1
|
Guo Z, Wu F, Guo C, Hu R, Ou Y, Zhu Y, Luo S, Song Y, He P, He C, Xu Y, Tang X, Qin M, Wang H, Du G, Sun X. Metalloparticle-Engineered Pickering Emulsion Displaying AAV-Vectored Vaccine for Enhancing Antigen Expression and Immunogenicity Against Pathogens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412627. [PMID: 39828538 DOI: 10.1002/adma.202412627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Recombinant adeno-associated viruses (rAAVs) have emerged as promising vaccine vectors due to their enduring efficacy with a single dose. However, insufficient cellular immune responses and the random and non-specific distribution of AAVs post-injection may hinder the development of AAV vaccines. Here, a novel Pickering emulsion platform stabilized by biomineralized manganese nanoparticles and aluminum hydroxide, which can rapidly and efficiently load AAVs, is reported. This platform confers AAVs with favorable in vivo distribution kinetics, diversifying AAV endocytic pathways with reduced dependency on the sialic acid receptor-mediated route, and ultimately enhancing AAV infection efficiency in antigen present cells (APCs). Concurrently, the Pickering emulsion substantially boosts endogenous 2'3'-cGAMP production, further activating the cGAS-STING pathway for stronger immune responses and improving protective efficacy in bacterial infection models. The STING pathway activation also increases AAV target gene expression, potently augmenting the cross-protective potential of AAV vaccines for COVID-19. These synergistic effects ensure that effective immune responses are induced even at one-fifth of the AAV vaccination dose, while the Pickering emulsion further reduces the accumulation of AAV in the liver, thereby improving their safety. The findings highlight the potential of Pickering emulsions as valuable enhancers for viral vectors, providing insights for their broader clinical applicability.
Collapse
Affiliation(s)
- Zhaofei Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Fuhua Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Chenqi Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yining Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shuang Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuanshuai Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xue Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Ming Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
2
|
Chauhan M, Daugherty AL, Khadir FE, Duzenli OF, Hoffman A, Tinklenberg JA, Kang PB, Aslanidi G, Pacak CA. AAV-DJ is superior to AAV9 for targeting brain and spinal cord, and de-targeting liver across multiple delivery routes in mice. J Transl Med 2024; 22:824. [PMID: 39237935 PMCID: PMC11375878 DOI: 10.1186/s12967-024-05599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Highly efficient adeno associated viruses (AAVs) targeting the central nervous system (CNS) are needed to deliver safe and effective therapies for inherited neurological disorders. The goal of this study was to compare the organ-specific transduction efficiencies of two AAV capsids across three different delivery routes. We compared AAV9-CBA-fLucYFP to AAV-DJ-CBA-fLucYFP using the following delivery routes in mice: intracerebroventricular (ICV) 1 × 1012 vg/kg, intrathecal (IT) 1 × 1012 vg/kg, and intravenous (IV) 1 × 1013 vg/kg body weight. Our evaluations revealed that following ICV and IT administrations, AAV-DJ demonstrated significantly increased vector genome (vg) uptake throughout the CNS as compared to AAV9. Through the IV route, AAV9 demonstrated significantly increased vg uptake in the CNS. However, significantly fewer vgs were detected in the off-target organs (kidney and liver) following administration of AAV-DJ using the IT and IV delivery routes as compared to AAV9. Distributions of vgs correlate well with transgene transcript levels, luciferase enzyme activities, and immunofluorescence detection of YFP. Overall, between the two vectors, AAV-DJ resulted in better targeting and expression in CNS tissues paired with de-targeting and reduced expression in liver and kidneys. Our findings support further examination of AAV-DJ as a gene therapy capsid for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Monika Chauhan
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Audrey L Daugherty
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Fatemeh Ellie Khadir
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Ozgun F Duzenli
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | | | - Jennifer A Tinklenberg
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Peter B Kang
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Christina A Pacak
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA.
| |
Collapse
|
3
|
Tang X, Wang L, Wang D, Zhang Y, Wang T, Zhu Z, Weng Y, Tao G, Wang Q, Tang L, Yan F, Wang Y. Maggot extracts chemo-prevent inflammation and tumorigenesis accompanied by changes in the intestinal microbiome and metabolome in AOM/DSS-induced mice. Front Microbiol 2023; 14:1143463. [PMID: 37200915 PMCID: PMC10185807 DOI: 10.3389/fmicb.2023.1143463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/29/2023] [Indexed: 05/20/2023] Open
Abstract
Inflammatory responses and intestinal microbiome play a crucial role in the progression of colitis-associated carcinoma (CAC). The traditional Chinese medicine maggot has been widely known owing to its clinical application and anti-inflammatory function. In this study, we investigated the preventive effects of maggot extract (ME) by intragastric administration prior to azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CAC in mice. The results showed that ME had superior advantages in ameliorating disease activity index score and inflammatory phenotype, in comparison with the AOM/DSS group. The number and size of polypoid colonic tumors were decreased after pre-administration of ME. In addition, ME was found to reverse the downregulation of tight junction proteins (zonula occluden-1 and occluding) while suppressing the levels of inflammatory factors (IL-1β and IL-6) in models. Moreover, Toll-like receptor 4 (TLR4) mediated intracellular nuclear factor-κB (NF-κB)-containing signaling cascades, including inducible nitric oxide synthase and cyclooxygenase-2, and exhibited decreasing expression in the mice model after ME pre-administration. 16s rRNA analysis and untargeted-metabolomics profiling of fecal samples inferred that ME revealed ideal prevention of intestinal dysbiosis in CAC mice, accompanied by and correlated with alterations in the composition of metabolites. Overall, ME pre-administration might be a chemo-preventive candidate in the initiation and development of CAC.
Collapse
Affiliation(s)
- Xun Tang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Lei Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Daojuan Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Tingyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhengquan Zhu
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yajing Weng
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Gaojian Tao
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin Wang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Li Tang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Feng Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
- *Correspondence: Feng Yan
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Nanjing University (Suzhou) High-Tech Institute, Nanjing University, Suzhou, China
- Yong Wang
| |
Collapse
|
4
|
Mollard A, Peccate C, Forand A, Chassagne J, Julien L, Meunier P, Guesmia Z, Marais T, Bitoun M, Piétri-Rouxel F, Benkhelifa-Ziyyat S, Lorain S. Muscle regeneration affects Adeno Associated Virus 1 mediated transgene transcription. Sci Rep 2022; 12:9674. [PMID: 35690627 PMCID: PMC9188557 DOI: 10.1038/s41598-022-13405-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy is a severe neuromuscular disease causing a progressive muscle wasting due to mutations in the DMD gene that lead to the absence of dystrophin protein. Adeno-associated virus (AAV)-based therapies aiming to restore dystrophin in muscles, by either exon skipping or microdystrophin expression, are very promising. However, the absence of dystrophin induces cellular perturbations that hinder AAV therapy efficiency. We focused here on the impact of the necrosis-regeneration process leading to nuclear centralization in myofiber, a common feature of human myopathies, on AAV transduction efficiency. We generated centronucleated myofibers by cardiotoxin injection in wild-type muscles prior to AAV injection. Intramuscular injections of AAV1 vectors show that transgene expression was drastically reduced in regenerated muscles, even when the AAV injection occurred 10 months post-regeneration. We show also that AAV genomes were not lost from cardiotoxin regenerated muscle and were properly localised in the myofiber nuclei but were less transcribed leading to muscle transduction defect. A similar defect was observed in muscles of the DMD mouse model mdx. Therefore, the regeneration process per se could participate to the AAV-mediated transduction defect observed in dystrophic muscles which may limit AAV-based therapies.
Collapse
Affiliation(s)
- Amédée Mollard
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Anne Forand
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Julie Chassagne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Laura Julien
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Pierre Meunier
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Thibaut Marais
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Marc Bitoun
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - France Piétri-Rouxel
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Stéphanie Lorain
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.,AFM-Téléthon, 1 rue de l'Internationale, BP59, 91002, Evry, France
| |
Collapse
|
5
|
Mehta N, Robbins DA, Yiu G. Ocular Inflammation and Treatment Emergent Adverse Events in Retinal Gene Therapy. Int Ophthalmol Clin 2021; 61:151-177. [PMID: 34196322 PMCID: PMC8259781 DOI: 10.1097/iio.0000000000000366] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Neesurg Mehta
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA
| | - Deborah Ahn Robbins
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA
| |
Collapse
|
6
|
Whitehead M, Osborne A, Yu-Wai-Man P, Martin K. Humoral immune responses to AAV gene therapy in the ocular compartment. Biol Rev Camb Philos Soc 2021; 96:1616-1644. [PMID: 33837614 DOI: 10.1111/brv.12718] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022]
Abstract
Viral vectors can be utilised to deliver therapeutic genes to diseased cells. Adeno-associated virus (AAV) is a commonly used viral vector that is favoured for its ability to infect a wide range of tissues whilst displaying limited toxicity and immunogenicity. Most humans harbour anti-AAV neutralising antibodies (NAbs) due to subclinical infections by wild-type virus during infancy and these pre-existing NAbs can limit the efficiency of gene transfer depending on the target cell type, route of administration and choice of serotype. Vector administration can also result in de novo NAb synthesis that could limit the opportunity for repeated gene transfer to diseased sites. A number of strategies have been described in preclinical models that could circumvent NAb responses in humans, however, the successful translation of these innovations into the clinical arena has been limited. Here, we provide a comprehensive review of the humoral immune response to AAV gene therapy in the ocular compartment. We cover basic AAV biology and clinical application, the role of pre-existing and induced NAbs, and possible approaches to overcoming antibody responses. We conclude with a framework for a comprehensive strategy for circumventing humoral immune responses to AAV in the future.
Collapse
Affiliation(s)
- Michael Whitehead
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K
| | - Andrew Osborne
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K
| | - Patrick Yu-Wai-Man
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K.,MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, U.K.,NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, U.K
| | - Keith Martin
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K.,Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, U.K.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Goodwin MS, Croft CL, Futch HS, Ryu D, Ceballos-Diaz C, Liu X, Paterno G, Mejia C, Deng D, Menezes K, Londono L, Arjona K, Parianos M, Truong V, Rostonics E, Hernandez A, Boye SL, Boye SE, Levites Y, Cruz PE, Golde TE. Utilizing minimally purified secreted rAAV for rapid and cost-effective manipulation of gene expression in the CNS. Mol Neurodegener 2020; 15:15. [PMID: 32122372 PMCID: PMC7053119 DOI: 10.1186/s13024-020-00361-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/13/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Recombinant adeno-associated virus (rAAV) is widely used in the neuroscience field to manipulate gene expression in the nervous system. However, a limitation to the use of rAAV vectors is the time and expense needed to produce them. To overcome this limitation, we evaluated whether unpurified rAAV vectors secreted into the media following scalable PEI transfection of HEK293T cells can be used in lieu of purified rAAV. METHODS We packaged rAAV2-EGFP vectors in 30 different wild-type and mutant capsids and subsequently collected the media containing secreted rAAV. Genomic titers of each rAAV vector were assessed and the ability of each unpurified virus to transduce primary mixed neuroglial cultures (PNGCs), organotypic brain slice cultures (BSCs) and the mouse brain was evaluated. RESULTS There was ~ 40-fold wide variance in the average genomic titers of the rAAV2-EGFP vector packaged in the 30 different capsids, ranging from a low ~ 4.7 × 1010 vector genomes (vg)/mL for rAAV2/5-EGFP to a high of ~ 2.0 × 1012 vg/mL for a capsid mutant of rAAV2/8-EGFP. In PNGC studies, we observed a wide range of transduction efficiency among the 30 capsids evaluated, with the rAAV2/6-EGFP vector demonstrating the highest overall transduction efficiency. In BSC studies, we observed robust transduction by wild-type capsid vectors rAAV2/6, 2/8 and 2/9, and by capsid mutants of rAAV2/1, 2/6, and 2/8. In the in vivo somatic brain transgenesis (SBT) studies, we found that intra-cerebroventricular injection of media containing unpurified rAAV2-EGFP vectors packaged with select mutant capsids resulted in abundant EGFP positive neurons and astrocytes in the hippocampus and forebrain of non-transgenic mice. We demonstrate that unpurified rAAV can express transgenes at equivalent levels to lysate-purified rAAV both in vitro and in vivo. We also show that unpurified rAAV is sufficient to drive tau pathology in BSC and neuroinflammation in vivo, recapitulating previous studies using purified rAAV. CONCLUSIONS Unpurified rAAV vectors secreted into the media can efficiently transduce brain cells in vitro and in vivo, providing a cost-effective way to manipulate gene expression. The use of unpurified virus will greatly reduce costs of exploratory studies and further increase the utility of rAAV vectors for standard laboratory use.
Collapse
Affiliation(s)
- Marshall S. Goodwin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Cara L. Croft
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Hunter S. Futch
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Daniel Ryu
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Carolina Ceballos-Diaz
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Xuefei Liu
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Catalina Mejia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Doris Deng
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Kimberly Menezes
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Laura Londono
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Kefren Arjona
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Mary Parianos
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Van Truong
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Eva Rostonics
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Amanda Hernandez
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Sanford L. Boye
- Department of Pediatrics and the Powell Gene Therapy Center, University of Florida, Gainesville, Florida USA
| | - Shannon E. Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida USA
| | - Yona Levites
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Pedro E. Cruz
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
| | - Todd E. Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida USA
| |
Collapse
|
8
|
Chen L, Kuang P, Liu H, Wei Q, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Sodium Fluoride (NaF) Induces Inflammatory Responses Via Activating MAPKs/NF-κB Signaling Pathway and Reducing Anti-inflammatory Cytokine Expression in the Mouse Liver. Biol Trace Elem Res 2019; 189:157-171. [PMID: 30062462 DOI: 10.1007/s12011-018-1458-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023]
Abstract
At present, no reports are focused on fluoride-induced hepatic inflammatory responses in human beings and animals. This study aimed to investigate the mRNA and protein levels of inflammatory cytokines and signaling molecules for evaluating the effect of different doses (0, 12, 24, and 48 mg/kg) of sodium fluoride (NaF) on inflammatory reaction in the mouse liver by using methods of experimental pathology, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot analysis. We found that NaF in excess of 12 mg/kg caused the hepatic inflammatory responses, and the results showed that NaF activated the mitogen-activated protein kinases (MAPKs) signaling pathway by markedly increasing (p < 0.01 or p < 0.05) mRNA and protein levels of apoptosis signal-regulating kinase 1 (ASK1), mitogen-activated protein kinase kinases 1/2 (MEK1/2), extracellular signal-regulated protein kinases 1/2 (Erk1/2), mitogen-activated protein kinase kinases 4/7 (MEK4/7), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (p38) and mitogen-activated protein kinase kinases 3/6 (MEK3/6), and the nuclear factor-kappa B (NF-κB) signaling pathway by increasing (p < 0.01 or p < 0.05) the production of NF-κB and inhibitor of nuclear factor kappa-B kinase subunit beta (IKK-β) and reducing (p < 0.01 or p < 0.05) the production of the inhibitory kappa B (IκB). Thus, NaF that caused the hepatic inflammatory responses was characterized by increasing (p < 0.01 or p < 0.05) the production of pro-inflammatory mediators such as interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), monocyte chemotactic protein 1 (MCP-1), and cyclooxygenase-2 (COX-2) via the activation of MAPKs and NF-κB pathways, and by significantly inhibiting (p < 0.01 or p < 0.05) the production of anti-inflammatory mediators including interleukin-4 (IL-4) and transforming growth factor beta (TGF-β).
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Ping Kuang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Huan Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Qin Wei
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China.
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China.
- Key Laboratory of Agricultural Information Engineering of Sichuan Province, Sichuan Agriculture University, Ya'an, 625014, Sichuan, China.
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| |
Collapse
|
9
|
Vila MC, Novak JS, Benny Klimek M, Li N, Morales M, Fritz AG, Edwards K, Boehler JF, Hogarth MW, Kinder TB, Zhang A, Mazala D, Fiorillo AA, Douglas B, Chen YW, van den Anker J, Lu QL, Hathout Y, Hoffman EP, Partridge TA, Nagaraju K. Morpholino-induced exon skipping stimulates cell-mediated and humoral responses to dystrophin in mdx mice. J Pathol 2019; 248:339-351. [PMID: 30883742 DOI: 10.1002/path.5263] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/02/2019] [Accepted: 03/11/2019] [Indexed: 01/16/2023]
Abstract
Exon skipping is a promising genetic therapeutic strategy for restoring dystrophin expression in the treatment of Duchenne muscular dystrophy (DMD). The potential for newly synthesized dystrophin to trigger an immune response in DMD patients, however, is not well established. We have evaluated the effect of chronic phosphorodiamidate morpholino oligomer (PMO) treatment on skeletal muscle pathology and asked whether sustained dystrophin expression elicits a dystrophin-specific autoimmune response. Here, two independent cohorts of dystrophic mdx mice were treated chronically with either 800 mg/kg/month PMO for 6 months (n = 8) or 100 mg/kg/week PMO for 12 weeks (n = 11). We found that significant muscle inflammation persisted after exon skipping in skeletal muscle. Evaluation of humoral responses showed serum-circulating antibodies directed against de novo dystrophin in a subset of mice, as assessed both by Western blotting and immunofluorescent staining; however, no dystrophin-specific antibodies were observed in the control saline-treated mdx cohorts (n = 8) or in aged (12-month-old) mdx mice with expanded 'revertant' dystrophin-expressing fibers. Reactive antibodies recognized both full-length and truncated exon-skipped dystrophin isoforms in mouse skeletal muscle. We found more antigen-specific T-cell cytokine responses (e.g. IFN-g, IL-2) in dystrophin antibody-positive mice than in dystrophin antibody-negative mice. We also found expression of major histocompatibility complex class I on some of the dystrophin-expressing fibers along with CD8+ and perforin-positive T cells in the vicinity, suggesting an activation of cell-mediated damage had occurred in the muscle. Evaluation of complement membrane attack complex (MAC) deposition on the muscle fibers further revealed lower MAC deposition on muscle fibers of dystrophin antibody-negative mice than on those of dystrophin antibody-positive mice. Our results indicate that de novo dystrophin expression after exon skipping can trigger both cell-mediated and humoral immune responses in mdx mice. Our data highlights the need to further investigate the autoimmune response and its long-term consequences after exon-skipping therapy. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maria C Vila
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - James S Novak
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Genomics and Precision Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Pediatrics, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Ning Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Melissa Morales
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Alexander G Fritz
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Katie Edwards
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Jessica F Boehler
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Travis B Kinder
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Aiping Zhang
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Davi Mazala
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Alyson A Fiorillo
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Genomics and Precision Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Pediatrics, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Bonnie Douglas
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Yi-Wen Chen
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Genomics and Precision Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Pediatrics, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - John van den Anker
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Center for Translational Science, Children's National Health System, Washington, DC, USA
| | - Qi L Lu
- Department of Neurology, McColl-Lockwood Laboratory for Muscular Dystrophy Research, Neuromuscular/ALS Center, Carolinas Medical Center, Charlotte, NC, USA
| | - Yetrib Hathout
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Eric P Hoffman
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Terence A Partridge
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Genomics and Precision Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Pediatrics, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine, Children's National Health System, Washington, DC, USA.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
10
|
Jia Y, Liu L, Sheng C, Cheng Z, Cui L, Li M, Zhao Y, Shi T, Yau TO, Li F, Chen L. Increased Serum Levels of Cortisol and Inflammatory Cytokines in People With Depression. J Nerv Ment Dis 2019; 207:271-276. [PMID: 30844940 DOI: 10.1097/nmd.0000000000000957] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
This cross-sectional study aimed at measuring the correlation and association between serum levels of cortisol, inflammatory cytokines, and depression and to measure the detection accuracy of serum levels of cortisol in serum samples. In total, 89 male participants were recruited into this study from June 15, 2017, to September 31, 2017. The Hamilton Depression Rating Scale, Beck Anxiety Inventory, and Pittsburgh Sleep Quality Index were used to investigate the mental health status of the participants. Serum concentrations of cortisol and inflammatory cytokines were determined. The serum cortisol concentration, anxiety level, and sleep quality were included in the final logistic regression model. Serum cortisol was able to accurately distinguish between patients with depression and those without depression. There was a significant positive correlation between serum cortisol levels and Hamilton Depression Rating Scale scores.
Collapse
Affiliation(s)
| | - Linlin Liu
- School of Public Health, Jilin University
| | - Chuqiao Sheng
- Department of Pediatric Intensive Care Unit, The First Hospital of Jilin University
| | - Zhaohua Cheng
- Hepatobiliary and Pancreatic Surgery Department, The Second Hospital of Jilin University
| | | | - Min Li
- Invasive Technology Department, The First Hospital of Jilin University
| | - Yawei Zhao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Tongfei Shi
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Tung On Yau
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | | |
Collapse
|
11
|
Vandamme C, Adjali O, Mingozzi F. Unraveling the Complex Story of Immune Responses to AAV Vectors Trial After Trial. Hum Gene Ther 2018; 28:1061-1074. [PMID: 28835127 PMCID: PMC5649404 DOI: 10.1089/hum.2017.150] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Over the past decade, vectors derived from adeno-associated virus (AAV) have established themselves as a powerful tool for in vivo gene transfer, allowing long-lasting and safe transgene expression in a variety of human tissues. Nevertheless, clinical trials demonstrated how B and T cell immune responses directed against the AAV capsid, likely arising after natural infection with wild-type AAV, might potentially impact gene transfer safety and efficacy in patients. Seroprevalence studies have evidenced that most individuals carry anti-AAV neutralizing antibodies that can inhibit recombinant AAV transduction of target cells following in vivo administration of vector particles. Likewise, liver- and muscle-directed clinical trials have shown that capsid-reactive memory CD8+ T cells could be reactivated and expanded upon presentation of capsid-derived antigens on transduced cells, potentially leading to loss of transgene expression and immune-mediated toxicities. In celebration of the 25th anniversary of the European Society of Gene and Cell Therapy, this review article summarizes progress made during the past decade in understanding and modulating AAV vector immunogenicity. While the knowledge generated has contributed to yield impressive clinical results, several important questions remain unanswered, making the study of immune responses to AAV a priority for the field of in vivo transfer.
Collapse
Affiliation(s)
- Céline Vandamme
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
- Correspondence: Dr. Céline Vandamme, Faculty of Health Sciences, Department of Clinical Microbiology, Yliopistonranta 1, 70210 Kuopio, Finland. E-mail:; Dr. Oumeya Adjali, IRS2 Nantes Biotech, 22, bd Bénoni Goullin, 44200 Nantes, France. E-mail:; Dr. Federico Mingozzi, 1 rue de l'Internationale, 91000 Evry, France. E-mail:
| | - Oumeya Adjali
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
- Correspondence: Dr. Céline Vandamme, Faculty of Health Sciences, Department of Clinical Microbiology, Yliopistonranta 1, 70210 Kuopio, Finland. E-mail:; Dr. Oumeya Adjali, IRS2 Nantes Biotech, 22, bd Bénoni Goullin, 44200 Nantes, France. E-mail:; Dr. Federico Mingozzi, 1 rue de l'Internationale, 91000 Evry, France. E-mail:
| | - Federico Mingozzi
- Genethon and IMSERM U951, Evry, France
- University Pierre and Marie Curie and INSERM U974, Paris, France
- Correspondence: Dr. Céline Vandamme, Faculty of Health Sciences, Department of Clinical Microbiology, Yliopistonranta 1, 70210 Kuopio, Finland. E-mail:; Dr. Oumeya Adjali, IRS2 Nantes Biotech, 22, bd Bénoni Goullin, 44200 Nantes, France. E-mail:; Dr. Federico Mingozzi, 1 rue de l'Internationale, 91000 Evry, France. E-mail:
| |
Collapse
|
12
|
Julien L, Chassagne J, Peccate C, Lorain S, Piétri-Rouxel F, Danos O, Benkhelifa-Ziyyat S. RFX1 and RFX3 Transcription Factors Interact with the D Sequence of Adeno-Associated Virus Inverted Terminal Repeat and Regulate AAV Transduction. Sci Rep 2018; 8:210. [PMID: 29317724 PMCID: PMC5760533 DOI: 10.1038/s41598-017-18604-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/13/2017] [Indexed: 12/31/2022] Open
Abstract
Adeno-associated virus (AAV) transduction efficiency depends on the way in which cellular proteins process viral genomes in the nucleus. In this study, we have investigated the binding of nuclear proteins to the double stranded D (dsD) sequence of the AAV inverted terminal repeat (ITRs) by electromobility shift assay. We present here several lines of evidence that transcription factors belonging to the RFX protein family bind specifically and selectively to AAV2 and AAV1 dsD sequences. Using supershift experiments, we characterize complexes containing RFX1 homodimers and RFX1/RFX3 heterodimers. Following transduction of HEK-293 cells, the AAV genome can be pulled-down by RFX1 and RFX3 antibodies. Moreover, our data suggest that RFX proteins which interact with transcriptional enhancers of several mammalian DNA viruses, can act as regulators of AAV mediated transgene expression.
Collapse
Affiliation(s)
- Laura Julien
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Julie Chassagne
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Cécile Peccate
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Stéphanie Lorain
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - France Piétri-Rouxel
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Olivier Danos
- REGENXBIO, 9600 Blackwell Rd, Rockville, MD, 20850, USA
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France.
| |
Collapse
|
13
|
Selot R, Arumugam S, Mary B, Cheemadan S, Jayandharan GR. Optimized AAV rh.10 Vectors That Partially Evade Neutralizing Antibodies during Hepatic Gene Transfer. Front Pharmacol 2017; 8:441. [PMID: 28769791 PMCID: PMC5511854 DOI: 10.3389/fphar.2017.00441] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
Of the 12 common serotypes used for gene delivery applications, Adeno-associated virus (AAV)rh.10 serotype has shown sustained hepatic transduction and has the lowest seropositivity in humans. We have evaluated if further modifications to AAVrh.10 at its phosphodegron like regions or predicted immunogenic epitopes could improve its hepatic gene transfer and immune evasion potential. Mutant AAVrh.10 vectors were generated by site directed mutagenesis of the predicted targets. These mutant vectors were first tested for their transduction efficiency in HeLa and HEK293T cells. The optimal vector was further evaluated for their cellular uptake, entry, and intracellular trafficking by quantitative PCR and time-lapse confocal microscopy. To evaluate their potential during hepatic gene therapy, C57BL/6 mice were administered with wild-type or optimal mutant AAVrh.10 and the luciferase transgene expression was documented by serial bioluminescence imaging at 14, 30, 45, and 72 days post-gene transfer. Their hepatic transduction was further verified by a quantitative PCR analysis of AAV copy number in the liver tissue. The optimal AAVrh.10 vector was further evaluated for their immune escape potential, in animals pre-immunized with human intravenous immunoglobulin. Our results demonstrate that a modified AAVrh.10 S671A vector had enhanced cellular entry (3.6 fold), migrate rapidly to the perinuclear region (1 vs. >2 h for wild type vectors) in vitro, which further translates to modest increase in hepatic gene transfer efficiency in vivo. More importantly, the mutant AAVrh.10 vector was able to partially evade neutralizing antibodies (~27-64 fold) in pre-immunized animals. The development of an AAV vector system that can escape the circulating neutralizing antibodies in the host will substantially widen the scope of gene therapy applications in humans.
Collapse
Affiliation(s)
- Ruchita Selot
- Department of Biological Sciences and Bioengineering, Indian Institute of TechnologyKanpur, India
| | - Sathyathithan Arumugam
- Department of Biological Sciences and Bioengineering, Indian Institute of TechnologyKanpur, India
| | - Bertin Mary
- Department of Biological Sciences and Bioengineering, Indian Institute of TechnologyKanpur, India
| | - Sabna Cheemadan
- Department of Hematology and Centre for Stem Cell Research (CSCR), Christian Medical CollegeVellore, India
| | - Giridhara R. Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of TechnologyKanpur, India
- Department of Hematology and Centre for Stem Cell Research (CSCR), Christian Medical CollegeVellore, India
| |
Collapse
|
14
|
Baruteau J, Waddington SN, Alexander IE, Gissen P. Gene therapy for monogenic liver diseases: clinical successes, current challenges and future prospects. J Inherit Metab Dis 2017; 40:497-517. [PMID: 28567541 PMCID: PMC5500673 DOI: 10.1007/s10545-017-0053-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/08/2023]
Abstract
Over the last decade, pioneering liver-directed gene therapy trials for haemophilia B have achieved sustained clinical improvement after a single systemic injection of adeno-associated virus (AAV) derived vectors encoding the human factor IX cDNA. These trials demonstrate the potential of AAV technology to provide long-lasting clinical benefit in the treatment of monogenic liver disorders. Indeed, with more than ten ongoing or planned clinical trials for haemophilia A and B and dozens of trials planned for other inherited genetic/metabolic liver diseases, clinical translation is expanding rapidly. Gene therapy is likely to become an option for routine care of a subset of severe inherited genetic/metabolic liver diseases in the relatively near term. In this review, we aim to summarise the milestones in the development of gene therapy, present the different vector tools and their clinical applications for liver-directed gene therapy. AAV-derived vectors are emerging as the leading candidates for clinical translation of gene delivery to the liver. Therefore, we focus on clinical applications of AAV vectors in providing the most recent update on clinical outcomes of completed and ongoing gene therapy trials and comment on the current challenges that the field is facing for large-scale clinical translation. There is clearly an urgent need for more efficient therapies in many severe monogenic liver disorders, which will require careful risk-benefit analysis for each indication, especially in paediatrics.
Collapse
Affiliation(s)
- Julien Baruteau
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK.
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ian E Alexander
- Gene Therapy Research Unit, The Children's Hospital at Westmead and Children's Medical Research Institute, Westmead, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia
| | - Paul Gissen
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
15
|
Plasmacytoid and conventional dendritic cells cooperate in crosspriming AAV capsid-specific CD8 + T cells. Blood 2017; 129:3184-3195. [PMID: 28468798 DOI: 10.1182/blood-2016-11-751040] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/01/2017] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) is a replication-deficient parvovirus that is extensively used as a gene therapy vector. CD8+ T-cell responses against the AAV capsid protein can, however, affect therapeutic efficacy. Little is known about the in vivo mechanism that leads to the crosspriming of CD8+ T cells against the input viral capsid antigen. In this study, we report that the Toll-like receptor 9 (TLR9)-MyD88 pattern-recognition receptor pathway is uniquely capable of initiating this response. By contrast, the absence of TLR2, STING, or the addition of TLR4 agonist has no effect. Surprisingly, both conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) are required for the crosspriming of capsid-specific CD8+ T cells, whereas other antigen-presenting cells are not involved. TLR9 signaling is specifically essential in pDCs but not in cDCs, indicating that sensing of the viral genome by pDCs activates cDCs in trans to cross-present capsid antigen during CD8+ T-cell activation. Cross-presentation and crosspriming depend not only on TLR9, but also on interferon type I signaling, and both mechanisms can be inhibited by administering specific molecules to prevent induction of capsid-specific CD8+ T cells. Thus, these outcomes directly point to therapeutic interventions and demonstrate that innate immune blockade can eliminate unwanted immune responses in gene therapy.
Collapse
|
16
|
Brempelis KJ, Yuen SY, Schwarz N, Mohar I, Crispe IN. Central role of the TIR-domain-containing adaptor-inducing interferon-β (TRIF) adaptor protein in murine sterile liver injury. Hepatology 2017; 65:1336-1351. [PMID: 28120431 PMCID: PMC5391172 DOI: 10.1002/hep.29078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 01/11/2017] [Accepted: 01/18/2017] [Indexed: 12/31/2022]
Abstract
UNLABELLED Multiple pathways drive the sterile injury response in the liver; however, it is unclear how the type of cells injured or the mechanism of injury activates these pathways. Here, we use a model of selective hepatocyte death to investigate sterile liver injury. In this model, the TIR-domain-containing adaptor-inducing interferon-β (TRIF) was a central mediator of the resulting intrahepatic inflammatory response that was independent of both upstream Toll-like receptor (TLR) 4 signaling and downstream type I interferon (IFN) signaling. TRIF was required for induction of interleukin (IL)-10, IL-6, and IL-1β cytokines. Conversely, although induction of C-C motif chemokine ligand (CCL) 2 and C-X-C motif chemokine ligand (CXCL) 1 chemokines and up-regulation of chemokine (Ccl2, Ccl7, Cxcl1, Cxcl2, and Cxcl10) and cell-adhesion (intracellular adhesion molecule 1 and vascular cell adhesion molecule 1) genes involved in myeloid cell recruitment was reduced in a majority of TRIF-/- mice, a subset of TRIF-/- mice showed breakthrough inflammation and the ability to induce these genes and proteins, indicating that redundant pathways exist to respond to hepatocyte death. Furthermore, we found that hepatocytes themselves were the main responders to hepatocyte death, increasing transcription of genes involved in myeloid cell recruitment more than either liver sinusoidal endothelial cells or Kupffer cells. CONCLUSION Our studies define a TRIF-dependent, TLR4- and type I IFN-independent pathway of sterile liver injury in which hepatocytes are both the targets of damage and the principal responding cell type. (Hepatology 2017;65:1336-1351).
Collapse
Affiliation(s)
- Katherine J. Brempelis
- Department of Global Health, University of Washington, Seattle, WA 98195, USA,Department of Pathology, University of Washington, Seattle, WA 98195, USA,Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Sebastian Y. Yuen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Nicole Schwarz
- Department of Pathology, University of Washington, Seattle, WA 98195, USA,Institute of Immunology, University of Medical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Isaac Mohar
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Ian N. Crispe
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Srivastava A. Adeno-Associated Virus: The Naturally Occurring Virus Versus the Recombinant Vector. Hum Gene Ther 2016; 27:1-6. [PMID: 26784640 DOI: 10.1089/hum.2015.29017.asr] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics and Department of Molecular Genetics and Microbiology, Powell Gene Therapy Center; Genetics Institute; Shands Cancer Center; University of Florida College of Medicine , Gainesville, Florida
| |
Collapse
|
18
|
Sauter MM, Brandt CR. Primate neural retina upregulates IL-6 and IL-10 in response to a herpes simplex vector suggesting the presence of a pro-/anti-inflammatory axis. Exp Eye Res 2016; 148:12-23. [PMID: 27170050 DOI: 10.1016/j.exer.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/11/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
Abstract
Injection of herpes simplex virus vectors into the vitreous of primate eyes induces an acute, transient uveitis. The purpose of this study was to characterize innate immune responses of macaque neural retina tissue to the herpes simplex virus type 1-based gene delivery vector hrR3. PCR array analysis demonstrated the induction of the pro-inflammatory cytokine IL-6, as well as the anti-inflammatory cytokine IL-10, following hrR3 exposure. Secretion of IL-6 was detected by ELISA and cone photoreceptors and Muller cells were the predominant IL-6 positive cell types. RNA in situ hybridization confirmed that IL-6 was expressed in photoreceptor and Muller cells. The IL-10 positive cells in the inner nuclear layer were identified as amacrine cells by immunofluorescence staining with calretinin antibody. hrR3 challenge resulted in activation of NFκB (p65) in Muller glial cells, but not in cone photoreceptors, suggesting a novel regulatory mechanism for IL-6 expression in cone cells. hrR3 replication was not required for IL-6 induction or NFκB (p65) activation. These data suggest a pro-inflammatory (IL-6)/anti-inflammatory (IL-10) axis exists in neural retina and the severity of acute posterior uveitis may be determined by this interaction. Further studies are needed to identify the trigger for IL-6 and IL-10 induction and the mechanism of IL-6 induction in cone cells.
Collapse
Affiliation(s)
- Monica M Sauter
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Curtis R Brandt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
19
|
Reprogramming Immune Response With Capsid-Optimized AAV6 Vectors for Immunotherapy of Cancer. J Immunother 2016; 38:292-8. [PMID: 26261893 DOI: 10.1097/cji.0000000000000093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the current studies we generated novel capsid-optimized adeno-associated virus (AAV) serotype 6 (AAV6) vectors expressing a tumor-associated antigen, and assessed their ability to activate a protective T-cell response in an animal model. First, we showed that specific mutations in the AAV6 capsid increase the transduction efficiency of these vectors in mouse bone marrow-derived dendritic cells in vitro for approximately 5-fold compared with the wild-type (WT) AAV6 vectors. Next, we evaluated the ability of the mutant AAV6 vectors to initiate specific T-cell clone proliferation in vivo. Our data indicate that the intramuscular administration of AAV6-S663V+T492V vectors expressing ovalbumin (OVA) led to a strong activation (approximately 9%) of specific T cells in peripheral blood compared with AAV6-WT treated animals (<1%). These OVA-specific T cells have a superior killing ability against mouse prostate cancer cell line RM1 stably expressing the OVA antigen when propagated in vitro. Finally, we evaluated the ability of capsid-optimized AAV6-S663V+T492V vectors to initiate a protective anticancer immune response in vivo. Our results document the suppression of subcutaneous tumor growth in animals immunized with AAV6-S663V+T492V vectors expressing prostatic acid phosphatase (PAP) for approximately 4 weeks in comparison with 1 week and 2 weeks for the negative controls, AAV6-EGFP, and AAV6-WT-PAP treated mice, respectively. These studies suggest that successful inhibition of tumor growth in an animal model would set the stage for potential clinical application of the capsid-optimized AAV6-S663V+T492V vectors.
Collapse
|
20
|
Yu X, Zhao Q, Zhang H, Fan C, Zhang X, Xie Q, Xu C, Liu Y, Wu X, Han Q, Zhang H. Gambogenic acid inhibits LPS-simulated inflammatory response by suppressing NF-κB and MAPK in macrophages. Acta Biochim Biophys Sin (Shanghai) 2016; 48:454-61. [PMID: 27025602 DOI: 10.1093/abbs/gmw021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/26/2016] [Indexed: 01/21/2023] Open
Abstract
Inflammation is a response of body tissues to injury and infection. Compounds that can inhibit inflammation have been shown to have potential therapeutic clinical application. Gambogenic acid (GEA) has potent antitumor and anti-inflammatory activities. Herein, the molecular mechanisms of GEA's anti-inflammatory effect were investigated in lipopolysaccharide (LPS)-stimulated macrophage cells. The results showed that pretreatment with GEA could markedly inhibit interleukin (IL)-1α, IL-1β, tumor necrosis factor-α, IFN-β, IL-12b, and IL-23a production in a dose-dependent manner in LPS-induced model. Furthermore, this drug significantly reduced the release of nitric oxide (NO), and impaired the protein level of inducible NO synthase and the cyclooxygenase 2. The finding also showed that the effect of GEA may be related to the suppression of the nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathway. These results indicate that GEA could suppress LPS-simulated inflammatory response partially by attenuating NO synthesis and NF-κB and MAPK activation, suggesting that it may become a potent therapeutic agent for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xianjun Yu
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qun Zhao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haiwei Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cunxian Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xixi Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qun Xie
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chengxian Xu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yongbo Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoxia Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Quanbin Han
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Haibing Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
21
|
Microglia-specific targeting by novel capsid-modified AAV6 vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16026. [PMID: 27308302 PMCID: PMC4909093 DOI: 10.1038/mtm.2016.26] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/14/2023]
Abstract
Recombinant adeno-associated viruses (rAAV) have been widely used in gene therapy
applications for central nervous system diseases. Though rAAV can efficiently target
neurons and astrocytes in mouse brains, microglia, the immune cells of the brain, are
refractile to rAAV. To identify AAV capsids with microglia-specific transduction
properties, we initially screened the most commonly used serotypes, AAV1–9 and
rh10, on primary mouse microglia cultures. While these capsids were not permissive, we
then tested the microglial targeting properties of a newly characterized set of modified
rAAV6 capsid variants with high tropism for monocytes. Indeed, these newly characterized
rAAV6 capsid variants, specially a triply mutated Y731F/Y705F/T492V form, carrying a
self-complementary genome and microglia-specific promoters (F4/80 or CD68) could
efficiently and selectively transduce microglia in vitro. Delivery of these
constructs in mice brains resulted in microglia-specific expression of green fluorescent
protein, albeit at modest levels. We further show that CD68 promoter–driven
expression of the inflammatory cytokine, interleukin-6, using this capsid variant leads to
increased astrogliosis in the brains of wild-type mice. Our study describes the first
instance of AAV-targeted microglial gene expression leading to functional modulation of
the innate immune system in mice brains. This provides the rationale for utilizing these
unique capsid/promoter combinations for microglia-specific gene targeting for modeling or
functional studies.
Collapse
|
22
|
Doerfler PA, Todd AG, Clément N, Falk DJ, Nayak S, Herzog RW, Byrne BJ. Copackaged AAV9 Vectors Promote Simultaneous Immune Tolerance and Phenotypic Correction of Pompe Disease. Hum Gene Ther 2016; 27:43-59. [PMID: 26603344 PMCID: PMC4741206 DOI: 10.1089/hum.2015.103] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/28/2015] [Indexed: 12/24/2022] Open
Abstract
Pompe disease is a progressive neuromuscular disorder caused by lysosomal accumulation of glycogen from a deficiency in acid alpha-glucosidase (GAA). Replacement of the missing enzyme is available by repeated protein infusions; however, efficacy is limited by immune response and inability to restore enzymatic function in the central nervous system. An alternative therapeutic option is adeno-associated virus (AAV)-mediated gene therapy, which results in widespread gene transfer and prolonged transgene expression. Both enzyme replacement therapy (ERT) and gene therapy can elicit anti-GAA immune reactions that dampen their effectiveness and pose life-threatening risks to patient safety. To modulate the immune responses related to gene therapy, we show that a human codon-optimized GAA (coGAA) driven by a liver-specific promoter (LSP) using AAV9 is capable of promoting immune tolerance in a Gaa(-/-) mouse model. Copackaging AAV9-LSP-coGAA with the tissue-restricted desmin promoter (AAV9-DES-coGAA) demonstrates the necessary cell autonomous expression in cardiac muscle, skeletal muscle, peripheral nerve, and the spinal cord. Simultaneous high-level expression in liver led to the expansion of GAA-specific regulatory T-cells (Tregs) and induction of immune tolerance. Transfer of Tregs into naïve recipients prevented pathogenic allergic reactions after repeated ERT challenges. Copackaged AAV9 also attenuated preexisting humoral and cellular immune responses, which enhanced the biochemical correction. Our data present a therapeutic design in which simultaneous administration of two copackaged AAV constructs may provide therapeutic benefit and resolve immune reactions in the treatment of multisystem disorders.
Collapse
Affiliation(s)
- Phillip A. Doerfler
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - Adrian G. Todd
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - Nathalie Clément
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - Darin J. Falk
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - Sushrusha Nayak
- Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Stockholm, Sweden
| | - Roland W. Herzog
- Division of Cellular & Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Barry J. Byrne
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| |
Collapse
|
23
|
Hareendran S, Ramakrishna B, Jayandharan GR. Synergistic inhibition of PARP-1 and NF-κB signaling downregulates immune response against recombinant AAV2 vectors during hepatic gene therapy. Eur J Immunol 2015; 46:154-66. [PMID: 26443873 DOI: 10.1002/eji.201545867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/04/2015] [Accepted: 09/30/2015] [Indexed: 02/02/2023]
Abstract
Host immune response remains a key obstacle to widespread application of adeno-associated virus (AAV) based gene therapy. Thus, targeted inhibition of the signaling pathways that trigger such immune responses will be beneficial. Previous studies have reported that DNA damage response proteins such as poly(ADP-ribose) polymerase-1 (PARP-1) negatively affect the integration of AAV in the host genome. However, the role of PARP-1 in regulating AAV transduction and the immune response against these vectors has not been elucidated. In this study, we demonstrate that repression of PARP-1 improves the transduction of single-stranded AAV vectors both in vitro (∼174%) and in vivo (two- to 3.4-fold). Inhibition of PARP-1, also significantly downregulated the expression of several proinflammatory and cytokine markers such as TLRs, ILs, NF-κB subunit proteins associated with the host innate response against self-complementary AAV2 vectors. The suppression of the inflammatory response targeted against these vectors was more effective upon combined inhibition of PARP-1 and NF-κB signaling. This strategy also effectively attenuated the AAV capsid-specific cytotoxic T-cell response, with minimal effect on vector transduction, as demonstrated in normal C57BL/6 and hemophilia B mice. These data suggest that targeting specific host cellular proteins could be useful to attenuate the immune barriers to AAV-mediated gene therapy.
Collapse
Affiliation(s)
- Sangeetha Hareendran
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Banumathi Ramakrishna
- Department of General Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Giridhara R Jayandharan
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India.,Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India.,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh, India
| |
Collapse
|
24
|
Tu M, Liu F, Chen S, Wang M, Cheng A. Role of capsid proteins in parvoviruses infection. Virol J 2015; 12:114. [PMID: 26239432 PMCID: PMC4524367 DOI: 10.1186/s12985-015-0344-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/15/2015] [Indexed: 11/30/2022] Open
Abstract
The parvoviruses are widely spread in many species and are among the smallest DNA animal viruses. The parvovirus is composed of a single strand molecule of DNA wrapped into an icosahedral capsid. In a viral infection, the massy capsid participates in the entire viral infection process, which is summarized in this review. The capsid protein VP1 is primarily responsible for the infectivity of the virus, and the nuclear localization signal (NLS) of the VP1 serves as a guide to assist the viral genome in locating the nucleus. The dominant protein VP2 provides an “anti-receptor”, which interacts with the cellular receptor and leads to the further internalization of virus, and, the N-terminal of VP2 also cooperates with the VP1 to prompt the process of nucleus translocation. Additionally, a cleavage protein VP3 is a part of the capsid, which exists only in several members of the parvovirus family; however, the function of this cleavage protein remains to be fully determined. Parvoviruses can suffer from the extreme environmental conditions such as low pH, or even escape from the recognition of pattern recognition receptors (PRRs), due to the protection of the stable capsid, which is thought to be an immune escape mechanism. The applications of the capsid proteins to the screening and the treatment of diseases are also discussed. The processes of viral infection should be noted, because understanding the virus-host interactions will contribute to the development of therapeutic vaccines.
Collapse
Affiliation(s)
- Mengyu Tu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China.
| | - Fei Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China.
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China.
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China.
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang District, Chengdu City, Sichuan Province, 611130, China.
| |
Collapse
|
25
|
Ling C, Wang Y, Lu Y, Wang L, Jayandharan GR, Aslanidi GV, Li B, Cheng B, Ma W, Lentz T, Ling C, Xiao X, Samulski RJ, Muzyczka N, Srivastava A. The Adeno-Associated Virus Genome Packaging Puzzle. J Mol Genet Med 2015; 9. [PMID: 26949410 DOI: 10.4172/1747-0862.1000175] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Yuan Wang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA; Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Lu
- Department of Orthopedics & Rehabilitative Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lina Wang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA; Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - George V Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Baozheng Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Binbin Cheng
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenqin Ma
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Thomas Lentz
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Changquan Ling
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiao Xiao
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Molecular Pharmaceutics, University of North Carolina School of Pharmacy, Chapel Hill, NC, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas Muzyczka
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA; Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; Genetics Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA; Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA; Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; Genetics Institute, University of Florida College of Medicine, Gainesville, FL, USA; Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
26
|
Rogers GL, Suzuki M, Zolotukhin I, Markusic DM, Morel LM, Lee B, Ertl HCJ, Herzog RW. Unique Roles of TLR9- and MyD88-Dependent and -Independent Pathways in Adaptive Immune Responses to AAV-Mediated Gene Transfer. J Innate Immun 2015; 7:302-14. [PMID: 25612611 DOI: 10.1159/000369273] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022] Open
Abstract
The immune system represents a significant barrier to successful gene therapy with adeno-associated viral (AAV) vectors. In particular, adaptive immune responses to the viral capsid or the transgene product are of concern. The sensing of AAV by toll-like receptors (TLRs) TLR2 and TLR9 has been suggested to play a role in innate immunity to the virus and may also shape subsequent adaptive immune responses. Here, we investigated the functions of TLR2, TLR9 and the downstream signaling adaptor MyD88 in antibody and CD8+ T-cell responses. Antibody formation against the transgene product occurred largely independently of TLR signaling following gene transfer with AAV1 or AAV2 vectors, whereas loss of signaling through the TLR9-MyD88 pathway substantially reduced CD8+ T-cell responses. In contrast, MyD88 (but neither of the TLRs) regulated antibody responses to capsid. B cell-intrinsic MyD88 was required for the formation of anti-capsid IgG2c independently of vector serotype or route of administration. However, MyD88(-/-) mice instead produced anti-capsid IgG1 that emerged with delayed kinetics but nonetheless completely prevented in vivo readministration. We conclude that there are distinct roles for TLR9 and MyD88 in promoting adaptive immune responses to AAV-mediated gene transfer and that there are redundant MyD88-dependent and MyD88-independent mechanisms that stimulate neutralizing antibody formation against AAV.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Fla., USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Hemophilia is an X-linked inherited bleeding disorder consisting of two classifications, hemophilia A and hemophilia B, depending on the underlying mutation. Although the disease is currently treatable with intravenous delivery of replacement recombinant clotting factor, this approach represents a significant cost both monetarily and in terms of quality of life. Gene therapy is an attractive alternative approach to the treatment of hemophilia that would ideally provide life-long correction of clotting activity with a single injection. In this review, we will discuss the multitude of approaches that have been explored for the treatment of both hemophilia A and B, including both in vivo and ex vivo approaches with viral and nonviral delivery vectors.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| | - Roland W Herzog
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| |
Collapse
|
28
|
Hong SH, Park SJ, Lee S, Cho CS, Cho MH. Aerosol gene delivery using viral vectors and cationic carriers forin vivolung cancer therapy. Expert Opin Drug Deliv 2014; 12:977-91. [DOI: 10.1517/17425247.2015.986454] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
29
|
Qin JD, Cao ZH, Li XF, Kang XL, Xue Y, Li YL, Zhang D, Liu XY, Xue YZ. Effect of ammonium pyrrolidine dithiocarbamate (PDTC) on NF-κB activation and CYP2E1 content of rats with immunological liver injury. PHARMACEUTICAL BIOLOGY 2014; 52:1460-1466. [PMID: 24963944 DOI: 10.3109/13880209.2014.898075] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT Ammonium pyrrolidine dithiocarbamate (PDTC) is a potent inhibitor of nuclear factor-κB (NF-κB). Recent studies have shown that NF-κB plays an essential role in the regulation of genes whose products are involved in the pathogenesis of immunological liver injury. OBJECTIVE To study the function of NF-κB in immunological liver injury of rat model and its effect on CYP2E1 content and metabolic activity. MATERIALS AND METHODS The present study investigated the effect of passivating NF-κB activation on CYP2E1 using Bacillus calmette Guérin (BCG)-induced immunological liver injury in Sprague-Dawley rats measured in terms of enzyme levels. The degree of hepatic injury of rats was measured by using biochemical parameters, hepatic tissue pathological changes, and physiological parameters. Protein localization of liver NF-κB was detected by immunohistochemical assay. Western blot analysis was used to detect the protein expression of NF-κB, IκBα, iNOS, and CYP2E1. The content of CYP2E1 of homogenate in the rat liver was detected by ELISA assay and the enzyme kinetics of CYP2E1 probe drug chlorzoxazone was evaluated by high-performance liquid chromatography (HPLC) assay. RESULTS The results showed that BCG-pretreatment (125 mg/kg) significantly (p < 0.01) increased the weight of liver and spleen (increased by 70% and 248%, respectively), serum levels of alanine transarninase (ALT) and aspartate aminotransferase (AST) (increased by 200% and 75.8%, respectively), the expression of NF-κB and iNOS (increased by 228% and 303%, respectively), and decreased CYP2E1 content and metabolic activity (p < 0.05). Administration of PDTC (50, 100, and 200 mg/kg) reversed above hepatic injury stimulated by BCG in vivo. Moreover, PDTC (ED50: 76 mg/kg) dose dependently inhibited down-regulation of CYP2E1 (p < 0.05). CONCLUSION Passivation of NF-κB can inhibit the down-regulation of CYP2E1 and iNOS to induce in rat liver tissue with immunological liver injury; NF-κB may be involved in the CYP2E1 regulation through iNOS.
Collapse
Affiliation(s)
- Jin-Dong Qin
- Department of Pharmacology, Baotou Medical College , Baotou , China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Enhanced transgene expression from recombinant single-stranded D-sequence-substituted adeno-associated virus vectors in human cell lines in vitro and in murine hepatocytes in vivo. J Virol 2014; 89:952-61. [PMID: 25355884 DOI: 10.1128/jvi.02581-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED We have previously reported that the removal of a 20-nucleotide sequence, termed the D sequence, from both ends of the inverted terminal repeats (ITRs) in the adeno-associated virus serotype 2 (AAV2) genome significantly impairs rescue, replication, and encapsidation of the viral genomes (X. S. Wang, S. Ponnazhagan, and A. Srivastava, J Mol Biol 250:573-580, 1995; X. S. Wang, S. Ponnazhagan, and A. Srivastava, J Virol 70:1668-1677, 1996). Here we describe that replacement of only one D sequence in either ITR restores each of these functions, but DNA strands of only single polarity are encapsidated in mature progeny virions. Since most commonly used recombinant AAV vectors contain a single-stranded DNA (ssDNA), which is transcriptionally inactive, efficient transgene expression from AAV vectors is dependent upon viral second-strand DNA synthesis. We have also identified a transcription suppressor sequence in one of the D sequences, which shares homology with the binding site for the cellular NF-κB-repressing factor (NRF). The removal of this D sequence from, and replacement with a sequence containing putative binding sites for transcription factors in, single-stranded AAV (ssAAV) vectors significantly augments transgene expression both in human cell lines in vitro and in murine hepatocytes in vivo. The development of these genome-modified ssAAV vectors has implications not only for the basic biology of AAV but also for the optimal use of these vectors in human gene therapy. IMPORTANCE The results of the studies described here not only have provided novel insights into some of the critical steps in the life cycle of a human virus, the adeno-associated virus (AAV), that causes no known disease but have also led to the development of novel recombinant AAV vectors which are more efficient in allowing increased levels of gene expression. Thus, these studies have significant implications for the potential use of these novel AAV vectors in human gene therapy.
Collapse
|
31
|
Doerfler PA, Byrne BJ, Clément N. Copackaging of multiple adeno-associated viral vectors in a single production step. Hum Gene Ther Methods 2014; 25:269-76. [PMID: 25143183 PMCID: PMC4346231 DOI: 10.1089/hgtb.2014.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/14/2014] [Indexed: 12/13/2022] Open
Abstract
Limiting factors in large preclinical and clinical studies utilizing adeno-associated virus (AAV) for gene therapy are focused on the restrictive packaging capacity, the overall yields, and the versatility of the production methods for single AAV vector production. Furthermore, applications where multiple vectors are needed to provide long expression cassettes, whether because of long cDNA sequences or the need of different regulatory elements, require that each vector be packaged and characterized separately, directly affecting labor and cost associated with such manufacturing strategies. To overcome these limitations, we propose a novel method of vector production that allows for the packaging of multiple expression cassettes in a single transfection step. Here we combined two expression cassettes in predetermined ratios before transfection and empirically demonstrate that the output vector recapitulates the predicted ratios. Titration by quantitative polymerase chain reaction of AAV vector genome copies using shared or unique genetic elements allowed for delineation of the individual vector contribution to the total preparation that showed the predicted differential packaging outcomes. By copackaging green fluorescent protein (GFP) and mCherry constructs, we demonstrate that both vector genome and infectious titers reiterated the ratios utilized to produce the constructs by transfection. Copackaged therapeutic constructs that only differ in transcriptional elements produced a heterogeneous vector population of both constructs in the predefined ratios. This study shows feasibility and reproducibility of a method that allows for two constructs, differing in either transgene or transcription elements, to be efficiently copackaged and characterized simultaneously, reducing cost of manufacturing and release testing.
Collapse
Affiliation(s)
- Phillip A Doerfler
- Powell Gene Therapy Center, Department of Pediatrics, College of Medicine, University of Florida , Gainesville, FL 32610
| | | | | |
Collapse
|
32
|
Sen D, Balakrishnan B, Jayandharan GR. Cellular unfolded protein response against viruses used in gene therapy. Front Microbiol 2014; 5:250. [PMID: 24904562 PMCID: PMC4033601 DOI: 10.3389/fmicb.2014.00250] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/07/2014] [Indexed: 01/21/2023] Open
Abstract
Viruses are excellent vehicles for gene therapy due to their natural ability to infect and deliver the cargo to specific tissues with high efficiency. Although such vectors are usually "gutted" and are replication defective, they are subjected to clearance by the host cells by immune recognition and destruction. Unfolded protein response (UPR) is a naturally evolved cyto-protective signaling pathway which is triggered due to endoplasmic reticulum (ER) stress caused by accumulation of unfolded/misfolded proteins in its lumen. The UPR signaling consists of three signaling pathways, namely PKR-like ER kinase, activating transcription factor 6, and inositol-requiring protein-1. Once activated, UPR triggers the production of ER molecular chaperones and stress response proteins to help reduce the protein load within the ER. This occurs by degradation of the misfolded proteins and ensues in the arrest of protein translation machinery. If the burden of protein load in ER is beyond its processing capacity, UPR can activate pro-apoptotic pathways or autophagy leading to cell death. Viruses are naturally evolved in hijacking the host cellular translation machinery to generate a large amount of proteins. This phenomenon disrupts ER homeostasis and leads to ER stress. Alternatively, in the case of gutted vectors used in gene therapy, the excess load of recombinant vectors administered and encountered by the cell can trigger UPR. Thus, in the context of gene therapy, UPR becomes a major roadblock that can potentially trigger inflammatory responses against the vectors and reduce the efficiency of gene transfer.
Collapse
Affiliation(s)
- Dwaipayan Sen
- Department of Hematology, Christian Medical College Vellore, India
| | | | - Giridhara R Jayandharan
- Department of Hematology, Christian Medical College Vellore, India ; Centre for Stem Cell Research, Christian Medical College Vellore, India
| |
Collapse
|
33
|
Wang LN, Wang Y, Lu Y, Yin ZF, Zhang YH, Aslanidi GV, Srivastava A, Ling CQ, Ling C. Pristimerin enhances recombinant adeno-associated virus vector-mediated transgene expression in human cell lines in vitro and murine hepatocytes in vivo. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2014; 12:20-34. [PMID: 24461592 DOI: 10.1016/s2095-4964(14)60003-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE In the present study, we systemically evaluated the ability of two bioactive compounds from traditional Chinese medicine, celastrol and pristimerin, to enhance recombinant adeno-associated virus (rAAV) serotype vector-mediated transgene expression both in human cell lines in vitro, and in murine hepatocytes in vivo. METHODS Human cell lines were infected with rAAV vectors with either mock treatment or treatment with celastrol or pristimerin. The transgene expression, percentage of nuclear translocated viral genomes and the ubiquitination of intracellular proteins were investigated post-treatment. In addition, nonobese diabetic/severe combined immunodeficient gamma (NSG) mice were tail vain-injected with rAAV vectors and co-administered with either dimethyl sulfoxide, celastrol, pristimerin or a positive control, bortezomib. The transgene expression in liver was detected and compared over time. RESULTS We observed that treatment with pristimerin, at as low as 1 μmol/L concentration, significantly enhanced rAAV2 vector-mediated transgene expression in vitro, and intraperitoneal co-administration with pristimerin at 4 mg/(kg·d) for 3 d dramatically facilitated viral transduction in murine hepatocytes in vivo. The transduction efficiency of the tyrosine-mutant rAAV2 vectors as well as that of rAAV8 vectors carrying oversized transgene cassette was also augmented significantly by pristimerin. The underlying molecular mechanisms by which pristimerin mediated the observed increase in the transduction efficiency of rAAV vectors include both inhibition of proteasomal degradation of the intracellular proteins and enhanced nuclear translocation of the vector genomes. CONCLUSION These studies suggest the potential beneficial use of pristimerin and pristimerin-containing herb extract in future liver-targeted gene therapy with rAAV vectors.
Collapse
Affiliation(s)
- Li-na Wang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yuan Wang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yuan Lu
- Department of Orthopaedics and Rehabilitation, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Zi-fei Yin
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yuan-hui Zhang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - George V Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Chang-quan Ling
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA; E-mail:
| |
Collapse
|
34
|
Sonsteng KM, Prigge JR, Talago EA, June RK, Schmidt EE. Hydrodynamic delivery of Cre protein to lineage-mark or time-stamp mouse hepatocytes in situ. PLoS One 2014; 9:e91219. [PMID: 24626158 PMCID: PMC3953374 DOI: 10.1371/journal.pone.0091219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/10/2014] [Indexed: 12/01/2022] Open
Abstract
Cre-responsive fluorescent marker alleles are powerful tools for cell lineage tracing in mice; however their utility is limited by regulation of Cre activity. When targeting hepatocytes, hydrodynamic delivery of a Cre-expression plasmid can convert Cre-responsive alleles without inducing the intracellular or systemic antiviral responses often associated with viral-derived Cre-expression vectors. In this method, rapid high-volume intravenous inoculation induces hepatocyte-targeted uptake of extracellular molecules. Here we tested whether hydrodynamic delivery of Cre protein or Cre fused to the HIV-TAT cell-penetrating peptide could convert Cre-responsive reporters in hepatocytes of mice. Hydrodynamic delivery of 2 nmol of either Cre or TAT-Cre protein converted the reporter allele in 5 to 20% of hepatocytes. Neither protein gave detectable Cre activity in endothelia, non-liver organs, or non-hepatocyte cells in liver. Using mice homozygous for a Cre-responsive marker that directs red- (Cre-naïve) or green- (Cre-converted) fluorescent proteins to the nucleus, we assessed sub-saturation Cre-activity. One month after hydrodynamic inoculation with Cre protein, 58% of hepatocyte nuclei that were green were also red, indicating that less than half of the hepatocytes that had obtained enough Cre to convert one marker allele to green were able to convert all alleles. For comparison, one month after hydrodynamic delivery of a Cre-expression plasmid with a weak promoter, only 26% of the green nuclei were also red. Our results show that hydrodynamic delivery of Cre protein allows rapid allelic conversion in hepatocytes, but Cre-activity is sub-saturating so many cells will not convert multiple Cre-responsive alleles.
Collapse
Affiliation(s)
- Katherine M. Sonsteng
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Justin R. Prigge
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Emily A. Talago
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Ronald K. June
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, Montana, United States of America
| | - Edward E. Schmidt
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
- * E-mail:
| |
Collapse
|
35
|
Autonomous parvoviruses neither stimulate nor are inhibited by the type I interferon response in human normal or cancer cells. J Virol 2014; 88:4932-42. [PMID: 24554651 DOI: 10.1128/jvi.03508-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Members of the genus Parvovirus are small, nonenveloped single-stranded DNA viruses that are nonpathogenic in humans but have potential utility as cancer therapeutics. Because the innate immune response to parvoviruses has received relatively little attention, we compared the response to parvoviruses to that of several other types of viruses in human cells. In normal human glia, fibroblasts, or melanocytes, vesicular stomatitis virus evoked robust beta interferon (IFN-β) responses. Cytomegalovirus, pseudorabies virus, and Sindbis virus all evoked a 2-log-unit or greater upregulation of IFN-β in glia; in contrast, LuIII and MVMp parvoviruses did not evoke a detectable IFN-β or interferon-stimulated gene (ISG; MX1, oligoadenylate synthetase [OAS], IFIT-1) response in the same cell types. The lack of response raised the question of whether parvoviral infection can be attenuated by IFN; interestingly, we found that IFN did not decrease parvovirus (MVMp, LuIII, and H-1) infectivity in normal human glia, fibroblasts, or melanocytes. The same was true in human cancers, including glioma, sarcoma, and melanoma. Similarly, IFN failed to attenuate transduction by the dependovirus vector adeno-associated virus type 2. Progeny production of parvoviruses was also unimpaired by IFN in both glioma and melanoma, whereas vesicular stomatitis virus replication was blocked. Sarcoma cells with upregulated IFN signaling that show high levels of resistance to other viruses showed strong infection by LuIII. Unlike many other oncolytic viruses, we found no evidence that impairment of innate immunity in cancer cells plays a role in the oncoselectivity of parvoviruses in human cells. Parvoviral resistance to the effects of IFN in cancer cells may constitute an advantage in the virotherapy of some tumors. IMPORTANCE Understanding the interactions between oncolytic viruses and the innate immune system will facilitate employing these viruses as therapeutic agents in cancer patients. The cancer-selective nature of some oncolytic viruses is based on the impaired innate immunity of many cancer cells. The parvoviruses H-1, LuIII, and MVM target cancer cells; however, their relationship with the innate immune system is relatively uncharacterized. Surprisingly, we found that these parvoviruses do not evoke an interferon response in normal human fibroblasts, glia, or melanocytes. Furthermore, unlike most other types of virus, we found that parvovirus infectivity is unaffected by interferon treatment of human normal or tumor cells. Finally, parvoviral replication was unimpaired by interferon in four human tumor types, including those with residual interferon functionality. We conclude that deficits in the interferon antiviral response of cancer cells do not contribute to parvoviral oncoselectivity in human cells. The interferon-resistant phenotype of parvoviruses may give them an advantage over interferon-sensitive oncolytic viruses in tumors showing residual interferon functionality.
Collapse
|
36
|
Rogers GL, Martino AT, Zolotukhin I, Ertl HCJ, Herzog RW. Role of the vector genome and underlying factor IX mutation in immune responses to AAV gene therapy for hemophilia B. J Transl Med 2014; 12:25. [PMID: 24460861 PMCID: PMC3904690 DOI: 10.1186/1479-5876-12-25] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/23/2014] [Indexed: 11/24/2022] Open
Abstract
Background Self-complementary adeno-associated virus (scAAV) vectors have become a desirable vector for therapeutic gene transfer due to their ability to produce greater levels of transgene than single-stranded AAV (ssAAV). However, recent reports have suggested that scAAV vectors are more immunogenic than ssAAV. In this study, we investigated the effects of a self-complementary genome during gene therapy with a therapeutic protein, human factor IX (hF.IX). Methods Hemophilia B mice were injected intramuscularly with ss or scAAV1 vectors expressing hF.IX. The outcome of gene transfer was assessed, including transgene expression as well as antibody and CD8+ T cell responses to hF.IX. Results Self-complementary AAV1 vectors induced similar antibody responses (which eliminated systemic hF.IX expression) but stronger CD8+ T cell responses to hF.IX relative to ssAAV1 in mice with F9 gene deletion. As a result, hF.IX-expressing muscle fibers were effectively eliminated in scAAV-treated mice. In contrast, mice with F9 nonsense mutation (late stop codon) lacked antibody or T cell responses, thus showing long-term expression regardless of the vector genome. Conclusions The nature of the AAV genome can impact the CD8+ T cell response to the therapeutic transgene product. In mice with endogenous hF.IX expression, however, this enhanced immunogenicity did not break tolerance to hF.IX, suggesting that the underlying mutation is a more important risk factor for transgene-specific immunity than the molecular form of the AAV genome.
Collapse
Affiliation(s)
| | | | | | | | - Roland W Herzog
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
37
|
Sen D, Gadkari RA, Sudha G, Gabriel N, Kumar YS, Selot R, Samuel R, Rajalingam S, Ramya V, Nair SC, Srinivasan N, Srivastava A, Jayandharan GR. Targeted modifications in adeno-associated virus serotype 8 capsid improves its hepatic gene transfer efficiency in vivo. Hum Gene Ther Methods 2013; 24:104-16. [PMID: 23442071 DOI: 10.1089/hgtb.2012.195] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recombinant adeno-associated virus vectors based on serotype 8 (AAV8) have shown significant promise for liver-directed gene therapy. However, to overcome the vector dose dependent immunotoxicity seen with AAV8 vectors, it is important to develop better AAV8 vectors that provide enhanced gene expression at significantly low vector doses. Since it is known that AAV vectors during intracellular trafficking are targeted for destruction in the cytoplasm by the host-cellular kinase/ubiquitination/proteasomal machinery, we modified specific serine/threonine kinase or ubiquitination targets on the AAV8 capsid to augment its transduction efficiency. Point mutations at specific serine (S)/threonine (T)/lysine (K) residues were introduced in the AAV8 capsid at the positions equivalent to that of the effective AAV2 mutants, generated successfully earlier. Extensive structure analysis was carried out subsequently to evaluate the structural equivalence between the two serotypes. scAAV8 vectors with the wild-type (WT) and each one of the S/T→Alanine (A) or K-Arginine (R) mutant capsids were evaluated for their liver transduction efficiency in C57BL/6 mice in vivo. Two of the AAV8-S→A mutants (S279A and S671A), and a K137R mutant vector, demonstrated significantly higher enhanced green fluorescent protein (EGFP) transcript levels (~9- to 46-fold) in the liver compared to animals that received WT-AAV8 vectors alone. The best performing AAV8 mutant (K137R) vector also had significantly reduced ubiquitination of the viral capsid, reduced activation of markers of innate immune response, and a concomitant two-fold reduction in the levels of neutralizing antibody formation in comparison to WT-AAV8 vectors. Vector biodistribution studies revealed that the K137R mutant had a significantly higher and preferential transduction of the liver (106 vs. 7.7 vector copies/mouse diploid genome) when compared to WT-AAV8 vectors. To further study the utility of the K137R-AAV8 mutant in therapeutic gene transfer, we delivered human coagulation factor IX (h.FIX) under the control of liver-specific promoters (LP1 or hAAT) into C57BL/6 mice. The circulating levels of h.FIX:Ag were higher in all the K137R-AAV8 treated groups up to 8 weeks post-hepatic gene transfer. These studies demonstrate the feasibility of the use of this novel AAV8 vectors for potential gene therapy of hemophilia B.
Collapse
Affiliation(s)
- Dwaipayan Sen
- Department of Hematology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hareendran S, Balakrishnan B, Sen D, Kumar S, Srivastava A, Jayandharan GR. Adeno-associated virus (AAV) vectors in gene therapy: immune challenges and strategies to circumvent them. Rev Med Virol 2013; 23:399-413. [DOI: 10.1002/rmv.1762] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/08/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Sangeetha Hareendran
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
| | - Balaji Balakrishnan
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Dwaipayan Sen
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Sanjay Kumar
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
| | - Alok Srivastava
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Giridhara R. Jayandharan
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| |
Collapse
|
39
|
Gil-Fariña I, Di Scala M, Vanrell L, Olagüe C, Vales A, High KA, Prieto J, Mingozzi F, Gonzalez-Aseguinolaza G. IL12-mediated liver inflammation reduces the formation of AAV transcriptionally active forms but has no effect over preexisting AAV transgene expression. PLoS One 2013; 8:e67748. [PMID: 23844082 PMCID: PMC3699534 DOI: 10.1371/journal.pone.0067748] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/21/2013] [Indexed: 02/04/2023] Open
Abstract
Recombinant adenoassociated viral vectors (rAAV) have proven to be excellent candidates for gene therapy clinical applications. Recent results showed that cellular immunity to AAV represents a major challenge facing the clinical use of systemic administration of these vectors. Interestingly, no preclinical animal model has previously fully reproduced the clinical findings. The aim of the present work was to enhance the T cell immune response against AAV capsid in mice by the administration of a rAAV expressing the immunostimulatory cytokine IL-12. Our results indicate that although IL-12 expression enhanced the AAV capsid-specific immune response it failed to eliminate transduced hepatocytes and long-term expression was achieved. We found that AAV-mediated transgene expression is altered by IL-12-induced liver inflammation. However, IL-12 expression has no effect over preexisting AAV-mediated transgene expression. IL-12 down-regulates AAV mediated transgene expression via induction of IFN-γ production by NK and T cells, but without altering the transduction efficiency measured by viral genomes. Our results indicate that liver inflammation affects the formation of transcriptionally active AAV vector genomes through an unknown mechanism that can be avoided by the use of DNA-demethylating or anti-inflammatory agents.
Collapse
Affiliation(s)
- Irene Gil-Fariña
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
| | - Marianna Di Scala
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
| | - Lucia Vanrell
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
| | - Cristina Olagüe
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
| | - Africa Vales
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
| | - Katherine A. High
- Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Howard Hughes Medical Institute, Philadelphia, Pennsylvania, United States of America
| | - Jesus Prieto
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, Pamplona, Spain
- Liver Unit and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas, University Clinic of Navarra, La Universidad de Navarra, Pamplona, Spain
| | - Federico Mingozzi
- Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Howard Hughes Medical Institute, Philadelphia, Pennsylvania, United States of America
| | | |
Collapse
|
40
|
AAV2 production with optimized N/P ratio and PEI-mediated transfection results in low toxicity and high titer for in vitro and in vivo applications. J Virol Methods 2013; 193:270-7. [PMID: 23791963 DOI: 10.1016/j.jviromet.2013.06.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 05/22/2013] [Accepted: 06/03/2013] [Indexed: 01/24/2023]
Abstract
The adeno-associated virus (AAV) is one of the most useful viral vectors for gene delivery for both in vivo and in vitro applications. A variety of methods have been established to produce and characterize recombinant AAV (rAAV) vectors; however most methods are quite cumbersome and obtaining consistently high titer can be problematic. This protocol describes a triple-plasmid co-transfection approach with 25 kDa linear polyethylenimine (PEI) in 293 T cells for the production of AAV serotype 2. Seventy-two hours post-transfection, supernatant and cells were harvested and purified by a discontinuous iodixanol density gradient ultracentrifugation, then dialyzed and concentrated with an Amicon 15 100,000 MWCO concentration unit. To optimize the protocol for AAV2 production using PEI, various N/P ratios and DNA amounts were compared. We found that an N/P ratio of 40 coupled with 1.05 μg DNA per ml of media (21 μg DNA/15 cm dish) was found to produce the highest yields for viral replication and assembly measured multiple ways. The infectious units, as determined by serial dilution, were between 1×10(8) and 2×10(9) IU/ml. The genomic titer of the viral stock was determined by qPCR and ranged from 2×10(12) to 6×10(13) VG/ml. These viral vectors showed high expression both in vivo within the brain and in vitro in cell culture. The use of linear 25 kDa polyethylenamine PEI as a transfection reagent is a simple, more cost-effective, and stable means of high-throughput production of high-titer AAV serotype 2. The use of PEI also eliminates the need to change cell medium post-transfection, lowering cost and workload, while producing high-titer, efficacious AAV2 vectors for routine gene transfer.
Collapse
|
41
|
Abstract
Gene therapy products for the treatment of genetic diseases are currently in clinical trials, and one of these, an adeno-associated viral (AAV) product, has recently been licensed. AAV vectors have achieved positive results in a number of clinical and preclinical settings, including hematologic disorders such as the hemophilias, Gaucher disease, hemochromatosis, and the porphyrias. Because AAV vectors are administered directly to the patient, the likelihood of a host immune response is high, as shown by human studies. Preexisting and/or recall responses to the wild-type virus from which the vector is engineered, or to the transgene product itself, can interfere with therapeutic efficacy if not identified and managed optimally. Small-scale clinical studies have enabled investigators to dissect the immune responses to the AAV vector capsid and to the transgene product, and to develop strategies to manage these responses to achieve long-term expression of the therapeutic gene. However, a comprehensive understanding of the determinants of immunogenicity of AAV vectors, and of potential associated toxicities, is still lacking. Careful immunosurveillance conducted as part of ongoing clinical studies will provide the basis for understanding the intricacies of the immune response in AAV-mediated gene transfer, facilitating safe and effective therapies for genetic diseases.
Collapse
|
42
|
Aslanidi GV, Rivers AE, Ortiz L, Song L, Ling C, Govindasamy L, Van Vliet K, Tan M, Agbandje-McKenna M, Srivastava A. Optimization of the capsid of recombinant adeno-associated virus 2 (AAV2) vectors: the final threshold? PLoS One 2013; 8:e59142. [PMID: 23527116 PMCID: PMC3602601 DOI: 10.1371/journal.pone.0059142] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/12/2013] [Indexed: 12/31/2022] Open
Abstract
The ubiquitin-proteasome pathway plays a critical role in the intracellular trafficking of AAV2 vectors, and phosphorylation of certain surface-exposed amino acid residues on the capsid provides the primary signal for ubiquitination. Removal of several critical tyrosine (Y) and serine (S) residues on the AAV2 capsid has been shown to significantly increase transduction efficiency compared with the wild-type (WT) vectors. In the present study, site-directed mutagenesis of each of the 17 surface-exposed threonine (T) residues was conducted, and the transduction efficiency of four of these mutants, T455V, T491V, T550V, and T659V, was observed to increase up to 4-fold in human HEK293 cells in vitro. The most critical Y, S, and T mutations were subsequently combined, and the quadruple-mutant (Y444+500+730F+T491V) AAV2 vector was identified as the most efficient. This vector increased the transduction efficiency ∼24-fold over the WT AAV2 vector, and ∼2-3-fold over the previously described triple-mutant (Y444+500+730F) vector in a murine hepatocyte cell line, H2.35, in vitro. Similar results were obtained in murine hepatocytes in vivo following tail vein injection of the Y444+500+730F+T491V scAAV2 vector, and whole-body bioluminescence imaging of C57BL/6 mice. The increase in the transduction efficiency of the Y-T quadruple-mutant over that of the Y triple-mutant correlated with an improved nuclear translocation of the vectors, which exceeded 90%. These observations suggest that further optimization of the AAV2 capsid by targeting amino acid residues involved in phosphorylation may not be possible. This study has thus led to the generation of a novel Y444+500+730F+T491V quadruple-mutant AAV2 vector with potential for use in liver-directed human gene therapy.
Collapse
Affiliation(s)
- George V Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Arsenic trioxide stabilizes accumulations of adeno-associated virus virions at the perinuclear region, increasing transduction in vitro and in vivo. J Virol 2013; 87:4571-83. [PMID: 23408604 DOI: 10.1128/jvi.03443-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Interactions with cellular stress pathways are central to the life cycle of many latent viruses. Here, we utilize adeno-associated virus (AAV) as a model to study these interactions, as previous studies have demonstrated that cellular stressors frequently increase transduction of recombinant AAV (rAAV) vectors and may even substitute for helper virus functions. Since several chemotherapeutic drugs are known to increase rAAV transduction, we investigated the effect of arsenic trioxide (As(2)O(3)), an FDA-approved chemotherapeutic agent with known effects on several other virus life cycles, on the transduction of rAAV. In vitro, As(2)O(3) caused a dose-dependent increase in rAAV2 transduction over a broad range of cell lines from various cell types and species (e.g., HEK-293, HeLa, HFF hTERT, C-12, and Cos-1). Mechanistically, As(2)O(3) treatment acted to prevent loss of virions from the perinuclear region, which correlated with increased cellular vector genome retention, and was distinguishable from proteasome inhibition. To extend our investigation of the cellular mechanism, we inhibited reactive oxygen species formation and determined that the As(2)O(3)-mediated increase in rAAV2 transduction was dependent upon production of reactive oxygen species. To further validate our in vitro data, we tested the effect of As(2)O(3) on rAAV transduction in vivo and determined that treatment initiated transgene expression as early as 2 days posttransduction and increased reporter expression by up to 10-fold. Moreover, the transduction of several other serotypes of rAAV was also enhanced in vivo, suggesting that As(2)O(3) affects a pathway used by several AAV serotypes. In summary, our data support a model wherein As(2)O(3) increases rAAV transduction both in vitro and in vivo and maintains perinuclear accumulations of capsids, facilitating productive nuclear trafficking.
Collapse
|
44
|
Charan RA, Niizawa G, Nakai H, Clemens PR. Adeno-associated virus serotype 8 (AAV8) delivery of recombinant A20 to skeletal muscle reduces pathological activation of nuclear factor (NF)-κB in muscle of mdx mice. Mol Med 2013; 18:1527-35. [PMID: 23154638 DOI: 10.2119/molmed.2012.00299] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 11/05/2012] [Indexed: 01/29/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic muscle disease caused by the absence of a functional dystrophin protein. Lack of dystrophin protein disrupts the dystrophin-glycoprotein complex causing muscle membrane instability and degeneration. One of the secondary manifestations resulting from lack of functional dystrophin in muscle tissue is an increased level of cytokines that recruit inflammatory cells, leading to chronic upregulation of the nuclear factor (NF)-κB. Negative regulators of the classical NF-κB pathway improve muscle health in the mdx mouse model for DMD. We have previously shown in vitro that a negative regulator of the NF-κB pathway, A20, plays a role in muscle regeneration. Here, we show that overexpression of A20 by using a muscle-specific promoter delivered with an adeno-associated virus serotype 8 (AAV8) vector to the mdx mouse decreases activation of the NF-κB pathway in skeletal muscle. Recombinant A20 expression resulted in a reduction in number of fibers with centrally placed nuclei and a reduction in the number of T cells infiltrating muscle transduced with the AAV8-A20 vector. Taken together, we conclude that overexpression of A20 in mdx skeletal muscle provides improved muscle health by reduction of chronic inflammation and muscle degeneration. These results suggest A20 is a potential therapeutic target to ameliorate symptoms of DMD.
Collapse
Affiliation(s)
- Rakshita A Charan
- Neurology Service, Department of Veterans Affairs Medical Center, Pittsburgh, Pennsylvania, United States of America
| | | | | | | |
Collapse
|
45
|
Differential type I interferon-dependent transgene silencing of helper-dependent adenoviral vs. adeno-associated viral vectors in vivo. Mol Ther 2013; 21:796-805. [PMID: 23319058 DOI: 10.1038/mt.2012.277] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We previously dissected the components of the innate immune response to Helper-dependent adenoviral vectors (HDAds) using genetic models, and demonstrated that multiple pattern recognition receptor signaling pathways contribute to this host response to HDAds in vivo. Based on analysis of cytokine expression profiles, type I interferon (IFN) mRNA is induced in host mouse livers at 1 hour post-injection. This type I IFN signaling amplifies cytokine expression in liver independent of the nature of vector DNA sequences after 3 hours post-injection. This type I IFN signaling in response to HDAds administration contributes to transcriptional silencing of both HDAd prokaryotic and eukaryotic DNA in liver. This silencing occurs early and is mediated by epigenetic modification as shown by in vivo chromatin immunoprecipitation (ChIP) with anti-histone deacetylase (HDAC) and promyelocytic leukemia protein (PML). In contrast, self-complementary adeno-associated viral vectors (scAAVs) showed significantly lower induction of type I IFN mRNA in liver compared to HDAds at both early and late time points. These results show that the type I IFN signaling dependent transgene silencing differs between AAV and HDAd vectors after liver-directed gene transfer.
Collapse
|
46
|
Balakrishnan B, Sen D, Hareendran S, Roshini V, David S, Srivastava A, Jayandharan GR. Activation of the cellular unfolded protein response by recombinant adeno-associated virus vectors. PLoS One 2013; 8:e53845. [PMID: 23320106 PMCID: PMC3540029 DOI: 10.1371/journal.pone.0053845] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 12/05/2012] [Indexed: 12/11/2022] Open
Abstract
The unfolded protein response (UPR) is a stress-induced cyto-protective mechanism elicited towards an influx of large amount of proteins in the endoplasmic reticulum (ER). In the present study, we evaluated if AAV manipulates the UPR pathways during its infection. We first examined the role of the three major UPR axes, namely, endoribonuclease inositol-requiring enzyme-1 (IRE1α), activating transcription factor 6 (ATF6) and PKR-like ER kinase (PERK) in AAV infected cells. Total RNA from mock or AAV infected HeLa cells were used to determine the levels of 8 different ER-stress responsive transcripts from these pathways. We observed a significant up-regulation of IRE1α (up to 11 fold) and PERK (up to 8 fold) genes 12–48 hours after infection with self-complementary (sc)AAV2 but less prominent with single-stranded (ss)AAV2 vectors. Further studies demonstrated that scAAV1 and scAAV6 also induce cellular UPR in vitro, with AAV1 vectors activating the PERK pathway (3 fold) while AAV6 vectors induced a significant increase on all the three major UPR pathways [6–16 fold]. These data suggest that the type and strength of UPR activation is dependent on the viral capsid. We then examined if transient inhibition of UPR pathways by RNA interference has an effect on AAV transduction. siRNA mediated silencing of PERK and IRE1α had a modest effect on AAV2 and AAV6 mediated gene expression (∼1.5–2 fold) in vitro. Furthermore, hepatic gene transfer of scAAV2 vectors in vivo, strongly elevated IRE1α and PERK pathways (2 and 3.5 fold, respectively). However, when animals were pre-treated with a pharmacological UPR inhibitor (metformin) during scAAV2 gene transfer, the UPR signalling and its subsequent inflammatory response was attenuated concomitant to a modest 2.8 fold increase in transgene expression. Collectively, these data suggest that AAV vectors activate the cellular UPR pathways and their selective inhibition may be beneficial during AAV mediated gene transfer.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Dwaipayan Sen
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sangeetha Hareendran
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Vaani Roshini
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sachin David
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Alok Srivastava
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Giridhara R. Jayandharan
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
47
|
The threefold protrusions of adeno-associated virus type 8 are involved in cell surface targeting as well as postattachment processing. J Virol 2012; 86:9396-408. [PMID: 22718833 DOI: 10.1128/jvi.00209-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adeno-associated virus (AAV) has attracted considerable interest as a vector for gene therapy owing its lack of pathogenicity and the wealth of available serotypes with distinct tissue tropisms. One of the most promising isolates for vector development, based on its superior gene transfer efficiency to the liver in small animals compared to AAV type 2 (AAV2), is AAV8. Comparison of the in vivo gene transduction of rAAV2 and rAAV8 in mice showed that single amino acid exchanges in the 3-fold protrusions of AAV8 in the surface loops comprised of residues 581 to 584 and 589 to 592 to the corresponding amino acids of AAV2 and vice versa had a strong influence on transduction efficiency and tissue tropism. Surprisingly, not only did conversion of AAV8 to AAV2 cap sequences increase the transduction efficiency and change tissue tropism but so did the reciprocal conversion of AAV2 to AAV8. Insertion of new peptide motifs at position 590 in AAV8 also enabled retargeting of AAV8 capsids to specific tissues, suggesting that these sequences can interact with receptors on the cell surface. However, a neutralizing monoclonal antibody that binds to amino acids (588)QQNTA(592) of AAV8 does not prevent cell binding and virus uptake, indicating that this region is not necessary for receptor binding but rather that the antibody interferes with an essential step of postattachment processing in which the 3-fold protrusion is also involved. This study supports a multifunctional role of the 3-fold region of AAV capsids in the infection process.
Collapse
|
48
|
Abstract
The administration of recombinant adeno-associated viral vectors (rAAV) for gene transfer induces strong humoral responses through mechanisms that remain incompletely characterized. To investigate the links between innate and adaptive immune responses to the vector, rAAVs were injected intravenously into mice deficient in cell-intrinsic components of innate responses (Toll-like receptors (TLRs), type-1 interferon (IFN) or inflammasome signaling molecules) and AAV-specific antibodies were measured. Of all molecules tested, only MyD88 was critically needed to mount immunoglobulin G (IgG) responses since MyD88(-/-) mice failed to develop high levels of AAV-specific IgG2 and IgG3, regardless of capsid serotype injected. None of the TLRs tested was essential here, but TLR9 ensured a Th1-biased antibody responses. Indeed, capsid-specific Th1 cells were induced upon injection of rAAV1, as directly confirmed with an epitope-tagged capsid, and the priming and development of these Th1 cells required T cell-extrinsic MyD88. Cell transfer experiments showed that autonomous MyD88 signaling in B cells, but not T cells, was sufficient to produce Th1-dependent IgGs. Therefore, rAAV triggers innate responses, at least via B cells, controlling the development of capsid-specific Th1-driven antibodies. MyD88 emerges as a critical and pivotal regulator of both T- and B-cell adaptive immunity against AAV.
Collapse
|
49
|
Mingozzi F, Chen Y, Murphy SL, Edmonson SC, Tai A, Price SD, Metzger ME, Zhou S, Wright JF, Donahue RE, Dunbar CE, High KA. Pharmacological modulation of humoral immunity in a nonhuman primate model of AAV gene transfer for hemophilia B. Mol Ther 2012; 20:1410-6. [PMID: 22565846 PMCID: PMC3392987 DOI: 10.1038/mt.2012.84] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Liver gene transfer for hemophilia B has shown very promising results in recent clinical studies. A potential complication of gene-based treatments for hemophilia and other inherited disorders, however, is the development of neutralizing antibodies (NAb) against the therapeutic transgene. The risk of developing NAb to the coagulation factor IX (F.IX) transgene product following adeno-associated virus (AAV)-mediated hepatic gene transfer for hemophilia is small but not absent, as formation of inhibitory antibodies to F.IX is observed in experimental animals following liver gene transfer. Thus, strategies to modulate antitransgene NAb responses are needed. Here, we used the anti-B cell monoclonal antibody rituximab (rtx) in combination with cyclosporine A (CsA) to eradicate anti-human F.IX NAb in rhesus macaques previously injected intravenously with AAV8 vectors expressing human F.IX. A short course of immunosuppression (IS) resulted in eradication of anti-F.IX NAb with restoration of plasma F.IX transgene product detection. In one animal, following IS anti-AAV6 antibodies also dropped below detection, allowing for successful AAV vector readministration and resulting in high levels (60% or normal) of F.IX transgene product in plasma. Though the number of animals is small, this study supports for the safety and efficacy of B cell-targeting therapies to eradicate NAb developed following AAV-mediated gene transfer.
Collapse
Affiliation(s)
- Federico Mingozzi
- Division of Hematology, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
High-efficiency transduction of human monocyte-derived dendritic cells by capsid-modified recombinant AAV2 vectors. Vaccine 2012; 30:3908-17. [PMID: 22497875 DOI: 10.1016/j.vaccine.2012.03.079] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/22/2012] [Accepted: 03/27/2012] [Indexed: 12/21/2022]
Abstract
Phosphorylation of surface-exposed tyrosine residues negatively impacts the transduction efficiency of recombinant AAV2 vectors. Pre-treatment of cells with specific cellular serine/threonine kinase inhibitors also significantly increased the transduction efficiency of AAV2 vectors. We reasoned that site-directed mutagenesis of surface-exposed serine residues might allow the vectors to evade phosphorylation and thus lead to higher transduction efficiency. Each of the 15 surface-exposed serine (S) residues was substituted with valine (V) residues, and the transduction efficiency of three of these mutants, S458V, S492V and S662V, was increased by up to ≈ 20-fold in different cell types. The S662V mutant was efficient in transducing human monocyte-derived dendritic cells (moDCs), a cell type not readily amenable to transduction by the conventional AAV vectors, and did not induce any phenotypic changes in these cells. Recombinant S662V-AAV2 vectors encoding a truncated human telomerase (hTERT) gene were generated and used to stimulate cytotoxic T cells (CTLs) against target cells. S662V-AAV2-hTERT vector-transduced DCs resulted in rapid, specific T-cell clone proliferation and generation of robust CTLs, which led to specific cell lysis of K562 cells. These studies suggest that high-efficiency transduction of moDCs by serine-modified AAV2 vectors is feasible, which supports the potential utility of these vectors for future human DCs vaccine studies.
Collapse
|