1
|
Fluorescent genetic barcoding for cellular multiplex analyses. Exp Hematol 2018; 67:10-17. [DOI: 10.1016/j.exphem.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
|
2
|
Maetzig T, Ruschmann J, Sanchez Milde L, Lai CK, von Krosigk N, Humphries RK. Lentiviral Fluorescent Genetic Barcoding for Multiplex Fate Tracking of Leukemic Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:54-65. [PMID: 28664166 PMCID: PMC5480982 DOI: 10.1016/j.omtm.2017.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/28/2017] [Indexed: 02/06/2023]
Abstract
Tracking the behavior of leukemic samples both in vitro and in vivo plays an increasingly large role in efforts to better understand the leukemogenic processes and the effects of potential new therapies. Such work can be accelerated and made more efficient by methodologies enabling the characterization of leukemia samples in multiplex assays. We recently developed three sets of lentiviral fluorescent genetic barcoding (FGB) vectors that create 26, 14, and 6 unique immunophenotyping-compatible color codes from GFP-, yellow fluorescent protein (YFP)-, and monomeric kusabira orange 2 (mKO2)-derived fluorescent proteins. These vectors allow for labeling and tracking of individual color-coded cell populations in mixed samples by real-time flow cytometry. Using the prototypical Hoxa9/Meis1 murine model of acute myeloid leukemia, we describe the application of the 6xFGB vector system for assessing leukemic cell characteristics in multiplex assays. By transplanting color-coded cell mixes, we investigated the competitive growth behavior of individual color-coded populations, determined leukemia-initiating cell frequencies, and assessed the dose-dependent potential of cells exposed to the histone deacetylase inhibitor Entinostat for bone marrow homing. Thus, FGB provides a useful tool for the multiplex characterization of leukemia samples in a wide variety of applications with a concomitant reduction in workload, processing times, and mouse utilization.
Collapse
Affiliation(s)
- Tobias Maetzig
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Jens Ruschmann
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Lea Sanchez Milde
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Courteney K Lai
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Niklas von Krosigk
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - R Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
3
|
Altrock PM, Brendel C, Renella R, Orkin SH, Williams DA, Michor F. Mathematical modeling of erythrocyte chimerism informs genetic intervention strategies for sickle cell disease. Am J Hematol 2016; 91:931-7. [PMID: 27299299 PMCID: PMC5093908 DOI: 10.1002/ajh.24449] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/11/2016] [Indexed: 01/24/2023]
Abstract
Recent advances in gene therapy and genome-engineering technologies offer the opportunity to correct sickle cell disease (SCD), a heritable disorder caused by a point mutation in the β-globin gene. The developmental switch from fetal γ-globin to adult β-globin is governed in part by the transcription factor (TF) BCL11A. This TF has been proposed as a therapeutic target for reactivation of γ-globin and concomitant reduction of β-sickle globin. In this and other approaches, genetic alteration of a portion of the hematopoietic stem cell (HSC) compartment leads to a mixture of sickling and corrected red blood cells (RBCs) in periphery. To reverse the sickling phenotype, a certain proportion of corrected RBCs is necessary; the degree of HSC alteration required to achieve a desired fraction of corrected RBCs remains unknown. To address this issue, we developed a mathematical model describing aging and survival of sickle-susceptible and normal RBCs; the former can have a selective survival advantage leading to their overrepresentation. We identified the level of bone marrow chimerism required for successful stem cell-based gene therapies in SCD. Our findings were further informed using an experimental mouse model, where we transplanted mixtures of Berkeley SCD and normal murine bone marrow cells to establish chimeric grafts in murine hosts. Our integrative theoretical and experimental approach identifies the target frequency of HSC alterations required for effective treatment of sickling syndromes in humans. Our work replaces episodic observations of such target frequencies with a mathematical modeling framework that covers a large and continuous spectrum of chimerism conditions. Am. J. Hematol. 91:931-937, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Philipp M. Altrock
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA 02138
| | - Christian Brendel
- Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Raffaele Renella
- Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Stuart H. Orkin
- Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
- Howard Hughes Medical Institute, Cambridge, MA 02138
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - David A. Williams
- Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
- Harvard Stem Cell Institute, Cambridge, MA 02138
- Corresponding Authors: David A. Williams, MD, Boston Children’s Hospital, 300 Longwood Ave., Karp 08125.3, Boston, MA 02115, Phone: 617-919-2697, Fax: 617-730-0868, , Franziska Michor, PhD, Dana-Farber Cancer Institute, Dept of Biostatistics and Computational Biology, Mailstop CLS-11007, 450 Brookline Avenue, Boston, MA 02115, Phone: 617-632-5045,
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115
- Corresponding Authors: David A. Williams, MD, Boston Children’s Hospital, 300 Longwood Ave., Karp 08125.3, Boston, MA 02115, Phone: 617-919-2697, Fax: 617-730-0868, , Franziska Michor, PhD, Dana-Farber Cancer Institute, Dept of Biostatistics and Computational Biology, Mailstop CLS-11007, 450 Brookline Avenue, Boston, MA 02115, Phone: 617-632-5045,
| |
Collapse
|
4
|
Verghese SC, Goloviznina NA, Skinner AM, Lipps HJ, Kurre P. S/MAR sequence confers long-term mitotic stability on non-integrating lentiviral vector episomes without selection. Nucleic Acids Res 2014; 42:e53. [PMID: 24474068 PMCID: PMC3985655 DOI: 10.1093/nar/gku082] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/27/2013] [Accepted: 01/06/2014] [Indexed: 01/11/2023] Open
Abstract
Insertional oncogene activation and aberrant splicing have proved to be major setbacks for retroviral stem cell gene therapy. Integrase-deficient human immunodeficiency virus-1-derived vectors provide a potentially safer approach, but their circular genomes are rapidly lost during cell division. Here we describe a novel lentiviral vector (LV) that incorporates human ß-interferon scaffold/matrix-associated region sequences to provide an origin of replication for long-term mitotic maintenance of the episomal LTR circles. The resulting 'anchoring' non-integrating lentiviral vector (aniLV) achieved initial transduction rates comparable with integrating vector followed by progressive establishment of long-term episomal expression in a subset of cells. Analysis of aniLV-transduced single cell-derived clones maintained without selective pressure for >100 rounds of cell division showed sustained transgene expression from episomes and provided molecular evidence for long-term episome maintenance. To evaluate aniLV performance in primary cells, we transduced lineage-depleted murine hematopoietic progenitor cells, observing GFP expression in clonogenic progenitor colonies and peripheral blood leukocyte chimerism following transplantation into conditioned hosts. In aggregate, our studies suggest that scaffold/matrix-associated region elements can serve as molecular anchors for non-integrating lentivector episomes, providing sustained gene expression through successive rounds of cell division and progenitor differentiation in vitro and in vivo.
Collapse
Affiliation(s)
- Santhosh Chakkaramakkil Verghese
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA, Department of Surgery/Surgical Oncology, Oregon Health & Science University, Portland, OR 97239, USA, Center for Biomedical Education and Research, Institute of Cell Biology, University of Witten/Herdecke, Witten 58453, Germany, Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA and Department of Cell & Developmental Biology Oregon Health & Science University, Portland, OR 97239, USA
| | - Natalya A. Goloviznina
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA, Department of Surgery/Surgical Oncology, Oregon Health & Science University, Portland, OR 97239, USA, Center for Biomedical Education and Research, Institute of Cell Biology, University of Witten/Herdecke, Witten 58453, Germany, Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA and Department of Cell & Developmental Biology Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy M. Skinner
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA, Department of Surgery/Surgical Oncology, Oregon Health & Science University, Portland, OR 97239, USA, Center for Biomedical Education and Research, Institute of Cell Biology, University of Witten/Herdecke, Witten 58453, Germany, Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA and Department of Cell & Developmental Biology Oregon Health & Science University, Portland, OR 97239, USA
| | - Hans J. Lipps
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA, Department of Surgery/Surgical Oncology, Oregon Health & Science University, Portland, OR 97239, USA, Center for Biomedical Education and Research, Institute of Cell Biology, University of Witten/Herdecke, Witten 58453, Germany, Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA and Department of Cell & Developmental Biology Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter Kurre
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA, Department of Surgery/Surgical Oncology, Oregon Health & Science University, Portland, OR 97239, USA, Center for Biomedical Education and Research, Institute of Cell Biology, University of Witten/Herdecke, Witten 58453, Germany, Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA and Department of Cell & Developmental Biology Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
5
|
DNA transfection of bone marrow stromal cells using microbubble-mediated ultrasound and polyethylenimine: an in vitro study. Cell Biochem Biophys 2014; 66:775-86. [PMID: 23404495 DOI: 10.1007/s12013-013-9523-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Non-viral vector transfection efficiency is an issue affecting the clinical application of stem cell gene therapy. This study makes use of the synergistic effect of combining ultrasound (US) with microbubbles (MB) and polyethylenimine (PEI) to increase DNA transfection efficiency, which will enhance the efficiency of gene transfer to bone marrow stromal cells (BMSCs). The optimal parameters for primary-cultured rat-BMSC DNA transfection were examined. The study was arranged based on uniform design. Using a construct containing hepatocyte growth factor (HGF) tagged with enhanced green fluorescent protein (pEGFP-HGF) as example, the mixture of BMSCs, MB, and PEI:DNA complex were exposed to US with frequency of 1 MHz and 10% duty cycle pulses. Other factors such as acoustic intensity (Q), MB dosage, and total treatment time (T) were also tested. The results were analyzed by regression analysis. Using the best match of parameters, Q = 0.6 W/cm(2), MB = 10(6)/ml, T = 30 s, different groups were compared. The cooperativity of MB-mediated US and PEI enhanced the gene transfection efficiency by nearly 38-times compared to the DNA without US group. Furthermore, the expression of HGF protein was confirmed by Western blot. The eGFP could be not only seen mainly at the cytoplasm, but also seen in the nucleus in a small proportion of the cells (<10%) for up to 7 observed days. The transfected BMSCs maintained their capability of multi-directional differentiation and reproductive activity. Our results provide useful information in establishing a novel non-viral transfection method, which may be applied to clinical application in stem cell gene therapy.
Collapse
|
6
|
Peng L, Gao Y, Xue YN, Huang SW, Zhuo RX. The effectiveness, cytotoxicity, and intracellular trafficking of nonviral vectors for gene delivery to bone mesenchymal stem cells. J BIOACT COMPAT POL 2013. [DOI: 10.1177/0883911513481893] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nonviral gene delivery that enables exogenous gene expression in bone mesenchymal stem cells could accelerate clinical application of cell-based gene therapy. This study systematically investigated and compared the potential of polyethylenimine and Lipofectamine 2000 as gene carriers to modify bone mesenchymal stem cells including transfection efficiency, cytotoxicity, intracellular trafficking as well as cell membrane damage and apoptosis/necrosis. Polyethylenimine at its optimal N/P ratio of 10 demonstrated the same toxic effects but lower transfection efficiency (17.1% vs 39.5%) compared to Lipofectamine. Intracellular trafficking resulted in over 80% of bone mesenchymal stem cells that were able to take up polyethylenimine polyplexes, but only 20.69% showed nuclear uptake; however, for Lipofectamine, about half bone mesenchymal stem cells were found to uptake lipoplexes but about 30% displayed nuclear localization. Moreover, the percentages of nuclear localization of both vectors were in close relationship with their transfection efficiency. We concluded that for bone mesenchymal stem cell transfection, polyethylenimine displayed high cellular uptake but Lipofectamine was more effective in delivering genes into the nucleus, which was likely the underlying basis for a more efficient gene expression. Further structure modification of polyethylenimine such as improving its nuclear entry ability will eventually make it a better candidate for bone mesenchymal stem cells’ in vitro gene delivery.
Collapse
Affiliation(s)
- Lin Peng
- State Key Laboratory of Oral Diseases, West China College & Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yuan Gao
- State Key Laboratory of Oral Diseases, West China College & Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Ya-Nan Xue
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P.R. China
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P.R. China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P.R. China
| |
Collapse
|
7
|
Polyclonal fluctuation of lentiviral vector–transduced and expanded murine hematopoietic stem cells. Blood 2011; 117:3053-64. [DOI: 10.1182/blood-2010-08-303222] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Gene therapy has proven its potential to cure diseases of the hematopoietic system. However, severe adverse events observed in clinical trials have demanded improved gene-transfer conditions. Whereas progress has been made to reduce the genotoxicity of integrating gene vectors, the role of pretransplantation cultivation is less well investigated. We observed that the STIF (stem cell factor [SCF], thrombopoietin [TPO], insulin-like growth factor-2 [IGF-2], and fibroblast growth factor-1 [FGF-1]) cytokine cocktail developed to effectively expand murine hematopoietic stem cells (HSCs) also supports the expansion of leukemia-initiating insertional mutants caused by gammaretroviral gene transfer. We compared 4 protocols to examine the impact of prestimulation and posttransduction culture in STIF in the context of lentiviral gene transfer. Observing 56 transplanted mice for up to 9.5 months, we found consistent engraftment and gene-marking rates after prolonged ex vivo expansion. Although a lentiviral vector with a validated insertional-mutagenic potential was used, longitudinal analysis identifying > 7000 integration sites revealed polyclonal fluctuations, especially in “expanded” groups, with de novo detection of clones even at late time points. Posttransduction expansion in STIF did not enrich clones with insertions in proto-oncogenes but rather increased clonal diversity. Our data indicate that lentiviral transduction in optimized media mediates intact polyclonal hematopoiesis without selection for growth-promoting hits by posttransduction expansion.
Collapse
|
8
|
O’Neill LS, Skinner AM, Woodward JA, Kurre P. Entry kinetics and cell-cell transmission of surface-bound retroviral vector particles. J Gene Med 2010; 12:463-76. [PMID: 20440757 PMCID: PMC2864923 DOI: 10.1002/jgm.1458] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Transduction with recombinant HIV-1 derived lentivirus vectors is a multi-step process initiated by surface attachment and subsequent receptor-directed uptake into the target cell. We previously reported the retention of vesicular stomatitis virus G protein pseudotyped particles on murine progenitor cells and their delayed cell-cell transfer. METHODS To examine the underlying mechanism in more detail, we used a combination of approaches focused on investigating the role of receptor-independent factors in modulating attachment. RESULTS The investigation of synchronized transduction reveals cell-type specific rates of vector particle clearance with substantial delays during particle entry into murine hematopoietic progenitor cells. The observed uptake kinetics from the surface of the 1 degrees cell correlate inversely with the magnitude of transfer to 2 degrees targets, corresponding with our initial observation of preferential cell-cell transfer in the context of brief vector exposures. We further demonstrate that vector particle entry into cells is associated with the cell-type specific abundance of extracellular matrix fibronectin. Residual particle-extracellular fibronectin matrix binding and 2 degrees transfer can be competitively disrupted by heparin exposure without affecting murine progenitor homing and repopulation. CONCLUSIONS Although cellular attachment factors, including fibronectin, aid gene transfer by colocalizing particles to cells and disfavoring early dissociation from targets, they also appear to stabilize particles on the cell surface. The present study highlights the inadvertent consequences for cell entry and cell-cell transfer.
Collapse
Affiliation(s)
- Lee S. O’Neill
- Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - Amy M. Skinner
- Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - Josha A. Woodward
- Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - Peter Kurre
- Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
- Papé Family Pediatric Research Institute, Department of Cell & Developmental Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
9
|
Laje P, Zoltick PW, Flake AW. SLAM-enriched hematopoietic stem cells maintain long-term repopulating capacity after lentiviral transduction using an abbreviated protocol. Gene Ther 2009; 17:412-8. [PMID: 19865179 DOI: 10.1038/gt.2009.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene transfer to long-term repopulating hematopoietic stem cells (HSCs) using integrating viral vectors is an important goal in gene therapy. The SLAM (signaling lymphocyte activation molecule)-family receptors have recently been used for the isolation of highly enriched murine HSCs. This HSC enrichment protocol is relatively simple, and results in an HSC population with comparable repopulating capacity to c-kit(+)lin(-)Sca-1(+) (KSL) HSCs. The capacity to withstand genetic manipulation and, most importantly, to maintain long-term repopulating capacity of SLAM-enriched HSC populations has not been reported. In this study, SLAM-enriched HSCs were assessed for transduction efficiency and in vivo long-term repopulating capacity after lentiviral transduction using an abbreviated transduction protocol and KSL-enriched HSCs as a reference population. SLAM- and KSL-enriched HSCs were efficiently transduced by lentiviral vector using a simple protocol that involves minimal in vitro manipulation and no pre-stimulation. SLAM-HSCs are at least equal to KSL-HSCs with respect to efficiency of transduction and maintenance of long-term repopulating capacity. Although there was a reduction in repopulating capacity related to enrichment and culture manipulations relative to freshly isolated bone marrow (BM) cells, no detrimental effects were identified on long-term competitive capacity related to transduction, as transduced cells maintained stable levels of chimerism in competition with non-transduced cells and freshly isolated BM cells. These results support the SLAM-HSC enrichment protocol as a simple and efficient method for HSC enrichment for gene transfer studies.
Collapse
Affiliation(s)
- P Laje
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA
| | | | | |
Collapse
|
10
|
Peng L, Liu M, Xue YN, Huang SW, Zhuo RX. Transfection and intracellular trafficking characteristics for poly(amidoamine)s with pendant primary amine in the delivery of plasmid DNA to bone marrow stromal cells. Biomaterials 2009; 30:5825-33. [DOI: 10.1016/j.biomaterials.2009.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Accepted: 07/06/2009] [Indexed: 12/18/2022]
|
11
|
Genetic correction of hematopoiesis in Fanconi anemia: the case for a non-HSC-autonomous defect. Mol Ther 2009; 17:1313-5. [PMID: 19644496 DOI: 10.1038/mt.2009.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
12
|
Müller LU, Williams DA. Finding the needle in the hay stack: hematopoietic stem cells in Fanconi anemia. Mutat Res 2009; 668:141-9. [PMID: 19508850 PMCID: PMC2815349 DOI: 10.1016/j.mrfmmm.2009.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 02/09/2009] [Accepted: 03/20/2009] [Indexed: 01/05/2023]
Abstract
Fanconi anemia is a rare bone marrow failure and cancer predisposition syndrome. Childhood onset of aplastic anemia is one of the hallmarks of this condition. Supportive therapy in the form of blood products, androgens, and hematopoietic growth factors may boost blood counts temporarily. However, allogeneic hematopoietic stem cell transplantation (HSCT) currently remains the only curative treatment option for the hematologic manifestations of Fanconi anemia (FA). Here we review current clinical and pre-clinical strategies for treating hematopoietic stem cell (HSC) failure, including the experience with mobilizing and collecting CD34+ hematopoietic stem and progenitor cells as target cells for somatic gene therapy, the current state of FA gene therapy trials, and future prospects for cell and gene therapy.
Collapse
Affiliation(s)
- Lars U.W. Müller
- Department of Medicine, Division of Pediatric Hematology Oncology, Children’s Hospital Boston, and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - David A. Williams
- Department of Medicine, Division of Pediatric Hematology Oncology, Children’s Hospital Boston, and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Millington M, Arndt A, Boyd M, Applegate T, Shen S. Towards a clinically relevant lentiviral transduction protocol for primary human CD34 hematopoietic stem/progenitor cells. PLoS One 2009; 4:e6461. [PMID: 19649289 PMCID: PMC2714083 DOI: 10.1371/journal.pone.0006461] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 06/19/2009] [Indexed: 11/21/2022] Open
Abstract
Background Hematopoietic stem cells (HSC), in particular mobilized peripheral blood stem cells, represent an attractive target for cell and gene therapy. Efficient gene delivery into these target cells without compromising self-renewal and multi-potency is crucial for the success of gene therapy. We investigated factors involved in the ex vivo transduction of CD34+ HSCs in order to develop a clinically relevant transduction protocol for gene delivery. Specifically sought was a protocol that allows for efficient transduction with minimal ex vivo manipulation without serum or other reagents of animal origin. Methodology/Principal Findings Using commercially available G-CSF mobilized peripheral blood (PB) CD34+ cells as the most clinically relevant target, we systematically examined factors including the use of serum, cytokine combinations, pre-stimulation time, multiplicity of infection (MOI), transduction duration and the use of spinoculation and/or retronectin. A self-inactivating lentiviral vector (SIN-LV) carrying enhanced green fluorescent protein (GFP) was used as the gene delivery vehicle. HSCs were monitored for transduction efficiency, surface marker expression and cellular function. We were able to demonstrate that efficient gene transduction can be achieved with minimal ex vivo manipulation while maintaining the cellular function of transduced HSCs without serum or other reagents of animal origin. Conclusions/Significance This study helps to better define factors relevant towards developing a standard clinical protocol for the delivery of SIN-LV into CD34+ cells.
Collapse
Affiliation(s)
| | - Allison Arndt
- Johnson and Johnson Research Pty Ltd., Eveleigh, New South Wales, Australia
| | - Maureen Boyd
- Johnson and Johnson Research Pty Ltd., Eveleigh, New South Wales, Australia
| | - Tanya Applegate
- Johnson and Johnson Research Pty Ltd., Eveleigh, New South Wales, Australia
| | - Sylvie Shen
- Johnson and Johnson Research Pty Ltd., Eveleigh, New South Wales, Australia
- * E-mail:
| |
Collapse
|
14
|
Avoiding lentiviral transduction culture induced MSC senescence. J Cell Mol Med 2009. [DOI: 10.1111/j.1582-4934.2009.00651.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
15
|
Lentiviral-mediated genetic correction of hematopoietic and mesenchymal progenitor cells from Fanconi anemia patients. Mol Ther 2009; 17:1083-92. [PMID: 19277017 DOI: 10.1038/mt.2009.26] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previous clinical trials based on the genetic correction of purified CD34(+) cells with gamma-retroviral vectors have demonstrated clinical efficacy in different monogenic diseases, including X-linked severe combined immunodeficiency, adenosine deaminase deficient severe combined immunodeficiency and chronic granulomatous disease. Similar protocols, however, failed to engraft Fanconi anemia (FA) patients with genetically corrected cells. In this study, we first aimed to correlate the hematological status of 27 FA patients with CD34(+) cell values determined in their bone marrow (BM). Strikingly, no correlation between these parameters was observed, although good correlations were obtained when numbers of colony-forming cells (CFCs) were considered. Based on these results, and because purified FA CD34(+) cells might have suboptimal repopulating properties, we investigated the possibility of genetically correcting unselected BM samples from FA patients. Our data show that the lentiviral transduction of unselected FA BM cells mediates an efficient phenotypic correction of hematopoietic progenitor cells and also of CD34(-) mesenchymal stromal cells (MSCs), with a reported role in hematopoietic engraftment. Our results suggest that gene therapy protocols appropriate for the treatment of different monogenic diseases may not be adequate for stem cell diseases like FA. We propose a new approach for the gene therapy of FA based on the rapid transduction of unselected hematopoietic grafts with lentiviral vectors (LVs).
Collapse
|
16
|
Lee HJ, Lee YS, Kim HS, Kim YK, Kim JH, Jeon SH, Lee HW, Kim S, Miyoshi H, Chung HM, Kim DK. Retronectin enhances lentivirus-mediated gene delivery into hematopoietic progenitor cells. Biologicals 2009; 37:203-9. [PMID: 19264508 DOI: 10.1016/j.biologicals.2009.01.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 01/18/2009] [Accepted: 01/19/2009] [Indexed: 11/26/2022] Open
Abstract
Genetic modification of hematopoietic stem cells holds great promise in the treatment of hematopoietic disorders. However, clinical application of gene delivery has been limited, in part, by low gene transfer efficiency. To overcome this problem, we investigated the effect of retronectin (RN) on lentiviral-mediated gene delivery into hematopoietic progenitor cells (HPCs) derived from bone marrow both in vitro and in vivo. RN has been shown to enhance transduction by promoting colocalization of lentivirus and target cells. We found that RN enhanced lentiviral transfer of the VENUS transgene into cultured c-Kit(+) Lin(-) HPCs. As a complementary approach, in vivo gene delivery was performed by subjecting mice to intra-bone marrow injection of lentivirus or a mixture of RN and lentivirus. We found that co-injection with RN increased the number of VENUS-expressing c-Kit(+) Lin(-) HPCs in bone marrow by 2-fold. Further analysis of VENUS expression in colony-forming cells from the bone marrow of these animals revealed that RN increased gene delivery among these cells by 4-fold. In conclusion, RN is effective in enhancing lentivirus-mediated gene delivery into HPCs.
Collapse
Affiliation(s)
- Hyun-Joo Lee
- Graduate School of Life Science and Biotechnology, Pochon CHA University, CHA Stem Cell Institute, 605 Yeoksam 1-dong, Kangnam-gu, Seoul 135-081, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Affiliation(s)
- Yung-Wei Pan
- Papé Family Pediatric Research Institute, Departments of Pediatrics and Cell & Developmental Biology, Oregon Health & Science UniversityPortland, OR, USA
| | - Peter Kurre
- Papé Family Pediatric Research Institute, Departments of Pediatrics and Cell & Developmental Biology, Oregon Health & Science UniversityPortland, OR, USA
- * Correspondence to: Peter KURRE, M.D., Associate Professor, Pediatrics and Cell & Developmental Biology, 3181 SW Sam Jackson Park Road, Oregon Health & Science University, Division of Pediatric Hematology/Oncology, CDRCP Portland, OR 97239, USA. Tel.: 503–494-0829 Fax: 503–494-0714 E-mail:
| |
Collapse
|
18
|
Modlich U, Schambach A, Li Z, Schiedlmeier B. Murine hematopoietic stem cell transduction using retroviral vectors. Methods Mol Biol 2009; 506:23-31. [PMID: 19110617 DOI: 10.1007/978-1-59745-409-4_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation and gene therapy applications. Their progeny cells carry the genetic information of the HSCs and replenish the blood and immune system. Therefore, in the setting of inherited diseases, transduction of HSCs with retroviral vectors (including gammaretro- and lentiviral vectors) offers the possibility to correct the phenotype in all blood lineages as demonstrated in clinical trials for immunodeficiencies (e.g., X-SCID). In the process of developing gene therapy strategies for patient applications, suitable mouse models for the human gene therapy are important to validate the concept. Stem-cell-enriched populations such as lineage negative cells as the functional equivalent of human CD34(+) cells can be isolated from murine bone marrow and efficiently transduced using retroviral vectors. This chapter provides a step-by-step protocol for retroviral transduction of murine lineage negative cells.
Collapse
Affiliation(s)
- Ute Modlich
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
19
|
Porter CC, DeGregori J. Interfering RNA-mediated purine analog resistance for in vitro and in vivo cell selection. Blood 2008; 112:4466-74. [PMID: 18587011 PMCID: PMC2597122 DOI: 10.1182/blood-2008-03-146571] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 06/17/2008] [Indexed: 12/14/2022] Open
Abstract
The advancement of gene therapy has been slowed, in part, by inefficient transduction of targeted cells and poor long-term engraftment of genetically modified cells. Thus, the ability to select for a desired population of cells within a recipient would be of great benefit for improving gene therapy. Proposed strategies for in vivo cell selection using drug resistance genes have had disappointing outcomes and/or require highly genotoxic medications to be effective. We hypothesized that resistance to purine analogs, a well-tolerated, relatively low-toxicity class of medications, could be provided to cells using interfering RNA against hypoxanthine phosphoribosyl transferase. Using a lentiviral vector, we found that interfering RNA-mediated purine analog resistance (iPAR) provided relative resistance to 6-thioguanine (6TG) in murine hematopoietic cells compared with control- and untransduced cells. iPAR attenuated 6TG-induced G(2)/M checkpoint activation, cell-cycle arrest, and apoptosis. Furthermore, in recipients of transplanted bone marrow cells with iPAR, treatment with 6TG resulted in increased percentages of transduced peripheral blood cells and hematopoietic progenitor cells in the bone marrow. Secondary transplantations resulted in higher hematopoietic contributions from 6TG-treated primary recipients relative to phosphate-buffered saline-treated recipients. These findings indicate that iPAR/6TG can be used for in vivo hematopoietic progenitor cell selection.
Collapse
Affiliation(s)
- Christopher C Porter
- Rick Wilson Center for Cancer and Blood Disorders, The Children's Hospital, Aurora, CO, USA.
| | | |
Collapse
|
20
|
Müller LUW, Milsom MD, Kim MO, Schambach A, Schuesler T, Williams DA. Rapid lentiviral transduction preserves the engraftment potential of Fanca(-/-) hematopoietic stem cells. Mol Ther 2008; 16:1154-1160. [PMID: 18398427 DOI: 10.1038/mt.2008.67] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Accepted: 03/10/2008] [Indexed: 01/02/2023] Open
Abstract
Fanconi anemia (FA) is a rare recessive syndrome, characterized by congenital anomalies, bone marrow failure, and predisposition to cancer. Two earlier clinical trials utilizing gamma-retroviral vectors for the transduction of autologous FA hematopoietic stem cells (HSCs) required extensive in vitro manipulation and failed to achieve detectable long-term engraftment of transduced HSCs. As a strategy for minimizing ex vivo manipulation, we investigated the use of a "rapid" lentiviral transduction protocol in a murine Fanca(-/-) model. Importantly, while this and most murine models of FA fail to completely mimic the human hematopoietic phenotype, we observed a high incidence of HSC transplant engraftment failure and low donor chimerism after conventional transduction (CT) of Fanca(-/-) donor cells. In contrast, rapid transduction (RT) of Fanca(-/-) HSCs preserved engraftment to the level achieved in wild-type cells, resulting in long-term multilineage engraftment of gene-modified cells. We also demonstrate the correction of the characteristic hypersensitivity of FA cells against the cross-linking agent mitomycin C (MMC), and provide evidence for the advantage of using pharmacoselection as a means of further increasing gene-modified cells after RT. Collectively, these data support the use of rapid lentiviral transduction for gene therapy in FA.
Collapse
Affiliation(s)
- Lars U W Müller
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | | | | | |
Collapse
|
21
|
Skinner AM, O'Neill SL, Grompe M, Kurre P. CXCR4 induction in hematopoietic progenitor cells from Fanca(-/-), -c(-/-), and -d2(-/-) mice. Exp Hematol 2008; 36:273-82. [PMID: 18279715 PMCID: PMC2335150 DOI: 10.1016/j.exphem.2007.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Revised: 11/16/2007] [Accepted: 11/19/2007] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Bone marrow failure is a near-universal occurrence in patients with Fanconi anemia (FA) and is thought to result from exhaustion of the hematopoietic stem cell (HSC) pool. Retrovirus-mediated expression of the deficient protein corrects this phenotype and makes FA a candidate disease for HSC-directed gene therapy. However, inherent repopulation deficits and stem cell attrition during conventional transduction culture prevent therapeutic chimerism. MATERIALS AND METHODS We previously reported rapid transduction protocols to limit stem cell losses after ex vivo culture. Here we describe a complementary strategy intended to improve repopulation through upregulation of chemokine receptor (CXCR) 4, a principal factor in hematopoietic homing. RESULTS Using murine models with transgenic disruption of Fanca, -c, and -d2, we found that c-kit(+) and sca-1(+) progenitor cells express levels of CXCR4 comparable with those of wild-type littermates. Lineage-depleted progenitor populations rapidly upregulated CXCR4 transcript and protein in response to cytokine stimulation or hypoxia, regardless of genotype. Hypoxia conditioning of lineage-depleted Fancc(-/-) progenitors also reduced oxidative stress, improved in vitro migration and led to improved chimerism in myeloablated recipients after transplantation. CONCLUSION These studies provide evidence that CXCR4 regulation in progenitor cells from transgenic mice representing multiple FA genotypes is intact and that modulation of homing offers a potential strategy to offset the FA HSC repopulation deficiency.
Collapse
Affiliation(s)
- Amy M Skinner
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Enhancing DNA repair activity of hematopoietic cells by stably integrating gene vectors that express O(6)-methylguanine-DNA-methyltransferase (MGMT) is of major interest for innovative approaches in tumor chemotherapy and for the control of hematopoietic chimerism in the treatment of multiple other acquired or inherited disorders. Crucial determinants of this selection principle are the stringency of treatment with O(6)-alkylating agents and the level of transgenic MGMT expression. Attempts to generate clinically useful MGMT vectors focus on the design of potent expression cassettes, an important component of which is formed by enhancer sequences that are active in primitive as well as more differentiated hematopoietic cells. However, recent studies have revealed that vectors harboring strong enhancer sequences are more likely to induce adverse events related to insertional mutagenesis. Safety-improved vectors that maintain high levels of MGMT expression may be constructed based on the following principles: choice of enhancer-promoter sequences with relatively mild long-distance effects despite a high transcription rate, improved RNA processing (export, stability and translation), and protein design. The need for optimizing MGMT protein design is supported by recent observations suggesting that the P140K mutant of MGMT, developed to be resistant to inhibitors such as O(6)-benzylguanine, may confer a selective disadvantage when expressed at high levels. Here, we provide a review of the literature exploring MGMT expression vectors for bone marrow chemoprotection, and describe experimental evidence suggesting that high expression of MGMT P140K induces a selective disadvantage in the absence of alkylating agents. We conclude that the appropriate design of expression vectors and MGMT protein features will be crucial for the long-term prospects of this promising selection principle.
Collapse
Affiliation(s)
- Axel Schambach
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
23
|
Incani V, Tunis E, Clements BA, Olson C, Kucharski C, Lavasanifar A, Uludag H. Palmitic acid substitution on cationic polymers for effective delivery of plasmid DNA to bone marrow stromal cells. J Biomed Mater Res A 2007; 81:493-504. [PMID: 17340629 DOI: 10.1002/jbm.a.31249] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nonviral gene carriers are actively explored in gene therapy due to safety concerns of the viral carriers. To design effective gene carriers for modification of bone marrow stromal cells (BMSC), an important cell phenotype for clinical application of gene therapy, cationic polymers polyethyleneimine (PEI), and poly-L-Lysine (PLL) were substituted with palmitic acid (PA) via amide linkages. Depending on the reaction conditions, PEI and PLL was substituted with 2.2-5.2 and 13.4-16.2 PA per polymer chain. The PA substituted polymers displayed slightly lower binding efficiency towards a plasmid containing Enhanced Green Fluorescent Protein (pEGFP) in an agarose gel binding assay. The cell binding of PLL-PA, but not PEI-PA, was particularly enhanced, resulting in higher percentage of the cells displaying a significant polymer uptake. pEGFP delivery into the BMSC was also significantly increased with the PLL-PA (vs. PLL), but not PEI-PA (vs. PEI). The transfection efficiency of PLL-PA was significantly higher ( approximately fivefold) than the unmodified polymer. We conclude that PA substitution on PLL provides an effective carrier for transfection of primary cells derived from the bone marrow.
Collapse
Affiliation(s)
- Vanessa Incani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
24
|
Farrell LL, Pepin J, Kucharski C, Lin X, Xu Z, Uludag H. A comparison of the effectiveness of cationic polymers poly-L-lysine (PLL) and polyethylenimine (PEI) for non-viral delivery of plasmid DNA to bone marrow stromal cells (BMSC). Eur J Pharm Biopharm 2006; 65:388-97. [PMID: 17240127 DOI: 10.1016/j.ejpb.2006.11.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2006] [Revised: 11/26/2006] [Accepted: 11/26/2006] [Indexed: 11/22/2022]
Abstract
Bone marrow stromal cells (BMSC) represent an important cell phenotype for pursuit of successful gene therapy. Non-viral methods to enable expression of exogenous genes in BMSC will accelerate clinical application of gene therapy, without the concerns associated with the viral means of gene transfer. Towards this end, this study investigated the potential of cationic polymers poly-L-lysine (PLL) and branched polyethylenimine (PEI) as gene carriers for modification of BMSC. Both polymers rapidly (approximately 30 min) condensed a 4.2 kb Enhanced Green Fluorescent Protein (pEGFP-N2) plasmid into 100-200 nm particles. PLL and PEI were both readily internalized with BMSC with >80% of BMSC exhibiting polymer uptake by flow cytometric analysis. The relative uptake of PEI, however, was significantly higher as compared to the PLL. The majority of the BMSC (>60%) exhibited nuclear presence of the polymers as analyzed by fluorescent microscopy. Although both polymers were able to deliver the pEGFP-N2 into the cells under microscopic evaluation, only a small fraction of the cells (<10%) displayed nuclear localization of the plasmid. Consistent with better uptake, PEI gave a higher delivery of pEGFP-N2 into the BMSC, which resulted in a more sustained expression of the model gene EGFP in short-term (7-day) culture. We conclude that both PLL and PEI readily displayed cellular uptake, but PEI was more effective in delivering plasmid DNA intracellularly, which was likely the underlying basis for a more sustained gene expression.
Collapse
Affiliation(s)
- Laura-Lee Farrell
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alta., Canada
| | | | | | | | | | | |
Collapse
|
25
|
Schambach A, Galla M, Modlich U, Will E, Chandra S, Reeves L, Colbert M, Williams DA, von Kalle C, Baum C. Lentiviral vectors pseudotyped with murine ecotropic envelope: increased biosafety and convenience in preclinical research. Exp Hematol 2006; 34:588-92. [PMID: 16647564 DOI: 10.1016/j.exphem.2006.02.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Revised: 02/06/2006] [Accepted: 02/06/2006] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Lentiviral vectors are increasingly used for preclinical models of gene therapy and other forms of experimental transgenesis. Due to the broad tropism and the ability for concentration by ultracentrifugation, most lentiviral vector preparations are produced using the vesicular stomatitis virus glycoprotein (VSV-g) protein as envelope. Recently, Hanawa and colleagues have demonstrated that the ecotropic envelope protein of murine leukemia viruses allows efficient pseudotyping of HIV-1-derived vector particles. However, this method has found little acceptance, despite potential advantages. MATERIALS AND METHODS We produced lentiviral vectors pseudotyped with murine ecotropic envelope using a four-plasmid transient transfection system and evaluated their performance in murine fibroblasts and hematopoietic cells. RESULTS Titers of lentiviral "ecotropic" supernatants were only slightly lower than those produced with VSV-g, could be concentrated by overnight centrifugation (13,000g), and efficiently transduced murine fibroblasts and hematopoietic cells but not human cells. Our Institutional Biosafety Committee agreed on the production and use of replication-defective lentiviral vectors pseudotyped with murine ecotropic envelope under biosafety level 1 (BL1) conditions with additional BL2 practices. We also obtained useful guidelines for the work with human infectious lentiviral vectors. CONCLUSIONS For the researcher, "ecotropic" lentiviral vectors significantly improve the convenience of daily work, compared to the conditions required for lentiviral pseudotypes that are capable of infecting human cells. High efficiency and superior biosafety in combination with convenient handling will certainly boost the potential applicability of this important vector system.
Collapse
Affiliation(s)
- Axel Schambach
- Department of Hematology, Hemostaseology and Oncology, Hannover Medical School, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|