1
|
Carrillo-Gálvez AB, Zurita F, Guerra-Valverde JA, Aguilar-González A, Abril-García D, Padial-Molina M, Olaechea A, Martín-Morales N, Martín F, O’Valle F, Galindo-Moreno P. NLRP3 and AIM2 inflammasomes expression is modified by LPS and titanium ions increasing the release of active IL-1β in alveolar bone-derived MSCs. Stem Cells Transl Med 2024; 13:826-841. [PMID: 39013640 PMCID: PMC11328940 DOI: 10.1093/stcltm/szae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/19/2024] [Indexed: 07/18/2024] Open
Abstract
Periodontitis and peri-implantitis are inflammatory diseases of infectious etiology that lead to the destruction of the supporting tissues located around teeth or implants. Although both pathologies share several characteristics, it is also known that they show important differences which could be due to the release of particles and metal ions from the implant surface. The activation of the inflammasome pathway is one of the main triggers of the inflammatory process. The inflammatory process in patients who suffer periodontitis or peri-implantitis has been mainly studied on cells of the immune system; however, it is also important to consider other cell types with high relevance in the regulation of the inflammatory response. In that context, mesenchymal stromal cells (MSCs) play an essential role in the regulation of inflammation due to their ability to modulate the immune response. This study shows that the induction of NLRP3 and absent in melanoma 2 (AIM2) inflammasome pathways mediated by bacterial components increases the secretion of active IL-1β and the pyroptotic process on human alveolar bone-derived mesenchymal stromal cells (hABSCs). Interestingly, when bacterial components are combined with titanium ions, NLRP3 expression is further increased while AIM2 expression is reduced. Furthermore, decrease of NLRP3 or AIM2 expression in hABSCs partially reverses the negative effect observed on the progression of the inflammatory process as well as on cell survival. In summary, our data suggest that the progression of the inflammatory process in peri-implantitis could be more acute due to the combined action of organic and inorganic components.
Collapse
Affiliation(s)
- Ana Belén Carrillo-Gálvez
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Federico Zurita
- Department of Genetics and Institute of Biotechnology, University of Granada, 18071 Granada, Spain
| | - José Antonio Guerra-Valverde
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Clinical Medicine and Public Health, University of Granada,18071 Granada, Spain
| | - Araceli Aguilar-González
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Bio-Medicine and the Environment, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Darío Abril-García
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Allinson Olaechea
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Clinical Medicine and Public Health, University of Granada,18071 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
| | - Natividad Martín-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Biomedicine, University of Granada, 18071 Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, 18071 Granada, Spain
| | - Francisco Martín
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18071 Granada, Spain
| | - Francisco O’Valle
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, 18071 Granada, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), University of Granada, 18071 Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| |
Collapse
|
2
|
Martinez-Navajas G, Ceron-Hernandez J, Simon I, Lupiañez P, Diaz-McLynn S, Perales S, Modlich U, Guerrero JA, Martin F, Sevivas T, Lozano ML, Rivera J, Ramos-Mejia V, Tersteeg C, Real PJ. Lentiviral gene therapy reverts GPIX expression and phenotype in Bernard-Soulier syndrome type C. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:75-92. [PMID: 37416759 PMCID: PMC10320622 DOI: 10.1016/j.omtn.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023]
Abstract
Bernard-Soulier syndrome (BSS) is a rare congenital disease characterized by macrothrombocytopenia and frequent bleeding. It is caused by pathogenic variants in three genes (GP1BA, GP1BB, or GP9) that encode for the GPIbα, GPIbβ, and GPIX subunits of the GPIb-V-IX complex, the main platelet surface receptor for von Willebrand factor, being essential for platelet adhesion and aggregation. According to the affected gene, we distinguish BSS type A1 (GP1BA), type B (GP1BB), or type C (GP9). Pathogenic variants in these genes cause absent, incomplete, or dysfunctional GPIb-V-IX receptor and, consequently, a hemorrhagic phenotype. Using gene-editing tools, we generated knockout (KO) human cellular models that helped us to better understand GPIb-V-IX complex assembly. Furthermore, we developed novel lentiviral vectors capable of correcting GPIX expression, localization, and functionality in human GP9-KO megakaryoblastic cell lines. Generated GP9-KO induced pluripotent stem cells produced platelets that recapitulated the BSS phenotype: absence of GPIX on the membrane surface and large size. Importantly, gene therapy tools reverted both characteristics. Finally, hematopoietic stem cells from two unrelated BSS type C patients were transduced with the gene therapy vectors and differentiated to produce GPIX-expressing megakaryocytes and platelets with a reduced size. These results demonstrate the potential of lentiviral-based gene therapy to rescue BSS type C.
Collapse
Affiliation(s)
- Gonzalo Martinez-Navajas
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Jorge Ceron-Hernandez
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Iris Simon
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Pablo Lupiañez
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Sofia Diaz-McLynn
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Sonia Perales
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Ute Modlich
- Department of Gene and Cell Therapy, Institute of Regenerative Medicine, University of Zürich, Wagistrasse 12, 8952 Schlieren-Zürich, Switzerland
| | - Jose A. Guerrero
- Department of Haematology, University of Cambridge, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Francisco Martin
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, Avenida Ilustracion S/n, 18016 Granada, Spain
| | - Teresa Sevivas
- Serviço de Sangue, Medicina Transfusional e Imunohemoterapia Do Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria L. Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, U765 Murcia, Spain
| | - Jose Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, U765 Murcia, Spain
- Grupo Español de Alteraciones Plaquetarias Congénitas (GEAPC), Madrid, Spain
| | - Veronica Ramos-Mejia
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Pedro J. Real
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| |
Collapse
|
3
|
Balmas E, Sozza F, Bottini S, Ratto ML, Savorè G, Becca S, Snijders KE, Bertero A. Manipulating and studying gene function in human pluripotent stem cell models. FEBS Lett 2023; 597:2250-2287. [PMID: 37519013 DOI: 10.1002/1873-3468.14709] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023]
Abstract
Human pluripotent stem cells (hPSCs) are uniquely suited to study human development and disease and promise to revolutionize regenerative medicine. These applications rely on robust methods to manipulate gene function in hPSC models. This comprehensive review aims to both empower scientists approaching the field and update experienced stem cell biologists. We begin by highlighting challenges with manipulating gene expression in hPSCs and their differentiated derivatives, and relevant solutions (transfection, transduction, transposition, and genomic safe harbor editing). We then outline how to perform robust constitutive or inducible loss-, gain-, and change-of-function experiments in hPSCs models, both using historical methods (RNA interference, transgenesis, and homologous recombination) and modern programmable nucleases (particularly CRISPR/Cas9 and its derivatives, i.e., CRISPR interference, activation, base editing, and prime editing). We further describe extension of these approaches for arrayed or pooled functional studies, including emerging single-cell genomic methods, and the related design and analytical bioinformatic tools. Finally, we suggest some directions for future advancements in all of these areas. Mastering the combination of these transformative technologies will empower unprecedented advances in human biology and medicine.
Collapse
Affiliation(s)
- Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Federica Sozza
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Sveva Bottini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Maria Luisa Ratto
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Giulia Savorè
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Silvia Becca
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Kirsten Esmee Snijders
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| |
Collapse
|
4
|
Duran AG, Schwestka M, Nazari-Shafti TZ, Neuber S, Stamm C, Gossen M. Limiting Transactivator Amounts Contribute to Transgene Mosaicism in Tet-On All-in-One Systems. ACS Synth Biol 2022; 11:2623-2635. [PMID: 35815862 DOI: 10.1021/acssynbio.2c00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MicroRNAs play an essential role in cell homeostasis and have been proposed as therapeutic agents. One strategy to deliver microRNAs is to genetically engineer target cells to express microRNAs of interest. However, to control dosage and timing, as well as to limit potential side-effects, microRNAs' expression should ideally be under exogenous, inducible control. Conditional expression of miRNA-based short hairpin RNAs (shRNAmirs) via gene regulatory circuits such as the Tet-system is therefore a promising strategy to control shRNAmirs' expression in research and therapy. Single vector approaches like Tet-On all-in-one designs are more compatible with potential clinical applications by providing the Tet-On system components in a single round of genetic engineering. However, all-in-one systems often come at the expense of heterogeneous and unstable expression. In this study, we aimed to understand the causes that lead to such erratic transgene expression. By using a reporter cell, we found that the degree of heterogeneity mostly correlated with reverse tetracycline transactivator (rtTA) expression levels. Moreover, the targeted integration of a potent rtTA expression cassette into a genomic safe harbor locus functionally rescued previously silenced rtTA-responsive transcription units. Overall, our results suggest that ensuring homogenous and stable rtTA expression is essential for the robust and reliable performance of future Tet-On all-in-one designs.
Collapse
Affiliation(s)
- Ana G Duran
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany.,Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Berlin 13353, Germany
| | - Marko Schwestka
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
| | - Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany
| | - Christof Stamm
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany.,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, 13353 Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
| |
Collapse
|
5
|
Tristán-Manzano M, Maldonado-Pérez N, Justicia-Lirio P, Muñoz P, Cortijo-Gutiérrez M, Pavlovic K, Jiménez-Moreno R, Nogueras S, Carmona MD, Sánchez-Hernández S, Aguilar-González A, Castella M, Juan M, Marañón C, Marchal JA, Benabdellah K, Herrera C, Martin F. Physiological lentiviral vectors for the generation of improved CAR-T cells. Mol Ther Oncolytics 2022; 25:335-349. [PMID: 35694446 PMCID: PMC9163403 DOI: 10.1016/j.omto.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/07/2022] [Indexed: 10/29/2022] Open
Abstract
Anti-CD19 chimeric antigen receptor (CAR)-T cells have achieved impressive outcomes for the treatment of relapsed and refractory B-lineage neoplasms. However, important limitations still remain due to severe adverse events (i.e., cytokine release syndrome and neuroinflammation) and relapse of 40%-50% of the treated patients. Most CAR-T cells are generated using retroviral vectors with strong promoters that lead to high CAR expression levels, tonic signaling, premature exhaustion, and overstimulation, reducing efficacy and increasing side effects. Here, we show that lentiviral vectors (LVs) expressing the transgene through a WAS gene promoter (AW-LVs) closely mimic the T cell receptor (TCR)/CD3 expression kinetic upon stimulation. These AW-LVs can generate improved CAR-T cells as a consequence of their moderate and TCR-like expression profile. Compared with CAR-T cells generated with human elongation factor α (EF1α)-driven-LVs, AW-CAR-T cells exhibited lower tonic signaling, higher proportion of naive and stem cell memory T cells, less exhausted phenotype, and milder secretion of tumor necrosis factor alpha (TNF-α) and interferon (IFN)-ɣ after efficient destruction of CD19+ lymphoma cells, both in vitro and in vivo. Moreover, we also showed their improved efficiency using an in vitro CD19+ pancreatic tumor model. We finally demonstrated the feasibility of large-scale manufacturing of AW-CAR-T cells in guanosine monophosphate (GMP)-like conditions. Based on these data, we propose the use of AW-LVs for the generation of improved CAR-T products.
Collapse
Affiliation(s)
- María Tristán-Manzano
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- LentiStem Biotech, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Noelia Maldonado-Pérez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Pedro Justicia-Lirio
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- LentiStem Biotech, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Pilar Muñoz
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- Department of Cellular Biology, Faculty of Sciences, University of Granada, Campus Fuentenueva, 18071 Granada, Spain
| | - Marina Cortijo-Gutiérrez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Kristina Pavlovic
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- Department of Cellular Biology, Faculty of Sciences, University of Granada, Campus Fuentenueva, 18071 Granada, Spain
| | - Rosario Jiménez-Moreno
- Maimonides Institute of Biomedical Research in Córdoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Sonia Nogueras
- Maimonides Institute of Biomedical Research in Córdoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - M. Dolores Carmona
- Maimonides Institute of Biomedical Research in Córdoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Sabina Sánchez-Hernández
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Araceli Aguilar-González
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Campus Cartuja, 18071 Granada, Spain
| | - María Castella
- Department of Hematology, ICMHO, Hospital Clínic de Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Manel Juan
- Department of Hematology, ICMHO, Hospital Clínic de Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Juan Antonio Marchal
- Biosanitary Research Institute of Granada (ibs.GRANADA), Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18071, Spain
| | - Karim Benabdellah
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Córdoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Francisco Martin
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Avda. de la Ilustración 114, 18016 Granada, Spain
- Departamento de Bioquimica y Biología Molecular 3 e Inmunología, Facultad de Medicina, Universidad de Granada, Avda. de la Investigacion 11, 18071 Granada, Spain
| |
Collapse
|
6
|
Aguilar-González A, González-Correa JE, Barriocanal-Casado E, Ramos-Hernández I, Lerma-Juárez MA, Greco S, Rodríguez-Sevilla JJ, Molina-Estévez FJ, Montalvo-Romeral V, Ronzitti G, Sánchez-Martín RM, Martín F, Muñoz P. Isogenic GAA-KO Murine Muscle Cell Lines Mimicking Severe Pompe Mutations as Preclinical Models for the Screening of Potential Gene Therapy Strategies. Int J Mol Sci 2022; 23:6298. [PMID: 35682977 PMCID: PMC9181599 DOI: 10.3390/ijms23116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Pompe disease (PD) is a rare disorder caused by mutations in the acid alpha-glucosidase (GAA) gene. Most gene therapies (GT) partially rely on the cross-correction of unmodified cells through the uptake of the GAA enzyme secreted by corrected cells. In the present study, we generated isogenic murine GAA-KO cell lines resembling severe mutations from Pompe patients. All of the generated GAA-KO cells lacked GAA activity and presented an increased autophagy and increased glycogen content by means of myotube differentiation as well as the downregulation of mannose 6-phosphate receptors (CI-MPRs), validating them as models for PD. Additionally, different chimeric murine GAA proteins (IFG, IFLG and 2G) were designed with the aim to improve their therapeutic activity. Phenotypic rescue analyses using lentiviral vectors point to IFG chimera as the best candidate in restoring GAA activity, normalising the autophagic marker p62 and surface levels of CI-MPRs. Interestingly, in vivo administration of liver-directed AAVs expressing the chimeras further confirmed the good behaviour of IFG, achieving cross-correction in heart tissue. In summary, we generated different isogenic murine muscle cell lines mimicking the severe PD phenotype, as well as validating their applicability as preclinical models in order to reduce animal experimentation.
Collapse
Affiliation(s)
- Araceli Aguilar-González
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Juan Elías González-Correa
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Eliana Barriocanal-Casado
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Iris Ramos-Hernández
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Miguel A. Lerma-Juárez
- Instituto de Investigación del Hospital Universitario La Paz, IdiPAZ, 28029 Madrid, Spain;
| | - Sara Greco
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Juan José Rodríguez-Sevilla
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
| | - Francisco Javier Molina-Estévez
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain
| | - Valle Montalvo-Romeral
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Giuseppe Ronzitti
- Généthon, Integrare Research Unit UMR_S951, INSERM, Université Paris-Saclay, Univ Evry, 91002 Evry, France; (V.M.-R.); (G.R.)
| | - Rosario María Sánchez-Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Francisco Martín
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departamento de Bioquímica y Biología Molecular 3 e Inmunología, Facultad de Medicina, Universidad de Granada, Avda. de la Investigación 11, 18071 Granada, Spain
| | - Pilar Muñoz
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government PTS Granada-Avenida de la Ilustración 114, 18016 Granada, Spain; (A.A.-G.); (J.E.G.-C.); (E.B.-C.); (I.R.-H.); (S.G.); (J.J.R.-S.); (F.J.M.-E.); (R.M.S.-M.)
- Departmento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain
| |
Collapse
|
7
|
Cortijo-Gutiérrez M, Sánchez-Hernández S, Tristán-Manzano M, Maldonado-Pérez N, Lopez-Onieva L, Real PJ, Herrera C, Marchal JA, Martin F, Benabdellah K. Improved Functionality of Integration-Deficient Lentiviral Vectors (IDLVs) by the Inclusion of IS 2 Protein Docks. Pharmaceutics 2021; 13:pharmaceutics13081217. [PMID: 34452178 PMCID: PMC8401568 DOI: 10.3390/pharmaceutics13081217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022] Open
Abstract
Integration-deficient lentiviral vectors (IDLVs) have recently generated increasing interest, not only as a tool for transient gene delivery, but also as a technique for detecting off-target cleavage in gene-editing methodologies which rely on customized endonucleases (ENs). Despite their broad potential applications, the efficacy of IDLVs has historically been limited by low transgene expression and by the reduced sensitivity to detect low-frequency off-target events. We have previously reported that the incorporation of the chimeric sequence element IS2 into the long terminal repeat (LTR) of IDLVs increases gene expression levels, while also reducing the episome yield inside transduced cells. Our study demonstrates that the effectiveness of IDLVs relies on the balance between two parameters which can be modulated by the inclusion of IS2 sequences. In the present study, we explore new IDLV configurations harboring several elements based on IS2 modifications engineered to mediate more efficient transgene expression without affecting the targeted cell load. Of all the insulators and configurations analysed, the insertion of the IS2 into the 3′LTR produced the best results. After demonstrating a DAPI-low nuclear gene repositioning of IS2-containing episomes, we determined whether, in addition to a positive effect on transcription, the IS2 could improve the capture of IDLVs on double strand breaks (DSBs). Thus, DSBs were randomly generated, using the etoposide or locus-specific CRISPR-Cas9. Our results show that the IS2 element improved the efficacy of IDLV DSB detection. Altogether, our data indicate that the insertion of IS2 into the LTR of IDLVs improved, not only their transgene expression levels, but also their ability to be inserted into existing DSBs. This could have significant implications for the development of an unbiased detection tool for off-target cleavage sites from different specific nucleases.
Collapse
Affiliation(s)
- Marina Cortijo-Gutiérrez
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
| | - Sabina Sánchez-Hernández
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
| | - María Tristán-Manzano
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
| | - Noelia Maldonado-Pérez
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
| | - Lourdes Lopez-Onieva
- GENYO, Centre for Genomics and Oncological Research, Molecular Oncology Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (L.L.-O.); (P.J.R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - Pedro J. Real
- GENYO, Centre for Genomics and Oncological Research, Molecular Oncology Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (L.L.-O.); (P.J.R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
- Personalized Oncology Group, Bio-Health Research Institute (ibs Granada), 18016 Granada, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain;
- Department of Haematology, Reina Sofía University Hospital, 14004 Cordoba, Spain
| | - Juan Antonio Marchal
- Biomedical Research Institute (ibs. Granada), 18012 Granada, Spain;
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Excellence Research Unit: Modeling Nature (MNat), University of Granada, 18016 Granada, Spain
| | - Francisco Martin
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
| | - Karim Benabdellah
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain; (M.C.-G.); (S.S.-H.); (M.T.-M.); (N.M.-P.); (F.M.)
- Correspondence: ; Tel.: +34-958-715-500
| |
Collapse
|
8
|
Page A, Fusil F, Cosset FL. Toward Tightly Tuned Gene Expression Following Lentiviral Vector Transduction. Viruses 2020; 12:v12121427. [PMID: 33322556 PMCID: PMC7764518 DOI: 10.3390/v12121427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Lentiviral vectors are versatile tools for gene delivery purposes. While in the earlier versions of retroviral vectors, transgene expression was controlled by the long terminal repeats (LTRs), the latter generations of vectors, including those derived from lentiviruses, incorporate internal constitutive or regulated promoters in order to regulate transgene expression. This allows to temporally and/or quantitatively control transgene expression, which is required for many applications such as for clinical applications, when transgene expression is required in specific tissues and at a specific timing. Here we review the main systems that have been developed for transgene regulated expression following lentiviral gene transfer. First, the induction of gene expression can be triggered either by external or by internal cues. Indeed, these regulated vector systems may harbor promoters inducible by exogenous stimuli, such as small molecules (e.g., antibiotics) or temperature variations, offering the possibility to tune rapidly transgene expression in case of adverse events. Second, expression can be indirectly adjusted by playing on inserted sequence copies, for instance by gene excision. Finally, synthetic networks can be developed to sense specific endogenous signals and trigger defined responses after information processing. Regulatable lentiviral vectors (LV)-mediated transgene expression systems have been widely used in basic research to uncover gene functions or to temporally reprogram cells. Clinical applications are also under development to induce therapeutic molecule secretion or to implement safety switches. Such regulatable approaches are currently focusing much attention and will benefit from the development of other technologies in order to launch autonomously controlled systems.
Collapse
|
9
|
Muñoz P, Tristán-Manzano M, Sánchez-Gilabert A, Santilli G, Galy A, Thrasher AJ, Martin F. WAS Promoter-Driven Lentiviral Vectors Mimic Closely the Lopsided WASP Expression during Megakaryocytic Differentiation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:220-235. [PMID: 33102615 PMCID: PMC7558809 DOI: 10.1016/j.omtm.2020.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/11/2020] [Indexed: 01/10/2023]
Abstract
Transplant of gene-modified autologous hematopoietic progenitors cells has emerged as a new therapeutic approach for Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency with microthrombocytopenia and abnormal lymphoid and myeloid functions. Despite the clinical benefits obtained in ongoing clinical trials, platelet restoration is suboptimal. The incomplete restoration of platelets in these patients can be explained either by a low number of corrected cells or by insufficient or inadequate WASP expression during megakaryocyte differentiation and/or in platelets. We therefore used in vitro models to study the endogenous WASP expression pattern during megakaryocytic differentiation and compared it with the expression profiles achieved by different therapeutic lentiviral vectors (LVs) driving WAS cDNA through different regions of the WAS promoter. Our data showed that all WAS promoter-driven LVs mimic very closely the endogenous WAS expression kinetic during megakaryocytic differentiation. However, LVs harboring the full-length (1.6-kb) WAS-proximal promoter (WW1.6) or a combination of the WAS alternative and proximal promoters (named AW) had the best behavior. Finally, all WAS-driven LVs restored the WAS knockout (WASKO) mice phenotype and functional defects of hematopoietic stem and progenitor cells (HSPCs) from a WAS patient with similar efficiency. In summary, our data back up the use of WW1.6 and AW LVs as physiological gene transfer tools for WAS therapy.
Collapse
Affiliation(s)
- Pilar Muñoz
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain.,University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Almudena Sánchez-Gilabert
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Giorgia Santilli
- University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - Anne Galy
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, 91000 Evry, France
| | - Adrian J Thrasher
- University College London (UCL) Great Ormond Street Institute of Child Health (ICH), 30 Guilford Street, WC1N 1EH London, UK
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Ilustracion 114, 18016 Granada, Spain
| |
Collapse
|
10
|
Tristán-Manzano M, Justicia-Lirio P, Maldonado-Pérez N, Cortijo-Gutiérrez M, Benabdellah K, Martin F. Externally-Controlled Systems for Immunotherapy: From Bench to Bedside. Front Immunol 2020; 11:2044. [PMID: 33013864 PMCID: PMC7498544 DOI: 10.3389/fimmu.2020.02044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/28/2020] [Indexed: 12/27/2022] Open
Abstract
Immunotherapy is a very promising therapeutic approach against cancer that is particularly effective when combined with gene therapy. Immuno-gene therapy approaches have led to the approval of four advanced therapy medicinal products (ATMPs) for the treatment of p53-deficient tumors (Gendicine and Imlygic), refractory acute lymphoblastic leukemia (Kymriah) and large B-cell lymphomas (Yescarta). In spite of these remarkable successes, immunotherapy is still associated with severe side effects for CD19+ malignancies and is inefficient for solid tumors. Controlling transgene expression through an externally administered inductor is envisioned as a potent strategy to improve safety and efficacy of immunotherapy. The aim is to develop smart immunogene therapy-based-ATMPs, which can be controlled by the addition of innocuous drugs or agents, allowing the clinicians to manage the intensity and durability of the therapy. In the present manuscript, we will review the different inducible, versatile and externally controlled gene delivery systems that have been developed and their applications to the field of immunotherapy. We will highlight the advantages and disadvantages of each system and their potential applications in clinics.
Collapse
Affiliation(s)
- María Tristán-Manzano
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Pedro Justicia-Lirio
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain.,LentiStem Biotech, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Noelia Maldonado-Pérez
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Marina Cortijo-Gutiérrez
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Karim Benabdellah
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Francisco Martin
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
11
|
Abstract
Mesenchymal stromal cell (MSC) therapy has produced very promising results for multiple diseases in animal models, with over 780 clinical trials on going or completed. However, most of the human clinical trials have not been as successful as trials using preclinical models. To improve the therapeutic potential of MSCs, different research groups have used gene transfer vectors to express factors involved in migration, survival, differentiation, and immunomodulation. The ideal gene transfer vector for most applications should achieve long-term, stable (constitutive or inducible) transgene expression in MSCs and their progeny. Given their efficiency and low impact on transduced cells, lentiviral vectors (LVs) are the vectors of choice. In this chapter we will describe a detailed protocol for the generation of genetically modified MSCs using lentiviral vectors (LVs). Although this protocol has been optimized for MSC lentiviral transduction, it can be easily adapted to other stem cells by changing culture conditions while maintaining volumes and incubation times.
Collapse
|
12
|
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: 1.5] [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
|
13
|
Wang K, Li Y, Zhu T, Zhang Y, Li W, Lin W, Li J, Zhu C. Overexpression of c-Met in bone marrow mesenchymal stem cells improves their effectiveness in homing and repair of acute liver failure. Stem Cell Res Ther 2017; 8:162. [PMID: 28679425 PMCID: PMC5499016 DOI: 10.1186/s13287-017-0614-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
Background Transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) has emerged as a novel therapy for acute liver failure (ALF). However, the homing efficiency of BMSCs to the injured liver sites appears to be poor. In this study, we aimed to determine if overexpression of c-Met in BMSCs could promote the homing ability of BMSCs to rat livers affected by ALF. Methods Overexpression of c-Met in BMSCs (c-Met-BMSCs) was attained by transfection of naive BMSCs with the lenti-c-Met-GFP. The impact of transplanted c-Met-BMSCs on both homing and repair of ALF was evaluated and compared with lenti-GFP empty vector transfected BMSCs (control BMSCs). Results After cells were transfected with the lenti-c-Met-GFP vector, the BMSCs displayed very high expression of c-Met protein as demonstrated by Western blot. In addition, in vitro transwell migration assays showed that the migration ability of c-Met-BMSCs was significantly increased in comparison with that of control BMSCs (P < 0.05), and was dependent on hepatocyte growth factor (HGF). Furthermore, rats with ALF that received transplanted c-Met-BMSCs showed significantly improved homing ability to the injured liver; this was accompanied by elevated survival rates and liver function in the ALF rats. Parallel pathological examination further confirmed that transplantation of c-Met-BMSCs ameliorated liver injury with reduced hepatic activity index (HAI) scores, and that the effects of c-Met-BMSCs were more profound than those of control BMSCs. Conclusions Overexpression of c-Met promotes the homing of BMSCs to injured hepatic sites in a rat model of ALF, thereby improving the efficacy of BMSC therapy for ALF repair.
Collapse
Affiliation(s)
- Kun Wang
- Department of Infectious Disease, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Department of Infectious Disease, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Yuwen Li
- Department of Pediatrics, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Tiantian Zhu
- Department of Infectious Disease, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Department of Infectious Disease, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Yongting Zhang
- Department of Infectious Disease, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wenting Li
- Department of Infectious Disease, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jun Li
- Department of Infectious Disease, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Chuanlong Zhu
- Department of Infectious Disease, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
14
|
Anderson KG, Stromnes IM, Greenberg PD. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies. Cancer Cell 2017; 31:311-325. [PMID: 28292435 PMCID: PMC5423788 DOI: 10.1016/j.ccell.2017.02.008] [Citation(s) in RCA: 468] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/13/2022]
Abstract
T cell dysfunction in solid tumors results from multiple mechanisms. Altered signaling pathways in tumor cells help produce a suppressive tumor microenvironment enriched for inhibitory cells, posing a major obstacle for cancer immunity. Metabolic constraints to cell function and survival shape tumor progression and immune cell function. In the face of persistent antigen, chronic T cell receptor signaling drives T lymphocytes to a functionally exhausted state. Here we discuss how the tumor and its microenvironment influences T cell trafficking and function with a focus on melanoma, and pancreatic and ovarian cancer, and discuss how scientific advances may help overcome these hurdles.
Collapse
Affiliation(s)
- Kristin G Anderson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Ingunn M Stromnes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Philip D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|