1
|
Morán-Plata FJ, Muñoz-García N, González-González M, Pozo J, Carretero-Domínguez S, Mateos S, Barrena S, Belhassen-García M, Lau C, Teixeira MDA, Santos AH, Yeguas A, Balanzategui A, García-Sancho AM, Orfao A, Almeida J. A novel NKp80-based strategy for universal identification of normal, reactive and tumor/clonal natural killer-cells in blood. Front Immunol 2024; 15:1423689. [PMID: 39040115 PMCID: PMC11260609 DOI: 10.3389/fimmu.2024.1423689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Purpose Natural killer (NK) cells are traditionally identified by flow cytometry using a combination of markers (CD16/CD56/CD3), because a specific NK-cell marker is still missing. Here we investigated the utility of CD314, CD335 and NKp80, compared to CD16/CD56/CD3, for more robust identification of NK-cells in human blood, for diagnostic purposes. Methods A total of 156 peripheral blood (PB) samples collected from healthy donors (HD) and patients with diseases frequently associated with loss/downregulation of classical NK-cell markers were immunophenotyped following EuroFlow protocols, aimed at comparing the staining profile of total blood NK-cells for CD314, CD335 and NKp80, and the performance of distinct marker combinations for their accurate identification. Results NKp80 showed a superior performance (vs. CD314 and CD335) for the identification of NK-cells in HD blood. Besides, NKp80 improved the conventional CD16/CD56/CD3-based strategy to identify PB NK-cells in HD and reactive processes, particularly when combined with CD16 for further accurate NK-cell-subsetting. Although NKp80+CD16 improved the identification of clonal/tumor NK-cells, particularly among CD56- cases (53%), aberrant downregulation of NKp80 was observed in 25% of patients, in whom CD56 was useful as a complementary NK-cell marker. As NKp80 is also expressed on T-cells, we noted increased numbers of NKp80+ cytotoxic T-cells at the more advanced maturation stages, mostly in adults. Conclusion Here we propose a new robust approach for the identification of PB NK-cells, based on the combination of NKp80 plus CD16. However, in chronic lymphoproliferative disorders of NK-cells, addition of CD56 is recommended to identify clonal NK-cells, due to their frequent aberrant NKp80- phenotype.
Collapse
Affiliation(s)
- F. Javier Morán-Plata
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Noemí Muñoz-García
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - María González-González
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Julio Pozo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Sonia Carretero-Domínguez
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Sheila Mateos
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cell-purification Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Susana Barrena
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Moncef Belhassen-García
- Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain
- Department of Infectious Diseases, University Hospital of Salamanca, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Salamanca, Spain
| | - Catarina Lau
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP), Porto, Portugal
| | - Maria Dos Anjos Teixeira
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP), Porto, Portugal
| | - Ana Helena Santos
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP), Porto, Portugal
| | - Ana Yeguas
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Ana Balanzategui
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
- Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Martín García-Sancho
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
- Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC – University of Salamanca), and Department of Medicine, University of Salamanca, Salamanca, Spain
- Cytometry Service, NUCLEUS, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Rückert T, Romagnani C. Extrinsic and intrinsic drivers of natural killer cell clonality. Immunol Rev 2024; 323:80-106. [PMID: 38506411 DOI: 10.1111/imr.13324] [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] [Indexed: 03/21/2024]
Abstract
Clonal expansion of antigen-specific lymphocytes is the fundamental mechanism enabling potent adaptive immune responses and the generation of immune memory. Accompanied by pronounced epigenetic remodeling, the massive proliferation of individual cells generates a critical mass of effectors for the control of acute infections, as well as a pool of memory cells protecting against future pathogen encounters. Classically associated with the adaptive immune system, recent work has demonstrated that innate immune memory to human cytomegalovirus (CMV) infection is stably maintained as large clonal expansions of natural killer (NK) cells, raising questions on the mechanisms for clonal selection and expansion in the absence of re-arranged antigen receptors. Here, we discuss clonal NK cell memory in the context of the mechanisms underlying clonal competition of adaptive lymphocytes and propose alternative selection mechanisms that might decide on the clonal success of their innate counterparts. We propose that the integration of external cues with cell-intrinsic sources of heterogeneity, such as variegated receptor expression, transcriptional states, and somatic variants, compose a bottleneck for clonal selection, contributing to the large size of memory NK cell clones.
Collapse
Affiliation(s)
- Timo Rückert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Chiara Romagnani
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| |
Collapse
|
3
|
Jiang S, Zheng Y, Lv B, He S, Yang W, Wang B, Zhou J, Liu S, Li D, Lin J. Single-cell landscape dissecting the transcription and heterogeneity of innate lymphoid cells in ischemic heart. Front Immunol 2023; 14:1129007. [PMID: 37228603 PMCID: PMC10203554 DOI: 10.3389/fimmu.2023.1129007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Background Until now, few articles have revealed the potential roles of innate lymphoid cells (ILCs) in cardiovascular diseases. However, the infiltration of ILC subsets in ischemic myocardium, the roles of ILC subsets in myocardial infarction (MI) and myocardial ischemia-reperfusion injury (MIRI) and the related cellular and molecular mechanisms have not been described with a sufficient level of detail. Method In the current study, 8-week-old male C57BL/6J mice were divided into three groups: MI, MIRI and sham group. Single-cell sequencing technology was used to perform dimensionality reduction clustering of ILC to analyze the ILC subset landscape at a single-cell resolution, and finally flow cytometry was used to confirm the existence of the new ILC subsets in different disease groups. Results Five ILC subsets were found, including ILC1, ILC2a, ILC2b, ILCdc and ILCt. It is worth noting that ILCdc, ILC2b and ILCt were identified as new ILC subclusters in the heart. The cellular landscapes of ILCs were revealed and signal pathways were predicted. Furthermore, pseudotime trajectory analysis exhibited different ILC statuses and traced related gene expression in normal and ischemic conditions. In addition, we established a ligand-receptor-transcription factor-target gene regulatory network to disclose cell communications among ILC clusters. Moreover, we further revealed the transcriptional features of the ILCdc and ILC2a subsets. Finally, the existence of ILCdc was confirmed by flow cytometry. Conclusion Collectively, by characterizing the spectrums of ILC subclusters, our results provide a new blueprint for understanding ILC subclusters' roles in myocardial ischemia diseases and further potential treatment targets.
Collapse
Affiliation(s)
- Shijiu Jiang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, The First Affiliated Hospital, Shihezi University, Shihezi, Xinjiang, China
| | - Yuqi Zheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenling Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boyuan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shangwei Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dazhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jibin Lin
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Kaur K, Kanayama K, Wu QQ, Gumrukcu S, Nishimura I, Jewett A. Zoledronic acid mediated differential activation of NK cells in different organs of WT and Rag2 mice; stark differences between the bone marrow and gingivae. Cell Immunol 2022; 375:104526. [DOI: 10.1016/j.cellimm.2022.104526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022]
|
5
|
Tissue-specific transcriptional imprinting and heterogeneity in human innate lymphoid cells revealed by full-length single-cell RNA-sequencing. Cell Res 2021; 31:554-568. [PMID: 33420427 PMCID: PMC8089104 DOI: 10.1038/s41422-020-00445-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The impact of the microenvironment on innate lymphoid cell (ILC)-mediated immunity in humans remains largely unknown. Here we used full-length Smart-seq2 single-cell RNA-sequencing to unravel tissue-specific transcriptional profiles and heterogeneity of CD127+ ILCs across four human tissues. Correlation analysis identified gene modules characterizing the migratory properties of tonsil and blood ILCs, and signatures of tissue-residency, activation and modified metabolism in colon and lung ILCs. Trajectory analysis revealed potential differentiation pathways from circulating and tissue-resident naïve ILCs to a spectrum of mature ILC subsets. In the lung we identified both CRTH2+ and CRTH2- ILC2 with lung-specific signatures, which could be recapitulated by alarmin-exposure of circulating ILC2. Finally, we describe unique TCR-V(D)J-rearrangement patterns of blood ILC1-like cells, revealing a subset of potentially immature ILCs with TCR-δ rearrangement. Our study provides a useful resource for in-depth understanding of ILC-mediated immunity in humans, with implications for disease.
Collapse
|
6
|
Fayez EA, Qazvini FF, Mahmoudi SM, Khoei S, Vesaltalab M, Teimourian S. Diagnosis of radiosensitive severe combined immunodeficiency disease (RS-SCID) by Comet Assay, management of bone marrow transplantation. Immunobiology 2020; 225:151961. [PMID: 32517885 DOI: 10.1016/j.imbio.2020.151961] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Severe combined immunodeficiency disease (SCID) is a rare inherited severe immunodeficiency, in which functions of T cells and B cells are impaired. SCID is inherited either in X-linked recessive, or autosomal recessive forms, and is either radiosensitive or radioresistant. Artemis (DCLRE1C gene), DNA ligase IV, DNA-PKC, and Cernunnos/XLF proteins are regarded as NHEJ (Non-Homologous End-Joining) proteins that are involved in the repair process of double-strand DNA breaks and their mutations would lead to cellular radiosensitivity. Diagnostic radiosensitivity assays are important for the management of clinical BMT (Bone Marrow Transplantation) conditions, such as what conditioning agents and doses should be used. MATERIALS AND METHODS In this study, five SCID patients and healthy controls were examined. Skin fibroblasts were cultured. After X-irradiation, cells either underwent clonogenic assay or incubated to allow DNA repair and examined by the alkaline comet assay. Finally, DCLRE1C, RAG-1, and RAG-2 genes sequenced. RESULTS By clonogenic assay, three patients were detected as radiosensitive with possible mutations in NHEJ genes such as DCLRE1C gene. The percentage of DNA in the tail measured by comet assay, in all three patients, was significantly different from the two other patients and the control group (p-value < 0.05). By using Sanger sequencing, a mutation in DCLRE1C gene was detected in one of the radiosensitive patients and two mutations in RAG-1, and RAG-2 genes were detected in the two radioresistant patients. CONCLUSION Our findings suggest that comet assay is a fast technique for the diagnosis of the radiosensitive form of SCID and is very suitable for the timely diagnosis of RS-SCID before BMT.
Collapse
Affiliation(s)
- Elham Alipour Fayez
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Farajihaye Qazvini
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Marzeyeh Mahmoudi
- Department of Cell and Molecular Biology, Islamic Azad University, Science and Research Branch. Tehran, Iran
| | - Samideh Khoei
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Matin Vesaltalab
- School of Medicine, Bandar Abbas University of Medical Science, Bandar Abbas, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Themeli M, Chhatta A, Boersma H, Prins HJ, Cordes M, de Wilt E, Farahani AS, Vandekerckhove B, van der Burg M, Hoeben RC, Staal FJT, Mikkers HMM. iPSC-Based Modeling of RAG2 Severe Combined Immunodeficiency Reveals Multiple T Cell Developmental Arrests. Stem Cell Reports 2020; 14:300-311. [PMID: 31956083 PMCID: PMC7013232 DOI: 10.1016/j.stemcr.2019.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022] Open
Abstract
RAG2 severe combined immune deficiency (RAG2-SCID) is a lethal disorder caused by the absence of functional T and B cells due to a differentiation block. Here, we generated induced pluripotent stem cells (iPSCs) from a RAG2-SCID patient to study the nature of the T cell developmental blockade. We observed a strongly reduced capacity to differentiate at every investigated stage of T cell development, from early CD7−CD5− to CD4+CD8+. The impaired differentiation was accompanied by an increase in CD7−CD56+CD33+ natural killer (NK) cell-like cells. T cell receptor D rearrangements were completely absent in RAG2SCID cells, whereas the rare T cell receptor B rearrangements were likely the result of illegitimate rearrangements. Repair of RAG2 restored the capacity to induce T cell receptor rearrangements, normalized T cell development, and corrected the NK cell-like phenotype. In conclusion, we succeeded in generating an iPSC-based RAG2-SCID model, which enabled the identification of previously unrecognized disorder-related T cell developmental roadblocks. RAG2-SCID cells show impaired differentiation at several stages of T cell development RAG2-SCID T and NK cells fail to undergo legitimate RAG-driven TCR rearrangements RAG2-SCID cells exhibit a skewed differentiation toward NK cell-like cells RAG2-SCID phenotype is rescued by gene correction
Collapse
Affiliation(s)
- Maria Themeli
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Amiet Chhatta
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Hester Boersma
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Henk Jan Prins
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Martijn Cordes
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Edwin de Wilt
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | - Aïda Shahrabi Farahani
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Bart Vandekerckhove
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Gent 9000, Belgium
| | - Mirjam van der Burg
- Department of Immunology, Erasmus Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Rob C Hoeben
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Frank J T Staal
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Harald M M Mikkers
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands; LUMC hiPSC Hotel, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands.
| |
Collapse
|
8
|
Abstract
DNA damage occurs on exposure to genotoxic agents and during physiological DNA transactions. DNA double-strand breaks (DSBs) are particularly dangerous lesions that activate DNA damage response (DDR) kinases, leading to initiation of a canonical DDR (cDDR). This response includes activation of cell cycle checkpoints and engagement of pathways that repair the DNA DSBs to maintain genomic integrity. In adaptive immune cells, programmed DNA DSBs are generated at precise genomic locations during the assembly and diversification of lymphocyte antigen receptor genes. In innate immune cells, the production of genotoxic agents, such as reactive nitrogen molecules, in response to pathogens can also cause genomic DNA DSBs. These DSBs in adaptive and innate immune cells activate the cDDR. However, recent studies have demonstrated that they also activate non-canonical DDRs (ncDDRs) that regulate cell type-specific processes that are important for innate and adaptive immune responses. Here, we review these ncDDRs and discuss how they integrate with other signals during immune system development and function.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barry P Sleckman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Theunissen PMJ, van Zessen D, Stubbs AP, Faham M, Zwaan CM, van Dongen JJM, Van Der Velden VHJ. Antigen receptor sequencing of paired bone marrow samples shows homogeneous distribution of acute lymphoblastic leukemia subclones. Haematologica 2017; 102:1869-1877. [PMID: 28860343 PMCID: PMC5664391 DOI: 10.3324/haematol.2017.171454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/24/2017] [Indexed: 01/30/2023] Open
Abstract
In B-cell precursor acute lymphoblastic leukemia, the initial leukemic cells share the same antigen receptor gene rearrangements. However, due to ongoing rearrangement processes, leukemic cells with different gene rearrangement patterns can develop, resulting in subclone formation. We studied leukemic subclones and their distribution in the bone marrow and peripheral blood at diagnosis. Antigen receptor gene rearrangements (IGH, IGK, TRG, TRD, TRB) were analyzed by next-generation sequencing in seven paired bone marrow samples and five paired bone marrow-peripheral blood samples. Background-thresholds were defined, which enabled identification of leukemic gene rearrangements down to very low levels. Paired bone marrow analysis showed oligoclonality in all 7 patients and up to 34 leukemic clones per patient. Additional analysis of evolutionary-related IGH gene rearrangements revealed up to 171 leukemic clones per patient. Interestingly, overall 86% of all leukemic gene rearrangements, including small subclones, were present in both bone marrow samples (range per patient: 72–100%). Paired bone marrow-peripheral blood analysis showed that 83% of all leukemic gene rearrangements in bone marrow were also found in peripheral blood (range per patient: 81–100%). Remarkably, in the paired bone marrow samples and paired bone marrow-peripheral blood samples the vast majority of leukemic gene rearrangements had a similar frequency (<5-fold frequency difference) (96% and 96% of leukemic rearrangements, respectively). Together, these results indicate that B-cell precursor acute lymphoblastic leukemia is generally highly oligoclonal. Nevertheless, the vast majority of leukemic clones, even the minor antigen receptor-defined subclones, are homogeneously distributed throughout the bone marrow and peripheral blood compartment.
Collapse
Affiliation(s)
- Prisca M J Theunissen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - David van Zessen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, the Netherlands.,Department of Bioinformatics, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Andrew P Stubbs
- Department of Bioinformatics, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Malek Faham
- Adaptive Biotechnologies Corp., South San Francisco, CA, USA
| | - Christian M Zwaan
- Department of Pediatric Oncology, Sophia Children's Hospital/Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jacques J M van Dongen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | |
Collapse
|
10
|
Dobbs K, Tabellini G, Calzoni E, Patrizi O, Martinez P, Giliani SC, Moratto D, Al-Herz W, Cancrini C, Cowan M, Bleesing J, Booth C, Buchbinder D, Burns SO, Chatila TA, Chou J, Daza-Cajigal V, Ott de Bruin LM, de la Morena M, Di Matteo G, Finocchi A, Geha R, Goyal RK, Hayward A, Holland S, Huang CH, Kanariou MG, King A, Kaplan B, Kleva A, Kuijpers TW, Lee BW, Lougaris V, Massaad M, Meyts I, Morsheimer M, Neven B, Pai SY, Parvaneh N, Plebani A, Prockop S, Reisli I, Soh JY, Somech R, Torgerson TR, Kim YJ, Walter JE, Gennery AR, Keles S, Manis JP, Marcenaro E, Moretta A, Parolini S, Notarangelo LD. Natural Killer Cells from Patients with Recombinase-Activating Gene and Non-Homologous End Joining Gene Defects Comprise a Higher Frequency of CD56 bright NKG2A +++ Cells, and Yet Display Increased Degranulation and Higher Perforin Content. Front Immunol 2017; 8:798. [PMID: 28769923 PMCID: PMC5511964 DOI: 10.3389/fimmu.2017.00798] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/23/2017] [Indexed: 11/13/2022] Open
Abstract
Mutations of the recombinase-activating genes 1 and 2 (RAG1 and RAG2) in humans are associated with a broad range of phenotypes. For patients with severe clinical presentation, hematopoietic stem cell transplantation (HSCT) represents the only curative treatment; however, high rates of graft failure and incomplete immune reconstitution have been observed, especially after unconditioned haploidentical transplantation. Studies in mice have shown that Rag−/− natural killer (NK) cells have a mature phenotype, reduced fitness, and increased cytotoxicity. We aimed to analyze NK cell phenotype and function in patients with mutations in RAG and in non-homologous end joining (NHEJ) genes. Here, we provide evidence that NK cells from these patients have an immature phenotype, with significant expansion of CD56bright CD16−/int CD57− cells, yet increased degranulation and high perforin content. Correlation was observed between in vitro recombinase activity of the mutant proteins, NK cell abnormalities, and in vivo clinical phenotype. Addition of serotherapy in the conditioning regimen, with the aim of depleting the autologous NK cell compartment, may be important to facilitate engraftment and immune reconstitution in patients with RAG and NHEJ defects treated by HSCT.
Collapse
Affiliation(s)
- Kerry Dobbs
- Laboratory of Host Defenses, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Enrica Calzoni
- "A. Nocivelli Institute for Molecular Medicine", Pediatric Clinic, University of Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Ornella Patrizi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paula Martinez
- Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Silvia Clara Giliani
- "A. Nocivelli Institute for Molecular Medicine", Pediatric Clinic, University of Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Daniele Moratto
- "A. Nocivelli Institute for Molecular Medicine", Pediatric Clinic, University of Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Caterina Cancrini
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy.,School of Medicine, University of Tor Vergata, Rome, Italy
| | - Morton Cowan
- Pediatric Allergy Immunology and Blood and Marrow Transplant Division, University of California San Francisco, Benioff Children's Hospital, San Francisco, CA, United States
| | - Jacob Bleesing
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Claire Booth
- Institute for Immunity and Transplantation, University College London, London, United Kingdom
| | - David Buchbinder
- Division of Pediatric Hematology, Children's Hospital Orange County, University of California Irvine, Orange County, CA, United States
| | - Siobhan O Burns
- Institute for Immunity and Transplantation, University College London, London, United Kingdom.,Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Vanessa Daza-Cajigal
- Institute for Immunity and Transplantation, University College London, London, United Kingdom
| | - Lisa M Ott de Bruin
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - MaiteTeresa de la Morena
- Division of Allergy and Immunology, Southwestern Medical Center, University of Texas, Dallas, TX, United States
| | - Gigliola Di Matteo
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy.,School of Medicine, University of Tor Vergata, Rome, Italy
| | - Andrea Finocchi
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy.,School of Medicine, University of Tor Vergata, Rome, Italy
| | - Raif Geha
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Rakesh K Goyal
- Division of Hematology/Oncology/BMT, Children's Mercy Hospital & Clinics, Kansas City, MO, United States
| | - Anthony Hayward
- Department of Pediatrics, Brown University, Providence, RI, United States
| | - Steven Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Chiung-Hui Huang
- Department of Paediatrics, National University Hospital, Singapore, Singapore
| | - Maria G Kanariou
- Department of Immunology-Histocompatibility, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Alejandra King
- Division of Pediatric Immunology, Hospital Luis Calvo Mackenna, Santiago, Chile
| | - Blanka Kaplan
- Department of Pediatrics, Division of Allergy and Immunology, Hofstra Northwell School of Medicine, Hofstra University, Great Neck, NY, United States
| | - Anastasiya Kleva
- Department of Pediatrics, Division of Allergy and Immunology, Hofstra Northwell School of Medicine, Hofstra University, Great Neck, NY, United States
| | - Taco W Kuijpers
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Bee Wah Lee
- Department of Paediatrics, National University Hospital, Singapore, Singapore
| | - Vassilios Lougaris
- Department of Experimental and Clinical Sciences, University of Brescia, Brescia, Italy
| | - Michel Massaad
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Megan Morsheimer
- Transplantation Branch, Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Benedicte Neven
- Pediatric Hematology-Immunology Department, Hospital Necker-Enfants Malades, Institut Imagine, AP-HP, Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA, United States
| | | | - Alessandro Plebani
- Department of Experimental and Clinical Sciences, University of Brescia, Brescia, Italy
| | - Susan Prockop
- Bone Marrow Transplant Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ismail Reisli
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Jian Yi Soh
- Department of Paediatrics, National University Hospital, Singapore, Singapore
| | - Raz Somech
- Pediatric Immunology Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Troy R Torgerson
- Department of Pediatrics and Immunology, Seattle Children's Hospital, University of Washingtin, Seattle, WA, United States
| | - Yae-Jaen Kim
- Division of Infectious Diseases and Immunodeficiency, Department of Pediatrics, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, South Korea
| | - Jolan E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Andrew R Gennery
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle Upon Tyne, United Kingdom.,Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sevgi Keles
- Division of Pediatric Immunology and Allergy, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - John P Manis
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Emanuela Marcenaro
- Molecular Immunology Laboratories, Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Alessandro Moretta
- Molecular Immunology Laboratories, Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi D Notarangelo
- Laboratory of Host Defenses, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Arya R, Bassing CH. V(D)J Recombination Exploits DNA Damage Responses to Promote Immunity. Trends Genet 2017; 33:479-489. [PMID: 28532625 PMCID: PMC5499712 DOI: 10.1016/j.tig.2017.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022]
Abstract
It has been recognized for 40 years that the variable (diversity) joining [V(D)J] recombination-mediated assembly of diverse B and T lymphocyte antigen receptor (AgR) genes is not only essential for adaptive immunity, but also a risk for autoimmunity and lymphoid malignancies. Over the past few years, several studies have revealed that recombination-activating gene (RAG) endonuclease-induced DNA double-strand breaks (DSBs) transcend hazardous intermediates during antigen receptor gene assembly. RAG cleavage within the genomes of lymphocyte progenitors and immature lymphocytes regulates the expression of ubiquitous and lymphocyte-specific gene transcripts to control the differentiation and function of both adaptive and innate immune cell lineages. These unexpected discoveries raise important new questions that have broad implications for basic immunology research and the screening, diagnosis, and treatment of human immunological disease.
Collapse
Affiliation(s)
- Rahul Arya
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Ettersperger J, Montcuquet N, Malamut G, Guegan N, Lopez-Lastra S, Gayraud S, Reimann C, Vidal E, Cagnard N, Villarese P, Andre-Schmutz I, Gomes Domingues R, Godinho-Silva C, Veiga-Fernandes H, Lhermitte L, Asnafi V, Macintyre E, Cellier C, Beldjord K, Di Santo JP, Cerf-Bensussan N, Meresse B. Interleukin-15-Dependent T-Cell-like Innate Intraepithelial Lymphocytes Develop in the Intestine and Transform into Lymphomas in Celiac Disease. Immunity 2016; 45:610-625. [PMID: 27612641 DOI: 10.1016/j.immuni.2016.07.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/12/2016] [Accepted: 06/02/2016] [Indexed: 01/19/2023]
Abstract
The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.
Collapse
Affiliation(s)
- Julien Ettersperger
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Nicolas Montcuquet
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Georgia Malamut
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Nicolas Guegan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Ségolène Gayraud
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Christian Reimann
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Elodie Vidal
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | | | - Patrick Villarese
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Isabelle Andre-Schmutz
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Rita Gomes Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | - Cristina Godinho-Silva
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | | | - Ludovic Lhermitte
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Vahid Asnafi
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Elizabeth Macintyre
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Christophe Cellier
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Kheira Beldjord
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France; Institut Universitaire d'Hématologie, Hôpital Saint-Louis, 75010 Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Bertrand Meresse
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| |
Collapse
|
13
|
Majumder K, Bassing CH, Oltz EM. Regulation of Tcrb Gene Assembly by Genetic, Epigenetic, and Topological Mechanisms. Adv Immunol 2015; 128:273-306. [PMID: 26477369 DOI: 10.1016/bs.ai.2015.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The adaptive immune system endows mammals with an ability to recognize nearly any foreign invader through antigen receptors that are expressed on the surface of all lymphocytes. This defense network is generated by V(D)J recombination, a set of sequentially controlled DNA cleavage and repair events that assemble antigen receptor genes from physically separated variable (V), joining (J), and sometimes diversity (D) gene segments. The recombination process itself must be stringently regulated to minimize oncogenic translocations involving chromosomes that harbor immunoglobulin and T cell receptor loci. Indeed, V(D)J recombination is controlled at several levels, including tissue-, developmental stage-, allele-, and gene segment-specificity. These levels of control are imposed by a collection of architectural and regulatory elements that are distributed throughout each antigen receptor locus. Together, the genetic elements regulate developmental changes in chromatin, transcription, and locus topology that promote or disfavor long-range recombination. This chapter focuses on the cross talk between these mechanisms at the T cell receptor beta (Tcrb) locus, and how they sculpt a diverse TCRβ repertoire while maintaining monospecificity of this antigen receptor on each mature T lymphocyte. We also discuss how insights obtained from studies of Tcrb are more generally relevant to our understanding of gene regulation strategies employed by mammals.
Collapse
Affiliation(s)
- Kinjal Majumder
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Abramson Family Cancer Research Institute, Cell and Molecular Biology Graduate Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eugene M Oltz
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
14
|
Abstract
The modular, noncontiguous architecture of the antigen receptor genes necessitates their assembly through V(D)J recombination. This program of DNA breakage and rejoining occurs during early lymphocyte development, and depends on the RAG1 and RAG2 proteins, whose collaborative endonuclease activity targets specific DNA motifs enriched in the antigen receptor loci. This essential gene shuffling reaction requires lymphocytes to traverse several developmental stages wherein DNA breakage is tolerated, while minimizing the expense to overall genome integrity. Thus, RAG activity is subject to stringent temporal and spatial regulation. The RAG proteins themselves also contribute autoregulatory properties that coordinate their DNA cleavage activity with target chromatin structure, cell cycle status, and DNA repair pathways. Even so, lapses in regulatory restriction of RAG activity are apparent in the aberrant V(D)J recombination events that underlie many lymphomas. In this review, we discuss the current understanding of the RAG endonuclease, its widespread binding in the lymphocyte genome, its noncleavage activities that restrain its enzymatic potential, and the growing evidence of its evolution from an ancient transposase.
Collapse
|
15
|
Karo JM, Sun JC. Novel molecular mechanism for generating NK-cell fitness and memory. Eur J Immunol 2015; 45:1906-15. [PMID: 26018782 DOI: 10.1002/eji.201445339] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 05/27/2015] [Indexed: 12/28/2022]
Abstract
The mammalian immune system has been traditionally subdivided into two compartments known as the innate and the adaptive. T cells and B cells, which rearrange their antigen-receptor genes using the RAG recombinase, comprise the adaptive arm of immunity. Meanwhile, every other white blood cell has been grouped together under the broad umbrella of innate immunity, including NK cells. NK cells are considered innate lymphocytes because of their rapid responses to stressed cells and their ability to develop without receptor gene rearrangement (i.e. in RAG-deficient mice). However, new findings implicate a critical function for RAG proteins during NK-cell ontogeny, and suggest a novel mechanism by which controlled DNA breaks during NK-cell development dictate the fitness, function, and longevity of these cells. This review highlights recent work describing how DNA break events can impact cellular differentiation and fitness in a variety of cell types and settings.
Collapse
Affiliation(s)
- Jenny M Karo
- Immunology Program and Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph C Sun
- Immunology Program and Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
16
|
Abstract
The RAG endonuclease generates DNA double strand breaks during antigen receptor gene assembly, an essential process for B- and T-lymphocyte development. However, a recent study reveals that RAG endonuclease activity affects natural killer cell function, demonstrating that such double strand breaks, and the responses they elicit, may have broad cellular effects.
Collapse
Affiliation(s)
- Andrea L Bredemeyer
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8118, St. Louis, MO 63110-1093, USA
| | - Barry P Sleckman
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8118, St. Louis, MO 63110-1093, USA.
| |
Collapse
|
17
|
Karo JM, Schatz DG, Sun JC. The RAG recombinase dictates functional heterogeneity and cellular fitness in natural killer cells. Cell 2015; 159:94-107. [PMID: 25259923 DOI: 10.1016/j.cell.2014.08.026] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/22/2014] [Accepted: 08/05/2014] [Indexed: 01/30/2023]
Abstract
The emergence of recombination-activating genes (RAGs) in jawed vertebrates endowed adaptive immune cells with the ability to assemble a diverse set of antigen receptor genes. In contrast, innate lymphocytes, such as natural killer (NK) cells, are not believed to require RAGs. Here, we report that NK cells unable to express RAGs or RAG endonuclease activity during ontogeny exhibit a cell-intrinsic hyperresponsiveness but a diminished capacity to survive following virus-driven proliferation, a reduced expression of DNA damage response mediators, and defects in the repair of DNA breaks. Evidence for this novel function of RAG has also been observed in T cells and innate lymphoid cells (ILCs), revealing an unexpected role for RAG proteins beyond V(D)J recombination. We propose that DNA cleavage events mediated by RAG endow developing adaptive and innate lymphocytes with a cellular "fitness" that safeguards their persistence later in life during episodes of rapid proliferation or cellular stress.
Collapse
Affiliation(s)
- Jenny M Karo
- Immunology Program and Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David G Schatz
- Department of Immunobiology and the Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joseph C Sun
- Immunology Program and Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
18
|
Dytrych P, Krol P, Kotrova M, Kuzilkova D, Hubacek P, Krol L, Katra R, Hrusak O, Kabelka Z, Dolezalova P, Kalina T, Fronkova E. Polyclonal, newly derived T cells with low expression of inhibitory molecule PD-1 in tonsils define the phenotype of lymphocytes in children with Periodic Fever, Aphtous Stomatitis, Pharyngitis and Adenitis (PFAPA) syndrome. Mol Immunol 2015; 65:139-47. [PMID: 25656804 DOI: 10.1016/j.molimm.2015.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/15/2014] [Accepted: 01/08/2015] [Indexed: 01/30/2023]
Abstract
PURPOSE PFAPA syndrome is a benign, recurrent inflammatory disease of childhood. Tonsillectomy is one of the therapeutic options with a yet unexplained biological mechanism. We tested whether specific lymphocyte subsets recruited from blood to human tonsils participate in PFAPA pathogenesis. METHODS Paired tonsils/peripheral blood (PB) samples were investigated (a) from children with PFAPA that successfully resolved after tonsillectomy (n=10) (b) from children with obstructive sleep apnoea syndrome as controls (n=10). The lymphocyte profiles were analysed using 8-colour flow cytometry, immunoglobulin (IGH) and T-cell receptor (TCR) gene rearrangements via PCR and next generation sequencing; a TREC/KREC analysis was performed using qPCR. RESULTS The PFAPA tonsils in the asymptomatic phase had a lower percentage of B-lymphocytes than controls; T-lymphocyte counts were significantly higher in PB. The percentages of cytotoxic CD8pos T-lymphocytes were approximately 2-fold higher in PFAPA tonsils; the transitional B cells and naïve stages of both the CD4pos and CD8pos T-lymphocytes with a low expression of PD-1 molecule and high numbers of TREC were also increased. With the exception of elevated plasmablasts, no other differences were significant in PB. The expression levels of CXCL10, CXCL9 and CCL19 genes were significantly higher in PFAPA tonsils. The IGH/TCR pattern showed no clonal/oligoclonal expansion. DNA from the Epstein-Barr virus, Human Herpervirus-6 or adenovirus was detected in 7 of 10 PFAPA tonsils but also in 7 of 9 controls. CONCLUSIONS Our findings suggest that the uninhibited, polyclonal response of newly derived lymphocytes participate in the pathogenesis of PFAPA. Because most of the observed changes were restricted to tonsils and were not present in PB, they partly explain the therapeutic success of tonsillectomy in PFAPA syndrome.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adenoviridae/isolation & purification
- B-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Chemokine CCL19/biosynthesis
- Chemokine CXCL10/biosynthesis
- Chemokine CXCL9/biosynthesis
- Child
- Child, Preschool
- Female
- Fever of Unknown Origin/complications
- Fever of Unknown Origin/immunology
- Fever of Unknown Origin/surgery
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/isolation & purification
- Herpesvirus 6, Human/genetics
- Herpesvirus 6, Human/isolation & purification
- Humans
- Infant
- Lymphadenitis/complications
- Lymphadenitis/immunology
- Lymphadenitis/surgery
- Lymphocyte Count
- Male
- Palatine Tonsil/cytology
- Palatine Tonsil/immunology
- Palatine Tonsil/surgery
- Pharyngitis/complications
- Pharyngitis/immunology
- Pharyngitis/surgery
- Programmed Cell Death 1 Receptor/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Sleep Apnea, Obstructive/immunology
- Sleep Apnea, Obstructive/surgery
- Stomatitis, Aphthous/complications
- Stomatitis, Aphthous/immunology
- Stomatitis, Aphthous/surgery
- T-Lymphocyte Subsets/immunology
- Tonsillectomy
Collapse
Affiliation(s)
- Petra Dytrych
- Department of ENT, Charles University, 2nd Faculty of Medicine, Charles University Prague and University Hospital Motol, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Large granular lymphocyte (LGL) syndrome comprises a clonal spectrum of T-cell and natural killer (NK)-cell LGL lymphoproliferative disorders associated with neutropenia. This review presents advances in diagnosis and therapy of LGL syndrome. RECENT FINDINGS Due to the lack of a single unique genetic or phenotypic feature and clinicopathological overlap between reactive and neoplastic entities, accurate LGL syndrome diagnosis should be based on the combination of morphologic, immunophenotypic, and molecular studies as well as clinical features. For diagnosis and monitoring of LGL proliferations, it is essential to perform flow cytometric blood and/or bone marrow analysis using a panel of monoclonal antibodies to conventional and novel T-cell and NK-cell antigens such as NK-cell receptors and T-cell receptor β-chain variable region families together with TCR gene rearrangement studies. Treatment of symptomatic cytopenias in patients with indolent LGL leukemia is still based on immunosuppressive therapy. Treatment with purine analogs and alemtuzumab may be considered as an alternative option. SUMMARY Progress in understanding the pathogenetic mechanisms of these entities, especially resistance of clonal LGLs to apoptosis, due to constitutive activation of survival signaling pathways, has its impact on identification of potential molecular therapeutic targets.
Collapse
|
20
|
CD2-positive B-cell precursor acute lymphoblastic leukemia with an early switch to the monocytic lineage. Leukemia 2013; 28:609-20. [PMID: 24270736 DOI: 10.1038/leu.2013.354] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/10/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022]
Abstract
Switches from the lymphoid to myeloid lineage during B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treatment are considered rare and thus far have been detected in MLL-rearranged leukemia. Here, we describe a novel BCP-ALL subset, switching BCP-ALL or swALL, which demonstrated monocytosis early during treatment. Despite their monocytic phenotype, 'monocytoids' share immunoreceptor gene rearrangements with leukemic B lymphoblasts. All swALLs demonstrated BCP-ALL with CD2 positivity and no MLL alterations, and the proportion of swALLs cases among BCP-ALLs was unexpectedly high (4%). The upregulation of CEBPα and demethylation of the CEBPA gene were significant in blasts at diagnosis, prior to the time when most of the switching occurs. Intermediate stages between CD14(neg)CD19(pos)CD34(pos) B lymphoblasts and CD14(pos)CD19(neg)CD34(neg) 'monocytoids' were detected, and changes in the expression of PAX5, PU1, M-CSFR, GM-CSFR and other genes accompanied the switch. Alterations in the Ikaros and ERG genes were more frequent in swALL patients; however, both were altered in only a minority of swALLs. Moreover, switching could be recapitulated in vitro and in mouse xenografts. Although children with swALL respond slowly to initial therapy, risk-based ALL therapy appears the treatment of choice for swALL. SwALL shows that transdifferentiating into monocytic lineage is specifically associated with CEBPα changes and CD2 expression.
Collapse
|
21
|
Mono/oligoclonal T and NK cells are common in chronic myeloid leukemia patients at diagnosis and expand during dasatinib therapy. Blood 2010; 116:772-82. [DOI: 10.1182/blood-2009-12-256800] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In a proportion of patients with chronic myeloid leukemia (CML) being treated with dasatinib, we recently observed large granular lymphocyte (LGL) expansions carrying clonal T-cell receptor (TCR) γ/δ gene rearrangements. To assess the prevalence and role of clonal lymphocytes in CML, we collected samples from patients (n = 34) at the time of diagnosis and during imatinib and dasatinib therapies and analyzed lymphocyte clonality with a sensitive polymerase chain reaction–based method of TCR γ and δ genes. Surprisingly, at CML diagnosis, 15 of 18 patients (83%) had a sizeable clonal, BCR-ABL1 negative lymphocyte population, which was uncommon in healthy persons (1 of 12; 8%). The same clone persisted at low levels in most imatinib-treated patients. In contrast, in a distinct population of dasatinib-treated patients, the diagnostic phase clone markedly expanded, resulting in absolute lymphocytosis in blood. Most patients with LGL expansions (90%) had TCR δ rearrangements, which were uncommon in patients without an LGL expansion (10%). The TCR δ clones were confined to γδ+ T- or natural killer–cell compartments and the TCR γ clones to CD4+/CD8+ αβ+ fractions. The functional importance of clonal lymphocytes as a part of leukemia immune surveillance and the putative anergy-reversing role of dasatinib require further evaluation.
Collapse
|
22
|
Mejstrikova E, Volejnikova J, Fronkova E, Zdrahalova K, Kalina T, Sterba J, Jabali Y, Mihal V, Blazek B, Cerna Z, Prochazkova D, Hak J, Zemanova Z, Jarosova M, Oltova A, Sedlacek P, Schwarz J, Zuna J, Trka J, Stary J, Hrusak O. Prognosis of children with mixed phenotype acute leukemia treated on the basis of consistent immunophenotypic criteria. Haematologica 2010; 95:928-35. [PMID: 20145275 DOI: 10.3324/haematol.2009.014506] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Mixed phenotype acute leukemia (MPAL) represents a diagnostic and therapeutic dilemma. The European Group for the Immunological Classification of Leukemias (EGIL) scoring system unambiguously defines MPAL expressing aberrant lineage markers. Discussions surrounding it have focused on scoring details, and information is limited regarding its biological, clinical and prognostic significance. The recent World Health Organization classification is simpler and could replace the EGIL scoring system after transformation into unambiguous guidelines. DESIGN AND METHODS Simple immunophenotypic criteria were used to classify all cases of childhood acute leukemia in order to provide therapy directed against acute lymphoblastic leukemia or acute myeloid leukemia. Prognosis, genotype and immunoglobulin/T-cell receptor gene rearrangement status were analyzed. RESULTS The incidences of MPAL were 28/582 and 4/107 for children treated with acute lymphoblastic leukemia and acute myeloid leukemia regimens, respectively. In immunophenotypic principal component analysis, MPAL treated as T-cell acute lymphoblastic leukemia clustered between cases of non-mixed T-cell acute lymphoblastic leukemia and acute myeloid leukemia, while other MPAL cases were included in the respective non-mixed B-cell progenitor acute lymphoblastic leukemia or acute myeloid leukemia clusters. Analogously, immunoglobulin/T-cell receptor gene rearrangements followed the expected pattern in patients treated as having acute myeloid leukemia (non-rearranged, 4/4) or as having B-cell progenitor acute lymphoblastic leukemia (rearranged, 20/20), but were missing in 3/5 analyzed cases of MPAL treated as having T-cell acute lymphobastic leukemia. In patients who received acute lymphoblastic leukemia treatment, the 5-year event-free survival of the MPAL cases was worse than that of the non-mixed cases (53+/-10% and 76+/-2% at 5 years, respectively, P=0.0075), with a more pronounced difference among B lineage cases. The small numbers of MPAL cases treated as T-cell acute lymphoblastic leukemia or as acute myeloid leukemia hampered separate statistics. We compared prognosis of all subsets with the prognosis of previously published cohorts. CONCLUSIONS Simple immunophenotypic criteria are useful for therapy decisions in MPAL. In B lineage leukemia, MPAL confers poorer prognosis. However, our data do not justify a preferential use of current acute myeloid leukemia-based therapy in MPAL.
Collapse
Affiliation(s)
- Ester Mejstrikova
- CLIP-Childhood Leukemia Investigation Prague Department of Pediatric Hematology and Oncology, Charles University, 2ndFaculty of Medicine and University Hospital Motol, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Legrand F, Driss V, Woerly G, Loiseau S, Hermann E, Fournié JJ, Héliot L, Mattot V, Soncin F, Gougeon ML, Dombrowicz D, Capron M. A functional gammadeltaTCR/CD3 complex distinct from gammadeltaT cells is expressed by human eosinophils. PLoS One 2009; 4:e5926. [PMID: 19536290 PMCID: PMC2693924 DOI: 10.1371/journal.pone.0005926] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 05/13/2009] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Eosinophils are effector cells during parasitic infections and allergic responses. However, their contribution to innate immunity has been only recently unravelled. METHODOLOGY/PRINCIPAL FINDINGS Here we show that human eosinophils express CD3 and gammadelta T Cell Receptor (TCR) but not alphabeta TCR. Surface expression of gammadeltaTCR/CD3 is heterogeneous between eosinophil donors and inducible by mycobacterial ligands. Surface immunoprecipitation revealed expression of the full gammadeltaTCR/CD3 complex. Real-time PCR amplification for CD3, gamma and delta TCR constant regions transcripts showed a significantly lower expression in eosinophils than in gammadeltaT cells. Limited TCR rearrangements occur in eosinophils as shown by spectratyping analysis of CDR3 length profiles and in situ hybridization. Release by eosinophils of Reactive Oxygen Species, granule proteins, Eosinophil Peroxidase and Eosinophil-Derived Neurotoxin and cytokines (IFN-gamma and TNF-alpha) was observed following activation by gammadeltaTCR-specific agonists or by mycobacteria. These effects were inhibited by anti-gammadeltaTCR blocking antibodies and antagonists. Moreover, gammadeltaTCR/CD3 was involved in eosinophil cytotoxicity against tumor cells. CONCLUSIONS/SIGNIFICANCE Our results provide evidence that human eosinophils express a functional gammadeltaTCR/CD3 with similar, but not identical, characteristics to gammadeltaTCR from gammadeltaT cells. We propose that this receptor contributes to eosinophil innate responses against mycobacteria and tumors and may represent an additional link between lymphoid and myeloid lineages.
Collapse
Affiliation(s)
- Fanny Legrand
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Virginie Driss
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Gaëtane Woerly
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Sylvie Loiseau
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Emmanuel Hermann
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | | | - Laurent Héliot
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
- CNRS UMR8161, Institut de Biologie de Lille, Lille, France
| | - Virginie Mattot
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
- CNRS UMR8161, Institut de Biologie de Lille, Lille, France
| | - Fabrice Soncin
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
- CNRS UMR8161, Institut de Biologie de Lille, Lille, France
| | | | - David Dombrowicz
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Monique Capron
- Inserm U547, Lille, France
- Université Lille - Nord de France, Lille, France
- Institut Pasteur de Lille, Lille, France
- * E-mail:
| |
Collapse
|
24
|
Pilbeam K, Basse P, Brossay L, Vujanovic N, Gerstein R, Vallejo AN, Borghesi L. The ontogeny and fate of NK cells marked by permanent DNA rearrangements. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:1432-41. [PMID: 18209038 PMCID: PMC4465768 DOI: 10.4049/jimmunol.180.3.1432] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A subset of NK cells bears incomplete V(D)J rearrangements, but neither the consequence to cell activities nor the precise developmental stages in which recombination occurs is known. These are important issues, as recombination errors cause cancers of the B and T lineages. Using transgenic recombination reporter mice to examine NK cell dynamics in vivo, we show that recombination(+) NK cells have distinct developmental patterns in the BM, including reduced homeostatic proliferation and diminished Stat5 phosphorylation. In the periphery, both recombination(+) and recombination(-) NK cells mediate robust functional responses including IFN-gamma production, cytolysis, and tumor homing, suggesting that NK cells with distinct developmental histories can be found together in the periphery. We also show that V(D)J rearrangement marks both human cytolytic (CD56(dim)) and immunoregulatory (CD56(bright)) populations, demonstrating the distribution of permanent DNA rearrangements across major NK cell subsets in man. Finally, direct quantification of rag transcripts throughout NK cell differentiation in both mouse and man establishes the specific developmental stages that are susceptible to V(D)J rearrangement. Together, these data demonstrate that multipotent progenitors rather than lineage-specified NK progenitors are targets of V(D)J recombination and that NK cells bearing the relics of earlier V(D)J rearrangements have different developmental dynamics but robust biological capabilities in vivo.
Collapse
Affiliation(s)
- Kristy Pilbeam
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Per Basse
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Nikola Vujanovic
- University of Pittsburgh Cancer Institute and Department of Pathology, Hillman Cancer Center, Pittsburgh, PA 15213
| | - Rachel Gerstein
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Abbe N. Vallejo
- Department of Pediatrics and Immunology, University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh Rangos Research Center, Pittsburgh, PA 15213
| | - Lisa Borghesi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
25
|
Lefort N, Benne C, Lelièvre JD, Dorival C, Balbo M, Sakano S, Coulombel L, Lévy Y. Short exposure to Notch ligand Delta-4 is sufficient to induce T-cell differentiation program and to increase the T cell potential of primary human CD34+ cells. Exp Hematol 2006; 34:1720-9. [PMID: 17157169 DOI: 10.1016/j.exphem.2006.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2006] [Revised: 08/09/2006] [Accepted: 08/10/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The Notch pathway plays a key role in cell fate choices and in T-cell development. The goal of our study was to evaluate whether a short in vitro stimulation of the Notch pathway may alter human progenitor cell behavior. METHODS CD34+ cord blood progenitors were exposed for 4 days to either immobilized Notch ligand Delta-4 or in control conditions. Phenotypic and molecular changes induced by the short stimulation were assessed at day 4. Next, long-term alteration of the fate of these progenitors was assessed in culture conditions suitable for B (coculture with MS5 stromal cells) and T (FTOC and OP9 stromal cells expressing Delta-4 systems) cell differentiation. RESULTS Notch activation was sufficient to trigger immunophenotypic and molecular changes consistent with early T-cell lineage differentiation. Delta-4 induced, in 4 days, CD7+cytCD3epsilon+ cells. This paralleled at the gene-transcription level with de novo expression of several T cell-related transcription factors and TCRgamma rearrangement, while B cell transcripts were simultaneous silenced. As compared to non-Delta-4 primed cells, these early changes translated to long-term alteration of the potential of cells. Delta-4 priming led to an acceleration of T-cell development, including a completion of the TCR rearrangement, when cells were cultured in systems suitable for T-cell development while B-cell development was inhibited. CONCLUSION A transient Notch activation is sufficient to promote T-cell differentiation from cord blood CD34+ cells. This system may be a useful tool for the amplification and the quantification of the T potential of CD34+ cells in various disease conditions.
Collapse
|