1
|
Pazhouhesh Far N, Hajiheidari Varnousafaderani M, Faghihkhorasani F, Etemad S, Abdulwahid AHRR, Bakhtiarinia N, Mousaei A, Dortaj E, Karimi S, Ebrahimi N, Aref AR. Breaking the barriers: Overcoming cancer resistance by targeting the NLRP3 inflammasome. Br J Pharmacol 2024. [PMID: 39394867 DOI: 10.1111/bph.17352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 10/14/2024] Open
Abstract
Inflammation has a pivotal role in the initiation and progression of various cancers, contributing to crucial processes such as metastasis, angiogenesis, cell proliferation and invasion. Moreover, the release of cytokines mediated by inflammation within the tumour microenvironment (TME) has a crucial role in orchestrating these events. The activation of inflammatory caspases, facilitated by the recruitment of caspase-1, is initiated by the activation of pattern recognition receptors on the immune cell membrane. This activation results in the production of proinflammatory cytokines, including IL-1β and IL-18, and participates in diverse biological processes with significant implications. The NOD-Like Receptor Protein 3 (NLRP3) inflammasome holds a central role in innate immunity and regulates inflammation through releasing IL-1β and IL-18. Moreover, it interacts with various cellular compartments. Recently, the mechanisms underlying NLRP3 inflammasome activation have garnered considerable attention. Disruption in NLRP3 inflammasome activation has been associated with a spectrum of inflammatory diseases, encompassing diabetes, enteritis, neurodegenerative diseases, obesity and tumours. The NLRP3 impact on tumorigenesis varies across different cancer types, with contrasting roles observed. For example, colorectal cancer associated with colitis can be suppressed by NLRP3, whereas gastric and skin cancers may be promoted by its activity. This review provides comprehensive insights into the structure, biological characteristics and mechanisms of the NLRP3 inflammasome, with a specific focus on the relationship between NLRP3 and tumour-related immune responses, and TME. Furthermore, the review explores potential strategies for targeting cancers via NLRP3 inflammasome modulation. This encompasses innovative approaches, including NLRP3-based nanoparticles, gene-targeted therapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | | | | | - Sareh Etemad
- Department of Pathology, Faculty of Anatomical Pathology, Ghaem Hospital, University of Medicine, Mashhad, Iran
| | | | | | - Afsaneh Mousaei
- Department of Biology, College of Science, Qaemshahr Branch, Islamic Azad University, Qaem Shahr, Iran
| | - Elahe Dortaj
- Department of Ergonomics, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Lu B, Lim JM, Yu B, Song S, Neeli P, Sobhani N, K P, Bonam SR, Kurapati R, Zheng J, Chai D. The next-generation DNA vaccine platforms and delivery systems: advances, challenges and prospects. Front Immunol 2024; 15:1332939. [PMID: 38361919 PMCID: PMC10867258 DOI: 10.3389/fimmu.2024.1332939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ming Lim
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Boyue Yu
- Department of Environmental Science, Policy, and Management, University of California at Berkeley, Berkeley, CA, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Pavithra K
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Yuan M, Han Z, Liang Y, Sun Y, He B, Chen W, Li F. mRNA nanodelivery systems: targeting strategies and administration routes. Biomater Res 2023; 27:90. [PMID: 37740246 PMCID: PMC10517595 DOI: 10.1186/s40824-023-00425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/26/2023] [Indexed: 09/24/2023] Open
Abstract
With the great success of coronavirus disease (COVID-19) messenger ribonucleic acid (mRNA) vaccines, mRNA therapeutics have gained significant momentum for the prevention and treatment of various refractory diseases. To function efficiently in vivo and overcome clinical limitations, mRNA demands safe and stable vectors and a reasonable administration route, bypassing multiple biological barriers and achieving organ-specific targeted delivery of mRNA. Nanoparticle (NP)-based delivery systems representing leading vector approaches ensure the successful intracellular delivery of mRNA to the target organ. In this review, chemical modifications of mRNA and various types of advanced mRNA NPs, including lipid NPs and polymers are summarized. The importance of passive targeting, especially endogenous targeting, and active targeting in mRNA nano-delivery is emphasized, and different cellular endocytic mechanisms are discussed. Most importantly, based on the above content and the physiological structure characteristics of various organs in vivo, the design strategies of mRNA NPs targeting different organs and cells are classified and discussed. Furthermore, the influence of administration routes on targeting design is highlighted. Finally, an outlook on the remaining challenges and future development toward mRNA targeted therapies and precision medicine is provided.
Collapse
Affiliation(s)
- Mujie Yuan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zeyu Han
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fan Li
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
4
|
Wang X, Lin J, Wang Z, Li Z, Wang M. Possible therapeutic targets for NLRP3 inflammasome-induced breast cancer. Discov Oncol 2023; 14:93. [PMID: 37300757 DOI: 10.1007/s12672-023-00701-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Inflammation plays a major role in the development and progression of breast cancer(BC). Proliferation, invasion, angiogenesis, and metastasis are all linked to inflammation and tumorigenesis. Furthermore, tumor microenvironment (TME) inflammation-mediated cytokine releases play a critical role in these processes. By recruiting caspase-1 through an adaptor apoptosis-related spot protein, inflammatory caspases are activated by the triggering of pattern recognition receptors on the surface of immune cells. Toll-like receptors, NOD-like receptors, and melanoma-like receptors are not triggered. It activates the proinflammatory cytokines interleukin (IL)-1β and IL-18 and is involved in different biological processes that exert their effects. The Nod-Like Receptor Protein 3 (NLRP3) inflammasome regulates inflammation by mediating the secretion of proinflammatory cytokines and interacting with other cellular compartments through the inflammasome's central role in innate immunity. NLRP3 inflammasome activation mechanisms have received much attention in recent years. Inflammatory diseases including enteritis, tumors, gout, neurodegenerative diseases, diabetes, and obesity are associated with abnormal activation of the NLRP3 inflammasome. Different cancer diseases have been linked to NLRP3 and its role in tumorigenesis may be the opposite. Tumors can be suppressed by it, as has been seen primarily in the context of colorectal cancer associated with colitis. However, cancers such as gastric and skin can also be promoted by it. The inflammasome NLRP3 is associated with breast cancer, but there are few specific reviews. This review focuses on the structure, biological characteristics and mechanism of inflammasome, the relationship between NLRP3 in breast cancer Non-Coding RNAs, MicroRNAs and breast cancer microenvironment, especially the role of NLRP3 in triple-negative breast cancer (TNBC). And the potential strategies of using NLRP3 inflammasome to target breast cancer, such as NLRP3-based nanoparticle technology and gene target therapy, are reviewed.
Collapse
Affiliation(s)
- Xixi Wang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Junyi Lin
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Zhe Wang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhi Li
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
- Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, China.
| | - Minghua Wang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
5
|
Yang W, Mixich L, Boonstra E, Cabral H. Polymer-Based mRNA Delivery Strategies for Advanced Therapies. Adv Healthc Mater 2023; 12:e2202688. [PMID: 36785927 PMCID: PMC11469255 DOI: 10.1002/adhm.202202688] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/31/2023] [Indexed: 02/15/2023]
Abstract
Messenger RNA (mRNA)-based therapies offer great promise for the treatment of a variety of diseases. In 2020, two FDA approvals of mRNA-based vaccines have elevated mRNA vaccines to global recognition. However, the therapeutic capabilities of mRNA extend far beyond vaccines against infectious diseases. They hold potential for cancer vaccines, protein replacement therapies, gene editing therapies, and immunotherapies. For realizing such advanced therapies, it is crucial to develop effective carrier systems. Recent advances in materials science have led to the development of promising nonviral mRNA delivery systems. In comparison to other carriers like lipid nanoparticles, polymer-based delivery systems often receive less attention, despite their unique ability to carefully tune their chemical features to promote mRNA protection, their favorable pharmacokinetics, and their potential for targeting delivery. In this review, the central features of polymer-based systems for mRNA delivery highlighting the molecular design criteria, stability, and biodistribution are discussed. Finally, the role of targeting ligands for the future of RNA therapies is analyzed.
Collapse
Affiliation(s)
- Wenqian Yang
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Lucas Mixich
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Eger Boonstra
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Horacio Cabral
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| |
Collapse
|
6
|
Pan P, Li J, Liu X, Hu C, Wang M, Zhang W, Li M, Liu Y. Plasmid containing VEGF-165 and ANG-1 dual genes packaged with fibroin-modified PEI to promote the regeneration of vascular network and dermal tissue. Colloids Surf B Biointerfaces 2023; 224:113210. [PMID: 36841206 DOI: 10.1016/j.colsurfb.2023.113210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/04/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Reducing the cytotoxicity of cationic polymers is the major issue to their use as a gene delivery carrier. In this study, plasmids containing encoding vascular endothelial cell growth factor 165 and angiopoietin-1 were packaged with the conjugates of cationic fibroin (CSF) and polyethylenimine (PEI), instead of packaging pDNA with PEI alone, to prepare nanocomplexes (CSF+PEI)/pDNA. The complexes were loaded into a silk fibroin scaffold to enhance its function to induce microvascular network generation and dermal tissue regeneration. The results of transfecting EA.hy926 cells with the complexes in vitro showed that (CSF+PEI)/pDNA had a stronger transfection ability than PEI/pDNA. Importantly, compared with PEI as the gene carrier alone, the cell viability was significantly increased and the cytotoxicity was effectively reduced after the conjugate of CSF and PEI was used as the gene carrier. The results of angiogenesis in chick embryo chorioallantoic membranes showed that compared with scaffolds loaded with PEI/pDNA, the neovascularization ratio in scaffolds loaded with (CSF+PEI)/pDNA was significantly increased. In vivo experimental results of scaffolds implantation for full-thickness skin defects in SD rats showed that, compared with loading PEI/pDNA complex, loading (CSF+PEI)/pDNA complex in the scaffold more effectively promoted the formation of vascular network in the scaffold and accelerated the regeneration of dermal tissue. The gene delivery system established in this study has application potential not only in the regeneration of vascular-containing tissues, but also in tumor gene therapy.
Collapse
Affiliation(s)
- Peng Pan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Jing Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Xueping Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Cheng Hu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Mengmeng Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Wenjing Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China
| | - Mingzhong Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China.
| | - Yu Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, China.
| |
Collapse
|
7
|
Sui S, Wang H, Song J, Tai W. Development of a spermine lipid for transient antibody expression. Bioorg Med Chem 2023; 78:117114. [PMID: 36563514 DOI: 10.1016/j.bmc.2022.117114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Transient expression is the only way to quickly obtain a small scale of antibodies for biomedical research and therapeutic evaluation. The agents for transfecting the suspension cells, e.g. PEI or commercial agents, either lack efficiency or excessively expensive. Herein, a novel spermine-based lipid was developed and fabricated into a cationic liposome for antibody expression. This new transfection agent, designated as sperminoliposome, is feasible, cheap, and highly effective to produce antibodies. Compared to PEI, a 3 times higher yield of antibody was obtained by sperminoliposome during the transient expression of cetuximab in suspension 293F cells. Characterizations confirmed that the expressed antibody is fully functional and eligible for further research. Our study provides an effective tool for the rapid production of antibodies economically and feasibly.
Collapse
Affiliation(s)
- Shaowei Sui
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hao Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiajie Song
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
8
|
Evaluation of the Cytotoxicity of Cationic Polymers on Glioblastoma Cancer Stem Cells. J Funct Biomater 2022; 14:jfb14010017. [PMID: 36662064 PMCID: PMC9862959 DOI: 10.3390/jfb14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Cationic polymers such as polyethylenimine (PEI) have found a pervasive place in laboratories across the world as gene delivery agents. However, their applications are not limited to this role, having found a place as delivery agents for drugs, in complexes known as polymer-drug conjugates (PDCs). Yet a potentially underexplored domain of research is in their inherent potential as anti-cancer therapeutic agents, which has been indicated by several studies. Even more interesting is the recent observation that certain polycations may present a significantly greater toxicity towards the clinically important cancer stem cell (CSC) niche than towards more differentiated bulk tumour cells. These cells, which possess the stem-like characteristics of self-renewal and differentiation, are highly implicated in cancer drug resistance, tumour recurrence and poor clinical prognosis. The search for compounds which may target and eliminate these cells is thus of great research interest. As such, the observation in our previous study on a PEI-based PDC which showed a considerably higher toxicity of PEI towards glioblastoma CSCs (GSCs) than on more differentiated glioma (U87) cells led us to investigate other cationic polymers for a similar effect. The evaluation of the toxicity of a range of different types of polycations, and an investigation into the potential source of GSC's sensitivity to such compounds is thus described.
Collapse
|
9
|
Jansen EM, Frijlink HW, Hinrichs WLJ, Ruigrok MJR. Are inhaled mRNA vaccines safe and effective? A review of preclinical studies. Expert Opin Drug Deliv 2022; 19:1471-1485. [DOI: 10.1080/17425247.2022.2131767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Evalyne M Jansen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Wouter LJ Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Mitchel JR Ruigrok
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
10
|
Street STG, Chrenek J, Harniman RL, Letwin K, Mantell JM, Borucu U, Willerth SM, Manners I. Length-Controlled Nanofiber Micelleplexes as Efficient Nucleic Acid Delivery Vehicles. J Am Chem Soc 2022; 144:19799-19812. [PMID: 36260789 DOI: 10.1021/jacs.2c06695] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Micelleplexes show great promise as effective polymeric delivery systems for nucleic acids. Although studies have shown that spherical micelleplexes can exhibit superior cellular transfection to polyplexes, to date there has been no report on the effects of micelleplex morphology on cellular transfection. In this work, we prepared precision, length-tunable poly(fluorenetrimethylenecarbonate)-b-poly(2-(dimethylamino)ethyl methacrylate) (PFTMC16-b-PDMAEMA131) nanofiber micelleplexes and compared their properties and transfection activity to those of the equivalent nanosphere micelleplexes and polyplexes. We studied the DNA complexation process in detail via a range of techniques including cryo-transmission electron microscopy, atomic force microscopy, dynamic light scattering, and ζ-potential measurements, thereby examining how nanofiber micelleplexes form, as well the key differences that exist compared to nanosphere micelleplexes and polyplexes in terms of DNA loading and colloidal stability. The effects of particle morphology and nanofiber length on the transfection and cell viability of U-87 MG glioblastoma cells with a luciferase plasmid were explored, revealing that short nanofiber micelleplexes (length < ca. 100 nm) were the most effective delivery vehicle examined, outperforming nanosphere micelleplexes, polyplexes, and longer nanofiber micelleplexes as well as the Lipofectamine 2000 control. This study highlights the potential importance of 1D micelleplex morphologies for achieving optimal transfection activity and provides a fundamental platform for the future development of more effective polymeric nucleic acid delivery vehicles.
Collapse
Affiliation(s)
- Steven T G Street
- School of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.,Department of Chemistry, University of Victoria, Victoria, BC V8W 3V6, Canada.,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada
| | - Josie Chrenek
- Department of Mechanical Engineering, Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | | | - Keiran Letwin
- Department of Mechanical Engineering, Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Judith M Mantell
- Wolfson Bioimaging Facility, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, U.K
| | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.,GW4 Facility for High-Resolution Electron Cryo-Microscopy, 24 Tyndall Ave, Bristol BS8 1TQ, U.K
| | - Stephanie M Willerth
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.,Department of Mechanical Engineering, Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ian Manners
- Department of Chemistry, University of Victoria, Victoria, BC V8W 3V6, Canada.,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada
| |
Collapse
|
11
|
Polyethylenimine, an Autophagy-Inducing Platinum-Carbene-Based Drug Carrier with Potent Toxicity towards Glioblastoma Cancer Stem Cells. Cancers (Basel) 2022; 14:cancers14205057. [PMID: 36291841 PMCID: PMC9599868 DOI: 10.3390/cancers14205057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
The difficulty involved in the treatment of many tumours due to their recurrence and resistance to chemotherapy is tightly linked to the presence of cancer stem cells (CSCs). This CSC sub-population is distinct from the majority of cancer cells of the tumour bulk. Indeed, CSCs have increased mitochondrial mass that has been linked to increased sensitivity to mitochondrial targeting compounds. Thus, a platinum-based polyethylenimine (PEI) polymer-drug conjugate (PDC) was assessed as a potential anti-CSC therapeutic since it has previously displayed mitochondrial accumulation. Our results show that CSCs have increased specific sensitivity to the PEI carrier and to the PDC. The mechanism of cell death seems to be necrotic in nature, with an absence of apoptotic markers. Cell death is accompanied by the induction of a protective autophagy. The interference in the balance of this pathway, which is highly important for CSCs, may be responsible for a partial reversion of the stem-like phenotype observed with prolonged PEI and PDC treatment. Several markers also indicate the cell death mode to be capable of inducing an anti-cancer immune response. This study thus indicates the potential therapeutic perspectives of polycations against CSCs.
Collapse
|
12
|
Kim HJ, Cho HB, Lee S, Lyu J, Kim HR, Lee S, Park JI, Park KH. Strategies for accelerating osteogenesis through nanoparticle-based DNA/mitochondrial damage repair. Am J Cancer Res 2022; 12:6409-6421. [PMID: 36168629 PMCID: PMC9475457 DOI: 10.7150/thno.77089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/18/2022] [Indexed: 11/05/2022] Open
Abstract
The efficiency of gene therapy is often dictated by the gene delivery system. Cationic polymers are essential elements of gene delivery systems. The relatively cheap cationic polymer, polyethyleneimine, has high gene delivery efficiency and is often used for gene delivery. However, the efficiency of gene therapy with polyethyleneimine-pDNA polyplex (PEI) is low. Human mesenchymal stem cells transfected with polyethyleneimine and a plasmid carrying the important osteogenic differentiation gene runt-related transcription factor 2 (RUNX2) accumulated DNA double-strand breaks and mitochondrial damage proportional to the amount of polyethyleneimine, reducing viability. Genomic/cellular stabilizer mediating RUNX2 delivery (GuaRD), a new reagent incorporating RS-1 NPs developed in this study, promoted DNA repair and prevented the accumulation of cell damage, allowing the delivery of pRUNX2 into hMSCs. while maintaining genome and mitochondrial stability. DNA damage was significantly lower and the expression of DNA repair-related genes significantly higher with GuaRD than with PEI. In addition, GuaRD improved mitochondrial stability, decreased the level of reactive oxygen species, and increased mitochondrial membrane potential. Osteogenic extracellular matrix (ECM) expression and calcification were higher with GuaRD than with PEI, suggesting improved osteogenic differentiation. These results indicate that lowering the cytotoxicity of PEI and improving cell stability are key to overcoming the limitations of conventional gene therapy, and that GuaRD can help resolve these limitations.
Collapse
Affiliation(s)
- Hye Jin Kim
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hui Bang Cho
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Sujin Lee
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Jiyon Lyu
- School of Medicine, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hye-Ryoung Kim
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Sujeong Lee
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Ji-In Park
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Keun-Hong Park
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| |
Collapse
|
13
|
Meenakshi Sundaram DN, Plianwong S, Kc R, Ostergaard H, Uludağ H. In Vitro Cytotoxicity and Cytokine Production by Lipid-Substituted Low Molecular Weight Branched PEIs Used for Gene Delivery. Acta Biomater 2022; 148:279-297. [PMID: 35738388 DOI: 10.1016/j.actbio.2022.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022]
Abstract
Lipid-modified low molecular weight branched polyethyleneimines (PEIs) are promising non-viral gene delivery systems that have been successfully explored for treatment of various diseases. The present study aims to determine in vitro safety of these delivery systems based on assessment of cytotoxicity with peripheral blood mononuclear cells (PBMCs), hemolysis with human red blood cells (RBC) and cytokine secretion from several sources of PBMCs. The viability of cells treated with lipopolymer/pDNA complexes was dependent on the polymer:pDNA ratio used but remained low at therapeutically relevant concentrations for most lipopolymers, except for the propionic acid substituted PEIs. The extent of hemolysis was minimal and below the accepted safety levels with most of the lipopolymers; however, some linoleic acid substituted PEIs yielded significant hemolysis activity. Unlike strong cytokine secretion from PMA/IO stimulated cells, most lipopolymer/pDNA complexes remained non-responsive, showing minimal changes in cytokine secretion (TNF-α, IL-6 and IFN-γ) irrespective of the lipopolymer/pDNA formulations. The 0.6 kDa PEI with lauric acid substituent displayed slight cytokine upregulation, however it remained low relative to the positive controls. This study demonstrated that the lipid modified LMW PEIs are expected to be safe in contact with blood components. However, close attention to lipopolymer concentration and ratio of polymer to pDNA in formulations might be required for individual lipopolymers for optimal safety response in nucleic acid therapies. STATEMENT OF SIGNIFICANCE: : This manuscript investigated the safety aspects of various lipid modified low molecular weight polyethylenimine (LMW-PEI) polymers employed for pDNA delivery through in vitro studies. Using peripheral blood mononuclear cells (PBMCs) from multiple sources, we show that the hemolysis ability was minimal for most polymers, although a particular lipid substituent (linoleic acid) at specific ratios exhibited hemolysis. The levels of pro-inflammatory cytokines (TNF-α, IL-6 and IFN-γ) were slightly upregulated only with a lauric acid substituted 0.6PEI, but remained low relative to positive control treatments. We further report the beneficial effect of polyacrylic acid additives on hemolysis and cytokine secretion to a reasonable extent. This study confirms the feasibility of using LMW-PEI as safe delivery agents for various therapeutic purposes.
Collapse
Affiliation(s)
| | - Samarwadee Plianwong
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Remant Kc
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - Hanne Ostergaard
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
14
|
Karam M, Fahs D, Maatouk B, Safi B, Jaffa AA, Mhanna R. Polymeric nanoparticles in the diagnosis and treatment of myocardial infarction: Challenges and future prospects. Mater Today Bio 2022; 14:100249. [PMID: 35434594 PMCID: PMC9006854 DOI: 10.1016/j.mtbio.2022.100249] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of morbidity and mortality worldwide. Despite extensive efforts to provide early diagnosis and adequate treatment regimens, detection of MI still faces major limitations and pathological MI complications continue to threaten the recovery of survivors. Polymeric nanoparticles (NPs) represent novel noninvasive drug delivery systems for the diagnosis and treatment of MI and subsequent prevention of fatal heart failure. In this review, we cover the recent advances in polymeric NP-based diagnostic and therapeutic approaches for MI and their application as multifunctional theranostic tools. We also discuss the in vivo behavior and toxicity profile of polymeric NPs, their application in noninvasive imaging, passive, and active drug delivery, and use in cardiac regenerative therapy. We conclude with the challenges faced with polymeric nanosystems and suggest future efforts needed for clinical translation.
Collapse
Affiliation(s)
- Mia Karam
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Duaa Fahs
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Batoul Maatouk
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Brouna Safi
- Department of Chemical Engineering, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
| | - Ayad A. Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
| |
Collapse
|
15
|
Nanoparticle-based delivery strategies of multifaceted immunomodulatory RNA for cancer immunotherapy. J Control Release 2022; 343:564-583. [DOI: 10.1016/j.jconrel.2022.01.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
|
16
|
Street STG, He Y, Harniman RL, Garcia-Hernandez JD, Manners I. Precision polymer nanofibers with a responsive polyelectrolyte corona designed as a modular, functionalizable nanomedicine platform. Polym Chem 2022. [DOI: 10.1039/d2py00152g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We describe the development of a modular, functionalizable platform for biocompatible core-shell block copolymer nanofibers of controlled length (22 nm – 1.3 μm) and low dispersity produced via living crystallization-driven...
Collapse
|
17
|
Zhang Y, Yuan Z, Jin Y, Zhang W, Yuan WE. Novel Fluorinated Spermine and Small Molecule PEI to Deliver Anti-PD-L1 and Anti-VEGF siRNA for Highly Efficient Tumor Therapy. Pharmaceutics 2021; 13:2058. [PMID: 34959340 PMCID: PMC8708240 DOI: 10.3390/pharmaceutics13122058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 11/17/2022] Open
Abstract
Small interfering RNA (siRNA) can specifically silence disease gene expression. This project investigated the overexpression of programmed death receptor ligand 1 (PD-L1) and vascular endothelial growth factor (VEGF) on the surface of tumor cells. However, the main obstacle to the development of gene therapy drugs is the lack of an efficient delivery vector, which should be able to overcome multiple delivery barriers and protect siRNA to enter the target cells. Therefore, a novel fluorine-modified endogenous molecular carrier TFSPEI was constructed by linking fluorinated groups with hydrophobic and hydrophilic characteristics on the surface of PEI and spermine. The results showed that lower toxicity, higher endocytosis, and silencing efficiency were achieved. We found that the inhibition of VEGF targets can indirectly activate the immune response to promote the tumor-killing and invasion effects of T cells. The combined delivery of anti-VEGF siRNA and anti-PD-L1 siRNA could inhibit the expression of corresponding proteins, restore the anti-tumor function of T cells and inhibit the growth of neovascularization, and obtained significant anti-tumor effects. Therefore, this safe and efficient fluorinated spermine and small molecule PEI-based anti-PD-L1 and anti-VEGF siRNA delivery system is expected to provide a new strategy for gene therapy of tumors.
Collapse
Affiliation(s)
| | | | | | | | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.Z.); (Z.Y.); (Y.J.); (W.Z.)
| |
Collapse
|
18
|
Park JJ, Kim Y, Lee C, Kim D, Choi W, Kwon H, Kim JH, Hwang KS, Lee JY. Morphological Analysis of PSMA/PEI Core-Shell Nanoparticles Synthesized by Soap-Free Emulsion Polymerization. NANOMATERIALS 2021; 11:nano11081958. [PMID: 34443787 PMCID: PMC8402240 DOI: 10.3390/nano11081958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/17/2022]
Abstract
Emulsion polymerization presents the disadvantage that the physical properties of polymer particles are altered by surfactant adsorption. Therefore, in the soap-free emulsion polymerization method, a hydrophilic initiator is utilized while inducing repulsion among particles on the polymer particle surface, resulting in stable polymer particle production. In this study, we developed a methodology wherein spherical and uniform poly(styrene-co-maleic anhydride) (PSMA)/polyethyleneimine (PEI) core–shell nanoparticles were prepared. Further, their morphology was analyzed. During PSMA polymerization, the addition of up to 30% maleic anhydride (MA) resulted in stable polymerization. In PSMA/PEI nanoparticle fabrication, the number of reactants increased with increased initial monomer feed amounts; consequently, the particle size increased, and as the complete monomer consumption time increased, the particle distribution widened. The styrene (St) copolymer acted as a stabilizer, reducing particle size and narrowing particle distribution. Furthermore, the monomers were more rapidly consumed at high initiator concentrations, irrespective of the initiator used, resulting in increased particle stability and narrowed particle distribution. The shell thickness and particle size were PEI feed ratio dependent, with 0.08 being the optimal PEI-to-MA ratio. The fabricated nanoparticles possess immense potential for application in environmental science and in chemical and health care industries.
Collapse
Affiliation(s)
- Jae-Jung Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 120749, Korea; (J.-J.P.); (D.K.); (W.C.); (H.K.); (J.-H.K.)
| | - Yongsoo Kim
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
| | - Chanmin Lee
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
| | - Donghyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 120749, Korea; (J.-J.P.); (D.K.); (W.C.); (H.K.); (J.-H.K.)
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
| | - Wonjun Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 120749, Korea; (J.-J.P.); (D.K.); (W.C.); (H.K.); (J.-H.K.)
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
| | - Hyukjun Kwon
- Department of Chemical and Biomolecular Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 120749, Korea; (J.-J.P.); (D.K.); (W.C.); (H.K.); (J.-H.K.)
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
| | - Jung-Hyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 120749, Korea; (J.-J.P.); (D.K.); (W.C.); (H.K.); (J.-H.K.)
| | - Ki-Seob Hwang
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
- Correspondence: (K.-S.H.); (J.-Y.L.); Tel.: +82-41-5898-424 (J.-Y.L.)
| | - Jun-Young Lee
- Research Institute of Sustainable Manufacturing System, Intelligent Sustainable Materials R&D Group, Korea Institute of Industrial Technology, 89 Yangdaegiro-gil, Ipjang-myeon, Seobuk-gu, Cheonan-si 31056, Korea; (Y.K.); (C.L.)
- Correspondence: (K.-S.H.); (J.-Y.L.); Tel.: +82-41-5898-424 (J.-Y.L.)
| |
Collapse
|
19
|
Rezaei A, Hashemi E. A pseudohomogeneous nanocarrier based on carbon quantum dots decorated with arginine as an efficient gene delivery vehicle. Sci Rep 2021; 11:13790. [PMID: 34215792 PMCID: PMC8253742 DOI: 10.1038/s41598-021-93153-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
A pseudohomogeneous carrier as an emerging term refers to subnanometric carbon-based vehicle with a high ability to interact with genetic materials to form stable carboplex and successfully transfer them into the cell which will result in inhibiting or expressing of therapeutic genes. Chitosan is a non-toxic polyaminosaccharide used as a precursor in the presence of citric acid to produce carbon quantum dots (CQDs), which decorated with arginine as a surface passivation agent with high amine density in hydrothermal methodology. The Arginine-CQDs are comprehensively characterized by Fourier-transform infrared spectroscopy (FT-IR), Ultraviolet-visible spectroscopy (UV-vis), Atomic force microscopy (AFM), field emission scanning electron microscope (FE-SEM), Energy-dispersive X-ray (EDX) mapping, fluorescence, High-resolution transmission electron microscopy (HR-TEM), zeta potential and X-ray powder diffraction (XRD). In this regard, for the first time, carboplex are formed by electrostatic conjugating of Arginine-CQDs with DNA to protect it from enzymatic degradation. Moreover, the carboplex, like the chitosan precursor, has not shown toxicity against AGS cell line. Interestingly, the Arginine-CQDs have exhibited an excellent ability to overcome cell barriers to deliver into cells compared to chitosan at the same weight ratio. The Arginine-CQDs/pEGFP (W/W) nanocomplex, not only lead to transfection with a relatively higher efficiency than PEI polymer, which is the "golden standard", but carboplex also demonstrates no significant toxicity. Indeed, the EGFP expression level has reached to 2.4 ± 0.2 via Arginine-CQDs carboplex at W/W 50 weight ratio. To the best of our knowledge, this is the first report includes chitosan-based CQDs functionalized by arginine which is applied to serve as a pseudohomogeneous vehicle for gene transfection.
Collapse
Affiliation(s)
- Aram Rezaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ehsan Hashemi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 14965-16, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Acri TM, Laird NZ, Jaidev LR, Meyerholz DK, Salem AK, Shin K. Nonviral Gene Delivery Embedded in Biomimetically Mineralized Matrices for Bone Tissue Engineering. Tissue Eng Part A 2020; 27:1074-1083. [PMID: 33086991 DOI: 10.1089/ten.tea.2020.0206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Research in bone tissue engineering aims to design materials that are effective at generating bone without causing significant side effects. The osteogenic potential of combining matrices and protein growth factors has been well documented, however, improvements are necessary to achieve optimal therapeutic benefits upon clinical translation. In this article, rat calvarial defects were treated with gene-activated matrices (GAMs). The GAMs used were collagen sponges mineralized with a simulated body fluid (SBF) containing a nonviral gene delivery system. Both in vitro and in vivo studies were performed to determine the optimal mode of gene delivery. After 6 weeks, the defects were extracted to assess bone formation and tissue quality through histological and microcomputed tomography analyses. The optimal GAM consisted of a collagen sponge with polyethylenimine plasmid DNA (PEI-pDNA) complexes embedded in a calcium phosphate coating produced by SBF, which increased total bone formation by 39% compared with 19% for control samples. A follow-up in vivo study was performed to optimize the ratio of growth factors included in the GAM. The optimal ratio for supporting bone formation after 6 weeks of implantation was five parts of pBMP-2 to three parts pFGF-2. These studies demonstrated that collagen matrices biomimetically mineralized and activated with plasmids encoding fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) can optimally improve bone regeneration outcomes. Impact statement Bone tissue engineering has explored both nonviral gene delivery and the concept of biomimetic mineralization. In this study, we combined these two concepts to further enhance bone regeneration outcomes. We demonstrated that embedding polyethylenimine (PEI)-based gene delivery within a mineral layer formed from simulated body fluid (SBF) immersion can increase bone formation rates. We also demonstrated that the ratio of growth factors utilized for matrix fabrication can impact the amount of bone formed in the defect site. This research highlights a combined approach using SBF and nonviral gene delivery both in vitro and in vivo and prepares the way for future optimization of synthetic gene activated matrices.
Collapse
Affiliation(s)
- Timothy M Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Noah Z Laird
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Leela R Jaidev
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Kyungsup Shin
- Department of Orthodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| |
Collapse
|
21
|
Rondon EP, Benabdoun HA, Vallières F, Segalla Petrônio M, Tiera MJ, Benderdour M, Fernandes JC. Evidence Supporting the Safety of Pegylated Diethylaminoethyl-Chitosan Polymer as a Nanovector for Gene Therapy Applications. Int J Nanomedicine 2020; 15:6183-6200. [PMID: 32922001 PMCID: PMC7450204 DOI: 10.2147/ijn.s252397] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Diethylaminoethyl-chitosan (DEAE-CH) is a derivative with excellent potential as a delivery vector for gene therapy applications. The aim of this study is to evaluate its toxicological profile for potential future clinical applications. METHODS An endotoxin-free chitosan (CH) modified with DEAE, folic acid (FA) and polyethylene glycol (PEG) was used to complex small interfering RNA (siRNA) and form nanoparticles (DEAE12-CH-PEG-FA2/siRNA). Based on the guidelines from the International Organization for Standardization (ISO), the American Society for Testing and Materials (ASTM), and the Nanotechnology Characterization Laboratory (NCL), we evaluated the effects of the interaction between these nanoparticles and blood components. In vitro screening assays such as hemolysis, hemagglutination, complement activation, platelet aggregation, coagulation times, cytokine production, and reactive species, such as nitric oxide (NO) and reactive oxygen species (ROS), were performed on erythrocytes, plasma, platelets, peripheral blood mononuclear cells (PBMC) and Raw 264.7 macrophages. Moreover, MTS and LDH assays on Raw 264.7 macrophages, PBMC and MG-63 cells were performed. RESULTS Our results show that a targeted theoretical plasma concentration (TPC) of DEAE12-CH-PEG-FA2/siRNA nanoparticles falls within the guidelines' thresholds: <1% hemolysis, 2.9% platelet aggregation, no complement activation, and no effect on coagulation times. ROS and NO production levels were comparable to controls. Cytokine secretion (TNF-α, IL-6, IL-4, and IL-10) was not affected by nanoparticles except for IL-1β and IL-8. Nanoparticles showed a slight agglutination. Cell viability was >70% for TPC in all cell types, although LDH levels were statistically significant in Raw 264.7 macrophages and PBMC after 24 and 48 h of incubation. CONCLUSION These DEAE12-CH-PEG-FA2/siRNA nanoparticles fulfill the existing ISO, ASTM and NCL guidelines' threshold criteria, and their low toxicity and blood biocompatibility warrant further investigation for potential clinical applications.
Collapse
Affiliation(s)
- Elsa Patricia Rondon
- Orthopedic Research Laboratory, Hôpital Du Sacré-Cœur De Montréal, Université De Montréal, Montréal, Québec, Canada
| | - Houda Abir Benabdoun
- Orthopedic Research Laboratory, Hôpital Du Sacré-Cœur De Montréal, Université De Montréal, Montréal, Québec, Canada
| | - Francis Vallières
- Orthopedic Research Laboratory, Hôpital Du Sacré-Cœur De Montréal, Université De Montréal, Montréal, Québec, Canada
| | - Maicon Segalla Petrônio
- Institute of Biosciences, Humanities and Exact Sciences, Department of Chemistry and Environmental Sciences, UNESP-São Paulo State University, São José Do Rio Preto, São Paulo State, Brazil
| | - Marcio José Tiera
- Institute of Biosciences, Humanities and Exact Sciences, Department of Chemistry and Environmental Sciences, UNESP-São Paulo State University, São José Do Rio Preto, São Paulo State, Brazil
| | - Mohamed Benderdour
- Orthopedic Research Laboratory, Hôpital Du Sacré-Cœur De Montréal, Université De Montréal, Montréal, Québec, Canada
| | - Julio Cesar Fernandes
- Orthopedic Research Laboratory, Hôpital Du Sacré-Cœur De Montréal, Université De Montréal, Montréal, Québec, Canada
| |
Collapse
|
22
|
Roy S, Zhu D, Parak WJ, Feliu N. Lysosomal Proton Buffering of Poly(ethylenimine) Measured In Situ by Fluorescent pH-Sensor Microcapsules. ACS NANO 2020; 14:8012-8023. [PMID: 32568521 DOI: 10.1021/acsnano.9b10219] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Poly(ethylenimine) (PEI) is frequently used as transfection agent for delivery of nucleic acids to the cytosol. After endocytosis of complexes of PEI and nucleic acids, a fraction of them can escape endosomes/lysosomes and reach the cytosol. One proposed mechanism is the so-called proton sponge effect, which involves buffering of the lysosomal pH by PEI. There are however also reports that report the absence of such buffering. In this work, the buffering capacity of PEI of the lysosomal pH was investigated in situ by combining PEI and pH-sensing ratiometric fluorophores in a single carrier particle. As carrier particles, hereby capsules were used, which were composed of polyelectrolyte walls based on layer-by-layer assembly, with the pH sensors located inside the capsule cavities. In this way, the local pH around individual particles could be monitored during the whole process of endocytosis. Results demonstrate the pH-buffering capability of PEI, which prevents the strong acidification of lysosomes containing PEI. This effect was related to the presence of PEI and was not related to the overall charge of the carrier particles. In case PEI was added in molecular form, no buffering of pH could be observed by endocytosed encapsulated pH-sensing ratiometric fluorophores. Co-localization experiments demonstrated that this was due to the fact that internalized free PEI and the encapsulated pH-sensing ratiometric fluorophores were not located in the same lysosomes. Missing co-localization might explain why also in other studies no pH buffering was found; in the case of co-delivery of PEI, the pH sensors could be clearly observed.
Collapse
Affiliation(s)
- Sathi Roy
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| | - Dingcheng Zhu
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
- CIC Biomagune, Miramon Pasealekua 182, 20014 San Sebastian, Spain
| | - Neus Feliu
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| |
Collapse
|
23
|
Hobson JJ, Rannard SP, Owen A, Liptrott NJ. Safety assessment of a new nanoemulsion-based drug-delivery system reveals unexpected drug-free anticoagulant activity. Nanomedicine (Lond) 2020; 15:1361-1373. [PMID: 32484393 DOI: 10.2217/nnm-2019-0447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: A preclinical safety assessment of a novel nanoemulsion drug-delivery system, initially developed to improve the posology of efavirenz (EFV), was conducted with a specific focus on possible immunological and hematological complications. Materials & methods: Assessment of common acute toxicities, such as complement activation and cytokine secretion, was performed using validated assays known to have good correlation with in vivo end points. Results & conclusion: Compared with a standard aqueous solution of EFV, the EFV nanoemulsion showed no significant effect on immune cell function or phenotype. Prolongation of activated partial thromboplastin time was observed for EFV-loaded nanoemulsions (88% at 4 μg/ml) as well as unloaded nanoemulsions (52%) highlighting the potential for drug-free anticoagulant activity and warranting further investigation of the mechanism and utility of these materials.
Collapse
Affiliation(s)
- James J Hobson
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD, UK
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD, UK
| | - Andrew Owen
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GF, UK.,European Nanomedicine Characterisation Laboratory, University of Liverpool, Liverpool, L7 3NY, UK
| | - Neill J Liptrott
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GF, UK.,European Nanomedicine Characterisation Laboratory, University of Liverpool, Liverpool, L7 3NY, UK
| |
Collapse
|
24
|
Liang S, Chen Y, Zhang S, Cao Y, Duan J, Wang Y, Sun Z. RhB-encapsulating silica nanoparticles modified with PEG impact the vascular endothelial function in endothelial cells and zebrafish model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 711:134493. [PMID: 32000304 DOI: 10.1016/j.scitotenv.2019.134493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/15/2019] [Indexed: 06/10/2023]
Abstract
Silica nanoparticles (SiNPs) have been widely used in human health related products, such as food additives, cosmetics and even drug delivery, gene therapy or bioimaging. Recently, a first-in-human clinical trial based on polyethylene glycol (PEG)-modified SiNPs had been approved by US FDA to trace melanoma. However, as a nano-based drug delivery system, its biocompatibility and vascular toxicity are still largely unknown. Thus, we synthesized the fluorescent SiNPs to explore the biocompatibility and vascular endothelial function, and compare different biological effects caused by PEG-modified and unmodified SiNPs in cells and zebrafish model. The characterizations of SiNPs and PEG-modified SiNPs were analyzed by TEM, SEM, AFM and DLS, which exhibited relatively good stable and dispersive. Compared with SiNPs, PEG-modified SiNPs had markedly reduced the inflammatory response and vascular damage in Tg (fli-1: EGFP) and Tg (mpo: GFP) transgenic zebrafish lines, respectively. Consistent with the in vivo results, the PEG-modified SiNPs had been found to significantly decline the levels of ROS, inflammatory cytokines and mitochondrial-mediated apoptosis in vascular endothelial cells compared to SiNPs, and the ROS scavenger NAC could effectively alleviate the above adverse effects induced by nanoparticles. Our results suggested that the PEG-modified SiNPs could become more safety via increasing the biocompatibility and decreasing cellular toxicities in living organisms.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Shiming Zhang
- Department of Chemistry, Renmin University of China, Beijing 100872, PR China
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Yapei Wang
- Department of Chemistry, Renmin University of China, Beijing 100872, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
25
|
da Silva AL, Silva LA, Cruz FF, Rocco PRM, Morales MM. Application of novel nanotechnologies in asthma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:159. [PMID: 32309307 PMCID: PMC7154405 DOI: 10.21037/atm.2019.12.40] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Adriana Lopes da Silva
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luisa Andrade Silva
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Gandhi NS, Godeshala S, Koomoa-Lange DLT, Miryala B, Rege K, Chougule MB. Bioreducible Poly(Amino Ethers) Based mTOR siRNA Delivery for Lung Cancer. Pharm Res 2018; 35:188. [PMID: 30105526 DOI: 10.1007/s11095-018-2460-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE Lung cancer is one of the leading causes of deaths in the United States, but currently available therapies for lung cancer are associated with reduced efficacy and adverse side effects. Small interfering RNA (siRNA) can knock down the expression of specific genes and result in therapeutic efficacy in lung cancer. Recently, mTOR siRNA has been shown to induce apoptosis in NSCLC cell lines but its use is limited due to poor stability in biological conditions. METHODS In this study, we modified an aminoglyocisde-derived cationic poly (amino-ether) by introducing a thiol group using Traut's reagent to generate a bio-reducible modified-poly (amino-ether) (mPAE). The mPAE polymer was used to encapsulate mTOR siRNA by nanoprecipitation method, resulting in the formation of stable and bio-reducible nanoparticles (NPs) which possessed an average diameter of 114 nm and a surface charge of approximately +27 mV. RESULTS The mTOR siRNA showed increased release from the mTS-mPAE NPs in the presence of 10 mM glutathione (GSH). The polymeric mTS-mPAE-NPs were also capable of efficient gene knockdown (60 and 64%) in A549 and H460 lung cancer cells, respectively without significant cytotoxicity at 30 μg/ml concentrations. The NPs also showed time-dependent cellular uptake for up to 24 h as determined using flow cytometry. Delivery of the siRNA using these NPs also resulted in significant inhibition of A549 and H460 cell proliferation in vitro, respectively. CONCLUSIONS The results demonstrate that the mPAE polymer based NPs show strong potential for siRNA delivery to lung cancer cells. It is anticipated that future modification can help improve the efficacy of nucleic acid delivery, leading to higher inhibition of lung cancer growth in vitro and in vivo.
Collapse
Affiliation(s)
- Nishant S Gandhi
- Department of Pharmaceutical Sciences, The Daniel K Inouye College of pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA
- Translational Bio-pharma Engineering Nanodelivery Research Laboratory, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, Faser Hall, University of Mississippi, University, MS, 38677, USA
| | - Sudhakar Godeshala
- Chemical Engineering, Arizona State University, Tempe, AZ, 85287-6106, USA
| | - Dana-Lynn T Koomoa-Lange
- Department of Pharmaceutical Sciences, The Daniel K Inouye College of pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA
| | - Bhavani Miryala
- Chemical Engineering, Arizona State University, Tempe, AZ, 85287-6106, USA
| | - Kaushal Rege
- Chemical Engineering, Arizona State University, Tempe, AZ, 85287-6106, USA
| | - Mahavir B Chougule
- Department of Pharmaceutical Sciences, The Daniel K Inouye College of pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA.
- Translational Bio-pharma Engineering Nanodelivery Research Laboratory, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, Faser Hall, University of Mississippi, University, MS, 38677, USA.
- Pii Center for Pharmaceutical Technology, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, 38677, USA.
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
27
|
Lin CW, Jan MS, Kuo JHS. The microRNA expression profiles in extracellular vesicles from HeLa cancer cells in response to cationic lipid- or polyethylenimine-mediated gene delivery. J Drug Target 2018; 27:94-102. [DOI: 10.1080/1061186x.2018.1491977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Chia-Wei Lin
- Institute of Biochemistry, Microbiology and Immunology, Taichung, Taiwan
| | - Ming-Shiou Jan
- Institute of Biochemistry, Microbiology and Immunology, Taichung, Taiwan
- Immunology Research Center, Medical College of Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Immunology, and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jung-Hua Steven Kuo
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
28
|
Yamamoto T, Tsunoda M, Ozono M, Watanabe A, Kotake K, Hiroshima Y, Yamada A, Terada H, Shinohara Y. Polyethyleneimine renders mitochondrial membranes permeable by interacting with negatively charged phospholipids in them. Arch Biochem Biophys 2018; 652:9-17. [PMID: 29886045 DOI: 10.1016/j.abb.2018.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 01/21/2023]
Abstract
Polyethyleneimines (PEIs) are used for transfection of cells with nucleic acids. Meanwhile, the interaction of PEI with mitochondria causes cytochrome c release prior to apoptosis; the mechanisms how PEI causes this permeabilization of mitochondrial membranes and the release of cytochrome c remain unclear. To clarify these mechanisms, we examined the effects of branched-type PEI and linear-type PEI, each of which was 25 kDa in size, on mitochondria. The permeabilization potency of mitochondrial membranes by branched PEI was stronger than that by linear PEI. The permeabilization by PEIs were insensitive to permeability-transition inhibitors, indicating that PEI-induced permeabilization was not attributed to permeability transition. Meanwhile, PEIs caused permeabilization of artificial lipid vesicles; again, the permeabilization potency of branched PEI was stronger than that of linear PEI. Such a difference in this potency was close to that in the case of isolated mitochondria, signifying that the PEI-induced permeabilization of mitochondrial membranes could be attributed to PEI's interaction with the phospholipid phase. Furthermore, this PEI-induced permeabilization of the lipid vesicles was observed only in the case of lipid vesicles including negatively charged phospholipids. These results indicate that PEIs interacted with negatively charged phospholipids in the mitochondrial membranes to directly lead to their permeabilization.
Collapse
Affiliation(s)
- Takenori Yamamoto
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan.
| | - Moe Tsunoda
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan
| | - Mizune Ozono
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan
| | - Akira Watanabe
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan
| | - Kazumasa Kotake
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan
| | - Yuka Hiroshima
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan
| | - Akiko Yamada
- School of Dentistry, Tokushima University, Kuramotocho-3, Tokushima 770-8504, Japan
| | - Hiroshi Terada
- Niigata University of Pharmacy and Applied Life Sciences, Niigata City 956-8603, Japan
| | - Yasuo Shinohara
- Institute for Genome Research, Tokushima University, Kuramotocho-3, Tokushima 770-8503, Japan; Faculty of Pharmaceutical Sciences, Tokushima University, Shomachi-1, Tokushima 770-8505, Japan
| |
Collapse
|
29
|
Alameh M, Lavertu M, Tran-Khanh N, Chang CY, Lesage F, Bail M, Darras V, Chevrier A, Buschmann MD. siRNA Delivery with Chitosan: Influence of Chitosan Molecular Weight, Degree of Deacetylation, and Amine to Phosphate Ratio on in Vitro Silencing Efficiency, Hemocompatibility, Biodistribution, and in Vivo Efficacy. Biomacromolecules 2017; 19:112-131. [PMID: 29211954 DOI: 10.1021/acs.biomac.7b01297] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chitosan (CS) shows in vitro and in vivo efficacy for siRNA delivery but with contradictory findings for incompletely characterized systems. For understanding which parameters produce effective delivery, a library of precisely characterized chitosans was produced at different degrees of deacetylation (DDAs) and average molecular weights (Mn). Encapsulation and transfection efficiencies were characterized in vitro. Formulations were selected to examine the influence of Mn and N:P ratio on nanoparticle uptake, metabolic activity, genotoxicity, and in vitro transfection. Hemocompatibility and in vivo biodistribution were then investigated for different Mn, N:P ratios, and doses. Nanoparticle uptake and gene silencing correlated with increased surface charge, which was obtained at high DDA and high Mn. A minimum polymer length of ∼60-70 monomers (∼10 kDa) was required for stability and knockdown. In vitro knockdown was equivalent to lipid control with no metabolic or genotoxicity. An inhibitory effect of serum on biological performance was dependent on DDA, Mn, and N:P. In vivo biodistribution in mice show accumulation of nanoparticles in kidney with 40-50% functional knockdown.
Collapse
Affiliation(s)
- Mohamad Alameh
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Marc Lavertu
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Nicolas Tran-Khanh
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Chi-Yuan Chang
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Frederic Lesage
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Martine Bail
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Vincent Darras
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Anik Chevrier
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| | - Michael D Buschmann
- Department of Chemical Engineering, ‡Institute of Biomedical Engineering, and §Department of Electrical Engineering, Polytechnique Montreal , Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
30
|
Hu Q, Zhao F, Guo F, Wang C, Fu Z. Polymeric Nanoparticles Induce NLRP3 Inflammasome Activation and Promote Breast Cancer Metastasis. Macromol Biosci 2017; 17. [PMID: 29131546 DOI: 10.1002/mabi.201700273] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/20/2017] [Indexed: 12/15/2022]
Abstract
Polymeric nanoparticles gain enormous interests in cancer therapy. Polyethylenimine (PEI) 25 kD is well known for its high transfection efficiency and cytotoxicity. PEI-CyD (PC) was previously synthesized by conjugating low molecular PEI (M w 600) with β-cyclodextrin (β-CyD), which is shown to induce lower cytotoxicity than PEI 25 kD. In the current study, the in vivo immune response of branched PEI 25 kD and PC is investigated. Compared to PC/pDNA, exposure of PEI 25kD/pDNA induces higher level of immune-stimulation evidenced by the increased spleen weight, phagocytic capacity of peritoneal macrophage, and proinflammatory cytokines in serum and liver. Importantly, administration of PEI 25 kD can greatly promote breast cancer metastasis in liver and lung tissues, which correlates with its ability to induce high oxidative stress and NLRP3-inflammasome activation. These results suggest that polymeric nanocarriers have the potential to induce immune-stimulation and cancer metastasis, which may affect their efficiency for cancer therapy.
Collapse
Affiliation(s)
- Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, 310032, Hangzhou, China
| | - Fenghui Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, 310032, Hangzhou, China
| | - Fengliang Guo
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, 310032, Hangzhou, China
| | - Chengcheng Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, 310032, Hangzhou, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, 310032, Hangzhou, China
| |
Collapse
|
31
|
Bengalli R, Gualtieri M, Capasso L, Urani C, Camatini M. Impact of zinc oxide nanoparticles on an in vitro model of the human air-blood barrier. Toxicol Lett 2017; 279:22-32. [DOI: 10.1016/j.toxlet.2017.07.877] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Accepted: 07/09/2017] [Indexed: 01/10/2023]
|
32
|
Marasini N, Haque S, Kaminskas LM. Polymer-drug conjugates as inhalable drug delivery systems: A review. Curr Opin Colloid Interface Sci 2017. [DOI: 10.1016/j.cocis.2017.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Strojan K, Lojk J, Bregar VB, Veranič P, Pavlin M. Glutathione reduces cytotoxicity of polyethyleneimine coated magnetic nanoparticles in CHO cells. Toxicol In Vitro 2017; 41:12-20. [DOI: 10.1016/j.tiv.2017.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/27/2023]
|
34
|
Almeida PV, Shahbazi MA, Correia A, Mäkilä E, Kemell M, Salonen J, Hirvonen J, Santos HA. A multifunctional nanocomplex for enhanced cell uptake, endosomal escape and improved cancer therapeutic effect. Nanomedicine (Lond) 2017; 12:1401-1420. [PMID: 28524813 DOI: 10.2217/nnm-2017-0034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate the chemotherapeutic potential of a novel multifunctional nanocomposite encapsulating both porous silicon (PSi) and gold (Au) nanoparticles in a polymeric nanocomplex. MATERIALS & METHODS The nanocomposite was physicochemically characterized and evaluated in vitro for biocompatibility, cellular internalization, endosomolytic properties, cytoplasmatic drug delivery and chemotherapeutic efficacy. RESULTS The nanocomposites were successfully produced and exhibited adequate physicochemical properties and superior in vitro cyto- and hemocompatibilities. The encapsulation of PSi nanoparticles in the nanocomplexes significantly enhanced their cellular internalization and enabled their endosomal escape, resulting in the efficient cytoplasmic delivery of these nanosystems. Sorafenib-loaded nanocomposites showed a potent in vitro antiproliferative effect on MDA-MB-231 breast cancer cells. CONCLUSION The multifunctional nanocomposite herein presented exhibits great potential as a chemotherapeutic nanoplatform.
Collapse
Affiliation(s)
- Patrick V Almeida
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland
| | - Mohammad-Ali Shahbazi
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland.,Department of Micro- & Nanotechnology, Technical University of Denmark, 2800 KGs. Lyngby, Denmark
| | - Alexandra Correia
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland
| | - Ermei Mäkilä
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland.,Laboratory of Industrial Physics, Department of Physics & Astronomy, University of Turku, FI-20014 Finland
| | - Marianna Kemell
- Laboratory of Inorganic Chemistry, Department of Chemistry, University of Helsinki, A.I. Virtasen aukio 1 (P.O. Box 55), FI-00014 Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics & Astronomy, University of Turku, FI-20014 Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry & Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E (P.O. Box 56), FI-00014 Finland.,Helsinki Institute of Life Science, HiLIFE, University of Helsinki, FI-00014Helsinki, Finland
| |
Collapse
|
35
|
Wang Y, Qiu Y, Yin S, Zhang L, Shi K, Gao H, Zhang Z, He Q. A functional nanocarrier that copenetrates extracellular matrix and multiple layers of tumor cells for sequential and deep tumor autophagy inhibitor and chemotherapeutic delivery. Autophagy 2017; 13:359-370. [PMID: 27911136 PMCID: PMC5324845 DOI: 10.1080/15548627.2016.1256523] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/19/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
To further enhance the intensity of deep tumor drug delivery and integrate a combined therapy, we herein report on a core-shell nanocarrier that could simultaneously overcome the double barriers of the extracellular matrix (ECM) and multiple layers of tumor cells (MLTC). A pH-triggered reversible swelling-shrinking core and an MMP2 (matrix metallopeptidase 2) degradable shell were developed to encapsulate chemotherapeutics and macroautophagy/autophagy inhibitors, respectively. MMP2 degraded the shell, which was followed by the autophagy inhibitors' release. The exposed core could diffuse along the pore within the ECM to deliver chemotherapeutics into deep tumors, and it was able to swell in lysosomes and shrink back in the cytoplasm or ECM. The swelling of the core resulted in the rapid release of chemotherapeutics to kill autophagy-inhibited cells. After leaving the dead cells, the shrinking core could act on neighboring cells that were closer to the center of the tumor. The core thus could also cross MLTC layer by layer to deliver chemotherapeutics into the deep tumor.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Qiu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Sheng Yin
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kairong Shi
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of education, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Leiro V, Garcia JP, Moreno PMD, Spencer AP, Fernandez-Villamarin M, Riguera R, Fernandez-Megia E, Paula Pêgo A. Biodegradable PEG–dendritic block copolymers: synthesis and biofunctionality assessment as vectors of siRNA. J Mater Chem B 2017; 5:4901-4917. [DOI: 10.1039/c7tb00279c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
New hybrid-biodegradable PEG–dendritic block copolymers as versatile delivery vectors for biomedical applications. Here, their biofunctionality as siRNA vectors is presented.
Collapse
Affiliation(s)
- Victoria Leiro
- i3S – Instituto de Investigação e Inovação em Saúde
- Universidade do Porto
- 4200-135 Porto
- Portugal
- INEB – Instituto de Engenharia Biomédica
| | - João Pedro Garcia
- i3S – Instituto de Investigação e Inovação em Saúde
- Universidade do Porto
- 4200-135 Porto
- Portugal
- INEB – Instituto de Engenharia Biomédica
| | - Pedro M. D. Moreno
- i3S – Instituto de Investigação e Inovação em Saúde
- Universidade do Porto
- 4200-135 Porto
- Portugal
- INEB – Instituto de Engenharia Biomédica
| | - Ana Patrícia Spencer
- i3S – Instituto de Investigação e Inovação em Saúde
- Universidade do Porto
- 4200-135 Porto
- Portugal
- INEB – Instituto de Engenharia Biomédica
| | - Marcos Fernandez-Villamarin
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| | - Ricardo Riguera
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| | - Ana Paula Pêgo
- i3S – Instituto de Investigação e Inovação em Saúde
- Universidade do Porto
- 4200-135 Porto
- Portugal
- INEB – Instituto de Engenharia Biomédica
| |
Collapse
|
37
|
Boyle WS, Senger K, Tolar J, Reineke TM. Heparin Enhances Transfection in Concert with a Trehalose-Based Polycation with Challenging Cell Types. Biomacromolecules 2016; 18:56-67. [DOI: 10.1021/acs.biomac.6b01297] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- William S. Boyle
- Department of Chemistry and ‡Stem Cell Institute and Division of Pediatric Blood
and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, United States
| | - Kyle Senger
- Department of Chemistry and ‡Stem Cell Institute and Division of Pediatric Blood
and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, United States
| | - Jakub Tolar
- Department of Chemistry and ‡Stem Cell Institute and Division of Pediatric Blood
and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, United States
| | - Theresa M. Reineke
- Department of Chemistry and ‡Stem Cell Institute and Division of Pediatric Blood
and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
38
|
Chiper M, Tounsi N, Kole R, Kichler A, Zuber G. Self-aggregating 1.8kDa polyethylenimines with dissolution switch at endosomal acidic pH are delivery carriers for plasmid DNA, mRNA, siRNA and exon-skipping oligonucleotides. J Control Release 2016; 246:60-70. [PMID: 27956144 DOI: 10.1016/j.jconrel.2016.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/23/2016] [Accepted: 12/05/2016] [Indexed: 11/24/2022]
Abstract
Efficiency of polyethylenimine (PEI) for nucleic acid delivery is affected by the size of the carrier and length of the nucleic acids. For instance, PEIs with molecular weights between 10-30kDa provide optimal DNA delivery activity whereas PEIs with molecular weights below 1.8kDa are ineffective. The activity of PEI is also severely diminished by substitution of DNA for shorter nucleic acids such as mRNA or siRNA. Here, through chemical modification of the primary amines to aromatic domains we achieved nucleic acid delivery by the 1.8kDa polyethylenimine (PEI) particles. This modification did not affect the PEI buffering abilities but enhanced its pH-sensitive aggregation, enabling stabilization of the polyplex outside the cell while still allowing nucleic acid release following cellular entry. The aromatic PEIs were then evaluated for their gene, mRNA, siRNA and 2'O-methyl phosphorothioate oligonucleotide in vitro transfection abilities. The salicylamide-grafted PEI showed to be a reliable carrier for delivering nucleic acids with cytoplasmic activity such as the mRNA and siRNA or nuclear diffusible oligonucleotide. It was then further equipped with polyethyleneglycol (PEG) and the delivery efficiency of the copolymer was tested in vivo for regeneration of dystrophin in the muscle of mdx mouse through a 2'O-methyl phosphorothioate-mediated splicing modulation. Intramuscular administration of polyplexes resulted in dystrophin-positive fibers in a mouse model of Duchenne muscular dystrophy without apparent toxicity. These findings indicate that precise modifications of low molecular weight PEI improve its bio-responsiveness and yield delivery vehicles for nucleic acids of various types in vitro and in vivo.
Collapse
Affiliation(s)
- Manuela Chiper
- Molecular and Pharmaceutical Engineering of Biologics, CNRS - Université de Strasbourg UMR 7242, Boulevard Sebastien Brant, 67412 Illkirch, France; Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France
| | - Nassera Tounsi
- Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France; Laboratory of Therapeutic Innovation UMR 7200, CNRS - Université de Strasbourg, France
| | - Ryszard Kole
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Antoine Kichler
- Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France; Laboratoire de Conception et d'Application de Molécules Bioactives, CNRS - Université de Strasbourg UMR 7199, France; Genethon, 91000 Evry, France
| | - Guy Zuber
- Molecular and Pharmaceutical Engineering of Biologics, CNRS - Université de Strasbourg UMR 7242, Boulevard Sebastien Brant, 67412 Illkirch, France.
| |
Collapse
|
39
|
Pandey AP, Sawant KK. Polyethylenimine: A versatile, multifunctional non-viral vector for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 68:904-918. [DOI: 10.1016/j.msec.2016.07.066] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/16/2016] [Accepted: 07/24/2016] [Indexed: 12/21/2022]
|
40
|
Vosen S, Rieck S, Heidsieck A, Mykhaylyk O, Zimmermann K, Plank C, Gleich B, Pfeifer A, Fleischmann BK, Wenzel D. Improvement of vascular function by magnetic nanoparticle-assisted circumferential gene transfer into the native endothelium. J Control Release 2016; 241:164-173. [PMID: 27667178 DOI: 10.1016/j.jconrel.2016.09.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022]
Abstract
Gene therapy is a promising approach for chronic disorders that require continuous treatment such as cardiovascular disease. Overexpression of vasoprotective genes has generated encouraging results in animal models, but not in clinical trials. One major problem in humans is the delivery of sufficient amounts of genetic vectors to the endothelium which is impeded by blood flow, whereas prolonged stop-flow conditions impose the risk of ischemia. In the current study we have therefore developed a strategy for the efficient circumferential lentiviral gene transfer in the native endothelium under constant flow conditions. For that purpose we perfused vessels that were exposed to specially designed magnetic fields with complexes of lentivirus and magnetic nanoparticles thereby enabling overexpression of therapeutic genes such as endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF). This treatment enhanced NO and VEGF production in the transduced endothelium and resulted in a reduction of vascular tone and increased angiogenesis. Thus, the combination of MNPs with magnetic fields is an innovative strategy for site-specific and efficient vascular gene therapy.
Collapse
Affiliation(s)
- Sarah Vosen
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | - Sarah Rieck
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | | | - Olga Mykhaylyk
- Institute of Experimental Oncology and Therapy Research, TU München, Germany
| | - Katrin Zimmermann
- Institute of Pharmacology and Toxicology, University Clinic of Bonn, Germany
| | - Christian Plank
- Institute of Experimental Oncology and Therapy Research, TU München, Germany
| | - Bernhard Gleich
- Zentralinstitut für Medizintechnik (IMETUM), TU München, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Clinic of Bonn, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | - Daniela Wenzel
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany.
| |
Collapse
|
41
|
Wang LH, Wu T, Wu DC, You YZ. Bioreducible Gene Delivery Vector Capable of Self-Scavenging the Intracellular-Generated ROS Exhibiting High Gene Transfection. ACS APPLIED MATERIALS & INTERFACES 2016; 8:19238-19244. [PMID: 27420138 DOI: 10.1021/acsami.6b04327] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cationic polymer vectors have received increasing attention for gene delivery in biotechnology over the past 2 decades, but few polymer vectors were used in clinical applications due to their low gene transfection efficacy. One of the major reasons is that the conventional cationic polymers can induce the increasing of intracellular reactive oxygen species (ROS) concentration and oxidative stress, which reduces the gene transfection efficacy. Here, we create a novel class of thioether dendron-branched polymer conjugate and self-assemble this conjugate into bioreducible cationic nanomicelles with disulfide bond connecting the thioether core to the cationic shell. The obtained nanomicelles have a unique ROS self-scavenging ability, thereby dramatically improving gene transfection efficacy.
Collapse
Affiliation(s)
- Long-Hai Wang
- Key Laboratory of Soft Matter Chemistry, Chinese Academy of Science, Department of Polymer Science and Engineering, University of Science and Technology of China , Hefei, Anhui 230026, China
| | - Ting Wu
- Key Laboratory of Soft Matter Chemistry, Chinese Academy of Science, Department of Polymer Science and Engineering, University of Science and Technology of China , Hefei, Anhui 230026, China
| | - De-Cheng Wu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
| | - Ye-Zi You
- Key Laboratory of Soft Matter Chemistry, Chinese Academy of Science, Department of Polymer Science and Engineering, University of Science and Technology of China , Hefei, Anhui 230026, China
| |
Collapse
|
42
|
Du J, Zhu W, Yang L, Wu C, Lin B, Wu J, Jin R, Shen T, Ai H. Reduction of polyethylenimine-coated iron oxide nanoparticles induced autophagy and cytotoxicity by lactosylation. Regen Biomater 2016; 3:223-9. [PMID: 27482464 PMCID: PMC4966295 DOI: 10.1093/rb/rbw023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide (SPIO) nanoparticles are excellent magnetic resonance contrast agents and surface engineering can expand their applications. When covered with amphiphilic alkyl-polyethyleneimine (PEI), the modified SPIO nanoparticles can be used as MRI visible gene/drug delivery carriers and cell tracking probes. However, the positively charged amines of PEI can also cause cytotoxicity and restricts their further applications. In this study, we used lactose to modify amphiphilic low molecular weight polyethylenimine (C12-PEI2K) at different lactosylation degree. It was found that the N-alkyl-PEI-lactobionic acid wrapped SPIO nanocomposites show better cell viability without compromising their labelling efficacy as well as MR imaging capability in RAW 264.7 cells, comparing to the unsubstituted ones. Besides, we found the PEI induced cell autophagy can be reduced via lactose modification, indicating the increased cell viability might rely on down-regulating autophagy. Thus, our findings provide a new approach to overcome the toxicity of PEI wrapped SPIO nanocomposites by lactose modification.
Collapse
Affiliation(s)
- Jiuju Du
- National Engineering Research Center for Biomaterials
| | - Wencheng Zhu
- National Engineering Research Center for Biomaterials; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China and
| | - Li Yang
- National Engineering Research Center for Biomaterials
| | - Changqiang Wu
- National Engineering Research Center for Biomaterials; School of Medical Imaging, North Sichuan Medical College, Nanchong, 637000, P.R. China
| | - Bingbing Lin
- National Engineering Research Center for Biomaterials
| | - Jun Wu
- National Engineering Research Center for Biomaterials
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials
| | - Taipeng Shen
- National Engineering Research Center for Biomaterials
| | - Hua Ai
- National Engineering Research Center for Biomaterials; Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610065, P.R. China
| |
Collapse
|
43
|
Quan S, Kumar P, Narain R. Cationic Galactose-Conjugated Copolymers for Epidermal Growth Factor (EGFR) Knockdown in Cervical Adenocarcinoma. ACS Biomater Sci Eng 2016; 2:853-859. [DOI: 10.1021/acsbiomaterials.6b00085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Stephen Quan
- Donadeo Innovation Centre for Engineering, Department of Chemical and Materials Engineering, and ‡Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Piyush Kumar
- Donadeo Innovation Centre for Engineering, Department of Chemical and Materials Engineering, and ‡Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ravin Narain
- Donadeo Innovation Centre for Engineering, Department of Chemical and Materials Engineering, and ‡Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
44
|
Zhao L, Zhang K, Bu W, Xu X, Jin H, Chang B, Wang B, Sun Y, Yang B, Zheng C, Sun H. Effective delivery of bone morphogenetic protein 2 gene using chitosan–polyethylenimine nanoparticle to promote bone formation. RSC Adv 2016. [DOI: 10.1039/c5ra24891d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Treating bone defects is still a challenge in clinical practice.
Collapse
|
45
|
Ma H, Liu J, Ali MM, Mahmood MAI, Labanieh L, Lu M, Iqbal SM, Zhang Q, Zhao W, Wan Y. Nucleic acid aptamers in cancer research, diagnosis and therapy. Chem Soc Rev 2015; 44:1240-56. [PMID: 25561050 DOI: 10.1039/c4cs00357h] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aptamers are single-stranded DNA or RNA oligomers, identified from a random sequence pool, with the ability to form unique and versatile tertiary structures that bind to cognate molecules with superior specificity. Their small size, excellent chemical stability and low immunogenicity enable them to rival antibodies in cancer imaging and therapy applications. Their facile chemical synthesis, versatility in structural design and engineering, and the ability for site-specific modifications with functional moieties make aptamers excellent recognition motifs for cancer biomarker discovery and detection. Moreover, aptamers can be selected or engineered to regulate cancer protein functions, as well as to guide anti-cancer drug design or screening. This review summarizes their applications in cancer, including cancer biomarker discovery and detection, cancer imaging, cancer therapy, and anti-cancer drug discovery. Although relevant applications are relatively new, the significant progress achieved has demonstrated that aptamers can be promising players in cancer research.
Collapse
Affiliation(s)
- Haitao Ma
- The Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Acute and subacute pulmonary toxicity and mortality in mice after intratracheal instillation of ZnO nanoparticles in three laboratories. Food Chem Toxicol 2015; 85:84-95. [DOI: 10.1016/j.fct.2015.08.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/28/2015] [Accepted: 08/04/2015] [Indexed: 01/06/2023]
|
47
|
Li H, Lee T, Dziubla T, Pi F, Guo S, Xu J, Li C, Haque F, Liang XJ, Guo P. RNA as a stable polymer to build controllable and defined nanostructures for material and biomedical applications. NANO TODAY 2015; 10:631-655. [PMID: 26770259 PMCID: PMC4707685 DOI: 10.1016/j.nantod.2015.09.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The value of polymers is manifested in their vital use as building blocks in material and life sciences. Ribonucleic acid (RNA) is a polynucleic acid, but its polymeric nature in materials and technological applications is often overlooked due to an impression that RNA is seemingly unstable. Recent findings that certain modifications can make RNA resistant to RNase degradation while retaining its authentic folding property and biological function, and the discovery of ultra-thermostable RNA motifs have adequately addressed the concerns of RNA unstability. RNA can serve as a unique polymeric material to build varieties of nanostructures including nanoparticles, polygons, arrays, bundles, membrane, and microsponges that have potential applications in biomedical and material sciences. Since 2005, more than a thousand publications on RNA nanostructures have been published in diverse fields, indicating a remarkable increase of interest in the emerging field of RNA nanotechnology. In this review, we aim to: delineate the physical and chemical properties of polymers that can be applied to RNA; introduce the unique properties of RNA as a polymer; review the current methods for the construction of RNA nanostructures; describe its applications in material, biomedical and computer sciences; and, discuss the challenges and future prospects in this field.
Collapse
Affiliation(s)
- Hui Li
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Taek Lee
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Republic of Korea
| | - Thomas Dziubla
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Fengmei Pi
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Sijin Guo
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jing Xu
- Laboratory of Nanomedicine and Nanosafety, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Chan Li
- Laboratory of Nanomedicine and Nanosafety, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Farzin Haque
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Xing-Jie Liang
- Laboratory of Nanomedicine and Nanosafety, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Peixuan Guo
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
48
|
Lin CW, Jan MS, Kuo JHS. Exploring MicroRNA Expression Profiles Related to the mTOR Signaling Pathway in Mouse Embryonic Fibroblast Cells Treated with Polyethylenimine. Mol Pharm 2015; 12:2858-68. [PMID: 26158199 DOI: 10.1021/acs.molpharmaceut.5b00329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although the toxicology of poly(ethylenimine) (PEI) in gene expression levels has been previously investigated, little is known about the effects of PEI on the expression of microRNAs (miRNAs) that regulate gene expression at the post-transcriptional level. In this study, we explored miRNA expression profiles related to cell death mechanisms in mouse embryonic fibroblast (MEF) cells treated with PEI by applying microarray analysis. Based on the analysis of the mTOR signaling pathway, three upregulated miRNAs (mmu-miR-3090-5p, mmu-miR-346-3p, and mmu-miR-494-3p) were verified in MEF cells treated with PEI at 24 h using real-time quantitative reverse transcriptase-polymerase chain reaction. We further demonstrated that these three upregulated miRNAs resulted in the decrease of gene and protein expressions of the target gene growth factor Igf1 in MEF cells treated with PEI or transfected with three upregulated miRNA mimics. However, these three upregulated miRNAs are not all cell-specific. Finally, we demonstrated that the mTOR signaling pathway is inhibited by autophagy induction and that the cell viability decreases in MEF cells treated with PEI or transfected with these three miRNA mimics. Collectively, our data suggested that PEI may affect the regulation of miRNAs in target cells.
Collapse
Affiliation(s)
| | | | - Jung-Hua Steven Kuo
- §Department of Pharmacy, Chia Nan University of Pharmacy and Science, 60 Erh-Jen Road, Section 1, Jen-Te, Tainan 717, Taiwan
| |
Collapse
|
49
|
Taranejoo S, Liu J, Verma P, Hourigan K. A review of the developments of characteristics of PEI derivatives for gene delivery applications. J Appl Polym Sci 2015. [DOI: 10.1002/app.42096] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Shahrouz Taranejoo
- Department of Chemical Engineering; Faculty of Engineering; Monash University; Melbourne Victoria Australia 3800
- Laboratory for Biomedical Engineering/Fluids Laboratory for Aeronautical and Industrial Research; Department of Mechanical and Aerospace Engineering; Faculty of Engineering; Monash University; Melbourne Victoria Australia 3800
| | - Jun Liu
- Stem Cell and Genetic Engineering Group; Department of Materials Engineering; Faculty of Engineering; Monash University Melbourne; Victoria Australia 3800
| | - Paul Verma
- Stem Cell and Genetic Engineering Group; Department of Materials Engineering; Faculty of Engineering; Monash University Melbourne; Victoria Australia 3800
- Turretfield Research Center South Australian Research and Development Institute; Adelaide South Australia Australia 5350
| | - Kerry Hourigan
- Laboratory for Biomedical Engineering/Fluids Laboratory for Aeronautical and Industrial Research; Department of Mechanical and Aerospace Engineering; Faculty of Engineering; Monash University; Melbourne Victoria Australia 3800
| |
Collapse
|
50
|
Abstract
RNA interference (RNAi) technology is a promising approach for efficient silencing of a particular gene for cancer gene therapy. However, the main obstacle for the development of RNAi-based therapeutic approaches is the delivery of the RNAi effector molecules to target cells. One promising strategy to surmount this challenge is the application of nonpathogenic bacteria as a delivery vector to target cells. In this chapter, the design of invasive Escherichia coli is described. The strain carries a plasmid encoding short hairpin RNAs (shRNAs), a protein (invasin) necessary for endocytotic absorption of the bacteria by target cells, and listeriolysin O required for the lysis of endocytotic vesicles within the target cells.
Collapse
Affiliation(s)
- Omar Ahmed
- Charité Campus Mitte, Institute of Pathology, Charitéplatz 1, Berlin, 10117, Germany
| | | | | |
Collapse
|