1
|
Tang PZ, Ding B, Reyes C, Papp D, Potter J. Target-seq: single workflow for detection of genome integration site, DNA translocation and off-target events. Biotechniques 2023. [PMID: 37161298 DOI: 10.2144/btn-2023-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Designed donor DNA delivery through viral or nonviral systems to target loci in the host genome is a critical step for gene therapy. Adeno-associated virus and lentivirus are leading vehicles for in vivo and ex vivo delivery of therapeutic genes due to their high delivery and editing efficiency. Nonviral editing tools, such as CRISPR/Cas9, are getting more attention for gene modification. However, there are safety concerns; for example, tumorigenesis due to off-target effects and DNA rearrangement. Analysis tools to detect and characterize on-target and off-target genome modification post editing in the host genome are pivotal for evaluating the success and safety of gene therapy. We developed Target-seq combined with different analysis tools to detect the genome integration site, DNA translocation and off-target events.
Collapse
Affiliation(s)
| | - Bo Ding
- Thermo Fisher Scientific, Inc., MA, USA
| | | | | | | |
Collapse
|
2
|
Gödecke N, Herrmann S, Hauser H, Mayer-Bartschmid A, Trautwein M, Wirth D. Rational Design of Single Copy Expression Cassettes in Defined Chromosomal Sites Overcomes Intraclonal Cell-to-Cell Expression Heterogeneity and Ensures Robust Antibody Production. ACS Synth Biol 2021; 10:145-157. [PMID: 33382574 DOI: 10.1021/acssynbio.0c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The expression of endogenous genes as well as transgenes depends on regulatory elements within and surrounding genes as well as their epigenetic modifications. Members of a cloned cell population often show pronounced cell-to-cell heterogeneity with respect to the expression of a certain gene. To investigate the heterogeneity of recombinant protein expression we targeted cassettes into two preselected chromosomal hot-spots in Chinese hamster ovary (CHO) cells. Depending on the gene of interest and the design of the expression cassette, we found strong expression variability that could be reduced by epigenetic modifiers, but not by site-specific recruitment of the modulator dCas9-VPR. In particular, the implementation of ubiquitous chromatin opening elements (UCOEs) reduced cell-to-cell heterogeneity and concomitantly increased expression. The application of this method to recombinant antibody expression confirmed that rational design of cell lines for production of transgenes with predictable and high titers is a promising approach.
Collapse
Affiliation(s)
- Natascha Gödecke
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Sabrina Herrmann
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Hansjörg Hauser
- Staff Unit Scientific Strategy, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | | | | | - Dagmar Wirth
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
- Institute of Experimental Hematology, Medical University Hannover, Hannover 30625, Germany
| |
Collapse
|
3
|
Cullmann K, Jahn M, Spindler M, Schenk F, Manukjan G, Mucci A, Steinemann D, Boller K, Schulze H, Bender M, Moritz T, Modlich U. Forming megakaryocytes from murine-induced pluripotent stem cells by the inducible overexpression of supporting factors. Res Pract Thromb Haemost 2020; 5:111-124. [PMID: 33537535 PMCID: PMC7845061 DOI: 10.1002/rth2.12453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/19/2023] Open
Abstract
Background Platelets are small anucleate cells that circulate in the blood in a resting state but can be activated by external cues. In case of need, platelets from blood donors can be transfused. As an alternative source, platelets can be produced from induced pluripotent stem cells (iPSCs); however, recovered numbers are low. Objectives To optimize megakaryocyte (MK) and platelet output from murine iPSCs, we investigated overexpression of the transcription factors GATA‐binding factor 1 (GATA1); nuclear factor, erythroid 2; and pre–B‐cell leukemia transcription factor 1 (Pbx1) and a hyperactive variant of the small guanosine triphosphatase RhoA (RhoAhc). Methods To avoid off‐target effects, we generated iPSCs carrying the reverse tetracycline‐responsive transactivator M2 (rtTA‐M2) in the Rosa26 locus and expressed the factors from Tet‐inducible gammaretroviral vectors. Differentiation of iPSCs was initiated by embryoid body (EB) formation. After EB dissociation, early hematopoietic progenitors were enriched and cocultivated on OP9 feeder cells with thrombopoietin and stem cell factor to induce megakaryocyte (MK) differentiation. Results Overexpression of GATA1 and Pbx1 increased MK output 2‐ to 2.5‐fold and allowed prolonged collection of MK. Cytologic and ultrastructural analyses identified typical MK with enlarged cells, multilobulated nuclei, granule structures, and an internal membrane system. However, GATA1 and Pbx1 expression did not improve MK maturation or platelet release, although in vitro–generated platelets were functional in spreading on fibrinogen or collagen‐related peptide. Conclusion We demonstrate that the use of rtTA‐M2 transgenic iPSCs transduced with Tet‐inducible retroviral vectors allowed for gene expression at later time points during differentiation. With this strategy we could identify factors that increased in vitro MK production. ![]()
Collapse
Affiliation(s)
- Katharina Cullmann
- RG Gene Modification in Stem Cells, Division of Veterinary Medicine Paul-Ehrlich-Institut Langen Germany
| | - Magdalena Jahn
- RG Gene Modification in Stem Cells, Division of Veterinary Medicine Paul-Ehrlich-Institut Langen Germany
| | - Markus Spindler
- Institute of Experimental Biomedicine I University Hospital and Rudolf Virchow Center University of Würzburg Würzburg Germany
| | - Franziska Schenk
- RG Gene Modification in Stem Cells, Division of Veterinary Medicine Paul-Ehrlich-Institut Langen Germany
| | - Georgi Manukjan
- Institute of Experimental Biomedicine I University Hospital and Rudolf Virchow Center University of Würzburg Würzburg Germany
| | - Adele Mucci
- RG Reprogramming and Gene Therapy, Institute of Experimental Hematology, Hannover Medical School Hannover Germany.,Present address: San Raffaele Telethon Institute for Gene Therapy Milano Italy
| | - Doris Steinemann
- Department of Human Genetics Hannover Medical School Hannover Germany
| | - Klaus Boller
- Morphology, Division of Immunology Paul-Ehrlich-Institut Langen Germany
| | - Harald Schulze
- Institute of Experimental Biomedicine I University Hospital and Rudolf Virchow Center University of Würzburg Würzburg Germany
| | - Markus Bender
- Institute of Experimental Biomedicine I University Hospital and Rudolf Virchow Center University of Würzburg Würzburg Germany
| | - Thomas Moritz
- RG Reprogramming and Gene Therapy, Institute of Experimental Hematology, Hannover Medical School Hannover Germany
| | - Ute Modlich
- RG Gene Modification in Stem Cells, Division of Veterinary Medicine Paul-Ehrlich-Institut Langen Germany
| |
Collapse
|
4
|
Kesselring L, Miskey C, Zuliani C, Querques I, Kapitonov V, Laukó A, Fehér A, Palazzo A, Diem T, Lustig J, Sebe A, Wang Y, Dinnyés A, Izsvák Z, Barabas O, Ivics Z. A single amino acid switch converts the Sleeping Beauty transposase into an efficient unidirectional excisionase with utility in stem cell reprogramming. Nucleic Acids Res 2020; 48:316-331. [PMID: 31777924 PMCID: PMC6943129 DOI: 10.1093/nar/gkz1119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 11/07/2019] [Accepted: 11/22/2019] [Indexed: 12/26/2022] Open
Abstract
The Sleeping Beauty (SB) transposon is an advanced tool for genetic engineering and a useful model to investigate cut-and-paste DNA transposition in vertebrate cells. Here, we identify novel SB transposase mutants that display efficient and canonical excision but practically unmeasurable genomic re-integration. Based on phylogenetic analyses, we establish compensating amino acid replacements that fully rescue the integration defect of these mutants, suggesting epistasis between these amino acid residues. We further show that the transposons excised by the exc+/int− transposase mutants form extrachromosomal circles that cannot undergo a further round of transposition, thereby representing dead-end products of the excision reaction. Finally, we demonstrate the utility of the exc+/int− transposase in cassette removal for the generation of reprogramming factor-free induced pluripotent stem cells. Lack of genomic integration and formation of transposon circles following excision is reminiscent of signal sequence removal during V(D)J recombination, and implies that cut-and-paste DNA transposition can be converted to a unidirectional process by a single amino acid change.
Collapse
Affiliation(s)
- Lisa Kesselring
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Csaba Miskey
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Cecilia Zuliani
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Irma Querques
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Vladimir Kapitonov
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | | | - Anita Fehér
- BioTalentum Ltd, Gödöllő, 2100 Gödöllő, Hungary
| | - Antonio Palazzo
- Department of Biology, University of Bari 'Aldo Moro', Italy
| | - Tanja Diem
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Janna Lustig
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Attila Sebe
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Yongming Wang
- Mobile DNA, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Zsuzsanna Izsvák
- Mobile DNA, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Orsolya Barabas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Zoltán Ivics
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| |
Collapse
|
5
|
Protein trap: a new Swiss army knife for geneticists? Mol Biol Rep 2019; 47:1445-1458. [PMID: 31728729 DOI: 10.1007/s11033-019-05181-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
The protein trap is a powerful tool for genetic and biochemical studies of gene function in the animal kingdom. Although the original protein trap was developed for flies, it can be easily adapted to other multicellular organisms, both known models and ones with an unsequenced genome. The protein trap has been successfully applied to the fruit fly, crustaceans Parhyale hawaiensis, zebrafish, and insect and animal cell cultures. This approach is based on the integration into genes of an artificial exon that carries DNA encoding a fluorescent marker, standardized immunoepitopes, an integrase docking site, and splice acceptor and donor sites. The protein trap for cell cultures additionally contains an antibiotic resistance gene, which facilitates the selection of trapped clones. Resulting chimeric tagged mRNAs can be interfered by dsRNA against GFP (iGFPi-in vivo GFP interference), or the chimeric proteins can be efficiently knocked down by deGradFP technology. Both RNA and protein knockdowns produce a strong loss of function phenotype in tagged cells. The fluorescent and protein affinity tags can be used for tagged protein localisation within the cell and for identifying their binding partners in their native complexes. Insertion into protein trap integrase docking sites allows the replacement of trap contents by any new constructs, including other markers, cell toxins, stop-codons, and binary expression systems such as GAL4/UAS, LexA/LexAop and QF/QUAS, that reliably reflect endogenous gene expression. A distinctive feature of the protein trap approach is that all manipulations with a gene or its product occur only in the endogenous locus, which cannot be achieved by any other method.
Collapse
|
6
|
Mucci A, Lopez-Rodriguez E, Hetzel M, Liu S, Suzuki T, Happle C, Ackermann M, Kempf H, Hillje R, Kunkiel J, Janosz E, Brennig S, Glage S, Bankstahl JP, Dettmer S, Rodt T, Gohring G, Trapnell B, Hansen G, Trapnell C, Knudsen L, Lachmann N, Moritz T. iPSC-Derived Macrophages Effectively Treat Pulmonary Alveolar Proteinosis in Csf2rb-Deficient Mice. Stem Cell Reports 2018; 11:696-710. [PMID: 30100408 PMCID: PMC6135208 DOI: 10.1016/j.stemcr.2018.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/13/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived hematopoietic cells represent a highly attractive source for cell and gene therapy. Given the longevity, plasticity, and self-renewal potential of distinct macrophage subpopulations, iPSC-derived macrophages (iPSC-Mφ) appear of particular interest in this context. We here evaluated the airway residence, plasticity, and therapeutic efficacy of iPSC-Mφ in a murine model of hereditary pulmonary alveolar proteinosis (herPAP). We demonstrate that single pulmonary macrophage transplantation (PMT) of 2.5–4 × 106 iPSC-Mφ yields efficient airway residence with conversion of iPSC-Mφ to an alveolar macrophage (AMφ) phenotype characterized by a distinct surface marker and gene expression profile within 2 months. Moreover, PMT significantly improves alveolar protein deposition and other critical herPAP disease parameters. Thus, our data indicate iPSC-Mφ as a source of functional macrophages displaying substantial plasticity and therapeutic potential that upon pulmonary transplantation will integrate into the lung microenvironment, adopt an AMφ phenotype and gene expression pattern, and profoundly ameliorate pulmonary disease phenotypes. iPSCs as a source of functional macrophages with substantial plasticity iPSC-derived macrophages have therapeutic potential in hereditary PAP Pulmonary-transplanted iPSC-Mφ integrate into the lung microenvironment iPSC-Mφ can adopt an AMφ phenotype and gene expression pattern
Collapse
Affiliation(s)
- Adele Mucci
- Research Group Reprogramming and Gene Therapy, Hannover Medical School (MHH), Hannover, Germany; Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany
| | - Elena Lopez-Rodriguez
- Department of Functional and Applied Anatomy, MHH, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Miriam Hetzel
- Research Group Reprogramming and Gene Therapy, Hannover Medical School (MHH), Hannover, Germany; Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany
| | - Serena Liu
- Department of Genome Sciences, Seattle, WA, USA
| | - Takuji Suzuki
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Pulmonary Medicine, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
| | - Christine Happle
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, MHH, Hannover, Germany
| | - Mania Ackermann
- Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Research Group Translational Hematology of Congenital Diseases, MHH, Hannover, Germany
| | - Henning Kempf
- Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Roman Hillje
- Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Research Group Translational Hematology of Congenital Diseases, MHH, Hannover, Germany
| | - Jessica Kunkiel
- Research Group Reprogramming and Gene Therapy, Hannover Medical School (MHH), Hannover, Germany; Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany
| | - Ewa Janosz
- Research Group Reprogramming and Gene Therapy, Hannover Medical School (MHH), Hannover, Germany; Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany
| | - Sebastian Brennig
- Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Research Group Translational Hematology of Congenital Diseases, MHH, Hannover, Germany
| | - Silke Glage
- Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Institute of Laboratory Animal Science and Central Animal Facility, MHH, Hannover, Germany
| | | | - Sabine Dettmer
- Department of Diagnostic and Interventional Radiology, MHH, Hannover, Germany
| | - Thomas Rodt
- Department of Diagnostic and Interventional Radiology, MHH, Hannover, Germany
| | | | - Bruce Trapnell
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gesine Hansen
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, MHH, Hannover, Germany
| | | | - Lars Knudsen
- Department of Functional and Applied Anatomy, MHH, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Nico Lachmann
- Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany; Research Group Translational Hematology of Congenital Diseases, MHH, Hannover, Germany.
| | - Thomas Moritz
- Research Group Reprogramming and Gene Therapy, Hannover Medical School (MHH), Hannover, Germany; Institute of Experimental Hematology, MHH, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; Cluster of Excellence REBIRTH, MHH, Hannover, Germany
| |
Collapse
|
7
|
Elsner C, Bohne J. The retroviral vector family: something for everyone. Virus Genes 2017; 53:714-722. [PMID: 28762206 DOI: 10.1007/s11262-017-1489-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/06/2017] [Indexed: 12/19/2022]
Abstract
After 30 years of retroviral vector research it became clear that the parental viruses can be both friend and foe. Especially human immunodeficiency virus sparked a global pandemic, but could be converted into a versatile tool for cell therapy. For all retroviral genera, the way from virus to vector was similar resulting in split-vector systems based on the separation of the genes needed for vector particle formation and transgene expression. The first gene therapy trials, although clinically effective, revealed the genotoxicity of retroviral vectors caused by insertional mutagenesis. This issue was solved using self-inactivating vectors carrying weaker cellular promoters. Further fine-tuning was able to generate inducible systems. The current toolbox also contains vectors for the generation of induced pluripotent stem cells or efficient RNA interference. More recently the application of CRISPR-Cas9-mediated gene editing led to the development of genome-wide small guide RNA libraries targeting all human genes and single lentiviral vectors for an easy delivery of Cas9.
Collapse
Affiliation(s)
- Carina Elsner
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jens Bohne
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
8
|
Geis FK, Galla M, Hoffmann D, Kuehle J, Zychlinski D, Maetzig T, Schott JW, Schwarzer A, Goffinet C, Goff SP, Schambach A. Potent and reversible lentiviral vector restriction in murine induced pluripotent stem cells. Retrovirology 2017; 14:34. [PMID: 28569216 PMCID: PMC5452410 DOI: 10.1186/s12977-017-0358-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
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. Electronic supplementary material The online version of this article (doi:10.1186/s12977-017-0358-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franziska K Geis
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Dirk Hoffmann
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Johannes Kuehle
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Daniela Zychlinski
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Tobias Maetzig
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Juliane W Schott
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Adrian Schwarzer
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Christine Goffinet
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infections Research, Hannover, Germany
| | - Stephen P Goff
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA.,Howard Hughes Medical Institute, Columbia University Medical Center, New York, NY, USA
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany. .,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany. .,Division of Hematology/Oncology, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Detailed comparison of retroviral vectors and promoter configurations for stable and high transgene expression in human induced pluripotent stem cells. Gene Ther 2017; 24:298-307. [PMID: 28346436 DOI: 10.1038/gt.2017.20] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/27/2017] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
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.
Collapse
|
10
|
LiPS-A3S, a human genomic site for robust expression of inserted transgenes. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e394. [PMID: 27898090 PMCID: PMC5155331 DOI: 10.1038/mtna.2016.99] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 10/06/2016] [Indexed: 01/14/2023]
Abstract
Transgenesis of human pluripotent stem cells (hPSCs) can enable and empower a variety of studies in stem cell research, including lineage tracing and functional genetics studies. While in recent years much progress has been made in the development of tools for gene targeting, little attention has been given to the identification of sites in the human genome where transgenes can be inserted and reliably expressed. In order to find human genomic sites capable of supporting long-term and high-level transgene expression in hPSCs, we performed a lentiviral screen in human induced pluripotent stem cells (iPSCs). We isolated 40 iPSC clones each harboring a single vector copy and characterized the level of transgene expression afforded by each unique integration site. We selected one clone, LiPS-A3 with an integration site in chromosome 15 maintaining robust expression without silencing and demonstrate that different transgenes can be inserted therein rapidly and efficiently through recombinase-mediated cassette exchange (RMCE). The LiPS-A3 line can greatly facilitate the insertion of reporter and other genes in hPSCs. Targeting transgenes in the LiPS-A3S genomic locus can find broad applications in stem cell research and possibly cell and gene therapy.
Collapse
|
11
|
Mucci A, Kunkiel J, Suzuki T, Brennig S, Glage S, Kühnel MP, Ackermann M, Happle C, Kuhn A, Schambach A, Trapnell BC, Hansen G, Moritz T, Lachmann N. Murine iPSC-Derived Macrophages as a Tool for Disease Modeling of Hereditary Pulmonary Alveolar Proteinosis due to Csf2rb Deficiency. Stem Cell Reports 2016; 7:292-305. [PMID: 27453007 PMCID: PMC4982988 DOI: 10.1016/j.stemcr.2016.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) represent an innovative source for the standardized in vitro generation of macrophages (Mφ). We here describe a robust and efficient protocol to obtain mature and functional Mφ from healthy as well as disease-specific murine iPSCs. With regard to morphology, surface phenotype, and function, our iPSC-derived Mφ (iPSC-Mφ) closely resemble their counterparts generated in vitro from bone marrow cells. Moreover, when we investigated the feasibility of our differentiation system to serve as a model for rare congenital diseases associated with Mφ malfunction, we were able to faithfully recapitulate the pathognomonic defects in GM-CSF signaling and Mφ function present in hereditary pulmonary alveolar proteinosis (herPAP). Thus, our studies may help to overcome the limitations placed on research into certain rare disease entities by the lack of an adequate supply of disease-specific primary cells, and may aid the development of novel therapeutic approaches for herPAP patients.
Collapse
Affiliation(s)
- Adele Mucci
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Junior Research Group Translational Hematology of Congenital Diseases, Cluster of Excellence REBIRTH, Hannover Medical School, Carl-Neuberg-Street 1, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Jessica Kunkiel
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Takuji Suzuki
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sebastian Brennig
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Junior Research Group Translational Hematology of Congenital Diseases, Cluster of Excellence REBIRTH, Hannover Medical School, Carl-Neuberg-Street 1, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Glage
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, 30625 Hannover, Germany
| | - Mark P Kühnel
- Department of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Mania Ackermann
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Junior Research Group Translational Hematology of Congenital Diseases, Cluster of Excellence REBIRTH, Hannover Medical School, Carl-Neuberg-Street 1, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Alexandra Kuhn
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Thomas Moritz
- Research Group Reprogramming and Gene Therapy, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Nico Lachmann
- Junior Research Group Translational Hematology of Congenital Diseases, Cluster of Excellence REBIRTH, Hannover Medical School, Carl-Neuberg-Street 1, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
12
|
Papapetrou EP, Schambach A. Gene Insertion Into Genomic Safe Harbors for Human Gene Therapy. Mol Ther 2016; 24:678-84. [PMID: 26867951 DOI: 10.1038/mt.2016.38] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/05/2016] [Indexed: 12/14/2022] Open
Abstract
Genomic safe harbors (GSHs) are sites in the genome able to accommodate the integration of new genetic material in a manner that ensures that the newly inserted genetic elements: (i) function predictably and (ii) do not cause alterations of the host genome posing a risk to the host cell or organism. GSHs are thus ideal sites for transgene insertion whose use can empower functional genetics studies in basic research and therapeutic applications in human gene therapy. Currently, no fully validated GSHs exist in the human genome. Here, we review our formerly proposed GSH criteria and discuss additional considerations on extending these criteria, on strategies for the identification and validation of GSHs, as well as future prospects on GSH targeting for therapeutic applications. In view of recent advances in genome biology, gene targeting technologies, and regenerative medicine, gene insertion into GSHs can potentially catalyze nearly all applications in human gene therapy.
Collapse
Affiliation(s)
- Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Thiesler CT, Cajic S, Hoffmann D, Thiel C, van Diepen L, Hennig R, Sgodda M, Weiβmann R, Reichl U, Steinemann D, Diekmann U, Huber NMB, Oberbeck A, Cantz T, Kuss AW, Körner C, Schambach A, Rapp E, Buettner FFR. Glycomic Characterization of Induced Pluripotent Stem Cells Derived from a Patient Suffering from Phosphomannomutase 2 Congenital Disorder of Glycosylation (PMM2-CDG). Mol Cell Proteomics 2016; 15:1435-52. [PMID: 26785728 PMCID: PMC4824866 DOI: 10.1074/mcp.m115.054122] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Indexed: 01/08/2023] Open
Abstract
PMM2-CDG, formerly known as congenital disorder of glycosylation-Ia (CDG-Ia), is caused by mutations in the gene encoding phosphomannomutase 2 (PMM2). This disease is the most frequent form of inherited CDG-diseases affecting protein N-glycosylation in human. PMM2-CDG is a multisystemic disease with severe psychomotor and mental retardation. In order to study the pathophysiology of PMM2-CDG in a human cell culture model, we generated induced pluripotent stem cells (iPSCs) from fibroblasts of a PMM2-CDG-patient (PMM2-iPSCs). Expression of pluripotency factors and in vitro differentiation into cell types of the three germ layers was unaffected in the analyzed clone PMM2-iPSC-C3 compared with nondiseased human pluripotent stem cells (hPSCs), revealing no broader influence of the PMM2 mutation on pluripotency in cell culture. Analysis of gene expression by deep-sequencing did not show obvious differences in the transcriptome between PMM2-iPSC-C3 and nondiseased hPSCs. By multiplexed capillary gel electrophoresis coupled to laser induced fluorescence detection (xCGE-LIF) we could show that PMM2-iPSC-C3 exhibit the common hPSC N-glycosylation pattern with high-mannose-type N-glycans as the predominant species. However, phosphomannomutase activity of PMM2-iPSC-C3 was 27% compared with control hPSCs and lectin staining revealed an overall reduced protein glycosylation. In addition, quantitative assessment of N-glycosylation by xCGE-LIF showed an up to 40% reduction of high-mannose-type N-glycans in PMM2-iPSC-C3, which was in concordance to the observed reduction of the Glc3Man9GlcNAc2 lipid-linked oligosaccharide compared with control hPSCs. Thus we could model the PMM2-CDG disease phenotype of hypoglycosylation with patient derived iPSCs in vitro. Knock-down of PMM2 by shRNA in PMM2-iPSC-C3 led to a residual activity of 5% and to a further reduction of the level of N-glycosylation. Taken together we have developed human stem cell-based cell culture models with stepwise reduced levels of N-glycosylation now enabling to study the role of N-glycosylation during early human development.
Collapse
Affiliation(s)
- Christina T Thiesler
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Samanta Cajic
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Dirk Hoffmann
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Thiel
- **Center for Child and Adolescent Medicine, Department Kinderheilkunde I, 69120 Heidelberg, Germany
| | - Laura van Diepen
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - René Hennig
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany; §§glyXera GmbH, 39120 Magdeburg, Germany
| | - Malte Sgodda
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ¶¶Translational Hepatology and Stem Cell Biology, Dept. of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Weiβmann
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - Udo Reichl
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Doris Steinemann
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖‖Institute of Human Genetics, Hannover Medical School, 30625 Hannover, Germany
| | - Ulf Diekmann
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Nicolas M B Huber
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Astrid Oberbeck
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Cantz
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ¶¶Translational Hepatology and Stem Cell Biology, Dept. of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas W Kuss
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - Christian Körner
- **Center for Child and Adolescent Medicine, Department Kinderheilkunde I, 69120 Heidelberg, Germany
| | - Axel Schambach
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Erdmann Rapp
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany; §§glyXera GmbH, 39120 Magdeburg, Germany
| | - Falk F R Buettner
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
14
|
Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Reports 2015; 4:282-96. [PMID: 25680479 PMCID: PMC4325194 DOI: 10.1016/j.stemcr.2015.01.005] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
Interleukin-3 (IL-3) is capable of supporting the proliferation of a broad range of hematopoietic cell types, whereas granulocyte colony-stimulating factor (G-CSF) and macrophage CSF (M-CSF) represent critical cytokines in myeloid differentiation. When this was investigated in a pluripotent-stem-cell-based hematopoietic differentiation model, IL-3/G-CSF or IL-3/M-CSF exposure resulted in the continuous generation of myeloid cells from an intermediate myeloid-cell-forming complex containing CD34+ clonogenic progenitor cells for more than 2 months. Whereas IL-3/G-CSF directed differentiation toward CD45+CD11b+CD15+CD16+CD66b+ granulocytic cells of various differentiation stages up to a segmented morphology displaying the capacity of cytokine-directed migration, respiratory burst response, and neutrophil-extracellular-trap formation, exposure to IL-3/M-CSF resulted in CD45+CD11b+CD14+CD163+CD68+ monocyte/macrophage-type cells capable of phagocytosis and cytokine secretion. Hence, we show here that myeloid specification of human pluripotent stem cells by IL-3/G-CSF or IL-3/M-CSF allows for prolonged and large-scale production of myeloid cells, and thus is suited for cell-fate and disease-modeling studies as well as gene- and cell-therapy applications. Myeloid specification of human PSCs by IL-3-/M-CSF, G-CSF, or GM-CSF Large-scale and continuous generation of M2-MΦ or granulocytes by M-CSF or G-CSF Functional iPSC-derived macrophages or granulocytes similar to in-vivo-derived cells
Collapse
|
15
|
Prel A, Caval V, Gayon R, Ravassard P, Duthoit C, Payen E, Maouche-Chretien L, Creneguy A, Nguyen TH, Martin N, Piver E, Sevrain R, Lamouroux L, Leboulch P, Deschaseaux F, Bouillé P, Sensébé L, Pagès JC. Highly efficient in vitro and in vivo delivery of functional RNAs using new versatile MS2-chimeric retrovirus-like particles. Mol Ther Methods Clin Dev 2015; 2:15039. [PMID: 26528487 PMCID: PMC4613645 DOI: 10.1038/mtm.2015.39] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 12/25/2022]
Abstract
RNA delivery is an attractive strategy to achieve transient gene expression in research projects and in cell- or gene-based therapies. Despite significant efforts investigating vector-directed RNA transfer, there is still a requirement for better efficiency of delivery to primary cells and in vivo. Retroviral platforms drive RNA delivery, yet retrovirus RNA-packaging constraints limit gene transfer to two genome-molecules per viral particle. To improve retroviral transfer, we designed a dimerization-independent MS2-driven RNA packaging system using MS2-Coat-retrovirus chimeras. The engineered chimeric particles promoted effective packaging of several types of RNAs and enabled efficient transfer of biologically active RNAs in various cell types, including human CD34(+) and iPS cells. Systemic injection of high-titer particles led to gene expression in mouse liver and transferring Cre-recombinase mRNA in muscle permitted widespread editing at the ROSA26 locus. We could further show that the VLPs were able to activate an osteoblast differentiation pathway by delivering RUNX2- or DLX5-mRNA into primary human bone-marrow mesenchymal-stem cells. Thus, the novel chimeric MS2-lentiviral particles are a versatile tool for a wide range of applications including cellular-programming or genome-editing.
Collapse
Affiliation(s)
- Anne Prel
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- UMR UPS/CNRS 5273, EFS-PM, INSERM U1031, Toulouse, France
| | - Vincent Caval
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
| | - Régis Gayon
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Philippe Ravassard
- Institut du Cerveau et de la Moelle (ICM), Université Pierre et Marie Curie, CNRS UMR7225; INSERM U1127, Biotechnologies and Biothérapies Team, Paris, France
| | - Christine Duthoit
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Emmanuel Payen
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | - Leila Maouche-Chretien
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | - Alison Creneguy
- INSERM UMRS 1064, Centre Hospitalier Universitaire (CHU) Hôtel Dieu, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), Université de Nantes, Nantes, France
| | - Tuan Huy Nguyen
- INSERM UMRS 1064, Centre Hospitalier Universitaire (CHU) Hôtel Dieu, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), Université de Nantes, Nantes, France
| | - Nicolas Martin
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Eric Piver
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- CHRU de Tours, Laboratoire de biochimie et biologie moléculaire, Tours, France
| | - Raphaël Sevrain
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Lucille Lamouroux
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Philippe Leboulch
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | | | - Pascale Bouillé
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Luc Sensébé
- UMR UPS/CNRS 5273, EFS-PM, INSERM U1031, Toulouse, France
| | - Jean-Christophe Pagès
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- CHRU de Tours, Laboratoire de biochimie et biologie moléculaire, Tours, France
| |
Collapse
|
16
|
Retrovirus-based vectors for transient and permanent cell modification. Curr Opin Pharmacol 2015; 24:135-46. [PMID: 26433198 DOI: 10.1016/j.coph.2015.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/04/2015] [Indexed: 01/19/2023]
Abstract
Retroviral vectors are commonly employed for long-term transgene expression via integrating vector technology. However, three alternative retrovirus-based platforms are currently available that allow transient cell modification. Gene expression can be mediated from either episomal DNA or RNA templates, or selected proteins can be directly transferred through retroviral nanoparticles. The different technologies are functionally graded with respect to safety, expression magnitude and expression duration. Improvement of the initial technologies, including modification of vector designs, targeted increase in expression strength and duration as well as improved safety characteristics, has allowed maturation of retroviral systems into efficient and promising tools that meet the technological demands of a wide variety of potential application areas.
Collapse
|
17
|
Gledhill K, Guo Z, Umegaki-Arao N, Higgins CA, Itoh M, Christiano AM. Melanin Transfer in Human 3D Skin Equivalents Generated Exclusively from Induced Pluripotent Stem Cells. PLoS One 2015; 10:e0136713. [PMID: 26308443 PMCID: PMC4550351 DOI: 10.1371/journal.pone.0136713] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 08/07/2015] [Indexed: 01/06/2023] Open
Abstract
The current utility of 3D skin equivalents is limited by the fact that existing models fail to recapitulate the cellular complexity of human skin. They often contain few cell types and no appendages, in part because many cells found in the skin are difficult to isolate from intact tissue and cannot be expanded in culture. Induced pluripotent stem cells (iPSCs) present an avenue by which we can overcome this issue due to their ability to be differentiated into multiple cell types in the body and their unlimited growth potential. We previously reported generation of the first human 3D skin equivalents from iPSC-derived fibroblasts and iPSC-derived keratinocytes, demonstrating that iPSCs can provide a foundation for modeling a complex human organ such as skin. Here, we have increased the complexity of this model by including additional iPSC-derived melanocytes. Epidermal melanocytes, which are largely responsible for skin pigmentation, represent the second most numerous cell type found in normal human epidermis and as such represent a logical next addition. We report efficient melanin production from iPSC-derived melanocytes and transfer within an entirely iPSC-derived epidermal-melanin unit and generation of the first functional human 3D skin equivalents made from iPSC-derived fibroblasts, keratinocytes and melanocytes.
Collapse
Affiliation(s)
- Karl Gledhill
- Department of Dermatology, Columbia University, New York, NY, United States of America
| | - Zongyou Guo
- Department of Dermatology, Columbia University, New York, NY, United States of America
| | - Noriko Umegaki-Arao
- Department of Dermatology, Columbia University, New York, NY, United States of America
| | - Claire A. Higgins
- Department of Dermatology, Columbia University, New York, NY, United States of America
| | - Munenari Itoh
- Department of Dermatology, Columbia University, New York, NY, United States of America
| | - Angela M. Christiano
- Department of Dermatology, Columbia University, New York, NY, United States of America
- Department of Genetics and Development, Columbia University, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
18
|
Tosato V, Bruschi CV. Per aspera ad astra: When harmful chromosomal translocations become a plus value in genetic evolution. Lessons from Saccharomyces cerevisiae. ACTA ACUST UNITED AC 2015; 2:363-375. [PMID: 28357264 PMCID: PMC5354581 DOI: 10.15698/mic2015.10.230] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this review we will focus on chromosomal translocations (either spontaneous or induced) in budding yeast. Indeed, very few organisms tolerate so well aneuploidy like Saccharomyces, allowing in depth studies on chromosomal numerical aberrations. Many wild type strains naturally develop chromosomal rearrangements while adapting to different environmental conditions. Translocations, in particular, are valuable not only because they naturally drive species evolution, but because they might allow the artificial generation of new strains that can be optimized for industrial purposes. In this area, several methodologies to artificially trigger chromosomal translocations have been conceived in the past years, such as the chromosomal fragmentation vector (CFV) technique, the Cre-loxP procedure, the FLP/FRT recombination method and, recently, the bridge - induced translocation (BIT) system. An overview of the methodologies to generate chromosomal translocations in yeast will be presented and discussed considering advantages and drawbacks of each technology, focusing in particular on the recent BIT system. Translocants are important for clinical studies because translocated yeast cells resemble cancer cells from morphological and physiological points of view and because the translocation event ensues in a transcriptional de-regulation with a subsequent multi-factorial genetic adaptation to new, selective environmental conditions. The phenomenon of post-translocational adaptation (PTA) is discussed, providing some new unpublished data and proposing the hypothesis that translocations may drive evolution through adaptive genetic selection.
Collapse
Affiliation(s)
- Valentina Tosato
- Yeast Molecular Genetics Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Carlo V Bruschi
- Yeast Molecular Genetics Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
19
|
Rahman SH, Kuehle J, Reimann C, Mlambo T, Alzubi J, Maeder ML, Riedel H, Fisch P, Cantz T, Rudolph C, Mussolino C, Joung JK, Schambach A, Cathomen T. Rescue of DNA-PK Signaling and T-Cell Differentiation by Targeted Genome Editing in a prkdc Deficient iPSC Disease Model. PLoS Genet 2015; 11:e1005239. [PMID: 26000857 PMCID: PMC4441453 DOI: 10.1371/journal.pgen.1005239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 04/26/2015] [Indexed: 12/22/2022] Open
Abstract
In vitro disease modeling based on induced pluripotent stem cells (iPSCs) provides a powerful system to study cellular pathophysiology, especially in combination with targeted genome editing and protocols to differentiate iPSCs into affected cell types. In this study, we established zinc-finger nuclease-mediated genome editing in primary fibroblasts and iPSCs generated from a mouse model for radiosensitive severe combined immunodeficiency (RS-SCID), a rare disorder characterized by cellular sensitivity to radiation and the absence of lymphocytes due to impaired DNA-dependent protein kinase (DNA-PK) activity. Our results demonstrate that gene editing in RS-SCID fibroblasts rescued DNA-PK dependent signaling to overcome radiosensitivity. Furthermore, in vitro T-cell differentiation from iPSCs was employed to model the stage-specific T-cell maturation block induced by the disease causing mutation. Genetic correction of the RS-SCID iPSCs restored T-lymphocyte maturation, polyclonal V(D)J recombination of the T-cell receptor followed by successful beta-selection. In conclusion, we provide proof that iPSC-based in vitro T-cell differentiation is a valuable paradigm for SCID disease modeling, which can be utilized to investigate disorders of T-cell development and to validate gene therapy strategies for T-cell deficiencies. Moreover, this study emphasizes the significance of designer nucleases as a tool for generating isogenic disease models and their future role in producing autologous, genetically corrected transplants for various clinical applications. Due to the limited availability and lifespan of some primary cells, in vitro disease modeling with induced pluripotent stem cells (iPSCs) offers a valuable complementation to in vivo studies. The goal of our study was to establish an in vitro disease model for severe combined immunodeficiency (SCID), a group of inherited disorders of the immune system characterized by the lack of T-lymphocytes. To this end, we generated iPSCs from fibroblasts of a radiosensitive SCID (RS-SCID) mouse model and established a protocol to recapitulate T-lymphopoiesis from iPSCs in vitro. We used designer nucleases to edit the underlying mutation in prkdc, the gene encoding DNA-PKcs, and demonstrated that genetic correction of the disease locus rescued DNA-PK dependent signaling, restored normal radiosensitivity, and enabled T-cell maturation and polyclonal T-cell receptor recombination. We hence provide proof that the combination of two promising technology platforms, iPSCs and designer nucleases, with a protocol to generate T-cells in vitro, represents a powerful paradigm for SCID disease modeling and the evaluation of therapeutic gene editing strategies. Furthermore, our system provides a basis for further development of iPSC-derived cell products with the potential for various clinical applications, including infusions of in vitro derived autologous T-cells to stabilize patients after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Shamim H. Rahman
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Johannes Kuehle
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Christian Reimann
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Tafadzwa Mlambo
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Jamal Alzubi
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Morgan L. Maeder
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Heimo Riedel
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Biochemistry and Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| | - Paul Fisch
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Tobias Cantz
- Translational Hepatology and Stem Cell Biology, REBIRTH cluster of excellence, Hannover Medical School, Hannover, Germany
| | - Cornelia Rudolph
- Institute for Cellular and Molecular Pathology, Hannover Medical School, Hannover, Germany
| | - Claudio Mussolino
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - J. Keith Joung
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- * E-mail: (AS); (TC)
| | - Toni Cathomen
- Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- * E-mail: (AS); (TC)
| |
Collapse
|
20
|
Accurate comparison of antibody expression levels by reproducible transgene targeting in engineered recombination-competent CHO cells. Appl Microbiol Biotechnol 2014; 98:9723-33. [PMID: 25158835 PMCID: PMC4231286 DOI: 10.1007/s00253-014-6011-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/30/2014] [Accepted: 08/01/2014] [Indexed: 11/23/2022]
Abstract
Over the years, Chinese hamster ovary (CHO) cells have emerged as the major host for expressing biotherapeutic proteins. Traditional methods to generate high-producer cell lines rely on random integration(s) of the gene of interest but have thereby left the identification of bottlenecks as a challenging task. For comparison of different producer cell lines derived from various transfections, a system that provides control over transgene expression behavior is highly needed. This motivated us to develop a novel “DUKX-B11 F3/F” cell line to target different single-chain antibody fragments into the same chromosomal target site by recombinase-mediated cassette exchange (RMCE) using the flippase (FLP)/FLP recognition target (FRT) system. The RMCE-competent cell line contains a gfp reporter fused to a positive/negative selection system flanked by heterospecific FRT (F) variants under control of an external CMV promoter, constructed as “promoter trap”. The expression stability and FLP accessibility of the tagged locus was demonstrated by successive rounds of RMCE. As a proof of concept, we performed RMCE using cassettes encoding two different anti-HIV single-chain Fc fragments, 3D6scFv-Fc and 2F5scFv-Fc. Both targeted integrations yielded homogenous cell populations with comparable intracellular product contents and messenger RNA (mRNA) levels but product related differences in specific productivities. These studies confirm the potential of the newly available “DUKX-B11 F3/F” cell line to guide different transgenes into identical transcriptional control regions by RMCE and thereby generate clones with comparable amounts of transgene mRNA. This new host is a prerequisite for cell biology studies of independent transfections and transgenes.
Collapse
|
21
|
Turan S, Qiao J, Madden S, Benham C, Kotz M, Schambach A, Bode J. Expanding Flp-RMCE options: the potential of Recombinase Mediated Twin-Site Targeting (RMTT). Gene 2014; 546:135-44. [DOI: 10.1016/j.gene.2014.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/02/2014] [Indexed: 01/02/2023]
|