Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Gene Ther ; 27(6): 297-306, 2020 06.
Article in English | MEDLINE | ID: mdl-32051561

ABSTRACT

Induced pluripotent stem cells (iPSCs) from patients with genetic disorders are a valuable source for in vitro disease models, which enable drug testing and validation of gene and cell therapies. We generated iPSCs from a severe congenital neutropenia (SCN) patient, who presented with a nonsense mutation in the glucose-6-phosphatase catalytic subunit 3 (G6PC3) gene causing profound defects in granulopoiesis, associated with increased susceptibility of neutrophils to apoptosis. Generated SCN iPSC clones exhibited the capacity to differentiate into hematopoietic cells of the myeloid lineage and we identified two cytokine conditions, i.e., using granulocyte-colony stimulating factor or granulocyte-macrophage colony stimulating factor in combination with interleukin-3, to model the SCN phenotype in vitro. Reduced numbers of granulocytes were produced by SCN iPSCs compared with control iPSCs in both settings, which reflected the phenotype in patients. Interestingly, our model showed increased monocyte/macrophage production from the SCN iPSCs. Most importantly, lentiviral genetic correction of SCN iPSCs with a codon-optimized G6PC3 transgene restored granulopoiesis and reduced apoptosis of in vitro differentiated myeloid cells. Moreover, addition of vitamin B3 clearly induced granulocytic differentiation of SCN iPSCs and increased the number of neutrophils to levels comparable with those obtained from healthy control iPSCs. In summary, we established an iPSC-derived in vitro disease model, which will serve as a tool to test the potency of alternative treatment options for SCN patients, such as small molecules and gene therapeutic vectors.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Genetic Therapy , Glucose-6-Phosphatase , Granulocyte Colony-Stimulating Factor , Humans , Niacinamide
2.
Retrovirology ; 14(1): 48, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29047401

ABSTRACT

The authors wish to apologize for an error within the scale bar of the microarray heatmap in Additional File 5 of the supplementary information. Two values were incorrectly displayed on the scale bar (11 instead of 10 and 13 instead of 12).

3.
Retrovirology ; 14(1): 34, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28569216

ABSTRACT

BACKGROUND: Retroviral vectors are derived from wild-type retroviruses, can be used to study retrovirus-host interactions and are effective tools in gene and cell therapy. However, numerous cell types are resistant or less permissive to retrovirus infection due to the presence of active defense mechanisms, or the absence of important cellular host co-factors. In contrast to multipotent stem cells, pluripotent stem cells (PSC) have potential to differentiate into all three germ layers. Much remains to be elucidated in the field of anti-viral immunity in stem cells, especially in PSC. RESULTS: In this study, we report that transduction with HIV-1-based, lentiviral vectors (LV) is impaired in murine PSC. Analyses of early retroviral events in induced pluripotent stem cells (iPSC) revealed that the restriction is independent of envelope choice and does not affect reverse transcription, but perturbs nuclear entry and proviral integration. Proteasomal inhibition by MG132 could not circumvent the restriction. However, prevention of cyclophilin A (CypA) binding to the HIV-1 capsid via use of either a CypA inhibitor (cyclosporine A) or CypA-independent capsid mutants improved transduction. In addition, application of higher vector doses also increased transduction. Our data revealed a CypA mediated restriction in iPSC, which was acquired during reprogramming, associated with pluripotency and relieved upon subsequent differentiation. CONCLUSIONS: We showed that murine PSC and iPSC are less susceptible to LV. The block observed in iPSC was CypA-dependent and resulted in reduced nuclear entry of viral DNA and proviral integration. Our study helps to improve transduction of murine pluripotent cells with HIV-1-based vectors and contributes to our understanding of retrovirus-host interactions in PSC.


Subject(s)
Genetic Vectors , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/virology , Lentivirus/genetics , Animals , Capsid Proteins/genetics , Carrier Proteins/genetics , Cell Line , Cyclophilin A/metabolism , Cyclosporine/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , HIV-1/genetics , Host-Pathogen Interactions , Induced Pluripotent Stem Cells/drug effects , Lentivirus/physiology , Leupeptins/pharmacology , Mice , Reverse Transcription/drug effects , Transduction, Genetic , Virus Integration/drug effects , Virus Internalization
4.
Curr Gene Ther ; 15(3): 245 - 254, 2015 04 09.
Article in English | MEDLINE | ID: mdl-25963902

ABSTRACT

The article entitled "Novel and safer self-inactivating vectors for gene therapy of Wiskott-Aldrich Syndrome", by E. G. Coci1, T. Maetzig, D. Zychlinski, M. Rothe, J. D. Suerth, C. Klein and A.Schambach has been retracted, on the request of the authors.Kindly see Bentham Science Policy on Article retraction at the link given below:(https://www.benthamscience.com/journals/current-medical-imaging/author-guidelines/).It is a pre-requisite for authors to declare explicitly that their work is original and has not been published elsewhere. Authors are advised to properly cite the original source to avoid plagiarism and copyright violation. As such this article represents a severe abuse of the scientific publishing system. Bentham Science Publishers takes a very strong view on this matter and apologizes to the readers of the journal for any inconvenience this may cause.

5.
Mol Ther ; 22(5): 919-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24434935

ABSTRACT

Methods for generating induced pluripotent stem cells (iPSCs) for disease modeling and cell therapies have progressed from integrating vectors to transient delivery of reprogramming factors, avoiding permanent genomic modification. A major limitation of unmodified iPSCs is the assessment of their distribution and contribution to adverse reactions in autologous cell therapy. Here, we report that polycistronic lentiviral vectors with single Flp recombinase (Flp) recognition target (FRT) sites can be used to generate murine iPSCs that are devoid of the reprogramming cassette but carry an intergenic 300-bp long terminal repeat sequence. Performing quantitative polymerase chain reaction on this marker, we could determine genetic identity and tissue contribution of iPSC-derived teratomas in mice. Moreover, we generated iPSCs carrying heterospecific FRT twin sites, enabling excision and recombinase-mediated cassette exchange (RMCE) of the reprogramming cassette for another expression unit of choice. Following screening of iPSCs for "safe harbor" integration sites, expression cassettes were introduced by RMCE into various previously silenced loci of selected single-copy iPSCs. Analysis of DNA methylation showed that RMCE reverted the local epigenetic signature, which allowed transgene expression in undifferentiated iPSCs and in differentiated progeny. These findings support the concept of creating clonotypically defined exchangeable and traceable pluripotent stem cells for disease research and cell therapy.


Subject(s)
Cell Differentiation/genetics , Cell- and Tissue-Based Therapy , DNA Nucleotidyltransferases/genetics , Induced Pluripotent Stem Cells , Terminal Repeat Sequences/genetics , Animals , Cellular Reprogramming , DNA Methylation , Genetic Vectors , Lentivirus/genetics , Mice
6.
Stem Cells Transl Med ; 2(9): 641-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23926210

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) hold great promise for studies on disease-related developmental processes and may serve as an autologous cell source for future treatment of many hereditary diseases. New genetic engineering tools such as zinc finger nucleases and transcription activator-like effector nuclease allow targeted correction of monogenetic disorders but are very cumbersome to establish. Aiming at studies on the knockdown of a disease-causing gene, lentiviral vector-mediated expression of short hairpin RNAs (shRNAs) is a valuable option, but it is limited by silencing of the knockdown construct upon epigenetic remodeling during differentiation. Here, we propose an approach for the expression of a therapeutic shRNA in disease-specific iPSCs using third-generation lentiviral vectors. Targeting severe α-1-antitrypsin (A1AT) deficiency, we overexpressed a human microRNA 30 (miR30)-styled shRNA directed against the PiZ variant of A1AT, which is known to cause chronic liver damage in affected patients. This knockdown cassette is traceable from clonal iPSC lines to differentiated hepatic progeny via an enhanced green fluorescence protein reporter expressed from the same RNA-polymerase II promoter. Importantly, the cytomegalovirus i/e enhancer chicken ß actin (CAG) promoter-driven expression of this construct is sustained without transgene silencing during hepatic differentiation in vitro and in vivo. At low lentiviral copy numbers per genome we confirmed a functional relevant reduction (-66%) of intracellular PiZ protein in hepatic cells after differentiation of patient-specific iPSCs. In conclusion, we have demonstrated that lentiviral vector-mediated expression of shRNAs can be efficiently used to knock down and functionally evaluate disease-related genes in patient-specific iPSCs.


Subject(s)
Gene Knockdown Techniques/methods , Genetic Therapy/methods , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Lentivirus/genetics , MicroRNAs/genetics , alpha 1-Antitrypsin Deficiency/therapy , Animals , Cell Differentiation , Cells, Cultured , Genetic Vectors , Green Fluorescent Proteins/genetics , Hepatocytes/cytology , Hepatocytes/virology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/virology , Mice , Mice, Transgenic , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA, Small Interfering/genetics , Transgenes , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/metabolism
7.
Hum Gene Ther ; 24(2): 132-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23311447

ABSTRACT

Over the past decades, lentiviral vectors have evolved as a benchmark tool for stable gene transfer into cells with a high replicative potential. Their relatively flexible genome and ability to transduce many forms of nondividing cells, combined with the potential for cell-specific pseudotyping, provides a rich resource for numerous applications in experimental platforms and therapeutic settings. Here, we give an overview of important biosafety features of lentiviral vectors, with detailed discussion of (i) the principles of the lentiviral split-genome design used for the construction of packaging cells; (ii) the relevance of modifications introduced into the lentiviral long terminal repeat (deletion of enhancer/promoter sequences and introduction of insulators); (iii) the basic features of mRNA processing, including the Rev/Rev-responsive element (RRE) interaction and the modifications of the 3' untranslated region of lentiviral vectors with various post-transcriptional regulatory elements affecting transcriptional termination, polyadenylation, and differentiation-specific degradation of mRNA; and (iv) the characteristic integration pattern with the associated risk of transcriptional interference with cellular genes. We conclude with considerations regarding the importance of cell targeting via envelope modifications. Along this course, we address canonical biosafety issues encountered with any type of viral vector: the risks of shedding, mobilization, germline transmission, immunogenicity, and insertional mutagenesis.


Subject(s)
Genetic Vectors/metabolism , Genome, Viral , Lentivirus/metabolism , Enhancer Elements, Genetic , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Lentivirus/physiology , Lentivirus Infections/prevention & control , Mutagenesis, Insertional , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Virus Activation , Virus Integration , Virus Replication , Virus Shedding
8.
EMBO J ; 31(20): 4035-44, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22968171

ABSTRACT

Biallelic mutations in the untranslated regions (UTRs) of mRNAs are rare causes for monogenetic diseases whose mechanisms remain poorly understood. We investigated a 3'UTR mutation resulting in a complex immunodeficiency syndrome caused by decreased mRNA levels of p14/robld3 by a previously unknown mechanism. Here, we show that the mutation creates a functional 5' splice site (SS) and that its recognition by the spliceosomal component U1 snRNP causes p14 mRNA suppression in the absence of splicing. Histone processing signals are able to rescue p14 expression. Therefore, the mutation interferes only with canonical poly(A)-site 3' end processing. Our data suggest that U1 snRNP inhibits cleavage or poly(A) site recognition. This is the first description of a 3'UTR mutation that creates a functional 5'SS causative of a monogenetic disease. Moreover, our data endorse the recently described role of U1 snRNP in suppression of intronic poly(A) sites, which is here deleterious for p14 mRNA biogenesis.


Subject(s)
3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/deficiency , Immunologic Deficiency Syndromes/genetics , Neutropenia/congenital , Polyadenylation/genetics , RNA Splice Sites/genetics , RNA, Small Nuclear/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Conserved Sequence , Endosomes/ultrastructure , Gene Expression Regulation/drug effects , Genes, Reporter , Histones/physiology , Humans , Introns/genetics , Mammals/genetics , Molecular Sequence Data , Morpholinos/pharmacology , Neutropenia/genetics , Point Mutation , Polyadenylation/drug effects , RNA Splicing/drug effects , RNA Stability , RNA, Messenger/biosynthesis , Sequence Alignment , Sequence Homology, Nucleic Acid , Species Specificity
9.
Biotechnol Bioeng ; 109(7): 1855-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22328297

ABSTRACT

For the aim of ex vivo engineering of functional tissue substitutes, Laser-assisted BioPrinting (LaBP) is under investigation for the arrangement of living cells in predefined patterns. So far three-dimensional (3D) arrangements of single or two-dimensional (2D) patterning of different cell types have been presented. It has been shown that cells are not harmed by the printing procedure. We now demonstrate for the first time the 3D arrangement of vital cells by LaBP as multicellular grafts analogous to native archetype and the formation of tissue by these cells. For this purpose, fibroblasts and keratinocytes embedded in collagen were printed in 3D as a simple example for skin tissue. To study cell functions and tissue formation process in 3D, different characteristics, such as cell localisation and proliferation were investigated. We further analysed the formation of adhering and gap junctions, which are fundamental for tissue morphogenesis and cohesion. In this study, it was demonstrated that LaBP is an outstanding tool for the generation of multicellular 3D constructs mimicking tissue functions. These findings are promising for the realisation of 3D in vitro models and tissue substitutes for many applications in tissue engineering.


Subject(s)
Collagen/chemistry , Fibroblasts/cytology , Keratinocytes/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioartificial Organs , Cell Line , Cell Proliferation , Fibroblasts/ultrastructure , Gap Junctions/ultrastructure , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Keratinocytes/ultrastructure , Lasers , Mice , Skin/cytology
10.
Hum Mol Genet ; 21(1): 32-45, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21949351

ABSTRACT

Many human diseases share a developmental origin that manifests during childhood or maturity. Aneuploid syndromes are caused by supernumerary or reduced number of chromosomes and represent an extreme example of developmental disease, as they have devastating consequences before and after birth. Investigating how alterations in gene dosage drive these conditions is relevant because it might help treat some clinical aspects. It may also provide explanations as to how quantitative differences in gene expression determine phenotypic diversity and disease susceptibility among natural populations. Here, we aimed to produce induced pluripotent stem cell (iPSC) lines that can be used to improve our understanding of aneuploid syndromes. We have generated iPSCs from monosomy X [Turner syndrome (TS)], trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome) and partial trisomy 11;22 (Emanuel syndrome), using either skin fibroblasts from affected individuals or amniocytes from antenatal diagnostic tests. These cell lines stably maintain the karyotype of the donors and behave like embryonic stem cells in all tested assays. TS iPSCs were used for further studies including global gene expression analysis and tissue-specific directed differentiation. Multiple clones displayed lower levels of the pseudoautosomal genes ASMTL and PPP2R3B than the controls. Moreover, they could be transformed into neural-like, hepatocyte-like and heart-like cells, but displayed insufficient up-regulation of the pseudoautosomal placental gene CSF2RA during embryoid body formation. These data support that abnormal organogenesis and early lethality in TS are not caused by a tissue-specific differentiation blockade, but rather involves other abnormalities including impaired placentation.


Subject(s)
Aneuploidy , Chromosome Disorders/genetics , Induced Pluripotent Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Chromosome Disorders/metabolism , Chromosome Disorders/physiopathology , Female , Gene Expression , Humans , Induced Pluripotent Stem Cells/metabolism , Infant , Male , Models, Genetic
11.
Mol Pharm ; 8(5): 1525-37, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21851067

ABSTRACT

Gene therapy is a promising therapeutic approach to treat primary immunodeficiencies. Indeed, the clinical trial for the Wiskott-Aldrich Syndrome (WAS) that is currently ongoing at the Hannover Medical School (Germany) has recently reported the correction of all affected cell lineages of the hematopoietic system in the first treated patients. However, an extensive study of the clonal inventory of those patients reveals that LMO2, CCND2 and MDS1/EVI1 were preferentially prevalent. Moreover, a first leukemia case was observed in this study, thus reinforcing the need of developing safer vectors for gene transfer into HSC in general. Here we present a novel self-inactivating (SIN) vector for the gene therapy of WAS that combines improved safety features. We used the elongation factor 1 alpha (EFS) promoter, which has been extensively evaluated in terms of safety profile, to drive a codon-optimized human WASP cDNA. To test vector performance in a more clinically relevant setting, we transduced murine HSPC as well as human CD34+ cells and also analyzed vector efficacy in their differentiated myeloid progeny. Our results show that our novel vector generates comparable WAS protein levels and is as effective as the clinically used LTR-driven vector. Therefore, the described SIN vectors appear to be good candidates for potential use in a safer new gene therapy protocol for WAS, with decreased risk of insertional mutagenesis.


Subject(s)
Genetic Vectors/adverse effects , Hematopoietic Stem Cells/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/metabolism , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Combined Modality Therapy/adverse effects , Feasibility Studies , Gene Expression Regulation, Viral , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hematopoietic Stem Cells/virology , Hepatitis B Virus, Woodchuck/genetics , Hepatitis B Virus, Woodchuck/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Cells/virology , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stem Cell Transplantation/adverse effects , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/therapy , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/therapeutic use
12.
Nucleic Acids Res ; 39(16): 7147-60, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21609958

ABSTRACT

The Sleeping Beauty (SB) transposase and its newly developed hyperactive variant, SB100X, are of increasing interest for genome modification in experimental models and gene therapy. The potential cytotoxicity of transposases requires careful assessment, considering that residual integration events of transposase expression vectors delivered by physicochemical transfection or episomal retroviral vectors may lead to permanent transposase expression and resulting uncontrollable transposition. Comparing retrovirus-based approaches for delivery of mRNA, episomal DNA or integrating DNA, we found that conventional SB transposase, SB100X and a newly developed codon-optimized SB100Xo may trigger premitotic arrest and apoptosis. Cell stress induced by continued SB overexpression was self-limiting due to the induction of cell death, which occurred even in the absence of a co-transfected transposable element. The cytotoxic effects of SB transposase were strictly dose dependent and heralded by induction of p53 and c-Jun. Inactivating mutations in SB's catalytic domain could not abrogate cytotoxicity, suggesting a mechanism independent of DNA cleavage activity. An improved approach of retrovirus particle-mediated mRNA transfer allowed transient and dose-controlled expression of SB100X, supported efficient transposition and prevented cytotoxicity. Transposase-mediated gene transfer can thus be tuned to maintain high efficiency in the absence of overt cell damage.


Subject(s)
RNA, Messenger/metabolism , Transduction, Genetic , Transposases/genetics , Caspase Inhibitors , Cell Cycle , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Genetic Vectors , HeLa Cells , Humans , Retroviridae/genetics , Transposases/metabolism , Virion/genetics
13.
Hum Mol Genet ; 20(16): 3176-87, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21593220

ABSTRACT

Directed hepatocyte differentiation from human induced pluripotent stem cells (iPSCs) potentially provides a unique platform for modeling liver genetic diseases and performing drug-toxicity screening in vitro. Wilson's disease is a genetic disease caused by mutations in the ATP7B gene, whose product is a liver transporter protein responsible for coordinated copper export into bile and blood. Interestingly, the spectrum of ATP7B mutations is vast and can influence clinical presentation (a variable spectrum of hepatic and neural manifestations), though the reason is not well understood. We describe the generation of iPSCs from a Chinese patient with Wilson's disease that bears the R778L Chinese hotspot mutation in the ATP7B gene. These iPSCs were pluripotent and could be readily differentiated into hepatocyte-like cells that displayed abnormal cytoplasmic localization of mutated ATP7B and defective copper transport. Moreover, gene correction using a self-inactivating lentiviral vector that expresses codon optimized-ATP7B or treatment with the chaperone drug curcumin could reverse the functional defect in vitro. Hence, our work describes an attractive model for studying the pathogenesis of Wilson's disease that is valuable for screening compounds or gene therapy approaches aimed to correct the abnormality. In the future, once relevant safety concerns (including the stability of the mature liver-like phenotype) and technical issues for the transplantation procedure are solved, hepatocyte-like cells from similarly genetically corrected iPSCs could be an option for autologous transplantation in Wilson's disease.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Curcumin/therapeutic use , Genetic Therapy , Hepatocytes/metabolism , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/pathology , Induced Pluripotent Stem Cells/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/therapeutic use , Base Sequence , Cation Transport Proteins/metabolism , Cation Transport Proteins/therapeutic use , Copper/metabolism , Copper-Transporting ATPases , Humans , Male , Middle Aged , Molecular Chaperones/therapeutic use , Molecular Sequence Data , Mutation/genetics , Protein Transport , Subcellular Fractions/metabolism
14.
Proc Natl Acad Sci U S A ; 107(17): 7805-10, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20385817

ABSTRACT

Retroviral particles assemble a few thousand units of the Gag polyproteins. Proteolytic cleavage mediated by the retroviral protease forms the bioactive retroviral protein subunits before cell entry. We hypothesized that this process could be exploited for targeted, transient, and dose-controlled transduction of nonretroviral proteins into cultured cells. We demonstrate that gammaretroviral particles tolerate the incorporation of foreign protein at several positions of their Gag or Gag-Pol precursors. Receptor-mediated and thus potentially cell-specific uptake of engineered particles occurred within minutes after cell contact. Dose and kinetics of nonretroviral protein delivery were dependent upon the location within the polyprotein precursor. Proteins containing nuclear localization signals were incorporated into retroviral particles, and the proteins of interest were released from the precursor by the retroviral protease, recognizing engineered target sites. In contrast to integration-defective lentiviral vectors, protein transduction by retroviral polyprotein precursors was completely transient, as protein transducing retrovirus-like particles could be produced that did not transduce genes into target cells. Alternatively, bifunctional protein-delivering particle preparations were generated that maintained their ability to serve as vectors for retroviral transgenes. We show the potential of this approach for targeted genome engineering of induced pluripotent stem cells by delivering the site-specific DNA recombinase, Flp. Protein transduction of Flp after proteolytic release from the matrix position of Gag allowed excision of a lentivirally transduced cassette that concomitantly expresses the canonical reprogramming transcription factors (Oct4, Klf4, Sox2, c-Myc) and a fluorescent marker gene, thus generating induced pluripotent stem cells that are free of lentivirally transduced reprogramming genes.


Subject(s)
Gene Products, gag/biosynthesis , Leukemia Virus, Murine/metabolism , Transduction, Genetic/methods , Virion/metabolism , Virus Internalization , Gene Products, gag/genetics , Genetic Engineering/methods , Green Fluorescent Proteins/metabolism , Kinetics , Leukemia Virus, Murine/genetics , Nuclear Localization Signals/metabolism , Peptide Hydrolases/metabolism , Virion/genetics
15.
Nucleic Acids Res ; 37(22): 7429-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19854941

ABSTRACT

Multiple types of regulation are used by cells and viruses to control alternative splicing. In murine leukemia virus, accessibility of the 5' splice site (ss) is regulated by an upstream region, which can fold into a complex RNA stem-loop structure. The underlying sequence of the structure itself is negligible, since most of it could be functionally replaced by a simple heterologous RNA stem-loop preserving the wild-type splicing pattern. Increasing the RNA duplex formation between U1 snRNA and the 5'ss by a compensatory mutation in position +6 led to enhanced splicing. Interestingly, this mutation affects splicing only in the context of the secondary structure, arguing for a dynamic interplay between structure and primary 5'ss sequence. The reduced 5'ss accessibility could also be counteracted by recruiting a splicing enhancer domain via a modified MS2 phage coat protein to a single binding site at the tip of the simple RNA stem-loop. The mechanism of 5'ss attenuation was revealed using hyperstable U1 snRNA mutants, showing that restricted U1 snRNP access is the cause of retroviral alternative splicing.


Subject(s)
Alternative Splicing , Leukemia Virus, Murine/genetics , RNA Splice Sites , RNA, Small Nuclear/chemistry , Base Sequence , Cell Line , Humans , Leukemia Virus, Murine/physiology , Molecular Sequence Data , Nuclear Proteins/chemistry , Nucleic Acid Conformation , Protein Structure, Tertiary , Proviruses/genetics , Proviruses/physiology , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , Serine-Arginine Splicing Factors , Suppression, Genetic , Virus Replication
16.
Mol Ther ; 17(11): 1919-28, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19672245

ABSTRACT

Gene transfer vectors may cause clonal imbalance and even malignant cell transformation by insertional upregulation of proto-oncogenes. Lentiviral vectors (LV) with their preferred integration in transcribed genes are considered less genotoxic than gammaretroviral vectors (GV) with their preference for integration next to transcriptional start sites and regulatory gene regions. Using a sensitive cell culture assay and a series of self-inactivating (SIN) vectors, we found that the lentiviral insertion pattern was approximately threefold less likely than the gammaretroviral to trigger transformation of primary hematopoietic cells. However, lentivirally induced mutants also showed robust replating, in line with the selection for common insertion sites (CIS) in the first intron of the Evi1 proto-oncogene. This potent proto-oncogene thus represents a CIS for both GV and LV, despite major differences in their integration mechanisms. Altering the vectors' enhancer-promoter elements had a greater effect on safety than the retroviral insertion pattern. Clinical grade LV expressing the Wiskott-Aldrich syndrome (WAS) protein under control of its own promoter had no transforming potential. Mechanistic studies support the conclusion that enhancer-mediated gene activation is the major cause for insertional transformation of hematopoietic cells, opening rational strategies for risk prevention.


Subject(s)
Bone Marrow Cells/metabolism , Gammaretrovirus/genetics , Genetic Vectors/genetics , Lentivirus/genetics , Mutagenesis, Insertional/methods , Transduction, Genetic/methods , Animals , Blotting, Northern , Bone Marrow Cells/virology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Proto-Oncogenes/genetics
17.
Mol Ther ; 16(4): 718-25, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18334985

ABSTRACT

The possible activation of cellular proto-oncogenes as a result of clonal transformation is a potential limitation in a therapeutic approach involving random integration of gene vectors. Given that enhancer promiscuity represents an important mechanism of insertional transformation, we assessed the enhancer activities of various cellular and retroviral promoters in transient transfection assays, and also in a novel experimental system designed to measure the activation of a minigene cassette contained in stably integrating retroviral vectors. Retroviral enhancer-promoters showed a significantly greater potential to activate neighboring promoters than did cellular promoters derived from human genes, elongation factor-1alpha (EF1alpha) and phosphoglycerate kinase (PGK). Self-inactivating (SIN) vector design reduced but did not abolish enhancer interactions. Using a recently established cell culture assay that detects insertional transformation by serial replating of primary hematopoietic cells, we found that SIN vectors containing the EF1alpha promoter greatly decrease the risk of insertional transformation. Despite integration of multiple copies per cell, activation of the crucial proto-oncogene Evi1 was not detectable when using SIN-EF1alpha vectors. On the basis of several quantitative indicators, the decrease in transforming activity was highly significant (more than tenfold, P < 0.01) when compared with similarly designed vectors containing a retroviral enhancer-promoter with or without a well-characterized genetic insulator core element. In this manner, the insertional biosafety of therapeutic gene vectors can be greatly enhanced and proactively evaluated in sensitive cell-based assays.


Subject(s)
Genetic Vectors/toxicity , Promoter Regions, Genetic , Retroviridae/genetics , Transfection/methods , Animals , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Fibroblasts/metabolism , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , Proto-Oncogene Mas , Proto-Oncogenes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
18.
J Virol ; 81(7): 3652-6, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17229710

ABSTRACT

Alternative splicing has been recognized as a major mechanism for creating proteomic diversity from a limited number of genes. However, not all determinants regulating this process have been characterized. Using subviral human immunodeficiency virus (HIV) env constructs we observed an enhanced splicing of the RNA when expression was under control of the cytomegalovirus (CMV) promoter instead of the HIV long terminal repeat (LTR). We extended these observations to LTR- or CMV-driven murine leukemia proviruses, suggesting that retroviral LTRs are adapted to inefficient alternative splicing at most sites in order to maintain balanced gene expression.


Subject(s)
Alternative Splicing/genetics , HIV/genetics , Leukemia Virus, Murine/genetics , Promoter Regions, Genetic/genetics , Cytomegalovirus/genetics , Gene Products, tat/genetics , Gene Products, tat/metabolism , HIV/metabolism , HIV Long Terminal Repeat , Leukemia Virus, Murine/metabolism , RNA, Viral/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus
19.
J Biol Chem ; 281(49): 37381-90, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17038324

ABSTRACT

Alternative splicing of the primary transcript plays a key role in retroviral gene expression. In contrast to all known mechanisms that mediate alternative splicing in retroviruses, we found that in murine leukemia virus, distinct elements located upstream of the 5' splice site either inhibited or activated splicing of the genomic RNA. Detailed analysis of the first untranslated exon showed that the primer binding site (PBS) activates splicing, whereas flanking sequences either downstream or upstream of the PBS are inhibitory. This new function of the PBS was independent of its orientation and primer binding but associated with a particular destabilizing role in a proposed secondary structure. On the contrary, all sequences surrounding the PBS that are involved in stem formation of the first exon were found to suppress splicing. Targeted mutations that destabilized the central stem and compensatory mutations of the counter strand clearly validated the concept that murine leukemia virus attenuates its 5' splice site by forming an inhibitory stem-loop in its first exon. Importantly, this mode of splice regulation was conserved in a complete proviral clone. Some of the mutants that increase splicing revealed an opposite effect on translation, implying that the first exon also regulates this process. Together, these findings suggest that sequences upstream of the 5' splice site play an important role in splice regulation of simple retroviruses, directly or indirectly attenuating the efficiency of splicing.


Subject(s)
Alternative Splicing , Leukemia Virus, Murine/genetics , 5' Untranslated Regions , Animals , Base Sequence , Cell Line , DNA, Viral/genetics , Exons , Gene Expression Regulation, Viral , Genetic Vectors , Leukemia Virus, Murine/metabolism , Mice , Models, Molecular , Nucleic Acid Conformation , Plasmids/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...