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: 0] [Impact Index Per Article: 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: 4] [Impact Index Per Article: 1.3] [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
|
Chatterjee S. Efficient Nuclease-free HR by Clade F AAV Requires High MOIs with High Quality Vectors. Mol Ther 2019; 27:2058-2061. [PMID: 31735603 DOI: 10.1016/j.ymthe.2019.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Saswati Chatterjee
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
4
|
Tsukamoto T. Gene Therapy Approaches to Functional Cure and Protection of Hematopoietic Potential in HIV Infection. Pharmaceutics 2019; 11:E114. [PMID: 30862061 PMCID: PMC6470728 DOI: 10.3390/pharmaceutics11030114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/28/2022] Open
Abstract
Although current antiretroviral drug therapy can suppress the replication of human immunodeficiency virus (HIV), a lifelong prescription is necessary to avoid viral rebound. The problem of persistent and ineradicable viral reservoirs in HIV-infected people continues to be a global threat. In addition, some HIV-infected patients do not experience sufficient T-cell immune restoration despite being aviremic during treatment. This is likely due to altered hematopoietic potential. To achieve the global eradication of HIV disease, a cure is needed. To this end, tremendous efforts have been made in the field of anti-HIV gene therapy. This review will discuss the concepts of HIV cure and relative viral attenuation and provide an overview of various gene therapy approaches aimed at a complete or functional HIV cure and protection of hematopoietic functions.
Collapse
Affiliation(s)
- Tetsuo Tsukamoto
- Department of Immunology, Kindai University Faculty of Medicine, Osaka 5898511, Japan.
| |
Collapse
|
5
|
Smith LJ, Wright J, Clark G, Ul-Hasan T, Jin X, Fong A, Chandra M, St Martin T, Rubin H, Knowlton D, Ellsworth JL, Fong Y, Wong KK, Chatterjee S. Stem cell-derived clade F AAVs mediate high-efficiency homologous recombination-based genome editing. Proc Natl Acad Sci U S A 2018; 115:E7379-E7388. [PMID: 30018062 PMCID: PMC6077703 DOI: 10.1073/pnas.1802343115] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The precise correction of genetic mutations at the nucleotide level is an attractive permanent therapeutic strategy for human disease. However, despite significant progress, challenges to efficient and accurate genome editing persist. Here, we report a genome editing platform based upon a class of hematopoietic stem cell (HSC)-derived clade F adeno-associated virus (AAV), which does not require prior nuclease-mediated DNA breaks and functions exclusively through BRCA2-dependent homologous recombination. Genome editing is guided by complementary homology arms and is highly accurate and seamless, with no evidence of on-target mutations, including insertion/deletions or inclusion of AAV inverted terminal repeats. Efficient genome editing was demonstrated at different loci within the human genome, including a safe harbor locus, AAVS1, and the therapeutically relevant IL2RG gene, and at the murine Rosa26 locus. HSC-derived AAV vector (AAVHSC)-mediated genome editing was robust in primary human cells, including CD34+ cells, adult liver, hepatic endothelial cells, and myocytes. Importantly, high-efficiency gene editing was achieved in vivo upon a single i.v. injection of AAVHSC editing vectors in mice. Thus, clade F AAV-mediated genome editing represents a promising, highly efficient, precise, single-component approach that enables the development of therapeutic in vivo genome editing for the treatment of a multitude of human gene-based diseases.
Collapse
Affiliation(s)
- Laura J Smith
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | | | - Gabriella Clark
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | - Taihra Ul-Hasan
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | - Xiangyang Jin
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | - Abigail Fong
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | - Manasa Chandra
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | | | | | | | | | - Yuman Fong
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010
| | - Kamehameha K Wong
- Department of Hematology and Stem Cell Transplantation, City of Hope Medical Center, Duarte, CA 91010
| | - Saswati Chatterjee
- Department of Surgery, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010;
| |
Collapse
|
6
|
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: 4.7] [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
|
7
|
Smith LJ, Ul-Hasan T, Carvaines SK, Van Vliet K, Yang E, Wong KK, Agbandje-McKenna M, Chatterjee S. Gene transfer properties and structural modeling of human stem cell-derived AAV. Mol Ther 2014; 22:1625-34. [PMID: 24925207 DOI: 10.1038/mt.2014.107] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/29/2014] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are proving to be remarkably successful for in vivo gene delivery. Based upon reports of abundant AAV in the human marrow, we tested CD34(+) hematopoietic stem cells for the presence of natural AAV. Here, we report for the first time, the presence of novel AAV variants in healthy CD34(+) human peripheral blood stem cells. The majority of healthy peripheral blood stem cell donors were found to harbor AAV in their CD34(+) cells. Every AAV isolated from CD34(+) cells mapped to AAV Clade F. Gene transfer vectors derived from these novel AAVs efficiently underwent entry and postentry processing in human cord blood stem cells and supported stable gene transfer into long-term, in vivo engrafting human HSCs significantly better than other serotypes. AAVHSC-transduced human CD34(+) cells engrafted in vivo and gave rise to differentiated transgene-expressing progeny. Importantly, gene-marked CD34(+) stem cells persisted long term in xenograft recipients, indicating transduction of primitive progenitors. Notably, correlation of structure with function permitted identification of potential capsid components important for HSC transduction. Thus, AAVHSCs represent a new class of genetic vectors for the manipulation of HSC genomes.
Collapse
Affiliation(s)
- Laura J Smith
- AAV Laboratory, Department of Virology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Taihra Ul-Hasan
- AAV Laboratory, Department of Virology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Sarah K Carvaines
- AAV Laboratory, Department of Virology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Kim Van Vliet
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute University of Florida, Gainesville, Florida, USA
| | - Ethel Yang
- AAV Laboratory, Department of Virology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Kamehameha K Wong
- Divisions of Hematology/Stem Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute University of Florida, Gainesville, Florida, USA
| | - Saswati Chatterjee
- AAV Laboratory, Department of Virology, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
8
|
Bennett MS, Akkina R. Gene therapy strategies for HIV/AIDS: preclinical modeling in humanized mice. Viruses 2013; 5:3119-41. [PMID: 24351796 PMCID: PMC3967164 DOI: 10.3390/v5123119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/04/2013] [Accepted: 12/03/2013] [Indexed: 12/28/2022] Open
Abstract
In the absence of an effective vaccine and lack of a complete cure, gene therapy approaches to control HIV infection offer feasible alternatives. Due to the chronic nature of infection, a wide window of opportunity exists to gene modify the HIV susceptible cells that continuously arise from the bone marrow source. To evaluate promising gene therapy approaches that employ various anti-HIV therapeutic molecules, an ideal animal model is necessary to generate important efficacy and preclinical data. In this regard, the humanized mouse models that harbor human hematopoietic cells susceptible to HIV infection provide a suitable in vivo system. This review summarizes the currently used humanized mouse models and different anti-HIV molecules utilized for conferring HIV resistance. Humanized mouse models are compared for their utility in this context and provide perspectives for new directions.
Collapse
Affiliation(s)
| | - Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, 1619 Campus delivery, Fort Collins, CO 80523, USA.
| |
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: 4.8] [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
|
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: 59] [Impact Index Per Article: 4.9] [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
|
11
|
Recombinant AAV2-mediated β-globin expression in human fetal hematopoietic cells from the aborted fetuses with β-thalassemia major. Int J Hematol 2011; 93:691-699. [PMID: 21617888 DOI: 10.1007/s12185-011-0823-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 02/21/2011] [Accepted: 03/22/2011] [Indexed: 10/18/2022]
Abstract
Genetic correction of autologous hematopoietic stem cells has been proposed as an attractive treatment method for β-thalassemia. Our previous study has shown that recombinant adeno-associated virus 2 (rAAV2) efficiently transduces human fetal liver hematopoietic cells, and mediates the expression of the human β-globin gene in vivo. In this study, we investigated whether rAAV2 could also mediate the expression of normal β-globin gene in human hematopoietic cells from β-thalassemia patients. Human hematopoietic cells were isolated from aborted β-thalassemia major fetuses, transduced with rAAV2-β-globin, and then transplanted into nude mice. We found that rAAV2-β-globin transduced human fetal hematopoietic cells, as determined by allele-specific PCR analysis. Furthermore, β-globin transgene expression was detected in human hematopoietic cells up to 70 days post-transplantation in the recipient mice. High-pressure liquid chromatography analysis showed that human β-globin expression levels increased significantly compared with control, as indicated by a 1.2-2.8-fold increase in the ratio of β/α-globin chain. These novel data demonstrate that rAAV2 can transduce and mediate the normal β-globin gene expression in fetal hematopoietic cells from β-thalassemia patients. Our findings further support the potential use of rAAV-based gene therapy in the treatment of human β-thalassemia.
Collapse
|
12
|
Kauss MA, Smith LJ, Zhong L, Srivastava A, Wong KK, Chatterjee S. Enhanced long-term transduction and multilineage engraftment of human hematopoietic stem cells transduced with tyrosine-modified recombinant adeno-associated virus serotype 2. Hum Gene Ther 2010; 21:1129-36. [PMID: 20486772 DOI: 10.1089/hum.2010.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The search for the ideal stem cell gene therapy vector continues as recognized problems persist. Although recombinant adeno-associated virus serotype 2 (rAAV2) mediates gene transfer into hematopoietic stem cells, identified restrictions to transgene expression reduce overall efficiency. Studies have shown that transduction efficiencies are significantly improved by preventing early proteasomal degradation after mutation of surface-exposed tyrosine residues on the capsid to phenylalanine. Here, we report that transduction of human cord blood CD34(+) stem cells by tyrosine-modified rAAV2 is significantly enhanced both in vitro and in vivo. Serial long-term in vivo bioluminescent imaging of immune-deficient recipients after xenotransplantation of CD34(+) cells transduced with tyrosine-modified rAAV2-luciferase revealed that modification of rAAV2 capsids led to a significant increase in the transduction of human CD34(+) cells, without adversely affecting engraftment capacity, or the ability to undergo multilineage differentiation and self-renewal. Together with observations of sustained high-level transgene expression in vivo and efficient persistence of rAAV genomes in human hematopoietic cells, these results suggest that, because of their ability to bypass restrictions to transduction, tyrosine-modified rAAV vectors, particularly Y500F, Y700F, Y444F, and Y704F, represent highly promising candidates for therapeutic evaluation for diseases of human hematopoietic stem cells.
Collapse
Affiliation(s)
- M Ariel Kauss
- Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
13
|
Aggarwal R, Pompili VJ, Das H. Genetic modification of ex-vivo expanded stem cells for clinical application. FRONT BIOSCI-LANDMRK 2010; 15:854-71. [PMID: 20515730 PMCID: PMC9930440 DOI: 10.2741/3650] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stem cell therapy is currently considered as an important regime for repairing, replacing or enhancing the biological functions of the damaged tissues. Among adult stem cells, hematopoietic stem cells (HSCs) are commonly used for cure of hematological disorders. However, the number of HSCs obtained from sources like bone marrow, peripheral or umbilical cord blood is not sufficient for routine clinical application. Thus, ex-vivo expansion of HSCs becomes critically important. Ex-vivo culture and expansion of stem cells are challenging, as stem cells differentiate in culture rather than self-renew. Lack of clarity about the factors responsible for quiescence and differentiation of HSCs, investigators struggled to optimize conditions for ex vivo expansion. As we understand better, various strategies can be incorporated to mimic in vivo conditions for successful expansion of stem cells. However, characterization and biological functionality should also be tested for expanded stem cells prior to clinical application. To treat ischemia by enhancing therapeutic angiogenesis and neo-vascularization, the role of genetic modification of HSCs with pro-angiogenic factors is the focus of this review.
Collapse
|
14
|
Veldwijk MR, Sellner L, Stiefelhagen M, Kleinschmidt JA, Laufs S, Topaly J, Fruehauf S, Zeller WJ, Wenz F. Pseudotyped recombinant adeno-associated viral vectors mediate efficient gene transfer into primary human CD34(+) peripheral blood progenitor cells. Cytotherapy 2010; 12:107-12. [PMID: 19929455 DOI: 10.3109/14653240903348293] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AND AIMS Because of their pluripotency, human CD34(+) peripheral blood progenitor cells (PBPC) are targets of interest for the treatment of many acquired and inherited disorders using gene therapeutic approaches. Unfortunately, most current vector systems lack either sufficient transduction efficiency or an appropriate safety profile. Standard single-stranded recombinant adeno-associated virus 2 (AAV2)-based vectors offer an advantageous safety profile, yet lack the required efficiency in human PBPC. METHODS A panel of pseudotyped AAV vectors (designated AAV2/x, containing the vector genome of serotype 2 and capsid of serotype x, AAV2/1-AAV2/6) was screened on primary human granulocyte-colony-stimulating factor (G-CSF)-mobilized CD34(+) PBPC to determine their gene transfer efficacy. Additionally, double-stranded self-complementary AAV (dsAAV) were used to determine possible second-strand synthesis limitations. RESULTS AAV2/6 vectors proved to be the most efficient [12.8% (1.8-25.4%) transgene-expressing PBPC after a single transduction], being significantly more efficient (all P<0.005) than the other vectors [AAV2/2, 2.0% (0.2-7.3%); AAV2/1, 1.3% (0.1-2.9%); others, <; 1% transgene-expressing PBPC]. In addition, the relevance of the single-to-double-strand conversion block in transduction of human PBPC could be shown using pseudotyped dsAAV vectors: for dsAAV2/2 [9.3% (8.3-20.3%); P<0.001] and dsAAV2/6 [37.7% (23.6-61.0%); P<0.001) significantly more PBPC expressed the transgene compared with their single-stranded counterparts; for dsAAV2/1, no significant increase could be observed. CONCLUSIONS We have shown that clinically relevant transduction efficiency levels using AAV-based vectors in human CD34(+) PBPC are feasible, thereby offering an efficient alternative vector system for gene transfer into this important target cell population.
Collapse
Affiliation(s)
- Marlon R Veldwijk
- Department of Radiation Oncology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Over the past three decades many techniques for expressing exogenous genes in a variety of cells and cell lines have been developed. Exogenous gene expression in macrophages has lagged behind that of other nonhematopioetic cells. There are many reasons for this, but most are due to technical difficulties associated with transfecting macrophages. As professional phagocytes, macrophages are endowed with many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process. This is especially true of activated macrophages which undergo a dramatic change in their physiology following exposure to immune or inflammatory stimuli. Viral transduction of these cells has been hampered because macrophages are end-stage cells that generally do not divide; therefore, some of the vectors that depend on integration into a replicative genome have met with limited success. Furthermore, macrophages are quite responsive to "danger signals," and therefore several of the original viral vectors that were used for gene transfer induced potent anti-viral responses in these cells making these vectors inappropriate for gene delivery. Many of these difficulties have been largely overcome, and relatively high efficiency gene expression in primary human or murine macrophages is becoming more routine. In the present chapter we discuss some of the gene expression techniques that have met with success and review the advantages and disadvantages of each.
Collapse
|
16
|
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
|
17
|
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: 0.6] [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
|
18
|
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: 1.4] [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
|
19
|
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: 1.0] [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
|
20
|
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: 1.8] [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
|
21
|
Paz H, Wong CA, Li W, Santat L, Wong KK, Chatterjee S. Quiescent subpopulations of human CD34-positive hematopoietic stem cells are preferred targets for stable recombinant adeno-associated virus type 2 transduction. Hum Gene Ther 2007; 18:614-26. [PMID: 17638572 DOI: 10.1089/hum.2006.188] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have previously demonstrated recombinant adeno-associated viral (rAAV) transduction of human CD34(+) hematopoietic stem cells (HSCs) capable of serial engraftment in vivo. Here we evaluated the capacity of rAAV2 to mediate gene transfer into nondividing, quiescent, primitive CD34(+) cells subdivided on the basis of metabolic, mitotic, and phenotypic properties. Results revealed that CD34(+)CD38() marrow cells are the most quiescent, exist primarily in G(0) at isolation and are the only population to remain nondividing during the entire exposure to free rAAV. Despite significant differences in the extended clonogenic capacities of CD34(+) subsets in stromal cultures, the frequency of rAAV marking of colonies derived from primitive progenitors was similar in all three populations, suggesting that both primitive and more differentiated progenitors were initially transduced at equal levels. After transduction, episomal and integrated rAAV genomes were detected in all CD34(+) subsets. However, the more quiescent cells displayed higher levels of integrated rAAV than did rapidly dividing cells. Importantly, stable long-term integration was observed only in the most primitive, quiescent CD34(+)CD38(-) subset, indicating that this HSC compartment comprises the preferred substrate for stable rAAV2 transduction. Previously described rate limitations to transgene expression were observed in transduced CD34(+) cells and could be overcome by tyrphostin pretreatment, which resulted in augmented second-strand synthesis. These results represent the first demonstration of rAAV-mediated gene transfer to primitive, quiescent human CD34(+)CD38(-) stem cells and reveal that nondividing CD34(+)CD38(-) HSCs are the optimal CD34(+) targets for rAAV transduction.
Collapse
Affiliation(s)
- Helicia Paz
- Division of Virology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
22
|
Pósfai G, Plunkett G, Fehér T, Frisch D, Keil GM, Umenhoffer K, Kolisnychenko V, Stahl B, Sharma SS, de Arruda M, Burland V, Harcum SW, Blattner FR. Emergent Properties of Reduced-Genome Escherichia coli. Science 2006; 312:1044-6. [PMID: 16645050 DOI: 10.1126/science.1126439] [Citation(s) in RCA: 510] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
With the use of synthetic biology, we reduced the Escherichia coli K-12 genome by making planned, precise deletions. The multiple-deletion series (MDS) strains, with genome reductions up to 15%, were designed by identifying nonessential genes and sequences for elimination, including recombinogenic or mobile DNA and cryptic virulence genes, while preserving good growth profiles and protein production. Genome reduction also led to unanticipated beneficial properties: high electroporation efficiency and accurate propagation of recombinant genes and plasmids that were unstable in other strains. Eradication of stress-induced transposition evidently stabilized the MDS genomes and provided some of the new properties.
Collapse
Affiliation(s)
- György Pósfai
- Institute of Biochemistry, Biological Research Center, H-6726 Szeged, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhong L, Li W, Li Y, Zhao W, Wu J, Li B, Maina N, Bischof D, Qing K, Weigel-Kelley KA, Zolotukhin I, Warrington KH, Li X, Slayton WB, Yoder MC, Srivastava A. Evaluation of primitive murine hematopoietic stem and progenitor cell transduction in vitro and in vivo by recombinant adeno-associated virus vector serotypes 1 through 5. Hum Gene Ther 2006; 17:321-33. [PMID: 16544981 DOI: 10.1089/hum.2006.17.321] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Conflicting data exist on hematopoietic cell transduction by AAV serotype 2 (AAV2) vectors, and additional AAV serotype vectors have not been evaluated for their efficacy in hematopoietic stem/progenitor cell transduction. We evaluated the efficacy of conventional, single-stranded AAV serotype vectors 1 through 5 in primitive murine hematopoietic stem/progenitor cells in vitro as well as in vivo. In progenitor cell assays using Sca1+ c-kit+ Lin- hematopoietic cells, 9% of the colonies in cultures infected with AAV1 expressed the transgene. Coinfection of AAV1 with self-complementary AAV vectors carrying the gene for T cell protein tyrosine phosphatase (scAAV-TC-PTP) increased the transduction efficiency to 24%, indicating that viral secondstrand DNA synthesis is a rate-limiting step. This was further corroborated by the use of scAAV vectors, which bypass this requirement. In bone marrow transplantation studies involving lethally irradiated syngeneic mice, Sca1+ c-kit+ Lin- cells coinfected with AAV1 +/- scAAV-TC-PTP vectors led to transgene expression in 2 and 7.5% of peripheral blood (PB) cells, respectively, 6 months posttransplantation. In secondary transplantation experiments, 7% of PB cells and 3% of bone marrow (BM) cells expressed the transgene 6 months posttransplantation. Approximately 21% of BM-derived colonies harbored the proviral DNA sequences in integrated forms. These results document that AAV1 is thus far the most efficient vector in transducing primitive murine hematopoietic stem/progenitor cells. Further studies involving scAAV genomes and hematopoietic cell-specific promoters should further augment the transduction efficiency of AAV1 vectors, which should have implications in the optimal use of these vectors in hematopoietic stem cell gene therapy.
Collapse
Affiliation(s)
- Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhong L, Li W, Li Y, Zhao W, Wu J, Li B, Maina N, Bischof D, Qing K, Weigel-Kelley KA, Zolotukhin I, Warrington KH, Li X, Slayton WB, Yoder MC, Srivastava A. Evaluation of Primitive Murine Hematopoietic Stem and Progenitor Cell Transduction In Vitro and In Vivo by Recombinant Adeno-Associated Virus Vector Serotypes 1 Through 5. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
25
|
Zhong L, Li W, Li Y, Zhao W, Wu J, Li B, Maina N, Bischof D, Qing K, Weigel-Kelley KA, Zolotukhin I, Warrington KH, Li X, Slayton WB, Yoder MC, Srivastava A. Evaluation of Primitive Murine Hematopoietic Stem and Progenitor Cell Transduction In Vitro and In Vivo by Recombinant Adeno-Associated Virus Vector Serotypes 1 Through 5. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-179] [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
|