1
|
Fiore D, Cappelli LV, Zhaoqi L, Kotlov N, Sorokina M, Phillip J, Zumbo P, Yoffe L, Ghione P, Wang A, Han X, Taylor A, Chiu W, Fragliasso V, Tabbo F, Zamponi N, Di Siervi N, Kayembe C, Medico G, Patel RP, Gaudiano M, Machiorlatti R, Astone G, Cacciapuoti MT, Zanetti G, Pignataro C, Eric RA, Patel S, Zammarchi F, Zanettini C, Queiroz L, Nikitina A, Kudryashova O, Karelin A, Nikitin D, Tychinin D, Postovalova E, Bagaev A, Svekolkin V, Belova E, Tikhonova K, Degryse S, Xu C, Novero D, Ponzoni M, Tiacci E, Falini B, Song J, Khodos I, De Stanchina E, Macari G, Cafforio L, Gardini S, Piva R, Medico E, Ng SY, Moskowitz A, Epstein Z, Intlekofer A, Ahmed D, Chan WC, Martin P, Ruan J, Bertoni F, Foà R, Brody JD, Weinstock DM, Osan J, Santambrogio L, Elemento O, Betel D, Tam W, Ruella M, Cerchietti L, Rabadan R, Horwitz S, Inghirami G. A patient-derived T cell lymphoma biorepository uncovers pathogenetic mechanisms and host-related therapeutic vulnerabilities. Cell Rep Med 2025; 6:102029. [PMID: 40147445 DOI: 10.1016/j.xcrm.2025.102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 04/24/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Peripheral T cell lymphomas (PTCLs) comprise heterogeneous malignancies with limited therapeutic options. To uncover targetable vulnerabilities, we generate a collection of PTCL patient-derived tumor xenografts (PDXs) retaining histomorphology and molecular donor-tumor features over serial xenografting. PDX demonstrates remarkable heterogeneity, complex intratumor architecture, and stepwise trajectories mimicking primary evolutions. Combining functional transcriptional stratification and multiparametric imaging, we identify four distinct PTCL microenvironment subtypes with prognostic value. Mechanistically, we discover a subset of PTCLs expressing Epstein-Barr virus-specific T cell receptors and uncover the capacity of cancer-associated fibroblasts of counteracting treatments. PDXs' pre-clinical testing captures individual vulnerabilities, mirrors donor patients' clinical responses, and defines effective patient-tailored treatments. Ultimately, we assess the efficacy of CD5KO- and CD30- Chimeric Antigen Receptor T Cells (CD5KO-CART and CD30_CART, respectively), demonstrating their therapeutic potential and the synergistic role of immune checkpoint inhibitors for PTCL treatment. This repository represents a resource for discovering and validating intrinsic and extrinsic factors and improving the selection of drugs/combinations and immune-based therapies.
Collapse
Affiliation(s)
- Danilo Fiore
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; Institute for Experimental Endocrinology and Oncology, "G.Salvatore" IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Luca Vincenzo Cappelli
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Liu Zhaoqi
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Jude Phillip
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Chemical and Biomolecular Engineering, Oncology, Sidney Kimmel Comprehensive Cancer Center, Core Member, Institute for Nanobiotechnology (INBT), Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Liron Yoffe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paola Ghione
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anqi Wang
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Xueshuai Han
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Abigail Taylor
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - William Chiu
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Valentina Fragliasso
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Laboratory of translational research, Azienda USL - IRCCS di Reggio Emilia, 42122 Reggio Emila, Italy
| | - Fabrizio Tabbo
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; SC Oncologia ASL CN2 Alba Bra Ospedale Michele e Pietro Ferrero, 12060 Verduno, (CN), Italy
| | - Nahuel Zamponi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Nicolás Di Siervi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Clarisse Kayembe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Medico
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ruchi P Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcello Gaudiano
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rodolfo Machiorlatti
- Department of Pathology, Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Giuseppina Astone
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Maria Teresa Cacciapuoti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giorgia Zanetti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Ruiz Arvin Eric
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sanjay Patel
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Claudio Zanettini
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lucio Queiroz
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Chengqi Xu
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Domenico Novero
- Division of Pathological Anatomy, Quality and Safety of Diagnosis and Treatment, Città della Salute e della Scienza, 10126 Turin, Italy
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute, Milan, Italy; Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Enrico Tiacci
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Brunangelo Falini
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Joo Song
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Inna Khodos
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | - Elisa De Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | | | | | | | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, TO, Italy; Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy
| | - Samuel Y Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; National Cancer Institute, Bethesda, MD 20892, USA
| | - Allison Moskowitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zachary Epstein
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew Intlekofer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dogan Ahmed
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Peter Martin
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Jia Ruan
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Francesco Bertoni
- Lymphoma Genomics, Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, EOC,6500 Bellinzona, Switzerland
| | - Robin Foà
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Joshua D Brody
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, US; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Jaspreet Osan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Oliver Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Doron Betel
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Wayne Tam
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematopathology, Northwell Health, New York, NY 11740, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leandro Cerchietti
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Steven Horwitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giorgio Inghirami
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
2
|
Rodríguez-Sánchez A, Quijada-Álamo M, Pérez-Carretero C, Herrero AB, Arroyo-Barea A, Dávila-Valls J, Rubio A, García de Coca A, Benito-Sánchez R, Rodríguez-Vicente AE, Hernández-Rivas JM, Hernández-Sánchez M. SAMHD1 dysfunction impairs DNA damage response and increases sensitivity to PARP inhibition in chronic lymphocytic leukemia. Sci Rep 2025; 15:10446. [PMID: 40140468 PMCID: PMC11947222 DOI: 10.1038/s41598-025-93629-7] [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: 10/02/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a clinically and genetically heterogenous disease. Recent next-generation sequencing (NGS) studies have uncovered numerous low-frequency mutated genes in CLL patients, with SAMHD1 emerging as a candidate driver gene. However, the biological and clinical implications of SAMHD1 mutations remain unclear. Using CRISPR/Cas9, we generated CLL models to investigate the impact of SAMHD1 deficiency on pathogenesis and explore therapeutic strategies. Moreover, we performed NGS in treatment-naïve CLL patients to characterize SAMHD1 mutations and employed RNA-sequencing to evaluate their clinical significance. Our study shows that SAMHD1 inactivation impairs the DNA damage response by reducing homologous recombination efficiency through BRCA1 and RAD51 dysregulation. Importantly, SAMHD1 colocalizes with BRCA1 at DNA damage sites in CLL cells. This research also identifies that SAMHD1-mutated cells are more sensitive to PARP inhibition. Clinically, SAMHD1 dysfunction negatively impacts clinical outcome of CLL cases: SAMHD1 mutations reduce failure-free survival (median 46 vs 57 months, p = 0.033), while low SAMHD1 expression associates with shorter time to first treatment (median 47 vs 77 months; p = 0.00073). Overall, this study elucidates that SAMHD1 dysfunction compromises DNA damage response mechanisms, potentially contributing to unfavorable clinical outcomes in CLL, and proposes PARP-inhibitors as a potential therapeutic approach for SAMHD1-mutated CLL cells.
Collapse
MESH Headings
- SAM Domain and HD Domain-Containing Protein 1/metabolism
- SAM Domain and HD Domain-Containing Protein 1/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Humans
- DNA Damage
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Mutation
- Cell Line, Tumor
- BRCA1 Protein/genetics
- BRCA1 Protein/metabolism
- Female
- Animals
- Male
- Mice
Collapse
Affiliation(s)
- Alberto Rodríguez-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Miguel Quijada-Álamo
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Pérez-Carretero
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ana B Herrero
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Departamento de Medicina, Unidad de Medicina Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Andrés Arroyo-Barea
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Julio Dávila-Valls
- Servicio de Hematología, Hospital Nuestra Señora de Sonsoles, SACYL, Ávila, Spain
| | - Araceli Rubio
- Servicio de Hematología, Hospital Miguel Servet, SERGAS, Zaragoza, Spain
| | | | - Rocío Benito-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ana E Rodríguez-Vicente
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - María Hernández-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain.
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
3
|
Wang M, Fu P, Chen Z, Wang X, Ma H, Zhang X, Gao G. Recruitment and rejoining of remote double-strand DNA breaks for enhanced and precise chromosome editing. Genome Biol 2025; 26:53. [PMID: 40069752 PMCID: PMC11895233 DOI: 10.1186/s13059-025-03523-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Chromosomal rearrangements, such as translocations, deletions, and inversions, underlie numerous genetic diseases and cancers, yet precise engineering of these rearrangements remains challenging. Here, we present a CRISPR-based homologous recombination-mediated rearrangement (HRMR) strategy that leverages homologous donor templates to align and repair broken chromosome ends. HRMR improves efficiency by approximately 80-fold compared to non-homologous end joining, achieving over 95% homologous recombination. Validated across multiple loci and cell lines, HRMR enables efficient and accurate chromosomal rearrangements. Live-cell imaging reveals that homologous donors mediate chromosome end proximity, enhancing rearrangement efficiency. Thus, HRMR provides a powerful tool for disease modeling, chromosomal biology, and therapeutic applications.
Collapse
Affiliation(s)
- Mingyao Wang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Pengchong Fu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Ziheng Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Xiangnan Wang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Hanhui Ma
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Xuedi Zhang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China.
- Department of Cell Biology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China.
| | - Guanjun Gao
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China.
| |
Collapse
|
4
|
Monchusi B, Dube P, Takundwa MM, Kenmogne VL, Thimiri Govinda Raj DB. Advances in CRISPR-Cas systems for blood cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 208:261-284. [PMID: 39266186 DOI: 10.1016/bs.pmbts.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
CRISPR-Cas systems have revolutionised precision medicine by enabling personalised treatments tailored to an individual's genetic profile. Various CRISPR technologies have been developed to target specific disease-causing genes in blood cancers, and some have advanced to clinical trials. Although some studies have explored the in vivo applications of CRISPR-Cas systems, several challenges continue to impede their widespread use. Furthermore, CRISPR-Cas technology has shown promise in improving the response of immunotherapies to blood cancers. The emergence of CAR-T cell therapy has shown considerable success in the targeting and correcting of disease-causing genes in blood cancers. Despite the promising potential of CRISPR-Cas in the treatment of blood cancers, issues related to safety, ethics, and regulatory approval remain significant hurdles. This comprehensive review highlights the transformative potential of CRISPR-Cas technology to revolutionise blood cancer therapy.
Collapse
Affiliation(s)
- Bernice Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Phumuzile Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Mutsa Monica Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Vanelle Larissa Kenmogne
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa; Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - Deepak Balaji Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
5
|
Tyagi S, Singh A, Sharma N, Chaturvedi R, Kushwaha HR. Insights into existing and futuristic treatment approach for chronic myeloid leukaemia. Indian J Med Res 2024; 159:455-467. [PMID: 39382408 PMCID: PMC11463244 DOI: 10.25259/ijmr_1716_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Indexed: 10/10/2024] Open
Abstract
Oncogenes play a crucial part in human cancer development, and when particular drugs obstruct the proteins produced by these oncogenes, the tumoural process can be ceased. For instance, in chronic myeloid leukaemia (CML), all pathological traits are associated with a single oncogene, BCR-ABL1. CML is a triphasic cancerous disorder of haematopoietic stem cells, marked by a balanced translocation between chromosomes 9 and 22, leading to the genesis of a Philadelphia chromosome encompassing the BCR-ABL1 fusion gene. This fusion oncogene further produces a constitutive active tyrosine kinase protein, enhancing the downstream signalling pathways and constitutes cancer. The treatment for CML has been entirely altered from chemotherapy and immunotherapy to targeted therapy with the emergence of tyrosine kinase inhibitors (TKIs) which inhibit BCR-ABL1 kinase activity. However, the inhibitory mechanism of TKIs is constrained by BCR-ABL1 dependent and independent resistance mechanisms, prompting the exploration of novel therapeutics through extensive clinical trials to develop next-generation drugs with enhanced potency. The persistent challenges posed by CML have motivated researchers to seek innovative strategies for its eradication, such as the application of the genome editing tool CRISPR/Cas9. This review provides insights into existing CML diagnoses, treatment modalities, resistance mechanisms, drugs under trial phases and new potential therapeutic drugs. Furthermore, the review looks ahead to a visionary perspective wherein the CRISPR/Cas9 approach holds the potential to evolve into a prospective curative measure for CML.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Protein Kinase Inhibitors/therapeutic use
- Gene Editing
- Drug Resistance, Neoplasm/genetics
- CRISPR-Cas Systems/genetics
Collapse
Affiliation(s)
- Sourabh Tyagi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Anu Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Naveen Sharma
- Biomedical Informatics Division, Indian Council of Medical Research, New Delhi, India
| | - Rupesh Chaturvedi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Hemant Ritturaj Kushwaha
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
6
|
Zeng J, Liang X, Duan L, Tan F, Chen L, Qu J, Li J, Li K, Luo D, Hu Z. Targeted disruption of the BCR-ABL fusion gene by Cas9/dual-sgRNA inhibits proliferation and induces apoptosis in chronic myeloid leukemia cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:525-537. [PMID: 38414349 DOI: 10.3724/abbs.2023280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024] Open
Abstract
The BCR-ABL fusion gene, formed by the fusion of the breakpoint cluster region protein ( BCR) and the Abl Oncogene 1, Receptor Tyrosine Kinase ( ABL) genes, encodes the BCR-ABL oncoprotein, which plays a crucial role in leukemogenesis. Current therapies have limited efficacy in patients with chronic myeloid leukemia (CML) because of drug resistance or disease relapse. Identification of novel strategies to treat CML is essential. This study aims to explore the efficiency of novel CRISPR-associated protein 9 (Cas9)/dual-single guide RNA (sgRNA)-mediated disruption of the BCR-ABL fusion gene by targeting BCR and cABL introns. A co-expression vector for Cas9 green fluorescent protein (GFP)/dual-BA-sgRNA targeting BCR and cABL introns is constructed to produce lentivirus to affect BCR-ABL expression in CML cells. The effects of dual-sgRNA virus-mediated disruption of BCR-ABL are analyzed via the use of a genomic sequence and at the protein expression level. Cell proliferation, cell clonogenic ability, and cell apoptosis are assessed after dual sgRNA virus infection, and phosphorylated BCR-ABL and its downstream signaling molecules are detected. These effects are further confirmed in a CML mouse model via tail vein injection of Cas9-GFP/dual-BA-sgRNA virus-infected cells and in primary cells isolated from patients with CML. Cas9-GFP/dual-BA-sgRNA efficiently disrupts BCR-ABL at the genomic sequence and gene expression levels in leukemia cells, leading to blockade of the BCR-ABL tyrosine kinase signaling pathway and disruption of its downstream molecules, followed by cell proliferation inhibition and cell apoptosis induction. This method prolongs the lifespan of CML model mice. Furthermore, the effect is confirmed in primary cells derived from patients with CML.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Apoptosis/genetics
- Cell Proliferation/genetics
- CRISPR-Cas Systems
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Genes, abl
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Proto-Oncogene Proteins c-bcr/genetics
- Proto-Oncogene Proteins c-bcr/metabolism
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Jianling Zeng
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Xinquan Liang
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Lili Duan
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Fenghua Tan
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Liujie Chen
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Jiayao Qu
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
| | - Jia Li
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
| | - Kai Li
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Dixian Luo
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518000, China
| | - Zheng Hu
- Translational Medicine Institute, the First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
- The First Affiliated Hospital of Xiangnan University, Chenzhou 423000, China
- National & Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, the First People's Hospital of Chenzhou, Chenzhou 423000, China
| |
Collapse
|
7
|
Wu A, Liu X, Fruhstorfer C, Jiang X. Clinical Insights into Structure, Regulation, and Targeting of ABL Kinases in Human Leukemia. Int J Mol Sci 2024; 25:3307. [PMID: 38542279 PMCID: PMC10970269 DOI: 10.3390/ijms25063307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/09/2024] Open
Abstract
Chronic myeloid leukemia is a multistep, multi-lineage myeloproliferative disease that originates from a translocation event between chromosome 9 and chromosome 22 within the hematopoietic stem cell compartment. The resultant fusion protein BCR::ABL1 is a constitutively active tyrosine kinase that can phosphorylate multiple downstream signaling molecules to promote cellular survival and inhibit apoptosis. Currently, tyrosine kinase inhibitors (TKIs), which impair ABL1 kinase activity by preventing ATP entry, are widely used as a successful therapeutic in CML treatment. However, disease relapses and the emergence of resistant clones have become a critical issue for CML therapeutics. Two main reasons behind the persisting obstacles to treatment are the acquired mutations in the ABL1 kinase domain and the presence of quiescent CML leukemia stem cells (LSCs) in the bone marrow, both of which can confer resistance to TKI therapy. In this article, we systemically review the structural and molecular properties of the critical domains of BCR::ABL1 and how understanding the essential role of BCR::ABL1 kinase activity has provided a solid foundation for the successful development of molecularly targeted therapy in CML. Comparison of responses and resistance to multiple BCR::ABL1 TKIs in clinical studies and current combination treatment strategies are also extensively discussed in this article.
Collapse
MESH Headings
- Humans
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Signal Transduction
Collapse
Affiliation(s)
- Andrew Wu
- Collings Stevens Chronic Leukemia Research Laboratory, Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (A.W.); (X.L.)
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Xiaohu Liu
- Collings Stevens Chronic Leukemia Research Laboratory, Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (A.W.); (X.L.)
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Clark Fruhstorfer
- Collings Stevens Chronic Leukemia Research Laboratory, Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (A.W.); (X.L.)
| | - Xiaoyan Jiang
- Collings Stevens Chronic Leukemia Research Laboratory, Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (A.W.); (X.L.)
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
8
|
Page EC, Heatley SL, Rehn J, Thomas PQ, Yeung DT, White DL. Gain of chromosome 21 increases the propensity for P2RY8::CRLF2 acute lymphoblastic leukemia via increased HMGN1 expression. Front Oncol 2023; 13:1177871. [PMID: 37483494 PMCID: PMC10358767 DOI: 10.3389/fonc.2023.1177871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) patients with a gain of chromosome 21, intrachromosomal amplification of chromosome 21 (iAMP21), or Down syndrome (DS), have increased expression of genes in the DS critical region (DSCR) of chromosome 21, including the high-mobility group nucleosome-binding protein 1, HMGN1. Children with DS are predisposed to develop hematologic malignancies, providing insight into the role of chromosome 21 in the development of leukemias. A 320-kb deletion in the pseudoautosomal region of the X/Y chromosome in leukemic cells, resulting in a gene fusion between the purinergic receptor and cytokine receptor-like factor-2 (P2Y Receptor Family Member 8 (P2RY8)::CRLF2), is a common feature in ~60% of DS-ALL and ~40% of iAMP21 patients, suggesting a link between chromosome 21 and P2RY8::CRLF2. In an Australian cohort of pediatric B-ALL patients with P2RY8::CRLF2 (n = 38), eight patients harbored gain of chromosome 21 (+21), and two patients had iAMP21, resulting in a significantly increased HMGN1 expression. An inducible CRISPR/Cas9 system was used to model P2RY8::CRLF2 and investigate its cooperation with HMGN1. This model was then used to validate HMGN1 as an influencing factor for P2RY8::CRLF2 development. Using Cas9 to cleave the DNA at the pseudoautosomal region without directed repair, cells expressing HMGN1 favored repair, resulting in P2RY8::CRLF2 generation, compared with cells without HMGN1. CRISPR/Cas9 P2RY8::CRLF2 cells expressing HMGN1 exhibit increased proliferation, thymic stromal lymphopoietin receptor (TSLPR) expression, and JAK/STAT signaling, consistent with cells from patients with P2RY8::CRLF2. Our patient expression data and unique CRISPR/Cas9 modeling, when taken together, suggest that HMGN1 increases the propensity for P2RY8::CRLF2 development. This has important implications for patients with DS, +21, or iAMP21.
Collapse
Affiliation(s)
- Elyse C. Page
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, Engineering, and Technology, University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | - Susan L. Heatley
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | - Jacqueline Rehn
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | - Paul Q. Thomas
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
- SA Gene Editing Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - David T. Yeung
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
- Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia
- Department of Hematology, Royal Adelaide Hospital and SA Pathology, Adelaide, SA, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, Australia
| | - Deborah L. White
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, Engineering, and Technology, University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
- Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia
- Australian and New Zealand Children’s Hematology/Oncology Group (ANZCHOG), Clayton, VIC, Australia
| |
Collapse
|
9
|
Lainšček D, Forstnerič V, Mikolič V, Malenšek Š, Pečan P, Benčina M, Sever M, Podgornik H, Jerala R. Coiled-coil heterodimer-based recruitment of an exonuclease to CRISPR/Cas for enhanced gene editing. Nat Commun 2022; 13:3604. [PMID: 35739111 PMCID: PMC9226073 DOI: 10.1038/s41467-022-31386-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
The CRISPR/Cas system has emerged as a powerful and versatile genome engineering tool, revolutionizing biological and biomedical sciences, where an improvement of efficiency could have a strong impact. Here we present a strategy to enhance gene editing based on the concerted action of Cas9 and an exonuclease. Non-covalent recruitment of exonuclease to Cas9/gRNA complex via genetically encoded coiled-coil based domains, termed CCExo, recruited the exonuclease to the cleavage site and robustly increased gene knock-out due to progressive DNA strand recession at the cleavage site, causing decreased re-ligation of the nonedited DNA. CCExo exhibited increased deletion size and enhanced gene inactivation efficiency in the context of several DNA targets, gRNA selection, Cas variants, tested cell lines and type of delivery. Targeting a sequence-specific oncogenic chromosomal translocation using CCExo in cells of chronic myelogenous leukemia patients and in an animal model led to the reduction or elimination of cancer, establishing it as a highly specific tool for treating CML and potentially other appropriate diseases with genetic etiology.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana, 1000, Slovenia
| | - Vida Forstnerič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia
| | - Veronika Mikolič
- Department of Hematology, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Peter Pečan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana, 1000, Slovenia
| | - Matjaž Sever
- Department of Hematology, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, 1000, Slovenia
- Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, 1000, Slovenia
| | - Helena Podgornik
- Department of Hematology, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, 1000, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana, 1000, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, 1000, Slovenia.
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana, 1000, Slovenia.
| |
Collapse
|
10
|
Vuelta E, Ordoñez JL, Sanz DJ, Ballesteros S, Hernández-Rivas JM, Méndez-Sánchez L, Sánchez-Martín M, García-Tuñón I. CRISPR/Cas9-Directed Gene Trap Constitutes a Selection System for Corrected BCR/ABL Leukemic Cells in CML. Int J Mol Sci 2022; 23:ijms23126386. [PMID: 35742831 PMCID: PMC9224210 DOI: 10.3390/ijms23126386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic myeloid leukaemia (CML) is a haematological neoplasm driven by the BCR/ABL fusion oncogene. The monogenic aspect of the disease and the feasibility of ex vivo therapies in haematological disorders make CML an excellent candidate for gene therapy strategies. The ability to abolish any coding sequence by CRISPR-Cas9 nucleases offers a powerful therapeutic opportunity to CML patients. However, a definitive cure can only be achieved when only CRISPR-edited cells are selected. A gene-trapping approach combined with CRISPR technology would be an ideal approach to ensure this. Here, we developed a CRISPR-Trap strategy that efficiently inserts a donor gene trap (SA-CMV-Venus) cassette into the BCR/ABL-specific fusion point in the CML K562 human cell line. The trapping cassette interrupts the oncogene coding sequence and expresses a reporter gene that enables the selection of edited cells. Quantitative mRNA expression analyses showed significantly higher level of expression of the BCR/Venus allele coupled with a drastically lower level of BCR/ABL expression in Venus+ cell fractions. Functional in vitro experiments showed cell proliferation arrest and apoptosis in selected Venus+ cells. Finally, xenograft experiments with the selected Venus+ cells showed a large reduction in tumour growth, thereby demonstrating a therapeutic benefit in vivo. This study represents proof of concept for the therapeutic potential of a CRISPR-Trap system as a novel strategy for gene elimination in haematological neoplasms.
Collapse
Affiliation(s)
- Elena Vuelta
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain; (E.V.); (S.B.); (J.M.H.-R.)
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), 37007 Salamanca, Spain;
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, 37007 Salamanca, Spain;
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José L. Ordoñez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Fisiología y Farmacología, Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain;
| | - David J. Sanz
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), 37007 Salamanca, Spain;
| | - Sandra Ballesteros
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain; (E.V.); (S.B.); (J.M.H.-R.)
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), 37007 Salamanca, Spain;
| | - Jesús M. Hernández-Rivas
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain; (E.V.); (S.B.); (J.M.H.-R.)
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), 37007 Salamanca, Spain;
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Lucía Méndez-Sánchez
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, 37007 Salamanca, Spain;
| | - Manuel Sánchez-Martín
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain; (E.V.); (S.B.); (J.M.H.-R.)
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, 37007 Salamanca, Spain;
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Correspondence: (M.S.-M.); (I.G.-T.)
| | - Ignacio García-Tuñón
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain; (E.V.); (S.B.); (J.M.H.-R.)
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), 37007 Salamanca, Spain;
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Correspondence: (M.S.-M.); (I.G.-T.)
| |
Collapse
|
11
|
Mojtahedi H, Yazdanpanah N, Rezaei N. Chronic myeloid leukemia stem cells: targeting therapeutic implications. Stem Cell Res Ther 2021; 12:603. [PMID: 34922630 PMCID: PMC8684082 DOI: 10.1186/s13287-021-02659-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/06/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm driven by BCR-ABL1 oncoprotein, which plays a pivotal role in CML pathology, diagnosis, and treatment as confirmed by the success of tyrosine kinase inhibitor (TKI) therapy. Despite advances in the development of more potent tyrosine kinase inhibitors, some mechanisms particularly in terms of CML leukemic stem cell (CML LSC) lead to intrinsic or acquired therapy resistance, relapse, and disease progression. In fact, the maintenance CML LSCs in patients who are resistance to TKI therapy indicates the role of CML LSCs in resistance to therapy through survival mechanisms that are not completely dependent on BCR-ABL activity. Targeting therapeutic approaches aim to eradicate CML LSCs through characterization and targeting genetic alteration and molecular pathways involving in CML LSC survival in a favorable leukemic microenvironment and resistance to apoptosis, with the hope of providing a functional cure. In other words, it is possible to develop the combination therapy of TKs with drugs targeting genes or molecules more specifically, which is required for survival mechanisms of CML LSCs, while sparing normal HSCs for clinical benefits along with TKIs.
Collapse
Affiliation(s)
- Hanieh Mojtahedi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Selvaraj D, Dawar R, Sivakumar PK, Devi A. Clustered regularly interspaced short palindromic repeats, a glimpse - impacts in molecular biology, trends and highlights. Horm Mol Biol Clin Investig 2021; 43:105-112. [PMID: 34881529 DOI: 10.1515/hmbci-2021-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/10/2021] [Indexed: 11/15/2022]
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) is a novel molecular tool. In recent days, it has been highlighted a lot, as the Nobel prize was awarded for this sector in 2020, and also for its recent use in Covid-19 related diagnostics. Otherwise, it is an eminent gene-editing technique applied in diverse medical zones of therapeutics in genetic diseases, hematological diseases, infectious diseases, etc., research related to molecular biology, cancer, hereditary diseases, immune and inflammatory diseases, etc., diagnostics related to infectious diseases like viral hemorrhagic fevers, Covid-19, etc. In this review, its discovery, working mechanisms, challenges while handling the technique, recent advancements, applications, alternatives have been discussed. It is a cheaper, faster technique revolutionizing the medicinal field right now. However, their off-target effects and difficulties in delivery into the desired cells make CRISPR, not easily utilizable. We conclude that further robust research in this field may promise many interesting, useful results.
Collapse
Affiliation(s)
- Dhivya Selvaraj
- Department of Biochemistry, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.,Department of Biochemistry, SGT University, Gurgaon, India
| | - Rajni Dawar
- Department of Biochemistry, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | | | - Anita Devi
- Department of Biochemistry, Dr Rajendra Prasad Government Medical College, Tanda, Kangra, India
| |
Collapse
|
13
|
Vuelta E, Ordoñez JL, Alonso-Pérez V, Méndez L, Hernández-Carabias P, Saldaña R, Sevilla J, Sebastián E, Muntión S, Sánchez-Guijo F, Hernández-Rivas JM, García-Tuñón I, Sánchez-Martín M. CRISPR-Cas9 Technology as a Tool to Target Gene Drivers in Cancer: Proof of Concept and New Opportunities to Treat Chronic Myeloid Leukemia. CRISPR J 2021; 4:519-535. [PMID: 34406033 DOI: 10.1089/crispr.2021.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a hematopoietic malignancy produced by a unique oncogenic event involving the constitutively active tyrosine-kinase (TK) BCR/ABL1. TK inhibitors (TKI) changed its prognosis and natural history. Unfortunately, ABL1 remains unaffected by TKIs. Leukemic stem cells (LSCs) remain, and resistant mutations arise during treatment. To address this problem, we have designed a therapeutic CRISPR-Cas9 deletion system targeting BCR/ABL1. The system was efficiently electroporated to cell lines, LSCs from a CML murine model, and LSCs from CML patients at diagnosis, generating a specific ABL1 null mutation at high efficiency and allowing the edited leukemic cells to be detected and tracked. The CRISPR-Cas9 deletion system triggered cell proliferation arrest and apoptosis in murine and human CML cell lines. Patient and murine-derived xenografts with CRISPR-edited LSCs in NOD SCID gamma niches revealed that normal multipotency and repopulation ability of CRISPR edited LSCs were fully restored. Normal hematopoiesis was restored, avoiding myeloid bias. To the best of our knowledge, we show for the first time how a CRISPR-Cas9 deletion system efficiently interrupts BCR/ABL1 oncogene in primary LSCs to bestow a therapeutic benefit. This study is a proof of concept for genome editing in all those diseases, like CML, sustained by a single oncogenic event, opening up new therapeutic opportunities.
Collapse
Affiliation(s)
- Elena Vuelta
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - José Luis Ordoñez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Verónica Alonso-Pérez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Lucía Méndez
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Patricia Hernández-Carabias
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Raquel Saldaña
- Servicio de Hematología, Hospital de Jerez, Cádiz, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Julián Sevilla
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Elena Sebastián
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Sandra Muntión
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- RETIC TerCel y CIBERONC, ISCIII, Madrid, Spain; and Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- RETIC TerCel y CIBERONC, ISCIII, Madrid, Spain; and Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ignacio García-Tuñón
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Manuel Sánchez-Martín
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| |
Collapse
|
14
|
Singh A, Bhatia P. Effective Downregulation of BCR-ABL Tumorigenicity by RNA Targeted CRISPR- Cas13a. Curr Gene Ther 2021; 21:270-277. [PMID: 33596804 DOI: 10.2174/1566523221666210217155233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/13/2020] [Accepted: 10/28/2020] [Indexed: 11/22/2022]
Abstract
AIM To induce BCR-ABL gene silencing using CRISPR Cas13a. BACKGROUND CML is a clonal myeloproliferative disorder of pluripotent stem cells driven by a reciprocal translocation between chromosomes 9 and 22 forming a BCR-ABL fusion gene. Tyrosine- kinase inhibitor drugs like imatinib are the mainstay of treatment and cases resistant to these drugs have a poor prognosis in the absence of a compatible stem-cell donor. However with rapid advancements in gene-editing technologies most studies are now focusing on developing a translational model targeting single-gene disorders with a prospective permanent cure. OBJECTIVE To explore the potential application of the RNA targeting CRISPR-Cas13a system for effective knockdown of BCR-ABL fusion transcript in a CML cell line K562. METHODS CRISPR Cas13a crRNA was designed specific to the chimeric BCR-ABL gene and the system was transfected as a two-plasmid system into a CML cell line K562. The effects were enumerated by evaluating the expression levels of downstream genes dependent on the expression of the BCR-ABL gene. Also next-generation sequencing was used to ascertain the effects of CRISPR on the gene. RESULTS The CRISPR system was successfully able to lower the expression of downstream genes [pCRKL and pCRK] dependent on the activated BCR-ABL kinase signal by up-to 4.3 folds. The viability of the CRISPR-treated cells was also significantly lowered by 373.83-fold [p-value= 0.000891196]. The time-dependent kinetics also highlighted the significant in-vitro suppressive activity to last up to 8 weeks [p-value: 0.025]. As per the cDNA sequencing data from the Oxford MinION next-generation sequencer the CRISPR treated cells show 62.37% suspected cleaved reads. CONCLUSION These preliminary results highlight an excellent potential application of RNA targeting CRISPRs in Haematological neoplasms like CML and should pave the way for further research in this direction.
Collapse
Affiliation(s)
- Aditya Singh
- Pediatric Hematology-Oncology Unit, Department of Pediatrics, Advanced Pediatric Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Pediatric Hematology-Oncology Unit, Department of Pediatrics, Advanced Pediatric Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
15
|
Kmiec EB, Bloh K. A toolmaker's perspective on CRISPR-directed gene editing as a therapeutic strategy for leukemia and beyond. Expert Rev Hematol 2021; 14:587-592. [PMID: 34047246 DOI: 10.1080/17474086.2021.1935853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Eric B Kmiec
- Gene Editing Institute, Helen F. Graham Cancer Center and Research Institute, Newark, USA
| | - Kevin Bloh
- Gene Editing Institute, Helen F. Graham Cancer Center and Research Institute, Newark, USA.,University of Delaware, Department of Medical and Molecular Sciences, College of Health Sciences, Newark, USA
| |
Collapse
|
16
|
Future Approaches for Treating Chronic Myeloid Leukemia: CRISPR Therapy. BIOLOGY 2021; 10:biology10020118. [PMID: 33557401 PMCID: PMC7915349 DOI: 10.3390/biology10020118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary In the last two decades, the therapeutic landscape of several tumors have changed profoundly with the introduction of drugs against proteins encoded by oncogenes. Oncogenes play an essential role in human cancer and when their encoded proteins are inhibited by specific drugs, the tumoral process can be reverted or stopped. An example of this is the case of the chronic myeloid leukemia, in which all the pathological features can be attributed by a single oncogene. Most patients with this disease now have a normal life expectancy thanks to a rationality designed inhibitor. However, the drug only blocks the protein, the oncogene continues unaffected and treatment discontinuation is only an option for a small subset of patients. With the advent of genome-editing nucleases and, especially, the CRISPR/Cas9 system, the possibilities to destroy oncogenes now is feasible. A novel therapeutic tool has been developed with unimaginable limits in cancer treatment. Recent studies support that CRISPR/Cas9 system could be a definitive therapeutic option in chronic myeloid leukemia. This work reviews the biology of chronic myeloid leukemia, the emergence of the CRISPR system, and its ability as a specific tool for this disease. Abstract The constitutively active tyrosine-kinase BCR/ABL1 oncogene plays a key role in human chronic myeloid leukemia development and disease maintenance, and determines most of the features of this leukemia. For this reason, tyrosine-kinase inhibitors are the first-line treatment, offering most patients a life expectancy like that of an equivalent healthy person. However, since the oncogene stays intact, lifelong oral medication is essential, even though this triggers adverse effects in many patients. Furthermore, leukemic stem cells remain quiescent and resistance is observed in approximately 25% of patients. Thus, new therapeutic alternatives are still needed. In this scenario, the interruption/deletion of the oncogenic sequence might be an effective therapeutic option. The emergence of CRISPR (clustered regularly interspaced short palindromic repeats) technology can offer a definitive treatment based on its capacity to induce a specific DNA double strand break. Besides, it has the advantage of providing complete and permanent oncogene knockout, while tyrosine kinase inhibitors (TKIs) only ensure that BCR-ABL1 oncoprotein is inactivated during treatment. CRISPR/Cas9 cuts DNA in a sequence-specific manner making it possible to turn oncogenes off in a way that was not previously feasible in humans. This review describes chronic myeloid leukemia (CML) disease and the main advances in the genome-editing field by which it may be treated in the future.
Collapse
|
17
|
Functional Comparison between VP64-dCas9-VP64 and dCas9-VP192 CRISPR Activators in Human Embryonic Kidney Cells. Int J Mol Sci 2021; 22:ijms22010397. [PMID: 33401508 PMCID: PMC7795359 DOI: 10.3390/ijms22010397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Reversal in the transcriptional status of desired genes has been exploited for multiple research, therapeutic, and biotechnological purposes. CRISPR/dCas9-based activators can activate transcriptionally silenced genes after being guided by gene-specific gRNA(s). Here, we performed a functional comparison between two such activators, VP64-dCas9-VP64 and dCas9-VP192, in human embryonic kidney cells by the concomitant targeting of POU5F1 and SOX2. We found 22- and 6-fold upregulations in the mRNA level of POU5F1 by dCas9-VP192 and VP64-dCas9-VP64, respectively. Likewise, SOX2 was up-regulated 4- and 2-fold using dCas9-VP192 and VP64dCas9VP64, respectively. For the POU5F1 protein level, we observed 3.7- and 2.2-fold increases with dCas9-VP192 and VP64-dCas9-VP64, respectively. Similarly, the SOX2 expression was 2.4- and 2-fold higher with dCas9-VP192 and VP64-dCas9-VP64, respectively. We also confirmed that activation only happened upon co-transfecting an activator plasmid with multiplex gRNA plasmid with a high specificity to the reference genes. Our data revealed that dCas9-VP192 is more efficient than VP64-dCas9-VP64 for activating reference genes.
Collapse
|
18
|
Chen SH, Hsieh YY, Tzeng HE, Lin CY, Hsu KW, Chiang YS, Lin SM, Su MJ, Hsieh WS, Lee CH. ABL Genomic Editing Sufficiently Abolishes Oncogenesis of Human Chronic Myeloid Leukemia Cells In Vitro and In Vivo. Cancers (Basel) 2020; 12:E1399. [PMID: 32485885 PMCID: PMC7352505 DOI: 10.3390/cancers12061399] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is the most common type of leukemia in adults, and more than 90% of CML patients harbor the abnormal Philadelphia chromosome (Ph) that encodes the BCR-ABL oncoprotein. Although the ABL kinase inhibitor (imatinib) has proven to be very effective in achieving high remission rates and improving prognosis, up to 33% of CML patients still cannot achieve an optimal response. Here, we used CRISPR/Cas9 to specifically target the BCR-ABL junction region in K562 cells, resulting in the inhibition of cancer cell growth and oncogenesis. Due to the variety of BCR-ABL junctions in CML patients, we utilized gene editing of the human ABL gene for clinical applications. Using the ABL gene-edited virus in K562 cells, we detected 41.2% indels in ABL sgRNA_2-infected cells. The ABL-edited cells reveled significant suppression of BCR-ABL protein expression and downstream signals, inhibiting cell growth and increasing cell apoptosis. Next, we introduced the ABL gene-edited virus into a systemic K562 leukemia xenograft mouse model, and bioluminescence imaging of the mice showed a significant reduction in the leukemia cell population in ABL-targeted mice, compared to the scramble sgRNA virus-injected mice. In CML cells from clinical samples, infection with the ABL gene-edited virus resulted in more than 30.9% indels and significant cancer cell death. Notably, no off-target effects or bone marrow cell suppression was found using the ABL gene-edited virus, ensuring both user safety and treatment efficacy. This study demonstrated the critical role of the ABL gene in maintaining CML cell survival and tumorigenicity in vitro and in vivo. ABL gene editing-based therapy might provide a potential strategy for imatinib-insensitive or resistant CML patients.
Collapse
Affiliation(s)
- Shu-Huey Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Huey-En Tzeng
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Division of Hematology/Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chun-Yu Lin
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30068, Taiwan;
- Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu 30068, Taiwan
| | - Kai-Wen Hsu
- Institute of New Drug Development, China Medical University, Taichung City 40402, Taiwan;
- Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan
| | - Yun-Shan Chiang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Su-Mei Lin
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan;
| | - Ming-Jang Su
- Department of Clinical Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Department of Family Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Wen-Shyang Hsieh
- Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
| | - Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei 11031, Taiwan
| |
Collapse
|
19
|
ABL Genomic Editing Sufficiently Abolishes Oncogenesis of Human Chronic Myeloid Leukemia Cells In Vitro and In Vivo. Cancers (Basel) 2020. [PMID: 32485885 DOI: 10.3390/cancers12061399.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is the most common type of leukemia in adults, and more than 90% of CML patients harbor the abnormal Philadelphia chromosome (Ph) that encodes the BCR-ABL oncoprotein. Although the ABL kinase inhibitor (imatinib) has proven to be very effective in achieving high remission rates and improving prognosis, up to 33% of CML patients still cannot achieve an optimal response. Here, we used CRISPR/Cas9 to specifically target the BCR-ABL junction region in K562 cells, resulting in the inhibition of cancer cell growth and oncogenesis. Due to the variety of BCR-ABL junctions in CML patients, we utilized gene editing of the human ABL gene for clinical applications. Using the ABL gene-edited virus in K562 cells, we detected 41.2% indels in ABL sgRNA_2-infected cells. The ABL-edited cells reveled significant suppression of BCR-ABL protein expression and downstream signals, inhibiting cell growth and increasing cell apoptosis. Next, we introduced the ABL gene-edited virus into a systemic K562 leukemia xenograft mouse model, and bioluminescence imaging of the mice showed a significant reduction in the leukemia cell population in ABL-targeted mice, compared to the scramble sgRNA virus-injected mice. In CML cells from clinical samples, infection with the ABL gene-edited virus resulted in more than 30.9% indels and significant cancer cell death. Notably, no off-target effects or bone marrow cell suppression was found using the ABL gene-edited virus, ensuring both user safety and treatment efficacy. This study demonstrated the critical role of the ABL gene in maintaining CML cell survival and tumorigenicity in vitro and in vivo. ABL gene editing-based therapy might provide a potential strategy for imatinib-insensitive or resistant CML patients.
Collapse
|
20
|
CRISPR/Cas9-generated models uncover therapeutic vulnerabilities of del(11q) CLL cells to dual BCR and PARP inhibition. Leukemia 2020; 34:1599-1612. [PMID: 31974435 PMCID: PMC7266745 DOI: 10.1038/s41375-020-0714-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/10/2019] [Accepted: 01/14/2020] [Indexed: 12/24/2022]
Abstract
The deletion of 11q (del(11q)) invariably comprises ATM gene in chronic lymphocytic leukemia (CLL). Concomitant mutations in this gene in the remaining allele have been identified in 1/3 of CLL cases harboring del(11q), being the biallelic loss of ATM associated with adverse prognosis. Although the introduction of targeted BCR inhibition has significantly favored the outcomes of del(11q) patients, responses of patients harboring ATM functional loss through biallelic inactivation are unexplored, and the development of resistances to targeted therapies have been increasingly reported, urging the need to explore novel therapeutic approaches. Here, we generated isogenic CLL cell lines harboring del(11q) and ATM mutations through CRISPR/Cas9-based gene-editing. With these models, we uncovered a novel therapeutic vulnerability of del(11q)/ATM-mutated cells to dual BCR and PARP inhibition. Ex vivo studies in the presence of stromal stimulation on 38 CLL primary samples confirmed a synergistic action of the combination of olaparib and ibrutinib in del(11q)/ATM-mutated CLL patients. In addition, we showed that ibrutinib produced a homologous recombination repair impairment through RAD51 dysregulation, finding a synergistic link of both drugs in the DNA damage repair pathway. Our data provide a preclinical rationale for the use of this combination in CLL patients with this high-risk cytogenetic abnormality.
Collapse
|
21
|
Peretti D, Kim S, Tufi R, Lev S. Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer. Front Cell Dev Biol 2020; 7:371. [PMID: 32039198 PMCID: PMC6989408 DOI: 10.3389/fcell.2019.00371] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/16/2019] [Indexed: 11/29/2022] Open
Abstract
Lipid-transfer proteins (LTPs) were initially discovered as cytosolic factors that facilitate lipid transport between membrane bilayers in vitro. Since then, many LTPs have been isolated from bacteria, plants, yeast, and mammals, and extensively studied in cell-free systems and intact cells. A major advance in the LTP field was associated with the discovery of intracellular membrane contact sites (MCSs), small cytosolic gaps between the endoplasmic reticulum (ER) and other cellular membranes, which accelerate lipid transfer by LTPs. As LTPs modulate the distribution of lipids within cellular membranes, and many lipid species function as second messengers in key signaling pathways that control cell survival, proliferation, and migration, LTPs have been implicated in cancer-associated signal transduction cascades. Increasing evidence suggests that LTPs play an important role in cancer progression and metastasis. This review describes how different LTPs as well as MCSs can contribute to cell transformation and malignant phenotype, and discusses how “aberrant” MCSs are associated with tumorigenesis in human.
Collapse
Affiliation(s)
- Diego Peretti
- UK Dementia Research Institute, Clinical Neurosciences Department, University of Cambridge, Cambridge, United Kingdom
| | - SoHui Kim
- Nakseongdae R&D Center, GPCR Therapeutics, Inc., Seoul, South Korea
| | - Roberta Tufi
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
22
|
Montaño A, Ordoñez JL, Alonso-Pérez V, Hernández-Sánchez J, Santos S, González T, Benito R, García-Tuñón I, Hernández-Rivas JM. ETV6/ RUNX1 Fusion Gene Abrogation Decreases the Oncogenicity of Tumour Cells in a Preclinical Model of Acute Lymphoblastic Leukaemia. Cells 2020; 9:E215. [PMID: 31952221 PMCID: PMC7017301 DOI: 10.3390/cells9010215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The t(12;21)(p13;q22), which fuses ETV6 and RUNX1 genes, is the most common genetic abnormality in children with B-cell precursor acute lymphoblastic leukaemia. The implication of the fusion protein in leukemogenesis seems to be clear. However, its role in the maintenance of the disease continues to be controversial. METHODS Generation of an in vitroETV6/RUNX1 knock out model using the CRISPR/Cas9 gene editing system. Functional characterization by RNA sequencing, proliferation assays, apoptosis and pharmacologic studies, and generation of edited-cell xenograft model. RESULTS The expression of ETV6/RUNX1 fusion gene was completely eliminated, thus generating a powerful model on which to study the role of the fusion gene in leukemic cells. The loss of fusion gene expression led to the deregulation of biological processes affecting survival such as apoptosis resistance and cell proliferation capacity. Tumour cells showed higher levels of apoptosis, lower proliferation rate and a greater sensitivity to PI3K inhibitors in vitro along as a decrease in tumour growth in xenografts models after ETV6/RUNX1 fusion gene abrogation. CONCLUSIONS ETV6/RUNX1 fusion protein seems to play an important role in the maintenance of the leukemic phenotype and could thus become a potential therapeutic target.
Collapse
Affiliation(s)
- Adrián Montaño
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jose Luis Ordoñez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Biochemistry and Molecular Biology, University of Salamanca, Campus Unamuno s/n, 37007 Salamanca, Spain
| | - Verónica Alonso-Pérez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jesús Hernández-Sánchez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Sandra Santos
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Teresa González
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Hematology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Rocío Benito
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Ignacio García-Tuñón
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jesús María Hernández-Rivas
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Hematology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Department of Medicine, Universidad de Salamanca and CIBERONC, 37007 Salamanca, Spain
| |
Collapse
|
23
|
Advances in Sphingolipidoses: CRISPR-Cas9 Editing as an Option for Modelling and Therapy. Int J Mol Sci 2019; 20:ijms20235897. [PMID: 31771289 PMCID: PMC6928934 DOI: 10.3390/ijms20235897] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/04/2023] Open
Abstract
Sphingolipidoses are inherited genetic diseases characterized by the accumulation of glycosphingolipids. Sphingolipidoses (SP), which usually involve the loss of sphingolipid hydrolase function, are of lysosomal origin, and represent an important group of rare diseases among lysosomal storage disorders. Initial treatments consisted of enzyme replacement therapy, but, in recent decades, various therapeutic approaches have been developed. However, these commonly used treatments for SP fail to be fully effective and do not penetrate the blood-brain barrier. New approaches, such as genome editing, have great potential for both the treatment and study of sphingolipidoses. Here, we review the most recent advances in the treatment and modelling of SP through the application of CRISPR-Cas9 genome editing. CRISPR-Cas9 is currently the most widely used method for genome editing. This technique is versatile; it can be used for altering the regulation of genes involved in sphingolipid degradation and synthesis pathways, interrogating gene function, generating knock out models, or knocking in mutations. CRISPR-Cas9 genome editing is being used as an approach to disease treatment, but more frequently it is utilized to create models of disease. New CRISPR-Cas9-based tools of gene editing with diminished off-targeting effects are evolving and seem to be more promising for the correction of individual mutations. Emerging Prime results and CRISPR-Cas9 difficulties are also discussed.
Collapse
|
24
|
Jiang C, Meng L, Yang B, Luo X. Application of CRISPR/Cas9 gene editing technique in the study of cancer treatment. Clin Genet 2019; 97:73-88. [PMID: 31231788 DOI: 10.1111/cge.13589] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
In recent years, gene editing, especially that using clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9, has made great progress in the field of gene function. Rapid development of gene editing techniques has contributed to their significance in the field of medicine. Because the CRISPR/Cas9 gene editing tool is not only powerful but also has features such as strong specificity and high efficiency, it can accurately and rapidly screen the whole genome, facilitating the administration of gene therapy for specific diseases. In the field of tumor research, CRISPR/Cas9 can be used to edit genomes to explore the mechanisms of tumor occurrence, development, and metastasis. In these years, this system has been increasingly applied in tumor treatment research. CRISPR/Cas9 can be used to treat tumors by repairing mutations or knocking out specific genes. To date, numerous preliminary studies have been conducted on tumor treatment in related fields. CRISPR/Cas9 holds great promise for gene-level tumor treatment. Personalized and targeted therapy based on CRISPR/Cas9 will possibly shape the development of tumor therapy in the future. In this study, we review the findings of CRISPR/Cas9 for tumor treatment research to provide references for related future studies on the pathogenesis and clinical treatment of tumors.
Collapse
Affiliation(s)
- Chunyang Jiang
- Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Lingxiang Meng
- Department of Anorectal Surgery, Anorectal Surgery Center, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Bingjun Yang
- Department of Thoracic Surgery, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Xin Luo
- Department of Radiotherapy, The Second Hospital of PingLiang City, Second Affiliated Hospital of Gansu Medical College, PingLiang, People's Republic of China
| |
Collapse
|
25
|
Luo Z, Gao M, Huang N, Wang X, Yang Z, Yang H, Huang Z, Feng W. Efficient disruption of bcr-abl gene by CRISPR RNA-guided FokI nucleases depresses the oncogenesis of chronic myeloid leukemia cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:224. [PMID: 31138265 PMCID: PMC6537404 DOI: 10.1186/s13046-019-1229-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
Background The bcr-abl fusion gene encodes BCR-ABL oncoprotein and plays a crucial role in the leukemogenesis of chronic myeloid leukemia (CML). Current therapeutic methods have limited treatment effect on CML patients with drug resistance or disease relapse. Therefore, novel therapeutic strategy for CML is essential to be explored and the CRISPR RNA-guided FokI nucleases (RFNs) meet the merits of variable target sites and specificity of cleavage enabled its suitability for gene editing of CML. The RFNs provide us a new therapeutic direction to obliterate this disease. Methods Guide RNA (gRNA) expression plasmids were constructed by molecular cloning technique. The modification rate of RFNs on bcr-abl was detected via NotI restriction enzyme digestion and T7 endonuclease 1 (T7E1) assay. The expression of BCR-ABL and its downstream signaling molecules were determined by western blotting. The effects of RFNs on cell proliferation and apoptosis of CML cell lines and CML stem/progenitor cells were evaluated by CCK-8 assay and flow cytometry. In addition, murine xenograft model was adopted to evaluate the capacity of RFNs in attenuating the tumorigenic ability of bcr-abl. Results The RFNs efficiently disrupted bcr-abl and prematurely terminated its translation. The destruction of bcr-abl gene suppressed cell proliferation and induced cell apoptosis in CML lines and in CML stem/progenitor cells. Moreover, the RFNs significantly impaired the leukemogenic capacity of CML cells in xenograft model. Conclusion These results illustrate that the RFNs can target to disrupt bcr-abl gene and may provide a new therapeutic option for CML patients affiliated by drug resistance or disease relapse. Electronic supplementary material The online version of this article (10.1186/s13046-019-1229-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenhong Luo
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Miao Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ningshu Huang
- Department of Clinical Laboratory, The Children's Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hao Yang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Zhenglan Huang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China.
| | - Wenli Feng
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China.
| |
Collapse
|
26
|
Luo Z, Gao M, Huang N, Wang X, Yang Z, Yang H, Huang Z, Feng W. Efficient disruption of bcr-abl gene by CRISPR RNA-guided FokI nucleases depresses the oncogenesis of chronic myeloid leukemia cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019. [PMID: 31138265 DOI: 10.1186/s13046-019-1229-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The bcr-abl fusion gene encodes BCR-ABL oncoprotein and plays a crucial role in the leukemogenesis of chronic myeloid leukemia (CML). Current therapeutic methods have limited treatment effect on CML patients with drug resistance or disease relapse. Therefore, novel therapeutic strategy for CML is essential to be explored and the CRISPR RNA-guided FokI nucleases (RFNs) meet the merits of variable target sites and specificity of cleavage enabled its suitability for gene editing of CML. The RFNs provide us a new therapeutic direction to obliterate this disease. METHODS Guide RNA (gRNA) expression plasmids were constructed by molecular cloning technique. The modification rate of RFNs on bcr-abl was detected via NotI restriction enzyme digestion and T7 endonuclease 1 (T7E1) assay. The expression of BCR-ABL and its downstream signaling molecules were determined by western blotting. The effects of RFNs on cell proliferation and apoptosis of CML cell lines and CML stem/progenitor cells were evaluated by CCK-8 assay and flow cytometry. In addition, murine xenograft model was adopted to evaluate the capacity of RFNs in attenuating the tumorigenic ability of bcr-abl. RESULTS The RFNs efficiently disrupted bcr-abl and prematurely terminated its translation. The destruction of bcr-abl gene suppressed cell proliferation and induced cell apoptosis in CML lines and in CML stem/progenitor cells. Moreover, the RFNs significantly impaired the leukemogenic capacity of CML cells in xenograft model. CONCLUSION These results illustrate that the RFNs can target to disrupt bcr-abl gene and may provide a new therapeutic option for CML patients affiliated by drug resistance or disease relapse.
Collapse
Affiliation(s)
- Zhenhong Luo
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Miao Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ningshu Huang
- Department of Clinical Laboratory, The Children's Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hao Yang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Zhenglan Huang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China.
| | - Wenli Feng
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016, China.
| |
Collapse
|
27
|
Abstract
CRISPR/Cas9 allows the generation of knockout cell lines and null zygotes by inducing site-specific double-stranded breaks. In most cases the DSB is repaired by non-homologous end joining, resulting in small nucleotide insertions or deletions that can be used to construct knockout alleles. However, these mutations do not produce the desired null result in all cases, but instead generate a similar, functionally active protein. This effect could limit the therapeutic efficiency of gene therapy strategies based on abrogating oncogene expression, and therefore needs to be considered carefully. If there is an acceptable degree of efficiency of CRISPR/Cas9 delivery to cells, the key step for success lies in the effectiveness of a specific sgRNA at knocking out the oncogene, when only one sgRNA can be used. This study shows that the null effect could be increased with an sgRNA targeting the splice donor site (SDS) of the chosen exon. Following this strategy, the generation of null alleles would be facilitated in two independent ways: the probability of producing a frameshift mutation and the probability of interrupting the canonical mechanism of pre-mRNA splicing. In these contexts, we propose to improve the loss-of-function yield driving the CRISPR system at the SDS of critical exons.
Collapse
|
28
|
García-Tuñón I, Alonso-Pérez V, Vuelta E, Pérez- Ramos S, Herrero M, Méndez L, Hernández-Sánchez JM, Martín-Izquierdo M, Saldaña R, Sevilla J, Sánchez- Guijo F, Hernández-Rivas JM, Sánchez-Martín M. Splice donor site sgRNAs enhance CRISPR/Cas9-mediated knockout efficiency. PLoS One 2019; 14:e0216674. [PMID: 31071190 PMCID: PMC6508695 DOI: 10.1371/journal.pone.0216674] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/26/2019] [Indexed: 01/08/2023] Open
Abstract
CRISPR/Cas9 allows the generation of knockout cell lines and null zygotes by inducing site-specific double-stranded breaks. In most cases the DSB is repaired by non-homologous end joining, resulting in small nucleotide insertions or deletions that can be used to construct knockout alleles. However, these mutations do not produce the desired null result in all cases, but instead generate a similar, functionally active protein. This effect could limit the therapeutic efficiency of gene therapy strategies based on abrogating oncogene expression, and therefore needs to be considered carefully. If there is an acceptable degree of efficiency of CRISPR/Cas9 delivery to cells, the key step for success lies in the effectiveness of a specific sgRNA at knocking out the oncogene, when only one sgRNA can be used. This study shows that the null effect could be increased with an sgRNA targeting the splice donor site (SDS) of the chosen exon. Following this strategy, the generation of null alleles would be facilitated in two independent ways: the probability of producing a frameshift mutation and the probability of interrupting the canonical mechanism of pre-mRNA splicing. In these contexts, we propose to improve the loss-of-function yield driving the CRISPR system at the SDS of critical exons.
Collapse
Affiliation(s)
- Ignacio García-Tuñón
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Verónica Alonso-Pérez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Elena Vuelta
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
| | - Sandra Pérez- Ramos
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
| | - María Herrero
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
| | - Lucía Méndez
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Sánchez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Marta Martín-Izquierdo
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Raquel Saldaña
- Servicio de Hematología, Hospital de Especialidades de Jerez, Zacatecas, Spain
| | - Julián Sevilla
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Fermín Sánchez- Guijo
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Manuel Sánchez-Martín
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
29
|
Rodríguez-Rodríguez DR, Ramírez-Solís R, Garza-Elizondo MA, Garza-Rodríguez MDL, Barrera-Saldaña HA. Genome editing: A perspective on the application of CRISPR/Cas9 to study human diseases (Review). Int J Mol Med 2019; 43:1559-1574. [PMID: 30816503 PMCID: PMC6414166 DOI: 10.3892/ijmm.2019.4112] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
Genome editing reemerged in 2012 with the development of CRISPR/Cas9 technology, which is a genetic manipulation tool derived from the defense system of certain bacteria against viruses and plasmids. This method is easy to apply and has been used in a wide variety of experimental models, including cell lines, laboratory animals, plants, and even in human clinical trials. The CRISPR/Cas9 system consists of directing the Cas9 nuclease to create a site-directed double-strand DNA break using a small RNA molecule as a guide. A process that allows a permanent modification of the genomic target sequence can repair the damage caused to DNA. In the present study, the basic principles of the CRISPR/Cas9 system are reviewed, as well as the strategies and modifications of the enzyme Cas9 to eliminate the off-target cuts, and the different applications of CRISPR/Cas9 as a system for visualization and gene expression activation or suppression. In addition, the review emphasizes on the potential application of this system in the treatment of different diseases, such as pulmonary, gastrointestinal, hematologic, immune system, viral, autoimmune and inflammatory diseases, and cancer.
Collapse
Affiliation(s)
- Diana Raquel Rodríguez-Rodríguez
- Universidad Autónoma de Nuevo León, Department of Biochemistry and Molecular Medicine, School of Medicine and University Hospital 'Dr. José E. González', Monterrey, Nuevo León 64460, México
| | - Ramiro Ramírez-Solís
- Institutional Core Laboratories, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mario Alberto Garza-Elizondo
- Universidad Autónoma de Nuevo León, Service of Rheumatology, School of Medicine and University Hospital 'Dr. José E. González', Monterrey, Nuevo León 64460, México
| | - María De Lourdes Garza-Rodríguez
- Universidad Autónoma de Nuevo León, Department of Biochemistry and Molecular Medicine, School of Medicine and University Hospital 'Dr. José E. González', Monterrey, Nuevo León 64460, México
| | - Hugo Alberto Barrera-Saldaña
- Universidad Autónoma de Nuevo León, Department of Biochemistry and Molecular Medicine, School of Medicine and University Hospital 'Dr. José E. González', Monterrey, Nuevo León 64460, México
| |
Collapse
|
30
|
Rein LA, Yang H, Chao NJ. Applications of Gene Editing Technologies to Cellular Therapies. Biol Blood Marrow Transplant 2018; 24:1537-1545. [DOI: 10.1016/j.bbmt.2018.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/23/2018] [Indexed: 12/26/2022]
|
31
|
Estêvão D, Rios Costa N, da Costa RG, Medeiros R. CRISPR-Cas9 therapies in experimental mouse models of cancer. Future Oncol 2018; 14:2083-2095. [PMID: 30027767 DOI: 10.2217/fon-2018-0028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The CRISPR-Cas9, a part of the defence mechanism from bacteria, has rapidly become the simplest, fastest and the most precise genome-editing tool available. The therapeutic applications of CRISPR are boundless: correction of mutations in several disorders, inactivation of oncogenes and viral oncoproteins, and activation of tumor suppressor genes. In this review, we expose recent advances concerning animal models of cancer that use CRISPR-Cas9, addressing also the current efforts to develop CRISPR-Cas9-based therapies, focusing on proof-of-concept studies. Finally, the review exposes some of the main challenges that this genome-editing tool faces. The key issue remains: does CRISPR-Cas9 have real potential for therapeutic application or will it just remain a wonderful research tool?
Collapse
Affiliation(s)
- Diogo Estêvão
- Molecular Oncology & Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), 4200-319 Porto, Portugal Porto, Portugal
| | - Natália Rios Costa
- Molecular Oncology & Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal Porto, Portugal
| | - Rui Gil da Costa
- Laboratory for Process Engineering, Environment, Biotechnology & Energy (LEPABE), Faculty of Engineering of the University of Porto (FEUP), 4200-465 Porto, Portugal.,Center for the Research & Technology of Agro-Environmental & Biological Sciences (CITAB), University of Trás-os-Montes & Alto Douro (UTAD), 5001-911 Vila Real, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), 4200-319 Porto, Portugal Porto, Portugal.,Research Department of the Portuguese League Against Cancer (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), 4200-177 Porto, Portugal Porto, Portugal.,Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal Porto, Portugal
| |
Collapse
|
32
|
Montaño A, Forero-Castro M, Hernández-Rivas JM, García-Tuñón I, Benito R. Targeted genome editing in acute lymphoblastic leukemia: a review. BMC Biotechnol 2018; 18:45. [PMID: 30016959 PMCID: PMC6050675 DOI: 10.1186/s12896-018-0455-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022] Open
Abstract
Background Genome editing technologies offers new opportunities for tackling diseases such as acute lymphoblastic leukemia (ALL) that have been beyond the reach of previous therapies. Results We show how the recent availability of genome-editing tools such as CRISPR-Cas9 are an important means of advancing functional studies of ALL through the incorporation, elimination and modification of somatic mutations and fusion genes in cell lines and mouse models. These tools not only broaden the understanding of the involvement of various genetic alterations in the pathogenesis of the disease but also identify new therapeutic targets for future clinical trials. Conclusions New approaches including CRISPR-Cas9 are crucial for functional studies of genetic aberrations driving cancer progression, and that may be responsible for treatment resistance and relapses. By using this approach, diseases can be more faithfully reproduced and new therapeutic targets and approaches found.
Collapse
Affiliation(s)
- Adrián Montaño
- IBSAL, IBMCC, University of Salamanca-CSIC, Cancer Research Center, Salamanca, Spain
| | - Maribel Forero-Castro
- School of Biological Sciences (GICBUPTC Research group), Universidad Pedagógica y Tecnológica de Colombia, Boyacá, Colombia
| | - Jesús-María Hernández-Rivas
- IBSAL, IBMCC, University of Salamanca-CSIC, Cancer Research Center, Salamanca, Spain. .,Department of Medicine, University of Salamanca, Spain, Department of Hematology, University Hospital of Salamanca, Salamanca, Spain. .,IBMCC, CIC University of Salamanca-CSIC, University Hospital of Salamanca, Salamanca, Spain.
| | - Ignacio García-Tuñón
- IBSAL, IBMCC, University of Salamanca-CSIC, Cancer Research Center, Salamanca, Spain
| | - Rocío Benito
- IBSAL, IBMCC, University of Salamanca-CSIC, Cancer Research Center, Salamanca, Spain
| |
Collapse
|