1
|
Cas9 protein delivery non-integrating lentiviral vectors for gene correction in sickle cell disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:121-132. [PMID: 33816645 PMCID: PMC8005818 DOI: 10.1016/j.omtm.2021.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 12/26/2022]
Abstract
Gene editing with the CRISPR-Cas9 system could revolutionize hematopoietic stem cell (HSC)-targeted gene therapy for hereditary diseases, including sickle cell disease (SCD). Conventional delivery of editing tools by electroporation limits HSC fitness due to its toxicity; therefore, efficient and non-toxic delivery remains crucial. Integrating lentiviral vectors are established for therapeutic gene delivery to engraftable HSCs in gene therapy trials; however, their sustained expression and size limitation preclude their use for CRISPR-Cas9 delivery. Here, we developed a Cas9 protein delivery non-integrating lentiviral system encoding guide RNA and donor DNA, allowing for transient endonuclease function and inclusion of all editing tools in a single vector (all-in-one). We demonstrated efficient one-time correction of the SCD mutation in the endogenous βs-globin gene up to 42% at the protein level (p < 0.01) with the Cas9 protein delivery non-integrating lentiviral all-in-one system without electroporation. Our findings improve prospects for efficient and safe genome editing.
Collapse
|
2
|
Kao CY, Papoutsakis ET. Engineering human megakaryocytic microparticles for targeted delivery of nucleic acids to hematopoietic stem and progenitor cells. SCIENCE ADVANCES 2018; 4:eaau6762. [PMID: 30417099 PMCID: PMC6221511 DOI: 10.1126/sciadv.aau6762] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/04/2018] [Indexed: 06/09/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are important target cells for gene therapy applications. Current genetic modifications of HSPCs rely on viral vectors in vivo or electroporation ex vivo. Here, we developed a nonviral system based on megakaryocytic microparticles (MPs) for targeted delivery of plasmid DNA (pDNA) and small RNAs to HSPCs. We have previously shown that megakaryocytic MPs, the most abundant MPs in blood circulation, target specifically and deliver cargo to HSPCs both in vitro and in vivo. With an optimized electroporation protocol, an average of 4200 plasmid copies per MP were loaded into MP, thus enabling effective delivery of green fluorescent protein (GFP)-encoding pDNA to HSPCs and HSPC nuclei, with up to 81% nuclei containing pDNA. Effective functional small interfering RNA (siRNA) and microRNA (miRNA) delivery were also demonstrated. As patient-specific or generic megakaryocytic MPs can be readily generated and stored frozen, our data suggest that this system has great potential for therapeutic applications targeting HSPCs.
Collapse
Affiliation(s)
- Chen-Yuan Kao
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
| | - Eleftherios T. Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
3
|
Diener Y, Bosio A, Bissels U. Delivery of RNA-based molecules to human hematopoietic stem and progenitor cells for modulation of gene expression. Exp Hematol 2016; 44:991-1001. [PMID: 27576131 DOI: 10.1016/j.exphem.2016.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/01/2016] [Accepted: 08/18/2016] [Indexed: 12/26/2022]
Abstract
Gene modulation of human hematopoietic stem and progenitor cells (HSPCs) harbors great potential for therapeutic application of these cells and presents a versatile tool in basic research to enhance our understanding of HSPC biology. However, stable genetic modification might be adverse, particularly in clinical settings. Here, we review a broad range of approaches to transient, nonviral modulation of protein expression with a focus on RNA-based methods. We compare different delivery methods and describe the usefulness of RNA molecules for overexpression as well as downregulation of proteins in HSPCs.
Collapse
Affiliation(s)
| | | | - Ute Bissels
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany.
| |
Collapse
|
4
|
Wright SE, Rewers-Felkins KA, Quinlin I, Chowdhury NI, Ahmed J, Eldridge PW, Srivastava SK, Pastan I. TGFα-PE38 enhances cytotoxic T-lymphocyte killing of breast cancer cells. Oncol Lett 2014; 7:2113-2117. [PMID: 24932299 PMCID: PMC4049764 DOI: 10.3892/ol.2014.1969] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 12/13/2013] [Indexed: 02/04/2023] Open
Abstract
The aim of the present study was to determine whether the combination of two modalities of immunotherapy, targeting two different tumor antigens, may be feasible and non-toxic, yet enhance the killing of a human breast cancer cell line. The first modality was tumor growth factor α-Pseudomonas exotoxin 38 (TGFα-PE38), which specifically targets and kills tumor cells that express the epidermal growth factor receptor. The second modality was mucin-1 (MUC1)-specific cytotoxic T lymphocytes (CTLs), generated by MUC1 stimulation of peripheral blood mononuclear cells, to target the human breast cancer cell line, MCF7. TGFα-PE38 exhibited specific lysis of the MCF7 cells in a concentration- and time-dependent manner. TGFα-PE38 did not kill the normal hematopoietic stem cells or CTLs. Furthermore, TGFα-PE38 was not inhibitory for the growth or differentiation of the normal human hematopoietic stem cells into erythroid and myeloid colonies. In addition, TGFα-PE38 did not inhibit the killing function of CTLs, either when preincubated or co-incubated with CTLs. Finally, therapeutic enhancement was observed, in that TGFα-PE38 and CTLs were additive in the specific lysis of the MCF7 cells. These two modalities of immunotherapy may be beneficial for humans with breast cancer with or without other therapies, including autologous hematopoietic stem cell transplantation, specifically for purging cancer cells from hematopoietic stem cells prior to transplantation.
Collapse
Affiliation(s)
- Stephen E Wright
- Women's Health Research Institute, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA ; Department of Microbiology and Immunology, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA ; Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA ; Harrington Cancer Center, Amarillo, TX 79106, USA
| | - Kathleen A Rewers-Felkins
- Women's Health Research Institute, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Imelda Quinlin
- Women's Health Research Institute, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Nazrul I Chowdhury
- Women's Health Research Institute, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Jewel Ahmed
- Women's Health Research Institute, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | - Sanjay K Srivastava
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| |
Collapse
|
5
|
Pati S, Kalra OP, Mukhopadhyay A. Foe turned friend: multiple functional roles attributable to hyper-activating stem cell factor receptor mutant in regeneration of the haematopoietic cell compartment. Cell Prolif 2011; 44:10-8. [PMID: 21199006 PMCID: PMC6496452 DOI: 10.1111/j.1365-2184.2010.00713.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Stem cell factor receptor, c-kit, is considered to be the master signalling molecule of haematopoietic stem cells. It develops the orchestral pattern of haematopoietic cell lineages, seen by its varying degree of omnipresence in progenitors, lineage committed and mature cells. We have investigated the effect of over-expressing c-kit on early recovery of the haematopoietic compartment, in irradiated hosts. MATERIALS AND METHODS Normal bone marrow cells (BMCs) were transfected with Kit(wt) (wild-type c-kit) or its variant Kit(mu) (asp814tyr) by electroporation. Lethally irradiated mice were transplanted with normal or transfected congeneic BMCs. The effect of ectopic expression of c-kit on haematopoietic cell recovery was determined by analysing donor-derived cells. Furthermore, effects of both types of c-kit over-expression on progenitor and lineage-committed cells were examined by flow cytometric analysis of Sca-1 and lineage-committed (Lin(+)) cells respectively. RESULTS Hyper-activating Kit(mu) significantly improved recovery of the haematopoietic system in irradiated hosts. In vivo results showed that the donor-derived c-kit(+) cell population was increased to more than 3-fold in the case of Kit(mu)-transfected cells compared to normal and Kit(wt) over-expressing BMCs. In general, survival of progenitor and committed cell was improved in the Kit(mu) over-expressing system compared to the other two cohorts. CONCLUSION These results suggest that recruitment of the hyper-activating variant of c-kit (Kit(mu)) lead to early recovery of the bone marrow of lethally irradiated mice.
Collapse
Affiliation(s)
- S Pati
- Stem Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | | |
Collapse
|
6
|
Ulrich-Vinther M. Gene therapy methods in bone and joint disorders. ACTA ORTHOPAEDICA. SUPPLEMENTUM 2010. [DOI: 10.1080/17453690610046512] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
7
|
Abstract
Dose-limiting toxicity of chemotherapeutic agents, i.e., myelosuppression, can limit their effectiveness. The transfer and expression of drug-resistance genes might decrease the risks associated with acute hematopoietic toxicity. Protection of hematopoietic stem/progenitor cells by transfer of drug-resistance genes provides the possibility of intensification or escalation of antitumor drug doses and consequently an improved therapeutic index. This chapter reviews drug-resistance gene transfer strategies for either myeloprotection or therapeutic gene selection. Selecting candidate drug-resistance gene(s), gene transfer methodology, evaluating the safety and the efficiency of the treatment strategy, relevant in vivo models, and oncoretroviral transduction of human hematopoietic stem/progenitor cells under clinically applicable conditions are described.
Collapse
Affiliation(s)
- Tulin Budak-Alpdogan
- Department of Medicine, The Cancer Institute of New Jersey, Robert Wood Johson Medical School, University of Medicine & Dentistry of New Jersey, New Brunswick, NJ, USA
| | | |
Collapse
|
8
|
Wiehe JM, Ponsaerts P, Rojewski MT, Homann JM, Greiner J, Kronawitter D, Schrezenmeier H, Hombach V, Wiesneth M, Zimmermann O, Torzewski J. mRNA-mediated gene delivery into human progenitor cells promotes highly efficient protein expression. J Cell Mol Med 2007; 11:521-30. [PMID: 17635643 PMCID: PMC3922358 DOI: 10.1111/j.1582-4934.2007.00038.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Gene transfer into human CD34+ haematopoietic progenitor cells (HPC) and multi-potent mesenchymal stromal cells (MSC) is an essential tool for numerous in vitro and in vivo applications including therapeutic strategies, such as tissue engineering and gene therapy. Virus based methods may be efficient, but bear risks like tumorigenesis and activation of immune responses. A safer alternative is non-viral gene transfer, which is considered to be less efficient and accomplished with high cell toxicity. The truncated low affinity nerve growth factor receptor (ÄLNGFR) is a marker gene approved for human in vivo application. Human CD34+ HPC and human MSC were transfected with in vitro-transcribed mRNA for ΔLNGFR using the method of nucleofection. Transfection efficiency and cell viability were compared to plasmid-based nucleofection. Protein expression was assessed using flow cytometry over a time period of 10 days. Nucleofection of CD34+ HPC and MSC with mRNA resulted in significantly higher transfection efficiencies compared to plasmid transfection. Cell differentiation assays were performed after selecting ΔLNGFR positive cells using a fluorescent activating cell sorter. Neither cell differentiation of MSC into chondrocytes, adipocytes and osteoblasts, nor differentiation of HPC into burst forming unit erythroid (BFU-E) colony forming unit-granulocyte, erythrocyte, macrophage and megakaryocyte (CFU-GEMM), and CFU-granulocyte-macrophage (GM) was reduced. mRNA based nucleofection is a powerful, highly efficient and non-toxic approach for transient labelling of human progenitor cells or, via transfection of selective proteins, for transient manipulation of stem cell function. It may be useful to transiently manipulate stem cell characteristics and thus combine principles of gene therapy and tissue engineering.
Collapse
Affiliation(s)
- Juliane M Wiehe
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
- *Correspondence to: Jan TORZEWSKI, University of Ulm; Department of Internal Medicine, II-Cardiology, Robert Koch Str. 8, 89081 Ulm, Germany. Tel: + 49 731 500-45026; Fax: + 49 731 500-45005; E-mail:
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Markus T Rojewski
- Institute for Transfusion Medicine, University of Ulm, and Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Germany
| | - Joerg M Homann
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Jochen Greiner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | | | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University of Ulm, and Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Germany
| | - Vinzenz Hombach
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Markus Wiesneth
- Institute for Transfusion Medicine, University of Ulm, and Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Germany
| | | | - Jan Torzewski
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
- *Correspondence to: Jan TORZEWSKI, University of Ulm; Department of Internal Medicine, II-Cardiology, Robert Koch Str. 8, 89081 Ulm, Germany. Tel: + 49 731 500-45026; Fax: + 49 731 500-45005; E-mail:
| |
Collapse
|
9
|
Papapetrou EP, Ziros PG, Micheva ID, Zoumbos NC, Athanassiadou A. Gene transfer into human hematopoietic progenitor cells with an episomal vector carrying an S/MAR element. Gene Ther 2006; 13:40-51. [PMID: 16094410 DOI: 10.1038/sj.gt.3302593] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Episomally maintained self-replicating systems present attractive alternative vehicles for gene therapy applications. Recent insights into the ability of chromosomal scaffold/matrix attachment regions (S/MARs) to mediate episomal maintenance of genetic elements allowed the development of a small circular episomal vector that functions independently of virally encoded proteins. In this study, we investigated the potential of this vector, pEPI-eGFP, to mediate gene transfer in hematopoietic progenitor cell lines and primary human cells. pEPI-eGFP was episomally maintained and conferred sustained eGFP expression even in nonselective conditions in the human cell line, K562, as well as in primary human fibroblast-like cells. In contrast, in the murine erythroleukemia cell line, MEL, transgene expression was silenced through histone deacetylation, despite the vector's episomal persistence. Hematopoietic semisolid cell colonies derived from transfected human cord blood CD34(+) cells expressed eGFP, albeit at low levels. After 4 weeks, the vector is retained in approximately 1% of progeny cells. Our results provide the first evidence that S/MAR-based plasmids can function as stable episomes in primary human cells, supporting long-term transgene expression. However, they do not display universal behavior in all cell types.
Collapse
Affiliation(s)
- E P Papapetrou
- Department of Biology, Faculty of Medicine, University Hospital of Patras, Rion, Patras, Greece.
| | | | | | | | | |
Collapse
|
10
|
Wiehe JMI, Niesler C, Torzewski J, Zimmermann O, Wiesneth M, Schmitt M, Schwarz K, Döhner H, Hombach V, Greiner J. Efficient transient genetic labeling of human CD34+ progenitor cells for in vivo application. Regen Med 2006; 1:223-34. [PMID: 17465806 DOI: 10.2217/17460751.1.2.223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Genetic labeling of human hematopoietic progenitor cells (HPC) and their consecutive fate-mapping in vivo is an approach to answer intriguing questions in stem cell biology. We recently reported efficient transient genetic labeling of human CD34+ HPC with the truncated low-affinity nerve growth factor receptor (ΔLNGFR) for in vivo application. Here we investigate whether HPC labeling with ΔLNGFR affects lineage-specific cell differentiation, whether ΔLNGFR expression is maintained during lineage-specific cell differentiation and which leukemia cell line might be an appropriate cell culture model for human CD34+ HPC. Human CD34+ peripheral blood stem cells and various leukemia cell lines were characterized by immunophenotyping. Cells were transfected using nucleofection. Hematopoietic differentiation was studied by colony-forming assays. ΔLNGFR expression was assessed using reverse transcription-PCR, immunofluorescence and flow cytometry. Nucleofection was efficient and did not significantly reduce hematopoietic cell differentiation. Mature myeloid cells (CD66b+) derived from human CD34+ HPC and Mutz2 cells maintained ΔLNGFR expression at a high percentage (70 ± 2% and 58 ± 2%, respectively). Mutz2 cells may serve as an in vitro model for human myeloid HPC. The method described herein has been adopted to Good Manufacturing Practices (GMP) guidelines and is ready for in vivo application.
Collapse
|
11
|
Papapetrou EP, Zoumbos NC, Athanassiadou A. Genetic modification of hematopoietic stem cells with nonviral systems: past progress and future prospects. Gene Ther 2006; 12 Suppl 1:S118-30. [PMID: 16231044 DOI: 10.1038/sj.gt.3302626] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Serious unwanted complications provoked by retroviral gene transfer into hematopoietic stem cells (HSCs) have recently raised the need for the development and assessment of alternative gene transfer vectors. Within this context, nonviral gene transfer systems are attracting increasing interest. Their main advantages include low cost, ease of handling and large-scale production, large packaging capacity and, most importantly, biosafety. While nonviral gene transfer into HSCs has been restricted in the past by poor transfection efficiency and transient maintenance, in recent years, biotechnological developments are converting nonviral transfer into a realistic approach for genetic modification of cells of hematopoietic origin. Herein we provide an overview of past accomplishments in the field of nonviral gene transfer into hematopoietic progenitor/stem cells and we point at future challenges. We argue that episomally maintained self-replicating vectors combined with physical methods of delivery show the greatest promise among nonviral gene transfer strategies for the treatment of disorders of the hematopoietic system.
Collapse
Affiliation(s)
- E P Papapetrou
- Department of Biology, Faculty of Medicine, University of Patras, Patras, Greece
| | | | | |
Collapse
|
12
|
Hackett PB, Ekker SC, Largaespada DA, McIvor RS. Sleeping Beauty Transposon‐Mediated Gene Therapy for Prolonged Expression. NON-VIRAL VECTORS FOR GENE THERAPY, SECOND EDITION: PART 2 2005; 54:189-232. [PMID: 16096013 DOI: 10.1016/s0065-2660(05)54009-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Sleeping Beauty (SB) transposon system represents a new vector for non-viral gene transfer that melds advantages of viruses and other forms of naked DNA transfer. The transposon itself is comprised of two inverted terminal repeats of about 340 base pairs each. The SB system directs precise transfer of specific constructs from a donor plasmid into a mammalian chromosome. The excision of the transposon from a donor plasmid and integration into a chromosomal site is mediated by Sleeping Beauty transposase, which can be delivered to cells vita its gene or its mRNA. As a result of its integration in chromosomes, and its lack of viral sequences that are often detected by poorly understood cellular defense mechanisms, a gene in a chromosomally integrated transposon can be expressed over the lifetime of a cell. SB transposons integrate nearly randomly into chromosomes at TA-dinucleotide base pairs although the sequences flanking the TAs can influence the probability of integration at a given site. Although random integration of vectors into human genomes is often thought to raise significant safety issues, evidence to date does not indicate that random insertions of SB transposons represent risks that are equal to those of viral vectors. Here we review the activities of the SB system in mice used as a model for human gene therapy, methods of delivery of the SB system, and its efficacy in ameliorating disorders that model human disease.
Collapse
Affiliation(s)
- Perry B Hackett
- Department of Genetics, Cell Biology and Development Arnold and Mabel Beckman Center for Transposon Research University of Minnesota Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
13
|
Smith SL, Kiss J, Siatskas C, Medin JA, Moldwin RL. Enhanced effect of vascular endothelial growth factor, thrombopoietin peptide agonist, SCF, and Flt3-L on LTC-IC and reporter gene transduction from umbilical cord blood CD34+ cells. Transfusion 2004; 44:438-49. [PMID: 14996204 DOI: 10.1111/j.1537-2995.2003.00661.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hemangioblastic precursors have been identified that give rise to both endothelial cells and HPCs, suggesting that common growth factor requirements may exist. STUDY DESIGN AND METHODS The effect of vascular endothelial growth factor (VEGF) in combination with thrombopoietin peptide agonist (TPOA), Flt-3 L (F), and SCF (S) on long-term culture-initiating cell (LTC-IC), CFU, differentiation, and transduction of cord blood (CB) CD34+ were evaluated up to 4 weeks in culture. RESULTS At Week 4, in cultures containing T/F/S and VEGF, the LTC-IC increased 1000-fold (from 37 +/- 8 to 37,012 +/- 14,329) with a frequency of 3.4 in 10,000 cells. In the T/F/S cultures without VEGF, the LTC-IC increased 675-fold (to 25,086 +/- 12,102) with a frequency of one LTC-IC in 10,000 cells. The addition of VEGF significantly increased (p < 0.05) the LTC-IC per 10,000 CB CD34+ cells. Transduction with reporter gene enhanced green fluorescent protein (EGFP), resulted in an increase in EGFP+ CFU at Week 1 and EGFP + LTC-IC at Weeks 1 and 4. The cells maintained their multilineage expression when cultured in conditions for erythroid, myeloid, or megakaryocytic differentiation. Peak percentage EGFP coexpression of GlyA and CD11b was 51 +/- 6 percent and 63 +/- 15 percent, respectively, at Week 2, while CD41a was 34 +/- 17 percent at Week 4. CONCLUSION T/F/S and VEGF have an enhanced effect on total LTC-IC output and frequency but do not appear to significantly alter the differentiation or transducibility characteristics of CB HPCs in vitro.
Collapse
Affiliation(s)
- Stephen L Smith
- Institute for Transfusion Medicine, Stem Cell Services, Glenview, Illinois, USA.
| | | | | | | | | |
Collapse
|
14
|
Weissinger F, Reimer P, Waessa T, Buchhofer S, Schertlin T, Kunzmann V, Wilhelm M. Gene transfer in purified human hematopoietic peripheral-blood stem cells by means of electroporation without prestimulation. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2003; 141:138-49. [PMID: 12577050 DOI: 10.1067/mlc.2003.14] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gene transfer in hematopoietic stem cells (HSCs) is an important tool, exploring regulation of the hematopoietic system and understanding the development and expansion of malignant cell clones. It is also a mandatory step for gene therapy of hematopoietic disorders. Although retroviral transduction of HSCs is effective, prestimulation of cells is generally required, also inducing differentiation of HSCs. Furthermore, the risk of viral recombination and insertional mutagenesis cannot be ruled out. Potential advantages of nonviral transfection are biosafety and easy management. However, experience in nonviral methods for transfecting peripheral-blood stem cells (PBSCs) is limited. To avoid differentiation, we evaluated the efficiency of gene transfer by means of electroporation without cytokine prestimulation. Compared with prestimulated (stem-cell factor, granulocyte-colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, interleukin-3, interleukin-6, erythropoietin, and monoclonal antibody to transforming growth factor-beta), transfection of thawed nonstimulated PBSCs was equally efficient, with a median transfection rate of 3.7%, transfection efficiency of 0.8%, and survival of 19.5% (n = 5). With freshly isolated HSCs, the rate of transfected cells could be increased to a median of 27.0% (range 8.3%-31.0%), transfection efficiency of 6.9% (range 4.5%-12.6%), and survival of 43% (range 22%-64%) (n = 5). However, the percentage of transfected cells declined with time; almost no cells were detectable by day 11. One cause for the lack of long-term expression of the heterologous gene in this system was induction of apoptosis in transgenic PBSCs, shown by up-regulation of CD95 (FAS antigen).
Collapse
Affiliation(s)
- Florian Weissinger
- Medizinische Poliklinik der Julius-Maximilians-Universität, Würzburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
15
|
Hui SW. The application of electroporation to transfect hematopoietic cells and to deliver drugs and vaccines transcutaneously for cancer treatment. Technol Cancer Res Treat 2002; 1:373-84. [PMID: 12625763 DOI: 10.1177/153303460200100508] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Electroporation and the associated phenomenon of electrofusion have been widely adapted as tools to a broad range of biomedical research and therapy. In this article, we summarize our adaptation of the electroporation and electrofusion technology in two fronts of cancer research and treatment. The first is genetic manipulation of hematopoietic cells for the purpose of cancer treatment. High efficiency transfection methods have been developed to transfect NK cells, peripheral blood stem cells, and bone marrow derived dendritic cells. Hybrids of tumor cells and bone marrow derived dendritic cells have been formed by electrofusion for the purpose of tumor vaccines. The second front is the use of transcutaneous electroporation to deliver anticancer drugs and vaccines across the skin. Methods to extend the upper molecular weight limit of transcutaneous electroporation have been developed. The pro-photosensitizer drug, delta-amino levulinic acid, the anticancer drug methotrexate, and peptide vaccines designed for cancer prevention and immunotherapy have been delivered transcutaneously by electroporation. These studies hold promise for the treatment of cancers in human.
Collapse
Affiliation(s)
- Sek-Wen Hui
- Molecular and Cellular Biophysics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| |
Collapse
|
16
|
Wu MH, Smith SL, Dolan ME. High efficiency electroporation of human umbilical cord blood CD34+ hematopoietic precursor cells. Stem Cells 2002; 19:492-9. [PMID: 11713340 DOI: 10.1634/stemcells.19-6-492] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human umbilical cord blood provides an alternative source of hematopoietic cells for purposes of transplantation or ex vivo genetic modification. The objective of this study was to evaluate electroporation as a means to introduce foreign genes into human cord blood CD34+ cells and evaluate gene expression in CD34+/CD38(dim) and committed myeloid progenitors (CD33+, CD11b+). CD34+ cells were cultured in X-VIVO 10 supplemented with thrombopoietin, stem cell factor, and Flt-3 ligand. Electroporation efficiency and cell viability measured by flow cytometry using enhanced green fluorescent protein (EGFP) as a reporter indicated 31% +/- 2% EGFP+ /CD34+ efficiency and 77% +/- 3% viability as determined 48 hours post-electroporation. The addition of allogeneic cord blood plasma increased the efficiency to 44% +/- 5% with no effect on viability. Of the total CD34+ cells 48 hours post-electroporation, 20% were CD38(dim)/EGFP+. CD34+ cells exposed to interleukin-3, GM-CSF and G-CSF for an additional 11 days differentiated into CD33+ and CD11b+ cells, and 9% +/- 3% and 8% +/- 7% were expressing the reporter gene, respectively. We show that electroporation can be used to introduce foreign genes into early hematopoietic stem cells (CD34+/CD38(dim)), and that the introduced gene is functionally expressed following expansion into committed myeloid progenitors (CD33+, CD11b+) in response to corresponding cytokines. Further investigation is needed to determine the transgene expression in functional terminal cells derived from the genetically modified CD34+ cells, such as T cells and dendritic cells.
Collapse
Affiliation(s)
- M H Wu
- Section of Hematology-Oncology, Department of Medicine and Cancer Research Center, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | | | | |
Collapse
|
17
|
Wu MH, Smith SL, Danet GH, Lin AM, Williams SF, Liebowitz DN, Dolan ME. Optimization of culture conditions to enhance transfection of human CD34+ cells by electroporation. Bone Marrow Transplant 2001; 27:1201-9. [PMID: 11551032 DOI: 10.1038/sj.bmt.1703054] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability to culture CD34+ stem cells, while maintaining their pluripotency, is essential for manipulations such as gene transfection for therapeutic trials. Human peripheral blood (PB) CD34+ cells (> or = 90% purity) were cultured for up to 4 days in serum-free culture medium supplemented with thrombopoietin (TPO), stem cell factor (SCF), Flt-3 ligand (Flt-3L), with or without PIXY321 (IL-3/GM-CSF fusion protein) and human serum. The CD34 mean fluorescence intensity (MFI) and cell cycle status were evaluated daily using flow cytometry and hypotonic propidium iodide. Prior to culture (day 0), 97.0 +/- 0.9%, 1.9 +/- 0.3% and 1.0 +/- 0.6% of the selected CD34+ cells were in G0-G1, S-phase, or G2-M, respectively. After 2-4 days in culture with TPO/SCF/Flt-3L, there was an increase in the percent of cells in S-phase to 26.4 +/- 0.1% without significant loss of CD34 MFI. The addition of PIXY321 increased.the percentage of CD34+ cells in S-phase to 36.3 +/- 4.0%, but the CD34 MFI and numbers of CFU (colony-forming units) were significantly decreased at day 3 when cultured with PIXY321 or various recombinant cytokine combinations that included IL-3 and IL-6. There is an increase from day 0 to day 4 in the percentages of CD34+ with CD38-, HLA-DR-, and c-kit(low), but not Thy-1+ cells. Electroporation with EGFP reporter gene showed that 1-2 days of pre-stimulation in X-VIVO 10 supplemented with TPO/SCF/Flt-3L was necessary and sufficient for efficient transfection. Flow cytometry analysis demonstrated that 22% of the viable cells are CD34+/EGFP+ 48 h post electroporation. The introduced reporter gene appears to be stable as determined by EGFP+/LTC-IC (long-term colony-initiating cells), at 30-40 positive colonies (16 +/- 7%) per 1 x 10(5) electroporated CD34+ cells.
Collapse
Affiliation(s)
- M H Wu
- Department of Medicine, and Cancer Research Center, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|