1
|
Ciobanu C, Cebotaru L. AAV1-CFTR preferentially transduces cysts and reduces cyst size in a mouse model of ADPKD. Am J Physiol Cell Physiol 2025; 328:C1783-C1792. [PMID: 40237039 DOI: 10.1152/ajpcell.00057.2025] [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: 01/19/2025] [Revised: 02/03/2025] [Accepted: 04/10/2025] [Indexed: 04/17/2025]
Abstract
To overcome the challenges of developing a gene therapy for autosomal dominant polycystic kidney disease (ADPKD), we focused on the surface receptors expressed in cystic epithelia. Importantly, we detected altered localization in the cystic epithelium of wheat germ agglutinin, WGA, staining of sialic acids. Throughout the membrane of the cysts, we saw altered staining with Maackia amurensis lectin (MAL) or with Sambucus nigra lectin (SNL) that are specific for α2,3- and α2,6-N-linked sialic acids, respectively. Given that these sialic acid glycoproteins facilitate the transduction of adeno-associated virus 1 (AAV1), we injected 1-mo-old, pkd1R3277C/R3277C, (RC/RC) ADPKD mice intraperitoneally with 2 × 1012 particles/kg of AAV1 containing either a green fluorescent protein (GFP) vector or a truncated cystic fibrosis transmembrane conductance regulator (CFTR) vector, Δ27-264-CFTR. Two months after treatment, the cyst area and size were significantly lower in the CFTR vector-treated mice compared with those untreated and those receiving the GFP. We detected vector genomes and mRNA expression only in their corresponding CFTR vector- or GFP vector-treated mice. We observed co-staining for GFP and CFTR immunofluorescence with either the Na+/H+ exchanger or epithelial Na+ channel, indicating proximal tubule or collecting duct expression, respectively. Expression of GFP and CFTR protein expression above background levels was detected. CFTR immunofluorescence was increased in the basolateral membrane after CFTR vector instillation. Finally, these data suggest that cysts are prime targets for AAV1 gene therapy and offer an exciting prospect for ADPKD gene therapy.NEW & NOTEWORTHY Current therapies for ADPKD involve treatment of the symptoms. A direct approach would involve a gene therapy. Here we show AAV1 is tropic for cystic epithelia, which have abundant expression of sialic acid resides known to enhance AAV1 transduction. We show that a CFTR-based vector can reduce cyst size, suggesting that it may be therapeutic. These data suggest that cysts are prime targets for AAV1 and offer an exciting prospect for ADPKD gene therapy.
Collapse
Affiliation(s)
- Cristian Ciobanu
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, United States
| | - Liudmila Cebotaru
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Sorroza-Martinez L, Pelletier M, Guay D, Gaillet B. Recent Advances in Therapeutics and Manufacturing Processes of Recombinant Adeno-Associated Virus for the Treatment of Lung Diseases. Curr Gene Ther 2025; 25:237-256. [PMID: 39225214 DOI: 10.2174/0115665232294935240826061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/01/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Developing delivery vectors capable of transducing genetic material across the lung epithelia and mucus barrier is a major challenge and of great interest to enable gene therapies to treat pulmonary diseases. Recombinant Adeno-associated Viruses (rAAVs) have emerged as attractive candidates among viral and non-viral vectors due to their broad tissue tropism, ability to transduce dividing and quiescent cells, and their safety profile in current human applications. While rAAVs have demonstrated safety in earlier clinical trials for lung disease applications, there are still some limitations regarding rAAV-transgene delivery in pulmonary cells. Thus, further improvements in rAAV engineering are needed to enhance the effectiveness of rAAV-based therapies for lung diseases. Such therapies could benefit patients with chronic lung diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary hypertension, and cystic fibrosis, among others, by regulating hereditary gene mutations or acquired gene deregulations causing these conditions. Alongside therapeutic development, advances in the rAAV production process are essential to meet increasing production demands, while reducing manufacturing costs. This review discusses current challenges and recent advances in the field of rAAV engineering and manufacturing to encourage the clinical development of new pulmonary gene therapy treatments.
Collapse
Affiliation(s)
- Luis Sorroza-Martinez
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Mia Pelletier
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - David Guay
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
3
|
Kwak G, Lee D, Suk JS. Advanced approaches to overcome biological barriers in respiratory and systemic routes of administration for enhanced nucleic acid delivery to the lung. Expert Opin Drug Deliv 2023; 20:1531-1552. [PMID: 37946533 PMCID: PMC10872418 DOI: 10.1080/17425247.2023.2282535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Numerous delivery strategies, primarily novel nucleic acid delivery carriers, have been developed and explored to enable therapeutically relevant lung gene therapy. However, its clinical translation is yet to be achieved despite over 30 years of efforts, which is attributed to the inability to overcome a series of biological barriers that hamper efficient nucleic acid transfer to target cells in the lung. AREAS COVERED This review is initiated with the fundamentals of nucleic acid therapy and a brief overview of previous and ongoing efforts on clinical translation of lung gene therapy. We then walk through the nature of biological barriers encountered by nucleic acid carriers administered via respiratory and/or systemic routes. Finally, we introduce advanced strategies developed to overcome those barriers to achieve therapeutically relevant nucleic acid delivery efficiency in the lung. EXPERT OPINION We are now stepping close to the clinical translation of lung gene therapy, thanks to the discovery of novel delivery strategies that overcome biological barriers via comprehensive preclinical studies. However, preclinical findings should be cautiously interpreted and validated to ultimately realize meaningful therapeutic outcomes with newly developed delivery strategies in humans. In particular, individual strategies should be selected, tailored, and implemented in a manner directly relevant to specific therapeutic applications and goals.
Collapse
Affiliation(s)
- Gijung Kwak
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daiheon Lee
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Sui H, Xu X, Su Y, Gong Z, Yao M, Liu X, Zhang T, Jiang Z, Bai T, Wang J, Zhang J, Xu C, Luo M. Gene therapy for cystic fibrosis: Challenges and prospects. Front Pharmacol 2022; 13:1015926. [PMID: 36304167 PMCID: PMC9592762 DOI: 10.3389/fphar.2022.1015926] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
Abstract
Cystic fibrosis (CF) is a life-threatening autosomal-recessive disease caused by mutations in a single gene encoding cystic fibrosis transmembrane conductance regulator (CFTR). CF effects multiple organs, and lung disease is the primary cause of mortality. The median age at death from CF is in the early forties. CF was one of the first diseases to be considered for gene therapy, and efforts focused on treating CF lung disease began shortly after the CFTR gene was identified in 1989. However, despite the quickly established proof-of-concept for CFTR gene transfer in vitro and in clinical trials in 1990s, to date, 36 CF gene therapy clinical trials involving ∼600 patients with CF have yet to achieve their desired outcomes. The long journey to pursue gene therapy as a cure for CF encountered more difficulties than originally anticipated, but immense progress has been made in the past decade in the developments of next generation airway transduction viral vectors and CF animal models that reproduced human CF disease phenotypes. In this review, we look back at the history for the lessons learned from previous clinical trials and summarize the recent advances in the research for CF gene therapy, including the emerging CRISPR-based gene editing strategies. We also discuss the airway transduction vectors, large animal CF models, the complexity of CF pathogenesis and heterogeneity of CFTR expression in airway epithelium, which are the major challenges to the implementation of a successful CF gene therapy, and highlight the future opportunities and prospects.
Collapse
Affiliation(s)
- Hongshu Sui
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Xinghua Xu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Yanping Su
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Zhaoqing Gong
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Minhua Yao
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Xiaocui Liu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ting Zhang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ziyao Jiang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Tianhao Bai
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Junzuo Wang
- The Affiliated Tai’an City Central Hospital of Qingdao University, Tai’an, Shandong, China
| | - Jingjun Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Changlong Xu
- The Reproductive Medical Center of Nanning Second People’s Hospital, Nanning, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Mingjiu Luo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| |
Collapse
|
5
|
McLachlan G, Alton EWFW, Boyd AC, Clarke NK, Davies JC, Gill DR, Griesenbach U, Hickmott JW, Hyde SC, Miah KM, Molina CJ. Progress in Respiratory Gene Therapy. Hum Gene Ther 2022; 33:893-912. [PMID: 36074947 PMCID: PMC7615302 DOI: 10.1089/hum.2022.172] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The prospect of gene therapy for inherited and acquired respiratory disease has energized the research community since the 1980s, with cystic fibrosis, as a monogenic disorder, driving early efforts to develop effective strategies. The fact that there are still no approved gene therapy products for the lung, despite many early phase clinical trials, illustrates the scale of the challenge: In the 1990s, first-generation non-viral and viral vector systems demonstrated proof-of-concept but low efficacy. Since then, there has been steady progress toward improved vectors with the capacity to overcome at least some of the formidable barriers presented by the lung. In addition, the inclusion of features such as codon optimization and promoters providing long-term expression have improved the expression characteristics of therapeutic transgenes. Early approaches were based on gene addition, where a new DNA copy of a gene is introduced to complement a genetic mutation: however, the advent of RNA-based products that can directly express a therapeutic protein or manipulate gene expression, together with the expanding range of tools for gene editing, has stimulated the development of alternative approaches. This review discusses the range of vector systems being evaluated for lung delivery; the variety of cargoes they deliver, including DNA, antisense oligonucleotides, messenger RNA (mRNA), small interfering RNA (siRNA), and peptide nucleic acids; and exemplifies progress in selected respiratory disease indications.
Collapse
Affiliation(s)
- Gerry McLachlan
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
| | - Eric W F W Alton
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - A Christopher Boyd
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Nora K Clarke
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jane C Davies
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah R Gill
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Uta Griesenbach
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jack W Hickmott
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen C Hyde
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Kamran M Miah
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Claudia Juarez Molina
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Effective viral-mediated lung gene therapy: is airway surface preparation necessary? Gene Ther 2022:10.1038/s41434-022-00332-7. [DOI: 10.1038/s41434-022-00332-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
AbstractGene-based therapeutics are actively being pursued for the treatment of lung diseases. While promising advances have been made over the last decades, the absence of clinically available lung-directed genetic therapies highlights the difficulties associated with this effort. Largely, progress has been hindered by the presence of inherent physical and physiological airway barriers that significantly reduce the efficacy of gene transfer. These barriers include surface mucus, mucociliary action, cell-to-cell tight junctions, and the basolateral cell membrane location of viral receptors for many commonly used gene vectors. Accordingly, airway surface preparation methods have been developed to disrupt these barriers, creating a more conducive environment for gene uptake into the target airway cells. The two major approaches have been chemical and physical methods. Both have proven effective for increasing viral-mediated gene transfer pre-clinically, although with variable effect depending on the specific strategy employed. While such methods have been explored extensively in experimental settings, they have not been used clinically. This review covers the airway surface preparation strategies reported in the literature, the advantages and disadvantages of each method, as well as a discussion about applying this concept in the clinic.
Collapse
|
7
|
Mercier J, Ruffin M, Corvol H, Guillot L. Gene Therapy: A Possible Alternative to CFTR Modulators? Front Pharmacol 2021; 12:648203. [PMID: 33967785 PMCID: PMC8097140 DOI: 10.3389/fphar.2021.648203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a rare genetic disease that affects several organs, but lung disease is the major cause of morbidity and mortality. The gene responsible for CF, the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene, has been discovered in 1989. Since then, gene therapy i.e., defective gene replacement by a functional one, remained the ultimate goal but unfortunately, it has not yet been achieved. However, patients care and symptomatic treatments considerably increased CF patients’ life expectancy ranging from 5 years old in the 1960s to 40 today. In the last decade, research works on CFTR protein structure and activity led to the development of new drugs which, by readdressing CFTR to the plasma membrane (correctors) or by enhancing its transport activity (potentiators), allow, alone or in combination, an improvement of CF patients’ lung function and quality of life. While expected, it is not yet known whether taking these drugs from an early age and for years will improve the quality of life of CF patients in the long term and further increase their life expectancy. Besides, these molecules are not available (specific variants of CFTR) or accessible (national health policies) for all patients and there is still no curative treatment. Another alternative that could benefit from new technologies, such as gene therapy, is therefore still attractive, although it is not yet offered to patients. Faced with the development of new CFTR correctors and potentiators, the question arises as to whether there is still a place for gene therapy and this is discussed in this perspective.
Collapse
Affiliation(s)
- J Mercier
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - M Ruffin
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - H Corvol
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - L Guillot
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| |
Collapse
|
8
|
Breaking the sound barrier: Towards next-generation AAV vectors for gene therapy of hearing disorders. Hear Res 2020; 413:108092. [PMID: 33268240 DOI: 10.1016/j.heares.2020.108092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/14/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
Owing to the advances in transgenic animal technology and the advent of the next-generation sequencing era, over 120 genes causing hereditary hearing loss have been identified by now. In parallel, the field of human gene therapy continues to make exciting and rapid progress, culminating in the recent approval of several ex vivo and in vivo applications. Despite these encouraging developments and the growing interest in causative treatments for hearing disorders, gene therapeutic interventions in the inner ear remain in their infancy and await clinical translation. This review focuses on the adeno-associated virus (AAV), which nowadays represents one of the safest and most promising vectors in gene therapy. We first provide an overview of AAV biology and outline the principles of therapeutic gene transfer with recombinant AAV vectors, before pointing out major challenges and solutions for clinical translation including vector manufacturing and species translatability. Finally, we highlight seminal technologies for engineering and selection of next-generation "designer" AAV capsids, and illustrate their power and potential with recent examples of their application for inner ear gene transfer in animals.
Collapse
|
9
|
Carneiro A, Lee H, Lin L, van Haasteren J, Schaffer DV. Novel Lung Tropic Adeno-Associated Virus Capsids for Therapeutic Gene Delivery. Hum Gene Ther 2020; 31:996-1009. [PMID: 32799685 DOI: 10.1089/hum.2020.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Efforts to identify mutations that underlie inherited genetic diseases combined with strides in the development of gene therapy vectors over the last three decades have culminated in the approval of several adeno-associated virus (AAV)-based gene therapies. Genetic diseases that manifest in the lung such as cystic fibrosis (CF) and surfactant deficiencies, however, have so far proven to be elusive targets. Early clinical trials in CF using AAV serotype 2 (AAV2) achieved safety, but not efficacy endpoints; however, importantly, these studies provided critical information on barriers that need to be surmounted to translate AAV lung gene therapy toward clinical success. Bolstered with an improved understanding of AAV biology and more clinically relevant lung models, next-generation molecular biology and bioinformatics approaches have given rise to novel AAV capsid variants that offer improvements in transduction efficiency, immunological profile, and the ability to circumvent physical barriers in the lung such as mucus. This review discusses the principal limiting barriers to clinical success in lung gene therapy and focuses on novel engineered AAV capsid variants that have been developed to overcome those challenges.
Collapse
Affiliation(s)
- Ana Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Hyuncheol Lee
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA
| | - Li Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Joost van Haasteren
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA.,Department of Bioengineering, University of California, Berkeley, California, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, California, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA.,Innovative Genomics Institute (IGI), University of California, Berkeley, California, USA
| |
Collapse
|
10
|
Tang Y, Yan Z, Engelhardt JF. Viral Vectors, Animal Models, and Cellular Targets for Gene Therapy of Cystic Fibrosis Lung Disease. Hum Gene Ther 2020; 31:524-537. [PMID: 32138545 PMCID: PMC7232698 DOI: 10.1089/hum.2020.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
After more than two decades since clinical trials tested the first use of recombinant adeno-associated virus (rAAV) to treat cystic fibrosis (CF) lung disease, gene therapy for this disorder has undergone a tremendous resurgence. Fueling this enthusiasm has been an enhanced understanding of rAAV transduction biology and cellular processes that limit transduction of airway epithelia, the development of new rAAV serotypes and other vector systems with high-level tropism for airway epithelial cells, an improved understanding of CF lung pathogenesis and the cellular targets for gene therapy, and the development of new animal models that reproduce the human CF disease phenotype. These advances have created a preclinical path for both assessing the efficacy of gene therapies in the CF lung and interrogating the target cell types in the lung required for complementation of the CF disease state. Lessons learned from early gene therapy attempts with rAAV in the CF lung have guided thinking for the testing of next-generation vector systems. Although unknown questions still remain regarding the cellular targets in the lung that are required or sufficient to complement CF lung disease, the field is now well positioned to tackle these challenges. This review will highlight the role that next-generation CF animal models are playing in the preclinical development of gene therapies for CF lung disease and the knowledge gaps in disease pathophysiology that these models are attempting to fill.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
11
|
Zengel J, Carette JE. Structural and cellular biology of adeno-associated virus attachment and entry. Adv Virus Res 2020; 106:39-84. [PMID: 32327148 DOI: 10.1016/bs.aivir.2020.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adeno-associated virus (AAV) is a nonenveloped, ssDNA virus in the parvovirus family, which has become one of the leading candidate vectors for human gene therapy. AAV has been studied extensively to identify host cellular factors involved in infection, as well as to identify capsid variants that confer clinically favorable transduction profiles ex vivo and in vivo. Recent advances in technology have allowed for direct genetic approaches to be used to more comprehensively characterize host factors required for AAV infection and allowed for identification of a critical multi-serotype receptor, adeno-associated virus receptor (AAVR). In this chapter, we will discuss the interactions of AAV with its glycan and proteinaceous receptors and describe the host and viral components involved in AAV entry, which requires cellular attachment, endocytosis, trafficking to the trans-Golgi network and nuclear import. AAV serves as a paradigm for entry of nonenveloped viruses. Furthermore, we will discuss the potential of utilizing our increased understanding of virus-host interactions during AAV entry to develop better AAV-based therapeutics, with a focus on host factors and capsid interactions involved in in vivo tropism.
Collapse
|
12
|
A CRISPR Screen Identifies the Cell Polarity Determinant Crumbs 3 as an Adeno-associated Virus Restriction Factor in Hepatocytes. J Virol 2019; 93:JVI.00943-19. [PMID: 31391273 DOI: 10.1128/jvi.00943-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022] Open
Abstract
Adeno-associated viruses (AAV) are helper-dependent parvoviruses that have been developed into promising gene therapy vectors. Many studies, including a recent unbiased genomic screen, have identified host factors essential for AAV cell entry, but no genome-wide screens that address inhibitory host factors have been reported. Here, we utilize a novel CRISPR screen to identify AAV restriction factors in a human hepatocyte cell line. The major hit from our gain-of-function screen is the apical polarity determinant Crumbs 3 (Crb3). Knockout (KO) of Crb3 enhances AAV transduction, while overexpression exerts the opposite effect. Further, Crb3 appears to restrict AAV transduction in a serotype- and cell type-specific manner. Particularly, for AAV serotype 9 and a rationally engineered AAV variant, we demonstrate that increased availability of galactosylated glycans on the surfaces of Crb3 KO cells, but not the universal AAV receptor, leads to increased capsid attachment and enhanced transduction. We postulate that Crb3 could serve as a key molecular determinant that restricts the availability of AAV glycan attachment factors on the cell surface by maintaining apical-basal polarity and tight junction integrity.IMPORTANCE Adeno-associated viruses (AAVs) have recently emerged at the forefront as gene therapy vectors; however, our understanding of host factors that influence AAV transduction in different cell types is still evolving. In the present study, we perform a genome-scale CRISPR knockout screen to identify cellular host factors that restrict AAV infection in hepatocyte cultures. We discover that Crumbs 3, which determines cellular polarity, also influences the distribution of certain carbohydrate attachment factors on the cell surface. This in turn affects the ability of virions to bind and enter the cells. This study underscores the importance of cell polarity in AAV transduction and provides a potential molecular basis for the differential infectious mechanism(s) in cell culture versus organ systems.
Collapse
|
13
|
Tamhankar M, Patterson JL. Directional entry and release of Zika virus from polarized epithelial cells. Virol J 2019; 16:99. [PMID: 31395061 PMCID: PMC6688342 DOI: 10.1186/s12985-019-1200-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/18/2019] [Indexed: 03/09/2023] Open
Abstract
Background Both vector borne and sexual transmission of Zika virus (ZIKV) involve infection of epithelial cells in the initial stages of infection. Epithelial cells are unique in their ability to form polarized monolayers and their barrier function. Cell polarity induces an asymmetry in the epithelial monolayer, which is maintained by tight junctions and specialized sorting machinery. This differential localization can have a potential impact of virus infection. Asymmetrical distribution of a viral receptor can restrict virus entry to a particular membrane while polarized sorting can lead to a directional release of virions. The present study examined the impact of cell polarity on ZIKV infection and release. Methods A polarized Caco-2 cell model we described previously was used to assess ZIKV infection. Transepithelial resistance (TEER) was used to assess epithelial cell polarity, and virus infection was measured by immunofluorescence microscopy and qRT-PCR. Cell permeability was measured using a fluorescein leakage assay. Statistical significance was calculated using one-way ANOVA and significance was set at p < 0.05. Results Using the Caco-2 cell model for polarized epithelial cells, we report that Zika virus preferentially infects polarized cells from the apical route and is released vectorially through the basolateral route. Our data also indicates that release occurs without disruption of cell permeability. Conclusions Our results show that ZIKV has directional infection and egress in a polarized cell system. This mechanism of directional infection may be one of the mechanisms that enables the cross the epithelial barrier effectively without a disruption in cell monolayer integrity. Elucidation of entry and release characteristics of Zika virus in polarized epithelial cells can lead to better understanding of virus dissemination in the host, and can help in developing effective therapeutic interventions. Electronic supplementary material The online version of this article (10.1186/s12985-019-1200-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manasi Tamhankar
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA.,Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jean L Patterson
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
14
|
Hamilton BA, Li X, Pezzulo AA, Abou Alaiwa MH, Zabner J. Polarized AAVR expression determines infectivity by AAV gene therapy vectors. Gene Ther 2019; 26:240-249. [PMID: 30962536 PMCID: PMC6588428 DOI: 10.1038/s41434-019-0078-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/04/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV) has been investigated to transfer the cystic fibrosis transmembrane conductance regulator (CFTR) to airways. Inhaled AAV2-CFTR in people with cystic fibrosis (CF) is safe, but inefficient. In vitro, AAV2 transduction of human airway epithelia on the apical (luminal) side is inefficient, but efficient basolaterally. We previously selected AAV2.5T, a novel capsid that apically transduces CF human airway epithelia and efficiently restores CFTR function. We hypothesize the AAV receptor (AAVR) is basolaterally localized, and that AAV2.5T utilizes an alternative apical receptor. We found AAVR in human airway epithelia by western blot and RNA-Seq analyses. Using immunocytochemistry we did not find endogenous AAVR at membranes but overexpression localized AAVR to the basolateral membrane, where it preferentially increased transduction. Anti-AAVR antibodies blocked transduction by AAV2 from the basolateral side but not AAV2.5T from the apical side, suggesting a unique apical receptor. Finally, we found infection by AAV2 but not AAV2.5T was blocked by CRISPR knockout of AAVR in cell lines. Our data suggest the absence of apical AAVR is rate limiting for AAV2, and efficient transduction by AAV2.5T is accomplished using an AAVR independent pathway. Our findings inform the development of gene therapy for CF, and AAV vectors in general.
Collapse
Affiliation(s)
- Bradley A Hamilton
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
- Molecular Medicine Program, The University of Iowa, Iowa City, IA, USA
| | - Xiaopeng Li
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - Alejandro A Pezzulo
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - Mahmoud H Abou Alaiwa
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, The University of Iowa, Iowa City, IA, USA
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA.
- Molecular Medicine Program, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Cooney AL, McCray PB, Sinn PL. Cystic Fibrosis Gene Therapy: Looking Back, Looking Forward. Genes (Basel) 2018; 9:genes9110538. [PMID: 30405068 PMCID: PMC6266271 DOI: 10.3390/genes9110538] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that encodes a cAMP-regulated anion channel. Although CF is a multi-organ system disease, most people with CF die of progressive lung disease that begins early in childhood and is characterized by chronic bacterial infection and inflammation. Nearly 90% of people with CF have at least one copy of the ΔF508 mutation, but there are hundreds of CFTR mutations that result in a range of disease severities. A CFTR gene replacement approach would be efficacious regardless of the disease-causing mutation. After the discovery of the CFTR gene in 1989, the in vitro proof-of-concept for gene therapy for CF was quickly established in 1990. In 1993, the first of many gene therapy clinical trials attempted to rescue the CF defect in airway epithelia. Despite the initial enthusiasm, there is still no FDA-approved gene therapy for CF. Here we discuss the history of CF gene therapy, from the discovery of the CFTR gene to current state-of-the-art gene delivery vector designs. While implementation of CF gene therapy has proven more challenging than initially envisioned; thanks to continued innovation, it may yet become a reality.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Patrick L Sinn
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
16
|
Stiles KM, Sondhi D, Kaminsky SM, De BP, Rosenberg JB, Crystal RG. Intrapleural Gene Therapy for Alpha-1 Antitrypsin Deficiency-Related Lung Disease. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:244-257. [PMID: 30723782 DOI: 10.15326/jcopdf.5.4.2017.0160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) manifests primarily as early-onset emphysema caused by the destruction of the lung by neutrophil elastase due to low amounts of the serine protease inhibitor alpha-1 antitrypsin (AAT). The current therapy involves weekly intravenous infusions of AAT-derived from pooled human plasma that is efficacious, yet costly. Gene therapy applications designed to provide constant levels of the AAT protein are currently under development. The challenge is for gene therapy to provide sufficient amounts of AAT to normalize the inhibitor level and anti-neutrophil elastase capacity in the lung. One strategy involves administration of an adeno-associated virus (AAV) gene therapy vector to the pleural space providing both local and systemic production of AAT to reach consistent therapeutic levels. This review focuses on the strategy, advantages, challenges, and updates for intrapleural administration of gene therapy vectors for the treatment of AATD.
Collapse
Affiliation(s)
- Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York.,KMS and DS contributed equally to this review
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York.,KMS and DS contributed equally to this review
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Bishnu P De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
17
|
Guggino WB, Benson J, Seagrave J, Yan Z, Engelhardt J, Gao G, Conlon TJ, Cebotaru L. A Preclinical Study in Rhesus Macaques for Cystic Fibrosis to Assess Gene Transfer and Transduction by AAV1 and AAV5 with a Dual-Luciferase Reporter System. HUM GENE THER CL DEV 2017; 28:145-156. [PMID: 28726496 DOI: 10.1089/humc.2017.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease that is potentially treatable by gene therapy. Since the identification of the gene encoding CF transmembrane conductance regulator, a number of preclinical and clinical trials have been conducted using the first generation of adeno-associated virus, AAV2. All these studies showed that AAV gene therapy for CF is safe, but clinical benefit was not clearly demonstrated. Thus, a new generation of AAV vectors based on other serotypes is needed to move the field forward. This study tested two AAV serotypes (AAV1 and AAV5) using a dual-luciferase reporter system with firefly and Renilla luciferase genes packaged into AAV1 or AAV5, respectively. Two male and two female Rhesus macaques were each instilled in their lungs with both serotypes using a Penn-Century microsprayer. Both AAV1 and AAV5 vector genomes were detected in all the lung samples when measured at the time of necropsy, 45 days after instillation. However, the vector genome number for AAV1 was at least 10-fold higher than for AAV5. Likewise, luciferase activity was also detected in the same samples at 45 days. AAV1-derived activity was not statistically greater than that derived from AAV5. These data suggest that gene transfer is greater for AAV1 than for AAV5 in macaque lungs. Serum neutralizing antibodies were increased dramatically against both serotypes but were less abundant with AAV1 than with AAV5. No adverse events were noted, again indicating that AAV gene therapy is safe. These results suggest that with more lung-tropic serotypes such as AAV1, new clinical studies of gene therapy using AAV are warranted.
Collapse
Affiliation(s)
- William B Guggino
- 1 Department of Physiology, Johns Hopkins University , Baltimore, Maryland
| | - Janet Benson
- 2 Lovelace Respiratory Research Institute , Albuquerque, New Mexico
| | | | - Ziying Yan
- 3 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - John Engelhardt
- 3 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Guangping Gao
- 4 Department of Microbiology & Physiological Systems, University of Massachusetts , Worcester, Massachusetts
| | - Thomas J Conlon
- 5 Department of Pediatrics, University of Florida , Gainesville, Florida
| | - Liudmila Cebotaru
- 6 Department of Medicine, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
18
|
Guggino WB, Cebotaru L. Adeno-Associated Virus (AAV) gene therapy for cystic fibrosis: current barriers and recent developments. Expert Opin Biol Ther 2017; 17:1265-1273. [PMID: 28657358 DOI: 10.1080/14712598.2017.1347630] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Since the cystic fibrosis (CF) gene was discovered in 1989, researchers have worked to develop a gene therapy. One of the most promising and enduring vectors is the AAV, which has been shown to be safe. In particular, several clinical trials have been conducted with AAV serotype 2. All of them detected viral genomes, but identification of mRNA transduction was not consistent; clinical outcomes in Phase II studies were also inconsistent. The lack of a positive outcome has been attributed to a less-than-efficient viral infection by AAV2, a weak transgene promoter and the host immune response to the vector. Areas covered: Herein, the authors focus on AAV gene therapy for CF, evaluating past experience with this approach and identifying ways forward, based on the progress that has already been made in identifying and overcoming the limitations of AAV gene therapy. Expert opinion: Such progress makes it clear that this is an opportune time to push forward toward the development of a gene therapy for CF. Drugs to treat the basic defect in CF represent a remarkable advance but cannot treat a significant cohort of patients with rare mutations. Thus, there is a critical need to develop a gene therapy for those individuals.
Collapse
Affiliation(s)
- William B Guggino
- a Departments of Medicine and Physiology , Johns Hopkins University , Baltimore , MD , USA
| | - Liudmila Cebotaru
- a Departments of Medicine and Physiology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
19
|
Optimization of adeno-associated virus vector-mediated gene transfer to the respiratory tract. Gene Ther 2017; 24:290-297. [PMID: 28346434 DOI: 10.1038/gt.2017.19] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 12/27/2022]
Abstract
An efficient adeno-associated virus (AAV) vector was constructed for the treatment of respiratory diseases. AAV serotypes, promoters and routes of administration potentially influencing the efficiency of gene transfer to airway cells were examined in the present study. Among the nine AAV serotypes (AAV1-9) screened in vitro and four serotypes (AAV1, 2, 6, 9) evaluated in vivo, AAV6 showed the strongest transgene expression. As for promoters, the cytomegalovirus (CMV) early enhancer/chicken β-actin (CAG) promoter resulted in more robust transduction than the CMV promoter. Regarding delivery routes, intratracheal administration resulted in strong transgene expression in the lung, whereas the intravenous and intranasal administration routes yielded negligible expression. The combination of the AAV6 capsid and CAG promoter resulted in sustained expression, and the intratracheally administered AAV6-CAG vector transduced bronchial cells and pericytes in the lung. These results suggest that AAV6-CAG vectors are more promising than the previously preferred AAV2 vectors for airway transduction, particularly when administered into the trachea. The present study offers an optimized strategy for AAV-mediated gene therapy for lung diseases, such as cystic fibrosis and pulmonary fibrosis.
Collapse
|
20
|
Castellani S, Orlando C, Carbone A, Di Gioia S, Conese M. Magnetofection Enhances Lentiviral-Mediated Transduction of Airway Epithelial Cells through Extracellular and Cellular Barriers. Genes (Basel) 2016; 7:genes7110103. [PMID: 27886077 PMCID: PMC5126789 DOI: 10.3390/genes7110103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/04/2016] [Accepted: 11/15/2016] [Indexed: 02/03/2023] Open
Abstract
Gene transfer to airway epithelial cells is hampered by extracellular (mainly mucus) and cellular (tight junctions) barriers. Magnetofection has been used to increase retention time of lentiviral vectors (LV) on the cellular surface. In this study, magnetofection was investigated in airway epithelial cell models mimicking extracellular and cellular barriers. Bronchiolar epithelial cells (H441 line) were evaluated for LV-mediated transduction after polarization onto filters and dexamethasone (dex) treatment, which induced hemicyst formation, with or without magnetofection. Sputum from cystic fibrosis (CF) patients was overlaid onto cells, and LV-mediated transduction was evaluated in the absence or presence of magnetofection. Magnetofection of unpolarized H441 cells increased the transduction with 50 MOI (multiplicity of infection, i.e., transducing units/cell) up to the transduction obtained with 500 MOI in the absence of magnetofection. Magnetofection well-enhanced LV-mediated transduction in mucus-layered cells by 20.3-fold. LV-mediated transduction efficiency decreased in dex-induced hemicysts in a time-dependent fashion. In dome-forming cells, zonula occludens-1 (ZO-1) localization at the cell borders was increased by dex treatment. Under these experimental conditions, magnetofection significantly increased LV transduction by 5.3-fold. In conclusion, these results show that magnetofection can enhance LV-mediated gene transfer into airway epithelial cells in the presence of extracellular (sputum) and cellular (tight junctions) barriers, representing CF-like conditions.
Collapse
Affiliation(s)
- Stefano Castellani
- Department of Medical and Surgical Sciences, University of Foggia, V. L. Pinto 1, 71122 Foggia, Italy.
| | - Clara Orlando
- Brainlab AG, Kapellenstrasse 12, 85622 Feldkirchen, Germany.
| | - Annalucia Carbone
- Department of Medical and Surgical Sciences, University of Foggia, V. L. Pinto 1, 71122 Foggia, Italy.
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, V. L. Pinto 1, 71122 Foggia, Italy.
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, V. L. Pinto 1, 71122 Foggia, Italy.
| |
Collapse
|
21
|
Srivastava A. In vivo tissue-tropism of adeno-associated viral vectors. Curr Opin Virol 2016; 21:75-80. [PMID: 27596608 DOI: 10.1016/j.coviro.2016.08.003] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 12/18/2022]
Abstract
In this review, a brief account of the historical perspective of the discovery of the first cellular receptor and co-receptor of the prototype adeno-associated virus serotype 2 (AAV2) will be presented. The Subsequent discovery of a number of AAV serotypes, and attempts to identify the cellular receptors and co-receptors for these serotype vectors has had significant implications in their use in human gene therapy. As additional AAV serotypes are discovered and isolated, a detailed understanding of their tropism is certainly likely to play a key role in all future studies, both basic science as well as clinical.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida College of Medicine, 2033 Mowry Road, Gainesville, FL 32611, United States; Department of Molecular Genetics & Microbiology, Powell Gene Therapy Center, University of Florida College of Medicine, 2033 Mowry Road, Gainesville, FL 32611, United States.
| |
Collapse
|
22
|
Chiuchiolo MJ, Crystal RG. Gene Therapy for Alpha-1 Antitrypsin Deficiency Lung Disease. Ann Am Thorac Soc 2016; 13 Suppl 4:S352-69. [PMID: 27564673 PMCID: PMC5059492 DOI: 10.1513/annalsats.201506-344kv] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/28/2015] [Indexed: 12/16/2022] Open
Abstract
Alpha-1 antitrypsin (AAT) deficiency, characterized by low plasma levels of the serine protease inhibitor AAT, is associated with emphysema secondary to insufficient protection of the lung from neutrophil proteases. Although AAT augmentation therapy with purified AAT protein is efficacious, it requires weekly to monthly intravenous infusion of AAT purified from pooled human plasma, has the risk of viral contamination and allergic reactions, and is costly. As an alternative, gene therapy offers the advantage of single administration, eliminating the burden of protein infusion, and reduced risks and costs. The focus of this review is to describe the various strategies for AAT gene therapy for the pulmonary manifestations of AAT deficiency and the state of the art in bringing AAT gene therapy to the bedside.
Collapse
Affiliation(s)
- Maria J Chiuchiolo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
23
|
Vidović D, Carlon MS, da Cunha MF, Dekkers JF, Hollenhorst MI, Bijvelds MJC, Ramalho AS, Van den Haute C, Ferrante M, Baekelandt V, Janssens HM, De Boeck K, Sermet-Gaudelus I, de Jonge HR, Gijsbers R, Beekman JM, Edelman A, Debyser Z. rAAV-CFTRΔR Rescues the Cystic Fibrosis Phenotype in Human Intestinal Organoids and Cystic Fibrosis Mice. Am J Respir Crit Care Med 2016; 193:288-98. [PMID: 26509335 DOI: 10.1164/rccm.201505-0914oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Gene therapy holds promise for a curative mutation-independent treatment applicable to all patients with cystic fibrosis (CF). The various viral vector-based clinical trials conducted in the past have demonstrated safety and tolerance of different vectors, but none have led to a clear and persistent clinical benefit. Recent clinical breakthroughs in recombinant adeno-associated viral vector (rAAV)-based gene therapy encouraged us to reexplore an rAAV approach for CF. OBJECTIVES We evaluated the preclinical potential of rAAV gene therapy for CF to restore chloride and fluid secretion in two complementary models: intestinal organoids derived from subjects with CF and a CF mouse model, an important milestone toward the development of a clinical rAAV candidate for CF gene therapy. METHODS We engineered an rAAV vector containing a truncated CF transmembrane conductance regulator (CFTRΔR) combined with a short promoter (CMV173) to ensure optimal gene expression. A rescue in chloride and fluid secretion after rAAV-CFTRΔR treatment was assessed by forskolin-induced swelling in CF transmembrane conductance regulator (CFTR)-deficient organoids and by nasal potential differences in ΔF508 mice. MEASUREMENTS AND MAIN RESULTS rAAV-CFTRΔR transduction of human CFTR-deficient organoids resulted in forskolin-induced swelling, indicating a restoration of CFTR function. Nasal potential differences demonstrated a clear response to low chloride and forskolin perfusion in most rAAV-CFTRΔR-treated CF mice. CONCLUSIONS Our study provides robust evidence that rAAV-mediated gene transfer of a truncated CFTR functionally rescues the CF phenotype across the nasal mucosa of CF mice and in patient-derived organoids. These results underscore the clinical potential of rAAV-CFTRΔR in offering a cure for all patients with CF in the future.
Collapse
Affiliation(s)
| | | | - Mélanie F da Cunha
- 2 INSERM U1151, University Paris Descartes, Faculté de Médecine Necker Enfants-Malades, Paris, France
| | - Johanna F Dekkers
- 3 Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, and.,4 Laboratory of Translational Immunology, University Medical Center, Utrecht, the Netherlands
| | - Monika I Hollenhorst
- 2 INSERM U1151, University Paris Descartes, Faculté de Médecine Necker Enfants-Malades, Paris, France
| | - Marcel J C Bijvelds
- 5 Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | | | - Marc Ferrante
- 9 Translational Research in Gastrointestinal Disorders, KU Leuven, Flanders, Belgium
| | | | - Hettie M Janssens
- 10 Department of Pediatric Pulmonology, Erasmus University Medical Centre/Sophia Children's Hospital, Rotterdam, the Netherlands; and
| | | | - Isabelle Sermet-Gaudelus
- 2 INSERM U1151, University Paris Descartes, Faculté de Médecine Necker Enfants-Malades, Paris, France
| | - Hugo R de Jonge
- 5 Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rik Gijsbers
- 1 Molecular Virology and Gene Therapy.,8 Leuven Viral Vector Core, and
| | - Jeffrey M Beekman
- 3 Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, and.,4 Laboratory of Translational Immunology, University Medical Center, Utrecht, the Netherlands
| | - Aleksander Edelman
- 2 INSERM U1151, University Paris Descartes, Faculté de Médecine Necker Enfants-Malades, Paris, France
| | | |
Collapse
|
24
|
Kim N, Duncan GA, Hanes J, Suk JS. Barriers to inhaled gene therapy of obstructive lung diseases: A review. J Control Release 2016; 240:465-488. [PMID: 27196742 DOI: 10.1016/j.jconrel.2016.05.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/29/2022]
Abstract
Knowledge of genetic origins of obstructive lung diseases has made inhaled gene therapy an attractive alternative to the current standards of care that are limited to managing disease symptoms. Initial lung gene therapy clinical trials occurred in the early 1990s following the discovery of the genetic defect responsible for cystic fibrosis (CF), a monogenic disorder. However, despite over two decades of intensive effort, gene therapy has yet to help patients with CF or any other obstructive lung disease. The slow progress is due in part to poor understanding of the biological barriers to inhaled gene therapy. Encouragingly, clinical trials have shown that inhaled gene therapy with various viral vectors and non-viral gene vectors is well tolerated by patients, and continued research has provided valuable lessons and resources that may lead to future success of this therapeutic strategy. In this review, we first introduce representative obstructive lung diseases and examine limitations of currently available therapeutic options. We then review key components for successful execution of inhaled gene therapy, including gene delivery systems, primary physiological barriers and strategies to overcome them, and advances in preclinical disease models with which the most promising systems may be identified for human clinical trials.
Collapse
Affiliation(s)
- Namho Kim
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregg A Duncan
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Justin Hanes
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Environmental and Health Sciences, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jung Soo Suk
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
25
|
Chirkova T, Lin S, Oomens AGP, Gaston KA, Boyoglu-Barnum S, Meng J, Stobart CC, Cotton CU, Hartert TV, Moore ML, Ziady AG, Anderson LJ. CX3CR1 is an important surface molecule for respiratory syncytial virus infection in human airway epithelial cells. J Gen Virol 2015; 96:2543-2556. [PMID: 26297201 DOI: 10.1099/vir.0.000218] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe pneumonia and bronchiolitis in infants and young children, and causes disease throughout life. Understanding the biology of infection, including virus binding to the cell surface, should help develop antiviral drugs or vaccines. The RSV F and G glycoproteins bind cell surface heparin sulfate proteoglycans (HSPGs) through heparin-binding domains. The G protein also has a CX3C chemokine motif which binds to the fractalkine receptor CX3CR1. G protein binding to CX3CR1 is not important for infection of immortalized cell lines, but reportedly is so for primary human airway epithelial cells (HAECs), the primary site for human infection. We studied the role of CX3CR1 in RSV infection with CX3CR1-transfected cell lines and HAECs with variable percentages of CX3CR1-expressing cells, and the effect of anti-CX3CR1 antibodies or a mutation in the RSV CX3C motif. Immortalized cells lacking HSPGs had low RSV binding and infection, which was increased markedly by CX3CR1 transfection. CX3CR1 was expressed primarily on ciliated cells, and ∼50 % of RSV-infected cells in HAECs were CX3CR1+. HAECs with more CX3CR1-expressing cells had a proportional increase in RSV infection. Blocking G binding to CX3CR1 with anti-CX3CR1 antibody or a mutation in the CX3C motif significantly decreased RSV infection in HAECs. The kinetics of cytokine production suggested that the RSV/CX3CR1 interaction induced RANTES (regulated on activation normal T-cell expressed and secreted protein), IL-8 and fractalkine production, whilst it downregulated IL-15, IL1-RA and monocyte chemotactic protein-1. Thus, the RSV G protein/CX3CR1 interaction is likely important in infection and infection-induced responses of the airway epithelium, the primary site of human infection.
Collapse
Affiliation(s)
- Tatiana Chirkova
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Songbai Lin
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Antonius G P Oomens
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Kelsey A Gaston
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Seyhan Boyoglu-Barnum
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Jia Meng
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Christopher C Stobart
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Calvin U Cotton
- Division of Pediatric Pulmonology, Case Western University, Cleveland, Ohio, USA
| | - Tina V Hartert
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine and Vanderbilt Center for Asthma and Environmental Health Sciences Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Martin L Moore
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Assem G Ziady
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Larry J Anderson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Yan Z, Sun X, Feng Z, Li G, Fisher JT, Stewart ZA, Engelhardt JF. Optimization of Recombinant Adeno-Associated Virus-Mediated Expression for Large Transgenes, Using a Synthetic Promoter and Tandem Array Enhancers. Hum Gene Ther 2015; 26:334-46. [PMID: 25763813 DOI: 10.1089/hum.2015.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The packaging capacity of recombinant adeno-associated viral (rAAV) vectors limits the size of the promoter that can be used to express the 4.43-kb cystic fibrosis transmembrane conductance regulator (CFTR) cDNA. To circumvent this limitation, we screened a set of 100-mer synthetic enhancer elements, composed of ten 10-bp repeats, for their ability to augment CFTR transgene expression from a short 83-bp synthetic promoter in the context of an rAAV vector designed for use in the cystic fibrosis (CF) ferret model. Our initial studies assessing transcriptional activity in monolayer (nonpolarized) cultures of human airway cell lines and primary ferret airway cells revealed that three of these synthetic enhancers (F1, F5, and F10) significantly promoted transcription of a luciferase transgene in the context of plasmid transfection. Further analysis in polarized cultures of human and ferret airway epithelia at an air-liquid interface (ALI), as well as in the ferret airway in vivo, demonstrated that the F5 enhancer produced the highest level of transgene expression in the context of an AAV vector. Furthermore, we demonstrated that increasing the size of the viral genome from 4.94 to 5.04 kb did not significantly affect particle yield of the vectors, but dramatically reduced the functionality of rAAV-CFTR vectors because of small terminal deletions that extended into the CFTR expression cassette of the 5.04-kb oversized genome. Because rAAV-CFTR vectors greater than 5 kb in size are dramatically impaired with respect to vector efficacy, we used a shortened ferret CFTR minigene with a 159-bp deletion in the R domain to construct an rAAV vector (AV2/2.F5tg83-fCFTRΔR). This vector yielded an ∼17-fold increase in expression of CFTR and significantly improved Cl(-) currents in CF ALI cultures. Our study has identified a small enhancer/promoter combination that may have broad usefulness for rAAV-mediated CF gene therapy to the airway.
Collapse
Affiliation(s)
- Ziying Yan
- 1 Department of Anatomy and Cell Biology, University of Iowa School of Medicine , Iowa City, IA 52242.,2 Center for Gene Therapy, University of Iowa School of Medicine , Iowa City, IA 52242
| | - Xingshen Sun
- 1 Department of Anatomy and Cell Biology, University of Iowa School of Medicine , Iowa City, IA 52242
| | - Zehua Feng
- 1 Department of Anatomy and Cell Biology, University of Iowa School of Medicine , Iowa City, IA 52242
| | - Guiying Li
- 3 Department of Surgery, University of Iowa School of Medicine , Iowa City, IA 52242
| | - John T Fisher
- 1 Department of Anatomy and Cell Biology, University of Iowa School of Medicine , Iowa City, IA 52242
| | - Zoe A Stewart
- 3 Department of Surgery, University of Iowa School of Medicine , Iowa City, IA 52242
| | - John F Engelhardt
- 1 Department of Anatomy and Cell Biology, University of Iowa School of Medicine , Iowa City, IA 52242.,2 Center for Gene Therapy, University of Iowa School of Medicine , Iowa City, IA 52242.,4 Department of Internal Medicine, University of Iowa School of Medicine , Iowa City, IA 52242
| |
Collapse
|
27
|
Yan Z, Stewart ZA, Sinn PL, Olsen JC, Hu J, McCray PB, Engelhardt JF. Ferret and pig models of cystic fibrosis: prospects and promise for gene therapy. HUM GENE THER CL DEV 2015; 26:38-49. [PMID: 25675143 PMCID: PMC4367511 DOI: 10.1089/humc.2014.154] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/26/2014] [Indexed: 12/13/2022] Open
Abstract
Large animal models of genetic diseases are rapidly becoming integral to biomedical research as technologies to manipulate the mammalian genome improve. The creation of cystic fibrosis (CF) ferrets and pigs is an example of such progress in animal modeling, with the disease phenotypes in the ferret and pig models more reflective of human CF disease than mouse models. The ferret and pig CF models also provide unique opportunities to develop and assess the effectiveness of gene and cell therapies to treat affected organs. In this review, we examine the organ disease phenotypes in these new CF models and the opportunities to test gene therapies at various stages of disease progression in affected organs. We then discuss the progress in developing recombinant replication-defective adenoviral, adeno-associated viral, and lentiviral vectors to target genes to the lung and pancreas in ferrets and pigs, the two most affected organs in CF. Through this review, we hope to convey the potential of these new animal models for developing CF gene and cell therapies.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Zoe A. Stewart
- Department of Surgery, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Patrick L. Sinn
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John C. Olsen
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jim Hu
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Paul B. McCray
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Internal Medicine, University of Iowa School of Medicine, Iowa City, IA 52242
| |
Collapse
|
28
|
Yan Z, Stewart ZA, Sinn PL, Olsen JC, Hu J, McCray, Jr. PB, Engelhardt JF. Ferret and Pig Models of Cystic Fibrosis: Prospects and Promise for Gene Therapy. HUM GENE THER CL DEV 2014. [DOI: 10.1089/hum.2014.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
29
|
Yan Z, Sun X, Evans IA, Tyler SR, Song Y, Liu X, Sui H, Engelhardt JF. Postentry processing of recombinant adeno-associated virus type 1 and transduction of the ferret lung are altered by a factor in airway secretions. Hum Gene Ther 2014; 24:786-96. [PMID: 23948055 DOI: 10.1089/hum.2013.137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
We recently created a cystic fibrosis ferret model that acquires neonatal lung infection. To develop lung gene therapies for this model, we evaluated recombinant adeno-associated virus (rAAV)-mediated gene transfer to the neonatal ferret lung. Unlike in vitro ferret airway epithelial (FAE) cells, in vivo infection of the ferret lung with rAAV1 required proteasome inhibitors to achieve efficient airway transduction. We hypothesized that differences in transduction between these two systems were because of an in vivo secreted factor that alter the transduction biology of rAAV1. Indeed, treatment of rAAV1 with ferret airway secretory fluid (ASF) strongly inhibited rAAV1, but not rAAV2, transduction of primary FAE and HeLa cells. Properties of the ASF inhibitory factor included a strong affinity for the AAV1 capsid, heat-stability, negative charge, and sensitivity to endoproteinase Glu-C. ASF-treated rAAV1 dramatically inhibited apical transduction of FAE ALI cultures (512-fold), while only reducing viral entry by 55-fold, suggesting that postentry processing of virus was influenced by the inhibitor factor. Proteasome inhibitors rescued transduction in the presence of ASF (~1600-fold) without effecting virus internalization, while proteasome inhibitors only enhanced transduction 45-fold in the absence of ASF. These findings demonstrate that a factor in lung secretions can influence intracellular processing of rAAV1 in a proteasome-dependent fashion.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242-1009, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Liu Y, Kim YJ, Ji M, Fang J, Siriwon N, Zhang LI, Wang P. Enhancing gene delivery of adeno-associated viruses by cell-permeable peptides. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:12. [PMID: 26015948 PMCID: PMC4365833 DOI: 10.1038/mtm.2013.12] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 12/08/2013] [Indexed: 12/21/2022]
Abstract
Adeno-associated virus type 2 (AAV2) is considered a promising gene delivery vector and has been extensively applied in several disease models; however, inefficient transduction in various cells and tissues has limited its widespread application in many areas of gene therapy. In this study, we have developed a general, but efficient, strategy to enhance viral transduction, both in vitro and in vivo, by incubating viral particles with cell-permeable peptides (CPPs). We show that CPPs increase internalization of viral particles into cells by facilitating both energy-independent and energy-dependent endocytosis. Moreover, CPPs can significantly enhance the endosomal escape process of viral particles, thus enhancing viral transduction to those cells that have exhibited very low permissiveness to AAV2 infection as a result of impaired intracellular viral processing. We also demonstrated that this approach could be applicable to other AAV serotypes. Thus, the membrane-penetrating ability of CPPs enables us to generate an efficient method for enhanced gene delivery of AAV vectors, potentially facilitating its applicability to human gene therapy.
Collapse
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California, USA
| | - Young Joo Kim
- Department of Physiology and Biophysics, University of Southern California , Los Angeles, California, USA
| | - Man Ji
- Department of Biochemistry and Molecular Biology, University of Southern California , Los Angeles, California, USA
| | - Jinxu Fang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California, USA
| | - Natnaree Siriwon
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California, USA
| | - Li I Zhang
- Department of Physiology and Biophysics, University of Southern California , Los Angeles, California, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California, USA ; Department of Biomedical Engineering, University of Southern California , Los Angeles, California, USA ; Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, California, USA
| |
Collapse
|
31
|
Abstract
This unit describes generation of and gene transfer to several commonly used airway models. Isolation and transduction of primary airway epithelial cells are first described. Next, the preparation of polarized airway epithelial monolayers is outlined. Transduction of these polarized cells is also described. Methods are presented for generation of tracheal xenografts, as well as both ex vivo and in vivo gene transfer to these xenografts. Finally, a method for in vivo gene delivery to the lungs of rodents is included. Methods for evaluating transgene expression are given in the support protocols.
Collapse
|
32
|
A novel chimeric adenoassociated virus 2/human bocavirus 1 parvovirus vector efficiently transduces human airway epithelia. Mol Ther 2013; 21:2181-94. [PMID: 23896725 DOI: 10.1038/mt.2013.92] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/11/2013] [Indexed: 12/30/2022] Open
Abstract
Human bocavirus virus-1 (HBoV1), a newly discovered autonomous parvovirus with a 5,500 nt genome, efficiently infects human-polarized airway epithelia (HAE) from the apical membrane. We hypothesized that the larger genome and high airway tropism of HBoV1 would be ideal for creating a viral vector for lung gene therapy. To this end, we successfully generated recombinant HBoV1 (rHBoV1) from an open reading frames-disrupted rHBoV1 genome that efficiently transduces HAE from the apical surface. We next evaluated whether HBoV1 capsids could package oversized rAAV2 genomes. These studies created a rAAV2/HBoV1 chimeric virus (5.5 kb genome) capable of apically transducing HAE at 5.6- and 70-fold greater efficiency than rAAV1 or rAAV2 (4.7-kb genomes), respectively. Molecular studies demonstrated that viral uptake from the apical surface was significantly greater for rAAV2/HBoV1 than for rAAV2 or rAAV1, and that polarization of airway epithelial cells was required for HBoV1 capsid-mediated gene transfer. Furthermore, rAAV2/HBoV1-CFTR virus containing the full-length cystic fibrosis transmembrane conductance regulator (CFTR) gene coding sequence and the strong CBA promoter efficiently corrected CFTR-dependent chloride transport in cystic fibrosis (CF) HAE. In summary, using the combined advantages of AAV and HBoV1, we have developed a novel and promising viral vector for CF lung gene therapy and also potentially HBoV1 vaccine development.
Collapse
|
33
|
Mastrangelo P, Hegele RG. RSV fusion: time for a new model. Viruses 2013; 5:873-85. [PMID: 23518574 PMCID: PMC3705301 DOI: 10.3390/v5030873] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/11/2013] [Accepted: 03/15/2013] [Indexed: 12/18/2022] Open
Abstract
In this review we propose a partially hypothetical model of respiratory syncytial virus (RSV) binding and entry to the cell that includes the recently discovered RSV receptor nucleolin, in an attempt to stimulate further inquiry in this research area. RSV binding and entry is likely to be a two-step process, the first involving the attachment of the virus to the cell membrane, which may be enhanced by electrostatic interactions with cellular glycoproteins/heparin and the viral G protein, and the second involving fusion to the cell membrane mediated by the viral F protein and a specific cellular fusion receptor. With our recent discovery of nucleolin as a functional fusion receptor for RSV, comes the possibility of a number of new approaches to the development of novel strategies for RSV prophylaxis and therapy, as well as raising some new questions concerning the pathobiology of RSV infection and tropism.
Collapse
Affiliation(s)
- Peter Mastrangelo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, M5S 1A8, Canada.
| | | |
Collapse
|
34
|
Polyak S, Mach A, Porvasnik S, Dixon L, Conlon T, Erger KE, Acosta A, Wright AJ, Campbell-Thompson M, Zolotukhin I, Wasserfall C, Mah C. Identification of adeno-associated viral vectors suitable for intestinal gene delivery and modulation of experimental colitis. Am J Physiol Gastrointest Liver Physiol 2012; 302:G296-308. [PMID: 22114116 DOI: 10.1152/ajpgi.00562.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective gene transfer with sustained gene expression is an important adjunct to the study of intestinal inflammation and future therapy in inflammatory bowel disease. Recombinant adeno-associated virus (AAV) vectors are ideal for gene transfer and long-term transgene expression. The purpose of our study was to identify optimal AAV pseudotypes for transduction of the epithelium in the small intestine and colon, which could be used for studies in experimental colitis. The tropism and transduction efficiencies of AAV pseudotypes 1-10 were examined in murine small intestine and colon 8 wk after administration by real-time PCR and immunohistochemistry. The clinical and histopathological effects of IL-10-mediated intestinal transduction delivered by AAVrh10 were examined in the murine IL-10⁻/⁻ enterocolitis model. Serum IL-10 levels and IL-10 expression were followed by ELISA and real-time PCR, respectively. AAV pseudotypes 4, 7, 8, 9, and 10 demonstrated optimal intestinal transduction. Transgene expression was sustained 8 wk after administration and was frequently observed in enteroendocrine cells. Long-term IL-10 gene expression and serum IL-10 levels were observed following AAV transduction in an IL-10-/- model of enterocolitis. Animals treated with AAVrh10-IL-10 had lower disease activity index scores, higher colon weight-to-length ratios, and lower microscopic inflammation scores. This study identifies novel AAV pseudotypes with small intestine and colon tropism and sustained transgene expression capable of modulating mucosal inflammation in a murine model of enterocolitis.
Collapse
Affiliation(s)
- Steven Polyak
- Univ. of Iowa College of Medicine, 200 Hawkins Dr., JCP4574, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Smith MK, Koch PJ, Reynolds SD. Direct and indirect roles for β-catenin in facultative basal progenitor cell differentiation. Am J Physiol Lung Cell Mol Physiol 2012; 302:L580-94. [PMID: 22227204 DOI: 10.1152/ajplung.00095.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The conducting airway epithelium is maintained and repaired by endogenous progenitor cells. Dysregulated progenitor cell proliferation and differentiation is thought to contribute to epithelial dysplasia in chronic lung disease. Thus modification of progenitor cell function is an attractive therapeutic goal and one that would be facilitated by knowledge of the molecular pathways that regulate their behavior. We modeled the human tracheobronchial epithelium using primary mouse tracheal epithelial cell cultures that were differentiated by exposure to the air-liquid-interface (ALI). A basal cell subset, termed facultative basal cell progenitors (FBP), initiate these cultures and are the progenitor for tracheal-specific secretory cells, the Clara-like cell, and ciliated cells. To test the hypothesis that β-catenin is necessary for FBP function, ALI cultures were generated from mice homozygous for the Ctnb(flox(E2-6)) allele. In this model, exons 2-6 of the β-catenin gene are flanked by LoxP sites, allowing conditional knockout of β-catenin. The β-catenin locus was modified through transduction with Adenovirus-5-encoding Cre recombinase. This approach generated a mosaic epithelium, comprised of β-catenin wild-type and β-catenin knockout cells. Dual immunostaining and quantitative histomorphometric analyses demonstrated that β-catenin played a direct role in FBP-to-ciliated cell differentiation and that it regulated cell-cell interactions that were necessary for FBP-to-Clara-like cell differentiation. β-catenin was also necessary for FBP proliferation and long-term FBP viability. We conclude that β-catenin is a critical determinant of FBP function and suggest that dysregulation of the β-catenin signaling pathway may contribute to disease pathology.
Collapse
|
36
|
Abstract
The Adeno-associated viruses (AAVs) are not associated with any diseases, and their ability to package non-genomic DNA and to transduce different cell/tissue populations has generated significant interest in understanding their basic biology in efforts to improve their utilization for corrective gene delivery. This includes their capsid structure, cellular tropism and interactions for entry, uncoating, replication, DNA packaging, capsid assembly, and antibody neutralization. The human and nonhuman primate AAVs are clustered into serologically distinct genetic clade and serotype groups, which have distinct cellular/tissue tropisms and transduction efficiencies. These properties are highly dependent upon the AAV capsid amino acid sequence, their capsid structure, and their interactions with host cell factors, including cell surface receptors, co-receptors, signaling molecules, proteins involved in host DNA replication, and host-derived antibodies. This chapter reviews the current structural information on AAV capsids and the capsid viral protein regions playing a role in the cellular interactions conferring an infective phenotype, which are then used to annotate the functional regions of the capsid. Based on the current data, the indication is that the AAVs, like other members of the Parvoviridae and other ssDNA viruses that form a T = 1 capsid, have evolved a multifunctional capsid with conserved core regions as is required for efficient capsid trafficking, capsid assembly, and genome packaging. Disparate surface loop structures confer differential receptor recognition and are involved in antibody recognition. The role of structural regions in capsid uncoating remains to be elucidated.
Collapse
Affiliation(s)
- Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA.
| | | |
Collapse
|
37
|
Martini SV, Rocco PRM, Morales MM. Adeno-associated virus for cystic fibrosis gene therapy. Braz J Med Biol Res 2011; 44:1097-104. [PMID: 21952739 DOI: 10.1590/s0100-879x2011007500123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 09/13/2011] [Indexed: 12/12/2022] Open
Abstract
Gene therapy is an alternative treatment for genetic lung disease, especially monogenic disorders such as cystic fibrosis. Cystic fibrosis is a severe autosomal recessive disease affecting one in 2500 live births in the white population, caused by mutation of the cystic fibrosis transmembrane conductance regulator (CFTR). The disease is classically characterized by pancreatic enzyme insufficiency, an increased concentration of chloride in sweat, and varying severity of chronic obstructive lung disease. Currently, the greatest challenge for gene therapy is finding an ideal vector to deliver the transgene (CFTR) to the affected organ (lung). Adeno-associated virus is the most promising viral vector system for the treatment of respiratory disease because it has natural tropism for airway epithelial cells and does not cause any human disease. This review focuses on the basic properties of adeno-associated virus and its use as a vector for cystic fibrosis gene therapy.
Collapse
Affiliation(s)
- S V Martini
- Laboratório de Fisiologia Celular e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
38
|
Hida K, Lai SK, Suk JS, Won SY, Boyle MP, Hanes J. Common gene therapy viral vectors do not efficiently penetrate sputum from cystic fibrosis patients. PLoS One 2011; 6:e19919. [PMID: 21637751 PMCID: PMC3103503 DOI: 10.1371/journal.pone.0019919] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 04/14/2011] [Indexed: 02/07/2023] Open
Abstract
Norwalk virus and human papilloma virus, two viruses that infect humans at mucosal surfaces, have been found capable of rapidly penetrating human mucus secretions. Viral vectors for gene therapy of Cystic Fibrosis (CF) must similarly penetrate purulent lung airway mucus (sputum) to deliver DNA to airway epithelial cells. However, surprisingly little is known about the rates at which gene delivery vehicles penetrate sputum, including viral vectors used in clinical trials for CF gene therapy. We find that sputum spontaneously expectorated by CF patients efficiently traps two viral vectors commonly used in CF gene therapy trials, adenovirus (d∼80 nm) and adeno-associated virus (AAV serotype 5; d∼20 nm), leading to average effective diffusivities that are ∼3,000-fold and 12,000-fold slower than their theoretical speeds in water, respectively. Both viral vectors are slowed by adhesion, as engineered muco-inert nanoparticles with diameters as large as 200 nm penetrate the same sputum samples at rates only ∼40-fold reduced compared to in pure water. A limited fraction of AAV exhibit sufficiently fast mobility to penetrate physiologically thick sputum layers, likely because of the lower viscous drag and smaller surface area for adhesion to sputum constituents. Nevertheless, poor penetration of CF sputum is likely a major contributor to the ineffectiveness of viral vector based gene therapy in the lungs of CF patients observed to date.
Collapse
Affiliation(s)
- Kaoru Hida
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Samuel K. Lai
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jung Soo Suk
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sang Y. Won
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Michael P. Boyle
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Adult Cystic Fibrosis Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Justin Hanes
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- The Center for Nanomedicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
39
|
Zhong Z, Wan Y, Han J, Shi S, Zhang Z, Sun X. Improvement of adenoviral vector-mediated gene transfer to airway epithelia by folate-modified anionic liposomes. Int J Nanomedicine 2011; 6:1083-93. [PMID: 21698075 PMCID: PMC3118681 DOI: 10.2147/ijn.s19745] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Indexed: 11/23/2022] Open
Abstract
Despite remarkable progress in the development of both viral and nonviral gene delivery vectors for airway disease treatment, poor gene transfer efficiency to the airway epithelium is a major obstacle in clinical application. To take advantage of the unique features of viral and nonviral vectors, we have developed complexes of adenovirus vector and anionic liposomes (AL-Ad5) by the calcium-induced phase change method. In the current study, based on the fact that there are overexpressed folate receptors on the surface of airway epithelia, we further modified the AL-Ad5 complexes with folate (F-AL-Ad5) to improve the transduction ability of Ad5 in airway epithelia. The transduction efficiencies of the obtained F-AL-Ad5 and AL-Ad5 complexes were assessed in primary-cultured airway epithelia in vitro. Our results indicated that compared with naked adenovirus vector, both AL-Ad5 and F-AL-Ad5 could significantly enhance the gene transduction efficiency of adenovirus vector in primary-cultured airway epithelial cells. Moreover, the enhancement mediated by F-AL-Ad5 was more dramatic than that by AL-Ad5. These results suggested that F-AL-Ad5 may be a useful strategy to deliver therapeutic genes to the airway epithelia and is promising in clinical application.
Collapse
Affiliation(s)
- Zhirong Zhong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | |
Collapse
|
40
|
Odom GL, Gregorevic P, Allen JM, Chamberlain JS. Gene therapy of mdx mice with large truncated dystrophins generated by recombination using rAAV6. Mol Ther 2011; 19:36-45. [PMID: 20859263 PMCID: PMC3017440 DOI: 10.1038/mt.2010.205] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 08/27/2010] [Indexed: 12/14/2022] Open
Abstract
Recombinant adeno-associated viral (rAAV) vector-mediated gene transfer represents a promising approach for many diseases. However, the applicability of rAAV vectors has long been hindered by the small (~4.8 kb) DNA packaging capacity. This limitation can hamper the packaging and delivery of critical regulatory elements and/or larger coding sequences, such as the ~14-kb dystrophin complementary DNA (cDNA) that is of interest for gene therapy of Duchenne muscular dystrophy (DMD). Here, we have demonstrated reconstitution of an expression cassette (7.3 kb) encoding a highly functional "minidystrophin" protein (ΔH2-R19, 222 kd) in vivo following intravascular co-delivery of two independent rAAV6 vectors sharing a central homologous recombinogenic region of 372 nucleotides. Similar to previously reported trans-splicing approaches, one rAAV vector provides the promoter with the ~1/2 initial portion of minidystrophin, while the second vector provides the remaining minidystrophin cDNA followed by the polyadenylation signal. Significantly, administering a modest dose [2 × 10(12) vector genomes (vg)] of the two minidystrophin-encoding rAAV vectors to dystrophic mice elicited an improvement of physiological performance indicative of prevention or amelioration of the disease state. These studies provide evidence that functional dystrophin transgenes larger than that typically carried by a single rAAV genome can be reconstituted in vivo by homologous recombination (HR) following intravascular co-delivery with rAAV6.
Collapse
Affiliation(s)
- Guy L Odom
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington 98195-7720, USA
| | | | | | | |
Collapse
|
41
|
Orlando C, Castellani S, Mykhaylyk O, Copreni E, Zelphati O, Plank C, Conese M. Magnetically guided lentiviral-mediated transduction of airway epithelial cells. J Gene Med 2010; 12:747-54. [PMID: 20821745 DOI: 10.1002/jgm.1494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lentiviral (LV) vectors are able to only slowly and inefficiently transduce nondividing cells such as those of the airway epithelium. To address this issue, we have exploited the magnetofection technique in in vitro models of airway epithelium. METHODS Magnetofectins were formed by noncovalent interaction between LV particles and polycation-coated iron oxide nanoparticles. Efficiency of LV-mediated transduction (as evaluated through green fluorescent protein (GFP) expression by cytofluorimetric analysis) was measured in bronchial epithelial cells in the presence or absence of a magnetic field. Cytotoxicity was evaluated by lactate dehydrogenase (LDH) release; cell monolayer integrity by measurement of transepithelial resistance (TER) and evaluation of correct zonula occludens-1 (ZO-1) localization at tight junctions (TJs) by immunofluorescence and confocal microscopy. RESULTS In nonpolarized cells, magnetofectins enhanced LV-mediated transduction at multiplicity of infection (MOI) of 50 up to 3.9-fold upon a 24-h incubation, to levels that approached those achieved at MOI of 200 for LV alone, in the presence or absence of the magnetic field. Magnetofection significantly increased the percentage of transduced cells up to 186-fold already after 15 min of incubation. In polarized cells, magnetofection increased GFP+ cells up to 24-fold compared to LV alone. Magnetofection did not enhance LDH release and slightly altered TER but not ZO-1 localization at the TJs. CONCLUSIONS We conclude that magnetofection can facilitate in vitro LV-mediated transduction of airway epithelial cells, in the absence of overt cytotoxicity and maintaining epithelial integrity, by lowering the necessary vector dose and reducing the incubation time required to achieve efficient transduction.
Collapse
Affiliation(s)
- Clara Orlando
- Institute for Experimental Treatment of Cystic Fibrosis, HS Raffaele, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Piro D, Rejman J, Conese M. Stem cell therapy for cystic fibrosis: current status and future prospects. Expert Rev Respir Med 2010; 2:365-80. [PMID: 20477199 DOI: 10.1586/17476348.2.3.365] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although cystic fibrosis (CF), an autosomal recessive disease caused by mutations in the gene encoding for the CF transmembrane conductance regulator (CFTR), seems a good candidate for gene therapy, 15 years of intense investigation and a number of clinical trials have not yet produced a viable clinical gene-therapy strategy. In addition, the duration of gene expression has been shown to be limited, only lasting 1-4 weeks. Therefore, alternative approaches involve the search for, and use of, stem cell populations. Bone marrow contains different stem cell types, including hematopoietic stem cells and multipotent mesenchymal stromal cells. Numerous studies have now demonstrated the ability of hematopoietic stem cells and mesenchymal stromal cells to home to the lung and differentiate into epithelial cells of both the conducting airways and the alveolar region. However, engraftment of bone marrow-derived stem cells into the airways is a very inefficient process. Detailed knowledge of the cellular and molecular determinants governing homing to the lung and transformation of marrow cells into lung epithelial cells would benefit this process. Despite a very low level of engraftment of donor cells into the nose and gut, significant CFTR mRNA expression and a measurable level of correction of the electrophysiological defect were observed after transplantation of wild-type marrow cells into CF mice. It is uncertain whether this effect is due to the presence of CFTR-expressing epithelial cells derived from donor cells or to the immunomodulatory role of transplanted cells. Finally, initial studies on the usefulness of umbilical cord blood and embryonic stem cells in the generation of airway epithelial cells will be discussed in this review.
Collapse
Affiliation(s)
- Donatella Piro
- Department of Biomedical Sciences, University of Foggia, c/o Ospedali Riuniti, Viale L. Pinto 1, 71100 Foggia, Italy.
| | | | | |
Collapse
|
43
|
Impact of lentiviral vector-mediated transduction on the tightness of a polarized model of airway epithelium and effect of cationic polymer polyethylenimine. J Biomed Biotechnol 2010; 2010:103976. [PMID: 20617131 PMCID: PMC2896616 DOI: 10.1155/2010/103976] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 03/31/2010] [Accepted: 05/09/2010] [Indexed: 11/17/2022] Open
Abstract
Lentiviral (LV) vectors are promising agents for efficient and long-lasting gene transfer into the lung and for gene therapy of genetically determined pulmonary diseases, such as cystic fibrosis, however, they have not been evaluated for cytotoxicity and impact on the tightness of the airway epithelium. In this study, we evaluated the transduction efficiency of a last-generation LV vector bearing Green Fluorescent Protein (GFP) gene as well as cytotoxicity and tight junction (TJ) integrity in a polarized model of airway epithelial cells. High multiplicities of infection (MOI) showed to be cytotoxic, as assessed by increase in propidium iodide staining and decrease in cell viability, and harmful for the epithelial tightness, as demonstrated by the decrease of transepithelial resistance (TER) and delocalization of occludin from the TJs. To increase LV efficiency at low LV:cell ratio, we employed noncovalent association with the polycation branched 25 kDa polyethylenimine (PEI). Transduction of cells with PEI/LV particles resulted in 2.5–3.6-fold increase of percentage of GFP-positive cells only at the highest PEI:LV ratios (1×107 PEI molecules/transducing units with 50 MOI LV) as compared to plain LV. At this dose PEI/LV transduction resulted in 6.5 ± 2.4% of propidium iodide-positive cells. On the other hand, PEI/LV particles did not determine any alteration of TER and occludin localization. We conclude that PEI may be useful for improving the efficiency of gene transfer mediated by LV vectors in airway epithelial cells, in the absence of high acute cytotoxicity and alteration in epithelial tightness.
Collapse
|
44
|
Ulrich-Vinther M. Gene therapy methods in bone and joint disorders. ACTA ORTHOPAEDICA. SUPPLEMENTUM 2010. [DOI: 10.1080/17453690610046512] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
45
|
Hida K, Won SY, Di Pasquale G, Hanes J, Chiorini JA, Ostermeier M. Sites in the AAV5 capsid tolerant to deletions and tandem duplications. Arch Biochem Biophys 2010; 496:1-8. [PMID: 20102698 PMCID: PMC2840209 DOI: 10.1016/j.abb.2010.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 01/18/2010] [Accepted: 01/20/2010] [Indexed: 01/30/2023]
Abstract
Gene therapy vectors based on adeno-associated virus (AAV) have shown much promise in clinical trials for the treatment of a variety of diseases. However, the ability to manipulate and engineer the viral surface for enhanced efficiency is necessary to overcome such barriers as pre-existing immunity and transduction of non-target cells that currently limit AAV applications. Although single amino acid changes and peptide insertions at select sites have been explored previously, the tolerance of AAV to small deletions and tandem duplications of sequence has not been globally addressed. Here, we have generated a large, diverse library of >10(5) members containing deletions and tandem duplications throughout the viral capsid of AAV5. Four unique mutants were identified that maintain the ability to form viral particles, with one showing improved transduction on both 293T and BEAS-2B cells. This approach may find potential use for the generation of novel variants with improved and altered properties or in the identification of sites that are tolerant to insertions of targeting ligands.
Collapse
Affiliation(s)
- Kaoru Hida
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218
| | - Sang Y. Won
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
| | - Giovanni Di Pasquale
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Justin Hanes
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| | - John A. Chiorini
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Marc Ostermeier
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
46
|
Castellani S, Conese M. Lentiviral vectors and cystic fibrosis gene therapy. Viruses 2010; 2:395-412. [PMID: 21994643 PMCID: PMC3185599 DOI: 10.3390/v2020395] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 01/26/2010] [Accepted: 01/27/2010] [Indexed: 01/01/2023] Open
Abstract
Cystic fibrosis (CF) is a chronic autosomic recessive syndrome, caused by mutations in the CF Transmembrane Conductance Regulator (CFTR) gene, a chloride channel expressed on the apical side of the airway epithelial cells. The lack of CFTR activity brings a dysregulated exchange of ions and water through the airway epithelium, one of the main aspects of CF lung disease pathophysiology. Lentiviral (LV) vectors, of the Retroviridae family, show interesting properties for CF gene therapy, since they integrate into the host genome and allow long-lasting gene expression. Proof-of-principle that LV vectors can transduce the airway epithelium and correct the basic electrophysiological defect in CF mice has been given. Initial data also demonstrate that LV vectors can be repeatedly administered to the lung and do not give rise to a gross inflammatory process, although they can elicit a T cell-mediated response to the transgene. Future studies will clarify the efficacy and safety profile of LV vectors in new complex animal models with CF, such as ferrets and pigs.
Collapse
Affiliation(s)
- Stefano Castellani
- Department of Biomedical Sciences, University of Foggia, Foggia, Italy; E-Mail: (S.C.)
| | - Massimo Conese
- Department of Biomedical Sciences, University of Foggia, Foggia, Italy; E-Mail: (S.C.)
| |
Collapse
|
47
|
Parrish CR. Structures and functions of parvovirus capsids and the process of cell infection. Curr Top Microbiol Immunol 2010; 343:149-76. [PMID: 20397069 DOI: 10.1007/82_2010_33] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To infect a cell, the parvovirus or adeno-associated virus (AAV) genome must be delivered from outside the plasma membrane to the nucleus, and in the process, the capsid must follow a series of binding and trafficking steps and also undergo necessary changes that result in exposure or release the ssDNA genome at the appropriate time and place within the cell. The 25 nm parvovirus capsid is comprised of two or three forms of a single protein, and although it is robust and stable, it is still sufficiently flexible to allow the exposure of several internal components at appropriate times during cell infection. The capsid can also accommodate insertion of peptides into surface loops, and capsid proteins from different viral serotypes can be shuffled to create novel functional variants. The capsids of the different viruses bind to one or more cell receptors, and for at least some viruses, the insertion of additional or alternative receptor binding sequences or structures into the capsid can expand or redirect its tropism. The infection process after cell binding involves receptor-mediated endocytosis followed by viral trafficking through the endosomal systems. That endosomal trafficking may be complex and prolonged for hours or be relatively brief. Generally only a small proportion of the particles taken up enter the cytoplasm after altering the endosomal membrane through the activity of a VP1-encoded phospholipase A2 domain that becomes released to the outside of the viral particle. Modifications to the capsid that can occur within the endosome or cytoplasm include structural changes to expose internal components, ubiquination and proteosomal processing, and possible trafficking of particles on molecular motors. It is still not clear how the genomes enter the nucleus, but nuclear pore-dependent entry of particles or permeabilization of nuclear membranes have been proposed. Those processes control the infection, pathogenesis, and host ranges of the autonomous viruses and determine the effectiveness of gene therapy using AAV capsids.
Collapse
Affiliation(s)
- Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
48
|
White AF, Mazur M, Sorscher EJ, Zinn KR, Ponnazhagan S. Genetic modification of adeno-associated viral vector type 2 capsid enhances gene transfer efficiency in polarized human airway epithelial cells. Hum Gene Ther 2009; 19:1407-14. [PMID: 18778196 DOI: 10.1089/hum.2008.117] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis (CF) is a common genetic disease characterized by defects in the expression of the CF transmembrane conductance regulator (CFTR) gene. Gene therapy offers better hope for the treatment of CF. Adeno-associated viral (AAV) vectors are capable of stable expression with low immunogenicity. Despite their potential in CF gene therapy, gene transfer efficiency by AAV is limited because of pathophysiological barriers in these patients. Although a few AAV serotypes have shown better transduction compared with the AAV2-based vectors, gene transfer efficiency in human airway epithelium has still not reached therapeutic levels. To engineer better AAV vectors for enhanced gene delivery in human airway epithelium, we developed and characterized mutant AAV vectors by genetic capsid modification, modeling the well-characterized AAV2 serotype. We genetically incorporated putative high-affinity peptide ligands to human airway epithelium on the GH loop region of AAV2 capsid protein. Six independent mutant AAV were constructed, containing peptide ligands previously reported to bind with high affinity for known and unknown receptors on human airway epithelial cells. The vectors were tested on nonairway cells and nonpolarized and polarized human airway epithelial cells for enhanced infectivity. One of the mutant vectors, with the peptide sequence THALWHT, not only showed the highest transduction in undifferentiated human airway epithelial cells but also indicated significant transduction in polarized cells. Interestingly, this modified vector was also able to infect cells independently of the heparan sulfate proteoglycan receptor. Incorporation of this ligand on other AAV serotypes, which have shown improved gene transfer efficiency in the human airway epithelium, may enhance the application of AAV vectors in CF gene therapy.
Collapse
Affiliation(s)
- April F White
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
49
|
Pinkenburg O, Vogelmeier C, Bossow S, Neubert WJ, Lutz RB, Ungerechts G, Lauer UM, Bitzer M, Bals R. RECOMBINANT SENDAI VIRUS FOR EFFICIENT GENE TRANSFER TO HUMAN AIRWAY EPITHELIUM. Exp Lung Res 2009; 30:83-96. [PMID: 14972769 DOI: 10.1080/01902140490266501] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Recombinant Sendai virus (rSeV) infects respiratory epithelial cells in animal models and cultures of undifferentiated human nasal cells. It was the aim of this study to investigate the capability of rSeV to express a transgene in human airway epithelium. Differentiated human airway epithelial cells were generated using air-liquid interface culture techniques. Application of rSeV coding for green fluorescence protein (GFP) onto the apical surface (using a multiplicity of infection of 3) resulted in expression of the transgene in more than 90% of the cells followed by decreasing numbers of positive cells during the observation time of 3 weeks. The infection of human respiratory epithelial cells is mediated by sialic acid residues at the apical surface. Despite the secretion of interleukin (IL)-8 and the replication of rSeV in the epithelial cells, the authors could not detect any cytopathic effect after the infection. In conclusion, rSeV infects differentiated human airway epithelial cells with high efficiency. Transgene expression is transient and accompanied by the secretion of an inflammatory cytokine.
Collapse
Affiliation(s)
- Olaf Pinkenburg
- Hospital of the University of Marburg, Department of Internal Medicine, Division of Pulmonology, Philipps-UniverstätMarburg, Marburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Cystic Fibrosis (CF) is an autosomal recessive disorder due to mutations in the CF transmembrane conductance regulator (CFTR) gene that lead to defective ion transport in the conducting pulmonary airways and exocrine glands. Through a process that is not fully understood, CFTR defects predispose affected patients to chronic endobronchial infections with organisms such as Pseudomonas aeruginosa and Staphylococcus aureus. Following the discovery of the CFTR gene in 1989, CF became one of the primary targets for gene therapy research. Early enthusiasm surrounded the new field of gene therapy during most of the 1990s and it led academics and clinicians on a big effort to apply gene therapy for cystic fibrosis. Clinical studies have been pursued using recombinant adenovirus, recombinant adeno-associated virus, cationic liposomes, and cationic polymer vectors. Although to this date no dramatic therapeutic benefits have been observed, a lot of information has been gained from the pre-clinical and clinical studies that were performed. This learning curve has led to the optimization of vector technology and an appreciation of immune and mechanical barriers that have to be overcome for successful delivery.
Collapse
|