1
|
In Vivo Hematopoietic Stem Cell Genome Editing: Perspectives and Limitations. Genes (Basel) 2022; 13:genes13122222. [PMID: 36553489 PMCID: PMC9778055 DOI: 10.3390/genes13122222] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
The tremendous evolution of genome-editing tools in the last two decades has provided innovative and effective approaches for gene therapy of congenital and acquired diseases. Zinc-finger nucleases (ZFNs), transcription activator- like effector nucleases (TALENs) and CRISPR-Cas9 have been already applied by ex vivo hematopoietic stem cell (HSC) gene therapy in genetic diseases (i.e., Hemoglobinopathies, Fanconi anemia and hereditary Immunodeficiencies) as well as infectious diseases (i.e., HIV), and the recent development of CRISPR-Cas9-based systems using base and prime editors as well as epigenome editors has provided safer tools for gene therapy. The ex vivo approach for gene addition or editing of HSCs, however, is complex, invasive, technically challenging, costly and not free of toxicity. In vivo gene addition or editing promise to transform gene therapy from a highly sophisticated strategy to a "user-friendly' approach to eventually become a broadly available, highly accessible and potentially affordable treatment modality. In the present review article, based on the lessons gained by more than 3 decades of ex vivo HSC gene therapy, we discuss the concept, the tools, the progress made and the challenges to clinical translation of in vivo HSC gene editing.
Collapse
|
2
|
Chatterjee S, Sivanandam V, Wong KKM. Adeno-Associated Virus and Hematopoietic Stem Cells: The Potential of Adeno-Associated Virus Hematopoietic Stem Cells in Genetic Medicines. Hum Gene Ther 2021; 31:542-552. [PMID: 32253938 DOI: 10.1089/hum.2020.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adeno-associated virus (AAV)-based vectors have transformed into powerful elements of genetic medicine with proven therapeutic efficacy and a good safety profile. Over the years, efforts to transduce hematopoietic stem cells (HSCs) with AAV2 vectors have, however, been challenging. While there was evidence that AAV2 delivered vector genomes to primitive, quiescent, multipotential, self-renewing, in vivo engrafting HSCs, transgene expression was elusive. In this study, we review the evolution of AAV transduction of HSC, starting with AAV2 vectors leading to the isolation of a family of naturally occurring AAVs from human CD34+ HSC, the AAVHSC. The stem cell-derived AAVHSCs have turned out to have remarkable potentials for genetic therapies well beyond the hematopoietic system. AAVHSCs have tropism for a wide variety of peripheral tissues, including the liver, muscle, and the retina. They cross the blood-brain barrier and transduce cells of the central nervous system. Preclinical gene therapy studies underway using AAVHSC vectors are discussed. We review the notable ability of AAVHSCs to mediate efficient, seamless homologous recombination in the absence of exogenous nuclease activity and discuss the therapeutic implications. We also discuss early results from an AAVHSC-based clinical gene therapy trial that is underway for the treatment of phenylketonuria. Thus, the stem cell-derived AAVHSC, offer a multifaceted platform for in vivo gene therapy and genome editing for the treatment of inherited diseases.
Collapse
Affiliation(s)
- Saswati Chatterjee
- Department of Surgery, Beckman Research Institute of City of Hope Medical Center, Duarte, California, USA
| | - Venkatesh Sivanandam
- Department of Surgery, Beckman Research Institute of City of Hope Medical Center, Duarte, California, USA
| | - Kamehameha Kai-Min Wong
- Department of Hematology and Stem Cell Transplantation, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
3
|
Grech L, Borg K, Borg J. Novel therapies in β-thalassaemia. Br J Clin Pharmacol 2021; 88:2509-2524. [PMID: 34004015 DOI: 10.1111/bcp.14918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 01/19/2023] Open
Abstract
Beta-thalassaemia is one of the most significant haemoglobinopathies worldwide resulting in the synthesis of little or no β-globin chains. Without treatment, β-thalassaemia major is lethal within the first decade of life due to the complex pathophysiology, which leads to wide clinical manifestations. Current clinical management for these patients depends on repeated transfusions followed by iron-chelating therapy. Several novel approaches to correct the resulting α/β-globin chain imbalance, treat ineffective erythropoiesis and improve iron overload are currently being developed. Up to now, the only curative treatment for β-thalassemia is haematopoietic stem-cell transplantation, but this is a risky and costly procedure. Gene therapy, gene editing and base editing are emerging as a powerful approach to treat this disease. In β-thalassaemia, gene therapy involves the insertion of a vector containing the normal β-globin or γ-globin gene into haematopoietic stem cells to permanently produce normal red blood cells. Gene editing and base editing involves the use of zinc finger nucleases, transcription activator-like nucleases and clustered regularly interspaced short palindromic repeats/Cas9 to either correct the causative mutation or else insert a single nucleotide variant that will increase foetal haemoglobin. In this review, we will examine the current management strategies used to treat β-thalassaemia and focus on the novel therapies targeting ineffective erythropoiesis, improving iron overload and correction of the globin chain imbalance.
Collapse
Affiliation(s)
- Laura Grech
- Centre for Molecular Medicine and Biobanking, University of Malta, Malta
| | - Karen Borg
- Department of Public Health Medicine, Ministry for Health, Malta
| | - Joseph Borg
- Centre for Molecular Medicine and Biobanking, University of Malta, Malta.,Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Malta
| |
Collapse
|
4
|
Brown N, Song L, Kollu NR, Hirsch ML. Adeno-Associated Virus Vectors and Stem Cells: Friends or Foes? Hum Gene Ther 2018; 28:450-463. [PMID: 28490211 DOI: 10.1089/hum.2017.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The infusion of healthy stem cells into a patient-termed "stem-cell therapy"-has shown great promise for the treatment of genetic and non-genetic diseases, including mucopolysaccharidosis type 1, Parkinson's disease, multiple sclerosis, numerous immunodeficiency disorders, and aplastic anemia. Stem cells for cell therapy can be collected from the patient (autologous) or collected from another "healthy" individual (allogeneic). The use of allogenic stem cells is accompanied with the potentially fatal risk that the transplanted donor T cells will reject the patient's cells-a process termed "graft-versus-host disease." Therefore, the use of autologous stem cells is preferred, at least from the immunological perspective. However, an obvious drawback is that inherently as "self," they contain the disease mutation. As such, autologous cells for use in cell therapies often require genetic "correction" (i.e., gene addition or editing) prior to cell infusion and therefore the requirement for some form of nucleic acid delivery, which sets the stage for the AAV controversy discussed herein. Despite being the most clinically applied gene delivery context to date, unlike other more concerning integrating and non-integrating vectors such as retroviruses and adenovirus, those based on adeno-associated virus (AAV) have not been employed in the clinic. Furthermore, published data regarding AAV vector transduction of stem cells are inconsistent in regards to vector transduction efficiency, while the pendulum swings far in the other direction with demonstrations of AAV vector-induced toxicity in undifferentiated cells. The variation present in the literature examining the transduction efficiency of AAV vectors in stem cells may be due to numerous factors, including inconsistencies in stem-cell collection, cell culture, vector preparation, and/or transduction conditions. This review summarizes the controversy surrounding AAV vector transduction of stem cells, hopefully setting the stage for future elucidation and eventual therapeutic applications.
Collapse
Affiliation(s)
- Nolan Brown
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , North Carolina.,2 Department of Ophthalmology, University of North Carolina at Chapel Hill , North Carolina
| | - Liujiang Song
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , North Carolina.,2 Department of Ophthalmology, University of North Carolina at Chapel Hill , North Carolina
| | - Nageswara R Kollu
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , North Carolina.,2 Department of Ophthalmology, University of North Carolina at Chapel Hill , North Carolina
| | - Matthew L Hirsch
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , North Carolina.,2 Department of Ophthalmology, University of North Carolina at Chapel Hill , North Carolina
| |
Collapse
|
5
|
Lu L, Ge Y, Li ZH, Freie B, Clapp DW, Broxmeyer HE. CD34+++ Stem/Progenitor Cells Purified from Cryopreserved Normal Cord Blood can be Transduced with High Efficiency by a Retroviral Vector and Expanded Ex Vivo with Stable Integration and Expression of Fanconi Anemia Complementation C Gene. Cell Transplant 2017; 4:493-503. [PMID: 8520833 DOI: 10.1177/096368979500400510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A future possibility for treatment of genetic diseases may be gene therapy using autologous cord blood (CB) stem/progenitor cells. This might require cryopreservation of CB stem/progenitor cells prior to purification, gene transduction, and ex vivo expansion of cells. To address this possibility, nonadherent low density T-lymphocyte depleted (NALT-) cells from fresh or cryopreserved cord blood were sorted for CD34+++ phenotype, transduced with a recombinant retroviral vector encoding Fanconi anemia complementation C (FACC) gene, and cells expanded ex vivo in suspension culture for 7 days with growth factors. The results demonstrate: 1) high recovery of viable cells after thawing; 2) high efficiency purification of CD34+++ cells from NALT- cells prior to and after cryopreservation; 3) high degree of expansion of nucleated cells and immature progenitors from CD34+++ cells before and after cryopreservation; 4) efficient transduction with stable integration and expression of newly introduced genes in cryopreserved and then sorted stem/progenitor cells, as detected prior to and after ex vivo expansion; and 5) high efficiency transduction of single isolated CD34+++ cells obtained from cryopreserved NALT- CB. This information should be of value for future studies evaluating the use of cryopreserved cord blood for gene transfer/gene therapy.
Collapse
Affiliation(s)
- L Lu
- Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis 46202, USA
| | | | | | | | | | | |
Collapse
|
6
|
Bartlett RJ, Secore SL, Singer JT, Bodo M, Sharma K, Ricordi C. Long-Term Expression of a Fluorescent Reporter Gene via Direct Injection of Plasmid Vector into Mouse Skeletal Muscle: Comparison of Human Creatine Kinase and Cmv Promoter Expression Levels in Vivo. Cell Transplant 2017; 5:411-9. [PMID: 8727010 DOI: 10.1177/096368979600500308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Expression of a fluorescent reporter gene has been studied using two alternate promoters to transcribe the green fluorescent protein (gfp) from Aequorea victoria. The human cytomegalovirus (CMV) enhancer/promoter or the human muscle-specific creatine kinase promoter (CKM) were inserted along with the gfp cDNA into a plasmid expression vector based on a modified adeno-associated virus genome. Naked plasmid DNA was injected into the hamstring muscle of mdx mice and gfp gene expression determined from frozen muscle sections taken at 4, 14, and 42 days postinjection. Fluorescence patterns obtained by photomicroscopy and quantitative fluorescence measurements indicated a near-linear increase in the accumulation of the gfp in skeletal muscle during the length of the study, with gfp expression at 42 days being roughly four times the values obtained at 4 days. The levels of expression of gfp from the CKM construct were consistantly higher than for the CMV construct. The CKM promoter/expression vector combination demonstrates significant potential for simple, direct delivery and long-term, high-level expression of genes in skeletal muscle.
Collapse
Affiliation(s)
- R J Bartlett
- Department of Neurology, University of Miami School of Medicine, FL 33136, USA.
| | | | | | | | | | | |
Collapse
|
7
|
Ling C, Bhukhai K, Yin Z, Tan M, Yoder MC, Leboulch P, Payen E, Srivastava A. High-Efficiency Transduction of Primary Human Hematopoietic Stem/Progenitor Cells by AAV6 Vectors: Strategies for Overcoming Donor-Variation and Implications in Genome Editing. Sci Rep 2016; 6:35495. [PMID: 27759036 PMCID: PMC5069717 DOI: 10.1038/srep35495] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/30/2016] [Indexed: 11/09/2022] Open
Abstract
We have reported that of the 10 commonly used AAV serotype vectors, AAV6 is the most efficient in transducing primary human hematopoietic stem/progenitor cells (HSPCs). However, the transduction efficiency of the wild-type (WT) AAV6 vector varies greatly in HSPCs from different donors. Here we report two distinct strategies to further increase the transduction efficiency in HSPCs from donors that are transduced less efficiently with the WT AAV6 vectors. The first strategy involved modifications of the viral capsid proteins where specific surface-exposed tyrosine (Y) and threonine (T) residues were mutagenized to generate a triple-mutant (Y705 + Y731F + T492V) AAV6 vector. The second strategy involved the use of ex vivo transduction at high cell density. The combined use of these strategies resulted in transduction efficiency exceeding ~90% in HSPCs at significantly reduced vector doses. Our studies have significant implications in the optimal use of capsid-optimized AAV6 vectors in genome editing in HSPCs.
Collapse
Affiliation(s)
- Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Powell Gene Therapy Center; University of Florida College of Medicine, Gainesville, FL, USA
| | - Kanit Bhukhai
- CEA/Paris Sud University (UMR-E 007), Institute of Emerging Diseases and Innovative Therapies (iMETI), Fontenay-aux-Roses, France
| | - Zifei Yin
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Powell Gene Therapy Center; University of Florida College of Medicine, Gainesville, FL, USA.,Department of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China
| | - Mengqun Tan
- Department of Physiology, Xiang-Ya School of Medicine, Central South University, Changsha, China
| | - Mervin C Yoder
- Herman B Well Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Philippe Leboulch
- CEA/Paris Sud University (UMR-E 007), Institute of Emerging Diseases and Innovative Therapies (iMETI), Fontenay-aux-Roses, France
| | - Emmanuel Payen
- CEA/Paris Sud University (UMR-E 007), Institute of Emerging Diseases and Innovative Therapies (iMETI), Fontenay-aux-Roses, France
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Powell Gene Therapy Center; University of Florida College of Medicine, Gainesville, FL, USA.,Department of Molecular Genetics &Microbiology; University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
8
|
Liu Q, Wang Y, Lin F, Zhang L, Li Y, Ge R, Hong Y. Gene transfer and genome-wide insertional mutagenesis by retroviral transduction in fish stem cells. PLoS One 2015; 10:e0127961. [PMID: 26029933 PMCID: PMC4451014 DOI: 10.1371/journal.pone.0127961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/22/2015] [Indexed: 12/15/2022] Open
Abstract
Retrovirus (RV) is efficient for gene transfer and integration in dividing cells of diverse organisms. RV provides a powerful tool for insertional mutagenesis (IM) to identify and functionally analyze genes essential for normal and pathological processes. Here we report RV-mediated gene transfer and genome-wide IM in fish stem cells from medaka and zebrafish. Three RVs were produced for fish cell transduction: rvLegfp and rvLcherry produce green fluorescent protein (GFP) and mCherry fluorescent protein respectively under control of human cytomegalovirus immediate early promoter upon any chromosomal integration, whereas rvGTgfp contains a splicing acceptor and expresses GFP only upon gene trapping (GT) via intronic in-frame integration and spliced to endogenous active genes. We show that rvLegfp and rvLcherry produce a transduction efficiency of 11~23% in medaka and zebrafish stem cell lines, which is as 30~67% efficient as the positive control in NIH/3T3. Upon co-infection with rvGTgfp and rvLcherry, GFP-positive cells were much fewer than Cherry-positive cells, consistent with rareness of productive gene trapping events versus random integration. Importantly, rvGTgfp infection in the medaka haploid embryonic stem (ES) cell line HX1 generated GTgfp insertion on all 24 chromosomes of the haploid genome. Similar to the mammalian haploid cells, these insertion events were presented predominantly in intergenic regions and introns but rarely in exons. RV-transduced HX1 retained the ES cell properties such as stable growth, embryoid body formation and pluripotency gene expression. Therefore, RV is proficient for gene transfer and IM in fish stem cells. Our results open new avenue for genome-wide IM in medaka haploid ES cells in culture.
Collapse
Affiliation(s)
- Qizhi Liu
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yunzhi Wang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Fan Lin
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Lei Zhang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yan Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ruowen Ge
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yunhan Hong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
9
|
Song L, Kauss MA, Kopin E, Chandra M, Ul-Hasan T, Miller E, Jayandharan GR, Rivers AE, Aslanidi GV, Ling C, Li B, Ma W, Li X, Andino LM, Zhong L, Tarantal AF, Yoder MC, Wong KK, Tan M, Chatterjee S, Srivastava A. Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy 2013; 15:986-98. [PMID: 23830234 PMCID: PMC3711144 DOI: 10.1016/j.jcyt.2013.04.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND AIMS Although recombinant adeno-associated virus serotype 2 (AAV2) vectors have gained attention because of their safety and efficacy in numerous phase I/II clinical trials, their transduction efficiency in hematopoietic stem cells (HSCs) has been reported to be low. Only a few additional AAV serotype vectors have been evaluated, and comparative analyses of their transduction efficiency in HSCs from different species have not been performed. METHODS We evaluated the transduction efficiency of all available AAV serotype vectors (AAV1 through AAV10) in primary mouse, cynomolgus monkey and human HSCs. The transduction efficiency of the optimized AAV vectors was also evaluated in human HSCs in a murine xenograft model in vivo. RESULTS We observed that although there are only six amino acid differences between AAV1 and AAV6, AAV1, but not AAV6, transduced mouse HSCs well, whereas AAV6, but not AAV1, transduced human HSCs well. None of the 10 serotypes transduced cynomolgus monkey HSCs in vitro. We also evaluated the transduction efficiency of AAV6 vectors containing mutations in surface-exposed tyrosine residues. We observed that tyrosine (Y) to phenylalanine (F) point mutations in residues 445, 705 and 731 led to a significant increase in transgene expression in human HSCs in vitro and in a mouse xenograft model in vivo. CONCLUSIONS These studies suggest that the tyrosine-mutant AAV6 serotype vectors are the most promising vectors for transducing human HSCs and that it is possible to increase further the transduction efficiency of these vectors for their potential use in HSC-based gene therapy in humans.
Collapse
Affiliation(s)
- Liujiang Song
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen 518057, China
| | - M. Ariel Kauss
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Etana Kopin
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Manasa Chandra
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Taihra Ul-Hasan
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Erin Miller
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Giridhara R. Jayandharan
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Haematology and Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Angela E. Rivers
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Division of Hematology/Oncology, Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - George V. Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Baozheng Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Wenqin Ma
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Xiaomiao Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Lourdes M. Andino
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Alice F. Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, and Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Mervin C. Yoder
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kamehameha K. Wong
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
- Division of Hematology/Stem Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Mengqun Tan
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen 518057, China
| | - Saswati Chatterjee
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
10
|
Shimizu A, Kobayashi N, Shimada K, Oura K, Tanaka T, Okamoto A, Kondo K. Novel gene therapy viral vector using non-oncogenic lymphotropic herpesvirus. PLoS One 2013; 8:e56027. [PMID: 23409116 PMCID: PMC3569415 DOI: 10.1371/journal.pone.0056027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 01/04/2013] [Indexed: 01/26/2023] Open
Abstract
Despite the use of retroviral vectors, efficiently introducing target genes into immunocytes such as T cells is difficult. In addition, retroviral vectors carry risks associated with the oncogenicity of the native virus and the potential for introducing malignancy in recipients due to genetic carryover from immortalized cells used during vector production. To address these issues, we have established a new virus vector that is based on human herpesvirus 6 (HHV-6), a non-oncogenic lymphotropic herpesvirus that infects CD4+ T cells, macrophages, and dendritic cells. In the present study, we have altered the cell specificity of the resulting recombinant HHV-6 by knocking out the U2–U8 genes. The resulting virus proliferated only in activated cord blood cells and not in peripheral blood cells. Umbilical cord blood cells produced replication-defective recombinant virus in sufficiently high titer to omit the use of immortalized cells during vector production. HHV-6 vectors led to high rates (>90%) of gene transduction in both CD4+ and CD8+ T cells. These viruses showed low-level replication of viral DNA that supported greater expression of the induced genes than that of other methods but that was insufficient to support the production of replication-competent virus. Furthermore, HHV-6 vectors containing short hairpin RNAs against CD4 and HIV Gag remarkably inhibited the production of these proteins and HIV particles. Here we demonstrate the utility of HHV-6 as a new non-carcinogenic viral vector for immunologic diseases and immunotherapy.
Collapse
Affiliation(s)
- Akihiro Shimizu
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nobuyuki Kobayashi
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuya Shimada
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kuniaki Oura
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tadao Tanaka
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuhiro Kondo
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
11
|
Drake AC, Chen Q, Chen J. Engineering humanized mice for improved hematopoietic reconstitution. Cell Mol Immunol 2012; 9:215-24. [PMID: 22425741 DOI: 10.1038/cmi.2012.6] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Humanized mice are immunodeficient animals engrafted with human hematopoietic stem cells that give rise to various lineages of human blood cells throughout the life of the mouse. This article reviews recent advances in the generation of humanized mice, focusing on practical considerations. We discuss features of different immunodeficient recipient mouse strains, sources of human hematopoietic stem cells, advances in expansion and genetic modification of hematopoietic stem cells, and techniques to modulate the cytokine environment of recipient mice, in order to enhance reconstitution of specific human blood lineage cells. We highlight the opportunities created by new technologies and discuss practical considerations on how to make best use of the widening array of basic models for specific research applications.
Collapse
Affiliation(s)
- Adam C Drake
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | |
Collapse
|
12
|
Zhong L, Jayandharan GR, Aslanidi GV, Zolotukhin S, Herzog RW, Srivastava A. Development of Novel Recombinant AAV Vectors and Strategies for the Potential Gene Therapy of Hemophilia. ACTA ACUST UNITED AC 2012; S1. [PMID: 23264889 DOI: 10.4172/2157-7412.s1-008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Recombinant vectors based on a non-pathogenic human parvovirus, the adeno-associated virus (AAV), have gained attention as a potentially safe and useful alternative to the more commonly used retroviral and adenoviral vectors. AAV vectors are currently in use in Phase I/II clinical trials for gene therapy of a number of diseases such as cystic fibrosis, α-1 antitrypsin deficiency, muscular dystrophy, Batten's disease, and Parkinson's disease, and have shown efficacy in patients with Leber's congenital amaurosis, and hemophilia B. For patients with hemophilia B, however, relatively large vector doses are needed to achieve therapeutic benefits. Large vector doses also trigger an immune response as significant fraction of the vectors fails to traffic efficiently to the nucleus, and is targeted for degradation by the host cell proteasome machinery. With a better understanding of the various steps in the life cycle of AAV vectors, strategies leading to the development of novel AAV vectors that are capable of high-efficiency transduction at lower doses are needed. In this review, we summarize our strategies to develop novel AAV vectors for the potential gene therapy of both hemophilia B and hemophilia A, based on our recent studies on the basic molecular biology of AAV. These strategies, including the development of novel AAV vectors by site-directed mutagenesis of critical surface-exposed tyrosine residues on AAV2 capsids to circumvent the ubiquitination step and the use of different AAV serotypes and self-complementary (sc) AAV2 vectors, and their use as helper vectors to circumvent the obstacles of second-strand DNA synthesis of single-stranded (ss) AAV, should dramatically accelerate the progress towards the potential gene therapy of both hemophilia A and hemophilia B.
Collapse
Affiliation(s)
- Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA ; Division of Hematology/Oncology, Department of Medicine, and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | | | | |
Collapse
|
13
|
Schuhmann NK, Pozzoli O, Sallach J, Huber A, Avitabile D, Perabo L, Rappl G, Capogrossi MC, Hallek M, Pesce M, Büning H. Gene transfer into human cord blood-derived CD34(+) cells by adeno-associated viral vectors. Exp Hematol 2010; 38:707-17. [PMID: 20447441 DOI: 10.1016/j.exphem.2010.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 04/08/2010] [Accepted: 04/27/2010] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Bone marrow-derived CD34(+) cells are currently used in clinical trials in patients with ischemic heart disease. An option to enhance activity of injected progenitors may be offered by genetic engineering of progenitor cells with angiogenic growth factors. Recombinant adeno-associated viral vectors (rAAV) have emerged as a leading gene transfer systems. In contrast to other vector systems in use for genetic engineering of CD34(+) cells, rAAV-mediated gene expression does not depend on vector integration. This is relevant for application in regenerative medicine of ischemic tissues, where transient transgene expression is likely sufficient to achieve therapeutic benefits. MATERIALS AND METHODS We compared three different human AAV serotypes, packaged as pseudotypes by a helper virus-free production method, for their transduction efficiency in human cord blood-derived CD34(+) cells. We further assessed the impact of vector genome conformation, of alpha(v)beta(5) and alpha(5)beta(1) integrin availability and of the transcription-modulating drugs retinoic acid and Trichostatin A on rAAV-mediated human CD34(+) cell transduction. RESULTS We provide, for the first time, evidence that hCD34(+) cells can be reproducibly transduced with high efficiency by self-complementary rAAV2 without inducing cytotoxicity or interfering with their differentiation potential. We further show the involvement of alpha(5)beta(1) integrin as a crucial AAV2 internalization receptor and a function for transcription-modulating drugs in enhancing rAAV-mediated transgene expression. CONCLUSION This study represents a first step toward translation of a combined cellular/rAAV-based therapy of ischemic disease.
Collapse
|
14
|
Abstract
Although the remarkable versatility and efficacy of recombinant adeno-associated virus 2 (AAV2) vectors in transducing a wide variety of cells and tissues in vitro, and in numerous pre-clinical animal models of human diseases in vivo, have been well established, the published literature is replete with controversies with regard to the efficacy of AAV2 vectors in hematopoietic stem cell (HSC) transduction. A number of factors have contributed to these controversies, the molecular bases of which have begun to come to light in recent years. With the availability of several novel serotypes (AAV1 through AAV12), rational design of AAV capsid mutants, and strategies (self-complementary vector genomes, hematopoietic cell-specific promoters), it is indeed becoming feasible to achieve efficient transduction of HSC by AAV vectors. Using a murine serial bone marrow transplantation model in vivo, we have recently documented stable integration of the proviral AAV genome into mouse chromosomes, which does not lead to any overt hematological abnormalities. Thus, a better understanding of the AAV-HSC interactions, and the availability of a vast repertoire of novel serotype and capsid mutant vectors, are likely to have significant implications in the use of AAV vectors in high-efficiency transduction of HSCs as well as in gene therapy applications involving the hematopoietic system.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular & Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32610-3633, USA.
| |
Collapse
|
15
|
Maina N, Zhong L, Li X, Zhao W, Han Z, Bischof D, Aslanidi G, Zolotukhin S, Weigel-Van Aken KA, Rivers AE, Slayton WB, Yoder MC, Srivastava A. Optimization of recombinant adeno-associated viral vectors for human beta-globin gene transfer and transgene expression. Hum Gene Ther 2008; 19:365-75. [PMID: 18399730 DOI: 10.1089/hum.2007.173] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Therapeutic levels of expression of the beta-globin gene have been difficult to achieve with conventional retroviral vectors without the inclusion of DNase I-hypersensitive site (HS2, HS3, and HS4) enhancer elements. We generated recombinant adeno-associated viral (AAV) vectors carrying an antisickling human beta-globin gene under the control of either the beta-globin gene promoter/enhancer or the erythroid cell-specific human parvovirus B19 promoter at map unit 6 (B19p6) without any enhancer, and tested their efficacy in a human erythroid cell line (K-562) and in primary murine hematopoietic progenitor cells (c-kit(+)lin()). We report here that (1) self-complementary AAV serotype 2 (scAAV2)-beta-globin vectors containing only the HS2 enhancer are more efficient than single-stranded AAV (ssAAV2)-beta-globin vectors containing the HS2+HS3+HS4 enhancers; (2) scAAV2-beta-globin vectors recombine with scAAV2-HS2+HS3+HS4 vectors after dual-vector transduction, leading to transgene expression; (3) scAAV2-beta-globin as well as scAAV1-beta-globin vectors containing the B19p6 promoter without the HS2 enhancer element are more efficient than their counterparts containing the HS2 enhancer/beta-globin promoter; and (4) scAAV2-B19p6-beta-globin vectors in K-562 cells, and scAAV1-B19p6-beta-globin vectors in murine c-kit(+)lin() cells, yield efficient expression of the beta-globin protein. Thus, the combined use of scAAV vectors and the parvovirus B19 promoter may lead to expression of therapeutic levels the beta-globin gene in human erythroid cells, which has implications in the use of these vectors in gene therapy of beta-thalassemia and sickle cell disease.
Collapse
Affiliation(s)
- Njeri Maina
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Maina N, Han Z, Li X, Hu Z, Zhong L, Bischof D, Weigel-Van Aken KA, Slayton WB, Yoder MC, Srivastava A. Recombinant self-complementary adeno-associated virus serotype vector-mediated hematopoietic stem cell transduction and lineage-restricted, long-term transgene expression in a murine serial bone marrow transplantation model. Hum Gene Ther 2008; 19:376-83. [PMID: 18370591 DOI: 10.1089/hum.2007.143] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although conventional recombinant single-stranded adeno-associated virus serotype 2 (ssAAV2) vectors have been shown to efficiently transduce numerous cells and tissues such as brain and muscle, their ability to transduce primary hematopoietic stem cells (HSCs) has been reported to be controversial. We have previously documented that among the ssAAV serotype 1 through 5 vectors, ssAAV1 vectors are more efficient in transducing primary murine HSCs, but that viral second-strand DNA synthesis continues to be a rate-limiting step. In the present studies, we evaluated the transduction efficiency of several novel serotype vectors (AAV1, AAV7, AAV8, and AAV10) and documented efficient transduction of HSCs in a murine serial bone marrow transplantation model. Self-complementary AAV (scAAV) vectors were found to be more efficient than ssAAV vectors, and the use of hematopoietic cell-specific enhancers/promoters, such as the human beta-globin gene DNase I-hypersensitive site 2 enhancer and promoter (HS2-betap) from the beta-globin locus control region (LCR), and the human parvovirus B19 promoter at map unit 6 (B19p6), allowed sustained transgene expression in an erythroid lineage-restricted manner in both primary and secondary transplant recipient mice. The proviral AAV genomes were stably integrated into progenitor cell chromosomal DNA, and did not lead to any overt hematological abnormalities in mice. These studies demonstrate the feasibility of the use of novel scAAV vectors for achieving high-efficiency transduction of HSCs as well as erythroid lineage-restricted expression of a therapeutic gene for the potential gene therapy of beta-thalassemia and sickle cell disease.
Collapse
Affiliation(s)
- Njeri Maina
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sellner L, Stiefelhagen M, Kleinschmidt JA, Laufs S, Wenz F, Fruehauf S, Zeller WJ, Veldwijk MR. Generation of efficient human blood progenitor-targeted recombinant adeno-associated viral vectors (AAV) by applying an AAV random peptide library on primary human hematopoietic progenitor cells. Exp Hematol 2008; 36:957-64. [PMID: 18495326 DOI: 10.1016/j.exphem.2008.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 02/27/2008] [Accepted: 03/11/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Currently standard recombinant adeno-associated virus serotype 2(rAAV2)-based vectors lack the efficiency for gene transfer into primary human CD34(+) peripheral blood progenitor cells (PBPC). MATERIALS AND METHODS An advancement in vector development now allows the generation of rAAV capsid mutants that offer higher target cell efficiency and specificity. To increase the gene transfer into hematopoietic progenitor cells, we applied this method for the first time on primary human CD34(+) PBPC cells. RESULTS On a panel of leukemia cell lines (CML/AML), significantly higher gene transfer efficiency of the rAAV capsid mutants (up to 100% gene transfer) was observed compared to standard rAAV2 vectors. A higher transduction efficiency in the imatinib-resistant cell line LAMA84-R than in their sensitive counterpart LAMA84-S and a pronounced difference in susceptibility for the capsid mutants vs rAAV2 in LAMA84-S were particularly striking. On solid tumor cell lines, on the other hand, rAAV2 was more efficient than the capsid mutants, suggesting an increased specificity of our capsid mutants for hematopoietic progenitor cells. On primary human CD34(+) PBPC significantly higher (up to eightfold; 16% green fluorescent protein-positive) gene transfer could be obtained with the newly generated vectors compared to standard rAAV2 vectors. CONCLUSION These novel vectors may enable efficient gene transfer using rAAV-based vectors into primary human blood progenitor cells for a future clinical application.
Collapse
Affiliation(s)
- Leopold Sellner
- Pharmacology of Cancer Treatment (G402), German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Han Z, Zhong L, Maina N, Hu Z, Li X, Chouthai NS, Bischof D, Weigel-Van Aken KA, Slayton WB, Yoder MC, Srivastava A. Stable Integration of Recombinant Adeno-Associated Virus Vector Genomes After Transduction of Murine Hematopoietic Stem Cells. Hum Gene Ther 2008; 19:267-78. [DOI: 10.1089/hum.2007.161] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zongchao Han
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| | - Njeri Maina
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Zhongbo Hu
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Xiaomiao Li
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Nitin S. Chouthai
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Pediatrics, Wayne State University, Detroit, MI 48201
| | - Daniela Bischof
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kirsten A. Weigel-Van Aken
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610
| | - William B. Slayton
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610
| |
Collapse
|
19
|
KELLY PATRICKF, CARRINGTON JODY, NATHWANI AMIT, VANIN ELIOF. RD114-Pseudotyped Oncoretroviral Vectors. Ann N Y Acad Sci 2006. [DOI: 10.1111/j.1749-6632.2001.tb03596.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Srivastava A. Hematopoietic stem cell transduction by recombinant adeno-associated virus vectors: problems and solutions. Hum Gene Ther 2005; 16:792-8. [PMID: 16000061 DOI: 10.1089/hum.2005.16.792] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recombinant adeno-associated virus 2 (AAV) vectors have taken center stage owing to their potentially safer profile compared with the more commonly used retroviral and adenoviral vectors in human gene therapy clinical trials. Their remarkable versatility and efficacy in a wide variety of preclinical animal models of human diseases have attracted further attention of a number of investigators. Although two particular cell types, muscle and brain, have been shown to be highly transducible by AAV vectors, controversies abound with reference to the efficacy of these vectors in transducing primary hematopoietic cells. Whereas some investigators have claimed that primitive hematopoietic cells are impervious to AAV vectors, others have reported that AAV vectors are capable of transducing these cells, but only at high vector-to-cell ratios. Still other investigators have reported successful transduction of primitive hematopoietic cells at relatively low vector-to-cell ratios. This review attempts to resolve these controversies, and provides a basis for the optimism that safe and high-efficiency transduction of hematopoietic stem and progenitor cells by AAV vectors is well within reach.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, Shands Cancer Center and Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
21
|
Srivastava A. Hematopoietic Stem Cell Transduction by Recombinant Adeno-Associated Virus Vectors: Problems and Solutions. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-81] [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
|
22
|
Ito H, Goater JJ, Tiyapatanaputi P, Rubery PT, O'Keefe RJ, Schwarz EM. Light-activated gene transduction of recombinant adeno-associated virus in human mesenchymal stem cells. Gene Ther 2004; 11:34-41. [PMID: 14681695 DOI: 10.1038/sj.gt.3302102] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Deficiencies in skeletal tissue repair and regeneration lead to conditions like osteoarthritis, osteoporosis and degenerative disc disease. While no cure for these conditions is available, the use of human bone marrow derived-mesenchymal stem cells (HuMSCs) has been shown to have potential for cell-based therapy. Furthermore, recombinant adeno-associated viruses (rAAV) could be used together with HuMSCs for in vivo or ex vivo gene therapy. Unfortunately, the poor transduction efficiency of these cells remains a significant obstacle. Here, we describe the properties of ultraviolet (UV) light-activated gene transduction (LAGT) with rAAV in HuMSCs, an advance toward overcoming this limitation. Using direct fluorescent image analysis and real-time quantitative PCR to evaluate enhanced green fluorescent protein (eGFP) gene expression, we found that the optimal effects of LAGT with limited cytotoxicity occurred at a UV dose of 200 J/m(2). Furthermore, this UV irradiation had no effect on either the chondrogenic or osteogenic potential of HuMSCs. Significant effects of LAGT in HuMSCs could be detected as early as 12 h after exposure and persisted over 21 days, in a time and energy-dependent manner. This LAGT effect was maintained for more than 8 h after irradiation and required only a 10-min exposure to rAAV after UV irradiation. Finally, we show that the production of secreted TGFbeta1 protein from rAAV-TGFbeta1-IRES-eGFP infected to HuMSCs is highly inducible by UV irradiation. These results demonstrate that LAGT combined with rAAV is a promising procedure to facilitate gene induction in HuMSCs for human gene therapy.
Collapse
Affiliation(s)
- H Ito
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
23
|
Srivastava A. Obstacles to human hematopoietic stem cell transduction by recombinant adeno-associated virus 2 vectors. J Cell Biochem 2002; 38:39-45. [PMID: 12046848 DOI: 10.1002/jcb.10053] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Recombinant adeno-associated virus 2 (AAV) vectors have proven to be a potentially useful alternative to the more commonly used retroviral and adenoviral vectors for gene therapy in humans. Their safety and efficacy in Phase I clinical trials for gene therapy of cystic fibrosis and hemophilia B have been well documented, and their remarkable versatility and efficacy in a wide variety of pre-clinical models of human diseases have catapulted these vectors to the forefront. AAV vectors have been shown to be particularly well suited for transduction of brain and muscle cells. However, controversies exist with regard to their utility as a vector for gene transfer into human hematopoietic stem cells. On the one hand, some investigators have concluded that AAV vectors do not transduce hematopoietic stem cells at all, and others have reported that stem cell transduction requires enormously high vector-to-cell ratios. On the other hand, some investigators have reported high-efficiency transduction of human hematopoietic stem cells at low vector-to cell ratios. This article will provide a historical perspective as well as attempt to elaborate the reasons behind these controversies which have become clearer by studies focused on understanding, at the molecular level, the fundamental aspects of the life cycle of recombinant AAV vectors.
Collapse
Affiliation(s)
- Arun Srivastava
- Department of Microbiology & Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis 46202-5120, USA.
| |
Collapse
|
24
|
Chiriva-Internati M, Liu Y, Salati E, Zhou W, Wang Z, Grizzi F, Roman JJ, Lim SH, Hermonat PL. Efficient generation of cytotoxic T lymphocytes against cervical cancer cells by adeno-associated virus/human papillomavirus type 16 E7 antigen gene transduction into dendritic cells. Eur J Immunol 2002; 32:30-38. [PMID: 11754001 DOI: 10.1002/1521-4141(200201)32:1<30::aid-immu30>3.0.co;2-e] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adeno-associated virus (AAV) is able to efficiently deliver a cytokine gene into dendritic cells (DC). Improvements in T cell priming by DC might be effected by the delivery of antigen genes into DC, resulting in continuous protein expression, as most proteins have short half-lives. In this study, a recombinant AAV vector containing the human papillomavirus (HPV)-16 E7 gene was used to pulse/infect DC and compared to the pulsing of DC by the lipofection of bacterially produced E7 protein. Pulsing of DC with AAV/antigen (Ag) gene was found to be superior to pulsing with protein in six different assay systems: (1) the level of antigen transfer into DC as determined by intracellular staining; (2) the level of MHC class I-restricted killing in cytotoxic T lymphocyte (CTL) assays; (3) the level of IFN-gamma expression; (4) the level of DC-T cell priming clusters generated; (5) the level of CD80 and CD83 expression on DC; and (6) in the resulting CD8:CD4 ratio. Finally, AAV/Ag gene pulsing resulted in strong CTL activity after only 7 days of priming. These data suggest that AAV vectors may offer advantages over the commonly used protein-pulsing technique and that AAV vectors may be useful for the stimulation of CTL activity and adoptive immunotherapy protocols.
Collapse
|
25
|
Liu Y, Chiriva-Internati M, Grizzi F, Salati E, Roman JJ, Lim S, Hermonat PL. Rapid induction of cytotoxic T-cell response against cervical cancer cells by human papillomavirus type 16 E6 antigen gene delivery into human dendritic cells by an adeno-associated virus vector. Cancer Gene Ther 2001; 8:948-957. [PMID: 11781657 DOI: 10.1038/sj.cgt.7700391] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2001] [Indexed: 11/09/2022]
Abstract
We have shown that the pulsing of dendritic cells (DCs) with human papillomavirus type 16 (HPV-16) antigen proteins by lipofection stimulates class I-restricted cytotoxic T lymphocyte (CTL) response against primary cervical cancer cells. Also, we have shown that adeno-associated virus (AAV) was able to effectively deliver a cytokine gene into DCs. It has been our hypothesis that the delivery of antigen genes into DCs, resulting in endogenous and continuous antigen protein expression, may result in an improvement in T-cell priming by DCs. Here, DCs are pulsed (infected) with an AAV vector containing the HPV-16 E6 gene. After infection, transduced E6 gene mRNA expression and vector chromosomal integration could be identified in infected DCs. Furthermore, priming rosettes formed at early times when the AAV/E6 vector was used. Most importantly, AAV/E6 vector pulsing of DCs induced, after only 7 days of priming, a strong CTL response against primary cervical cancer cell lines, compared to bacterial E6 protein lipofection. Killing was significantly blocked by the addition of anti-MHC class I antibodies. Fluorescence-activated cell sorter (FACS) analysis of resulting primed cell populations revealed higher levels of CD8+ T cells by AAV-based pulsing, with little evidence of CD56 (NK). FACS analysis of the DC populations revealed that AAV/E6 vector-pulsed DCs had higher levels of CD80 and lower levels of CD86 than protein-pulsed DCs. These data suggest that rAAV may be appropriate for antigen pulsing of DCs for immunotherapy protocols. Finally, our protocol represents an advance in regards to the time needed for generating a CTL response compared to other techniques.
Collapse
Affiliation(s)
- Y Liu
- Departments of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Kogure K, Urabe M, Mizukami H, Kume A, Sato Y, Monahan J, Ozawa K. Targeted integration of foreign DNA into a defined locus on chromosome 19 in K562 cells using AAV-derived components. Int J Hematol 2001; 73:469-475. [PMID: 11503961 DOI: 10.1007/bf02994009] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Targeted integration of foreign DNA is ideal for gene therapy, particularly when target cells such as hematopoietic cells actively divide and proliferate. Adeno-associated virus (AAV) has been shown to integrate its genome into a defined locus, AAVS1 (19q13.3-qter). The inverted terminal repeat (ITR) and Rep proteins are responsible for this site-specific integration, and a system has been developed that delivers a gene preferentially into AAVS1 by using these components of AAV. We examined whether this system could be applied to gene transfer into K562 cells. Two rep expression plasmids were tested, 1 driven by the cytomegalovirus (CMV) promoter (pCMVR78) and the other under the translational control of an internal ribosome entry site (pMGiR78) with mouse mammary tumor virus promoter. K562 cells were cotransfected with a rep plasmid and a plasmid containing a neo gene flanked by the ITRs. G418-resistant clones were isolated and analyzed by Southern blot analysis and fluorescence in situ hybridization (FISH). Southern blot analysis suggested AAVS1-specific integration of the neo gene in 6 (35%) of 17 clones when K562 cells were transfected with pMGiR78 by lipofection. FISH located the neo gene on chromosome 19 in 5 of these 6 clones (29%). Eight (32%) of 25 clones obtained by electroporation with pCMVR78 had the neo gene at AAVS1, according to Southern blot analysis, and 4 of these 8 clones (16%) were positive according to FISH analysis. These results suggest that site-specific integration of foreign DNA can be achieved at a significantly high rate in human hematopoietic cells using the AAV components.
Collapse
Affiliation(s)
- Katsuhiro Kogure
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi
- CREST, Japan Science and Technology Corporation, Tochigi
| | - Masashi Urabe
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi
- CREST, Japan Science and Technology Corporation, Tochigi
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi
- CREST, Japan Science and Technology Corporation, Tochigi
| | - Akihiro Kume
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi
- CREST, Japan Science and Technology Corporation, Tochigi
| | - Yuko Sato
- Department of Intractable Diseases, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | | | - Keiya Ozawa
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi.
- CREST, Japan Science and Technology Corporation, Tochigi.
- Division of Hematology, Department of Medicine, Jichi Medical School, Tochigi, Japan.
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, 3311-1 Yakushiji, Minami-kawachi, 329-0498, Tochigi, Japan.
| |
Collapse
|
27
|
Wagner T, Fritsch G, Thalhammer R, Höcker P, Lanzer G, Lechner K, Geissler K. IL-10 increases the number of CFU-GM generated by ex vivo expansion of unmanipulated human MNCs and selected CD34+ cells. Transfusion 2001; 41:659-66. [PMID: 11346703 DOI: 10.1046/j.1537-2995.2001.41050659.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Ex vivo expansion strategies with different cytokine combinations are currently used by several groups as a means of increasing the number of HPCs for a variety of special clinical applications. Because there is little information on the potential role of IL-10 in such ex vivo expansion models, the effect of this cytokine on the generation of myeloid progenitor cells in suspension cultures was investigated. STUDY DESIGN AND METHODS On the basis of data from the literature and from new experiments, the combination of SCF and IL-3 at concentrations of 100 ng per mL and 100 U per mL, respectively, was chosen as the standard cocktail. The addition of IL-10 to such cultures resulted in a marked and dose-dependent potentiation of myeloid progenitor cell production. RESULTS Using unmanipulated leukapheresis components from 13 individuals (including lymphoma and cancer patients and normal donors), the expansion multiple of CFU-GM after 14 days as compared with pre-expansion values was 9.54 +/- 2.31 times by SCF/IL-3 and 46.38 +/- 7.37 times by the combination of SCF/IL-3 and 100 ng per mL of IL-10 (p<0.001). IL-10 also potentiated CFU-GM generation from selected CD34 PBMNCs (n = 9) with an expansion of 17.22 +/- 7.04 times versus 45.67 +/- 16.78 times using the SCF/IL-3 and SCF/IL-3/IL-10 combination, respectively (p<0.05). Moreover, expansion-promoting effects of IL-10 were observed in liquid cultures containing MNCs from bone marrow (n = 4) and cord blood (n = 3), but did not reach statistical significance because of the small number of samples. CONCLUSION These results suggest IL-10 as a useful cytokine to optimize progenitor cell-expansion strategies for clinical application.
Collapse
Affiliation(s)
- T Wagner
- Department of Blood Group Serology and Transfusion Medicine, University Clinics of Graz, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
28
|
Prasad KM, Yang Z, Bleich D, Nadler JL. Adeno-associated virus vector mediated gene transfer to pancreatic beta cells. Gene Ther 2000; 7:1553-61. [PMID: 11021593 DOI: 10.1038/sj.gt.3301279] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Insulin-dependent diabetes mellitus (IDDM) or type 1 diabetes is an autoimmune disease that results in destruction of the insulin-producing pancreatic islet beta cells. Several factors induce the invasion of immune cells into islets and trigger inflammation. Gene therapy approaches targeting the islet cells could be an effective treatment to prevent the onset or reverse type 1 diabetes. Allogeneic islet transplantation provides short-term treatment. However, genetically modified islets, which resist the host immune response, could provide long-term solutions. Adeno-associated virus (AAV) is emerging as a prominent vector system for delivering therapeutic genes for human gene therapy. AAV vector can transduce nondividing cells and provide long-term gene expression by integrating into host chromosome. Therefore, it is an appropriate vector system for islet cell gene therapy. To test the efficacy of AAV vector to transduce pancreatic endocrine cells, we constructed AAV vectors using plasmid pSub201. Wild-type AAV DNA analogue from plasmid psub201 was subcloned into a cloning plasmid pSP72 and AAV vectors were constructed by inserting the transgenes with heterologous promoter in place of AAV open reading frames (rep and cap). In this report we demonstrate the transduction of pancreatic islet cells with AAV vectors encoding bacterial -galactosidase enzyme or enhanced green fluorescent protein (EGFP) as reporter gene. Dispersed porcine and rat islet cells can be transduced by AAV vector, with an efficiency of 47% and 38%, respectively. In particular porcine islet insulin producing beta cells were transduced with an efficiency of 39%. Intact rat islet cells were transduced with an efficiency of 26% as estimated by FACS analysis following transduction with an AAV vector encoding EGFP. Transduction of intact rat islets with an AAV vector did not alter glucose-induced insulin secretion. AAV vector transduction was higher in transformed islet cell lines INS-1 and RIN m5F with an efficiency of 65% and 57%, respectively. These new results suggest that AAV vectors will provide an improved method of gene delivery to pancreatic islets and isolated pancreatic beta cells.
Collapse
Affiliation(s)
- K M Prasad
- Division of Endocrinology and Metabolism, University of Virginia Health Science Center, Charlottesville 22908-1405, USA
| | | | | | | |
Collapse
|
29
|
Wu P, Xiao W, Conlon T, Hughes J, Agbandje-McKenna M, Ferkol T, Flotte T, Muzyczka N. Mutational analysis of the adeno-associated virus type 2 (AAV2) capsid gene and construction of AAV2 vectors with altered tropism. J Virol 2000; 74:8635-47. [PMID: 10954565 PMCID: PMC102071 DOI: 10.1128/jvi.74.18.8635-8647.2000] [Citation(s) in RCA: 297] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adeno-associated virus type 2 (AAV2) has proven to be a valuable vector for gene therapy. Characterization of the functional domains of the AAV capsid proteins can facilitate our understanding of viral tissue tropism, immunoreactivity, viral entry, and DNA packaging, all of which are important issues for generating improved vectors. To obtain a comprehensive genetic map of the AAV capsid gene, we have constructed 93 mutants at 59 different positions in the AAV capsid gene by site-directed mutagenesis. Several types of mutants were studied, including epitope tag or ligand insertion mutants, alanine scanning mutants, and epitope substitution mutants. Analysis of these mutants revealed eight separate phenotypes. Infectious titers of the mutants revealed four classes. Class 1 mutants were viable, class 2 mutants were partially defective, class 3 mutants were temperature sensitive, and class 4 mutants were noninfectious. Further analysis revealed some of the defects in the class 2, 3, and 4 mutants. Among the class 4 mutants, a subset completely abolished capsid formation. These mutants were located predominantly, but not exclusively, in what are likely to be beta-barrel structures in the capsid protein VP3. Two of these mutants were insertions at the N and C termini of VP3, suggesting that both ends of VP3 play a role that is important for capsid assembly or stability. Several class 2 and 3 mutants produced capsids that were unstable during purification of viral particles. One mutant, R432A, made only empty capsids, presumably due to a defect in packaging viral DNA. Additionally, five mutants were defective in heparan binding, a step that is believed to be essential for viral entry. These were distributed into two amino acid clusters in what is likely to be a cell surface loop in the capsid protein VP3. The first cluster spanned amino acids 509 to 522; the second was between amino acids 561 and 591. In addition to the heparan binding clusters, hemagglutinin epitope tag insertions identified several other regions that were on the surface of the capsid. These included insertions at amino acids 1, 34, 138, 266, 447, 591, and 664. Positions 1 and 138 were the N termini of VP1 and VP2, respectively; position 34 was exclusively in VP1; the remaining surface positions were located in putative loop regions of VP3. The remaining mutants, most of them partially defective, were presumably defective in steps of viral entry that were not tested in the preliminary screening, including intracellular trafficking, viral uncoating, or coreceptor binding. Finally, in vitro experiments showed that insertion of the serpin receptor ligand in the N-terminal regions of VP1 or VP2 can change the tropism of AAV. Our results provide information on AAV capsid functional domains and are useful for future design of AAV vectors for targeting of specific tissues.
Collapse
Affiliation(s)
- P Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32610-0266, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The efficient delivery of therapeutic genes and appropriate gene expression are the crucial issues for clinically relevant gene therapy. Viruses are naturally evolved vehicles which efficiently transfer their genes into host cells. This ability made them desirable for engineering virus vector systems for the delivery of therapeutic genes. The viral vectors recently in laboratory and clinical use are based on RNA and DNA viruses processing very different genomic structures and host ranges. Particular viruses have been selected as gene delivery vehicles because of their capacities to carry foreign genes and their ability to efficiently deliver these genes associated with efficient gene expression. These are the major reasons why viral vectors derived from retroviruses, adenovirus, adeno-associated virus, herpesvirus and poxvirus are employed in more than 70% of clinical gene therapy trials worldwide. Among these vector systems, retrovirus vectors represent the most prominent delivery system, since these vectors have high gene transfer efficiency and mediate high expression of therapeutic genes. Members of the DNA virus family such as adenovirus-, adeno-associated virus or herpesvirus have also become attractive for efficient gene delivery as reflected by the fast growing number of clinical trials using these vectors. The first clinical trials were designed to test the feasibility and safety of viral vectors. Numerous viral vector systems have been developed for ex vivo and in vivo applications. More recently, increasing efforts have been made to improve infectivity, viral targeting, cell type specific expression and the duration of expression. These features are essential for higher efficacy and safety of RNA- and DNA-virus vectors. From the beginning of development and utilisation of viral vectors it was apparent that they harbour risks such as toxicities, immunoresponses towards viral antigens or potential viral recombination, which limit their clinical use. However, many achievements have been made in vector safety, the retargeting of virus vectors and improving the expression properties by refining vector design and virus production. This review addresses important issues of the current status of viral vector design and discusses their key features as delivery systems in gene therapy of human inherited and acquired diseases at the level of laboratory developments and of clinical applications.
Collapse
Affiliation(s)
- W Walther
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | | |
Collapse
|
31
|
Handa A, Muramatsu SI, Qiu J, Mizukami H, Brown KE. Adeno-associated virus (AAV)-3-based vectors transduce haematopoietic cells not susceptible to transduction with AAV-2-based vectors. J Gen Virol 2000; 81:2077-2084. [PMID: 10900047 DOI: 10.1099/0022-1317-81-8-2077] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although adeno-associated virus (AAV)-2 has a broad tissue-host range and can transduce a wide variety of tissue types, some cells, such as erythro-megakaryoblastoid cells, are non-permissive and appear to lack the AAV-2 receptor. However, limited studies have been reported with the related dependovirus AAV-3. We have previously cloned this virus, characterized its genome and produced an infectious clone. In this study, the gene for green fluorescent protein (GFP) was inserted into AAV-2- and AAV-3-based plasmids and recombinant viruses were produced. These viruses were then used to transduce haematopoietic cells and the transduction efficiencies were compared. In contrast to recombinant (r) AAV-2, rAAV-3 successfully transduced erythroid and megakaryoblastoid cells, although rAAV-2 was superior in transduction of lymphocyte-derived cell lines. Recently, it was reported that heparan sulphate can act as a receptor of AAV-2. The infectivity of rAAV-2 and rAAV-3 was tested with mutant cell lines of Chinese hamster ovary cells that were defective for heparin or heparan sulphate expression on the cell surface. There was no correlation between the ability of rAAV-2 or rAAV-3 to infect cells and the cell surface expression of heparan sulphate and, although heparin blocked both rAAV-2 and rAAV-3 transduction, the ID(50) of rAAV-3 was higher than that of rAAV-2. In addition, virus-binding overlay assays indicated that AAV-2 and AAV-3 bound different membrane proteins. These results suggest not only that there are different cellular receptors for AAV-2 and AAV-3, but that rAAV-3 vectors may be preferred for transduction of some haematopoietic cell types.
Collapse
Affiliation(s)
- Atsushi Handa
- Hematology Branch, National Heart, Lung and Blood Institute, Bldg 10/Rm 7C218, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-1652, USA1
| | - Shin-Ichi Muramatsu
- Hematology Branch, National Heart, Lung and Blood Institute, Bldg 10/Rm 7C218, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-1652, USA1
| | - Jianming Qiu
- Hematology Branch, National Heart, Lung and Blood Institute, Bldg 10/Rm 7C218, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-1652, USA1
| | - Hiroaki Mizukami
- Hematology Branch, National Heart, Lung and Blood Institute, Bldg 10/Rm 7C218, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-1652, USA1
| | - Kevin E Brown
- Hematology Branch, National Heart, Lung and Blood Institute, Bldg 10/Rm 7C218, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-1652, USA1
| |
Collapse
|
32
|
Nathwani AC, Hanawa H, Vandergriff J, Kelly P, Vanin EF, Nienhuis AW. Efficient gene transfer into human cord blood CD34+ cells and the CD34+CD38- subset using highly purified recombinant adeno-associated viral vector preparations that are free of helper virus and wild-type AAV. Gene Ther 2000; 7:183-95. [PMID: 10694794 DOI: 10.1038/sj.gt.3301068] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recombinant adeno-associated viral (rAAV) vectors have been evaluated for their ability to transduce primitive hematopoietic cells. Early studies documented rAAV-mediated gene expression during progenitor derived colony formation in vitro, but studies examining genome integration and long-term gene expression in hematopoietic cells have yielded conflicting results. Such studies were performed with crude vector preparations. Using improved methodology, we have generated high titer, biologically active preparations of rAAV free of wild-type AAV (less than 1/107particles) and adenovirus. Transduction of CD34+ cells from umbilical cord blood was evaluated with a bicistronic rAAV vector encoding the green fluorescent protein (GFP) and a trimetrexate resistant variant of dihydrofolate reductase (DHFR). Freshly isolated, quiescent CD34+ cells were resistant to transduction (less than 4%), but transduction increased to 23 +/- 2% after 2 days of cytokine stimulation and was further augmented by addition of tumor necrosis factor alpha (51 +/- 4%) at a multiplicity of infection of 106. rAAV-mediated gene expression was transient in that progenitor derived colony formation was inhibited by trimetrexate. Primitive CD34+ and CD34+, CD38- subsets were sequentially transduced with a rAAV vector encoding the murine ecotropic receptor followed by transduction with an ecotropic retroviral vector encoding GFP and DHFR. Under optimal conditions 41 +/- 7% of CD34+ progenitors and 21 +/- 6% of CD34+, CD38- progenitors became trimetrexate resistant. These results document that highly purified rAAV transduce primitive human hematopoietic cells efficiently but gene expression appears to be transient. Gene Therapy (2000) 7, 183-195.
Collapse
Affiliation(s)
- A C Nathwani
- Division of Experimental Hematology, Department of Hematology/Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | |
Collapse
|
33
|
Intraneuronal aggregate formation and cell death after viral expression of expanded polyglutamine tracts in the adult rat brain. J Neurosci 2000. [PMID: 10627599 DOI: 10.1523/jneurosci.20-01-00219.2000] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Expanded polyglutamine (polyQ) tracts have been linked to a new class of human disease characterized by psychiatric/motor syndromes associated with specific patterns of neurodegeneration. We have used a direct viral approach to locally express expanded polyglutamine tracts fused to the green fluorescent protein (97Q-GFP) in the adult rat brain. We show that intrastriatal expression of 97Q-GFP causes the rapid formation of fibrillar, cytoplasmic, and ubiquitinated nuclear aggregates in neurons. 97Q-GFP expression also results in a specific temporal pattern of cell death in the striatum. Co-infection studies suggest that high level 97Q-GFP-expressing cells die during the first month, whereas low level 97Q-GFP-expressing neurons persist for up to 6 months after infection. These data indicate that cumulative expression of polyQ repeats throughout the life of the animal is not required to induce neuronal death, but rather acute overexpression of polyQ is toxic to adult neurons in vivo.
Collapse
|
34
|
Senut MC, Suhr ST, Kaspar B, Gage FH. Intraneuronal aggregate formation and cell death after viral expression of expanded polyglutamine tracts in the adult rat brain. J Neurosci 2000; 20:219-29. [PMID: 10627599 PMCID: PMC6774116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/1999] [Revised: 10/06/1999] [Accepted: 10/14/1999] [Indexed: 02/15/2023] Open
Abstract
Expanded polyglutamine (polyQ) tracts have been linked to a new class of human disease characterized by psychiatric/motor syndromes associated with specific patterns of neurodegeneration. We have used a direct viral approach to locally express expanded polyglutamine tracts fused to the green fluorescent protein (97Q-GFP) in the adult rat brain. We show that intrastriatal expression of 97Q-GFP causes the rapid formation of fibrillar, cytoplasmic, and ubiquitinated nuclear aggregates in neurons. 97Q-GFP expression also results in a specific temporal pattern of cell death in the striatum. Co-infection studies suggest that high level 97Q-GFP-expressing cells die during the first month, whereas low level 97Q-GFP-expressing neurons persist for up to 6 months after infection. These data indicate that cumulative expression of polyQ repeats throughout the life of the animal is not required to induce neuronal death, but rather acute overexpression of polyQ is toxic to adult neurons in vivo.
Collapse
Affiliation(s)
- M C Senut
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
35
|
Kurpad C, Mukherjee P, Wang XS, Ponnazhagan S, Li L, Yoder MC, Srivastava A. Adeno-associated virus 2-mediated transduction and erythroid lineage-restricted expression from parvovirus B19p6 promoter in primary human hematopoietic progenitor cells. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 1999; 8:585-92. [PMID: 10645765 DOI: 10.1089/152581699319740] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Human parvovirus B19 gene expression from the viral p6 promoter (B19p6) is restricted to primary human hematopoietic cells undergoing erythroid differentiation. We have demonstrated that expression from this promoter does not occur in established human erythroid cell lines in the context of a recombinant parvovirus genome (Ponnazhagan et al. J Virol 69:8096-8101, 1995). However, abundant expression from this promoter can be readily detected in primary human bone marrow cells (Wang et al. Proc Natl Acad Sci USA 92:12416-12420, 1995; Ponnazhagan et al. J Gen Virol 77:1111-1122, 1996). In the present studies, we investigated the pattern of expression from the B19p6 promoter in primary human bone marrow-derived CD34+ HPC undergoing differentiation into myeloid and erythroid lineages. CD34+ cells were transduced with recombinant adeno-associated virus 2 (AAV) vectors containing the beta-galactosidase (lacZ) gene under the control of the following promoters/enhancers: the cytomegalovirus promoter (vCMVp-lacZ), B19p6 promoter (vB19p6-lacZ), B19p6 promoter with an upstream erythroid cell-specific enhancer element (HS-2) from the locus control region (LCR) from the human beta-globin gene cluster (vHS2-B19p6-lacZ), and the human beta-globin gene promoter with the HS-2 enhancer (vHS2-beta p-lacZ). Transgene expression was evaluated either 48 h after infection or following erythroid differentiation in vitro for 3 weeks. Whereas high-level expression from the CMV promoter 48 h after infection diminished with time, low-level expression from the B19p6 and the beta-globin promoters increased significantly following erythroid differentiation. Furthermore, in HPC assays, there was no significant difference in the level of expression from the CMV promoter in myeloid or erythroid cell-derived colonies. Expression from the B19p6 and the beta-globin promoters, on the other hand, was restricted to erythroid cell colonies. These data further corroborate that the B19p6 promoter is erythroid cell-specific and suggest that the recombinant AAV-B19 hybrid vectors may prove useful in gene therapy of human hemoglobinopathies in general and sickle cell anemia and beta-thalassemia in particular.
Collapse
MESH Headings
- Anemia, Sickle Cell/genetics
- Anemia, Sickle Cell/therapy
- Antigens, CD34/analysis
- Cells, Cultured
- Colony-Forming Units Assay
- Cytomegalovirus/genetics
- Dependovirus/genetics
- Enhancer Elements, Genetic
- Erythroid Precursor Cells/metabolism
- Erythroid Precursor Cells/virology
- Erythropoiesis/genetics
- Flow Cytometry
- Gene Expression Regulation, Developmental
- Genes, Reporter
- Genes, Viral
- Genetic Therapy
- Genetic Vectors/genetics
- Globins/genetics
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/virology
- Humans
- Lac Operon
- Organ Specificity
- Parvovirus B19, Human/genetics
- Promoter Regions, Genetic
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Transfection
- beta-Galactosidase/biosynthesis
- beta-Galactosidase/genetics
- beta-Thalassemia/genetics
- beta-Thalassemia/therapy
Collapse
Affiliation(s)
- C Kurpad
- Department of Microbiology & Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis 46202-5120, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Hartung SD, Reddy RG, Whitley CB, McIvor RS. Enzymatic correction and cross-correction of mucopolysaccharidosis type I fibroblasts by adeno-associated virus-mediated transduction of the alpha-L-iduronidase gene. Hum Gene Ther 1999; 10:2163-72. [PMID: 10498248 DOI: 10.1089/10430349950017158] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I), a deficiency in the lysosomal enzyme alpha-L-iduronidase (IDUA), is characterized by skeletal abnormalities, hepatosplenomegaly and neurological dysfunction. To evaluate the potential for treatment of the disease using a gene delivery approach, recombinant adeno-associated virus (rAAV) vectors were constructed and evaluated for expression of the human IDUA cDNA in transduced cells. 293 cells transduced with these AAV vectors contained IDUA activity at 0.5 to 1.4 micromol/mg x hr, 50- to 140-fold above background (control-transduced) levels. In time course studies of transduced 293 cells, IDUA activity levels peaked 1 week after transduction and persisted at 50% of the peak level for at least 6 weeks. Transduced MPS I fibroblasts also expressed high levels of IDUA activity (114-290 nmol/mg x hr), which persisted for at least 3 weeks in the absence of selection. In addition, transduced MPS I fibroblasts were capable of clearing intracellular radiolabeled glycosaminoglycan (GAG). As a test of the ability of these vectors to mediate metabolic cross-correction, transduced HuH7 human hepatoma cells were demonstrated to release enzyme that was subsequently taken up by nontransduced MPS I fibroblasts. These results illustrate the effectiveness of AAV vectors for delivery and expression of human IDUA gene sequences and for potential treatment of MPS I.
Collapse
Affiliation(s)
- S D Hartung
- Institute of Human Genetics, Department of Laboratory Medicine, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
37
|
Romano G, Pacilio C, Giordano A. Gene transfer technology in therapy: current applications and future goals. Stem Cells 1999; 17:191-202. [PMID: 10437982 DOI: 10.1002/stem.170191] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Gene therapy has attracted much interest since the first submissions of phase I clinical trials in the early 1990s, for the treatment of inherited genetic diseases. Preliminary results were very encouraging and prompted many investigators to submit protocols for phase I and phase II clinical trials for the treatment of inherited genetic diseases and cancer. The possible application of gene transfer technology to treat AIDS, cardiopathies, and neurologic diseases is under evaluation. Some viral vectors have already been used to deliver HIV-1 subunits to immunize volunteers who are participating in the AIDS vaccine programs in the USA. However, gene delivery systems still need to be optimized in order to achieve effective therapeutic interventions. The purpose of this review is to summarize the latest achievements in improving gene delivery systems, their current application in preclinical studies and in therapy, and the most pressing issues that must be addressed in the area of vector design.
Collapse
Affiliation(s)
- G Romano
- Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | |
Collapse
|
38
|
|
39
|
Russell DW, Kay MA. Adeno-associated virus vectors and hematology. Blood 1999; 94:864-74. [PMID: 10419876 PMCID: PMC3739711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023] Open
Affiliation(s)
- D W Russell
- Markey Molecular Medicine Center, Department of Medicine, University of Washington, Seattle, WA, USA.
| | | |
Collapse
|
40
|
Veldwijk MR, Schiedlmeier B, Kleinschmidt JA, Zeller WJ, Fruehauf S. Superior gene transfer into solid tumour cells than into human mobilised peripheral blood progenitor cells using helpervirus-free adeno-associated viral vector stocks. Eur J Cancer 1999; 35:1136-42. [PMID: 10533460 DOI: 10.1016/s0959-8049(99)00075-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Autologous peripheral blood progenitor cell (PBPC) grafts can be contaminated with tumour cells that potentially give rise to relapse following myeloablative therapy and PBPC transplantation. Adeno-associated virus (AAV)-based vectors produced by a new adenovirus-free technique are a gene delivery system which may be applicable for tumour cell purging. To test for the host range of these vectors, solid tumours of clinical relevance and normal CD34+ PBPC were selected as target cells for an AAV-vector, encoding the green-fluorescent protein (GFP) as the indicator gene. At a multiplicity of infection (MOI) of 100: 79.94% +/- 14.36% (mean +/- SEM) of the connective tissue sarcoma cell line (HS-1) and 64.84% +/- 6.91% of the cervical carcinoma cell line cells (HeLa-RC) expressed GFP while the other cell lines tested (1 ovarian tumour, 1 germ cell tumour, 1 osteosarcoma, 2 small cell lung cancer) ranged between 2.82% and 11.94%. Optimising the transduction protocol by use of higher MOIs of up to 500 and by pretreatment with the tyrosine kinase inhibitor, genistein, resulted in up to 95.97% and 94.10% green-fluorescent HS-1 and HeLa-RC cells, respectively. In contrast, only 1.39% +/- 0.51% of the normal haematopoietic CD34+ progenitor cells expressed GFP at a MOI of 100. The differential infectivity between HS-1 and CD34+ cells was maintained after tumour cell spiking in leucapheresis products. Our observations suggest that AAV-based vectors may prove useful for purging of autologous PBPC grafts from solid tumour cells.
Collapse
Affiliation(s)
- M R Veldwijk
- German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
41
|
Ueba T, Kaspar B, Zhao X, Gage FH. Repression of human fibroblast growth factor 2 by a novel transcription factor. J Biol Chem 1999; 274:10382-7. [PMID: 10187827 DOI: 10.1074/jbc.274.15.10382] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Here we describe the cloning of the regulator of fibroblast growth factor 2 (FGF-2) transcription (RFT) using a yeast one-hybrid screening with a defined motif in FGF-2 promoter as a target sequence. Overexpression of human RFT (RFT-A) reduces FGF-2 RNA and protein levels in both normal and tumor cell lines. Its splice variants, RFT-A' and RFT-B, have deletions in the putative DNA binding domain and fail to bind FGF-2 promoter and repress FGF-2 gene expression. The ratios of RFT isoforms differ between normal and tumor cells, with the splice variants dominating in tumor cells. Overexpression of RFT-A induces glioma cell death. Our data suggest that regulation of FGF-2 by RFT is important for cellular functions and may be impaired in certain tumors.
Collapse
Affiliation(s)
- T Ueba
- Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
42
|
Transduction of Primitive Human Marrow and Cord Blood-Derived Hematopoietic Progenitor Cells With Adeno-Associated Virus Vectors. Blood 1999. [DOI: 10.1182/blood.v93.6.1882.406k03_1882_1894] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We evaluated the capacity of adeno-associated virus (AAV) vectors to transduce primitive human myeloid progenitor cells derived from marrow and cord blood in long-term cultures and long-term culture-initiating cell (LTC-IC) assays. Single-colony analyses showed that AAV vectors transduced CD34+ and CD34+38− clonogenic cells in long-term culture. Gene transfer was readily observed in LTC-ICs derived from 5-, 8-, and 10-week cultures. Recombinant AAV (rAAV) transduction was observed in every donor analyzed, although a wide range of gene transfer frequencies (5% to 100%) was noted. AAV transduction of LTC-ICs was stable, with week-8 and -10 LTC-ICs showing comparable or better transduction relative to week-5 LTC-ICs. Fluorescence in situ hybridization (FISH) analyses performed to determine the fate of AAV vectors in transduced cells showed that 9% to 28% of CD34+ and CD34+38− cells showed stable vector integration as evidenced by chromosome-associated signals in metaphase spreads. Comparisons of interphase and metaphase FISH suggested that a fraction of cells also contained episomal vector at early time points after transduction. Despite the apparent loss of the episomal forms with continued culture, the number of metaphases containing integrated vector genomes remained stable long term. Transgene transcription and placental alkaline phosphatase (PLAP) expression was observed in CD34+, CD34+38−LTC-ICs in the absence of selective pressure. These results suggest that primitive myeloid progenitors are amenable to genetic modification with AAV vectors.
Collapse
|
43
|
Zhang PX, Fuleihan RL. Transfer of activation-dependent gene expression into T cell lines by recombinant adeno-associated virus. Gene Ther 1999; 6:182-9. [PMID: 10435102 DOI: 10.1038/sj.gt.3300803] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examined the ability of recombinant adeno-associated virus (rAAV) to transfer regulated gene expression into T cell lines. An AAV-based vector containing the neomycin resistance gene and expressing the firefly luciferase (luc) gene under the regulatory control of the interleukin 2 promoter (pAAV-luc) was generated and adenovirus-free rAAV (rAAV-luc) was produced from this vector. Transfection of pAAV-luc into the human T cell line Jurkat resulted in luciferase expression while infection of Jurkat T cells with rAAV-luc resulted in significant luciferase expression only after selection for neomycin-resistant cells. Long-term growth of transduced Jurkat T cells showed that there was no detectable constitutive expression of luciferase and that luciferase gene expression remained inducible for at least 180 days. Luciferase expression was activated by PMA and ionomycin and by anti-CD3 antibodies and was inhibited by cyclosporin A. Examination of G418-resistant clones showed that rAAV-luc had integrated into the host chromosomes but that some of the clones lost some of the transferred DNA or lost expression from the transferred DNA. These results indicate that rAAV can transfer and integrate regulated gene expression into T cell lines but that the transferred genetic material may be lost or its expression may be silenced over time.
Collapse
Affiliation(s)
- P X Zhang
- Yale Child Health Research Center, Yale University School of Medicine, New Haven, CT 06520-8081, USA
| | | |
Collapse
|
44
|
Schimmenti S, Boesen J, Claassen EA, Valerio D, Einerhand MP. Long-term genetic modification of rhesus monkey hematopoietic cells following transplantation of adenoassociated virus vector-transduced CD34+ cells. Hum Gene Ther 1998; 9:2727-34. [PMID: 9874271 DOI: 10.1089/hum.1998.9.18-2727] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have explored the potential of recombinant adenoassociated virus (AAV) vectors for gene transfer of the human beta-globin gene and the genetic modification of primate pluripotent hematopoietic stem cells (P-PHSCs). Transduction of P-PHSCs was tested in a preclinical bone marrow transplantation model in rhesus monkeys. CD34+ cells were transduced ex vivo and autologously transplanted without prior selection into irradiated rhesus monkeys. Vector-transduced peripheral blood mononuclear cells and granulocytes were present in the circulation for more than 15 months after transplantation. Approximately 1 in 10(5) cells in the circulation was vector modified. The vector was detected in the bone marrow, in granulocytes, and in purified populations of B and T cells, thus demonstrating multilineage repopulation by vector-transduced stem cells. Comparison of transduction protocols suggested that short-term culture of P-PHSCs enhances transduction and subsequent repopulation by rAAV-transduced cells. These results demonstrate that rAAV vectors can be used for the permanent genetic modification of a rhesus monkey hematopoietic system in the absence of selective pressure.
Collapse
|
45
|
Li LL. Reconstitution of NADPH oxidase activity in human X-linked chronic granulomatous disease myeloid cells after stable gene transfer using a recombinant adeno-associated virus 2 vector. Blood Cells Mol Dis 1998; 24:522-38. [PMID: 9880243 DOI: 10.1006/bcmd.1998.0216] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
X-linked chronic granulomatous disease (X-CGD) is an inherited disorder of host defense that results from mutations in the gene encoding gp91phox, the large subunit of the phagocyte NADPH oxidase flavocytochrome b. In this study, we constructed a recombinant adeno-associated virus-2 (AAV) vector in which the constitutively active promoter from the human elongation factor- 1alpha (EF-1alpha) gene drives expression of the murine gp91phox cDNA, and tested its ability to integrate and express in a human X-CGD myeloid cell line. The nitroblue tetrazolium (NBT) test of NADPH oxidase activity was used to screen transduced cells for vector-mediated expression of recombinant gp91phox. Between 2 - 14% of cells were NBT-positive in the first several weeks after transduction. Clones with NBT-positive cells persisting several months after transduction had integrated vector by Southern blot analyses, with high level reconstitution of NADPH oxidase activity. In some clones, oxidase activity persisted for at least 8 to 14 months. In the majority, however, vector-derived RNA transcripts declined, although integrated rAAV genomes persisted. Decreased transgene expression was not directly correlated with methylation of the provirus. This study indicates that rAAV vectors can be successfully used for stable gene transfer, integration, and expression of recombinant gp91phoxin a human myeloid cell line for at least 8 - 14 months in the absence of any selection. The EF-1alpha promotor, however, was subject to silencing in a high percentage of clones with integrated rAAV, suggesting that alternative promotors may be desirable for achieving long-term expression in myeloid cells.
Collapse
Affiliation(s)
- L L Li
- Herman B Wells Center for Pediatric Research, Department of Pediatrics,Indianapolis, IN 46202, USA
| |
Collapse
|
46
|
Yang Q, Mamounas M, Yu G, Kennedy S, Leaker B, Merson J, Wong-Staal F, Yu M, Barber JR. Development of novel cell surface CD34-targeted recombinant adenoassociated virus vectors for gene therapy. Hum Gene Ther 1998; 9:1929-37. [PMID: 9741431 DOI: 10.1089/hum.1998.9.13-1929] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant adenoassociated virus (rAAV) type 2 vectors have been used to transduce a wide variety of cell types, including hematopoietic progenitor cells. For in vivo gene transfer, it is desirable to have an rAAV vector that specifically transduces selected target cells. As a first step toward generating an rAAV vector capable of targeting delivery in vivo, we have engineered a chimeric protein combining the AAV capsid protein and the variable region of a single-chain antibody against human CD34 molecules, a cell surface marker for hematopoietic stem/progenitor cells. Inclusion of the chimeric CD34 single-chain antibody-AAV capsid proteins within an rAAV virion significantly increased the preferential infectivity of rAAV for the CD34+ human myoleukemia cell line KG-1, which is normally refractory to rAAV transduction. Antibodies against the single-chain antibody and the CD34 protein blocked this transduction. This chimeric vector represents a significant improvement in the host range of rAAV and the first step toward specific gene delivery by rAAV vectors to cells of choice, in this case, hematopoietic progenitor cells, for the treatment of human disease.
Collapse
MESH Headings
- Antibodies, Anti-Idiotypic
- Antibodies, Blocking
- Antibodies, Monoclonal
- Antigens, CD34/immunology
- Antigens, CD34/metabolism
- Binding, Competitive
- Biomarkers
- Capsid/genetics
- Cloning, Molecular
- Dependovirus/genetics
- Dependovirus/isolation & purification
- Gene Targeting/methods
- Genetic Therapy/methods
- Genetic Vectors/genetics
- HeLa Cells
- Hematopoietic Stem Cells
- Humans
- Immunoglobulin Variable Region/genetics
- Leukemia, Myeloid, Acute
- RNA, Messenger/biosynthesis
- RNA, Viral/biosynthesis
- Recombinant Fusion Proteins
- Tumor Cells, Cultured
- Viral Proteins/biosynthesis
- Virion
Collapse
Affiliation(s)
- Q Yang
- Immusol, Inc., San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Mayani H, Lansdorp PM. Biology of human umbilical cord blood-derived hematopoietic stem/progenitor cells. Stem Cells 1998; 16:153-65. [PMID: 9617891 DOI: 10.1002/stem.160153] [Citation(s) in RCA: 180] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reported in 1989, studies by Broxmeyer, Gluckman, and colleagues demonstrated that umbilical cord blood (UCB) is a rich source of hematopoietic stem/progenitor cells (HSPC) and that UCB could be used in clinical settings for hematopoietic cell transplantation. Since then, a great interest has been generated on the biological characterization of these cells. Over the last nine years, several groups have focused on the study of UCB HSPC, addressing different aspects, such as the frequency of these cells in UCB, the identification of different HSPC subsets based on their immunophenotype, their ability to respond to hematopoietic cytokines, the factors that control their proliferation and expansion potentials, and their capacity to reconstitute hematopoiesis in animal models. Most of these studies have shown that significant functional differences exist between HSPC from UCB and adult bone marrow (i.e., the former possess higher proliferation and expansion potential than the latter). It is also noteworthy that genetic manipulation of UCB HSPC has been achieved by several groups and that genetically modified UCB cells have already been used in the clinic. In spite of the significant advances in the characterization of these cells, we are still in the process of trying to fully understand their biology, both at the cellular and the molecular levels. In the present article, we describe and discuss what is currently known about the biology of UCB HSPC.
Collapse
Affiliation(s)
- H Mayani
- Oncological Research Unit, Oncology Hospital, National Medical Center, Mexico City, Mexico
| | | |
Collapse
|
49
|
Maass G, Bogedain C, Scheer U, Michl D, Hörer M, Braun-Falco M, Volkenandt M, Schadendorf D, Wendtner CM, Winnacker EL, Kotin RM, Hallek M. Recombinant adeno-associated virus for the generation of autologous, gene-modified tumor vaccines: evidence for a high transduction efficiency into primary epithelial cancer cells. Hum Gene Ther 1998; 9:1049-59. [PMID: 9607416 DOI: 10.1089/hum.1998.9.7-1049] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To explore the potential of recombinant vectors based on recombinant adeno-associated virus (rAAV) for cancer vaccination, we investigated the transduction efficiency of rAAV into cancer cells ex vivo. Infection of human epithelial cancer cell lines with rAAV carrying reporter genes encoding beta-galactosidase (rAAV/LacZ) or luciferase (rAAV/Luc) resulted in high levels of reporter gene expression (>90% positive cells). In marked contrast, rAAV poorly transduced all murine tumor cell lines, as well as human hematopoietic cell lines. Either irradiation or adenovirus infection of tumor cells prior to rAAV infection induced a 10- to 100-fold increase of reporter gene expression. To determine the transduction efficiency of rAAV into primary cancer cells, freshly isolated, irradiated tumor cells from malignant melanoma and ovarian carcinoma patients were infected with rAAV/Luc, resulting in up to 6.9-fold higher levels of gene expression than in a HeLa tumor cell line. Time course experiments with freshly isolated tumor cells infected with rAAV/Luc showed maximal levels of luciferase expression between days 3 and 9 posttransduction. Simultaneous infection of primary tumor cells with up to three rAAV vectors containing genes encoding the immunostimulatory proteins B7-2 (CD86), p35 subunit of IL-12, and p40 subunit of IL-12 resulted in high expression of B7-2 in more than 90% of the tumor cells and in the secretion of high levels of IL-12. Taken together, our results demonstrate that rAAV efficiently transduces freshly isolated human, epithelial tumor cells and might therefore be a potent tool to produce improved, gene-modified cancer vaccines.
Collapse
Affiliation(s)
- G Maass
- MediGene AG, Martinsried/Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Doerflinger N, Miclea JM, Lopez J, Chomienne C, Bougnères P, Aubourg P, Cartier N. Retroviral transfer and long-term expression of the adrenoleukodystrophy gene in human CD34+ cells. Hum Gene Ther 1998; 9:1025-36. [PMID: 9607414 DOI: 10.1089/hum.1998.9.7-1025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adrenoleukodystrophy (ALD) is a demyelinating disease of the central nervous system that results from a genetic deficiency of ALDP, an ABC protein involved in the transport of very long-chain fatty acids (VLCFAs). The cloning of the ALD gene and the positive effects of allogeneic bone marrow transplantation support the feasibility of a gene therapy approach. We report the retroviral transfer of the ALD cDNA to peripheral blood and bone marrow CD34+ cells from control donors and ALD patients. Prestimulation of these cells with cytokines, followed by infection with the M48-ALD retroviral vector, resulted in 20% transduction efficiency (4-40%) and expression of the vector-encoded ALDP in 20% of CD34+ cells (7.3-50%). Long-term culture (LTC) of transduced CD34+ cells from two ALD patients showed efficient transduction (24-28%) and stable expression (25-32%) of ALDP in derived clonogenic progenitors at 3 weeks of culture. The expression of ALDP in CFU cells derived from 5 and 6 weeks of LTC confirmed the effective transduction of LTC-initiating cells. Expression of ALDP was observed in CD68+ CFU-derived cells, suggesting that monocyte-macrophages, the target bone marrow cells in ALD, were produced from transduced progenitor cells. VL-CFA content was corrected in LTC and CFU-derived cells in proportion to the percentage of transduced cells, indicating that the vector-encoded ALDP was functional. Although not efficient yet to allow a clinical perspective, these results demonstrate the feasibility of ALD gene transfer into CD34+ cells of ALD patients.
Collapse
Affiliation(s)
- N Doerflinger
- INSERM U342, Hôpital Saint-Vincent de Paul, Paris, France
| | | | | | | | | | | | | |
Collapse
|