1
|
Nemirov K, Authié P, Souque P, Moncoq F, Noirat A, Blanc C, Bourgine M, Majlessi L, Charneau P. Preclinical proof of concept of a tetravalent lentiviral T-cell vaccine against dengue viruses. Front Immunol 2023; 14:1208041. [PMID: 37654495 PMCID: PMC10466046 DOI: 10.3389/fimmu.2023.1208041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/17/2023] [Indexed: 09/02/2023] Open
Abstract
Dengue virus (DENV) is responsible for approximately 100 million cases of dengue fever annually, including severe forms such as hemorrhagic dengue and dengue shock syndrome. Despite intensive vaccine research and development spanning several decades, a universally accepted and approved vaccine against dengue fever has not yet been developed. The major challenge associated with the development of such a vaccine is that it should induce simultaneous and equal protection against the four DENV serotypes, because past infection with one serotype may greatly increase the severity of secondary infection with a distinct serotype, a phenomenon known as antibody-dependent enhancement (ADE). Using a lentiviral vector platform that is particularly suitable for the induction of cellular immune responses, we designed a tetravalent T-cell vaccine candidate against DENV ("LV-DEN"). This vaccine candidate has a strong CD8+ T-cell immunogenicity against the targeted non-structural DENV proteins, without inducing antibody response against surface antigens. Evaluation of its protective potential in the preclinical flavivirus infection model, i.e., mice knockout for the receptor to the type I IFN, demonstrated its significant protective effect against four distinct DENV serotypes, based on reduced weight loss, viremia, and viral loads in peripheral organs of the challenged mice. These results provide proof of concept for the use of lentiviral vectors for the development of efficient polyvalent T-cell vaccine candidates against all DENV serotypes.
Collapse
Affiliation(s)
- Kirill Nemirov
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Baron Y, Sens J, Lange L, Nassauer L, Klatt D, Hoffmann D, Kleppa MJ, Barbosa PV, Keisker M, Steinberg V, Suerth JD, Vondran FW, Meyer J, Morgan M, Schambach A, Galla M. Improved alpharetrovirus-based Gag.MS2 particles for efficient and transient delivery of CRISPR-Cas9 into target cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:810-823. [PMID: 35141043 PMCID: PMC8801357 DOI: 10.1016/j.omtn.2021.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/29/2021] [Indexed: 12/12/2022]
Abstract
DNA-modifying technologies, such as the CRISPR-Cas9 system, are promising tools in the field of gene and cell therapies. However, high and prolonged expression of DNA-modifying enzymes may cause cytotoxic and genotoxic side effects and is therefore unwanted in therapeutic approaches. Consequently, development of new and potent short-term delivery methods is of utmost importance. Recently, we developed non-integrating gammaretrovirus- and MS2 bacteriophage-based Gag.MS2 (g.Gag.MS2) particles for transient transfer of non-retroviral CRISPR-Cas9 RNA into target cells. In the present study, we further improved the technique by transferring the system to the alpharetroviral vector platform (a.Gag.MS2), which significantly increased CRISPR-Cas9 delivery into target cells and allowed efficient targeted knockout of endogenous TP53/Trp53 genes in primary murine fibroblasts as well as primary human fibroblasts, hepatocytes, and cord-blood-derived CD34+ stem and progenitor cells. Strikingly, co-packaging of Cas9 mRNA and multiple single guide RNAs (sgRNAs) into a.Gag.MS2 chimera displayed efficient targeted knockout of up to three genes. Co-transfection of single-stranded DNA donor oligonucleotides during CRISPR-Cas9 particle production generated all-in-one particles, which mediated up to 12.5% of homology-directed repair in primary cell cultures. In summary, optimized a.Gag.MS2 particles represent a versatile tool for short-term delivery of DNA-modifying enzymes into a variety of target cells, including primary murine and human cells.
Collapse
Affiliation(s)
- Yvonne Baron
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Johanna Sens
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Lucas Lange
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Larissa Nassauer
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Denise Klatt
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Dirk Hoffmann
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Marc-Jens Kleppa
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Philippe Vollmer Barbosa
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover 30625, Germany
| | - Maximilian Keisker
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Viviane Steinberg
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Julia D. Suerth
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Florian W.R. Vondran
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover 30625, Germany
- German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover Medical School, Hannover 30625, Germany
| | - Johann Meyer
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
3
|
Du M, Li Y, Chen Z. Sonoporation-Mediated Gene Transfection: A Novel Direction for Cell Reprogramming In Vivo. Front Bioeng Biotechnol 2022; 9:803055. [PMID: 35174147 PMCID: PMC8841490 DOI: 10.3389/fbioe.2021.803055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/09/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Li
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Zhiyi Chen,
| |
Collapse
|
4
|
Sii-Felice K, Castillo Padilla J, Relouzat F, Cheuzeville J, Tantawet S, Maouche L, Le Grand R, Leboulch P, Payen E. Enhanced Transduction of Macaca fascicularis Hematopoietic Cells with Chimeric Lentiviral Vectors. Hum Gene Ther 2019; 30:1306-1323. [DOI: 10.1089/hum.2018.179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Karine Sii-Felice
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Javier Castillo Padilla
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Francis Relouzat
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Joëlle Cheuzeville
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- bluebird bio France, Fontenay aux Roses, France
| | - Siriporn Tantawet
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Leïla Maouche
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- INSERM, Paris, France
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, UMR 1184, IDMIT Department, Institute of Biology François Jacob, INSERM, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
| | - Philippe Leboulch
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- Ramathibodi Hospital and Mahidol University, Bangkok, Thailand
- Harvard Medical School and Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston Massachusetts
| | - Emmanuel Payen
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Paris-Sud University, Paris-Saclay University, Fontenay aux Roses, France
- INSERM, Paris, France
| |
Collapse
|
5
|
Bax M, McKenna J, Do-Ha D, Stevens CH, Higginbottom S, Balez R, Cabral-da-Silva MEC, Farrawell NE, Engel M, Poronnik P, Yerbury JJ, Saunders DN, Ooi L. The Ubiquitin Proteasome System Is a Key Regulator of Pluripotent Stem Cell Survival and Motor Neuron Differentiation. Cells 2019; 8:cells8060581. [PMID: 31200561 PMCID: PMC6627164 DOI: 10.3390/cells8060581] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The ubiquitin proteasome system (UPS) plays an important role in regulating numerous cellular processes, and a dysfunctional UPS is thought to contribute to motor neuron disease. Consequently, we sought to map the changing ubiquitome in human iPSCs during their pluripotent stage and following differentiation to motor neurons. Ubiquitinomics analysis identified that spliceosomal and ribosomal proteins were more ubiquitylated in pluripotent stem cells, whilst proteins involved in fatty acid metabolism and the cytoskeleton were specifically ubiquitylated in the motor neurons. The UPS regulator, ubiquitin-like modifier activating enzyme 1 (UBA1), was increased 36-fold in the ubiquitome of motor neurons compared to pluripotent stem cells. Thus, we further investigated the functional consequences of inhibiting the UPS and UBA1 on motor neurons. The proteasome inhibitor MG132, or the UBA1-specific inhibitor PYR41, significantly decreased the viability of motor neurons. Consistent with a role of the UPS in maintaining the cytoskeleton and regulating motor neuron differentiation, UBA1 inhibition also reduced neurite length. Pluripotent stem cells were extremely sensitive to MG132, showing toxicity at nanomolar concentrations. The motor neurons were more resilient to MG132 than pluripotent stem cells but demonstrated higher sensitivity than fibroblasts. Together, this data highlights the important regulatory role of the UPS in pluripotent stem cell survival and motor neuron differentiation.
Collapse
Affiliation(s)
- Monique Bax
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Jessie McKenna
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Dzung Do-Ha
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Claire H Stevens
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Sarah Higginbottom
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Mauricio E Castro Cabral-da-Silva
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Natalie E Farrawell
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Martin Engel
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Philip Poronnik
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia.
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia.
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| |
Collapse
|
6
|
Petrillo C, Thorne LG, Unali G, Schiroli G, Giordano AMS, Piras F, Cuccovillo I, Petit SJ, Ahsan F, Noursadeghi M, Clare S, Genovese P, Gentner B, Naldini L, Towers GJ, Kajaste-Rudnitski A. Cyclosporine H Overcomes Innate Immune Restrictions to Improve Lentiviral Transduction and Gene Editing In Human Hematopoietic Stem Cells. Cell Stem Cell 2018; 23:820-832.e9. [PMID: 30416070 PMCID: PMC6292841 DOI: 10.1016/j.stem.2018.10.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/09/2018] [Accepted: 10/02/2018] [Indexed: 01/01/2023]
Abstract
Innate immune factors may restrict hematopoietic stem cell (HSC) genetic engineering and contribute to broad individual variability in gene therapy outcomes. Here, we show that HSCs harbor an early, constitutively active innate immune block to lentiviral transduction that can be efficiently overcome by cyclosporine H (CsH). CsH potently enhances gene transfer and editing in human long-term repopulating HSCs by inhibiting interferon-induced transmembrane protein 3 (IFITM3), which potently restricts VSV glycoprotein-mediated vector entry. Importantly, individual variability in endogenous IFITM3 levels correlated with permissiveness of HSCs to lentiviral transduction, suggesting that CsH treatment will be useful for improving ex vivo gene therapy and standardizing HSC transduction across patients. Overall, our work unravels the involvement of innate pathogen recognition molecules in immune blocks to gene correction in primary human HSCs and highlights how these roadblocks can be overcome to develop innovative cell and gene therapies.
Collapse
Affiliation(s)
- Carolina Petrillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, MI 20132, Italy
| | - Lucy G Thorne
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Giulia Unali
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, MI 20132, Italy
| | - Giulia Schiroli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy
| | - Anna M S Giordano
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, MI 20132, Italy
| | - Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, MI 20132, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy
| | - Sarah J Petit
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Fatima Ahsan
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Pietro Genovese
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, MI 20132, Italy
| | - Greg J Towers
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, MI 20132, Italy.
| |
Collapse
|
7
|
Cullmann K, Blokland KEC, Sebe A, Schenk F, Ivics Z, Heinz N, Modlich U. Sustained and regulated gene expression by Tet-inducible "all-in-one" retroviral vectors containing the HNRPA2B1-CBX3 UCOE ®. Biomaterials 2018; 192:486-499. [PMID: 30508767 DOI: 10.1016/j.biomaterials.2018.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022]
Abstract
Genetic modification of induced pluripotent stem (iPS) cells may be necessary for the generation of effector cells for cellular therapies. Hereby, it can be important to induce transgene expression at restricted and defined time windows, especially if it interferes with pluripotency or differentiation. To achieve this, inducible expression systems can be used such as the tetracycline-inducible retroviral vector system, however, retroviral expression can be subjected to epigenetic silencing or to position-effect variegation. One strategy to overcome this is the incorporation of ubiquitous chromatin opening elements (UCOE®'s) into retroviral vectors to maintain a transcriptionally permissive chromatin state at the integration site. In this study, we developed Tet-inducible all-in-one gammaretroviral vectors carrying different sized UCOE®'s derived from the A2UCOE. The ability to prevent vector silencing by preserving the Tet-regulatory potential was investigated in different cell lines, and in murine and human iPS cells. A 670-bp fragment spanning the CBX3 promoter region of A2UCOE (U670) was the most potent element in preventing silencing, and conferred the strongest expression from the vector in the induced state. While longer fragments of A2UCOEs also sustained expression, vector titers and induction efficiencies were impaired. Finally, we demonstrate that U670 can be used for constitutive expression of the transactivator in the all-in-one vector for faithful regulation of transgenes by doxycycline, including the thrombopoietin receptor Mpl conferring cytokine-dependent cell growth.
Collapse
Affiliation(s)
- Katharina Cullmann
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Kaj E C Blokland
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Attila Sebe
- Div. of Medical Biotechnology, Paul-Ehrlich-Institute, Langen, Germany
| | - Franziska Schenk
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Zoltán Ivics
- Div. of Medical Biotechnology, Paul-Ehrlich-Institute, Langen, Germany
| | - Niels Heinz
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany; BioNTech Innovative Manufacturing Services GmbH, Idar-Oberstein, Germany
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany.
| |
Collapse
|
8
|
Parekh U, Wu Y, Zhao D, Worlikar A, Shah N, Zhang K, Mali P. Mapping Cellular Reprogramming via Pooled Overexpression Screens with Paired Fitness and Single-Cell RNA-Sequencing Readout. Cell Syst 2018; 7:548-555.e8. [PMID: 30448000 DOI: 10.1016/j.cels.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
Abstract
Understanding the effects of genetic perturbations on the cellular state has been challenging using traditional pooled screens, which typically rely on the delivery of a single perturbation per cell and unidimensional phenotypic readouts. Here, we use barcoded open reading frame overexpression libraries coupled with single-cell RNA sequencing to assay cell state and fitness, a technique we call SEUSS (scalable functional screening by sequencing). Using SEUSS, we perturbed hPSCs with a library of developmentally critical transcription factors (TFs) and assayed the impact of TF overexpression on fitness and transcriptomic states. We further leveraged the versatility of the ORF library approach to assay mutant genes and whole gene families. From the transcriptomic responses, we built genetic co-regulatory networks to identify altered gene modules and found that KLF4 and SNAI2 drive opposing effects along the epithelial-mesenchymal transition axis. From the fitness responses, we identified ETV2 as a driver of reprogramming toward an endothelial-like state.
Collapse
Affiliation(s)
- Udit Parekh
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Yan Wu
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Dongxin Zhao
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Atharv Worlikar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Neha Shah
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA
| | - Kun Zhang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Hammerschmidt SI, Werth K, Rothe M, Galla M, Permanyer M, Patzer GE, Bubke A, Frenk DN, Selich A, Lange L, Schambach A, Bošnjak B, Förster R. CRISPR/Cas9 Immunoengineering of Hoxb8-Immortalized Progenitor Cells for Revealing CCR7-Mediated Dendritic Cell Signaling and Migration Mechanisms in vivo. Front Immunol 2018; 9:1949. [PMID: 30210501 PMCID: PMC6120996 DOI: 10.3389/fimmu.2018.01949] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
Abstract
To present antigens to cognate T cells, dendritic cells (DCs) exploit the chemokine receptor CCR7 to travel from peripheral tissue via afferent lymphatic vessels to directly enter draining lymph nodes through the floor of the subcapsular sinus. Here, we combined unlimited proliferative capacity of conditionally Hoxb8-immortalized hematopoietic progenitor cells with CRISPR/Cas9 technology to create a powerful experimental system to investigate DC migration and function. Hematopoietic progenitor cells from the bone marrow of Cas9-transgenic mice were conditionally immortalized by lentiviral transduction introducing a doxycycline-regulated form of the transcription factor Hoxb8 (Cas9-Hoxb8 cells). These cells could be stably cultured for weeks in the presence of doxycycline and puromycin, allowing us to introduce additional genetic modifications applying CRISPR/Cas9 technology. Importantly, modified Cas9-Hoxb8 cells retained their potential to differentiate in vitro into myeloid cells, and GM-CSF-differentiated Cas9-Hoxb8 cells showed the classical phenotype of GM-CSF-differentiated bone marrow-derived dendritic cells. Following intralymphatic delivery Cas9-Hoxb8 DCs entered the lymph node in a CCR7-dependent manner. Finally, we used two-photon microscopy and imaged Cas9-Hoxb8 DCs that expressed the genetic Ca2+ sensor GCaMP6S to visualize in real-time chemokine-induced Ca2+ signaling of lymph-derived DCs entering the LN parenchyma. Altogether, our study not only allows mechanistic insights in DC migration in vivo, but also provides a platform for the immunoengineering of DCs that, in combination with two-photon imaging, can be exploited to further dissect DC dynamics in vivo.
Collapse
Affiliation(s)
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Anja Bubke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - David N. Frenk
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anton Selich
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Lucas Lange
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Geis FK, Galla M, Hoffmann D, Kuehle J, Zychlinski D, Maetzig T, Schott JW, Schwarzer A, Goffinet C, Goff SP, Schambach A. Correction to: Potent and reversible lentiviral vector restriction in murine induced pluripotent stem cells. Retrovirology 2017; 14:48. [PMID: 29047401 PMCID: PMC5648417 DOI: 10.1186/s12977-017-0372-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
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
|