1
|
Romero P, Richart L, Aflaki S, Petitalot A, Burton M, Michaud A, Masliah-Planchon J, Kuhnowski F, Le Cam S, Baliñas-Gavira C, Méaudre C, Luscan A, Hamza A, Legoix P, Vincent-Salomon A, Wassef M, Holoch D, Margueron R. EZH2 mutations in follicular lymphoma distort H3K27me3 profiles and alter transcriptional responses to PRC2 inhibition. Nat Commun 2024; 15:3452. [PMID: 38658543 PMCID: PMC11043461 DOI: 10.1038/s41467-024-47701-x] [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: 01/19/2023] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in chromatin regulators are widespread in cancer. Among them, the histone H3 lysine 27 methyltransferase Polycomb Repressive Complex 2 (PRC2) shows distinct alterations according to tumor type. This specificity is poorly understood. Here, we model several PRC2 alterations in one isogenic system to reveal their comparative effects. Focusing then on lymphoma-associated EZH2 mutations, we show that Ezh2Y641F induces aberrant H3K27 methylation patterns even without wild-type Ezh2, which are alleviated by partial PRC2 inhibition. Remarkably, Ezh2Y641F rewires the response to PRC2 inhibition, leading to induction of antigen presentation genes. Using a unique longitudinal follicular lymphoma cohort, we further link EZH2 status to abnormal H3K27 methylation. We also uncover unexpected variability in the mutational landscape of successive biopsies, pointing to frequent co-existence of different clones and cautioning against stratifying patients based on single sampling. Our results clarify how oncogenic PRC2 mutations disrupt chromatin and transcription, and the therapeutic vulnerabilities this creates.
Collapse
Affiliation(s)
- Pierre Romero
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
- Institut Curie, Department of Pathology, Paris Sciences et Lettres Research University, Paris, France
| | - Laia Richart
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Setareh Aflaki
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Ambre Petitalot
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Megan Burton
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Audrey Michaud
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Julien Masliah-Planchon
- Institut Curie, Pharmacogenetics Unit, Department of Genetics, Paris Sciences et Lettres Research University, Paris, France
| | - Frédérique Kuhnowski
- Institut Curie, Department of Clinical Hematology, Paris Sciences et Lettres Research University, Paris, France
| | - Samuel Le Cam
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Carlos Baliñas-Gavira
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Céline Méaudre
- Institut Curie, Department of Pathology, Paris Sciences et Lettres Research University, Paris, France
| | - Armelle Luscan
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Abderaouf Hamza
- Institut Curie, Pharmacogenetics Unit, Department of Genetics, Paris Sciences et Lettres Research University, Paris, France
| | - Patricia Legoix
- Institut Curie, Genomics of Excellence (ICGex) Platform, Paris Sciences et Lettres Research University, Paris, France
| | - Anne Vincent-Salomon
- Institut Curie, Department of Pathology, Paris Sciences et Lettres Research University, Paris, France
| | - Michel Wassef
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Daniel Holoch
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.
| | - Raphaël Margueron
- Institut Curie, INSERM U934/CNRS UMR 3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.
| |
Collapse
|
2
|
Pacot L, Sabbagh A, Sohier P, Hadjadj D, Ye M, Boland-Auge A, Bacq-Daian D, Laurendeau I, Briand-Suleau A, Deleuze JF, Margueron R, Vidaud M, Ferkal S, Parfait B, Vidaud D, Pasmant E, Wolkenstein P. Identification of potential common genetic modifiers of neurofibromas: a genome-wide association study in 1333 patients with neurofibromatosis type 1. Br J Dermatol 2024; 190:226-243. [PMID: 37831592 DOI: 10.1093/bjd/ljad390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is characterized by the highly variable and unpredictable development of benign peripheral nerve sheath tumours: cutaneous (cNFs), subcutaneous (scNFs) and plexiform (pNFs) neurofibromas. OBJECTIVES To identify neurofibroma modifier genes, in order to develop a database of patients with NF1. METHODS All patients were phenotypically evaluated by a medical practitioner using a standardized questionnaire and the causal NF1 variant identified. We enrolled 1333 patients with NF1 who were genotyped for > 7 million common variants. RESULTS A genome-wide association case-only study identified a significant association with 9q21.33 in the pNF phenotype in the discovery cohort. Twelve, three and four regions suggestive of association at the P ≤ 1 × 10-6 threshold were identified for pNFs, cNFs and scNFs, respectively. Evidence of replication was observed for 4, 2 and 6 loci, including 168 candidate modifier protein-coding genes. Among the candidate modifier genes, some were implicated in the RAS-mitogen-activated protein kinase pathway, cell-cycle control and myelination. Using an original CRISPR/Cas9-based functional assay, we confirmed GAS1 and SPRED2 as pNF and scNF candidate modifiers, as their inactivation specifically affected NF1-mutant Schwann cell growth. CONCLUSIONS Our study may shed new light on the pathogenesis of NF1-associated neurofibromas and will, hopefully, contribute to the development of personalized care for patients with this deleterious and life-threatening condition.
Collapse
Affiliation(s)
- Laurence Pacot
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Audrey Sabbagh
- UMR 261 MERIT, Institut de Recherche pour le Développement, UFR de Pharmacie de Paris, Université Paris Cité, Paris, France
| | - Pierre Sohier
- Service de Pathologie, Hôpital Cochin, AP-HP, Centre-Université Paris Cité, Paris, France
| | - Djihad Hadjadj
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Manuela Ye
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Anne Boland-Auge
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Delphine Bacq-Daian
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Ingrid Laurendeau
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Audrey Briand-Suleau
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Raphaël Margueron
- Institut Curie, INSERM U934/CNRS UMR3215, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France
| | - Michel Vidaud
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Salah Ferkal
- Department of Dermatology, Hôpital Henri Mondor, Assistance Publique-Hôpital Paris (AP-HP), Créteil, France
- INSERM, Clinical Investigation Center 1430, Referral Center of Neurofibromatosis, Hôpital Henri Mondor, AP-HP, Faculté de Santé Paris Est Créteil, Créteil, France
| | - Béatrice Parfait
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Dominique Vidaud
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Eric Pasmant
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP, Centre-Université Paris Cité, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Pierre Wolkenstein
- Department of Dermatology, Hôpital Henri Mondor, Assistance Publique-Hôpital Paris (AP-HP), Créteil, France
- INSERM, Clinical Investigation Center 1430, Referral Center of Neurofibromatosis, Hôpital Henri Mondor, AP-HP, Faculté de Santé Paris Est Créteil, Créteil, France
| |
Collapse
|
3
|
XIST loss impairs mammary stem cell differentiation and increases tumorigenicity through Mediator hyperactivation. Cell 2022; 185:2164-2183.e25. [PMID: 35597241 DOI: 10.1016/j.cell.2022.04.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/10/2022] [Accepted: 04/27/2022] [Indexed: 12/27/2022]
Abstract
X inactivation (XCI) is triggered by upregulation of XIST, which coats the chromosome in cis, promoting formation of a heterochromatic domain (Xi). XIST role beyond initiation of XCI is only beginning to be elucidated. Here, we demonstrate that XIST loss impairs differentiation of human mammary stem cells (MaSCs) and promotes emergence of highly tumorigenic and metastatic carcinomas. On the Xi, XIST deficiency triggers epigenetic changes and reactivation of genes overlapping Polycomb domains, including Mediator subunit MED14. MED14 overdosage results in increased Mediator levels and hyperactivation of the MaSC enhancer landscape and transcriptional program, making differentiation less favorable. We further demonstrate that loss of XIST and Xi transcriptional instability is common among human breast tumors of poor prognosis. We conclude that XIST is a gatekeeper of human mammary epithelium homeostasis, thus unveiling a paradigm in the control of somatic cell identity with potential consequences for our understanding of gender-specific malignancies.
Collapse
|
4
|
Kim B, Kim Y, Shin S, Lee ST, Cho JY, Lee KA. Application of CRISPR/Cas9-based mutant enrichment technique to improve the clinical sensitivity of plasma EGFR testing in patients with non-small cell lung cancer. Cancer Cell Int 2022; 22:82. [PMID: 35168603 PMCID: PMC8845274 DOI: 10.1186/s12935-022-02504-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Approximately 50%–60% of secondary resistance to primary EGFR- tyrosine kinase inhibitors (TKI) therapy is caused by acquired p.Thr790Met (T790M) mutation; however, highly fragmented, low-quantity circulating tumor DNA is an obstacle for detecting mutations. Therefore, more sensitive mutation detection techniques are required. Here, we report a new mutant enrichment technology, the CRISPR system combined with post-polymerase chain reaction (PCR) cell-free DNA (cfDNA) (CRISPR-CPPC) to detect the T790M mutation using droplet digital PCR (ddPCR) from cfDNA. Methods The CRISPR-CPPC process comprises the following three steps: (1) cfDNA PCR, (2) assembly of post-PCR cfDNA and CRISPR/CRISPR associated protein 9 complex, and (3) enrichment of the target DNA template. After CRISPR-CPPC, the target DNA was detected using ddPCR. We optimized and validated CRISPR-CPPC using reference cfDNA standards and cfDNA from patients with non-small cell lung cancer who underwent TKI therapy. We then compared the detection sensitivity of CRISPR-CPPC assay with the results of real-time PCR and those of ddPCR. Results CRISPR-CPPC aided detection of T790M with 93.9% sensitivity and 100% specificity. T790M mutant copies were sensitively detected achieving an approximately 13-fold increase in the detected allele frequency. Furthermore, positive rate of detecting a low T790M copy number (< 10 copies/mL) were 93.8% (15/16) and 43.8% (7/16) for CRISPR-CPPC assay and ddPCR, respectively. Conclusions CRISPR-CPPC is a useful mutant enrichment tool for the sensitive detection of target mutation. When tested in patients with progressive disease, the diagnostic performance of CRISPR-CPPC assay is exceptionally better than that of any other currently available methods. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02504-2.
Collapse
Affiliation(s)
- Boyeon Kim
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Yoonjung Kim
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jae Yong Cho
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Kyung-A Lee
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
5
|
Holoch D, Wassef M, Lövkvist C, Zielinski D, Aflaki S, Lombard B, Héry T, Loew D, Howard M, Margueron R. A cis-acting mechanism mediates transcriptional memory at Polycomb target genes in mammals. Nat Genet 2021; 53:1686-1697. [PMID: 34782763 DOI: 10.1038/s41588-021-00964-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/05/2021] [Indexed: 11/09/2022]
Abstract
Epigenetic inheritance of gene expression states enables a single genome to maintain distinct cellular identities. How histone modifications contribute to this process remains unclear. Using global chromatin perturbations and local, time-controlled modulation of transcription, we establish the existence of epigenetic memory of transcriptional activation for genes that can be silenced by the Polycomb group. This property emerges during cell differentiation and allows genes to be stably switched after a transient transcriptional stimulus. This transcriptional memory state at Polycomb targets operates in cis; however, rather than relying solely on read-and-write propagation of histone modifications, the memory is also linked to the strength of activating inputs opposing Polycomb proteins, and therefore varies with the cellular context. Our data and computational simulations suggest a model whereby transcriptional memory arises from double-negative feedback between Polycomb-mediated silencing and active transcription. Transcriptional memory at Polycomb targets thus depends not only on histone modifications but also on the gene-regulatory network and underlying identity of a cell.
Collapse
Affiliation(s)
- Daniel Holoch
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,INSERM U934/CNRS UMR 3215, Paris, France
| | - Michel Wassef
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,INSERM U934/CNRS UMR 3215, Paris, France
| | - Cecilia Lövkvist
- John Innes Centre, Norwich Research Park, Norwich, UK. .,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Dina Zielinski
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,INSERM U934/CNRS UMR 3215, Paris, France.,INSERM U900, Mines ParisTech, Paris, France
| | - Setareh Aflaki
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,INSERM U934/CNRS UMR 3215, Paris, France
| | - Bérangère Lombard
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,Proteomics Mass Spectrometry Laboratory, Paris, France
| | - Tiphaine Héry
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,INSERM U934/CNRS UMR 3215, Paris, France
| | - Damarys Loew
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France.,Proteomics Mass Spectrometry Laboratory, Paris, France
| | - Martin Howard
- John Innes Centre, Norwich Research Park, Norwich, UK
| | - Raphaël Margueron
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, Paris, France. .,INSERM U934/CNRS UMR 3215, Paris, France.
| |
Collapse
|
6
|
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: 1.7] [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
|
7
|
Lam AJ, Lin DTS, Gillies JK, Uday P, Pesenacker AM, Kobor MS, Levings MK. Optimized CRISPR-mediated gene knockin reveals FOXP3-independent maintenance of human Treg identity. Cell Rep 2021; 36:109494. [PMID: 34348163 DOI: 10.1016/j.celrep.2021.109494] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
Regulatory T cell (Treg) therapy is a promising curative approach for a variety of immune-mediated conditions. CRISPR-based genome editing allows precise insertion of transgenes through homology-directed repair, but its use in human Tregs has been limited. We report an optimized protocol for CRISPR-mediated gene knockin in human Tregs with high-yield expansion. To establish a benchmark of human Treg dysfunction, we target the master transcription factor FOXP3 in naive and memory Tregs. Although FOXP3-ablated Tregs upregulate cytokine expression, effects on suppressive capacity in vitro manifest slowly and primarily in memory Tregs. Moreover, FOXP3-ablated Tregs retain their characteristic protein, transcriptional, and DNA methylation profile. Instead, FOXP3 maintains DNA methylation at regions enriched for AP-1 binding sites. Thus, although FOXP3 is important for human Treg development, it has a limited role in maintaining mature Treg identity. Optimized gene knockin with human Tregs will enable mechanistic studies and the development of tailored, next-generation Treg cell therapies.
Collapse
Affiliation(s)
- Avery J Lam
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - David T S Lin
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Jana K Gillies
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Prakruti Uday
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Anne M Pesenacker
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Michael S Kobor
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
8
|
Flow Assisted Mutation Enrichment (FAME): A highly efficacious and efficient method to enrich Double Knockouts (DKO) after gene editing. PLoS One 2021; 16:e0247375. [PMID: 33661977 PMCID: PMC7932066 DOI: 10.1371/journal.pone.0247375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 02/05/2021] [Indexed: 11/19/2022] Open
Abstract
Gene editing has become an essential tool for interrogation of gene function in biomedical research and is also a promising approach for gene therapy. Despite recent progresses, the gene-editing procedure is still a tedious process involving manually isolating large number of single cell colonies to screen for desired mutations. For diploid eukaryotic cells, there is the additional challenge to inactivate both alleles for genes-of-interest, i.e., generating double knockouts (DKOs), for the desired phenotypes or therapeutic effects. In this report, we present a novel method based on Fluorescence Assisted Cell Sorting (FACS) to enrich for DKO cells, using a cell surface marker β2-microglobulin (B2M) as a basis for negative selection. This method significantly increased percentage of DKOs in isolated cells after gene editing, and in the meantime, significantly improve the efficiency of workflow by automating colony isolation. It would greatly facilitate future biomedical research including potential gene/cell therapies.
Collapse
|
9
|
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: 8] [Impact Index Per Article: 2.7] [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
|
10
|
Liu J, Ge Y, Wang N, Wen J, Wang W, Zeng B, Chen GL. A Simple and Efficient Method to Generate Gene-Knockout and Transgenic Cell Lines. DNA Cell Biol 2020; 40:239-246. [PMID: 33395371 DOI: 10.1089/dna.2020.6183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Knockout (KO) or exogenous expression of a gene of interest in cultured cells is one of the most important ways to study the function of the gene. Compared with transient transfection, stable cell lines possess great advantages such as excellent cell homogeneity and feasibility for long-term use. However, technical challenges in generating stable cell lines still exist in many laboratories using conventional techniques like limiting dilution or cloning cylinders. In this study we describe an optimized method to efficiently create stable cell lines for functional studies. This method was successfully used to generate a PIEZO1 gene-KO cell line with the CRISPR/Cas9 technology, and TRPC5/GCaMP6f-mCherry-coexpressing cell lines without antibiotic selection. Monoclonal cell lines can be obtained in 2-4 weeks after transfection. This method does not require any special equipment or consumables and can be conducted in all laboratories with general cell-culture facility.
Collapse
Affiliation(s)
- Jieyu Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yan Ge
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Na Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
11
|
López-Manzaneda S, Ojeda-Pérez I, Zabaleta N, García-Torralba A, Alberquilla O, Torres R, Sánchez-Domínguez R, Torella L, Olivier E, Mountford J, Ramírez JC, Bueren JA, González-Aseguinolaza G, Segovia JC. In Vitro and In Vivo Genetic Disease Modeling via NHEJ-Precise Deletions Using CRISPR-Cas9. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:426-437. [PMID: 33294491 PMCID: PMC7683234 DOI: 10.1016/j.omtm.2020.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/11/2020] [Indexed: 12/01/2022]
Abstract
The development of advanced gene and cell therapies for the treatment of genetic diseases requires reliable animal and cellular models to test their efficacy. Moreover, the availability of the target human primary cells of these therapies is reduced in many diseases. The development of endonucleases that can cut into specific sites of the cell genome, as well as the repair of the generated break by non-homologous end-joining, results in a variety of outcomes, insertions, deletions, and inversions that can induce the disruption of any specific gene. Among the many methods that have been developed for gene editing, CRISPR-Cas9 technology has become one of the most widely used endonuclease tools due to its easy design and its low cost. It has also been reported that the use of two guides, instead of just the one required, reduces the outcomes of non-homologous end joining mainly to the precise genomic sequences between the cutting sites of the guides used. We have explored this strategy to generate useful cellular and animal models. Different distances between the two guides have been tested (from 8 to 500 bp apart), and using the optimal range of 30–60 bp we have obtained a human primary cellular model of a genetic disease, pyruvate kinase deficiency, where the availability of the target cells is limited. We have also generated an in vivo model of glycolate oxidase (GO) deficiency, which is an enzyme involved in the glyoxylate metabolism following the same strategy. We demonstrate that the use of two-guide CRISPR-Cas9-induced non-homologous end joining is a feasible and useful tool for disease modeling, and it is most relevant to those diseases in which it is difficult to get the cells that will be genetically manipulated.
Collapse
Affiliation(s)
- Sergio López-Manzaneda
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Isabel Ojeda-Pérez
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | | | - Aída García-Torralba
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Omaira Alberquilla
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Raúl Torres
- Molecular Cytogenetics Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Rebeca Sánchez-Domínguez
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | | | - Emmanuel Olivier
- Scottish National Blood Transfusion Service and ICAMS, University of Glasgow, Glasgow, UK
| | - Joanne Mountford
- Scottish National Blood Transfusion Service and ICAMS, University of Glasgow, Glasgow, UK
| | | | - Juan A Bueren
- Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.,Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - Jose-Carlos Segovia
- Cell Differentiation and Cytometry Unit. Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Unidad Mixta de Terapias Avanzadas. Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| |
Collapse
|
12
|
Rondelet A, Pozniakovsky A, Namboodiri D, Cardoso da Silva R, Singh D, Leuschner M, Poser I, Ssykor A, Berlitz J, Schmidt N, Röhder L, Vader G, Hyman AA, Bird AW. ESI mutagenesis: a one-step method for introducing mutations into bacterial artificial chromosomes. Life Sci Alliance 2020; 4:4/2/e202000836. [PMID: 33293335 PMCID: PMC7756954 DOI: 10.26508/lsa.202000836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 01/23/2023] Open
Abstract
A simple and efficient recombineering-based method for introducing point mutations into bacterial artificial chromosomes using an artificial intron cassette. Bacterial artificial chromosome (BAC)–based transgenes have emerged as a powerful tool for controlled and conditional interrogation of protein function in higher eukaryotes. Although homologous recombination-based recombineering methods have streamlined the efficient integration of protein tags onto BAC transgenes, generating precise point mutations has remained less efficient and time-consuming. Here, we present a simplified method for inserting point mutations into BAC transgenes requiring a single recombineering step followed by antibiotic selection. This technique, which we call exogenous/synthetic intronization (ESI) mutagenesis, relies on co-integration of a mutation of interest along with a selectable marker gene, the latter of which is harboured in an artificial intron adjacent to the mutation site. Cell lines generated from ESI-mutated BACs express the transgenes equivalently to the endogenous gene, and all cells efficiently splice out the synthetic intron. Thus, ESI mutagenesis provides a robust and effective single-step method with high precision and high efficiency for mutating BAC transgenes.
Collapse
Affiliation(s)
- Arnaud Rondelet
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrei Pozniakovsky
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | - Divya Singh
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marit Leuschner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrea Ssykor
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Julian Berlitz
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Lea Röhder
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Gerben Vader
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | |
Collapse
|
13
|
Lu X, Guo Y, Gu S, Tan D, Cheng B, Li Z, Huang W. An efficient and precise method for generating knockout cell lines based on CRISPR-Cas9 system. Eng Life Sci 2020; 20:585-593. [PMID: 33304232 PMCID: PMC7708952 DOI: 10.1002/elsc.202000032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/07/2020] [Accepted: 08/11/2020] [Indexed: 11/25/2022] Open
Abstract
Although the efficiency and versatility of CRISPR-Cas9 system has been greatly improved over conventional genome editing methods such as zinc finger or TALEN, it is still time-consuming and labor-intensive for screening knockout/knock-in cell clones due to differences of the targeted location or efficacies of guide RNAs (gRNAs). Here, we adapted a targeted knock-in strategy with CRISPR-Cas9 system and characterized the efficiency for generating single or double knockout cell lines. Specifically, a homology-arm based donor cassette consisting of genes encoding a fluorescence protein and antibiotic selection marker driven by a constitutive promoter was co-transfected with a gRNA expressing unit. Based on FACS sorting and antibiotic drug selection, positive cell clones were confirmed by genotyping and at the protein expression level. The results indicated that more than 70% of analyzed clones identified by cell sorting and selection were successfully targeted in both single and double knockout experiments. The procedure takes less than three weeks to obtain knockout cell lines. We believe that this methodology could be applicable and versatile in generating knockout cell clones with high efficiency in most cell lines.
Collapse
Affiliation(s)
- Xibin Lu
- Core Research FacilitiesSouthern University of Science and TechnologyShenzhenP. R. China
| | - Yuhan Guo
- Forward Pharmaceuticals Limited CoShenzhenP. R. China
| | - Shu Gu
- Department of BiologySouthern University of Science and TechnologyShenzhenP. R. China
| | - Deng Tan
- Department of BiologySouthern University of Science and TechnologyShenzhenP. R. China
| | - Baoyun Cheng
- Department of BiologySouthern University of Science and TechnologyShenzhenP. R. China
| | - Zhoufang Li
- Core Research FacilitiesSouthern University of Science and TechnologyShenzhenP. R. China
| | - Wei Huang
- Department of BiologySouthern University of Science and TechnologyShenzhenP. R. China
| |
Collapse
|
14
|
Davidson PM, Battistella A, Déjardin T, Betz T, Plastino J, Borghi N, Cadot B, Sykes C. Nesprin-2 accumulates at the front of the nucleus during confined cell migration. EMBO Rep 2020; 21:e49910. [PMID: 32419336 DOI: 10.15252/embr.201949910] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
The mechanisms by which cells exert forces on their nuclei to migrate through openings smaller than the nuclear diameter remain unclear. We use CRISPR/Cas9 to fluorescently label nesprin-2 giant, which links the cytoskeleton to the nuclear interior. We demonstrate that nesprin-2 accumulates at the front of the nucleus during nuclear deformation through narrow constrictions, independently of the nuclear lamina. We find that nesprins are mobile at time scales similar to the accumulation. Using artificial constructs, we show that the actin-binding domain of nesprin-2 is necessary and sufficient for this accumulation. Actin filaments are organized in a barrel structure around the nucleus in the direction of movement. Using two-photon ablation and cytoskeleton-inhibiting drugs, we demonstrate an actomyosin-dependent pulling force on the nucleus from the front of the cell. The elastic recoil upon ablation is dampened when nesprins are reduced at the nuclear envelope. We thus show that actin redistributes nesprin-2 giant toward the front of the nucleus and contributes to pulling the nucleus through narrow constrictions, in concert with myosin.
Collapse
Affiliation(s)
- Patricia M Davidson
- Laboratoire Physico-Chimie Curie, Institut Curie, CNRS UMR168, Sorbonne Université, PSL, Paris, France.,Center for Research in Myology, INSERM UMR974, Sorbonne Université, Paris, France
| | - Aude Battistella
- Laboratoire Physico-Chimie Curie, Institut Curie, CNRS UMR168, Sorbonne Université, PSL, Paris, France
| | - Théophile Déjardin
- Laboratoire Physico-Chimie Curie, Institut Curie, CNRS UMR168, Sorbonne Université, PSL, Paris, France.,Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Paris, France
| | - Timo Betz
- Institute of Cell Biology, ZMBE, Münster, Germany
| | - Julie Plastino
- Laboratoire Physico-Chimie Curie, Institut Curie, CNRS UMR168, Sorbonne Université, PSL, Paris, France
| | - Nicolas Borghi
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Paris, France
| | - Bruno Cadot
- Center for Research in Myology, INSERM UMR974, Sorbonne Université, Paris, France
| | - Cécile Sykes
- Laboratoire Physico-Chimie Curie, Institut Curie, CNRS UMR168, Sorbonne Université, PSL, Paris, France
| |
Collapse
|
15
|
Dalmasso B, Ghiorzo P. Evolution of approaches to identify melanoma missing heritability. Expert Rev Mol Diagn 2020; 20:523-531. [PMID: 32124637 DOI: 10.1080/14737159.2020.1738221] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Around 10% of melanoma patients have a positive family history of melanoma and/or related cancers. Although a germline pathogenic variant in a high-risk gene can be identified in up to 40% of these patients, the remaining part of melanoma heritability remains largely unexplained.Areas covered: The aim of this review is to provide an overview of the impact that new technologies and new research approaches had and are having on finding more efficient ways to unravel the missing heritability in melanoma.Expert opinion: High-throughput sequencing technologies have been crucial in increasing the number of genes/loci that might be implicated in melanoma predisposition. However, results from these approaches may have been inferior to the expectations, due to an increase in quantitative information which hasn't been followed at the same speed by an improvement of the methods to correctly interpret these data. Optimal approaches for improving our knowledge on melanoma heritability are currently based on segregation analysis coupled with functional assessment of candidate genes. An improvement of computational methods to infer genotype-phenotype correlations could help address the issue of missing heritability.
Collapse
Affiliation(s)
- Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| |
Collapse
|
16
|
García-Tuñón I, Vuelta E, Lozano L, Herrero M, Méndez L, Palomero-Hernandez J, Pérez-Caro M, Pérez-García J, González-Sarmiento R, Sánchez-Martín M. Establishment of a conditional Nomo1 mouse model by CRISPR/Cas9 technology. Mol Biol Rep 2020; 47:1381-1391. [PMID: 31833031 DOI: 10.1007/s11033-019-05214-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022]
Abstract
The Nomo1 gene mediates a wide range of biological processes of importance in embryonic development. Accordingly, constitutive perturbation of Nomo1 function may result in myriad developmental defects that trigger embryonic lethality. To extend our understanding of Nomo1 function in postnatal stages and in a tissue-specific manner, we generated a conditional knockout mouse model of Nomo1. To achieve this, we used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology in C57Bl/6J mouse zygotes to generate a new mouse model in which exon 3 of the Nomo1 gene is specifically flanked (or floxed) by LoxP sites (Nomo1f/f). Nomo1f/f mouse embryonic fibroblasts were transduced with a Cre adenovirus and efficiently recombined between LoxP sites. Genomic and expression studies in Nomo1-transduced MEFs demonstrated that the Nomo1 exon 3 is ablated. Western blot assay showed that no protein or early truncated protein is produced. In vivo assay crossing Nomo1f/f mouse with a Msi1-CRE transgenic mouse corroborated the previous findings and it showed Nomo1 exon 3 deletion at msi1+ cell compartment. This short technical report demonstrates that CRISPR/Cas9 technology is a simple and easy method for creating conditional mouse models. The Nomo1f/f mouse will be useful to researchers who wish to explore the role of Nomo1 in any developmental stage or in a tissue-specific manner.
Collapse
Affiliation(s)
- Ignacio García-Tuñón
- IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Elena Vuelta
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Laura Lozano
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - María Herrero
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Lucía Méndez
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Javier Palomero-Hernandez
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - María Pérez-Caro
- Banco de ADN, Nucleus, Universidad de Salamanca, Salamanca, Spain
| | - Jessica Pérez-García
- IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Rogelio González-Sarmiento
- IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, Spain
- Departamento de Medicina, Universidad 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
|
17
|
Spiegel A, Bachmann M, Jurado Jiménez G, Sarov M. CRISPR/Cas9-based knockout pipeline for reverse genetics in mammalian cell culture. Methods 2019; 164-165:49-58. [PMID: 31051255 DOI: 10.1016/j.ymeth.2019.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/04/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
We present a straightforward protocol for reverse genetics in cultured mammalian cells, using CRISPR/Cas9-mediated homology-dependent repair (HDR) based insertion of a protein trap cassette, resulting in a termination of the endogenous gene expression. Complete loss of function can be achieved with monoallelic trap cassette insertion, as the second allele is frequently disrupted by an error-prone non-homologous end joining (NHEJ) mechanism. The method should be applicable to any expressed gene in most cell lines, including those with low HDR efficiency, as the knockout alleles can be directly selected for.
Collapse
Affiliation(s)
- Aleksandra Spiegel
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Mandy Bachmann
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Gabriel Jurado Jiménez
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
18
|
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
|
19
|
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: 15] [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
|
20
|
Alka K, Casey JR. Ophthalmic Nonsteroidal Anti-Inflammatory Drugs as a Therapy for Corneal Dystrophies Caused by SLC4A11 Mutation. Invest Ophthalmol Vis Sci 2019; 59:4258-4267. [PMID: 30140924 DOI: 10.1167/iovs.18-24301] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose SLC4A11 is a plasma membrane protein of corneal endothelial cells. Some mutations of the SLC4A11 gene result in SLC4A11 protein misfolding and failure to mature to the plasma membrane. This gives rise to some cases of Fuchs' endothelial corneal dystrophy (FECD) and congenital hereditary endothelial dystrophy (CHED). We screened ophthalmic nonsteroidal anti-inflammatory drugs (NSAIDs) for their ability to correct SLC4A11 folding defects. Methods Five ophthalmic NSAIDs were tested for their therapeutic potential in some genetic corneal dystrophy patients. HEK293 cells expressing CHED and FECD-causing SLC4A11 mutants were grown on 96-well dishes in the absence or presence of NSAIDs. Ability of NSAIDs to correct mutant SLC4A11 cell-surface trafficking was assessed with a bioluminescence resonance energy transfer (BRET) assay and by confocal microscopy. The ability of mutant SLC4A11-expressing cells to mediate water flux (SLC4A11 mediates water flux across the corneal endothelial cell basolateral membrane as part of the endothelial water pump) was measured upon treatment with ophthalmic NSAIDs. Results BRET-assays revealed significant rescue of SLC4A11 mutants to the cell surface by 4 of 5 NSAIDs tested. The NSAIDs, diclofenac and nepafenac, were effective in moving endoplasmic reticulum-retained missense mutant SLC4A11 to the cell surface, as measured by confocal immunofluorescence. Among intracellular-retained SLC4A11 mutants, 20 of 30 had significant restoration of cell surface abundance upon treatment with diclofenac. Diclofenac restored mutant SLC4A11 water flux activity to the level of wild-type SLC4A11 in some cases. Conclusions These results encourage testing diclofenac eye drops as a treatment for corneal dystrophy in patients whose disease is caused by some SLC4A11 missense mutations.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph R Casey
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Khan SH. Genome-Editing Technologies: Concept, Pros, and Cons of Various Genome-Editing Techniques and Bioethical Concerns for Clinical Application. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:326-334. [PMID: 30965277 PMCID: PMC6454098 DOI: 10.1016/j.omtn.2019.02.027] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022]
Abstract
The traditional healthcare system is at the doorstep for entering into the arena of molecular medicine. The enormous knowledge and ongoing research have now been able to demonstrate methodologies that can alter DNA coding. The techniques used to edit or change the genome evolved from the earlier attempts like nuclease technologies, homing endonucleases, and certain chemical methods. Molecular techniques like meganuclease, transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs) initially emerged as genome-editing technologies. These initial technologies suffer from lower specificity due to their off-targets side effects. Moreover, from biotechnology's perspective, the main obstacle was to develop simple but effective delivery methods for host cell entry. Later, small RNAs, including microRNA (miRNA) and small interfering RNA (siRNA), have been widely adopted in the research laboratories to replace lab animals and cell lines. The latest discovery of CRISPR/Cas9 technology seems more encouraging by providing better efficiency, feasibility, and multi-role clinical application. This later biotechnology seem to take genome-engineering techniques to the next level of molecular engineering. This review generally discusses the various gene-editing technologies in terms of the mechanisms of action, advantages, and side effects.
Collapse
Affiliation(s)
- Sikandar Hayat Khan
- Department of Pathology, PNS HAFEEZ Hospital, Pathology E-8, Islamabad, Islamabad 44400, Pakistan.
| |
Collapse
|
22
|
Gueneau F, Ravn U, Fischer N. Round optimization for improved discovery of native bispecific antibodies. Methods 2019; 154:51-59. [PMID: 30448477 DOI: 10.1016/j.ymeth.2018.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 01/07/2023] Open
Abstract
The assembly of bispecific antibodies (bsAb) that retain the structure of a standard IgG can be challenging as the correct pairing of the different heavy and light chains has to be ensured while unwanted side products kept to a minimum. The use of antibodies sharing a common chain facilitates assembly of such bsAb formats but requires additional efforts during the initial discovery phase. We have developed a native bsAb format called κλ body based on antibodies that, while being specific for different antigens, share the same heavy chain. Such antibodies can readily be isolated from antibody libraries incorporating a single VH combined with light chain diversity. However, in order to improve the discovery process of such fixed VH antibodies, we developed a method to optimize populations of light chains by recovering and shuffling CDRL3 sequences that have been enriched for antigen binding by phage display selection. This approach allowed for the isolation of a more diverse and potent panel of antibodies blocking the interaction between PD-1 and PD-L1 when compared to our standard in vitro selection approach, thus providing better building blocks for subsequent bsAb generation.
Collapse
Affiliation(s)
- F Gueneau
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - U Ravn
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - N Fischer
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland.
| |
Collapse
|
23
|
Campagne A, Lee MK, Zielinski D, Michaud A, Le Corre S, Dingli F, Chen H, Shahidian LZ, Vassilev I, Servant N, Loew D, Pasmant E, Postel-Vinay S, Wassef M, Margueron R. BAP1 complex promotes transcription by opposing PRC1-mediated H2A ubiquitylation. Nat Commun 2019; 10:348. [PMID: 30664650 PMCID: PMC6341105 DOI: 10.1038/s41467-018-08255-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 12/21/2018] [Indexed: 11/09/2022] Open
Abstract
In Drosophila, a complex consisting of Calypso and ASX catalyzes H2A deubiquitination and has been reported to act as part of the Polycomb machinery in transcriptional silencing. The mammalian homologs of these proteins (BAP1 and ASXL1/2/3, respectively), are frequently mutated in various cancer types, yet their precise functions remain unclear. Using an integrative approach based on isogenic cell lines generated with CRISPR/Cas9, we uncover an unanticipated role for BAP1 in gene activation. This function requires the assembly of an enzymatically active BAP1-associated core complex (BAP1.com) containing one of the redundant ASXL proteins. We investigate the mechanism underlying BAP1.com-mediated transcriptional regulation and show that it does not participate in Polycomb-mediated silencing. Instead, our results establish that the function of BAP1.com is to safeguard transcriptionally active genes against silencing by the Polycomb Repressive Complex 1. In Drosophila, the Calypso–ASX complex catalyzes H2A deubiquitination and aids Polycomb in transcriptional silencing. Here the authors show that the orthologous complex, BAP1.com, promotes gene activation by counteracting PRC1-mediated gene silencing.
Collapse
Affiliation(s)
- Antoine Campagne
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Ming-Kang Lee
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Dina Zielinski
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Audrey Michaud
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Stéphanie Le Corre
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Florent Dingli
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France
| | - Hong Chen
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Lara Z Shahidian
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Ivaylo Vassilev
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Nicolas Servant
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Damarys Loew
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France
| | - Eric Pasmant
- Department of Molecular Genetics Pathology, Cochin Hospital, HUPC AP-HP, EA7331, Faculty of Pharmacy, University of Paris Descartes, Paris, 75014, France
| | - Sophie Postel-Vinay
- Département d'Innovation Thérapeutique et Essais Précoces, INSERM U981, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Michel Wassef
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France. .,INSERM U934/CNRS UMR3215, 75005, Paris, France.
| | - Raphaël Margueron
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France. .,INSERM U934/CNRS UMR3215, 75005, Paris, France.
| |
Collapse
|
24
|
Xu JJ, Smeets MF, Tan SY, Wall M, Purton LE, Walkley CR. Modeling human RNA spliceosome mutations in the mouse: not all mice were created equal. Exp Hematol 2018; 70:10-23. [PMID: 30408513 DOI: 10.1016/j.exphem.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 01/23/2023]
Abstract
Myelodysplastic syndromes (MDS) and related myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) are clonal stem cell disorders, primarily affecting patients over 65 years of age. Mapping of the MDS and MDS/MPN genome identified recurrent heterozygous mutations in the RNA splicing machinery, with the SF3B1, SRSF2, and U2AF1 genes being frequently mutated. To better understand how spliceosomal mutations contribute to MDS pathogenesis in vivo, numerous groups have sought to establish conditional murine models of SF3B1, SRSF2, and U2AF1 mutations. The high degree of conservation of hematopoiesis between mice and human and the well-established phenotyping and genetic modification approaches make murine models an effective tool with which to study how a gene mutation contributes to disease pathogenesis. The murine models of spliceosomal mutations described to date recapitulate human MDS or MDS/MPN to varying extents. Reasons for the differences in phenotypes reported between alleles of the same mutation are varied, but the nature of the genetic modification itself and subsequent analysis methods are important to consider. In this review, we summarize recently reported murine models of SF3B1, SRSF2, and U2AF1 mutations, with a particular focus on the genetically engineered modifications underlying the models and the experimental approaches applied.
Collapse
Affiliation(s)
- Jane Jialu Xu
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Monique F Smeets
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Shuh Ying Tan
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Department of Hematology, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Meaghan Wall
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Victorian Cancer Cytogenetics Service, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Louise E Purton
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Carl R Walkley
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
25
|
Gu Y, Chang X, Dai S, Song Q, Zhao H, Lei P. Identification of four novel XPC mutations in two xeroderma pigmentosum complementation group C patients and functional study of XPC Q320X mutant. Gene 2017; 628:162-169. [PMID: 28669926 DOI: 10.1016/j.gene.2017.06.057] [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] [Received: 04/06/2017] [Revised: 06/24/2017] [Accepted: 06/28/2017] [Indexed: 11/12/2022]
Abstract
Xeroderma pigmentosum (XP) is a rare, recessive hereditary disease characterized by sunlight hypersensitivity and high incidence of skin cancer with clinical and genetic heterogeneity. We collected two unrelated Chinese patients showing typical symptoms of XPC without neurologic symptoms. Direct sequencing of XPC gene revealed that patient 1 carried IVS1+1G>A and c.958 C>T mutations, and patient 2 carried c.545_546delTA and c.2257_2258insC mutations. All these four mutations introduced premature terminal codons (PTCs) in XPC gene. The nonsense mutation c.958 C>T yielded truncated mutant Q320X, and we studied its function for global genome repair kinetics. Overexpressed Q320X mutant can localize to site of DNA damage, but it is defective in CPD and 6-4PP repair. Readthrough of PTCs is a new approach to treatment of genetic diseases. We found that aminoglycosides could significantly increase the full length protein expression of Q320X mutant, but NER defects were not rescued in vitro.
Collapse
Affiliation(s)
- Yajuan Gu
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaodan Chang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Shan Dai
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Qinghua Song
- Department of Dermatology, Peking University Third Hospital, Beijing, China.
| | - Hongshan Zhao
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China; Human Disease Genomics Center, Peking University, Beijing, China.
| | - Pengcheng Lei
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
26
|
Concordet JP, Giovannangeli C. CRISPR-Cas systems for genome engineering and investigation. Methods 2017. [DOI: 10.1016/j.ymeth.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|