1
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
2
|
Seo H, Jeon L, Kwon J, Lee H. High-Precision Synthesis of RNA-Loaded Lipid Nanoparticles for Biomedical Applications. Adv Healthc Mater 2023; 12:e2203033. [PMID: 36737864 DOI: 10.1002/adhm.202203033] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/26/2023] [Indexed: 02/05/2023]
Abstract
The recent development of RNA-based therapeutics in delivering nucleic acids for gene editing and regulating protein translation has led to the effective treatment of various diseases including cancer, inflammatory and genetic disorder, as well as infectious diseases. Among these, lipid nanoparticles (LNP) have emerged as a promising platform for RNA delivery and have shed light by resolving the inherent instability issues of naked RNA and thereby enhancing the therapeutic potency. These LNP consisting of ionizable lipid, helper lipid, cholesterol, and poly(ethylene glycol)-anchored lipid can stably enclose RNA and help them release into the cells' cytosol. Herein, the significant progress made in LNP research starting from the LNP constituents, formulation, and their diverse applications is summarized first. Moreover, the microfluidic methodologies which allow precise assembly of these newly developed constituents to achieve LNP with controllable composition and size, high encapsulation efficiency as well as scalable production are highlighted. Furthermore, a short discussion on current challenges as well as an outlook will be given on emerging approaches to resolving these issues.
Collapse
Affiliation(s)
- Hanjin Seo
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Leekang Jeon
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Jaeyeong Kwon
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Hyomin Lee
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| |
Collapse
|
3
|
Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid Nanoparticles─From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS NANO 2021; 15:16982-17015. [PMID: 34181394 DOI: 10.1021/acsnano.1c04996] [Citation(s) in RCA: 806] [Impact Index Per Article: 268.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Lipid nanoparticles (LNPs) have emerged across the pharmaceutical industry as promising vehicles to deliver a variety of therapeutics. Currently in the spotlight as vital components of the COVID-19 mRNA vaccines, LNPs play a key role in effectively protecting and transporting mRNA to cells. Liposomes, an early version of LNPs, are a versatile nanomedicine delivery platform. A number of liposomal drugs have been approved and applied to medical practice. Subsequent generations of lipid nanocarriers, such as solid lipid nanoparticles, nanostructured lipid carriers, and cationic lipid-nucleic acid complexes, exhibit more complex architectures and enhanced physical stabilities. With their ability to encapsulate and deliver therapeutics to specific locations within the body and to release their contents at a desired time, LNPs provide a valuable platform for treatment of a variety of diseases. Here, we present a landscape of LNP-related scientific publications, including patents and journal articles, based on analysis of the CAS Content Collection, the largest human-curated collection of published scientific knowledge. Rising trends are identified, such as nanostructured lipid carriers and solid lipid nanoparticles becoming the preferred platforms for numerous formulations. Recent advancements in LNP formulations as drug delivery platforms, such as antitumor and nucleic acid therapeutics and vaccine delivery systems, are discussed. Challenges and growth opportunities are also evaluated in other areas, such as medical imaging, cosmetics, nutrition, and agrochemicals. This report is intended to serve as a useful resource for those interested in LNP nanotechnologies, their applications, and the global research effort for their development.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS, a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Allison E Curtze
- CAS, a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Qiongqiong Zhou
- CAS, a division of the American Chemical Society, Columbus, Ohio 43210, United States
| |
Collapse
|
4
|
Bialas F, Reichinger D, Becker CF. Biomimetic and biopolymer-based enzyme encapsulation. Enzyme Microb Technol 2021; 150:109864. [DOI: 10.1016/j.enzmictec.2021.109864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022]
|
5
|
Grilo LF, Martins JD, Cavallaro CH, Nathanielsz PW, Oliveira PJ, Pereira SP. Development of a 96-well based assay for kinetic determination of catalase enzymatic-activity in biological samples. Toxicol In Vitro 2020; 69:104996. [DOI: 10.1016/j.tiv.2020.104996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
|
6
|
He X, Feng J, Yan S, Zhang Y, Zhong C, Liu Y, Shi D, Abagyan R, Xiang T, Zhang J. Biomimetic microbioreactor-supramolecular nanovesicles improve enzyme therapy of hepatic cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102311. [PMID: 33011392 DOI: 10.1016/j.nano.2020.102311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/05/2020] [Accepted: 09/20/2020] [Indexed: 12/24/2022]
Abstract
A novel biomimetic nanovesicle-loaded supramolecular enzyme-based therapeutics has been developed. Here, using a biomimetic lipid-D-α-tocopherol polyethylene glycol succinate (TPGS) hybrid semi-permeable membrane, cyclodextrin supramolecular docking, metal-ion-aided coordination complexing, we combined multiple functional motifs into a single biomimetic microbioreactor-supramolecular nanovesicle (MiSuNv) that allowed effective transport of arginine deiminase (ADI) to hepatic tumor cells to enhance arginine depletion. We compared two intercalated enzyme-carrying supermolecular motifs mainly comprising of 2-hydroxypropyl-β-cyclodextrin and sulfobutyl-ether-β-cyclodextrin, the only two cyclodextrin derivatives approved for injection by the United States Food and Drug Administration. The ADI-specific antitumor effects were enhanced by TPGS (one constituent of MiSuNv, having synergistic antitumor effects), as ADI was separated from adverse external environment by a semi-permeable membrane and sequestered in a favorable internal microenvironment with an optimal pH and metal-ion combination. ADI@MiSuNv contributed to cell cycle arrest, apoptosis and autophagy through the enhanced efficacy of enzyme treatment against Hep3B xenograft tumors in rats.
Collapse
Affiliation(s)
- Xiaoqian He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiao Feng
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Shenglei Yan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yonghong Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yuying Liu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Bertoni S, Tedesco D, Bartolini M, Prata C, Passerini N, Albertini B. Solid Lipid Microparticles for Oral Delivery of Catalase: Focus on the Protein Structural Integrity and Gastric Protection. Mol Pharm 2020; 17:3609-3621. [PMID: 32786955 PMCID: PMC8009523 DOI: 10.1021/acs.molpharmaceut.0c00666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 01/22/2023]
Abstract
Protein inactivation either during the production process or along the gastrointestinal tract is the major problem associated with the development of oral delivery systems for biological drugs. This work presents an evaluation of the structural integrity and the biological activity of a model protein, catalase, after its encapsulation in glyceryl trimyristate-based solid lipid microparticles (SLMs) obtained by the spray congealing technology. Circular dichroism and fluorescence spectroscopies were used to assess the integrity of catalase released from SLMs. The results confirmed that no conformational change occurred during the production process and both the secondary and tertiary structures were retained. Catalase is highly sensitive to temperature and undergoes denaturation above 60 °C; nevertheless, spray congealing allowed the retention of most biological activity due to the loading of the drug at the solid state, markedly reducing the risk of denaturation. Catalase activity after exposure to simulated gastric conditions (considering both acidic pH and the presence of gastric digestive hydrolases) ranged from 35 to 95% depending on the carrier: increasing of both the fatty acid chain length and the degree of substitution of the glyceride enhanced residual enzyme activity. SLMs allowed the protein release in a simulated intestinal environment and were not cytotoxic against HT29 cells. In conclusion, the encapsulation of proteins into SLMs by spray congealing might be a promising strategy for the formulation of nontoxic and inexpensive oral biotherapeutic products.
Collapse
Affiliation(s)
- Serena Bertoni
- PharmTech
Lab, Department of Pharmacy and Biotechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Daniele Tedesco
- Bio-Pharmaceutical
Analysis Section (Bio-PhASe), Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Bio-Pharmaceutical
Analysis Section (Bio-PhASe), Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Cecilia Prata
- Biochemistry
Lab, Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Nadia Passerini
- PharmTech
Lab, Department of Pharmacy and Biotechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Beatrice Albertini
- PharmTech
Lab, Department of Pharmacy and Biotechnology, University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| |
Collapse
|
8
|
Yang L, Zhang Y, Xie J, Zhong C, He D, Wang T, Li K, Li Y, Shi D, Abagyan R, Yang L, Zhang J. Biomimetic polysaccharide-cloaked lipidic nanovesicles/microassemblies for improving the enzymatic activity and prolonging the action time for hyperuricemia treatment. NANOSCALE 2020; 12:15222-15235. [PMID: 32639489 DOI: 10.1039/d0nr02651d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The improvement and maintenance of enzymatic activities represent major challenges. However, to address these we developed novel biomimetic polysaccharide hyaluronan (Hn)-cloaked lipidic nanovesicles (BHLN) and microassemblies (BHLNM) as enzyme carriers that function by entrapping enzymes in the core or by tethering them to the inner/outer surfaces via covalent interactions. The effectiveness of these enzyme carriers was demonstrated through an evaluation of the enzymatic activity and anti-hyperuricemia bioactivity of urate oxidase (also called uricase, Uase). We showed that Uase was effectively loaded within the BHLN/BHLNM (UHLN/UHLNM) and maintained good enzymatic bioactivity through a range of effects, including isolation from the external environment due to the vesicle-carrying (shielding effect), avoidance of recognition by the reticuloendothelial system due to Hn-cloaking (long-term effect), production of beneficial conformational changes (allosteric effect) due to a favorable internal microenvironment of construction and vesicle loading, and stabilization due to the reversible conjugation of Uase or vesicle and serum albumin (deposit effect). UHLN/UHLNM had significantly increased bioavailability (∼533% and ∼331% compared to Uase) and demonstrated greatly improved efficacy, whereby the time required for UHLN/UHLNM to lower the plasma uric acid concentration to a normal level was much shorter than that for free Uase. The interactions of the therapeutic enzyme (Uase), biomimetic membrane components (Hn and phospholipid), and serum albumin were investigated with a fluorescent probe and computational simulations to help understand the superior properties of UHLN/UHLNM.
Collapse
Affiliation(s)
- Lan Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yonghong Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jiangchuan Xie
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Tingting Wang
- Biochemistry and Molecular Biology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 401331, China
| | - Kailing Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yao Li
- Division of Infectious Disease, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Lin Yang
- Department of Pharmacology, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
9
|
Gu J, Huang Y, Yan Z, He D, Zhang Y, Xu J, Li Y, Xie X, Xie J, Shi D, Abagyan R, Zhang J, Tan Q. Biomimetic Membrane-Structured Nanovesicles Carrying a Supramolecular Enzyme to Cure Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:31112-31123. [PMID: 32544316 DOI: 10.1021/acsami.0c06207] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Platforms for enzyme delivery must simultaneously have plasma stability, high catalytic activity, and low/no immunogenicity of the enzyme. Here, we designed a novel biomimetic membrane-structured nanovesicle (BNV) to efficiently carry supramolecular enzymes to meet the above requirements. We complexed l-asparaginase (Aase) with hydroxypropyl-β-cyclodextrin (HPCD) to form a supramolecular amphiphile (AS) by self-assembly via noncovalent reversible interactions. We then used the first synthesized polyethylene glycol (PEG 2 kDa)-decorated hyaluronan (12 kDa) and HPCD to self-assemble a semipermeable biomimetic membrane-structured nanovesicle (BNV) together with AS loading. As compared to native Aase, AS@BNV exhibited superior catalytic activity preservation, improved catalytic activity, better pharmacokinetics in rats, enhanced cytotoxic effects, increased antitumor efficacy, and decreased side effects. The underlying mechanisms, such as the autophagy inhibition action against tumor cells, protein-protein docking of the interaction between Aase-serum albumin, and decreased hepatic enzymatic activity, were investigated. This approach paves the way for new types of powerful biomimetic-, supramolecular-, and nanocarrier-based enzymatic therapies.
Collapse
Affiliation(s)
- Jing Gu
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing 400042, China
| | - Yongjia Huang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Zijun Yan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yonghong Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jingyu Xu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Yao Li
- Division of Infectious Disease, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Xuemei Xie
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jiaxi Xie
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing 400042, China
| |
Collapse
|
10
|
Huang Y, Gu J, Yan Z, Hu X, He D, Zhang Y, Li Y, Zhong C, Yang J, Shi D, Abagyan R, Tan Q, Zhang J. Cytomembrane-mimicking nanocarriers with a scaffold consisting of a CD44-targeted endogenous component for effective asparaginase supramolecule delivery. NANOSCALE 2020; 12:12083-12097. [PMID: 32478361 DOI: 10.1039/d0nr02588g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Highly effective and safe delivery of therapeutic enzymes is pivotal to the success of antitumor therapy. Herein, we report on a targeted enzyme delivery system based on cytomembrane-mimicking nanocarriers (CmN) and a supramolecular technique (SmT). Specifically, each CmN had a scaffold that mainly consisted of a CD44-targeted endogenous component conjugated with polyethylene glycol 2000 (HA-g-PEG) that self-assembled with α-cyclodextrin (ACD). The CmN acted as a microbioreactor with an inner hollow space with the capacity to confine the large molecule asparaginase (Asp) in an Asp/ACD-supramolecular complex conjugated to the inner region. The supramolecular Asp loaded into the CmN (A-S-CmN) exhibited superior stability, kinetic properties, catalytic activity and antitumor effects compared to free Asp due to the dual protection of the supramolecular complex and the nanovesicle, the CD44 targeting-homing ability, the prolonged effects of HA-g-PEG, and the favorable inner microenvironment of the constructed supramolecular CmN. The A-S-CmN also showed a decrease in in vivo toxicity and immunogenicity. CmN combined with SmT therapeutics are easy to implement and extend for use in the delivery of various enzymes and for many types of cancer treatment.
Collapse
Affiliation(s)
- Yongjia Huang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Gu
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing 400042, China.
| | - Zijun Yan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Xueyuan Hu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yonghong Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yao Li
- Division of infectious disease, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jie Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing 400042, China.
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
11
|
Mousavi M, Hakimian S, Mustafa TA, Aziz FM, Salihi A, Ale-Ebrahim M, Mirpour M, Rasti B, Akhtari K, Shahpasand K, Abou-Zied OK, Falahati M. The interaction of silica nanoparticles with catalase and human mesenchymal stem cells: biophysical, theoretical and cellular studies. Int J Nanomedicine 2019; 14:5355-5368. [PMID: 31409992 PMCID: PMC6643057 DOI: 10.2147/ijn.s210136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/04/2019] [Indexed: 11/23/2022] Open
Abstract
Aim Nanoparticles (NPs) have been receiving potential interests in protein delivery and cell therapy. As a matter of fact, NPs may be used as great candidates in promoting cell therapy by catalase (CAT) delivery into high oxidative stress tissues. However, for using NPs like SiO2 as carriers, the interaction of NPs with proteins and mesenchymal stem cells (MSCs) should be explored in advance. Methods In the present study, the interaction of SiO2 NPs with CAT and human MSCs (hMSCs) was explored by various spectroscopic methods (fluorescence, circular dichroism (CD), UV-visible), molecular docking and dynamics studies, and cellular (MTT, cellular morphology, cellular uptake, lactate dehydrogenase, ROS, caspase-3, flow cytometry) assays. Results Fluorescence study displayed that both dynamic and static quenching mechanisms and hydrophobic interactions are involved in the spontaneous interaction of SiO2 NPs with CAT. CD spectra indicated that native structure of CAT remains stable after interaction with SiO2 NPs. UV-visible study also revealed that the kinetic parameters of CAT such as Km, Vmax, Kcat, and enzyme efficiency were not changed after the addition of SiO2 NPs. Molecular docking and dynamics studies showed that Si and SiO2 clusters interact with hydrophobic residues of CAT and SiO2 cluster causes minor changes in the CAT structure at a total simulation time of 200 ps. Cellular assays depicted that SiO2 NPs induce significant cell mortality, change in cellular morphology, cellular internalization, ROS elevation, and apoptosis in hMSCs at higher concentration than 100 µg/mL (170 µM). Conclusion The current results suggest that low concentrations of SiO2 NPs induce no substantial change or mortality against CAT and hMSCs, and potentially useful carriers in CAT delivery to hMSC.
Collapse
Affiliation(s)
- Mina Mousavi
- Department of Biochemistry and Biophysics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Hakimian
- Department of Biochemistry and Biophysics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Twana Ahmed Mustafa
- Department of Medical Laboratory Technology, Health Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Falah Mohammad Aziz
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq.,Department of Medical Analysis, Faculty of Science, Tishk International University, Erbil, Iraq
| | - Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mirsasan Mirpour
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology (RI-SCBT), Tehran, Iran
| | - Osama K Abou-Zied
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, P.O. Box 36, Postal Code 123 Muscat, Oman
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
12
|
Mukerabigwi JF, Ge Z, Kataoka K. Therapeutic Nanoreactors as In Vivo Nanoplatforms for Cancer Therapy. Chemistry 2018; 24:15706-15724. [DOI: 10.1002/chem.201801159] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Jean Felix Mukerabigwi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 China
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine Institute of Industrial Promotion-Kawasaki 3-25-14 Tonomachi Kawasaki-ku Kawasaki 210-0821 Japan
- Policy Alternatives Research Institute The University of Tokyo Tokyo 113-0033 Japan
| |
Collapse
|