Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Gene Ther ; 24(5): 298-307, 2017 05.
Article in English | MEDLINE | ID: mdl-28346436

ABSTRACT

Correction of patient-specific induced pluripotent stem cells (iPSC) upon gene delivery through retroviral vectors offers new treatment perspectives for monogenetic diseases. Gene-modified iPSC clones can be screened for safe integration sites and differentiated into transplantable cells of interest. However, the current bottleneck is epigenetic vector silencing. In order to identify the most suitable retroviral expression system in iPSC, we systematically compared vectors from different retroviral genera, different promoters and their combination with ubiquitous chromatin opening elements (UCOE), and several envelope pseudotypes. Lentiviral vectors (LV) pseudotyped with vesicular stomatitis virus glycoprotein were superior to gammaretroviral and alpharetroviral vectors and other envelopes tested. The elongation factor 1α short (EFS) promoter mediated the most robust expression, whereas expression levels were lower from the potent but more silencing-prone spleen focus forming virus (SFFV) promoter. Both full-length (A2UCOE) and minimal (CBX3) UCOE juxtaposed to two physiological and one viral promoter reduced transgene silencing with equal efficiency. However, a promoter-specific decline in expression levels was not entirely prevented. Upon differentiation of transgene-positive iPSC into endothelial cells, A2UCOE.EFS and CBX3.EFS vectors maintained highest transgene expression in a larger fraction of cells as compared with all other constructs tested here. The function of UCOE diminished, but did not fully counteract, vector silencing and possibilities for improvements remain. Nevertheless, the CBX3.EFS in a LV background exhibited the most promising promoter and vector configuration for both high titer production and long-term genetic modification of human iPSC and their progeny.


Subject(s)
Genetic Vectors/genetics , Induced Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Retroviridae/genetics , Transgenes , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Silencing , HeLa Cells , Humans , Induced Pluripotent Stem Cells/cytology , Peptide Elongation Factor 1/genetics , Transfection/methods , Transfection/standards
2.
Gene Ther ; 21(11): 938-49, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25102011

ABSTRACT

Retroviral vectors are versatile gene transfer vehicles widely used in basic research and gene therapy. Mutation of retroviral integrase converts these vectors into transient, integration-deficient gene delivery vehicles associated with a high degree of biosafety. We explored the option to use integration-deficient retroviral vectors to achieve transient ectopic expression of transcription factors, which is considered an important tool for induced cell fate conversion. Stepwise optimization of the retroviral episome transfer as exemplified for the transcription factor Oct4 enabled to improve both expression magnitude and endurance. Long terminal repeat-driven γ-retroviral vectors were identified as the most suitable vector architecture. Episomal expression was enhanced by epigenetic modifiers, and Oct4 activity was increased following fusion to a minimal transactivation motif of herpes simplex virus VP16. Based on kinetic analyses, we identified optimal time intervals for repeated vector administration and established prolonged expression windows of choice. Providing proof-of-concept, episomal transfer of Oct4 was potent to mediate conversion of human fibroblasts stably expressing Klf4, Sox2 and c-Myc into induced pluripotent stem cells, which were mainly free of residual Oct4 vector integration. This study provides evidence for suitability of retroviral episome transfer of transcription factors for cell fate conversion, allowing the generation of distinct patient- or disease-specific cell types.


Subject(s)
Plasmids/genetics , Retroviridae/genetics , Transcription Factors/genetics , Transduction, Genetic/methods , Cell Differentiation/genetics , Cell Line , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Integrases/genetics , Kruppel-Like Factor 4 , Octamer Transcription Factor-3/genetics
3.
Cytogenet Genome Res ; 142(1): 14-20, 2014.
Article in English | MEDLINE | ID: mdl-24192547

ABSTRACT

The fate of cultivated primary hematopoietic stem cells (HSCs) with respect to genetic instability and telomere attrition has not yet been described in great detail. Thus, knowledge of the genetic constitution of HSCs is important when interpreting results of HSCs in culture. While establishing a cell culture model for myelodysplastic syndrome with a deletion in 5q by performing RPS14 knockdown, we found surprising data that may be of importance for any CD34+ cell culture experiments. We performed cytogenetic analyses and telomere length measurement on transduced CD34+ cells and untransduced control cells to observe the effects of long-term culturing. Initially, CD34+ cells had a normal median telomere length of about 12 kb and showed no signs of chromosomal instability. During follow-up, the median telomere length seemed to decrease and, simultaneously, increased chromosomal instability could be observed - in modified and control cells. One culture showed a clonal monosomy 7 - independent of prior RPS14 knockdown. During further culturing, it seemed that the telomeres re-elongated, and chromosomes stabilized, while TERT expression was not elevated. In summary, irrespective of our results of RPS14 knockdown in the long-term culture of CD34+ cells, it becomes clear that cell culture artefacts inducing telomere shortening and chromosomal instability have to be taken into account and regular cytogenetic analyses should always be performed.


Subject(s)
Artifacts , Cell Culture Techniques , Chromosomal Instability/genetics , Chromosomes, Human, Pair 5/genetics , Hematopoietic Stem Cells/ultrastructure , Ribosomal Proteins/genetics , Telomere Shortening/genetics , Antigens, CD34/analysis , Cells, Cultured , Chromosome Deletion , Chromosomes, Human, Pair 5/ultrastructure , Colony-Forming Units Assay , DNA Repair , Fetal Blood/cytology , Genes, Reporter , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization, Fluorescence , K562 Cells , Karyotyping , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering/genetics , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/deficiency , Ribosomal Proteins/physiology , Telomerase/metabolism , Transduction, Genetic
4.
Gene Ther ; 20(3): 298-307, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22592598

ABSTRACT

Regulated transgene expression may reduce transgene-specific and genotoxic risks associated with gene therapy. To prove this concept, we have investigated the suitability of doxycycline (Dox)-inducible human cytidine deaminase (hCDD) overexpression from lentiviral vectors to mediate effective myeloprotection while circumventing the lymphotoxicity observed with constitutive CDD activity. Rapid Dox-mediated transgene induction associated with a 6-17-fold increase in drug resistance was observed in 32D and primary murine bone marrow (BM) cells. Moreover, robust Dox-regulated transgene expression in the entire haematopoietic system was demonstrated for primary and secondary recipients of hCDD-transduced R26-M2rtTA transgenic BM cells. Furthermore, mice were significantly protected from myelosuppressive chemotherapy as evidenced by accelerated recovery of granulocytes (1.9±0.6 vs 1.3±0.3, P=0.034) and platelets (883±194 vs 584±160 10(3) per µl, P=0.011). Minimal transgene expression in the non-induced state and no overt cellular toxicities including lymphotoxicity were detected. Thus, using a relevant murine transplant model our data provide conclusive evidence that drug-resistance transgenes can be expressed in a regulated fashion in the lymphohaematopoietic system, and that Dox-inducible systems may be used to reduce myelotoxic side effect of anticancer chemotherapy or to avoid side effects of high constitutive transgene expression.


Subject(s)
Cytidine Deaminase/genetics , Doxycycline/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hematopoietic System/metabolism , Lentivirus/genetics , Animals , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Line , Cells, Cultured , Cytarabine/pharmacology , Cytidine Deaminase/metabolism , Dose-Response Relationship, Drug , Female , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic System/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism , Time-Lapse Imaging/methods , Transgenes/genetics
5.
Gene Ther ; 19(1): 15-24, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21562592

ABSTRACT

Gene transfer-based therapeutic approaches have greatly benefited from the ability of some viral vectors to efficiently integrate within the cell genome and ensure persistent transmission of newly acquired transgenes to the target cell progeny. However, integration of provirus has been associated with epigenetic repercussions that may influence the expression of both the transgene and cellular genes close to vector integration loci. The exploitation of genetic insulator elements may overcome both issues through their ability to act as barriers that limit transgene silencing and/or as enhancer-blockers preventing the activation of endogenous genes by the vector enhancer. We established quantitative plasmid-based assay systems to screen enhancer-blocker and barrier genetic elements. Short synthetic insulators that bind to nuclear factor-I protein family transcription factors were identified to exert both enhancer-blocker and barrier functions, and were compared to binding sites for the insulator protein CTCF (CCCTC-binding factor). Gamma-retroviral vectors enclosing these insulator elements were produced at titers similar to their non-insulated counterparts and proved to be less genotoxic in an in vitro immortalization assay, yielding lower activation of Evi1 oncogene expression and reduced clonal expansion of bone marrow cells.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/metabolism , Insulator Elements , NFI Transcription Factors/metabolism , Animals , Binding Sites , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CCCTC-Binding Factor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Friend murine leukemia virus/genetics , Friend murine leukemia virus/metabolism , Gene Silencing , Genetic Vectors/genetics , HeLa Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred C57BL , NFI Transcription Factors/genetics , Plasmids/genetics , Plasmids/metabolism , Proto-Oncogenes/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Transgenes , Virus Integration
6.
Gene Ther ; 19(9): 915-24, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21975463

ABSTRACT

Endogenous microRNA (miRNA) expression can be exploited for cell type-specific transgene expression as the addition of miRNA target sequences to transgenic cDNA allows for transgene downregulation specifically in cells expressing the respective miRNAs. Here, we have investigated the potential of miRNA-150 target sequences to specifically suppress gene expression in lymphocytes and thereby prevent transgene-induced lymphotoxicity. Abundance of miRNA-150 expression specifically in differentiated B and T cells was confirmed by quantitative reverse transcriptase PCR. Mono- and bicistronic lentiviral vectors were used to investigate the effect of miRNA-150 target sequences on transgene expression in the lymphohematopoietic system. After in vitro studies demonstrated effective downregulation of transgene expression in murine B220(+) B and CD3(+) T cells, the concept was further verified in a murine transplant model. Again, marked suppression of transgene activity was observed in B220(+) B and CD4(+) or CD8(+) T cells whereas expression in CD11b(+) myeloid cells, lin(-) and lin(-)/Sca1(+) progenitors, or lin(-)/Sca1(+)/c-kit(+) stem cells remained almost unaffected. No toxicity of miRNA-150 targeting in transduced lymphohematopoietic cells was noted. Thus, our results demonstrate the suitability of miRNA-150 targeting to specifically suppress transgene expression in lymphocytes and further support the concept of miRNA targeting for cell type-specific transgene expression in gene therapy approaches.


Subject(s)
B-Lymphocytes/immunology , Down-Regulation , Gene Targeting , Genetic Vectors , Hematopoiesis/genetics , MicroRNAs/genetics , T-Lymphocytes/immunology , Animals , Cell Line , Female , Gene Targeting/adverse effects , Male , Mice , Mice, Nude , Transgenes
7.
Gene Ther ; 19(4): 425-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21850050

ABSTRACT

Partial resistance of primary mouse hepatocytes to lentiviral (LV) vector transduction poses a challenge for ex vivo gene therapy protocols in models of monogenetic liver disease. We thus sought to optimize ex vivo LV gene transfer while preserving the hepatocyte integrity for subsequent transplantation into recipient animals. We found that culture media supplemented with epidermal growth factor (EGF) and, to a lesser extent, hepatocyte growth factor (HGF) markedly improved transduction efficacy at various multiplicities of infection. Up to 87% of primary hepatocytes were transduced in the presence of 10 ng EGF, compared with ~30% in standard culture medium (SCMs). The increased number of transgene-expressing cells correlated with increased nuclear import and more integrated pro-viral copies per cell. Higher LV transduction efficacy was not associated with proliferation, as transduction capacity of gammaretroviral vectors remained low (<1%). Finally, we developed an LV transduction protocol for short-term (maximum 24 h) adherent hepatocyte cultures. LV-transduced hepatocytes showed liver repopulation capacities similar to freshly isolated hepatocytes in alb-uPA mouse recipients. Our findings highlight the importance of EGF for efficient LV transduction of primary hepatocytes in culture and should facilitate studies of LV gene transfer in mouse models of monogenetic liver disease.


Subject(s)
Epidermal Growth Factor/pharmacology , Genetic Vectors , Hepatocytes/metabolism , Lentivirus/genetics , Transduction, Genetic , Animals , Cells, Cultured , Culture Media , Gene Transfer Techniques , Hepatocyte Growth Factor/pharmacology , Hepatocytes/transplantation , Mice , Mice, Inbred C57BL
8.
Gene Ther ; 19(3): 246-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21753795

ABSTRACT

The need for γ-retroviral (gRV) vectors with a self-inactivating (SIN) design for clinical application has prompted a shift in methodology of vector manufacturing from the traditional use of stable producer lines to transient transfection-based techniques. Herein, we set out to define and optimize a scalable manufacturing process for the production of gRV vectors using transfection in a closed-system bioreactor in compliance with current good manufacturing practices (cGMP). The process was based on transient transfection of 293T cells on Fibra-Cel disks in the Wave Bioreactor. Cells were harvested from tissue culture flasks and transferred to the bioreactor containing Fibra-Cel in the presence of vector plasmid, packaging plasmids and calcium-phosphate in Dulbecco's modified Eagle's medium and 10% fetal bovine serum. Virus supernatant was harvested at 10-14 h intervals. Using optimized procedures, a total of five ecotropic cGMP-grade gRV vectors were produced (9 liters each) with titers up to 3.6 × 10(7) infectious units per milliliter on 3T3 cells. One GMP preparation of vector-like particles was also produced. These results describe an optimized process for the generation of SIN viral vectors by transfection using a disposable platform that allows for the generation of clinical-grade viral vectors without the need for cleaning validation in a cost-effective manner.


Subject(s)
Bioreactors , Gammaretrovirus/genetics , Genetic Vectors/isolation & purification , Genetic Vectors/standards , Transfection/methods , Animals , Batch Cell Culture Techniques/methods , Batch Cell Culture Techniques/standards , Biotechnology , Cell Line , Gammaretrovirus/isolation & purification , Humans , Mice , Quality Control
9.
Gene Ther ; 19(8): 872-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22551777

ABSTRACT

Patients with X-linked severe combined immunodeficiency (SCID-X1) were successfully cured following gene therapy with a gamma-retroviral vector (gRV) expressing the common gamma chain of the interleukin-2 receptor (IL2RG). However, 5 of 20 patients developed leukemia from activation of cellular proto-oncogenes by viral enhancers in the long-terminal repeats (LTR) of the integrated vector. These events prompted the design of a gRV vector with self-inactivating (SIN) LTRs to enhance vector safety. Herein we report on the production of a clinical-grade SIN IL2RG gRV pseudotyped with the Gibbon Ape Leukemia Virus envelope for a new gene therapy trial for SCID-X1, and highlight variables that were found to be critical for transfection-based large-scale SIN gRV production. Successful clinical production required careful selection of culture medium without pre-added glutamine, reduced exposure of packaging cells to cell-dissociation enzyme, and presence of cations in wash buffer. The clinical vector was high titer; transduced 68-70% normal human CD34(+) cells, as determined by colony-forming unit assays and by xenotransplantation in immunodeficient NOD.CB17-Prkdc(scid)/J (nonobese diabetic/severe combined immunodeficiency (NOD/SCID)) and NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ (NOD/SCID gamma (NSG))) mice; and resulted in the production of T cells in vitro from human SCID-X1 CD34(+) cells. The vector was certified and released for the treatment of SCID-X1 in a multi-center international phase I/II trial.


Subject(s)
Genetic Vectors , Interleukin Receptor Common gamma Subunit/genetics , Retroviridae/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , Animals , Gene Transfer Techniques , Humans , Leukemia Virus, Gibbon Ape/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Terminal Repeat Sequences , Transduction, Genetic
10.
Tissue Antigens ; 79(5): 340-50, 2012 May.
Article in English | MEDLINE | ID: mdl-22489943

ABSTRACT

Matching of human leukocyte antigen (HLA) alleles between donors and recipients plays a major role in hematopoietic stem cell transplantation (HSCT). Null or questionably expressed HLA allelic variants are a major issue in HLA matching, because the aberrant expression of such alleles can have a major impact on the outcome of HSCT and/or its complications such as graft-versus-host disease. The goal of this study was to investigate the potential of a recently developed cytokine-induced secretion assay to differentiate the expression levels of HLA-A*32:11Q (questionable) into a null (N) or low (L) expression variant. An amino acid mutation at position 164 of HLA-A*32:11Q disrupts the disulfide bridge in the α2 domain. HLA-A*32:11Q is not detectable by standard microlymphocytotoxicity assay. To this end, we cloned soluble HLA-A*32:11Q and a reference allele (HLA-A*32:01) into expression vectors and transfected/transduced HEK293 and K562 cells. Allele-expressing K562 cells were simultaneously transfected/transduced with a ß2-microglobulin (B2M)-encoding vector to ensure the intact HLA structure with B2M. After treatment with proinflammatory cytokines, secreted soluble HLA molecules were determined by enzyme-linked immunosorbent assay in the supernatant and intracellular accumulation of the recombinant proteins by flow cytometry. HLA-A*32:11Q was nearly undetectable in untreated transfectants. Cytokine treatment increased the secretion of HLA-A*32:11Q to detectable levels and resulted in intracellular accumulation of the allele. There was no difference in mRNA transcription between the A*32 alleles. On the basis of these results, we recommend reclassification of HLA-A*32:11Q as a low expression (L) variant.


Subject(s)
Gene Expression/immunology , HLA-A Antigens/genetics , Alleles , Cloning, Molecular , Genetic Vectors , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , HEK293 Cells , HLA-A Antigens/immunology , Hematopoietic Stem Cell Transplantation , Histocompatibility Testing , Humans , Interferon-gamma/immunology , Interferon-gamma/pharmacology , K562 Cells , Mutation , Protein Isoforms/genetics , Protein Isoforms/immunology , Transfection , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
11.
Gene Ther ; 17(3): 400-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19847204

ABSTRACT

Bidirectional lentiviral vectors mediate expression of two or more cDNAs from a single internal promoter. In this study, we examined mechanisms that control titer and expression properties of this vector system. To address whether the bidirectional design depends on lentiviral (LV) backbone components, especially the Rev/Rev responsive element (RRE) system, we constructed similar expression cassettes for LV and gammaretroviral (GV) vectors. Bidirectional expression levels could be adjusted by the use of different internal promoters. Furthermore, removal of the constitutive RNA transport element of Mason-Pfizer monkey virus, used in first generation bidirectional LV vectors, improved gene expression. Titers of bidirectional vectors were approximately 10-fold reduced in comparison to unidirectional vectors, independent of the Rev/RRE interaction. We reasoned that titer reductions were due to the formation of interfering double-stranded RNA in packaging cells. Indeed, cotransfection of Nodamuravirus B2 protein, an RNA interference suppressor, increased bidirectional vector titers at least fivefold. We validated the potential of high titer bidirectional vectors by coexpressing a fluorescent marker with O(6)-methylguanine-DNA methyltransferase from integrating, or with Cre recombinase from integrating and non-integrating GV and LV backbones. This allowed for the tracking of chemoprotected and recombined cells by fluorescence marker expression.


Subject(s)
Gammaretrovirus/genetics , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Lentivirus/genetics , Viral Load/genetics , Animals , Cell Line , Genes, env , Humans , Mason-Pfizer monkey virus/genetics , Mice , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic , RNA, Double-Stranded/genetics
12.
Gene Ther ; 17(2): 272-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19865181

ABSTRACT

The clinical application of self-inactivating (SIN) retroviral vectors has been hampered by the lack of reliable and efficient vector production technologies. To enable production of SIN gamma-retroviral vectors from stable producer clones, a new PG13-based packaging cell, known as PG368, was developed. Viral vector expression constructs can be reliably inserted at a predefined genomic locus of PG368 packaging cells by an Flp-recombinase-mediated targeted cassette exchange (RMCE) reaction. A new, carefully designed vector-targeting construct, pEMTAR-1, eliminated the co-packaging of the selectable marker gene used for the identification of successful recombination at the predefined genomic locus and thus, improved the safety of the production system. Selected clones produced vector supernatants at consistent titers. The targeted insertion of therapeutically relevant SIN vectors for chronic granulomatous disease and X-linked severe combined immunodeficiency into PG368 cells results in stable titers within the range necessary for clinical application. The production of retroviral SIN vectors from stable clinical-grade producer cells is feasible and will contribute to the safe production and application of SIN gamma-retroviral vectors for clinical trials.


Subject(s)
DNA Nucleotidyltransferases , Gene Transfer Techniques , Genetic Vectors , Retroviridae/genetics , Cell Line , Feasibility Studies , Gene Targeting , Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Humans , Severe Combined Immunodeficiency/therapy
13.
Gene Ther ; 16(1): 111-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18784749

ABSTRACT

Gene therapy has proven to be of potential value for the correction of inherited hematopoietic disorders. However, the occurrence of severe side effects in some of the clinical trials has questioned the safety of this approach and has hampered the use of long terminal repeat-driven vectors for the treatment of a large number of patients. The development of self-inactivating (SIN) vectors with reduced genotoxicity provides an alternative to the currently used vectors. Our initial attempts to use SIN vectors for the correction of a myeloid disorder, chronic granulomatous disease, failed due to low vector titers and poor transgene expression. The optimization of the transgene cDNA (gp91(phox)) resulted in substantially increased titers and transgene expression. Most notably, transgene optimization significantly improved expression of a second cistron located downstream of gp91(phox). Thus, optimization of the transgene sequence results in higher expression levels and increased therapeutic index allowing the use of low vector copy numbers per transduced cell and weaker internal promoters.


Subject(s)
Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/metabolism , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Animals , Cell Line, Tumor , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Granulomatous Disease, Chronic/metabolism , Hematopoietic Stem Cells/virology , Humans , Immunomagnetic Separation , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Superoxides/analysis , Transduction, Genetic/methods , Transgenes , Virus Inactivation
14.
Hum Gene Ther ; 30(10): 1284-1296, 2019 10.
Article in English | MEDLINE | ID: mdl-31407607

ABSTRACT

Endothelial cells (EC) are targets in gene therapy and regenerative medicine, but they are inefficiently transduced with adeno-associated virus (AAV) vectors of various serotypes. To identify barriers hampering efficient transduction and to develop an optimized AAV variant for EC transduction, we screened an AAV serotype 2-based peptide display library on primary human macrovascular EC. Using a new high-throughput selection and monitoring protocol, we identified a capsid variant, AAV-VEC, which outperformed the parental serotype as well as first-generation targeting vectors in EC transduction. AAV vector uptake was improved, resulting in significantly higher transgene expression levels from single-stranded vector genomes detectable within a few hours post-transduction. Notably, AAV-VEC transduced not only proliferating EC but also quiescent EC, although higher particle-per-cell ratios had to be applied. Also, induced pluripotent stem cell-derived endothelial progenitor cells, a novel tool in regenerative medicine and gene therapy, were highly susceptible toward AAV-VEC transduction. Thus, overcoming barriers by capsid engineering significantly expands the AAV tool kit for a wide range of applications targeting EC.


Subject(s)
Capsid/chemistry , Dependovirus/genetics , Genetic Engineering/methods , Genetic Vectors/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Transduction, Genetic/methods , Amino Acid Sequence , Capsid/metabolism , Cell Differentiation , Dependovirus/metabolism , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Peptide Library
15.
Nucleic Acids Res ; 28(4): 901-10, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10648781

ABSTRACT

Retroviral gene expression requires nuclear export and translation of incompletely spliced RNA. In the case of human immunodeficiency virus (HIV), this is facilitated by the viral Rev protein binding to its cognate RNA response element (RRE), while other retroviruses contain constitutive transport elements (CTE) binding to cellular factors. These CTE can substitute for the HIV-1 Rev/RRE system, albeit with reduced efficiency. Here, we show that multimeric copies of the CTE restore HIV-1 protein expression to levels comparable to or higher than Rev/RRE in various cell lines from different species. We suggest that multimerization of export factors is important for CTE function, as reported for Rev. CTE function was not affected when the element was displaced from its natural position close to the poly(A) signal, while insertion of an intron into the 3'-untranslated region (3'-UTR) severely reduced CTE activity. In this case, cytoplasmic RNA degradation was observed, which may be mediated by nonsense-mediated RNA decay. In contrast, Rev-dependent gene expression was insensitive to an intron in the 3'-UTR. Finally, we show that the putative CTE-binding protein RNA helicase A is not specifically translocated into the cytoplasm upon overexpression of CTE-containing RNA.


Subject(s)
Gene Products, gag/genetics , HIV-1/genetics , Mason-Pfizer monkey virus/genetics , RNA, Viral/genetics , 3' Untranslated Regions , Base Sequence , Biological Transport , Cytoplasm/metabolism , DNA Primers , Gene Products, gag/biosynthesis , HeLa Cells , Humans , Introns , Transduction, Genetic
17.
Leukemia ; 29(7): 1530-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25652739

ABSTRACT

Co-transplantation of hematopoietic stem cells with those engineered to express leukemia-reactive T-cell receptors (TCRs) and differentiated ex vivo into precursor T cells (preTs) may reduce the risk of leukemia relapse. As expression of potentially self-(leukemia-) reactive TCRs will lead to negative selection or provoke autoimmunity upon thymic maturation, we investigated a novel concept whereby TCR expression set under the control of an inducible promoter would allow timely controlled TCR expression. After in vivo maturation and gene induction, preTs developed potent anti-leukemia effects. Engineered preTs provided protection even after repeated leukemia challenges by giving rise to effector and central memory cells. Importantly, adoptive transfer of TCR-transduced allogeneic preTs mediated anti-leukemia effect without evoking graft-versus-host disease (GVHD). Earlier transgene induction forced CD8(+) T-cell development was required to obtain a mature T-cell subset of targeted specificity, allowed engineered T cells to efficiently pass positive selection and abrogated the endogenous T-cell repertoire. Later induction favored CD4 differentiation and failed to produce a leukemia-reactive population emphasizing the dominant role of positive selection. Taken together, we provide new functional insights for the employment of TCR-engineered precursor cells as a controllable immunotherapeutic modality with significant anti-leukemia activity.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Leukemia, Myeloid/immunology , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell/genetics , Adoptive Transfer , Animals , Flow Cytometry , Genetic Engineering , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid/mortality , Leukemia, Myeloid/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Promoter Regions, Genetic/genetics , Receptors, Antigen, T-Cell/immunology , Transplantation, Homologous
18.
Curr Mol Med ; 13(5): 765-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23642058

ABSTRACT

Reprogramming of somatic cells into patient-specific pluripotent analogues of human embryonic stem cells (ESCs) emerges as a prospective therapeutic angle in molecular medicine and a tool for basic stem cell biology. However, the combination of relative inefficiency and high variability of non-defined culture conditions precluded the use of this technique in a clinical setting and impeded comparability between laboratories. To overcome these obstacles, we sequentially devised a reprogramming protocol using one lentiviral-based polycistronic reprogramming construct, optimized for high co-expression of OCT4, SOX2, KLF4 and MYC in conjunction with small molecule inhibitors of non-permissive signaling cascades, such as transforming growth factor ß (SB431542), MEK/ERK (PD0325901) and Rho-kinase signaling (Thiazovivin), in a defined extracellular environment. Based on human fetal liver fibroblasts we could efficiently derive induced pluripotent stem cells (iPSCs) within 14 days. We attained efficiencies of up to 10.97±1.71% resulting in 79.5- fold increase compared to non-defined reprogramming using four singular vectors. We show that the overall increase of efficiency and temporal kinetics is a combinatorial effect of improved lentiviral vector design, signaling inhibition and definition of extracellular matrix (Matrigel®) and culture medium (mTESR®1). Using this protocol, we could derive iPSCs from patient fibroblasts, which were impermissive to classical reprogramming efforts, and from a patient suffering from familial platelet disorder. Thus, our defined protocol for highly efficient reprogramming to generate patient-specific iPSCs, reflects a big step towards therapeutic and broad scientific application of iPSCs, even in previously unfeasible settings.


Subject(s)
Collagen/chemistry , Induced Pluripotent Stem Cells/physiology , Laminin/chemistry , Proteoglycans/chemistry , Animals , Benzamides/pharmacology , Blood Platelet Disorders/genetics , Blood Platelet Disorders/pathology , Cell Proliferation , Cells, Cultured , Cellular Reprogramming , Core Binding Factor Alpha 2 Subunit/genetics , Culture Media/chemistry , Dioxoles/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Combinations , Embryonic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Infant, Newborn , Kruppel-Like Factor 4 , Mice , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics
19.
Curr Mol Med ; 13(5): 842-55, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23642065

ABSTRACT

Based on their almost unlimited self-renewal capacity and their ability to differentiate into derivatives of all three germ layers, human induced pluripotent stem cells (hiPSCs) might serve as a preferable source for hepatic transplants in metabolic liver disorders or acute liver failure. Furthermore, the generation of patient specific hiPSCs might facilitate the development of innovative therapeutic strategies by accurately modelling disease in vitro. In our study, we aimed for an efficient hepatic differentiation protocol that is applicable for both human embryonic stem cells (hESCs) and hiPSCs. We attempted to accomplish this goal by using a cytokine and small molecule-based protocol for direct differentiation of hESCs and hiPSCs into hepatic cells. Selecting differentiated hepatic cells was possible using an albumin promoter-driven G418 resistance system. Due to IRES-dependent dTomato reporter expression, we were able to track hepatic differentiated cells and we evaluated the most efficient time frame for G418 selection. The status of hepatic differentiation was determined by qRT-PCR comparing the expression of hepatic markers such as AFP, ALB, SOX17, and HNF4 to standard hepatic cells. Functional analysis of the hepatic phenotype was obtained by measuring secreted albumin levels and by analysis of cytochrome P450 type 1A1 activity (EROD). The percentage of differentiated cells was quantified by FACS analysis. In conclusion, our improved protocol demonstrates that both pluripotent cell sources (hESC and hiPSC) can efficiently be differentiated into mature hepatic cells with functional characteristics similar to those of standard hepatic cell lines such as HepG2.


Subject(s)
Cell Differentiation , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/physiology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cell Culture Techniques , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Fibroblasts/physiology , Gene Expression , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Liver/cytology , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics
20.
Leukemia ; 27(5): 1127-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23212151

ABSTRACT

The transcription factor Evi1 has an outstanding role in the formation and transformation of hematopoietic cells. Its activation by chromosomal rearrangement induces a myelodysplastic syndrome with progression to acute myeloid leukemia of poor prognosis. Similarly, retroviral insertion-mediated upregulation confers a competitive advantage to transplanted hematopoietic cells, triggering clonal dominance or even leukemia. To study the molecular and functional response of primary murine hematopoietic progenitor cells to the activation of Evi1, we established an inducible lentiviral expression system. EVI1 had a biphasic effect with initial growth inhibition and retarded myeloid differentiation linked to enhanced survival of myeloblasts in long-term cultures. Gene expression microarray analysis revealed that within 24 h EVI1 upregulated 'stemness' genes characteristic for long-term hematopoietic stem cells (Aldh1a1, Abca1, Cdkn1b, Cdkn1c, Epcam, among others) but downregulated genes involved in DNA replication (Cyclins and their kinases, among others) and DNA repair (including Brca1, Brca2, Rad51). Cell cycle analysis demonstrated EVI1's anti-proliferative effect to be strictly dose-dependent with accumulation of cells in G0/G1, but preservation of a small fraction of long-term proliferating cells. Although confined to cultured cells, our study contributes to new hypotheses addressing the mechanisms and molecular targets involved in preleukemic clonal dominance or leukemic transformation by Evi1.


Subject(s)
Cell Cycle , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/cytology , Proto-Oncogenes/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Line , Cell Survival , Granulocyte Precursor Cells/physiology , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL