1
|
Siow KM, Güngör M, Wrona D, Raimondi F, Pastukhov O, Tsapogas P, Menzi T, Schmitz M, Kulcsár PI, Schwank G, Schulz A, Jinek M, Modlich U, Siler U, Reichenbach J. Targeted knock-in of NCF1 cDNA into the NCF2 locus leads to myeloid phenotypic correction of p47 phox -deficient chronic granulomatous disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102229. [PMID: 38952440 PMCID: PMC11215332 DOI: 10.1016/j.omtn.2024.102229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 05/22/2024] [Indexed: 07/03/2024]
Abstract
p47 phox -deficient chronic granulomatous disease (p47-CGD) is a primary immunodeficiency caused by mutations in the neutrophil cytosolic factor 1 (NCF1) gene, resulting in defective NADPH oxidase function in phagocytes. Due to its complex genomic context, the NCF1 locus is not suited for safe gene editing with current genome editing technologies. Therefore, we developed a targeted NCF1 coding sequence knock-in by CRISPR-Cas9 ribonucleoprotein and viral vector template delivery, to restore p47 phox expression under the control of the endogenous NCF2 locus. NCF2 encodes for p67 phox , an NADPH oxidase subunit that closely interacts with p47 phox and is predominantly expressed in myeloid cells. This approach restored p47 phox expression and NADPH oxidase function in p47-CGD patient hematopoietic stem and progenitor cells (HSPCs) and in p47 phox -deficient mouse HSPCs, with the transgene expression following a myeloid differentiation pattern. Adeno-associated viral vectors performed favorably over integration-deficient lentiviral vectors for template delivery, with fewer off-target integrations and higher correction efficacy in HSPCs. Such myeloid-directed gene editing is promising for clinical CGD gene therapy, as it leads to the co-expression of p47 phox and p67 phox , ensuring spatiotemporal and near-physiological transgene expression in myeloid cells.
Collapse
Affiliation(s)
- Kah Mun Siow
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Merve Güngör
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Dominik Wrona
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Federica Raimondi
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Oleksandr Pastukhov
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Panagiotis Tsapogas
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Timon Menzi
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Michael Schmitz
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Péter István Kulcsár
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, 89075 Ulm, Germany
| | - Martin Jinek
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Ute Modlich
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
| | - Ulrich Siler
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
- School of Life Sciences, Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, 4132 Muttenz, Switzerland
| | - Janine Reichenbach
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952 Zurich, Switzerland
- Department of Somatic Gene Therapy, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
2
|
Zhao JZ, Li QY, Lin JJ, Yang LY, Du MY, Wang Y, Liu KX, Jiang ZA, Li HH, Wang SF, Sun B, Mu SQ, Li B, Liu K, Gong M, Sun SG. Integrated analysis of tRNA-derived small RNAs in proliferative human aortic smooth muscle cells. Cell Mol Biol Lett 2022; 27:47. [PMID: 35705912 PMCID: PMC9199163 DOI: 10.1186/s11658-022-00346-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
Background Abnormal proliferation of vascular smooth muscle cells (VSMCs) contributes to vascular remodeling diseases. Recently, it has been discovered that tRNA-derived small RNAs (tsRNAs), a new type of noncoding RNAs, are related to the proliferation and migration of VSMCs. tsRNAs regulate target gene expression through miRNA-like functions. This study aims to explore the potential of tsRNAs in human aortic smooth muscle cell (HASMC) proliferation. Methods High-throughput sequencing was performed to analyze the tsRNA expression profile of proliferative and quiescent HASMCs. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to validate the sequence results and subcellular distribution of AS-tDR-001370, AS-tDR-000067, AS-tDR-009512, and AS-tDR-000076. Based on the microRNA-like functions of tsRNAs, we predicted target promoters and mRNAs and constructed tsRNA–promoter and tsRNA–mRNA interaction networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to reveal the function of target genes. EdU incorporation assay, Western blot, and dual-luciferase reporter gene assay were utilized to detect the effects of tsRNAs on HASMC proliferation. Results Compared with quiescent HASMCs, there were 1838 differentially expressed tsRNAs in proliferative HASMCs, including 887 with increased expression (fold change > 2, p < 0.05) and 951 with decreased expression (fold change < ½, p < 0.05). AS-tDR-001370, AS-tDR-000067, AS-tDR-009512, and AS-tDR-000076 were increased in proliferative HASMCs and were mainly located in the nucleus. Bioinformatics analysis suggested that the four tsRNAs involved a variety of GO terms and pathways related to VSMC proliferation. AS-tDR-000067 promoted HASMC proliferation by suppressing p53 transcription in a promoter-targeted manner. AS-tDR-000076 accelerated HASMC proliferation by attenuating mitofusin 2 (MFN2) levels in a 3′-untranslated region (UTR)-targeted manner. Conclusions During HASMC proliferation, the expression levels of many tsRNAs are altered. AS-tDR-000067 and AS-tDR-000076 act as new factors promoting VSMC proliferation. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00346-4.
Collapse
Affiliation(s)
- Jian-Zhi Zhao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China.,Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi-Yao Li
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jia-Jie Lin
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Li-Yun Yang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Mei-Yang Du
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yu Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Ke-Xin Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Ze-An Jiang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Huan-Huan Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Si-Fan Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Bo Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Shi-Qing Mu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Bin Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Kun Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Miao Gong
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Shao-Guang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
3
|
Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies. J Immunol Res 2018; 2018:4054815. [PMID: 30306093 PMCID: PMC6166361 DOI: 10.1155/2018/4054815] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are powerful immune effectors whose antitumor activity is regulated through a sophisticated network of activating and inhibitory receptors. As effectors of cancer immunotherapy, NK cells are attractive as they do not attack healthy self-tissues nor do they induce T cell-driven inflammatory cytokine storm, enabling their use as allogeneic adoptive cellular therapies. Clinical responses to adoptive NK-based immunotherapy have been thwarted, however, by the profound immunosuppression induced by the tumor microenvironment, particularly severe in the context of solid tumors. In addition, the short postinfusion persistence of NK cells in vivo has limited their clinical efficacy. Enhancing the antitumor immunity of NK cells through genetic engineering has been fueled by the promise that impaired cytotoxic functionality can be restored or augmented with the use of synthetic genetic approaches. Alongside expressing chimeric antigen receptors to overcome immune escape by cancer cells, enhance their recognition, and mediate their killing, NK cells have been genetically modified to enhance their persistence in vivo by the expression of cytokines such as IL-15, avoid functional and metabolic tumor microenvironment suppression, or improve their homing ability, enabling enhanced targeting of solid tumors. However, NK cells are notoriously adverse to endogenous gene uptake, resulting in low gene uptake and transgene expression with many vector systems. Though viral vectors have achieved the highest gene transfer efficiencies with NK cells, nonviral vectors and gene transfer approaches—electroporation, lipofection, nanoparticles, and trogocytosis—are emerging. And while the use of NK cell lines has achieved improved gene transfer efficiencies particularly with viral vectors, challenges with primary NK cells remain. Here, we discuss the genetic engineering of NK cells as they relate to NK immunobiology within the context of cancer immunotherapy, highlighting the most recent breakthroughs in viral vectors and nonviral approaches aimed at genetic reprogramming of NK cells for improved adoptive immunotherapy of cancer, and, finally, address their clinical status.
Collapse
|
4
|
Montagna C, Petris G, Casini A, Maule G, Franceschini GM, Zanella I, Conti L, Arnoldi F, Burrone OR, Zentilin L, Zacchigna S, Giacca M, Cereseto A. VSV-G-Enveloped Vesicles for Traceless Delivery of CRISPR-Cas9. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:453-462. [PMID: 30195783 PMCID: PMC6041463 DOI: 10.1016/j.omtn.2018.05.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/16/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
The method of delivery of CRISPR-Cas9 into target cells is a strong determinant of efficacy and specificity in genome editing. Even though high efficiency of Cas9 delivery is necessary for optimal editing, its long-term and high levels of expression correlate with increased off-target activity. We developed vesicles (VEsiCas) carrying CRISPR-SpCas9 ribonucleoprotein complexes (RNPs) that are efficiently delivered into target cells through the fusogenic glycoprotein of the vesicular stomatitis virus (VSV-G). A crucial step for VEsiCas production is the synthesis of the single guide RNA (sgRNA) mediated by the T7 RNA polymerase in the cytoplasm of producing cells as opposed to canonical U6-driven Pol III nuclear transcription. In VEsiCas, the absence of DNA encoding SpCas9 and sgRNA allows rapid clearance of the nuclease components in target cells, which correlates with reduced genome-wide off-target cleavages. Compared with SpCas9 RNPs electroporation, which is currently the method of choice to obtain transient SpCas9 activity, VEsiCas deliver the nuclease with higher efficiency and lower toxicity. We show that a wide variety of cells can be edited through VEsiCas, including a variety of transformed cells, induced pluripotent stem cells (iPSCs), and cardiomyocytes, in vivo. VEsiCas is a traceless CRISPR-Cas9 delivery tool for efficient and safe genome editing that represents a further advancement toward the therapeutic use of the CRISPR-Cas9 technology.
Collapse
Affiliation(s)
- Claudia Montagna
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Gianluca Petris
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy.
| | - Antonio Casini
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Giulia Maule
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Gian Marco Franceschini
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Ilaria Zanella
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Luciano Conti
- Laboratory of Stem Cell Biology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy
| | - Francesca Arnoldi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Oscar R Burrone
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Serena Zacchigna
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Anna Cereseto
- Laboratory of Molecular Virology, University of Trento, Centre for Integrative Biology, 38123 Trento, Italy.
| |
Collapse
|
5
|
Darbey A, Smith LB. Deliverable transgenics & gene therapy possibilities for the testes. Mol Cell Endocrinol 2018; 468:81-94. [PMID: 29191697 DOI: 10.1016/j.mce.2017.11.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 11/30/2022]
Abstract
Male infertility and hypogonadism are clinically prevalent conditions with a high socioeconomic burden and are both linked to an increased risk in cardiovascular-metabolic diseases and earlier mortality. Therefore, there is an urgent need to better understand the causes and develop new treatments for these conditions that affect millions of men. The accelerating advancement in gene editing and delivery technologies promises improvements in both diagnosis as well as affording the opportunity to develop bespoke treatment options which would both prove beneficial for the millions of individuals afflicted with these reproductive disorders. In this review, we summarise the systems developed and utilised for the delivery of gene therapy and discuss how each of these systems could be applied for the development of a gene therapy system in the testis and how they could be of use for the future diagnosis and repair of common male reproductive disorders.
Collapse
Affiliation(s)
- Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
6
|
Hit and go CAS9 delivered through a lentiviral based self-limiting circuit. Nat Commun 2017; 8:15334. [PMID: 28530235 PMCID: PMC5458126 DOI: 10.1038/ncomms15334] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/21/2017] [Indexed: 12/26/2022] Open
Abstract
In vivo application of the CRISPR-Cas9 technology is still limited by unwanted Cas9 genomic cleavages. Long-term expression of Cas9 increases the number of genomic loci non-specifically cleaved by the nuclease. Here we develop a Self-Limiting Cas9 circuit for Enhanced Safety and specificity (SLiCES) which consists of an expression unit for Streptococcus pyogenes Cas9 (SpCas9), a self-targeting sgRNA and a second sgRNA targeting a chosen genomic locus. The self-limiting circuit results in increased genome editing specificity by controlling Cas9 levels. For its in vivo utilization, we next integrate SLiCES into a lentiviral delivery system (lentiSLiCES) via circuit inhibition to achieve viral particle production. Upon delivery into target cells, the lentiSLiCES circuit switches on to edit the intended genomic locus while simultaneously stepping up its own neutralization through SpCas9 inactivation. By preserving target cells from residual nuclease activity, our hit and go system increases safety margins for genome editing.
Collapse
|
7
|
Fornai F, Carrizzo A, Forte M, Ambrosio M, Damato A, Ferrucci M, Biagioni F, Busceti C, Puca AA, Vecchione C. The inflammatory protein Pentraxin 3 in cardiovascular disease. IMMUNITY & AGEING 2016; 13:25. [PMID: 27559355 PMCID: PMC4995820 DOI: 10.1186/s12979-016-0080-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/15/2016] [Indexed: 12/12/2022]
Abstract
The acute phase protein Pentraxin 3 (PTX3) plays a non-redundant role as a soluble pattern recognition receptor for selected pathogens and it represents a rapid biomarker for primary local activation of innate immunity and inflammation. Recent evidence indicates that PTX3 exerts an important role in modulating the cardiovascular system in humans and experimental models. In particular, there are conflicting points concerning the effects of PTX3 in cardiovascular diseases (CVD) since several observations indicate a cardiovascular protective effect of PTX3 while others speculate that the increased plasma levels of PTX3 in subjects with CVD correlate with disease severity and with poor prognosis in elderly patients. In the present review, we discuss the multifaceted effects of PTX3 on the cardiovascular system focusing on its involvement in atherosclerosis, endothelial function, hypertension, myocardial infarction and angiogenesis. This may help to explain how the specific modulation of PTX3 such as the use of different dosing, time, and target organs could help to contain different vascular diseases. These opposite actions of PTX3 will be emphasized concerning the modulation of cardiovascular system where potential therapeutic implications of PTX3 in humans are discussed.
Collapse
Affiliation(s)
- Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy ; I.R.C.C.S. Neuromed, Pozzilli, IS Italy
| | | | | | | | | | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Annibale A Puca
- Vascular Physiopathology Unit, I.R.C.C.S. Multimedica, Milan, Italy ; Department of Medicine and Surgery, University of Salerno, Via S. Allende, Baronissi, SA 84081 Italy
| | - Carmine Vecchione
- I.R.C.C.S. Neuromed, Pozzilli, IS Italy ; Department of Medicine and Surgery, University of Salerno, Via S. Allende, Baronissi, SA 84081 Italy
| |
Collapse
|
8
|
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: 42] [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
|
9
|
Generation of a stable packaging cell line producing high-titer PPT-deleted integration-deficient lentiviral vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15025. [PMID: 26229972 PMCID: PMC4510976 DOI: 10.1038/mtm.2015.25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 12/29/2022]
Abstract
The risk of insertional mutagenesis inherent to all integrating exogenous expression cassettes was the impetus for the development of various integration-defective lentiviral vector (IDLV) systems. These systems were successfully employed in a plethora of preclinical applications, underscoring their clinical potential. However, current production of IDLVs by transient plasmid transfection is not optimal for large-scale production of clinical grade vectors. Here, we describe the development of the first tetracycline-inducible stable IDLV packaging cell line comprising the D64E integrase mutant and the VSV-G envelope protein. A conditional self-inactivating (cSIN) vector and a novel polypurine tract (PPT)-deleted vector were incorporated into the newly developed stable packaging cell line by transduction and stable transfection, respectively. High-titer (~10(7) infectious units (IU)/ml) cSIN vectors were routinely generated. Furthermore, screening of single-cell clones stably transfected with PPT-deleted vector DNA resulted in the identification of highly efficient producer cell lines generating IDLV titers higher than 10(8) IU/mL, which upon concentration increased to 10(10) IU/ml. IDLVs generated by stable producer lines efficiently transduce CNS tissues of rodents. Overall, the availability of high-titer IDLV lentivirus packaging cell line described here will significantly facilitate IDLV-based basic science research, as well as preclinical and clinical applications.
Collapse
|
10
|
Nogo-B protects mice against lipopolysaccharide-induced acute lung injury. Sci Rep 2015; 5:12061. [PMID: 26174362 PMCID: PMC4502524 DOI: 10.1038/srep12061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/09/2015] [Indexed: 12/11/2022] Open
Abstract
Nogo-B, a member of the reticulon 4 protein family, plays a critical role in tissue repair and acute inflammation. Its role in acute lung injury (ALI) remains unclear. Here, we assessed the function of Nogo-B during tissue injury in a lipopolysaccharide (LPS)-induced ALI mouse model. We found that pulmonary Nogo-B was significantly repressed after LPS instillation in C57BL/6 mice. Over-expression of pulmonary Nogo-B using an adenovirus vector carrying the Nogo-B-RFP-3flag gene (Ad-Nogo-B) significantly prolonged the survival of mice challenged with a lethal dose of LPS. The Ad-Nogo-B-treated mice also had less severe lung injury, less alveolar protein exudation, and a higher number of macrophages but less neutrophil infiltration compared with Ad-RFP-treated mice. Interestingly, microarray analysis showed that the Ad-Nogo-B-treated mice had different gene expression profiles compared with the controls and the prominent expression of genes related to wound healing and the humoral immune response after LPS induction. Of the 49 differently expressed genes, we found that the expression of PTX3 was significantly up-regulated following Nogo-B over-expression as observed in lung tissues and RAW264.7 cells. In conclusion, Nogo-B plays a protective role against LPS-induced ALI, and this effect might be exerted through the modulation of alveolar macrophage recruitment and PTX3 production.
Collapse
|
11
|
Novel potential targets for prevention of arterial restenosis: insights from the pre-clinical research. Clin Sci (Lond) 2014; 127:615-34. [PMID: 25072327 DOI: 10.1042/cs20140131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Restenosis is the pathophysiological process occurring in 10-15% of patients submitted to revascularization procedures of coronary, carotid and peripheral arteries. It can be considered as an excessive healing reaction of the vascular wall subjected to arterial/venous bypass graft interposition, endarterectomy or angioplasty. The advent of bare metal stents, drug-eluting stents and of the more recent drug-eluting balloons, have significantly reduced, but not eliminated, the incidence of restenosis, which remains a clinically relevant problem. Biomedical research in pre-clinical animal models of (re)stenosis, despite its limitations, has contributed enormously to the identification of processes involved in restenosis progression, going well beyond the initial dogma of a primarily proliferative disease. Although the main molecular and cellular mechanisms underlying restenosis have been well described, new signalling molecules and cell types controlling the progress of restenosis are continuously being discovered. In particular, microRNAs and vascular progenitor cells have recently been shown to play a key role in this pathophysiological process. In addition, the advanced highly sensitive high-throughput analyses of molecular alterations at the transcriptome, proteome and metabolome levels occurring in injured vessels in animal models of disease and in human specimens serve as a basis to identify novel potential therapeutic targets for restenosis. Molecular analyses are also contributing to the identification of reliable circulating biomarkers predictive of post-interventional restenosis in patients, which could be potentially helpful in the establishment of an early diagnosis and therapy. The present review summarizes the most recent and promising therapeutic strategies identified in experimental models of (re)stenosis and potentially translatable to patients subjected to revascularization procedures.
Collapse
|
12
|
Lu-Nguyen NB, Broadstock M, Schliesser MG, Bartholomae CC, von Kalle C, Schmidt M, Yáñez-Muñoz RJ. Transgenic expression of human glial cell line-derived neurotrophic factor from integration-deficient lentiviral vectors is neuroprotective in a rodent model of Parkinson's disease. Hum Gene Ther 2014; 25:631-41. [PMID: 24635742 DOI: 10.1089/hum.2014.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Standard integration-proficient lentiviral vectors (IPLVs) are effective at much lower doses than other vector systems and have shown promise for gene therapy of Parkinson's disease (PD). Their main drawback is the risk of insertional mutagenesis. The novel biosafety-enhanced integration-deficient lentiviral vectors (IDLVs) may offer a significant enhancement in biosafety, but have not been previously tested in a model of a major disease. We have assessed biosafety and transduction efficiency of IDLVs in a rat model of PD, using IPLVs as a reference. Genomic insertion of lentivectors injected into the lesioned striatum was studied by linear amplification-mediated polymerase chain reaction (PCR), followed by deep sequencing and insertion site analysis, demonstrating lack of significant IDLV integration. Reporter gene expression studies showed efficient, long-lived, and transcriptionally targeted expression from IDLVs injected ahead of lesioning in the rat striatum, although at somewhat lower expression levels than from IPLVs. Transgenic human glial cell line-derived neurotrophic factor (hGDNF) expression from IDLVs was used for a long-term investigation of lentivector-mediated, transcriptionally targeted neuroprotection in this PD rat model. Vectors were injected before striatal lesioning, and the results showed improvements in nigral dopaminergic neuron survival and behavioral tests regardless of lentiviral integration proficiency, although they confirmed lower expression levels of hGDNF from IDLVs. These data demonstrate the effectiveness of IDLVs in a model of a major disease and indicate that these vectors could provide long-term PD treatment at low dose, combining efficacy and biosafety for targeted central nervous system applications.
Collapse
Affiliation(s)
- Ngoc B Lu-Nguyen
- 1 School of Biological Sciences, Royal Holloway, University of London , Egham, Surrey TW20 0EX, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
13
|
Recent progresses in gene delivery-based bone tissue engineering. Biotechnol Adv 2013; 31:1695-706. [DOI: 10.1016/j.biotechadv.2013.08.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/24/2013] [Accepted: 08/19/2013] [Indexed: 12/18/2022]
|
14
|
Suwanmanee T, Hu G, Gui T, Bartholomae CC, Kutschera I, von Kalle C, Schmidt M, Monahan PE, Kafri T. Integration-deficient lentiviral vectors expressing codon-optimized R338L human FIX restore normal hemostasis in Hemophilia B mice. Mol Ther 2013; 22:567-574. [PMID: 23941813 DOI: 10.1038/mt.2013.188] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/31/2013] [Indexed: 12/31/2022] Open
Abstract
Integration-deficient lentiviral vectors (IDLVs) have been shown to transduce a wide spectrum of target cells and organs in vitro and in vivo and to maintain long-term transgene expression in nondividing cells. However, epigenetic silencing of episomal vector genomes reduces IDLV transgene expression levels and renders these safe vectors less efficient. In this article, we describe for the first time a complete correction of factor IX (FIX) deficiency in hemophilia B mice by IDLVs carrying a novel, highly potent human FIX cDNA. A 50-fold increase in human FIX cDNA potency was achieved by combining two mechanistically independent yet synergistic strategies: (i) optimization of the human FIX cDNA codon usage to increase human FIX protein production per vector genome and (ii) generation of a highly catalytic mutant human FIX protein in which the arginine residue at position 338 was substituted with leucine. The enhanced human FIX activity was not associated with liver damage or with the formation of human FIX-directed inhibitory antibodies and rendered IDLV-treated FIX-knockout mice resistant to a challenging tail-clipping assay. A novel S1 nuclease-based B1-quantitative polymerase chain reaction assay showed low levels of IDLV integration in mouse liver. Overall, this study demonstrates that IDLVs carrying an improved human FIX cDNA safely and efficiently cure hemophilia B in a mouse model.
Collapse
Affiliation(s)
- Thipparat Suwanmanee
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Genlin Hu
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Tong Gui
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cynthia C Bartholomae
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ina Kutschera
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pediatrics, Division of Hematology/Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.
| |
Collapse
|