1
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Wang J, Huang H, Jia M, Chen S, Wang F, He G, Wu C, Lou K, Zheng X, Zhang H, Qin C, Yuan Y, Zen K, Liang H. Autologous platelet delivery of siRNAs by autologous plasma protein self-assembled nanoparticles for the treatment of acute kidney injury. J Nanobiotechnology 2025; 23:256. [PMID: 40156015 PMCID: PMC11954310 DOI: 10.1186/s12951-025-03338-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Acute kidney injury (AKI) involves the activation of intrarenal hemostatic and inflammatory pathways. Platelets rapidly migrate to affected sites of AKI and release extracellular vesicles (EVs) laden with bioactive mediators that regulate inflammation and hemostasis. While small interfering RNA (siRNA) is a potent gene-silencing tool for biomedical applications, its therapeutic application in vivo remains challenging. We developed an innovative nucleic acid delivery platform by hybridizing synthetic transformation-related protein 53 (p53) siRNA with autologous plasma and incubating the complex with autologous platelets. These engineered platelets selectively delivered p53 siRNA to injured renal tubular cells via EV-mediated cargo release, resulting in targeted p53 suppression in renal cells and subsequent attenuation of AKI progression. This platelet-centric translational strategy demonstrates significant potential for advancing precision therapies in AKI by exploiting endogenous platelet trafficking to deliver therapeutics directly to injury sites.
Collapse
Affiliation(s)
- Jiafan Wang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Hai Huang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Meng Jia
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Si Chen
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengjuan Wang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Guiyang He
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Chong Wu
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Kaibin Lou
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Xuexue Zheng
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Heng Zhang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yanggang Yuan
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ke Zen
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, China.
| | - Hongwei Liang
- Department of Emergency, School of Life Science and Technology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
3
|
Fei S, Ma Y, Zhou B, Chen X, Zhang Y, Yue K, Li Q, Gui Y, Xiang T, Liu J, Yang B, Wang L, Huang X. Platelet membrane biomimetic nanoparticle-targeted delivery of TGF-β1 siRNA attenuates renal inflammation and fibrosis. Int J Pharm 2024; 659:124261. [PMID: 38782155 DOI: 10.1016/j.ijpharm.2024.124261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
The progression of renal fibrosis to end-stage renal disease (ESRD) is significantly influenced by transforming growth factor-beta (TGF-beta) signal pathway. This study aimed to develop nanoparticles (PMVs@PLGA complexes) with platelet membrane camouflage, which can transport interfering RNA to target and regulate the TGF-β1 pathway in damaged renal tissues. The aim is to reduce the severity of acute kidney injury and to reduce fibrosis in chronic kidney disease. Hence, we formulated PMVs@TGF-β1-siRNA NP complexes and employed them for both in vitro and in vivo therapy. From the experimental findings we know that the PMVs@siRNA NPs could effectively target the kidneys in unilateral ureteral obstruction (UUO) mice and ischemia/reperfusion injury (I/R) mice. In animal models of treatment, PMVs@siRNA NP complexes effectively decreased the expression of TGF-β1 and mitigated inflammation and fibrosis in the kidneys by blocking the TGF-β1/Smad3 pathway. Therefore, these PMVs@siRNA NP complexes can serve as a promising biological delivery system for treating kidney diseases.
Collapse
Affiliation(s)
- Shengnan Fei
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Yidan Ma
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Bing Zhou
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Xu Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China
| | - Yuan Zhang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China
| | - Kun Yue
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Qingxin Li
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Yuanyuan Gui
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Tianya Xiang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Jianhang Liu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China; Medical School of Nantong University, Nantong 226001, PR China
| | - Bin Yang
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester LE1 9HN, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Lei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China; Nantong Egens Biotechnology Co., Ltd, Nantong 226001, Jiangsu, PR China.
| | - Xinzhong Huang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong 226001, PR China.
| |
Collapse
|
4
|
Shin DH, Jiang H, Gillard AG, Kim D, Fan X, Singh SK, Nguyen TT, Sohoni SS, Lopez-Rivas AR, Parthasarathy A, Ene CI, Gumin J, Lang FF, Alonso MM, Gomez-Manzano C, Fueyo J. Chimeric oncolytic adenovirus evades neutralizing antibodies from human patients and exhibits enhanced anti-glioma efficacy in immunized mice. Mol Ther 2024; 32:722-733. [PMID: 38311852 PMCID: PMC10928285 DOI: 10.1016/j.ymthe.2024.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/23/2023] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Oncolytic viruses are a promising treatment for patients with high-grade gliomas, but neutralizing antibodies can limit their efficacy in patients with prior virus exposure or upon repeated virus injections. Data from a previous clinical trial using the oncolytic adenovirus Delta-24-RGD showed that generation of anti-viral neutralizing antibodies may affect the long-term survival of glioma patients. Past studies have examined the effects of neutralizing antibodies during systemic virus injections, but largely overlooked their impact during local virus injections into the brain. We found that immunoglobulins colocalized with viral proteins upon local oncolytic virotherapy of brain tumors, warranting a strategy to prevent virus neutralization and maximize oncolysis. Thus, we generated a chimeric virus, Delta-24-RGD-H43m, by replacing the capsid protein HVRs from the serotype 5-based Delta-24-RGD with those from the rare serotype 43. Delta-24-RGD-H43m evaded neutralizing anti-Ad5 antibodies and conferred a higher rate of long-term survival than Delta-24-RGD in glioma-bearing mice. Importantly, Delta-24-RGD-H43m activity was significantly more resistant to neutralizing antibodies present in sera of glioma patients treated with Delta-24-RGD during a phase 1 clinical trial. These findings provide a framework for a novel treatment of glioma patients that have developed immunity against Delta-24-RGD.
Collapse
Affiliation(s)
- Dong Ho Shin
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Jiang
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew G Gillard
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Debora Kim
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuejun Fan
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sanjay K Singh
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Teresa T Nguyen
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sagar S Sohoni
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andres R Lopez-Rivas
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Akhila Parthasarathy
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chibawanye I Ene
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joy Gumin
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Frederick F Lang
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marta M Alonso
- Department of Pediatrics, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Candelaria Gomez-Manzano
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Juan Fueyo
- MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
5
|
Kyaw T, Drummond G, Bobik A, Peter K. Myocarditis: causes, mechanisms, and evolving therapies. Expert Opin Ther Targets 2023; 27:225-238. [PMID: 36946552 DOI: 10.1080/14728222.2023.2193330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Myocarditis is a severe lymphocyte-mediated inflammatory disorder of the heart, mostly caused by viruses and immune checkpoint inhibitors (ICIs). Recently, myocarditis as a rare adverse event of mRNA vaccines for SARS-CoV-2 has caused global attention. The clinical consequences of myocarditis can be very severe, but specific treatment options are lacking or not yet clinically proven. AREAS COVERED This paper offers a brief overview of the biology of viruses that frequently cause myocarditis, focusing on mechanisms important for viral entry and replication following host infection. Current and new potential therapeutic targets/strategies especially for viral myocarditis are reviewed systematically. In particular, the immune system in myocarditis is dissected with respect to infective viral and non-infective, ICI-induced myocarditis. EXPERT OPINION Vaccination is an excellent emerging preventative strategy for viral myocarditis, but most vaccines still require further development. Anti-viral treatments that inhibit viral replication need to be considered following viral infection in host myocardium, as lower viral load reduces inflammation severity. Understanding how the immune system continues to damage the heart even after viral clearance will define novel therapeutic targets/strategies. We propose that viral myocarditis can be best treated using a combination of antiviral agents and immunotherapies that control cytotoxic T cell activity.
Collapse
Affiliation(s)
- Tin Kyaw
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
| | - Grant Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University Melbourne Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Alex Bobik
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
- Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Karlheinz Peter
- Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute
- Department of Cardiometabolic Health, University of Melbourne Melbourne Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University Melbourne Australia
- Heart Centre, Alfred Hospital, Melbourne, Australia
- Department of Immunology, Monash University Melbourne Australia
| |
Collapse
|
6
|
Wang T, Zhou T, Xu M, Wang S, Wu A, Zhang M, Zhou YL, Shi J. Platelet membrane-camouflaged nanoparticles carry microRNA inhibitor against myocardial ischaemia‒reperfusion injury. J Nanobiotechnology 2022; 20:434. [PMID: 36195952 PMCID: PMC9531416 DOI: 10.1186/s12951-022-01639-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/18/2022] [Indexed: 12/04/2022] Open
Abstract
The incidence of myocardial ischaemia‒reperfusion injury (MIRI) is increasing every year, and there is an urgent need to develop new therapeutic approaches. Nrf2 is thought to play a protective role during MIRI and it is regulated by microRNAs (miRNAs). This study focused on PLGA nanoparticles camouflaged by platelet membrane vesicles (PMVs) (i.e., PMVs@PLGA complexes) carrying microRNA inhibitors, which regulate Nrf2 and can play a therapeutic role in the MIRI process. In vitro and in vivo characterization showed that PMVs@PLGA has excellent transfection efficiency, low toxicity and good targeting. MicroRNAs that effectively regulate Nrf2 were identified, and then PMVs@PLGA-miRNA complexes were prepared and used for in vitro and in vivo treatment. PMVs@PLGA-miRNA complexes can effectively target the delivery of inhibitors to cardiomyocytes. Our results suggest that PMVs@PLGA complexes are a novel delivery system and a novel biological approach to the treatment of MIRI.
Collapse
Affiliation(s)
- Tianyi Wang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
| | - Tingting Zhou
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
| | - Mingming Xu
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
| | - Shuo Wang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
| | - Anqi Wu
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
| | - Mingyang Zhang
- Department of Forensic Sciences, Soochow University, NO.178, Ganjiang Road, Suzhou, 215000, Jiangsu, China.
| | - You Lang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Jiahai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China. .,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China. .,School of Public Health, Nantong University, NO.9, Seyuan Road, Nantong, 226019, Jiangsu, China.
| |
Collapse
|
7
|
Rojas JM, Sevilla N, Martín V. A New Look at Vaccine Strategies Against PPRV Focused on Adenoviral Candidates. Front Vet Sci 2021; 8:729879. [PMID: 34568477 PMCID: PMC8455998 DOI: 10.3389/fvets.2021.729879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is a virus that mainly infects goats and sheep causing significant economic loss in Africa and Asia, but also posing a serious threat to Europe, as recent outbreaks in Georgia (2016) and Bulgaria (2018) have been reported. In order to carry out the eradication of PPRV, an objective set for 2030 by the Office International des Epizooties (OIE) and the Food and Agriculture Organization of the United Nations (FAO), close collaboration between governments, pharmaceutical companies, farmers and researchers, among others, is needed. Today, more than ever, as seen in the response to the SARS-CoV2 pandemic that we are currently experiencing, these goals are feasible. We summarize in this review the current vaccination approaches against PPRV in the field, discussing their advantages and shortfalls, as well as the development and generation of new vaccination strategies, focusing on the potential use of adenovirus as vaccine platform against PPRV and more broadly against other ruminant pathogens.
Collapse
Affiliation(s)
| | | | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA-CSIC), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
8
|
Xing SG, Zhou YL, Yang QQ, Ju F, Zhang L, Tang JB. Effects of nanoparticle-mediated growth factor gene transfer to the injured microenvironment on the tendon-to-bone healing strength. Biomater Sci 2020; 8:6611-6624. [PMID: 33231577 DOI: 10.1039/d0bm01222j] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The tendon-to-bone healing after trauma is usually slow and weak, and the repair site is easily disrupted during early mobilization exercise. bFGF and VEGFA gene therapy may hold promise in augmenting the tendon-to-bone healing process through enhancing cell proliferation and angiogenesis. This study is conducted to determine the effects of nanoparticle-mediated co-delivery of bFGF and VEGFA genes to the tendon-to-bone repair interface on the healing strength and biological responses in a chicken model. The PLGA nanoparticle/pEGFP-bFGF + pEGFP-VEGFA plasmid complexes were prepared and were characterized in vitro and in vivo. The nanoparticle/plasmid complexes can effectively transfer bFGF and VEGFA genes to the tendon-to-bone interface. Nanoparticle-mediated co-delivery of bFGF and VEGFA genes significantly improved the tendon-to-bone healing in terms of healing strengths and histology in a chicken flexor tendon repair model. Our results suggest a new biological approach to accelerate the tendon-to-bone healing.
Collapse
Affiliation(s)
- Shu Guo Xing
- The Nanomedicine Research Laboratory, Research for Frontier Medicine and Hand Surgery Research Center, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China.
| | | | | | | | | | | |
Collapse
|
9
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
10
|
Mohd-Lila MA, Yee LK, Cen LS, Bala JA, Balakrishnan KN, Allaudin ZN, Abdul Rahman SO, Hani H, Abdullah R. The application of naked DNA plasmid (DrZP3) and recombinant adenovirus (Ad-rZP3) in rat animal model to determine comparative efficacy of ZP3-Immunocontraceptive vaccines. Microb Pathog 2019; 134:103572. [PMID: 31163251 DOI: 10.1016/j.micpath.2019.103572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023]
Abstract
The common physical and chemical methods for controlling rat pest are less than satisfactory and inhumane. Immunocontraception approach has been considered more humane and it can be accomplished by inducing the relevant host immune response that block further development of reproductive gametes. ZP3 proteins are known to play very important role during sperm-ovum fertilization. It is a self-antigen and only localized in female ovaries. Therefore, an immunization with ZP3 protein elsewhere will induce a generalize host immune response against ZP3 protein. This study employed rat ZP3 (rZP3) gene prepared from its cDNA of Rattus rattus diardii. It was delivered and expressed in vivo by naked plamid DNA (DrZP3) or recombinant ZP3-Adenovirus (Ad-rZP3). Expression studies in vitro with DrZP3 or Ad-ZP3 showed rZP3 proteins were successfully expressed in Vero cells. Hyperimmune serum against rZP3 that were prepared by immunizing several rats with purified rZP3-pichia yeast fusion protein showed it blocked sperms from binding DrZP3-transfected Vero cells. Female Sprague Dawley rats immunized with DrZP3 demonstrated a long-term effect for significant reduction of fertility up to 92.6%. Ovaries from rats immunized with DrZP3 were severely atrophied with disappearance of primordial follicles from ovarian cortex with an increased in the amount of oocyte-free cell clusters. Female rats immunized with Ad-rZP3 demonstrated 27% reduction of fertility. The infertility induced by Ad-rZP3 is comparatively low and ineffective. This could be due to a strong host immune response that suppresses the recombinant virus itself resulted in minimum rZP3 protein presentation to the host immune system. As a result, low antibody titers produced against rZP3 is insufficient to block oocytes from maturity and fertilization. Therefore, immunization with DrZP3 for immunocontraception is more effective than Ad-rZP3 recombinant adenovirus. It is proposed to explore further on the use of adenovirus or other alternative viruses to deliver ZP3 protein and for the development of enhanced expression of rZP3 in target host.
Collapse
Affiliation(s)
- Mohd-Azmi Mohd-Lila
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia.
| | - Lai Kit Yee
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Lo Sewn Cen
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Jamilu Abubakar Bala
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Microbiology Unit, Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University Kano, Nigeria, P.M.B. 3011, Kano, Nigeria
| | | | - Zeenathul Nazariah Allaudin
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Sheikh-Omar Abdul Rahman
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Homayoun Hani
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| | - Rasedee Abdullah
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
11
|
Heiniö C, Sorsa S, Siurala M, Grönberg-Vähä-Koskela S, Havunen R, Haavisto E, Koski A, Hemminki O, Zafar S, Cervera-Carrascon V, Munaro E, Kanerva A, Hemminki A. Effect of Genetic Modifications on Physical and Functional Titers of Adenoviral Cancer Gene Therapy Constructs. Hum Gene Ther 2019; 30:740-752. [PMID: 30672366 DOI: 10.1089/hum.2018.240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After the discovery and characterization of the adenovirus in the 1950s, this prevalent cause of the common cold and other usually mild diseases has been modified and utilized in biomedicine in several ways. To date, adenoviruses are the most frequently used vectors and therapeutic (e.g., oncolytic) agents with a number of beneficial features. They infect both dividing and nondividing cells, enable high-level, transient protein expression, and are easy to amplify to high concentrations. As an important and versatile research tool, it is of essence to understand the limits and advantages that genetic modification of adenovirus vectors may entail. Therefore, a retrospective analysis was performed of adenoviral gene therapy constructs produced in the same laboratory with similar methods. The aim was to assess the impact of various modifications on the physical and functional titer of the virus. It was found that genome size (designed within "the 105% golden rule") did not significantly affect the physical titer of the adenovirus preparations, regardless of the type of transgene (e.g., immunostimulatory vs. other), number of engineered changes, and size of the mutated virus genome. One statistically significant exception was noted, however. Chimeric adenoviruses (5/3) had a slightly lower physical titer compared to Ad5-based viruses, although a trend for the opposite was true for functional titers. Thus, 5/3 chimeric viruses may in fact be appealing from a safety versus efficacy viewpoint. Armed viruses had lower functional and physical titers than unarmed viruses, while five genomic modifications started to decrease functional titer. Importantly, even highly modified armed viruses generally had good titers compatible with clinical testing. In summary, this paper shows the plasticity of adenovirus for various vector, oncolytic, and armed oncolytic uses. These results inform future generations of adenovirus-based drugs for human use. This information is directly transferable to academic laboratories and the biomedical industry involved in vector design and production optimization.
Collapse
Affiliation(s)
- Camilla Heiniö
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,2 TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Mikko Siurala
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,2 TILT Biotherapeutics Ltd., Helsinki, Finland
| | | | - Riikka Havunen
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,2 TILT Biotherapeutics Ltd., Helsinki, Finland
| | | | - Anniina Koski
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Otto Hemminki
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,3 Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Sadia Zafar
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Víctor Cervera-Carrascon
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,2 TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Eleonora Munaro
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Anna Kanerva
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,4 Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Akseli Hemminki
- 1 Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,2 TILT Biotherapeutics Ltd., Helsinki, Finland.,5 Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
12
|
Xia W, Wu Z, Guo C, Zhu S, Zhang X, Xia X, Sun H. Recombinant adenovirus-delivered soluble CD163 and sialoadhesin receptors protected pigs from porcine reproductive and respiratory syndrome virus infection. Vet Microbiol 2018; 219:1-7. [PMID: 29778179 DOI: 10.1016/j.vetmic.2018.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 11/26/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most important swine diseases affecting pig industry worldwide. Sialoadehesin (Sn) and CD163 are the two specific receptors for PRRSV infection of porcine alveolar macrophages. Our previous study showed that the soluble Sn receptor Sn4D-Fc and soluble CD163 receptor SRCR59-Fc expressed by the two recombinant adenoviral (rAd) vectors have an additive anti-PRRSV effect in vitro. In the present study, rAd-Sn4D-Fc and rAd-SRCR59-Fc were inoculated into pigs, and the efficient expression of Sn4D-Fc and SRCR59-Fc proteins was detected by ELISA. Then, PRRSV-naïve pigs were inoculated with rAd-Sn4D-Fc and/or rAd-SRCR59-Fc before contagious infection with different PRRSV strains. Among the three rAd inoculation groups, simultaneous inoculation with the two rAd vectors provided the best protection against highly pathogenic JXA1 strain PRRSV, followed by rAd-SRCR59-Fc inoculation and rAd-Sn4D-Fc inoculation. Clinical observation and quantitative RT-PCR analyses showed that all of the double rAd-inoculated pigs (n = 9) survived from the contagious infection with highly pathogenic JXA1, JS07 or SH1705 strain PRRSV with significantly alleviated clinical scores, viremia, fecal viral emission and tissue virus loads. These data suggest that rAd-Sn4D-Fc and rAd-SRCR59-Fc can be developed further as the universal therapeutic vaccine to facilitate PRRSV eradication.
Collapse
Affiliation(s)
- Wenlong Xia
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhi Wu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Changming Guo
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Xinyu Zhang
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiaoli Xia
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Huaichang Sun
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
13
|
Yang QQ, Shao YX, Zhang LZ, Zhou YL. Therapeutic strategies for flexor tendon healing by nanoparticle-mediated co-delivery of bFGF and VEGFA genes. Colloids Surf B Biointerfaces 2018; 164:165-176. [DOI: 10.1016/j.colsurfb.2018.01.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 01/10/2023]
|
14
|
Immunodominance of Adenovirus-Derived CD8 + T Cell Epitopes Interferes with the Induction of Transgene-Specific Immunity in Adenovirus-Based Immunization. J Virol 2017; 91:JVI.01184-17. [PMID: 28768877 DOI: 10.1128/jvi.01184-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Adenovirus (Ad)-based immunization is a popular approach in vaccine development, and Ad-based vectors are renowned for their potential to induce strong CD8+ T cell responses to the encoded transgene. Surprisingly, we previously found in the mouse Friend retrovirus (FV) model that Ad-based immunization did not induce CD8+ T cell responses to the FV Leader-Gag-derived immunodominant epitope GagL85-93 We show now that induction of GagL85-93-specific CD8+ T cells was highly effective when leader-Gag was delivered by plasmid DNA immunization, implying a role for Ad-derived epitopes in mediating unresponsiveness. By immunizing with DNA constructs encoding strings of GagL85-93 and the two Ad-derived epitopes DNA-binding protein418-426 (DBP418-426) and hexon486-494, we confirmed that Ad epitopes prevent induction of GagL85-93-specific CD8+ T cells. Interestingly, while DBP418-426 did not interfere with GagL85-93-specific CD8+ T cell induction, the H-2Dd-restricted hexon486-494 suppressed the CD8+ T cell response to the H-2Db-restricted GagL85-93 strongly in H-2b/d mice but not in H-2b/b mice. This finding indicates that competition occurs at the level of responding CD8+ T cells, and we could indeed demonstrate that coimmunization with an interleukin 2 (IL-2)-encoding plasmid restored GagL85-93-specific CD8+ T cell responses to epitope strings in the presence of hexon486-494 IL-2 codelivery did not restore GagL85-93 responsiveness in Ad-based immunization, however, likely due to the presence of further epitopes in the Ad vector. Our findings show that seemingly immunodominant transgene epitopes can be dominated by Ad-derived epitopes. These findings underline the importance of thorough characterization of vaccine vectors, and modifications of vectors or immunogens may be required to prevent impaired transgene-specific immune responses.IMPORTANCE Ad-based vectors are widely used in experimental preclinical and clinical immunization studies against numerous infectious agents, such as human immunodeficiency virus, Ebola virus, Plasmodium falciparum, or Mycobacterium tuberculosis Preexisting immunity to Ad-based vectors is widely recognized as a hindrance to the widespread use of Ad-based vectors for immunizations in humans; however, our data show that an immune response to Ad-derived T cell epitopes can also result in loss or impairment of transgene-specific immune responses in prenaive vaccinees due to immune competition. Our results highlight that seemingly immunodominant epitopes may be affected by dominance of vector-derived epitopes, and modifications of the vector design or the immunogens employed in immunization may lead to more effective vaccines.
Collapse
|
15
|
Fougeroux C, Holst PJ. Future Prospects for the Development of Cost-Effective Adenovirus Vaccines. Int J Mol Sci 2017; 18:ijms18040686. [PMID: 28420073 PMCID: PMC5412272 DOI: 10.3390/ijms18040686] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 01/13/2023] Open
Abstract
Vaccination is one of the most efficient tools for disease prevention, and a continuously growing field of research. However, despite progress, we still need more efficient and cost-effective vaccines that would improve access to those in need. In this review, we will describe the status of virus-vectored vaccine technology with a focus on adenoviral-based vaccines. Adenovirus (Ad) vaccines have proven to be efficient in military vaccinations against Ad4 and Ad7 and as highly efficient vectored vaccines against rabies. The question of how other adenovirus-based vaccines can become as efficient as the rabies vaccine is the underlying theme in this review. Here, we will first give an overview of the basic properties of vectored vaccines, followed by an introduction to the characteristics of adenoviral vectors and previously tested modifications of the vector backbone and expression cassettes, with a focus on how they can contribute to increased vaccine cost-effectiveness. Finally, we will highlight a few successful examples of research that have attempted to improve the use of adenoviral-based vaccines by improving the transgene immunogenicity.
Collapse
Affiliation(s)
- Cyrielle Fougeroux
- Department of Immunology and Microbiology, Copenhagen University, København K 1014, Denmark.
| | - Peter J Holst
- Department of Immunology and Microbiology, Copenhagen University, København K 1014, Denmark.
| |
Collapse
|
16
|
Shen CF, Wang SM, Ho TS, Liu CC. Clinical features of community acquired adenovirus pneumonia during the 2011 community outbreak in Southern Taiwan: role of host immune response. BMC Infect Dis 2017; 17:196. [PMID: 28270104 PMCID: PMC5341368 DOI: 10.1186/s12879-017-2272-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/17/2017] [Indexed: 11/11/2022] Open
Abstract
Background Human adenovirus 7 (HAdV-7) was responsible for a significant number of fatalities during the 2011 community outbreak in Taiwan. The mechanisms underlying the pathogenesis of severe adenovirus infections in non-immunocompromised individuals remain unclear. Adenovirus pneumonia was associated with pleural effusion in a number of patients from the 2011 outbreak suggesting that similar to bacterial pneumonia, patients diagnosed with adenovirus pneumonia who have pleural effusion are more severely and systemically infected, and may have a more protracted disease course. We hypothesized that the host immunological response determines the severity of adenoviral infection. Methods This retrospective case series study included patients diagnosed with severe lower respiratory tract infections at the National Cheng Kung University Hospital in southern Taiwan between December 2010 and October 2011. The main inclusion criteria were 1) presence of multifocal patchy infiltrates, lobar consolidation or reticular interstitial opacities in chest X-rays, and 2) presence of adenovirus isolated from respiratory specimens. All patients had adenovirus isolated from respiratory specimens, and were negative for other viruses. Pleural effusion was confirmed in all patients using chest echography. Clinical features and laboratory data were compared in patients with (n = 12) and without (n = 15) parapneumonic effusion. Results Presence of parapneumonic effusion was significantly associated with a longer febrile duration, more complicated clinical management, and a greater risk of extrapulmonary involvement, notably hepatitis. Patients without pleural effusion had significantly higher numbers of WBCs, platelets, and absolute segment cell counts (ASCs) compared to patients with pleural effusion (all p < 0.05). Patients without pleural effusion had significantly higher counts of CD4+, CD8+, and CD20+ T cells (all p < 0.05) compared to patients with pleural effusion. Conclusion Our data indicated that presence of parapneumonic effusion in adenoviral pneumonia was associated with longer febrile duration, more complicated clinical management, a greater risk of hepatitis, and suppression of host cellular immunity. Further prospective, large-scale studies are needed to validate our results.
Collapse
Affiliation(s)
- Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, North Dist., Tainan, 70403, Taiwan
| | - Shih-Min Wang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzong-Shiann Ho
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, North Dist., Tainan, 70403, Taiwan. .,Center of Infectious Disease and Signaling Research, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center for Infection Control, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
17
|
Shapir N, Miari R, Blum S, Schwartz D, Chernin G, Neil GA, Afik D, Panet A. Preclinical and Preliminary Clinical Evaluation of Genetically Transduced Dermal Tissue Implants for the Sustained Secretion of Erythropoietin and Interferon α. HUM GENE THER CL DEV 2016; 26:216-27. [PMID: 26684446 DOI: 10.1089/humc.2015.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Protein drugs are currently delivered by bolus injection and although treatment frequently is successful, these methods also have major drawbacks, which call for the development of alternative technologies allowing prolonged delivery of these drugs. We developed a new ex vivo gene therapy platform called Transduced Autologous Restorative Gene Therapy (TARGT) for sustained long term production and secretion of autologous therapeutic proteins. A biopsy of dermal tissue taken from the patient is transduced ex vivo with a viral vector encoding the required gene under a constitutive promoter. Following measurement of protein secretion ex vivo, the transduced dermal tissue is implanted back into the patient, where it secretes the therapeutic protein into the circulation for several months or longer. A major hurdle to this approach is potential immunogenicity of the transduced tissue following implantation. In this paper we describe the preclinical and early clinical development of this technology, which allowed for overcoming these hurdles. To that end, we have used the helper dependent (HD) adenoviral vector with newly designed expression cassette containing genetic elements to optimize transgene expression. Moreover, we have developed procedures for TARGT tissue implantation, with measures to improve engraftment and reduce inflammation and rejection. Implantation of human TARGT to severe combined immune deficient (SCID) mice indicated long-term production of active proteins in the blood. Preliminary results of a clinical trial from two anemic end-stage renal disease patients, implanted with TARGTs expressing the human erythropoietin (EPO) gene, demonstrated prolonged secretion with physiologic blood level of the hormone and hemoglobin maintenance in the desired range, for a period of at least 5 months without exogenous EPO administration. We believe that the TARGT technology has the potential to become a platform for the sustained delivery of therapeutic proteins in various clinical indications.
Collapse
Affiliation(s)
- Nir Shapir
- 1 Medgenics Medical Israel, Ltd., Misgav, Israel
| | - Reem Miari
- 1 Medgenics Medical Israel, Ltd., Misgav, Israel
| | - Shany Blum
- 1 Medgenics Medical Israel, Ltd., Misgav, Israel
| | - Doron Schwartz
- 2 Nephrology and Dialysis Department, The Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Gil Chernin
- 2 Nephrology and Dialysis Department, The Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Daniel Afik
- 1 Medgenics Medical Israel, Ltd., Misgav, Israel
| | - Amos Panet
- 4 Department of Biochemistry (IMRIC), The Hebrew University-Hadassah Medical School , Jerusalem, Israel
| |
Collapse
|
18
|
Matthews QL, Farrow AL, Rachakonda G, Gu L, Nde P, Krendelchtchikov A, Pratap S, Sakhare SS, Sabbaj S, Lima MF, Villalta F. Epitope Capsid-Incorporation: New Effective Approach for Vaccine Development for Chagas Disease. Pathog Immun 2016; 1:214-233. [PMID: 27709126 PMCID: PMC5046838 DOI: 10.20411/pai.v1i2.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Previously we reported that a hexon-modified adenovirus (Ad) vector containing the invasive neutralizing epitope of Trypanosoma cruzi (T. cruzi) trypomastigote gp83 (Ad5-gp83) provided immunoprotection against T. cruzi infection. The purpose of this work was to design an improved vaccine for T. cruzi using a novel epitope capsid incorporation strategy. Thus, we evaluated the immunoprotection raised by co-immunization with Ad5-gp83 and an Ad vector containing an epitope (ASP-M) of the T. cruzi amastigote surface protein 2. Methods: Protein IX (pIX)-modified Ad vector (Ad5-pIX-ASP-M) was generated, characterized, and validated. C3H/He mice were immunized with Ad5-pIX-ASP-M and Ad5-gp83 and the cell-mediated responses were evaluated by enzyme-linked immunospot (ELISPOT) assay and intracellular staining. Immunized mice were challenged with T. cruzi to evaluate the vaccine efficacy. Results: Our findings indicate that Ad5-pIX-ASP-M was viable. Specific CD8+ T-cell mediated responses prior to the challenge show an increase in IFNγ and TNFα production. A single immunization with Ad5-pIX-ASP-M provided protection from T. cruzi infection, but co-immunizations with Ad5-pIX-ASP-M and Ad5-gp83 provided a higher immunoprotection and increased survival rate of mice. Conclusions: Overall, these results suggest that the combination of gp83 and ASP-M specific epitopes onto the capsid-incorporated adenoviruses would provide superior protection against Chagas disease as compared with Ad5-gp83 alone.
Collapse
Affiliation(s)
- Qiana L Matthews
- Department of Biological Sciences, Alabama State University, Montgomery, AL; Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Anitra L Farrow
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Girish Rachakonda
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Linlin Gu
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Pius Nde
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | | | - Siddharth Pratap
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Shruti S Sakhare
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Steffanie Sabbaj
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Maria F Lima
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Fernando Villalta
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| |
Collapse
|
19
|
Wang D, Mou H, Li S, Li Y, Hough S, Tran K, Li J, Yin H, Anderson DG, Sontheimer EJ, Weng Z, Gao G, Xue W. Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses. Hum Gene Ther 2016; 26:432-42. [PMID: 26086867 DOI: 10.1089/hum.2015.087] [Citation(s) in RCA: 265] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
CRISPR/Cas9 derived from the bacterial adaptive immunity pathway is a powerful tool for genome editing, but the safety profiles of in vivo delivered Cas9 (including host immune responses to the bacterial Cas9 protein) have not been comprehensively investigated in model organisms. Nonalcoholic steatohepatitis (NASH) is a prevalent human liver disease characterized by excessive fat accumulation in the liver. In this study, we used adenovirus (Ad) vector to deliver a Streptococcus pyogenes-derived Cas9 system (SpCas9) targeting Pten, a gene involved in NASH and a negative regulator of the PI3K-AKT pathway, in mouse liver. We found that the Ad vector mediated efficient Pten gene editing even in the presence of typical Ad vector-associated immunotoxicity in the liver. Four months after vector infusion, mice receiving the Pten gene-editing Ad vector showed massive hepatomegaly and features of NASH, consistent with the phenotypes following Cre-loxP-induced Pten deficiency in mouse liver. We also detected induction of humoral immunity against SpCas9 and the potential presence of an SpCas9-specific cellular immune response. Our findings provide a strategy to model human liver diseases in mice and highlight the importance considering Cas9-specific immune responses in future translational studies involving in vivo delivery of CRISPR/Cas9.
Collapse
Affiliation(s)
- Dan Wang
- 1 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Haiwei Mou
- 2 RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shaoyong Li
- 1 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Yingxiang Li
- 3 Department of Bioinformatics, School of Life Science and Technology, Tongji University , Shanghai, P.R. China
| | - Soren Hough
- 2 RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Karen Tran
- 1 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Jia Li
- 1 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Hao Yin
- 4 David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Daniel G Anderson
- 4 David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts.,5 Department of Chemical Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts.,6 Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts.,7 Institute of Medical Engineering and Science, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Erik J Sontheimer
- 2 RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Zhiping Weng
- 8 Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Guangping Gao
- 1 Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Wen Xue
- 2 RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
20
|
A Novel Vaccine Approach for Chagas Disease Using Rare Adenovirus Serotype 48 Vectors. Viruses 2016; 8:78. [PMID: 26978385 PMCID: PMC4810268 DOI: 10.3390/v8030078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/26/2016] [Accepted: 03/03/2016] [Indexed: 12/28/2022] Open
Abstract
Due to the increasing amount of people afflicted worldwide with Chagas disease and an increasing prevalence in the United States, there is a greater need to develop a safe and effective vaccine for this neglected disease. Adenovirus serotype 5 (Ad5) is the most common adenovirus vector used for gene therapy and vaccine approaches, but its efficacy is limited by preexisting vector immunity in humans resulting from natural infections. Therefore, we have employed rare serotype adenovirus 48 (Ad48) as an alternative choice for adenovirus/Chagas vaccine therapy. In this study, we modified Ad5 and Ad48 vectors to contain T. cruzi’s amastigote surface protein 2 (ASP-2) in the adenoviral early gene. We also modified Ad5 and Ad48 vectors to utilize the “Antigen Capsid-Incorporation” strategy by adding T. cruzi epitopes to protein IX (pIX). Mice that were immunized with the modified vectors were able to elicit T. cruzi-specific humoral and cellular responses. This study indicates that Ad48-modified vectors function comparable to or even premium to Ad5-modified vectors. This study provides novel data demonstrating that Ad48 can be used as a potential adenovirus vaccine vector against Chagas disease.
Collapse
|
21
|
Solovyeva VV, Kiyasov AP, Rizvanov AA. Genetically Engineered Dental Stem Cells for Regenerative Medicine. DENTAL STEM CELLS 2016. [DOI: 10.1007/978-3-319-28947-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Shapir N, Miari R, Blum S, Schwartz D, Chernin G, Neil G, Afik D, Panet A. Preclinical and preliminary clinical evaluation of genetically transduced dermal tissue implants for the sustained secretion of erythropoietin and interferon α. HUM GENE THER CL DEV 2015. [DOI: 10.1089/hum.2015.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Teng Y, Bai M, Sun Y, Wang Q, Li F, Xing J, Du L, Gong T, Duan Y. Enhanced delivery of PEAL nanoparticles with ultrasound targeted microbubble destruction mediated siRNA transfection in human MCF-7/S and MCF-7/ADR cells in vitro. Int J Nanomedicine 2015; 10:5447-57. [PMID: 26346350 PMCID: PMC4556292 DOI: 10.2147/ijn.s81172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The gene knockdown activity of small interfering RNA (siRNA) has led to their use as potential therapeutics for a variety of diseases. However, successful gene therapy requires safe and efficient delivery systems. In this study, we choose mPEG-PLGA-PLL nanoparticles (PEAL NPs) with ultrasound targeted microbubble destruction (UTMD) to efficiently deliver siRNA into cells. An emulsification-solvent evaporation method was used to prepare siRNA-loaded PEAL NPs. The NPs possessed an average size of 132.6±10.3 nm (n=5), with a uniform spherical shape, and had an encapsulation efficiency (EE) of more than 98%. As demonstrated by MTT assay, neither PEAL NPs nor siRNA-loaded PEAL NPs showed cytotoxicity even at high concentrations. The results of cellular uptake showed, with the assistance of UTMD, the siRNA-loaded PEAL NPs can be effectively internalized and can subsequently release siRNA in cells. Taken together, PEAL NPs with UTMD may be highly promising for siRNA delivery, making it possible to fully exploit the potential of siRNA-based therapeutics.
Collapse
Affiliation(s)
- Yanwei Teng
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China ; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Min Bai
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Qi Wang
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China ; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fan Li
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jinfang Xing
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Lianfang Du
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
24
|
Moilanen AM, Rysä J, Kaikkonen L, Karvonen T, Mustonen E, Serpi R, Szabó Z, Tenhunen O, Bagyura Z, Näpänkangas J, Ohukainen P, Tavi P, Kerkelä R, Leósdóttir M, Wahlstrand B, Hedner T, Melander O, Ruskoaho H. WDR12, a Member of Nucleolar PeBoW-Complex, Is Up-Regulated in Failing Hearts and Causes Deterioration of Cardiac Function. PLoS One 2015; 10:e0124907. [PMID: 25915632 PMCID: PMC4411154 DOI: 10.1371/journal.pone.0124907] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 03/09/2015] [Indexed: 01/13/2023] Open
Abstract
Aims In a recent genome-wide association study, WD-repeat domain 12 (WDR12) was associated with early-onset myocardial infarction (MI). However, the function of WDR12 in the heart is unknown. Methods and Results We characterized cardiac expression of WDR12, used adenovirus-mediated WDR12 gene delivery to examine effects of WDR12 on left ventricular (LV) remodeling, and analyzed relationship between MI associated WDR12 allele and cardiac function in human subjects. LV WDR12 protein levels were increased in patients with dilated cardiomyopathy and rats post-infarction. In normal adult rat hearts, WDR12 gene delivery into the anterior wall of the LV decreased interventricular septum diastolic and systolic thickness and increased the diastolic and systolic diameters of the LV. Moreover, LV ejection fraction (9.1%, P<0.05) and fractional shortening (12.2%, P<0.05) were declined. The adverse effects of WDR12 gene delivery on cardiac function were associated with decreased cellular proliferation, activation of p38 mitogen–activated protein kinase (MAPK)/heat shock protein (HSP) 27 pathway, and increased protein levels of Block of proliferation 1 (BOP1), essential for ribosome biogenesis. Post-infarction WDR12 gene delivery decreased E/A ratio (32%, P<0.05) suggesting worsening of diastolic function. In human subjects, MI associated WDR12 allele was associated significantly with diastolic dysfunction and left atrial size. Conclusions WDR12 triggers distinct deterioration of cardiac function in adult rat heart and the MI associated WDR12 variant is associated with diastolic dysfunction in human subjects.
Collapse
Affiliation(s)
- Anne-Mari Moilanen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Department of Pathology, The Institute of Diagnostics, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jaana Rysä
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Teemu Karvonen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Erja Mustonen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Raisa Serpi
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Zoltán Szabó
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Olli Tenhunen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Zsolt Bagyura
- Heart Center, Semmelweis University, Budapest, Hungary
| | - Juha Näpänkangas
- Department of Pathology, The Institute of Diagnostics, University of Oulu, Oulu, Finland
| | - Pauli Ohukainen
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Risto Kerkelä
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Margrét Leósdóttir
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Björn Wahlstrand
- Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Hedner
- Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Heikki Ruskoaho
- The Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
25
|
Uhrig-Schmidt S, Geiger M, Luippold G, Birk G, Mennerich D, Neubauer H, Grimm D, Wolfrum C, Kreuz S. Gene delivery to adipose tissue using transcriptionally targeted rAAV8 vectors. PLoS One 2014; 9:e116288. [PMID: 25551639 PMCID: PMC4281237 DOI: 10.1371/journal.pone.0116288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/08/2014] [Indexed: 02/02/2023] Open
Abstract
In recent years, the increasing prevalence of obesity and obesity-related co-morbidities fostered intensive research in the field of adipose tissue biology. To further unravel molecular mechanisms of adipose tissue function, genetic tools enabling functional studies in vitro and in vivo are essential. While the use of transgenic animals is well established, attempts using viral and non-viral vectors to genetically modify adipocytes in vivo are rare. Therefore, we here characterized recombinant Adeno-associated virus (rAAV) vectors regarding their potency as gene transfer vehicles for adipose tissue. Our results demonstrate that a single dose of systemically applied rAAV8-CMV-eGFP can give rise to remarkable transgene expression in murine adipose tissues. Upon transcriptional targeting of the rAAV8 vector to adipocytes using a 2.2 kb fragment of the murine adiponectin (mAP2.2) promoter, eGFP expression was significantly decreased in off-target tissues while efficient transduction was maintained in subcutaneous and visceral fat depots. Moreover, rAAV8-mAP2.2-mediated expression of perilipin A – a lipid-droplet-associated protein – resulted in significant changes in metabolic parameters only three weeks post vector administration. Taken together, our findings indicate that rAAV vector technology is applicable as a flexible tool to genetically modify adipocytes for functional proof-of-concept studies and the assessment of putative therapeutic targets in vivo.
Collapse
Affiliation(s)
| | - Matthias Geiger
- Swiss Federal Institute of Technology, ETH Zurich, SLA C92, Institute of Food Nutrition and Health, Schwerzenbach, Switzerland
| | - Gerd Luippold
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gerald Birk
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Detlev Mennerich
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Heike Neubauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Dirk Grimm
- Centre for Infectious Diseases/Virology, Heidelberg University Hospital and Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, Heidelberg, Germany
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, ETH Zurich, SLA C92, Institute of Food Nutrition and Health, Schwerzenbach, Switzerland
| | - Sebastian Kreuz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- * E-mail:
| |
Collapse
|
26
|
Liao ZX, Peng SF, Chiu YL, Hsiao CW, Liu HY, Lim WH, Lu HM, Sung HW. Enhancement of efficiency of chitosan-based complexes for gene transfection with poly(γ-glutamic acid) by augmenting their cellular uptake and intracellular unpackage. J Control Release 2014; 193:304-15. [DOI: 10.1016/j.jconrel.2014.04.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/02/2014] [Accepted: 04/10/2014] [Indexed: 12/01/2022]
|
27
|
Ueyama K, Mori K, Shoji T, Omata H, Gehlbach PL, Brough DE, Wei LL, Yoneya S. Ocular localization and transduction by adenoviral vectors are serotype-dependent and can be modified by inclusion of RGD fiber modifications. PLoS One 2014; 9:e108071. [PMID: 25232844 PMCID: PMC4169476 DOI: 10.1371/journal.pone.0108071] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 08/24/2014] [Indexed: 11/18/2022] Open
Abstract
Purpose To evaluate localization and transgene expression from adenoviral vector of serotypes 5, 35, and 28, ± an RGD motif in the fiber following intravitreal or subretinal administration. Methods Ocular transduction by adenoviral vector serotypes ± RGD was studied in the eyes of mice receiving an intravitreous or subretinal injection. Each serotype expressed a CMV-GFP expression cassette and histological sections of eyes were examined. Transgene expression levels were examined using luciferase (Luc) regulated by the CMV promoter. Results GFP localization studies revealed that serotypes 5 and 28 given intravitreously transduced corneal endothelial, trabecular, and iris cells. Intravitreous delivery of the unmodified Ad35 serotype transduced only trabecular meshwork cells, but, the modification of the RGD motif into the fiber of the Ad35 viral vector base expanded transduction to corneal endothelial and iris cells. Incorporation of the RGD motif into the fiber knob with deletion of RGD from the penton base did not affect the transduction ability of the Ad5 vector base. Subretinal studies showed that RGD in the Ad5 knob shifted transduction from RPE cells to photoreceptor cells. Using a CMV-Luc expression cassette, intravitreous delivery of all the tested vectors, such as Ad5-, Ad35- and Ad28- resulted in an initial rapid induction of luciferase activity that thereafter declined. Subretinal administration of vectors showed a marked difference in transgene activity. Ad35-Luc gene expression peaked at 7 days and remained elevated for 6 months. Ad28-Luc expression was high after 1 day and remained sustained for one month. Conclusions Different adenoviral vector serotypes ± modifications transduce different cells within the eye. Transgene expression can be brief or extended and is serotype and delivery route dependent. Thus, adenoviral vectors provide a versatile platform for the delivery of therapeutic agents for ocular diseases.
Collapse
Affiliation(s)
- Kazuhiro Ueyama
- Department of Ophthalmology, Saitama Medical University, Moroyama, Iruma, Saitama, Japan
- * E-mail:
| | - Keisuke Mori
- Department of Ophthalmology, Saitama Medical University, Moroyama, Iruma, Saitama, Japan
| | - Takuhei Shoji
- Department of Ophthalmology, Saitama Medical University, Moroyama, Iruma, Saitama, Japan
| | - Hidekazu Omata
- Department of Ophthalmology, Saitama Medical University, Moroyama, Iruma, Saitama, Japan
| | - Peter L. Gehlbach
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | - Lisa L. Wei
- GenVec, Inc., Gaithersburg, Maryland, United States of America
- National Eye Institute, Bethesda, Maryland, United States of America
| | - Shin Yoneya
- Department of Ophthalmology, Saitama Medical University, Moroyama, Iruma, Saitama, Japan
| |
Collapse
|
28
|
Farrow AL, Rachakonda G, Gu L, Krendelchtchikova V, Nde PN, Pratap S, Lima MF, Villalta F, Matthews QL. Immunization with Hexon modified adenoviral vectors integrated with gp83 epitope provides protection against Trypanosoma cruzi infection. PLoS Negl Trop Dis 2014; 8:e3089. [PMID: 25144771 PMCID: PMC4140675 DOI: 10.1371/journal.pntd.0003089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/30/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi is the causative agent of Chagas disease. Chagas disease is an endemic infection that affects over 8 million people throughout Latin America and now has become a global challenge. The current pharmacological treatment of patients is unsuccessful in most cases, highly toxic, and no vaccines are available. The results of inadequate treatment could lead to heart failure resulting in death. Therefore, a vaccine that elicits neutralizing antibodies mediated by cell-mediated immune responses and protection against Chagas disease is necessary. METHODOLOGY/PRINCIPAL FINDINGS The "antigen capsid-incorporation" strategy is based upon the display of the T. cruzi epitope as an integral component of the adenovirus' capsid rather than an encoded transgene. This strategy is predicted to induce a robust humoral immune response to the presented antigen, similar to the response provoked by native Ad capsid proteins. The antigen chosen was T. cruzi gp83, a ligand that is used by T. cruzi to attach to host cells to initiate infection. The gp83 epitope, recognized by the neutralizing MAb 4A4, along with His6 were incorporated into the Ad serotype 5 (Ad5) vector to generate the vector Ad5-HVR1-gp83-18 (Ad5-gp83). This vector was evaluated by molecular and immunological analyses. Vectors were injected to elicit immune responses against gp83 in mouse models. Our findings indicate that mice immunized with the vector Ad5-gp83 and challenged with a lethal dose of T. cruzi trypomastigotes confer strong immunoprotection with significant reduction in parasitemia levels, increased survival rate and induction of neutralizing antibodies. CONCLUSIONS/SIGNIFICANCE This data demonstrates that immunization with adenovirus containing capsid-incorporated T. cruzi antigen elicits a significant anti-gp83-specific response in two different mouse models, and protection against T. cruzi infection by eliciting neutralizing antibodies mediated by cell-mediated immune responses, as evidenced by the production of several Ig isotypes. Taken together, these novel results show that the recombinant Ad5 presenting T. cruzi gp83 antigen is a useful candidate for the development of a vaccine against Chagas disease.
Collapse
Affiliation(s)
- Anitra L. Farrow
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Girish Rachakonda
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Linlin Gu
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Valentina Krendelchtchikova
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Pius N. Nde
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Qiana L. Matthews
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
29
|
Arora S, Swaminathan SK, Kirtane A, Srivastava SK, Bhardwaj A, Singh S, Panyam J, Singh AP. Synthesis, characterization, and evaluation of poly (D,L-lactide-co-glycolide)-based nanoformulation of miRNA-150: potential implications for pancreatic cancer therapy. Int J Nanomedicine 2014; 9:2933-42. [PMID: 24971005 PMCID: PMC4069140 DOI: 10.2147/ijn.s61949] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs are small (18–22 nucleotide long) noncoding RNAs that play important roles in biological processes through posttranscriptional regulation of gene expression. Their aberrant expression and functional significance are reported in several human malignancies, including pancreatic cancer. Recently, we identified miR-150 as a novel tumor suppressor microRNA in pancreatic cancer. Furthermore, expression of miR-150 was downregulated in the majority of tumor cases, suggesting that its restoration could serve as an effective approach for pancreatic cancer therapy. In the present study, we developed a nanoparticle-based miR-150 delivery system and tested its therapeutic efficacy in vitro. Using double emulsion solvent evaporation method, we developed a poly (D,L-lactide-co-glycolide) (PLGA)-based nanoformulation of miR-150 (miR-150-NF). Polyethyleneimine (a cationic polymer) was incorporated in PLGA matrix to increase the encapsulation of miR-150. Physical characterization of miR-150-NF demonstrated that these nanoparticles had high encapsulation efficiency (~78%) and exhibited sustained release profile. Treatment of pancreatic cancer cells with miR-150-NF led to efficient intracellular delivery of miR-150 mimics and caused significant downregulation of its target gene (MUC4) expression. Inhibition of MUC4 correlated with a concomitant decrease in the expression of its interacting partner, HER2, and repression of its downstream signaling. Furthermore, treatment of pancreatic cancer cells with miR-150-NF suppressed their growth, clonogenicity, motility, and invasion. Together, these findings suggest that PLGA-based nanoformulation could potentially serve as a safe and effective nanovector platform for miR-150 delivery to pancreatic tumor cells.
Collapse
Affiliation(s)
- Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | | | - Ameya Kirtane
- Department of Pharmaceutics, The University of Minnesota, Minneapolis, USA
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Jayanth Panyam
- Department of Pharmaceutics, The University of Minnesota, Minneapolis, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA ; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
30
|
EMMPRIN reduction via scFv-M6-1B9 intrabody affects α3β1-integrin and MCT1 functions and results in suppression of progressive phenotype in the colorectal cancer cell line Caco-2. Cancer Gene Ther 2014; 21:246-55. [DOI: 10.1038/cgt.2014.24] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/30/2014] [Accepted: 04/30/2014] [Indexed: 12/15/2022]
|
31
|
Rocca CJ, Ur SN, Harrison F, Cherqui S. rAAV9 combined with renal vein injection is optimal for kidney-targeted gene delivery: conclusion of a comparative study. Gene Ther 2014; 21:618-28. [PMID: 24784447 PMCID: PMC4047163 DOI: 10.1038/gt.2014.35] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/19/2022]
Abstract
Effective gene therapy strategies for the treatment of kidney disorders remain elusive. We report an optimized kidney-targeted gene delivery strategy using recombinant adeno-associated virus (rAAV) administered via retrograde renal vein injection in mice. Renal vein injection of rAAV consistently resulted in superior kidney transduction compared with tail vein injection using as little as half the tail vein dose. We compared rAAV5, 6, 8 and 9, containing either green fluorescent protein (GFP) or luciferase reporter genes driven by the Cytomegalovirus promoter. We demonstrated that although rAAV6 and 8 injected via renal vein transduced the kidney, transgene expression was mainly restricted to the medulla. Transgene expression was systematically low after rAAV5 injection, attributed to T-cell immune response, which could be overcome by transient immunosuppression. However, rAAV9 was the only serotype that permitted high-transduction efficiency of both the cortex and medulla. Moreover, both the glomeruli and tubules were targeted, with a higher efficiency within the glomeruli. To improve the specificity of kidney-targeted gene delivery with rAAV9, we used the parathyroid hormone receptor 'kidney-specific' promoter. We obtained a more efficient transgene expression within the kidney, and a significant reduction in other tissues. Our work represents the first comprehensive and clinically relevant study for kidney gene delivery.
Collapse
Affiliation(s)
- Céline J. Rocca
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Sarah N. Ur
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Frank Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| |
Collapse
|
32
|
Ismailov SM, Barykova YA, Shmarov MM, Tarantul VZ, Barskov IV, Kucheryanu VG, Brylev LV, Logunov DY, Tutykhina IL, Bocharov EV, Zakharova MN, Narodizky BS, Illarioshkin SN. Experimental approach to the gene therapy of motor neuron disease with the use of genes hypoxia-inducible factors. RUSS J GENET+ 2014. [DOI: 10.1134/s1022795414050056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Xue C, Tian X, Li X, Zhou Z, Su X, Zhou R. Construction and characterization of a recombinant human adenovirus type 3 vector containing two foreign neutralizing epitopes in hexon. Virus Res 2014; 183:67-74. [PMID: 24518297 DOI: 10.1016/j.virusres.2014.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/25/2014] [Accepted: 01/31/2014] [Indexed: 01/09/2023]
Abstract
The "antigen capsid-incorporation" strategy has been developed for adenovirus-based vaccines in the context of several diseases. Exogenous antigenic peptides incorporated into the adenovirus capsid structure can induce a robust and boosted antigen-specific immune response. Recently, we sought to generate a multivalent adenovirus type 3 (Ad3) vaccine vector by incorporating multiple epitopes into the major adenovirus capsid protein, hexon. In the present study, a multivalent recombinant Ad3 vaccine (R1R2A3) was constructed by homologous recombination, displaying two neutralizing epitopes from enterovirus type 71 (EV71) in hexon. The recombinant virus was confirmed by PCR, immunoblotting, and enzyme-linked immunosorbent assay, and injected into mice to analyze the epitope-specific humoral response. No differences were found between the viruses with two epitopes incorporated into the hypervariable regions (HVR1 and HVR2) of hexon and Ad3EGFP, based on thermostability and growth kinetic tests. Both the epitopes are thought to be exposed on the hexon-modified intact virion surface. The repeated administration of the modified adenovirus R1R2A3 to BALB/c mice boosted the humoral immune response against both epitopes. Immunization with recombinant virus R1R2A3 elicited higher IgG titers and higher neutralization titers against EV71 in vitro than immunization with the modified adenovirus with only one epitope incorporated into HVR1. In this study, the recombinant R1R2A3 virus expressing two exogenous neutralizing epitopes in hexon HVR1 and HVR2 induced specific immune responses to both foreign epitopes. Our study contributes to a better understanding of hexon-modified Ad vector as a multiple-epitope delivery vehicle.
Collapse
Affiliation(s)
- Chunyan Xue
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120 , China.
| | - Xingui Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120 , China.
| | - Xiao Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120 , China.
| | - Zhichao Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120 , China.
| | - Xiaobo Su
- Department of Medical Genetics and Cell Biology, School of Basic Science, Guangzhou Medical University, Guangzhou 510120, China.
| | - Rong Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120 , China.
| |
Collapse
|
34
|
Gherardini L, Bardi G, Gennaro M, Pizzorusso T. Novel siRNA delivery strategy: a new "strand" in CNS translational medicine? Cell Mol Life Sci 2014; 71:1-20. [PMID: 23508806 PMCID: PMC11113879 DOI: 10.1007/s00018-013-1310-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 12/12/2022]
Abstract
RNA interference has been envisaged as a powerful tool for molecular and clinical investigation with a great potential for clinical applications. In recent years, increased understanding of cancer biology and stem cell biology has dramatically accelerated the development of technology for cell and gene therapy in these areas. This paper is a review of the most recent report of innovative use of siRNA to benefit several central nervous system diseases. Furthermore, a description is made of innovative strategies of delivery into the brain by means of viral and non-viral vectors with high potential for translation into clinical use. Problems are also highlighted that might hamper the transition from bench to bed, analyzing the lack of reliable preclinical models with predictive validity and the lack of effective delivery systems, which are able to overcome biological barriers and specifically reach the brain site of action.
Collapse
Affiliation(s)
| | - Giuseppe Bardi
- Center for MicroBioRobotics @SSSA, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | | | - Tommaso Pizzorusso
- Institute of Neuroscience, CNR, Via Moruzzi, 1 56124 Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Florence, Italy
| |
Collapse
|
35
|
Gehlbach PL, Chuck RS, Park CG, Park CY. Viral Transgene Expression Delivered by Repeat Intraocular Adenoviral Vector Injection: in Vivo Live Imaging Study. Mol Imaging 2012. [DOI: 10.2310/7290.2011.00053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Peter L. Gehlbach
- From the Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD; Department of Ophthalmology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Cancer Research Institute, Tumor Immunity Medical Research Center, Seoul National University, College of Medicine, Seoul, South Korea; and Department of Ophthalmology, Dongguk University Ilsan Hospital, Koyang, South Korea
| | - Roy S. Chuck
- From the Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD; Department of Ophthalmology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Cancer Research Institute, Tumor Immunity Medical Research Center, Seoul National University, College of Medicine, Seoul, South Korea; and Department of Ophthalmology, Dongguk University Ilsan Hospital, Koyang, South Korea
| | - Chung-Gyu Park
- From the Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD; Department of Ophthalmology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Cancer Research Institute, Tumor Immunity Medical Research Center, Seoul National University, College of Medicine, Seoul, South Korea; and Department of Ophthalmology, Dongguk University Ilsan Hospital, Koyang, South Korea
| | - Choul Yong Park
- From the Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD; Department of Ophthalmology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Cancer Research Institute, Tumor Immunity Medical Research Center, Seoul National University, College of Medicine, Seoul, South Korea; and Department of Ophthalmology, Dongguk University Ilsan Hospital, Koyang, South Korea
| |
Collapse
|
36
|
Characterization of malleability and immunological properties of human adenovirus type 3 hexon hypervariable region 1. Arch Virol 2012; 157:1709-18. [PMID: 22669318 DOI: 10.1007/s00705-012-1364-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 04/26/2012] [Indexed: 12/20/2022]
Abstract
Adenovirus (Ad) capsids that display exogenous epitopes can be potently immunogenic, eliciting a potent humoral response against components of the capsid. We used the epitopes flag, his(6)flag, his(6)lgsflag and AdV4HVR5 as model antigens to characterize the hexon hypervariable region (HVR) 1 as a site for epitope insertion. A peptide of up to 17 amino acids could be incorporated into HVR1 of the Ad3 hexon without adversely affecting the biological characteristics of the virus. Multiple vaccinations with capsid-modified Ad3 induced a humoral response against the epitope inserted in HVR1. However, antiserum against the his(6)flag or his(6)lgsflag epitope did not recognize glutathione S-transferase (GST)-his(6) and GST-flag fusion protein. Our study illustrates that there is an immune response against the new epitope within the amino acids of his(6)flag or his(6)lgsflag epitopes. This discovery could be a warning for the generation of multivalent vaccine vectors by incorporation of multiple epitopes into single HVRs.
Collapse
|
37
|
Local delivery of siRNA using a biodegradable polymer application to enhance BMP-induced bone formation. Biomaterials 2011; 32:9642-8. [DOI: 10.1016/j.biomaterials.2011.08.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 08/09/2011] [Indexed: 02/04/2023]
|
38
|
Green WJF, James PA, Ratan HL. Potential use of RNA interference as therapeutic strategy in urologic cancer. Urology 2011; 78:500-4. [PMID: 21741681 DOI: 10.1016/j.urology.2011.04.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 03/20/2011] [Accepted: 04/16/2011] [Indexed: 11/28/2022]
|
39
|
Pavelko KD, Girtman MA, Mitsunaga Y, Mendez-Fernandez YV, Bell MP, Hansen MJ, Allen KS, Rodriguez M, Pease LR. Theiler's murine encephalomyelitis virus as a vaccine candidate for immunotherapy. PLoS One 2011; 6:e20217. [PMID: 21625449 PMCID: PMC3098865 DOI: 10.1371/journal.pone.0020217] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 04/15/2011] [Indexed: 12/29/2022] Open
Abstract
The induction of sterilizing T-cell responses to tumors is a major goal in the development of T-cell vaccines for treating cancer. Although specific components of anti-viral CD8+ immunity are well characterized, we still lack the ability to mimic viral CD8+ T-cell responses in therapeutic settings for treating cancers. Infection with the picornavirus Theiler's murine encephalomyelitis virus (TMEV) induces a strong sterilizing CD8+ T-cell response. In the absence of sterilizing immunity, the virus causes a persistent infection. We capitalized on the ability of TMEV to induce strong cellular immunity even under conditions of immune deficiency by modifying the virus to evaluate its potential as a T-cell vaccine. The introduction of defined CD8+ T-cell epitopes into the leader sequence of the TMEV genome generates an attenuated vaccine strain that can efficiently drive CD8+ T-cell responses to the targeted antigen. This virus activates T-cells in a manner that is capable of inducing targeted tissue damage and glucose dysregulation in an adoptive T-cell transfer model of diabetes mellitus. As a therapeutic vaccine for the treatment of established melanoma, epitope-modified TMEV can induce strong cytotoxic T-cell responses and promote infiltration of the T-cells into established tumors, ultimately leading to a delay in tumor growth and improved survival of vaccinated animals. We propose that epitope-modified TMEV is an excellent candidate for further development as a human T-cell vaccine for use in immunotherapy.
Collapse
Affiliation(s)
- Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Megan A. Girtman
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Yoshihiro Mitsunaga
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | - Michael P. Bell
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael J. Hansen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kathleen S. Allen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Moses Rodriguez
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Larry R. Pease
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
40
|
Gregory SM, Nazir SA, Metcalf JP. Implications of the innate immune response to adenovirus and adenoviral vectors. Future Virol 2011; 6:357-374. [PMID: 21738557 DOI: 10.2217/fvl.11.6] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenovirus (AdV) is a common cause of respiratory illness in both children and adults. Respiratory symptoms can range from those of the common cold to severe pneumonia. Infection can also cause significant disease in the immunocompromised and among immunocompetent subjects in close quarters. Fortunately, infection with AdV in the normal host is generally mild. This is one reason why its initial use as a gene-therapy vector appeared to be so promising. Unfortunately, both innate and adaptive responses to the virus have limited the development of AdV vectors as a tool of gene therapy by increasing toxicity and limiting duration of transgene expression. This article will focus on the innate immune response to infection with wild-type AdV and exposure to AdV gene-therapy vectors. As much of the known information relates to the pulmonary inflammatory response, this organ system will be emphasized. This article will also discuss how that understanding has led to the creation of new vectors for use in gene therapy.
Collapse
Affiliation(s)
- Seth M Gregory
- Division of Pulmonary & Critical Care Medicine of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | |
Collapse
|
41
|
Horst D, Ressing ME, Wiertz EJHJ. Exploiting human herpesvirus immune evasion for therapeutic gain: potential and pitfalls. Immunol Cell Biol 2011; 89:359-66. [PMID: 21301483 DOI: 10.1038/icb.2010.129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herpesviruses stand out for their capacity to establish lifelong infections of immunocompetent hosts, generally without causing overt symptoms. Herpesviruses are equipped with sophisticated immune evasion strategies, allowing these viruses to persist for life despite the presence of a strong antiviral immune response. Although viral evasion tactics appear to target virtually any stage of the innate and adaptive host immune response, detailed knowledge is now available on the molecular mechanisms underlying herpesvirus obstruction of MHC class I-restricted antigen presentation to T cells. This opens the way for clinical application. Here, we review and discuss recent efforts to exploit human herpesvirus MHC class I evasion strategies for the rational design of novel strategies for vaccine development, cancer treatment, transplant protection and gene therapy.
Collapse
Affiliation(s)
- Daniëlle Horst
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
42
|
Kakinoki K, Nakamoto Y, Kagaya T, Tsuchiyama T, Sakai Y, Nakahama T, Mukaida N, Kaneko S. Prevention of intrahepatic metastasis of liver cancer by suicide gene therapy and chemokine ligand 2/monocyte chemoattractant protein-1 delivery in mice. J Gene Med 2010; 12:1002-1013. [PMID: 21157824 DOI: 10.1002/jgm.1528] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The prognosis of patients with hepatocellular carcinoma (HCC) remains poor, largely as a result of intrahepatic metastasis. Using a mouse model of intrahepatic metastasis, we investigated whether chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) could potentiate the antitumor effects of the herpes simplex virus thymidine kinase/ganciclovir (HSV-tk/GCV) system. METHODS Mouse hepatoma cells infected with recombinant adenovirus vectors expressing HSV-tk, CCL2/MCP-1 and LacZ at multiplicities of infection of Ad-tk/Ad-MCP1 = 3/0.03 (T/M(Low)), 3/3 (T/M(High)) and Ad-tk/Ad-LacZ = 3/3 (T/L) were injected into BALB/c mice. RESULTS Intrahepatic tumor growth was significantly lower in T/M(Low) mice. By contrast, no tumor suppression was observed in T/M(High) mice. The tumor-specific cytolytic activities of splenocytes from T/M(Low) and T/M(High) mice were comparable. Immunohistochemical analysis of liver tissues showed similar infiltration by Mac-1(+) and T cells in these animals, whereas the proportions of classical activated (M1) monocytes/macrophages were significantly higher in T/M(Low) mice. In addition, interleukin-12 production was elevated in these tissues. Vascular endothelial growth factor-A expression and CD31(+) microvessels were increased in T/M(High) mice. CONCLUSIONS Collectively, these results demonstrate that an adequate amount of CCL2/MCP-1, together with the HSV-tk/GCV system, may induce T helper 1-polarized antitumor effects without inducing tumor angiogenesis in the microenvironment of intrahepatic HCC progression.
Collapse
Affiliation(s)
- Kaheita Kakinoki
- Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Matthews QL. Capsid-incorporation of antigens into adenovirus capsid proteins for a vaccine approach. Mol Pharm 2010; 8:3-11. [PMID: 21047139 DOI: 10.1021/mp100214b] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Some viral vectors are potent inducers of cellular and humoral responses; therefore, viral vectors can be used to vaccinate against cancer or infectious diseases. This report will focus on adenovirus (Ad)-based vectors. Traditional viral-vector vaccination embodies the concept that the vector uses the host-cell machinery to express antigens that are encoded as transgenes within the viral vector. Several preclinical successes have used this approach in animal model systems. However, in some instances, these conventional Ad-based vaccines have yielded suboptimal clinical results. These suboptimal results are ascribed, in part, to preexisting Ad serotype 5 (Ad5) immunity. To address this issue, the "antigen capsid-incorporation" strategy has been developed to circumvent the drawbacks associated with conventional transgene expression of antigens by Ad vectors. This strategy embodies the incorporation of antigenic peptides within the capsid structure of viral vectors. Incorporating immunogenic peptides into the Ad capsid offers potential advantages. Importantly, vaccination by means of the antigen capsid-incorporated approach results in a strong humoral response, similar to the response generated by native Ad capsid proteins. This strategy also allows for the boosting of antigenic specific responses. This strategy may be the way forward for improved vaccine schemes, especially for those infections requiring a strong humoral antigenic response.
Collapse
Affiliation(s)
- Qiana L Matthews
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, the Gene Therapy Center, and Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
44
|
Seregin SS, Amalfitano A. Improving adenovirus based gene transfer: strategies to accomplish immune evasion. Viruses 2010; 2:2013-2036. [PMID: 21994718 PMCID: PMC3185744 DOI: 10.3390/v2092013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/03/2010] [Accepted: 09/15/2010] [Indexed: 12/20/2022] Open
Abstract
Adenovirus (Ad) based gene transfer vectors continue to be the platform of choice for an increasing number of clinical trials worldwide. In fact, within the last five years, the number of clinical trials that utilize Ad based vectors has doubled, indicating growing enthusiasm for the numerous positive characteristics of this gene transfer platform. For example, Ad vectors can be easily and relatively inexpensively produced to high titers in a cGMP compliant manner, can be stably stored and transported, and have a broad applicability for a wide range of clinical conditions, including both gene therapy and vaccine applications. Ad vector based gene transfer will become more useful as strategies to counteract innate and/or pre-existing adaptive immune responses to Ads are developed and confirmed to be efficacious. The approaches attempting to overcome these limitations can be divided into two broad categories: pre-emptive immune modulation of the host, and selective modification of the Ad vector itself. The first category of methods includes the use of immunosuppressive drugs or specific compounds to block important immune pathways, which are known to be induced by Ads. The second category comprises several innovative strategies inclusive of: (1) Ad-capsid-display of specific inhibitors or ligands; (2) covalent modifications of the entire Ad vector capsid moiety; (3) the use of tissue specific promoters and local administration routes; (4) the use of genome modified Ads; and (5) the development of chimeric or alternative serotype Ads. This review article will focus on both the promise and the limitations of each of these immune evasion strategies, and in the process delineate future directions in developing safer and more efficacious Ad-based gene transfer strategies.
Collapse
Affiliation(s)
- Sergey S. Seregin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; E-Mail:
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; E-Mail:
- Department of Pediatrics, Michigan State University, East Lansing, MI 48824, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-517-884-5324; Fax: +1-517-353-8957
| |
Collapse
|
45
|
Matthews QL, Fatima A, Tang Y, Perry BA, Tsuruta Y, Komarova S, Timares L, Zhao C, Makarova N, Borovjagin AV, Stewart PL, Wu H, Blackwell JL, Curiel DT. HIV antigen incorporation within adenovirus hexon hypervariable 2 for a novel HIV vaccine approach. PLoS One 2010; 5:e11815. [PMID: 20676400 PMCID: PMC2910733 DOI: 10.1371/journal.pone.0011815] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 06/27/2010] [Indexed: 11/18/2022] Open
Abstract
Adenoviral (Ad) vectors have been used for a variety of vaccine applications including cancer and infectious diseases. Traditionally, Ad-based vaccines are designed to express antigens through transgene expression of a given antigen. However, in some cases these conventional Ad-based vaccines have had sub-optimal clinical results. These sub-optimal results are attributed in part to pre-existing Ad serotype 5 (Ad5) immunity. In order to circumvent the need for antigen expression via transgene incorporation, the “antigen capsid-incorporation” strategy has been developed and used for Ad-based vaccine development in the context of a few diseases. This strategy embodies the incorporation of antigenic peptides within the capsid structure of viral vectors. The major capsid protein hexon has been utilized for these capsid incorporation strategies due to hexon's natural role in the generation of anti-Ad immune response and its numerical representation within the Ad virion. Using this strategy, we have developed the means to incorporate heterologous peptide epitopes specifically within the major surface-exposed domains of the Ad capsid protein hexon. Our study herein focuses on generation of multivalent vaccine vectors presenting HIV antigens within the Ad capsid protein hexon, as well as expressing an HIV antigen as a transgene. These novel vectors utilize HVR2 as an incorporation site for a twenty-four amino acid region of the HIV membrane proximal ectodomain region (MPER), derived from HIV glycoprotein gp41 (gp41). Our study herein illustrates that our multivalent anti-HIV vectors elicit a cellular anti-HIV response. Furthermore, vaccinations with these vectors, which present HIV antigens at HVR2, elicit a HIV epitope-specific humoral immune response.
Collapse
Affiliation(s)
- Qiana L. Matthews
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Aiman Fatima
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Yizhe Tang
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Vision Science Graduate Program, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Brian A. Perry
- Davidson College, Davidson, North Carolina, United States of America
| | - Yuko Tsuruta
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Svetlana Komarova
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Laura Timares
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Chunxia Zhao
- Emory University, Atlanta, Georgia, United States of America
| | | | - Anton V. Borovjagin
- Institute of Oral Health Research, University of Alabama at Birmingham, School of Dentistry, Birmingham, Alabama, United States of America
| | - Phoebe L. Stewart
- Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Hongju Wu
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Pathology, Surgery, Obstetrics and Gynecology, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
46
|
Hom C, Lu J, Liong M, Luo H, Li Z, Zink JI, Tamanoi F. Mesoporous silica nanoparticles facilitate delivery of siRNA to shutdown signaling pathways in mammalian cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2010; 6:1185-90. [PMID: 20461725 PMCID: PMC2953950 DOI: 10.1002/smll.200901966] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Affiliation(s)
- Christopher Hom
- Department of Microbiology, Immunology, and Molecular Genetics, California NanoSystems Institute, JCCC, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| | - Jie Lu
- Department of Microbiology, Immunology, and Molecular Genetics, California NanoSystems Institute, JCCC, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| | - Monty Liong
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, 605 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| | - Hanzhi Luo
- School of Basic Medical Sciences, Health Science Center, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191 (P.R. China)
| | - Zongxi Li
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, 605 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| | - Jeffrey I. Zink
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, 605 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| | - Fuyuhiko Tamanoi
- Department of Microbiology, Immunology, and Molecular Genetics, California NanoSystems Institute, JCCC, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, CA 90095 (USA)
| |
Collapse
|
47
|
Wonganan P, Croyle MA. PEGylated Adenoviruses: From Mice to Monkeys. Viruses 2010; 2:468-502. [PMID: 21994645 PMCID: PMC3185605 DOI: 10.3390/v2020468] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 01/20/2010] [Accepted: 01/25/2010] [Indexed: 12/13/2022] Open
Abstract
Covalent modification with polyethylene glycol (PEG), a non-toxic polymer used in food, cosmetic and pharmaceutical preparations for over 60 years, can profoundly influence the pharmacokinetic, pharmacologic and toxciologic profile of protein and peptide-based therapeutics. This review summarizes the history of PEGylation and PEG chemistry and highlights the value of this technology in the context of the design and development of recombinant viruses for gene transfer, vaccination and diagnostic purposes. Specific emphasis is placed on the application of this technology to the adenovirus, the most potent viral vector with the most highly characterized toxicity profile to date, in several animal models.
Collapse
Affiliation(s)
- Piyanuch Wonganan
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; E-Mail:
| | - Maria A. Croyle
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; E-Mail:
- Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-512-471-1972; Fax: +1-512-471-7474
| |
Collapse
|
48
|
Expression of Green Fluorescent Protein Gene in Somatic Chimeric Chickens Produced by Transplantation of Transfected Chicken Embryonic Fibroblasts. J Poult Sci 2009. [DOI: 10.2141/jpsa.46.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
49
|
Patil Y, Panyam J. Polymeric nanoparticles for siRNA delivery and gene silencing. Int J Pharm 2008; 367:195-203. [PMID: 18940242 DOI: 10.1016/j.ijpharm.2008.09.039] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 09/15/2008] [Accepted: 09/22/2008] [Indexed: 01/01/2023]
Abstract
Gene silencing using small interfering RNA (siRNA) has several potential therapeutic applications. In the present study, we investigated nanoparticles formulated using the biodegradable polymer, poly(d,l-lactide-co-glycolide) (PLGA) for siRNA delivery. A cationic polymer, polyethylenimine (PEI), was incorporated in the PLGA matrix to improve siRNA encapsulation in PLGA nanoparticles. PLGA-PEI nanoparticles were formulated using double emulsion-solvent evaporation technique and characterized for siRNA encapsulation and in vitro release. The effectiveness of siRNA-loaded PLGA-PEI nanoparticles in silencing a model gene, fire-fly luciferase, was investigated in cell culture. Presence of PEI in PLGA nanoparticle matrix increased siRNA encapsulation by about 2-fold and also improved the siRNA release profile. PLGA-PEI nanoparticles carrying luciferase-targeted siRNA enabled effective silencing of the gene in cells stably expressing luciferase as well as in cells that could be induced to overexpress the gene. Quantitative studies indicated that presence of PEI in PLGA nanoparticles resulted in 2-fold higher cellular uptake of nanoparticles while fluorescence microscopy studies showed that PLGA-PEI nanoparticles delivered the encapsulated siRNA in the cellular cytoplasm; both higher uptake and greater cytosolic delivery could have contributed to the gene silencing effectiveness of PLGA-PEI nanoparticles. Serum stability and lack of cytotoxicity further add to the potential of PLGA-PEI nanoparticles in gene silencing-based therapeutic applications.
Collapse
Affiliation(s)
- Yogesh Patil
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | | |
Collapse
|
50
|
Abstract
The ultimate goal of gene therapy is the replacement of a defective gene sequence with a corrected version to eliminate disease for the lifetime of the patient. This challenging task is not yet accomplished, however significant progress is evident. An initial spate of clinical trials attempting the treatment of haemophilia with gene transfer primarily resulted in the demonstration of good safety profiles, but without efficacy. Subsequent reengineering of vector plasmids and delivery systems resulted in markedly improved outcomes in animal models of the disease. The most recent clinical trial for the treatment of haemophilia B with gene transfer showed transient achievement of efficacy in the highest dose cohort tested, but also exposed a previously hidden barrier to the future success of these treatments. The progress and problems of gene therapies for haemorrhagic disorders will be discussed. This review will concentrate on approaches in or near clinical application.
Collapse
Affiliation(s)
- Samuel L Murphy
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | |
Collapse
|