1
|
Namipashaki A, Pugsley K, Liu X, Abrehart K, Lim SM, Sun G, Herold MJ, Polo JM, Bellgrove MA, Hawi Z. Integration of xeno-free single-cell cloning in CRISPR-mediated DNA editing of human iPSCs improves homogeneity and methodological efficiency of cellular disease modeling. Stem Cell Reports 2023; 18:2515-2527. [PMID: 37977144 PMCID: PMC10724053 DOI: 10.1016/j.stemcr.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
The capability to generate induced pluripotent stem cell (iPSC) lines, in tandem with CRISPR-Cas9 DNA editing, offers great promise to understand the underlying genetic mechanisms of human disease. The low efficiency of available methods for homogeneous expansion of singularized CRISPR-transfected iPSCs necessitates the coculture of transfected cells in mixed populations and/or on feeder layers. Consequently, edited cells must be purified using labor-intensive screening and selection, culminating in inefficient editing. Here, we provide a xeno-free method for single-cell cloning of CRISPRed iPSCs achieving a clonal survival of up to 70% within 7-10 days. This is accomplished through improved viability of the transfected cells, paralleled with provision of an enriched environment for the robust establishment and proliferation of singularized iPSC clones. Enhanced cell survival was accompanied by a high transfection efficiency exceeding 97%, and editing efficiencies of 50%-65% for NHEJ and 10% for HDR, indicative of the method's utility in stem cell disease modeling.
Collapse
Affiliation(s)
- Atefeh Namipashaki
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Kealan Pugsley
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Xiaodong Liu
- Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Kirra Abrehart
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Sue Mei Lim
- Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Guizhi Sun
- Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Marco J Herold
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jose M Polo
- Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia; Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Mark A Bellgrove
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Ziarih Hawi
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
Li XL, Li GH, Fu J, Fu YW, Zhang L, Chen W, Arakaki C, Zhang JP, Wen W, Zhao M, Chen WV, Botimer GD, Baylink D, Aranda L, Choi H, Bechar R, Talbot P, Sun CK, Cheng T, Zhang XB. Highly efficient genome editing via CRISPR-Cas9 in human pluripotent stem cells is achieved by transient BCL-XL overexpression. Nucleic Acids Res 2019; 46:10195-10215. [PMID: 30239926 PMCID: PMC6212847 DOI: 10.1093/nar/gky804] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022] Open
Abstract
Genome editing of human induced pluripotent stem cells (iPSCs) is instrumental for functional genomics, disease modeling, and regenerative medicine. However, low editing efficiency has hampered the applications of CRISPR–Cas9 technology in creating knockin (KI) or knockout (KO) iPSC lines, which is largely due to massive cell death after electroporation with editing plasmids. Here, we report that the transient delivery of BCL-XL increases iPSC survival by ∼10-fold after plasmid transfection, leading to a 20- to 100-fold increase in homology-directed repair (HDR) KI efficiency and a 5-fold increase in non-homologous end joining (NHEJ) KO efficiency. Treatment with a BCL inhibitor ABT-263 further improves HDR efficiency by 70% and KO efficiency by 40%. The increased genome editing efficiency is attributed to higher expressions of Cas9 and sgRNA in surviving cells after electroporation. HDR or NHEJ efficiency reaches 95% with dual editing followed by selection of cells with HDR insertion of a selective gene. Moreover, KO efficiency of 100% can be achieved in a bulk population of cells with biallelic HDR KO followed by double selection, abrogating the necessity for single cell cloning. Taken together, these simple yet highly efficient editing strategies provide useful tools for applications ranging from manipulating human iPSC genomes to creating gene-modified animal models.
Collapse
Affiliation(s)
- Xiao-Lan Li
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Guo-Hua Li
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Juan Fu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian 116044, China.,Department of Obstetrics and Gynecology, the First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Ya-Wen Fu
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Lu Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Wanqiu Chen
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Cameron Arakaki
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jian-Ping Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin 300020, China
| | - Wei Wen
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Mei Zhao
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | | | - Gary D Botimer
- Department of Orthopaedic Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - David Baylink
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Leslie Aranda
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hannah Choi
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Rachel Bechar
- UCR Stem Cell Center and Core, University of California at Riverside, Riverside, CA 92521, USA
| | - Prue Talbot
- UCR Stem Cell Center and Core, University of California at Riverside, Riverside, CA 92521, USA
| | - Chang-Kai Sun
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian 116044, China.,Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China.,State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China.,Collaborative Innovation Center for Cancer Medicine, Tianjin 300020, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
3
|
Kirikovich SS, Taranov OS, Omigov VV, Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Bogachev SS. Ultrastructural analysis of the Krebs-2 ascites cancer cells treated with extracellular double-stranded DNA preparation. Ultrastruct Pathol 2019; 43:56-65. [PMID: 30758240 DOI: 10.1080/01913123.2019.1575499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Electron-microscopic analysis of the ultrastructure of the Krebs-2 carcinoma ascites cells in the first 90 min immediately after their exposure to fragmented double-stranded DNA has been performed. Morphological attributes of the treated cancer cells indicate the induction in these cells of destructive processes of presumably apoptotic type. The predominance of dystrophic-destructive changes in cells after the addition of DNA is supposed to be a consequence of the disturbance in metabolic processes caused by the experimental action.
Collapse
Affiliation(s)
- Svetlana S Kirikovich
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia
| | - Oleg S Taranov
- b State Research Center of Virology and Biotechnology VECTOR , Rospotrebnadzor , Novosibirsk region , Russia
| | - Vladimir V Omigov
- b State Research Center of Virology and Biotechnology VECTOR , Rospotrebnadzor , Novosibirsk region , Russia
| | - Ekaterina A Potter
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia
| | - Evgenia V Dolgova
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia
| | - Anastasia S Proskurina
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia
| | - Yaroslav R Efremov
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia.,c Department of natural sciences , Novosibirsk State University , Novosibirsk , Russia
| | - Sergey S Bogachev
- a Institute of Cytology and Genetics , Siberian Branch of the Russian Academy of Sciences , Novosibirsk , Russia
| |
Collapse
|
4
|
Brown AJ, Kalsi D, Fernandez-Martell A, Cartwright J, Barber NOW, Patel YD, Turner R, Bryant CL, Johari YB, James DC. Expression Systems for Recombinant Biopharmaceutical Production by Mammalian Cells in Culture. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527699124.ch13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Adam J. Brown
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Devika Kalsi
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Joe Cartwright
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Nicholas O. W. Barber
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yash D. Patel
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Claire L. Bryant
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yusuf B. Johari
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - David C. James
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| |
Collapse
|
5
|
Achyut BR, Arbab AS. Taming immune suppressor: application of myeloid-derived suppressor cells in anti-cancer gene therapy. Transl Cancer Res 2017; 6:S160-S162. [PMID: 28503409 DOI: 10.21037/tcr.2017.02.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Bhagelu R Achyut
- Tumor Angiogenesis Lab, Department of Biochemistry and Molecular Biology, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Ali S Arbab
- Tumor Angiogenesis Lab, Department of Biochemistry and Molecular Biology, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
6
|
How transient alterations of organelles in mammalian cells submitted to electric field may explain some aspects of gene electrotransfer process. Bioelectrochemistry 2016; 112:166-72. [DOI: 10.1016/j.bioelechem.2016.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/10/2016] [Accepted: 02/17/2016] [Indexed: 11/22/2022]
|
7
|
In vitro exploration of a myeloid-derived suppressor cell line as vehicle for cancer gene therapy. Cancer Gene Ther 2016; 24:149-155. [PMID: 27857057 DOI: 10.1038/cgt.2016.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
Recent research indicates that cell-mediated gene therapy can be an interesting method to obtain intratumoral expression of therapeutic proteins. This paper explores the possibility of using transfected myeloid-derived suppressor cells (MDSCs), derived from a murine cell line, as cellular vehicles for transporting plasmid DNA (pDNA) encoding interleukin-12 (IL-12) to tumors. Transfecting these cells via electroporation caused massive cell death. This was not due to electroporation-induced cell damage, but was mainly the result of the intracellular presence of plasmids. In contrast, pDNA transfection using Lipofectamine 2000 (LF2000) did not result in a significant loss of viability. Differences in delivery mechanism may explain the distinctive effects on cell viability. Indeed, electroporation is expected to cause a rapid and massive influx of pDNA resulting in cytosolic pDNA levels that most likely surpass the activation threshold of the intracellular DNA sensors leading to cell death. In contrast, a more sustained intracellular release of the pDNA is expected with LF2000. After lipofection with LF2000, 56% of the MDSCs were transfected and transgene expression lasted for at least 24 h. Moreover, biologically relevant amounts of IL-12 were produced by the MDSCs after lipofection with an IL-12 encoding pDNA. In addition, IL-12 transfection caused a significant upregulation of CD80 and considerably reduced the immunosuppressive capacity of the MDSCs. IL-12-transfected MDSCs were still able to migrate to tumor cells, albeit that lipofection of the MDSCs seemed to slightly decrease their migration capacity.
Collapse
|
8
|
Bjurström CF, Mojadidi M, Phillips J, Kuo C, Lai S, Lill GR, Cooper A, Kaufman M, Urbinati F, Wang X, Hollis RP, Kohn DB. Reactivating Fetal Hemoglobin Expression in Human Adult Erythroblasts Through BCL11A Knockdown Using Targeted Endonucleases. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e351. [PMID: 28131278 PMCID: PMC5023398 DOI: 10.1038/mtna.2016.52] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 04/18/2016] [Indexed: 12/17/2022]
Abstract
We examined the efficiency, specificity, and mutational signatures of zinc finger nucleases (ZFNs), transcriptional activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 systems designed to target the gene encoding the transcriptional repressor BCL11A, in human K562 cells and human CD34+ progenitor cells. ZFNs and TALENs were delivered as in vitro transcribed mRNA through electroporation; CRISPR/Cas9 was codelivered by Cas9 mRNA with plasmid-encoded guideRNA (gRNA) (pU6.g1) or in vitro transcribed gRNA (gR.1). Analyses of efficacy revealed that for these specific reagents and the delivery methods used, the ZFNs gave rise to more allelic disruption in the targeted locus compared to the TALENs and CRISPR/Cas9, which was associated with increased levels of fetal hemoglobin in erythroid cells produced in vitro from nuclease-treated CD34+ cells. Genome-wide analysis to evaluate the specificity of the nucleases revealed high specificity of this specific ZFN to the target site, while specific TALENs and CRISPRs evaluated showed off-target cleavage activity. ZFN gene-edited CD34+ cells had the capacity to engraft in NOD-PrkdcSCID-IL2Rγnull mice, while retaining multi-lineage potential, in contrast to TALEN gene-edited CD34+ cells. CRISPR engraftment levels mirrored the increased relative plasmid-mediated toxicity of pU6.g1/Cas9 in hematopoietic stem/progenitor cells (HSPCs), highlighting the value for the further improvements of CRISPR/Cas9 delivery in primary human HSPCs.
Collapse
Affiliation(s)
- Carmen F Bjurström
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Michelle Mojadidi
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - John Phillips
- Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Caroline Kuo
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, California, USA
| | - Stephen Lai
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Georgia R Lill
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Aaron Cooper
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Michael Kaufman
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Fabrizia Urbinati
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, Los Angeles, California, USA
| | - Roger P Hollis
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Donald B Kohn
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA; Department of Pediatrics, University of California, Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
9
|
Zhao X, Huang X, Wang X, Wu Y, Eisfeld AK, Schwind S, Gallego-Perez D, Boukany PE, Marcucci GI, Lee LJ. Nanochannel Electroporation as a Platform for Living Cell Interrogation in Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500111. [PMID: 27980918 PMCID: PMC5115302 DOI: 10.1002/advs.201500111] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/03/2015] [Indexed: 05/29/2023]
Abstract
A living cell interrogation platform based on nanochannel electroporation is demonstrated with analysis of RNAs in single cells. This minimally invasive process is based on individual cells and allows both multi-target analysis and stimulus-response analysis by sequential deliveries. The unique platform possesses a great potential to the comprehensive and lysis-free nucleic acid analysis on rare or hard-to-transfect cells.
Collapse
Affiliation(s)
- Xi Zhao
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA; William G. Lowrie, Department of Chemical and Biomolecular Engineering The Ohio State University Columbus OH 43210 USA
| | - Xiaomeng Huang
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA
| | - Xinmei Wang
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA
| | - Yun Wu
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA
| | - Ann-Kathrin Eisfeld
- Department of Internal Medicine Comprehensive Cancer Center The Ohio State University Columbus OH 43210 USA
| | - Sebastian Schwind
- Department of Internal Medicine Comprehensive Cancer Center The Ohio State University Columbus OH 43210 USA
| | - Daniel Gallego-Perez
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA
| | - Pouyan E Boukany
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA
| | - Guido I Marcucci
- Department of Internal Medicine Comprehensive Cancer Center The Ohio State University Columbus OH 43210 USA
| | - Ly James Lee
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices The Ohio State University Columbus OH 43212 USA; William G. Lowrie, Department of Chemical and Biomolecular Engineering The Ohio State University Columbus OH 43210 USA
| |
Collapse
|
10
|
García-Sánchez T, Guitart M, Rosell-Ferrer J, Gómez-Foix AM, Bragós R. A new spiral microelectrode assembly for electroporation and impedance measurements of adherent cell monolayers. Biomed Microdevices 2014; 16:575-90. [DOI: 10.1007/s10544-014-9860-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
11
|
Hyland KA, Olson ER, Clark KJ, Aronovich EL, Hackett PB, Blazar BR, Tolar J, Scott McIvor R. Sleeping Beauty-mediated correction of Fanconi anemia type C. J Gene Med 2012; 13:462-9. [PMID: 21766398 DOI: 10.1002/jgm.1589] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The Sleeping Beauty (SB) transposon system can insert defined sequences into chromosomes to direct the extended expression of therapeutic genes. Our goal is to develop the SB system for nonviral complementation of Fanconi anemia (FA), a rare autosomal recessive disorder accompanied by progressive bone marrow failure. METHODS We used a CytoPulse electroporation system (CytoPulse, Glen Burnie, MD, USA) to introduce SB transposons into human lymphoblastoid cells (LCL) derived from both Fanconi anemia type C (FA-C) defective and normal patients. Correction of the FA-C defect was assessed by resistance to mitomycin C, a DNA-crosslinking agent. RESULTS Culture of both cell types with the antioxidant N-acetyl- l-cysteine improved cell viability after electroporation. Co-delivery of enhanced green fluorescent protein (GFP) transposon with SB100X transposase-encoding plasmid supported a 50- to 90-fold increase in stable GFP expression compared to that observed in the absence of SB100X for normal LCL, but in FA-C defective LCL SB100X enhancement of stable GFP-expression was a more moderate five- to 13-fold. SB-mediated integration and expression of the FA-C gene was demonstrated by the emergence of a mitomycin C-resistant population bearing characteristic transposon-chromosome junction sequences and exhibiting a mitomycin dose response identical to that of normal LCL. CONCLUSIONS The SB transposon system achieved stable expression of therapeutic FA-C genes, complementing the genetic defect in patient-derived cells by nonviral gene transfer.
Collapse
|
12
|
Faurie C, Rebersek M, Golzio M, Kanduser M, Escoffre JM, Pavlin M, Teissie J, Miklavcic D, Rols MP. Electro-mediated gene transfer and expression are controlled by the life-time of DNA/membrane complex formation. J Gene Med 2010; 12:117-25. [PMID: 19941315 DOI: 10.1002/jgm.1414] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electroporation is a physical method used to transfer molecules into cells and tissues. Clinical applications have been developed for antitumor drug delivery. Clinical trials of gene electrotransfer are under investigation. However, knowledge about how DNA enters cells is not complete. By contrast to small molecules that have direct access to the cytoplasm, DNA forms a long lived complex with the plasma membrane and is transferred into the cytoplasm with a considerable delay. METHODS To increase our understanding of the key step of DNA/membrane complex formation, we investigated the dependence of DNA/membrane interaction and gene expression on electric pulse polarity and repetition frequency. RESULTS We observed that both are affected by reversing the polarity and by increasing the repetition frequency of pulses. The results obtained in the present study reveal the existence of two classes of DNA/membrane interaction: (i) a metastable DNA/membrane complex from which DNA can leave and return to external medium and (ii) a stable DNA/membrane complex, where DNA cannot be removed, even by applying electric pulses of reversed polarity. Only DNA belonging to the second class leads to effective gene expression. CONCLUSIONS The life-time of DNA/membrane complex formation is of the order of 1 s and has to be taken into account to improve protocols of electro-mediated gene delivery.
Collapse
Affiliation(s)
- Cécile Faurie
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Buzder T, Yin X, Wang X, Banfalvi G, Basnakian AG. Uptake of foreign nucleic acids in kidney tubular epithelial cells deficient in proapoptotic endonucleases. DNA Cell Biol 2009; 28:435-42. [PMID: 19558214 DOI: 10.1089/dna.2008.0850] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Degradation of DNA during gene delivery is an obstacle for gene transfer and for gene therapy. DNases play a major role in degrading foreign DNA. However, which of the DNases are involved and whether their inactivation can improve gene delivery have not been studied. We have recently identified deoxyribonuclease I (DNase I) and endonuclease G (EndoG) as the major degradative enzymes in the mouse kidney proximal tubule epithelial (TKPTS) cells. In this study, we used immortalized mouse TKPTS cells and primary tubular epithelial cells isolated from DNase I or EndoG knockout (KO) mice and examined the degradation of plasmid DNA during its uptake. DNase I and EndoG KO cells showed a higher rate of transfection by pECFP-N1 plasmid than wild-type cells. In addition, EndoG KO cells prevented the uptake of fluorescent-labeled RNA. Complete inhibition of secreted DNase I by G-actin did not improve plasmid transfection, indicating that only intracellular DNase I affects DNA stability. Data demonstrate the importance of DNase I and EndoG in host cell defense against gene and RNA delivery to renal tubular epithelial cells in vitro.
Collapse
Affiliation(s)
- Timea Buzder
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
Chronic lymphocytic leukemia (CLL) consists of at least two major prognostic subgroups, characterized by different cellular and molecular markers. This observation sparked studies on the function and clinical importance of these markers. In order to address their function adequately, an efficient and reliable method for gene transfer is needed. In this study, we compared efficiency and utility of different gene transfer techniques in CLL. Lenti-, retro- and adenoviral transduction did not yield appreciable numbers of marker gene enhanced green fluorescent protein (EGFP) positive CLL cells, despite various prestimulation protocols. Efficient transgene expression was observed after nucleofection of CLL cells with plasmid DNA, at the expense of low survival rates. After optimization, electroporation of in vitro transcribed mRNA yielded up to 90% EGFP+CLL cells without affecting survival. Transgene expression remained detectable for at least 2 weeks after electroporation. Furthermore, we could demonstrate overexpression of ZAP70 and of a ZAP70-EGFP fusion protein after electroporation with ZAP70 or ZAP70-EGFP mRNA. We conclude that mRNA electroporation is a novel and straightforward method for highly efficient gene transfer in CLL. The application of this technique should facilitate functional studies on CLL cells, as well as clinical research.
Collapse
|
15
|
Seiffert M, Stilgenbauer S, Döhner H, Lichter P. Efficient nucleofection of primary human B cells and B-CLL cells induces apoptosis, which depends on the microenvironment and on the structure of transfected nucleic acids. Leukemia 2007; 21:1977-83. [PMID: 17637809 DOI: 10.1038/sj.leu.2404863] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Accumulation of neoplastic cells in B-cell chronic lymphocytic leukemia (B-CLL) is thought to be due to intrinsic defects in the apoptotic machinery of the leukemic cells or to an altered, survival-stimulating microenvironment in vivo. Despite their long survival in vivo, B-CLL cells undergo rapid spontaneous apoptosis ex vivo. To maintain survival in vitro, we established a coculture system using the human bone marrow-derived stromal cell line HS-5. The microenvironment in these cocultures lead to B-CLL cell survival for at least several months and therefore provided a tool for valid in vitro analysis, mimicking the in vivo situation. Although primary B lymphocytes are notoriously resistant to most gene transfer techniques, we achieved high transfection efficiency and cell viability in this coculture system by using a nucleofection-based strategy. Surprisingly, the introduction of circular plasmid DNA into B cells and B-CLL cells induced rapid apoptosis, which was independent of the type of transgene used, but dependent on the DNA concentration. However, transfection of these cells with mRNA was highly efficient and resulted in sustained cell viability and potent transgene expression. The described procedure represents a new approach to study gene function in primary B cells and B-CLL cells.
Collapse
Affiliation(s)
- M Seiffert
- Department for Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | | | | | | |
Collapse
|
16
|
Gray JS, Pestka JJ. Transcriptional regulation of deoxynivalenol-induced IL-8 expression in human monocytes. Toxicol Sci 2007; 99:502-11. [PMID: 17636245 DOI: 10.1093/toxsci/kfm182] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The trichothecene mycotoxin deoxynivalenol (DON), commonly present in contaminated grains worldwide, induces expression of the chemokine interleukin (IL)-8 in human monocytes. The purpose of this study was to test the hypothesis that DON modulates transcriptional and posttranscriptional regulation of IL-8 expression in the U937 human monocyte model. When U937 cells were transfected with a wild-type IL-8 promoter luciferase construct (-162/+44 IL-8 LUC) and incubated with DON (1 mug/ml) or the positive control, lipopolysaccharide (LPS) (1 mug/ml), there was a significant increase in luciferase expression. Mutation of the nuclear factor-kappaB (NF-kappaB) binding site significantly impaired both DON- and LPS-induced luciferase expression. In contrast, mutating the activator protein-1 binding site resulted in significantly increased DON- and LPS-induced luciferase expression. CCAAT/enhancer-binding protein beta, octamer-1, or NF-kappaB repressing factor binding site mutations did not affect DON-induced luciferase activity. Consistent with reporter studies, the NF-kappaB inhibitor caffeic acid phenethyl ester completely ablated both DON-induced IL-8 mRNA and protein expression. When NF-kappaB subunit binding to a specific IL-8 promoter probe was evaluated by enzyme-linked immunosorbent assay (ELISA), DON was observed to increase p65 binding by 21-fold, have no effect on p50 binding and decrease p52 binding. DON was not found to stabilize IL-8 mRNA in U937 cells. Taken together, these data suggest that DON-induced IL-8 expression is likely to be mediated at the transcriptional level by NF-kappaB, specifically p65, but does not appear to involve mRNA stabilization.
Collapse
Affiliation(s)
- Jennifer S Gray
- Department of Microbiology and Molecular Genetics, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824-1224, USA
| | | |
Collapse
|
17
|
Li LH, Biagi E, Allen C, Shivakumar R, Weiss JM, Feller S, Yvon E, Fratantoni JC, Liu LN. Rapid and efficient nonviral gene delivery of CD154 to primary chronic lymphocytic leukemia cells. Cancer Gene Ther 2006; 13:215-24. [PMID: 16082377 DOI: 10.1038/sj.cgt.7700883] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interactions between CD40 and CD40 ligand (CD154) are essential in the regulation of both humoral and cellular immune responses. Forced expression of human CD154 in B chronic lymphocytic leukemia (B-CLL) cells can upregulate costimulatory and adhesion molecules and restore antigen-presenting capacity. Unfortunately, B-CLL cells are resistant to direct gene manipulation with most currently available gene transfer systems. In this report, we describe the use of a nonviral, clinical-grade, electroporation-based gene delivery system and a standard plasmid carrying CD154 cDNA, which achieved efficient (64+/-15%) and rapid (within 3 h) transfection of primary B-CLL cells. Consistent results were obtained from multiple human donors. Transfection of CD154 was functional in that it led to upregulated expression of CD80, CD86, ICAM-I and MHC class II (HLA-DR) on the B-CLL cells and induction of allogeneic immune responses in MLR assays. Furthermore, sustained transgene expression was demonstrated in long-term cryopreserved transfected cells. This simple and rapid gene delivery technology has been validated under the current Good Manufacturing Practice conditions, and multiple doses of CD154-expressing cells were prepared for CLL patients from one DNA transfection. Vaccination strategies using autologous tumor cells manipulated ex vivo for patients with B-CLL and perhaps with other hematopoietic malignancies could be practically implemented using this rapid and efficient nonviral gene delivery system.
Collapse
Affiliation(s)
- L H Li
- MaxCyte, Inc., Gaithersburg, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lee SY, Kim JW, Jin JO, Song MG, Park JI, Min DS, Kwak JY. Delayed apoptosis and modulation of phospholipase D activity by plasmid containing mammalian cDNA in human neutrophils. Biochem Biophys Res Commun 2006; 347:1039-47. [PMID: 16870152 DOI: 10.1016/j.bbrc.2006.07.004] [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] [Received: 06/21/2006] [Accepted: 07/01/2006] [Indexed: 10/24/2022]
Abstract
Phospholipase D (PLD) has been reported to have an anti-apoptotic role in neutrophils. This study examined the effects of plasmids containing the cDNA of PLD on the apoptosis of neutrophils. The apoptotic rate of neutrophils treated with the pCDNA3.1 plasmid was similar to that of the untreated cells after 24 h culture. However, the addition of pCDNA3.1 containing the cDNA of either human PLD1 (pCDNA3.1-PLD1) or -PLD2 (pCDNA3.1-PLD2) to the culture media with or without transfection reagent significantly decreased the rate of spontaneous apoptosis but not Fas-stimulated apoptosis and the decreased apoptosis was blocked by 1-butanol. pCDNA3.1-PLD blocked the cleavage of procaspase-3 and -8. The phorbol myristate acetate stimulated the PLD activities of pCDNA3.1-PLD-treated neutrophils but did not stimulate the activities of untreated or pCDNA3.1-treated neutrophils. The level of the PLD1 protein was higher in the cultured neutrophils with pCDNA3.1-PLD than with the media or pCDNA3.1. The spontaneous apoptosis of neutrophils was inhibited and the PLD1 expression level was increased by the linearized or promoterless forms of pCDNA3.1-PLD1 and the plasmids containing the cDNA of the enhanced green fluorescent protein (pEGFP) and EGFP-PLD1. These results suggest that the plasmids containing mammalian cDNA inhibit the spontaneous apoptosis of neutrophils and modulate PLD.
Collapse
Affiliation(s)
- Sun-Young Lee
- Department of Biochemistry, College of Medicine and Medical Research Center for Cancer Molecular Therapy, Dong-A University, Busan 602-714, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
von Levetzow G, Spanholtz J, Beckmann J, Fischer J, Kögler G, Wernet P, Punzel M, Giebel B. Nucleofection, an efficient nonviral method to transfer genes into human hematopoietic stem and progenitor cells. Stem Cells Dev 2006; 15:278-85. [PMID: 16646674 DOI: 10.1089/scd.2006.15.278] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The targeted manipulation of the genetic program of single cells as well as of complete organisms has strongly enhanced our understanding of cellular and developmental processes and should also help to increase our knowledge of primary human stem cells, e.g., hematopoietic stem cells (HSCs), within the next few years. An essential requirement for such genetic approaches is the existence of a reliable and efficient method to introduce genetic elements into living cells. Retro- and lentiviral techniques are efficient in transducing primary human HSCs, but remain labor and time consuming and require special safety conditions, which do not exist in many laboratories. In our study, we have optimized the nucleofection technology, a modified electroporation strategy, to introduce plasmid DNA into freshly isolated human HSC-enriched CD34(+) cells. Using enhanced green fluorescent protein (eGFP)-encoding plasmids, we obtained transfection efficiencies of approximately 80% and a mean survival rate of 50%. Performing functional assays using GFU-GEMM and long-term culture initiating cells (LTC-IC), we demonstrate that apart from a reduction in the survival rate the nucleofection method itself does not recognizably change the short- or long-term cell fate of primitive hematopoietic cells. Therefore, we conclude, the nucleofection method is a reliable and efficient method to manipulate primitive hematopoietic cells genetically.
Collapse
Affiliation(s)
- Gregor von Levetzow
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ghartey-Tagoe EB, Babbin BA, Nusrat A, Neish AS, Prausnitz MR. Plasmid DNA and siRNA transfection of intestinal epithelial monolayers by electroporation. Int J Pharm 2006; 315:122-33. [PMID: 16564652 DOI: 10.1016/j.ijpharm.2006.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Revised: 12/01/2005] [Accepted: 02/13/2006] [Indexed: 11/28/2022]
Abstract
This study was conducted to evaluate the ability of electroporation to efficiently transfect differentiated intestinal epithelial monolayers with plasmid DNA and to determine whether electroporation can transfect these monolayers with short-interfering RNA (siRNA) to cause gene silencing. Confluent T84 monolayers were transfected with reporter plasmids expressing luciferase or green-fluorescent protein or with siRNA directed against the nuclear envelope proteins lamin A/C using electroporation. Optimized electroporation conditions resulted in luciferase and GFP expression. Both intracellular uptake of fluorescently labeled plasmid and expression of the reporter genes increased with increasing electroporation strength and DNA concentration. When monolayers were transfected by lipofection with the reporter plasmids, expression and DNA uptake were less than for electroporation. Electroporation was also found to transfect monolayers with siRNA, which resulted in up to 90% inhibition of targeted protein production. Silencing occurred within 24h of transfection and increased with increasing siRNA concentration. These results suggest that electroporation can provide a valuable research tool for transfection of intestinal epithelial monolayers and other differentiated cell systems, and may ultimately be useful for clinical gene therapy applications.
Collapse
Affiliation(s)
- Esi B Ghartey-Tagoe
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | | | | |
Collapse
|
21
|
Shimokawa T, Ra C. C/EBPα functionally and physically interacts with GABP to activate the human myeloid IgA Fc receptor (FcαR, CD89) gene promoter. Blood 2005; 106:2534-42. [PMID: 15928042 DOI: 10.1182/blood-2004-06-2413] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractHuman Fcα receptor (FcαR; CD89), the receptor for the crystallizable fragment (Fc) of immunoglobulin A (IgA), is expressed exclusively in myeloid cells, including granulocytes and monocytes/macrophages, and is considered to define a crucial role of these cells in immune and inflammatory responses. A 259-base pair fragment of the FCAR promoter is sufficient to direct myeloid expression of a reporter gene and contains functionally important binding sites for CCAAT/enhancer-binding protein α (C/EBPα) (CE1, CE2, and CE3) and an unidentified Ets-like nuclear protein. Here, we show that the Ets-binding site is bound by a heterodimer composed of GA-binding protein α (GABPα), an Ets-related factor, and GABPβ, a Notch-related protein. Cotransfection of GABP increased FCAR promoter activity 3.7-fold through the Ets-binding site. GABP and C/EBPα synergistically activated the FCAR promoter 280-fold. Consistent with these observations, in vitro binding analyses revealed a physical interaction between the GABPα subunit and C/EBPα. This is the first report demonstrating both physical and functional interactions between GABP and C/EBPα and will provide new insights into the molecular basis of myeloid gene expression. (Blood. 2005;106:2534-2542)
Collapse
Affiliation(s)
- Toshibumi Shimokawa
- Division of Molecular Cell Immunology and Allergology, Advanced Medical Research Center, Nihon University Graduate School of Medical Sciences, Itabashi-ku, Tokyo, Japan
| | | |
Collapse
|
22
|
Nateri AS, Tzavelas C, Bildirici L, Rickwood D. Transfection of Human Peripheral Blood Mononuclear Cells Using Immunoporation. J Immunoassay Immunochem 2005; 26:169-77. [PMID: 16011143 DOI: 10.1081/ias-200062469] [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: 01/11/2023]
Abstract
Immunoporation has been found to be able to efficiently transfect a wide range of human cultured cell lines. This report shows that peripheral blood mononuclear cells can also be efficiently transfected using immunoporation. The immunoporation of the cells with fluorescent TMR-Dextran, using Immunofect MG beads, indicates that transient holes of 5.4nm in diameter or larger are formed during immunoporation. The efficiencies of transfection of lymphocytes transfected with vectors coding for EGFP and lacZ were found to be within the range of 15-30% with high levels of cell viability of more than 90%. In addition, it was observed that mononuclear cells stimulated with PHA expressed transfected reporter genes with a higher efficiency. In conclusion, these results demonstrate that immunoporation using Immunofect MG beads can be used for the efficient transfection of primary lymphocytes with DNA or other macromolecules.
Collapse
Affiliation(s)
- A S Nateri
- Department of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK
| | | | | | | |
Collapse
|
23
|
Van De Parre TJL, Martinet W, Schrijvers DM, Herman AG, De Meyer GRY. mRNA but not plasmid DNA is efficiently transfected in murine J774A.1 macrophages. Biochem Biophys Res Commun 2005; 327:356-60. [PMID: 15629470 DOI: 10.1016/j.bbrc.2004.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Indexed: 10/26/2022]
Abstract
Previous studies demonstrated that macrophages are difficult to transfect. In the present study, we investigated whether J774A.1 macrophages can be efficiently transfected using nucleofector technology. Nucleofection of J774A.1 macrophages with mRNA resulted in transfection efficiencies up to 75% without cell death as compared to control pulsed macrophages. In contrast, introduction of DNA into J774A.1 cells caused apoptosis without expression of the gene of interest. Our results show that mRNA nucleofection is a new high-speed transfection method for macrophages.
Collapse
Affiliation(s)
- Tim J L Van De Parre
- Division of Pharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.
| | | | | | | | | |
Collapse
|
24
|
Beebe SJ, White J, Blackmore PF, Deng Y, Somers K, Schoenbach KH. Diverse effects of nanosecond pulsed electric fields on cells and tissues. DNA Cell Biol 2004; 22:785-96. [PMID: 14683589 DOI: 10.1089/104454903322624993] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The application of pulsed electric fields to cells is extended to include nonthermal pulses with shorter durations (10-300 ns), higher electric fields (< or =350 kV/cm), higher power (gigawatts), and distinct effects (nsPEF) compared to classical electroporation. Here we define effects and explore potential application for nsPEF in biology and medicine. As the pulse duration is decreased below the plasma membrane charging time constant, plasma membrane effects decrease and intracellular effects predominate. NsPEFs induced apoptosis and caspase activation that was calcium-dependent (Jurkat cells) and calcium-independent (HL-60 and Jurkat cells). In mouse B10-2 fibrosarcoma tumors, nsPEFs induced caspase activation and DNA fragmentation ex vivo, and reduced tumor size in vivo. With conditions below thresholds for classical electroporation and apoptosis, nsPEF induced calcium release from intracellular stores and subsequent calcium influx through store-operated channels in the plasma membrane that mimicked purinergic receptor-mediated calcium mobilization. When nsPEF were applied after classical electroporation pulses, GFP reporter gene expression was enhanced above that observed for classical electroporation. These findings indicate that nsPEF extend classical electroporation to include events that primarily affect intracellular structures and functions. Potential applications for nsPEF include inducing apoptosis in cells and tumors, probing signal transduction mechanisms that determine cell fate, and enhancing gene expression.
Collapse
Affiliation(s)
- Stephen J Beebe
- Eastern Virginia Medical School, Norfolk, Virginia 23510, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Gabriel B, Sureau F, Casselyn M, Teissié J, Petit PX. Retroactive pathway involving mitochondria in electroloaded cytochrome c-induced apoptosis. Protective properties of Bcl-2 and Bcl-XL. Exp Cell Res 2003; 289:195-210. [PMID: 14499621 DOI: 10.1016/s0014-4827(03)00255-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cytochrome c release is thought to play an important role in the initiation of apoptosis. The nature of the control exerted by Bcl-2 and Bcl-XL on such a pathway is not precisely known. We addressed this issue by square-wave pulse electroloading of exogenous cytochrome c into Jurkat cells. Three hours after cytochrome c loading into the cells, characteristic phenotypes of apoptosis were observed. However, a significant drop in the mitochondrial membrane potential (Deltapsim) was also observed, while cytochrome c was generally considered to act downstream from the mitochondria. Related to the Deltapsim drop, there was a release of proapoptotic proteins such as AIF and Smac from the mitochondria. This release, as well as NAD(P)H and cardiolipids oxidation, are linked to previous caspase activation. Cytochrome c-linked caspase activation also led to potassium efflux out of the cell. Overexpression of Bcl-2 and Bcl-XL or N-acetyl-DEVD-aldehyde treatment not only prevented the mitochondrial membrane potential decrease, but also protected cells from the apoptosis directly induced by cytochrome c electroloading. Bcl-2 and Bcl-XL protection is based on the inhibition of the caspase-dependent retroactive pathway affecting the mitochondrial compartment.
Collapse
Affiliation(s)
- Bruno Gabriel
- Institut de Pharmacologie et Biologie Structurale-CNRS, UMR 5089, 205, Route de Narbonne, 31077 Toulouse Cedex 04, France
| | | | | | | | | |
Collapse
|
26
|
Beebe SJ, Fox PM, Rec LJ, Willis ELK, Schoenbach KH. Nanosecond, high-intensity pulsed electric fields induce apoptosis in human cells. FASEB J 2003; 17:1493-5. [PMID: 12824299 DOI: 10.1096/fj.02-0859fje] [Citation(s) in RCA: 254] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Electroporation by using pulsed electric fields with long durations compared with the charging time of the plasma membrane can induce cell fusion or introduce xenomolecules into cells. Nanosecond pulse power technology generates pulses with high-intensity electric fields, but with such short durations that the charging time of the plasma membrane is not reached, but intracellular membranes are affected. To determine more specifically their effects on cell structure and function, human cells were exposed to high intensity (up to 300 kV/cm) nanosecond (10-300 ns) pulsed electric fields (nsPEF) and were analyzed at the cellular and molecular levels. As the pulse duration decreased, plasma membrane electroporation decreased and appearances of apoptosis markers were delayed. NsPEF induced apoptosis within tens of minutes, depending on the pulse duration. Annexin-V binding, caspase activation, decreased forward light scatter, and cytochrome c release into the cytoplasm were coincident. Apoptosis was caspase- and mitochondria-dependent but independent of plasma membrane electroporation and thermal changes. The results suggest that with decreasing pulse durations, nsPEF modulate cell signaling from the plasma membrane to intracellular structures and functions. NsPEF technology provides a unique, high-power, energy-independent tool to recruit plasma membrane and/or intracellular signaling mechanisms that can delete aberrant cells by apoptosis.
Collapse
Affiliation(s)
- Stephen J Beebe
- Center for Pediatric Research, 855 W. Brambleton Ave., Norfolk, VA 23510, USA.
| | | | | | | | | |
Collapse
|
27
|
Li LH, Shivakumar R, Feller S, Allen C, Weiss JM, Dzekunov S, Singh V, Holaday J, Fratantoni J, Liu LN. Highly efficient, large volume flow electroporation. Technol Cancer Res Treat 2002; 1:341-50. [PMID: 12625759 DOI: 10.1177/153303460200100504] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Electroporation is widely used to transfect and load cells with various molecules. Traditional electroporation using a static mode is typically restricted to volumes less than 1 mL, which limits its use in clinical and industrial bioprocessing applications. Here we report efficient, large volume transfection results by using a scalable-volume electroporation system. Suspended (Jurkat) and adherent cells (10T1/2 and Huh-7) were tested. A large macromolecule, FITC-conjugated dextran (MW=500 kD) was used to measure cell uptake, while a plasmid carrying the gene coding for enhanced green fluorescence protein (eGFP) was used to quantitate the flow electrotransfection efficiency as determined by flow cytometry. The flow electroloading efficiency of FITC-dextran was >90%, while the cell viability was highly maintained (>90%). High flow electrotransfection efficiency (up to 75%) and cell viability (up to 90%) were obtained with processing volumes ranging from 1.5 to 50 mL. No significant difference of electrotransfection efficiency was observed between flow and static electrotransfection. When 50 mL of cell volume was processed and samples collected at different time points during electroporation, the transgene expression and cell viability results were identical. We also demonstrated that DNA plasmid containing EBNA1-OriP elements from Epstein-Barr virus were more efficient in transgene expression than standard plasmid without the elements (at least 500 too 1000-fold increase in expression level). Finally, to examine the feasibility of utilizing flow electrotransfected cells as a gene delivery vehicle, 10T1/2 cells were transfected with a DNA plasmid containing the gene coding for mIL12. mIL12 transfected cells were injected subcutaneously into mice, and produced functional mIL12, as demonstrated by anti-angiogenic activity. This is the first demonstration of efficient, large volume, flow electroporation and the in vivo efficacy of flow electrotransfected cells. This technology may be useful for clinical gene therapy and large-scale bioprocesses.
Collapse
Affiliation(s)
- Lin-Hong Li
- MaxCyte, Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hayes MM, Lane BR, King SR, Markovitz DM, Coffey MJ. Peroxisome proliferator-activated receptor gamma agonists inhibit HIV-1 replication in macrophages by transcriptional and post-transcriptional effects. J Biol Chem 2002; 277:16913-9. [PMID: 11847231 DOI: 10.1074/jbc.m200875200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies have demonstrated that cyclopentenone prostaglandins (cyPG) inhibit human immunodeficiency virus type 1 (HIV-1) replication in various cell types. We investigated the role of PG in the replication of HIV-1 in primary macrophages. The cyPG, PGA(1) and PGA(2), inhibited HIV-1 replication in acutely infected human monocyte-derived macrophages (MDM). Because PGA(1) and PGA(2) have previously been shown to be peroxisome proliferator-activated receptor gamma (PPARgamma) agonists, we examined the effect of synthetic PPARgamma agonists on HIV replication. The PPARgamma agonist ciglitazone inhibited HIV-1 replication in a dose-dependent manner in acutely infected human MDM. In addition, cyPG and ciglitazone reduced HIV replication in latently infected and viral entry-independent U1 cells, suggesting an effect at the level of HIV gene expression. Ciglitazone also suppressed HIV-1 mRNA levels as measured by reverse transcriptase PCR, in parallel with the decrease in reverse transcriptase activity. Co-transfection of PPARgamma wild type vectors and treatment with PPARgamma agonists inhibited HIV-1 promoter activity in U937 cells. Activation of PPARgamma also decreased HIV-1 mRNA stability following actinomycin D treatment. In summary, our experimental findings implicate PPARgamma as an important factor in the suppression of HIV-1 gene expression in MDM by cyPG. Thus natural and synthetic PPARgamma agonists may play a role in controlling HIV-1 infection in macrophages.
Collapse
Affiliation(s)
- Michael M Hayes
- Divisions of Pulmonary and Critical Care Medicine, Rheumatology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
29
|
Hayes MM, Lane BR, King SR, Markovitz DM, Coffey MJ. Prostaglandin E(2) inhibits replication of HIV-1 in macrophages through activation of protein kinase A. Cell Immunol 2002; 215:61-71. [PMID: 12142037 DOI: 10.1016/s0008-8749(02)00017-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since macrophages are a source of increased PGE(2) in AIDS, we investigated the role of PGE(2) in the replication of HIV-1 in these cells. PGE(2) inhibited HIV-1 replication measured by reverse transcriptase in human monocyte-derived macrophage (MDM). Treatment of MDM with the PGE(1) analog misoprostol, the adenylate cyclase activator forskolin, and the cyclic AMP analog dibutyryl-cyclic AMP (db-cAMP) suppressed HIV replication. The protein kinase A (PKA) activator 8-bromo-cyclic AMP also inhibited HIV-1 replication. Similar results were observed with the entry-independent, latently HIV-infected U1 cells. There was a parallel decrease in HIV-1 mRNA levels following PGE(2) treatment. Co-transfection of the HIV-1 promoter LTR.luciferase, with the vector CMV.Calpha, which expresses the PKA catalytic unit increasing PKA activity, reduced HIV-1 promoter activity. Inhibition of PKA activity with the pMT.RAB vector, a mutant regulatory unit of PKA, augmented HIV-1 promoter activity. In summary, PGE(2) inhibits HIV-1 gene expression in MDM through a PKA-dependent mechanism.
Collapse
Affiliation(s)
- Michael M Hayes
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor 48109, USA
| | | | | | | | | |
Collapse
|
30
|
Matsumoto T, Komori K, Shoji T, Kuma S, Kume M, Yamaoka T, Mori E, Furuyama T, Yonemitsu Y, Sugimachi K. Successful and optimized in vivo gene transfer to rabbit carotid artery mediated by electronic pulse. Gene Ther 2001; 8:1174-9. [PMID: 11509948 DOI: 10.1038/sj.gt.3301502] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2000] [Accepted: 05/10/2001] [Indexed: 11/09/2022]
Abstract
Several gene transfer methods, including viral or nonviral vehicles have been developed, however, efficacy, safety or handling continue to present problems. We developed a nonviral and plasmid-based method for arterial gene transfer by in vivo electronic pulse, using a newly designed T-shaped electrode. Using rabbit carotid arteries, we first optimized gene transfer efficiency, and firefly luciferase gene transfer via electronic pulse under 20 voltage (the pulse length: P(on)time 20 ms, the pulse interval: P(off) time 80 ms, number of pulse: 10 times) showed the highest gene expression. Exogenous gene expression was detectable for at least up to 14 days. Electroporation-mediated gene transfer of E. coli lacZ with nuclear localizing signal revealed successful gene transfer to luminal endothelial cells and to medial cells. Histological damage was recognized as the voltage was increased but neointima formation 4 weeks after gene transfer was not induced. In vivo electroporation-mediated arterial gene transfer is readily facilitated, is safe and may prove to be an alternative form of gene transfer to the vasculature.
Collapse
Affiliation(s)
- T Matsumoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Li LH, McCarthy P, Hui SW. High-efficiency electrotransfection of human primary hematopoietic stem cells. FASEB J 2001; 15:586-8. [PMID: 11259375 DOI: 10.1096/fj.00-0447fje] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A major obstacle to gene transfer into hematopoietic stem cells, a key step for many gene therapy and tissue replacement applications, is its low efficiency. High cell mortality is responsible for the low efficiency of electrotransfection when this technique is applied to certain 'refractory' cell types such as hematopoietic stem cells. Using human primary CD-34+ cells from peripheral blood as a model, we found that transfection-induced apoptosis and, to a lesser extent, postpulse colloidal-osmotic swelling are two main factors for the poor transfection of these cells. By applying caspase inhibitors (B-D-Fluomethyl Ketone and Z-VAD-FMK) to reduce apoptosis, and by using the postpulse pelleting method to suppress colloidal-osmotic swelling, we achieved a transfection efficiency of ~20%, regardless of the presence of cytokines in the suspension medium. This effort brings the ex vivo electrotransfection efficiency within the reach of therapeutic applications.
Collapse
Affiliation(s)
- L H Li
- Department of Molecular and Cellular Biophysics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | |
Collapse
|