the nonionic hydrophilic hydroxyl groups in pullulan might neutralize the excessive detrimental cationic charges. There was no obvious cytotoxicity and hemolysis, but also enhancement of transfection efficiency with regard to OGG3P in comparison with that of native G3P in Hela and HEK293T cells. More importantly, we found that the uptake efficiency in Hela cells between OGG3P and G3P complexes was not markedly different. However, guanidination caused changes in uptake pathway and led to macropinocytosis pathway, which may be a crucial reason for improved transfection efficiency. After introducing a therapeutic pKillerRed-mem plasmid, OGG3P complexes achieved significantly enhanced KillerRed protein expression and ROS production under irradiation. ROS-induced cancer cells proliferation suppression was also confirmed. This study highlights the guanidine-decorated dendronized pullulan could emerge as a reliable nonviral gene carrier to specifically deliver therapeutic genes.
Collapse
Affiliation(s)
- Jie Zhou
- National Engineering Research Center for Biomaterials , Sichuan University , No. 29, Wangjiang Road , Chengdu 610065 , Sichuan , People's Republic of China
| | - Aisha Roshan Mohamed Wali
- School of Pharmacy, Faculty of Science and Engineering , University of Wolverhampton , Wulfruna Street , Wolverhampton WV1 1LY , United Kingdom
| | - Shengnan Ma
- National Engineering Research Center for Biomaterials , Sichuan University , No. 29, Wangjiang Road , Chengdu 610065 , Sichuan , People's Republic of China
| | - Yiyan He
- College of Materials Science and Engineering , Nanjing Tech University , 30 South Puzhu Road , Nanjing 211816 , People's Republic of China
| | - Dong Yue
- National Engineering Research Center for Biomaterials , Sichuan University , No. 29, Wangjiang Road , Chengdu 610065 , Sichuan , People's Republic of China
| | - James Zhenggui Tang
- School of Pharmacy, Faculty of Science and Engineering , University of Wolverhampton , Wulfruna Street , Wolverhampton WV1 1LY , United Kingdom
| | - Zhongwei Gu
- National Engineering Research Center for Biomaterials , Sichuan University , No. 29, Wangjiang Road , Chengdu 610065 , Sichuan , People's Republic of China.,College of Materials Science and Engineering , Nanjing Tech University , 30 South Puzhu Road , Nanjing 211816 , People's Republic of China
| |
Collapse
|
26
|
Elamin KM, Yamashita Y, Higashi T, Motoyama K, Arima H. Supramolecular Complex of Methyl-β-cyclodextrin with Adamantane-Grafted Hyaluronic Acid as a Novel Antitumor Agent. Chem Pharm Bull (Tokyo) 2017; 66:277-285. [PMID: 29269686 DOI: 10.1248/cpb.c17-00824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Methyl-β-cyclodextrin (M-β-CyD) exhibits cytotoxic activity, and has the potentials as an antitumor agent. However, a tumor-selectivity of M-β-CyD is low, leading to low antitumor activity and the adverse effects. Meanwhile, hyaluronic acid (HA) is known as a promising tumor targeting ligand, because various cancer cells overexpress CD44, a HA-binding glycoprotein. In the present study, to develop a tumor-selective delivery system for M-β-CyD, we designed a supramolecular complex of M-β-CyD with adamantane-grafted HA (Ad-HA/M-β-CyD) and evaluated it as a tumor-selective antitumor agent. M-β-CyD formed a stable complex with Ad-HA (Kc>104 M-1). In addition, Ad-HA/M-β-CyD formed slightly a negative-charged nanoparticle with ca. 140 nm of a particle size, indicating the favorable physicochemical properties for antitumor agents. Ad-HA/M-β-CyD showed the superior cytotoxic activity via CD44-mediated endosomal pathways in HCT116 cells (CD44(+)), a human colon cancer cell line. In addition, cytotoxic activity of Ad-HA/M-β-CyD was induced by apoptosis. These results suggest that Ad-HA/M-β-CyD has the potentials as a tumor-selective supramolecular antitumor agent.
Collapse
Affiliation(s)
- Khaled Mohamed Elamin
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Yamashita
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Taishi Higashi
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Keiichi Motoyama
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Hidetoshi Arima
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University.,Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program," Kumamoto University
| |
Collapse
|
27
|
Dual-targeting nanoparticles with excellent gene transfection efficiency for gene therapy of peritoneal metastasis of colorectal cancer. Oncotarget 2017; 8:89837-89847. [PMID: 29163792 PMCID: PMC5685713 DOI: 10.18632/oncotarget.21159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/26/2017] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer has been one of the most common cancers in the worldwide. Poor patient compliance and serious side effects often associated with conventional therapy (e.g. surgery, radiation, and chemotherapy). Gene therapy may be an alternative strategy. Herein, we developed a dual-targeting nanoparticle with excellent gene transfection efficiency for gene therapy of peritoneal metastasis of colorectal cancer. This nanoparticle can facilitate efficient cellular uptake and promote penetration into nucleus. Meanwhile, this nanoparticle mediated efficient gene transfection in medium with or without serum, which significantly surpassed that of commercial transfection reagents, Lipofectamine 2000 and Lipofectamine 3000. After systemic administration, this nanoparticle loaded with hTRAIL plasmid significantly inhibited peritoneal metastasis of colorectal cancer in vivo. In conclusion, this dual-targeting nanoparticle has great potential to be a gene delivery vector for colorectal cancer therapy.
Collapse
|
28
|
Lallana E, Rios de la Rosa JM, Tirella A, Pelliccia M, Gennari A, Stratford IJ, Puri S, Ashford M, Tirelli N. Chitosan/Hyaluronic Acid Nanoparticles: Rational Design Revisited for RNA Delivery. Mol Pharm 2017; 14:2422-2436. [PMID: 28597662 DOI: 10.1021/acs.molpharmaceut.7b00320] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chitosan/hyaluronic acid (HA) nanoparticles can be used to deliver an RNA/DNA cargo to cells overexpressing HA receptors such as CD44. For these systems, unequivocal links have not been established yet between chitosan macromolecular (molecular weight; degree of deacetylation, i.e., charge density) and nanoparticle variables (complexation strength, i.e., stability; nucleic acid protection; internalization rate) on one hand, and transfection efficiency on the other hand. Here, we have focused on the role of avidity on transfection efficiency in the CD44-expressing HCT-116 as a cellular model; we have employed two differently sized payloads (a large luciferase-encoding mRNA and a much smaller anti-Luc siRNA), and a small library of chitosans (variable molecular weight and degree of deactylation). The RNA avidity for chitosan showed-as expected-an inverse relationship: higher avidity-higher polyplex stability-lower transfection efficiency. The avidity of chitosan for RNA appears to lead to opposite effects: higher avidity-higher polyplex stability but also higher transfection efficiency. Surprisingly, the best transfecting particles were those with the lowest propensity for RNA release, although this might be a misleading relationship: for example, the same macromolecular parameters that increase avidity can also boost chitosan's endosomolytic activity, with a strong enhancement in transfection. The performance of these nonviral vectors appears therefore difficult to predict simply on the basis of carrier- or payload-related variables, and a more holistic consideration of the journey of the nanoparticle, from cell uptake to cytosolic bioavailability of payload, is needed. It is also noteworthy that the nanoparticles used in this study showed optimal performance under slightly acidic conditions (pH 6.4), which is promising for applications in a tumoral extracellular environment. It is also worth pointing out that under these conditions we have for the first time successfully delivered mRNA with chitosan/HA nanoparticles.
Collapse
Affiliation(s)
- Enrique Lallana
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Julio M Rios de la Rosa
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Annalisa Tirella
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Maria Pelliccia
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Arianna Gennari
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Ian J Stratford
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Sanyogitta Puri
- Innovative Medicines-Pharmaceutical Sciences, AstraZeneca , Silk Road Business Park, Macclesfield, SK10 2NA, United Kingdom
| | - Marianne Ashford
- Innovative Medicines-Pharmaceutical Sciences, AstraZeneca , Silk Road Business Park, Macclesfield, SK10 2NA, United Kingdom
| | - Nicola Tirelli
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester , Oxford Road, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
29
|
Xie F, Zhang L, Peng J, Li C, Pu J, Xu Y, Du Z. Hepatic Carcinoma Selective Nucleic Acid Nanovector Assembled by Endogenous Molecules Based on Modular Strategy. Mol Pharm 2017; 14:1841-1851. [DOI: 10.1021/acs.molpharmaceut.6b00709] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Fang Xie
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Luchen Zhang
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jinliang Peng
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Chong Li
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Pu
- School
of Medicine, Shanghai Jiao Tong University, 280 Chongqing South Road, Shanghai 200127, China
| | - Yuhong Xu
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zixiu Du
- School
of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
30
|
Li L, Li X, Wu Y, Song L, Yang X, He T, Wang N, Yang S, Zeng Y, Wu Q, Qian Z, Wei Y, Gong C. Multifunctional Nucleus-targeting Nanoparticles with Ultra-high Gene Transfection Efficiency for In Vivo Gene Therapy. Theranostics 2017; 7:1633-1649. [PMID: 28529641 PMCID: PMC5436517 DOI: 10.7150/thno.17588] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/14/2017] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cell-like cells (CSCL) are responsible for tumor recurrence associated with conventional therapy (e.g. surgery, radiation, and chemotherapy). Here, we developed a novel multifunctional nucleus-targeting nanoparticle-based gene delivery system which is capable of targeting and eradicating CSCL. These nanoparticles can facilitate efficient endosomal escape and spontaneously penetrate into nucleus without additional nuclear localization signal. They also induced extremely high gene transfection efficiency (>95%) even in culture medium containing 30% serum, which significantly surpassed that of some commercial transfection reagents, such as Lipofectamine 2000 and Lipofectamine 3000 etc. Especially, when loaded with the TRAIL gene, this system mediated remarkable depletion of CSCL. Upon systemic administration, the nanoparticles accumulated in tumor sites while sparing the non-cancer tissues and significantly inhibited the growth of tumors with no evident systemic toxicity. Taken together, our results suggest that these novel multifunctional, nucleus-targeting nanoparticles are a very promising in vivo gene delivery system capable of targeting CSCL and represent a new treatment candidate for improving the survival of cancer patients.
Collapse
|
31
|
Kuang H, Ku SH, Kokkoli E. The design of peptide-amphiphiles as functional ligands for liposomal anticancer drug and gene delivery. Adv Drug Deliv Rev 2017; 110-111:80-101. [PMID: 27539561 DOI: 10.1016/j.addr.2016.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/12/2016] [Accepted: 08/05/2016] [Indexed: 12/25/2022]
Abstract
Liposomal nanomedicine has led to clinically useful cancer therapeutics like Doxil and DaunoXome. In addition, peptide-functionalized liposomes represent an effective drug and gene delivery vehicle with increased cancer cell specificity, enhanced tumor-penetrating ability and high tumor growth inhibition. The goal of this article is to review the recently published literature of the peptide-amphiphiles that were used to functionalize liposomes, to highlight successful designs that improved drug and gene delivery to cancer cells in vitro, and cancer tumors in vivo, and to discuss the current challenges of designing these peptide-decorated liposomes for effective cancer treatment.
Collapse
|
32
|
Li S, Cai Y, Cao J, Cai M, Chen Y, Luo X. Phosphorylcholine micelles decorated by hyaluronic acid for enhancing antitumor efficiency. Polym Chem 2017. [DOI: 10.1039/c6py02032a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
DOX-loaded PCL-PDEAMPC micelles coated with HA by electrostatic attraction for enhancing antitumor efficiency and prolonging blood circulation time.
Collapse
Affiliation(s)
- Shuai Li
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu, 610065
- China
| | - Yuanyuan Cai
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu, 610065
- China
| | - Jun Cao
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu, 610065
- China
| | - Mengtan Cai
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu, 610065
- China
| | - Yuanwei Chen
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu, 610065
- China
| | - Xianglin Luo
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu, 610065
- China
- State Key Laboratory of Polymer Materials Engineering
| |
Collapse
|
33
|
Yamada Y, Harashima H. MITO-Porter for Mitochondrial Delivery and Mitochondrial Functional Analysis. Handb Exp Pharmacol 2017; 240:457-472. [PMID: 27830347 DOI: 10.1007/164_2016_4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria are attractive organelles that have the potential to contribute greatly to medical therapy, the maintenance of beauty and health, and the development of the life sciences. Therefore, it would be expected that the further development of mitochondrial drug delivery systems (DDSs) would exert a significant impact on the medical and life sciences. To achieve such an innovative objective, it will be necessary to deliver various cargoes to mitochondria in living cells. However, only a limited number of approaches are available for accomplishing this. We recently proposed a new concept for mitochondrial delivery, a MITO-Porter, a liposome-based carrier that introduces macromolecular cargoes into mitochondria via membrane fusion. To date, we have demonstrated the utility of mitochondrial therapeutic strategy by MITO-Porter using animal models of diseases. We also showed that the mitochondrial delivery of antisense oligo-RNA by the MITO-Porter results in mitochondrial RNA knockdown and has a functional impact on mitochondria. Here, we summarize the current state of mitochondrial DDS focusing on our research and show some examples of mitochondrial functional regulations using mitochondrial DDS.
Collapse
Affiliation(s)
- Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan.
| |
Collapse
|
34
|
Wang Z, Tian Y, Zhang H, Qin Y, Li D, Gan L, Wu F. Using hyaluronic acid-functionalized pH stimuli-responsive mesoporous silica nanoparticles for targeted delivery to CD44-overexpressing cancer cells. Int J Nanomedicine 2016; 11:6485-6497. [PMID: 27980406 PMCID: PMC5147400 DOI: 10.2147/ijn.s117184] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In this study, novel hyaluronic acid-pH stimuli-responsive lipid membrane mesoporous silica nanoparticles (HA-PL-MSNs) were designed and assembled, with the chemotherapeutic agent doxorubicin (DOX) as the model drug. HA-PL-MSNs exhibited a well-defined mesostructure covered by lipid bilayer and particle size of ~150 nm. The drug loading capacity was up to ~18.2%. DOX release could be effectively retained by the lipid bilayer in pH 7.4 buffer and exhibited a pH-triggered burst release in the acidic condition. Confocal laser scanning microscopy and fluorescence-activated cell sorting showed that HA-PL-MSNs exhibited higher cellular uptake efficiency via CD44 receptor-mediated endocytosis compared with PL-MSNs in HeLa cells. In vitro cytotoxicity studies demonstrated that HA-PL-MSNs could effectively enhance the targeted delivery of DOX and restrain the growth of HeLa cells. This might provide a promising alternative for the development of a targeted anticancer drug delivery system.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Yongfeng Tian
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Hua Zhang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Yanmei Qin
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Dong Li
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Li Gan
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Fanhong Wu
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| |
Collapse
|
35
|
Zhang L, Li Z, Sun F, Xu Y, Du Z. Effect of inserted spacer in hepatic cell-penetrating multifunctional peptide component on the DNA intracellular delivery of quaternary complexes based on modular design. Int J Nanomedicine 2016; 11:6283-6295. [PMID: 27920533 PMCID: PMC5126006 DOI: 10.2147/ijn.s115381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A safe and efficient quaternary gene delivery system (named Q-complexes) was constructed based on self-assembly of molecules through noncovalent bonds. This system was formulated through the cooperation and competing interactions of cationic liposomes, multifunctional peptides, and DNA, followed by coating hyaluronic acid on the surface of the ternary complexes. The multifunctional peptide was composed of two functional domains: penetrating hepatic tumor-targeted cell moiety (KRPTMRFRYTWNPMK) and a wrapping gene sequence (polyarginine 16). The effect of spacer insertion between the two domains of multifunctional peptide on the intracellular transfection of Q-complexes was further studied. Experimental results showed that the formulations assembled with various peptides in the spacer elements possessed different intercellular pathways and transfection efficiencies. The Q-complexes containing peptide in the absence of spacer element (Pa) showed the highest gene expression among all samples. The Q-complexes containing peptides with a noncleavable spacer GA (Pc) had no ability of intracellular nucleic acid delivery, whereas those with a cleavable spacer RVRR (Pd) showed moderate transfection activity. These results demonstrated that the different spacers inserted in the multifunctional peptide played an important role in in vitro DNA transfection efficiency. Atomic force microscopy images showed that the morphologies of ternary complexes (LPcD) and Q-complexes (HLcPD) were crystal lamellas, whereas those of other nanocomplexes were spheres. Circular dichroism showed the changed configuration of peptide with spacer GA in nanocomplexes compared with that of its free state, whereas the Pa configuration without spacer in nanocomplexes was consistent with that of its free state. The present study contributed to the structural understanding of Q-complexes, and further effective modification is in progress.
Collapse
Affiliation(s)
- Luchen Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai
| | - Zhenbo Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai
| | - Fangli Sun
- School of Engineering, Zhejiang Agriculture and Forestry University, Lin'an, Zhejiang Province, People's Republic of China
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai
| | - Zixiu Du
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai
| |
Collapse
|
36
|
He Y, Zhou J, Ma S, Nie Y, Yue D, Jiang Q, Wali ARM, Tang JZ, Gu Z. Multi-Responsive "Turn-On" Nanocarriers for Efficient Site-Specific Gene Delivery In Vitro and In Vivo. Adv Healthc Mater 2016; 5:2799-2812. [PMID: 27717282 DOI: 10.1002/adhm.201600710] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/23/2016] [Indexed: 12/19/2022]
Abstract
Systemic gene delivery is a complicated and multistep process that confronts numerous biological barriers. It remains a formidable challenge to exploit a single gene carrier with multiple features to combat all obstacles collectively. Herein, a multi-responsive "turn-on" polyelectrolyte complex (DNA/OEI-SSx /HA-SS-COOH, DSS) delivery system is demonstrated with a sequential self-assembly of disulfide-conjugated oligoethylenimine (OEI-SSx ) and disulfide bond-modified hyaluronic acid envelope (HA-SS-COOH) that can combat multiple biological barriers collectively when administered intravenously. DSS is designed to effectively accumulate at the tumor tissue and to be internalized into tumor cells by recognizing CD44. The multi-responsive "turn-on" DSS can respond to the alterations of hyaluronidases and glutathione at both the tumor site and at the intracellular milieu. Sequential degradation and detachment of the HA-SS-COOH envelope followed by the dissociation of the OEI-SSx/DNA inner core contributes to the activation of the endosomal escape and gene release functions, thus greatly enhancing nuclear gene delivery. A systematic investigation of DSS has revealed that the tumor accumulation ability, internalization, and endosome escape of the DSS nanocarriers, DNA unpacking and nuclear transportation are all remarkably improved by the multi-responsive "turn-on" design resulting in highly efficient gene transfection in vitro and in vivo.
Collapse
Affiliation(s)
- Yiyan He
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Jie Zhou
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Shengnan Ma
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Dong Yue
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Qian Jiang
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Aisha Roshan Mohamed Wali
- Faculty of Science and Engineering; School of Pharmacy; University of Wolverhampton; Wulfruna Street Wolverhampton WV1 1SB UK
| | - James Zhenggui Tang
- Faculty of Science and Engineering; School of Pharmacy; University of Wolverhampton; Wulfruna Street Wolverhampton WV1 1SB UK
| | - Zhongwei Gu
- National Engineering Research Center for Biomaterials; Sichuan University; 29 Wangjiang Road Chengdu 610064 P. R. China
- College of Materials Science and Engineering; Nanjing Tech University; 30 South Puzhu Road 211816 Nanjing P. R. China
| |
Collapse
|
37
|
Torrecilla J, Del Pozo-Rodríguez A, Solinís MÁ, Apaolaza PS, Berzal-Herranz B, Romero-López C, Berzal-Herranz A, Rodríguez-Gascón A. Silencing of hepatitis C virus replication by a non-viral vector based on solid lipid nanoparticles containing a shRNA targeted to the internal ribosome entry site (IRES). Colloids Surf B Biointerfaces 2016; 146:808-17. [PMID: 27451369 DOI: 10.1016/j.colsurfb.2016.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 02/05/2023]
Abstract
Gene silencing mediated by RNAi has gained increasing interest as an alternative for the treatment of infectious diseases such as refractory hepatitis C virus (HCV) infection. In this work we have designed and evaluated a non-viral vector based on solid lipid nanoparticles (SLN) bearing hyaluronic acid, protamine and a short hairpin RNA (shRNA74) targeted to the Internal Ribosome Entry Site (IRES) of the HCV. The vector was able to inhibit the expression of the HCV IRES in Huh-7 cells, with the inhibition level dependent on the shRNA74 to SLN ratio and on the shRNA74 dose added to the culture cells. The nanocarrier was also able to inhibit the replication in human hepatoma cells supporting a subgenomic HCV replicon (Huh-7 NS3-3'). The vector was quickly and efficiently internalized by the cells, and endocytosis was the most productive uptake mechanism for silencing. Clathrin-mediated endocytosis and to a lesser extent caveolae/lipid raft-mediated endocytosis were identified as endocytic mechanisms involved in the cell uptake. Internalization via the CD44 receptor was also involved, although this entry route seems to be less productive for silencing than endocytosis. The vector did not induce either hemolysis or agglutination of red cells in vitro, which was indicative of good biocompatibility. In summary, we have shown for the first time the ability of a non-viral SLN-based vector to silence a HCV replicon.
Collapse
Affiliation(s)
- Josune Torrecilla
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7. 01006, Vitoria-Gasteiz, Spain.
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7. 01006, Vitoria-Gasteiz, Spain.
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7. 01006, Vitoria-Gasteiz, Spain.
| | - Paola S Apaolaza
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7. 01006, Vitoria-Gasteiz, Spain.
| | - Beatriz Berzal-Herranz
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), PTS Granada, Av del conocimiento s/n, Armilla, 18016, Granada, Spain.
| | - Cristina Romero-López
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), PTS Granada, Av del conocimiento s/n, Armilla, 18016, Granada, Spain.
| | - Alfredo Berzal-Herranz
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), PTS Granada, Av del conocimiento s/n, Armilla, 18016, Granada, Spain.
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7. 01006, Vitoria-Gasteiz, Spain.
| |
Collapse
|
38
|
Li L, Song L, Yang X, Li X, Wu Y, He T, Wang N, Yang S, Zeng Y, Yang L, Wu Q, Wei Y, Gong C. Multifunctional "core-shell" nanoparticles-based gene delivery for treatment of aggressive melanoma. Biomaterials 2016; 111:124-137. [PMID: 27728812 DOI: 10.1016/j.biomaterials.2016.09.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/27/2016] [Accepted: 09/29/2016] [Indexed: 02/05/2023]
Abstract
Gene therapy may be a promising and powerful strategy for cancer treatment, but efficient targeted gene delivery in vivo has so far remained challenging. Here, we developed a well-tailored and versatile "core-shell" ternary system (RRPHC) of systemic gene delivery for treatment of aggressive melanoma. The capsid-like "shell" of this system was engineered to mediate depth penetration to tissues, simultaneously target the CD44 receptors and integrin αvβ3 receptors overexpressed on neovasculature and most malignant tumor cells, while the "core" was responsible for nucleus-targeting and effective transfection. The RRPHC ternary complexes enhanced cellular uptake via dual receptor-mediated endocytosis, improved the endosomal escape and significantly promoted the plasmid penetration into the nucleus. Notably, RRPHC ternary complexes exhibited ultra-high gene transfection efficiency (∼100% in B16F10 cells), which surpassed that of commercial transfection agents, PEI 25K, Lipofectamine 2000 and even Lipofectamine 3000. Especially, RRPHC ternary complexes showed excellent serum resistance and remained high gene transfection efficacy (∼100%) even in medium containing 30% serum. In vivo biodistribution imaging demonstrated RRPHC ternary complexes possessed much more accumulation and extensive distribution throughout tumor regions while minimal location in other organs. Furthermore, systemic delivery of the pro-apoptotic mTRAIL gene to tumor xenografts by RRPHC ternary complexes resulted in remarkable inhibition of melanoma, with no systemic toxicity. These results demonstrated that the designed novel RRPHC ternary complexes might be a promising gene delivery system for targeted cancer therapy in vivo.
Collapse
Affiliation(s)
- Ling Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Linjiang Song
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Xi Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Xia Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Yuzhe Wu
- College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Tao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Suleixin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Yan Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Li Yang
- Carl Zeiss (Shanghai) Co., Ltd., Chengdu Branch, PR China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China.
| |
Collapse
|
39
|
Wang R, Luo Y, Yang S, Lin J, Gao D, Zhao Y, Liu J, Shi X, Wang X. Hyaluronic acid-modified manganese-chelated dendrimer-entrapped gold nanoparticles for the targeted CT/MR dual-mode imaging of hepatocellular carcinoma. Sci Rep 2016; 6:33844. [PMID: 27653258 PMCID: PMC5032118 DOI: 10.1038/srep33844] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common malignant tumor of the liver. The early and effective diagnosis has always been desired. Herein, we present the preparation and characterization of hyaluronic acid (HA)-modified, multifunctional nanoparticles (NPs) targeting CD44 receptor-expressing cancer cells for computed tomography (CT)/magnetic resonance (MR) dual-mode imaging. We first modified amine-terminated generation 5 poly(amidoamine) dendrimers (G5.NH2) with an Mn chelator, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), fluorescein isothiocyanate (FI), and HA. Then, gold nanoparticles (AuNPs) were entrapped within the above raw product, denoted as G5.NH2-FI-DOTA-HA. The designed multifunctional NPs were formed after further Mn chelation and purification and were denoted as {(Au0)100G5.NH2-FI-DOTA(Mn)-HA}. These NPs were characterized via several different techniques. We found that the {(Au0)100G5.NH2-FI-DOTA(Mn)-HA} NPs exhibited good water dispersibility, stability under different conditions, and cytocompatibility within a given concentration range. Because both AuNPs and Mn were present in the product, {(Au0)100G5.NH2-FI-DOTA(Mn)-HA} displayed a high X-ray attenuation intensity and favorable r1 relaxivity, which are advantageous properties for targeted CT/MR dual-mode imaging. This approach was used to image HCC cells in vitro and orthotopically transplanted HCC tumors in a unique in vivo model through the CD44 receptor-mediated endocytosis pathway. This work introduces a novel strategy for preparing multifunctional NPs via dendrimer nanotechnology.
Collapse
Affiliation(s)
- Ruizhi Wang
- Shanghai Institute of Medical Imaging, Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yu Luo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China
| | - Shuohui Yang
- Shanghai Institute of Medical Imaging, Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jiang Lin
- Shanghai Institute of Medical Imaging, Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jinguo Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China
| | - Xiaolin Wang
- Shanghai Institute of Medical Imaging, Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
40
|
Jang HE, Mok H. Polydopamine-Coated Porous Microspheres Conjugated with Immune Stimulators for Enhanced Cytokine Induction in Macrophages. Macromol Biosci 2016; 16:1562-1569. [PMID: 27503807 DOI: 10.1002/mabi.201600195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/18/2016] [Indexed: 11/09/2022]
Abstract
Polydopamine-coated porous microsphere (PPM) is investigated as a simple and versatile immobilization strategy for immune-stimulating biomolecules to enhance delivery efficiency and immune-stimulating effects such as cytokine induction in macrophages. The PPMs, with diameters of about 2 μm, exhibit simultaneous and efficient incorporation of biomolecules (nucleotides and proteins), which is comparable to that achieved using microspheres carrying biomolecules internally by virtue of their porous structure. Ovalbumin-conjugated PPMs are internalized into macrophages efficiently and selectively via the phagocytic pathway, without any noticeable toxicity. Internalized CpG oligodeoxynucleotide (ODN)-conjugated PPMs (PPM-CpG) greatly enhance the induction of selected cytokines (TNF-α and IL-6) in RAW 264.7 cells compared to that by the soluble CpG ODN and ionic complexes. Therefore, PPMs generated in this study may serve as effective carriers of immune-stimulating biomolecules such as diverse toll-like receptor agonists.
Collapse
Affiliation(s)
- Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
41
|
Lin WJ, Lee WC, Shieh MJ. Hyaluronic acid conjugated micelles possessing CD44 targeting potential for gene delivery. Carbohydr Polym 2016; 155:101-108. [PMID: 27702492 DOI: 10.1016/j.carbpol.2016.08.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/28/2016] [Accepted: 08/08/2016] [Indexed: 11/17/2022]
Abstract
The high- and low-molecular weight hyaluronic acid (HHA and LHA) were used to conjugate with PLGA-PEG copolymers which were applied to encapsulate DOTAP/pDNA (D/P) lipoplex as a CD44-targeted micelle delivery system. The size and zeta potential of DNA loaded micelles were measured. The cytotoxicity and cellular transfection of DNA loaded micelles were performed in CD44-positive MDA-MB-231 and MCF-7 cancer cells and CD44-negative HepG2 cells. The endocytosis mechanism of micelles was investigated further. The DNA loaded HA-conjugated micelles possessed negative-charged character which prevented erythrocytes from agglutination. Both LHA-PEG-PLGA and HHA-PEG-PLGA micelles had comparable cellular viability in L929 normal cells. The cellular transfection of HHA-PEG-PLGA micelles was much higher than of LHA-PEG-PLGA micelles in CD44-positive cells. The specific and strong binding of HHA to CD44-positive cells resulted in the cellular transfection of HHA-PEG-PLGA micelles in CD44-positive cells significantly higher than in CD44-negative cells.
Collapse
Affiliation(s)
- Wen Jen Lin
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, National Taiwan University, Taipei, Taiwan; Drug Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Wei-Chi Lee
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
42
|
Cai Y, Tang Q, Wu X, Si W, Zhang Q, Huang W, Dong X. Bromo-Substituted Diketopyrrolopyrrole Derivative with Specific Targeting and High Efficiency for Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:10737-10742. [PMID: 27064605 DOI: 10.1021/acsami.6b01533] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Novel photosensitizers with high reactive oxygen species generation and precise targeting to tumors are crucial for photodynamic therapy (PDT). Here, a bromo-substituted diketopyrrolopyrrole derivative (2,5-bis(6-bromohexyl)-3,6-bis(5-bromothiophene-2-yl)-2,5-dihydropyrrolo[3,4-c]pyrrole-1,4-dione)-grafted hyaluronic acid is synthesized, which presents excellent targeting and PDT efficiency both in vitro and in vivo.
Collapse
Affiliation(s)
- Yu Cai
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Qianyun Tang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Xiujuan Wu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Qi Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), and ‡School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech) , 30 South Puzhu Road, Nanjing 211816, China
| |
Collapse
|
43
|
A Dual-Ligand Liposomal System Composed of a Cell-Penetrating Peptide and a Mitochondrial RNA Aptamer Synergistically Facilitates Cellular Uptake and Mitochondrial Targeting. J Pharm Sci 2016; 105:1705-1713. [PMID: 27056631 DOI: 10.1016/j.xphs.2016.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/08/2016] [Accepted: 03/01/2016] [Indexed: 01/22/2023]
Abstract
It has been reported that the use of mitochondrial RNA aptamers including RNase P (RP) results in the selective mitochondrial delivery of endogenous and exogenous RNAs. The issue of whether these aptamers would be useful ligands for the mitochondrial targeting of a nanoparticle has not been demonstrated to date because nanocarriers modified with these RNA aptamers are insufficiently internalized by cells. We report here on the development of a dual-ligand liposomal system composed of octaarginine (R8), a device that enhances cellular uptake, and an RP aptamer for mitochondrial targeting to permit a nanocarrier to be efficiently delivered to mitochondria. Surprisingly, the cellular uptake of the R8-modified nanocarrier was facilitated by modification with an RP aptamer. The optimal composition of a nanocarrier needed for efficient cellular uptake and mitochondrial targeting was determined. In a confocal laser scanning microscopy analysis, the dual-ligand-modified nanocarrier was found to result in effective mitochondrial targeting through an ATP-dependent pathway and was much more effective than a single-ligand R8-modified nanocarrier. This is the first report of the regulation of intracellular trafficking by a mitochondrial RNA aptamer-modified nanocarrier system.
Collapse
|
44
|
Liang K, Bae KH, Lee F, Xu K, Chung JE, Gao SJ, Kurisawa M. Self-assembled ternary complexes stabilized with hyaluronic acid-green tea catechin conjugates for targeted gene delivery. J Control Release 2016; 226:205-16. [PMID: 26855049 DOI: 10.1016/j.jconrel.2016.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/08/2016] [Accepted: 02/03/2016] [Indexed: 11/25/2022]
Abstract
Nanosized polyelectrolyte complexes are attractive delivery vehicles for the transfer of therapeutic genes to diseased cells. Here we report the application of self-assembled ternary complexes constructed with plasmid DNA, branched polyethylenimine and hyaluronic acid-green tea catechin conjugates for targeted gene delivery. These conjugates not only stabilize plasmid DNA/polyethylenimine complexes via the strong DNA-binding affinity of green tea catechin, but also facilitate their transport into CD44-overexpressing cells via receptor-mediated endocytosis. The hydrodynamic size, surface charge and physical stability of the complexes are characterized. We demonstrate that the stabilized ternary complexes display enhanced resistance to nuclease attack and polyanion-induced dissociation. Moreover, the ternary complexes can efficiently transfect the difficult-to-transfect HCT-116 colon cancer cell line even in serum-supplemented media due to their enhanced stability and CD44-targeting ability. Confocal microscopic analysis demonstrates that the stabilized ternary complexes are able to promote the nuclear transport of plasmid DNA more effectively than binary complexes and hyaluronic acid-coated ternary complexes. The present study suggests that the ternary complexes stabilized with hyaluronic acid-green tea catechin conjugates can be widely utilized for CD44-targeted delivery of nucleic acid-based therapeutics.
Collapse
Affiliation(s)
- Kun Liang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Ki Hyun Bae
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Fan Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Keming Xu
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Joo Eun Chung
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Shu Jun Gao
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore
| | - Motoichi Kurisawa
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore.
| |
Collapse
|
45
|
Badwaik V, Liu L, Gunasekera D, Kulkarni A, Thompson DH. Mechanistic Insight into Receptor-Mediated Delivery of Cationic-β-Cyclodextrin:Hyaluronic Acid-Adamantamethamidyl Host:Guest pDNA Nanoparticles to CD44(+) Cells. Mol Pharm 2016; 13:1176-84. [PMID: 26900622 DOI: 10.1021/acs.molpharmaceut.6b00078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Targeted delivery is a key element for improving the efficiency and safety of nonviral vectors for gene therapy. We have recently developed a CD44 receptor targeted, hyaluronic acid-adamantamethamidyl based pendant polymer system (HA-Ad), capable of forming complexes with cationic β-cyclodextrins (CD-PEI(+)) and pDNA. Complexes formed using these compounds (HA-Ad:CD-PEI(+):pDNA) display high water solubility, good transfection efficiency, and low cytotoxicity. Spatial and dynamic tracking of the transfection complexes by confocal microscopy and multicolor flow cytometry techniques was used to evaluate the target specificity, subcellular localization, and endosomal escape process. Our data shows that cells expressing the CD44 receptor undergo enhanced cellular uptake and transfection efficiency with HA-Ad:CD-PEI(+):pDNA complexes. This transfection system, comprised noncovalent assembly of cyclodextrin:adamantamethamidyl-modified hyaluronic acid via host:guest interactions to condense pDNA, is a potentially useful tool for targeted delivery of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Vivek Badwaik
- Department of Chemistry and Purdue University Center for Cancer Research, Multidisciplinary Cancer Research Facility, Bindley Bioscience Center, Purdue University , 1203 West State Street, West Lafayette, Indiana 47907, United States
| | - Linjia Liu
- Department of Chemistry and Purdue University Center for Cancer Research, Multidisciplinary Cancer Research Facility, Bindley Bioscience Center, Purdue University , 1203 West State Street, West Lafayette, Indiana 47907, United States
| | - Dinara Gunasekera
- Department of Chemistry and Purdue University Center for Cancer Research, Multidisciplinary Cancer Research Facility, Bindley Bioscience Center, Purdue University , 1203 West State Street, West Lafayette, Indiana 47907, United States
| | - Aditya Kulkarni
- Department of Chemistry and Purdue University Center for Cancer Research, Multidisciplinary Cancer Research Facility, Bindley Bioscience Center, Purdue University , 1203 West State Street, West Lafayette, Indiana 47907, United States
| | - David H Thompson
- Department of Chemistry and Purdue University Center for Cancer Research, Multidisciplinary Cancer Research Facility, Bindley Bioscience Center, Purdue University , 1203 West State Street, West Lafayette, Indiana 47907, United States
| |
Collapse
|
46
|
Zulliger R, Conley SM, Naash MI. Non-viral therapeutic approaches to ocular diseases: An overview and future directions. J Control Release 2015; 219:471-487. [PMID: 26439665 PMCID: PMC4699668 DOI: 10.1016/j.jconrel.2015.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 12/31/2022]
Abstract
Currently there are no viable treatment options for patients with debilitating inherited retinal degeneration. The vast variability in disease-inducing mutations and resulting phenotypes has hampered the development of therapeutic interventions. Gene therapy is a logical approach, and recent work has focused on ways to optimize vector design and packaging to promote optimized expression and phenotypic rescue after intraocular delivery. In this review, we discuss ongoing ocular clinical trials, which currently use viral gene delivery, but focus primarily on new advancements in optimizing the efficacy of non-viral gene delivery for ocular diseases. Non-viral delivery systems are highly customizable, allowing functionalization to improve cellular and nuclear uptake, bypassing cellular degradative machinery, and improving gene expression in the nucleus. Non-viral vectors often yield transgene expression levels lower than viral counterparts, however their favorable safety/immune profiles and large DNA capacity (critical for the delivery of large ocular disease genes) make their further development a research priority. Recent work on particle coating and vector engineering presents exciting ways to overcome limitations of transient/low gene expression levels, but also highlights the fact that further refinements are needed before use in the clinic.
Collapse
Affiliation(s)
- Rahel Zulliger
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States.
| |
Collapse
|
47
|
Hu P, Chen Y, Liu Y. Hyaluronan/Ru( ii)-cyclodextrin supramolecular assemblies for colorimetric sensor of hyaluronidase activity. RSC Adv 2015. [DOI: 10.1039/c5ra19122j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A hyaluronidase-induced colorimetric change was found in a hyaluronan/Ru(ii)-cyclodextrin supramolecular assembly under a laser (532 nm) irradiation.
Collapse
Affiliation(s)
- Ping Hu
- Department of Chemistry
- State Key Laboratory of Elemento-Organic Chemistry
- Nankai University
- Tianjin 300071
- P. R. China
| | - Yong Chen
- Department of Chemistry
- State Key Laboratory of Elemento-Organic Chemistry
- Nankai University
- Tianjin 300071
- P. R. China
| | - Yu Liu
- Department of Chemistry
- State Key Laboratory of Elemento-Organic Chemistry
- Nankai University
- Tianjin 300071
- P. R. China
| |
Collapse
|