1
|
Blanco E, Camps C, Bahal S, Kerai MD, Ferla MP, Rochussen AM, Handel AE, Golwala ZM, Spiridou Goncalves H, Kricke S, Klein F, Zhang F, Zinghirino F, Evans G, Keane TM, Lizot S, Kusters MA, Iro MA, Patel SV, Morris EC, Burns SO, Radcliffe R, Vasudevan P, Price A, Gillham O, Valdebenito GE, Stewart GS, Worth A, Adams SP, Duchen M, André I, Adams DJ, Santili G, Gilmour KC, Holländer GA, Davies EG, Taylor JC, Griffiths GM, Thrasher AJ, Dhalla F, Kreins AY. Dominant negative variants in ITPR3 impair T cell Ca2+ dynamics causing combined immunodeficiency. J Exp Med 2025; 222:e20220979. [PMID: 39560673 PMCID: PMC11577440 DOI: 10.1084/jem.20220979] [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: 06/06/2022] [Revised: 09/09/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
The importance of calcium (Ca2+) as a second messenger in T cell signaling is exemplified by genetic deficiencies of STIM1 and ORAI1, which abolish store-operated Ca2+ entry (SOCE) resulting in combined immunodeficiency (CID). We report five unrelated patients with de novo missense variants in ITPR3, encoding a subunit of the inositol 1,4,5-trisphosphate receptor (IP3R), which forms a Ca2+ channel in the endoplasmic reticulum (ER) membrane responsible for the release of ER Ca2+ required to trigger SOCE, and for Ca2+ transfer to other organelles. The patients presented with CID, abnormal T cell Ca2+ homeostasis, incompletely penetrant ectodermal dysplasia, and multisystem disease. Their predominant T cell immunodeficiency is characterized by significant T cell lymphopenia, defects in late stages of thymic T cell development, and impaired function of peripheral T cells, including inadequate NF-κB- and NFAT-mediated, proliferative, and metabolic responses to activation. Pathogenicity is not due to haploinsufficiency, rather ITPR3 protein variants interfere with IP3R channel function leading to depletion of ER Ca2+ stores and blunted SOCE in T cells.
Collapse
Affiliation(s)
- Elena Blanco
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Carme Camps
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sameer Bahal
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mohit D. Kerai
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Immunology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Matteo P. Ferla
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Adam M. Rochussen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Adam E. Handel
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Zainab M. Golwala
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Helena Spiridou Goncalves
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Susanne Kricke
- SIHMDS-Haematology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fabian Klein
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Fang Zhang
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Federica Zinghirino
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Grace Evans
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Thomas M. Keane
- Wellcome Sanger Institute, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sabrina Lizot
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | - Maaike A.A. Kusters
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Mildred A. Iro
- Department of Paediatric Infectious Diseases and Immunology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Faculty of Medicine and Institute of Life Sciences, University of Southampton, Southampton, UK
| | - Sanjay V. Patel
- Department of Paediatric Infectious Diseases and Immunology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Emma C. Morris
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
- Institute for Immunity and Transplantation, University College London, London, UK
| | - Siobhan O. Burns
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
- Institute for Immunity and Transplantation, University College London, London, UK
| | - Ruth Radcliffe
- Department of Immunology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Pradeep Vasudevan
- Department of Clinical Genetics, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Arthur Price
- Department of Immunology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Olivia Gillham
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Gabriel E. Valdebenito
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Grant S. Stewart
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Austen Worth
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Stuart P. Adams
- SIHMDS-Haematology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Michael Duchen
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Isabelle André
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | | | - Giorgia Santili
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Kimberly C. Gilmour
- Immunology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Georg A. Holländer
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children’s Hospital, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - E. Graham Davies
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jenny C. Taylor
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gillian M. Griffiths
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Adrian J. Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fatima Dhalla
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Clinical Immunology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alexandra Y. Kreins
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
2
|
Yu H, Yang W, Cao M, Lei Q, Yuan R, Xu H, Cui Y, Chen X, Su X, Zhuo H, Lin L. Mechanism study of ubiquitination in T cell development and autoimmune disease. Front Immunol 2024; 15:1359933. [PMID: 38562929 PMCID: PMC10982411 DOI: 10.3389/fimmu.2024.1359933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
T cells play critical role in multiple immune processes including antigen response, tumor immunity, inflammation, self-tolerance maintenance and autoimmune diseases et. Fetal liver or bone marrow-derived thymus-seeding progenitors (TSPs) settle in thymus and undergo T cell-lineage commitment, proliferation, T cell receptor (TCR) rearrangement, and thymic selections driven by microenvironment composed of thymic epithelial cells (TEC), dendritic cells (DC), macrophage and B cells, thus generating T cells with diverse TCR repertoire immunocompetent but not self-reactive. Additionally, some self-reactive thymocytes give rise to Treg with the help of TEC and DC, serving for immune tolerance. The sequential proliferation, cell fate decision, and selection during T cell development and self-tolerance establishment are tightly regulated to ensure the proper immune response without autoimmune reaction. There are remarkable progresses in understanding of the regulatory mechanisms regarding ubiquitination in T cell development and the establishment of self-tolerance in the past few years, which holds great potential for further therapeutic interventions in immune-related diseases.
Collapse
Affiliation(s)
- Hui Yu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Wenyong Yang
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Min Cao
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Qingqiang Lei
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Renbin Yuan
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - He Xu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yuqian Cui
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xuerui Chen
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xu Su
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Hui Zhuo
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Liangbin Lin
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| |
Collapse
|
3
|
Bacsa B, Hopl V, Derler I. Synthetic Biology Meets Ca 2+ Release-Activated Ca 2+ Channel-Dependent Immunomodulation. Cells 2024; 13:468. [PMID: 38534312 DOI: 10.3390/cells13060468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Many essential biological processes are triggered by the proximity of molecules. Meanwhile, diverse approaches in synthetic biology, such as new biological parts or engineered cells, have opened up avenues to precisely control the proximity of molecules and eventually downstream signaling processes. This also applies to a main Ca2+ entry pathway into the cell, the so-called Ca2+ release-activated Ca2+ (CRAC) channel. CRAC channels are among other channels are essential in the immune response and are activated by receptor-ligand binding at the cell membrane. The latter initiates a signaling cascade within the cell, which finally triggers the coupling of the two key molecular components of the CRAC channel, namely the stromal interaction molecule, STIM, in the ER membrane and the plasma membrane Ca2+ ion channel, Orai. Ca2+ entry, established via STIM/Orai coupling, is essential for various immune cell functions, including cytokine release, proliferation, and cytotoxicity. In this review, we summarize the tools of synthetic biology that have been used so far to achieve precise control over the CRAC channel pathway and thus over downstream signaling events related to the immune response.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Division of Medical Physics und Biophysics, Medical University of Graz, A-8010 Graz, Austria
| | - Valentina Hopl
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
4
|
Peng Z, Zhang H, Hu H. The Function of Ubiquitination in T-Cell Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1466:135-159. [PMID: 39546141 DOI: 10.1007/978-981-97-7288-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Thymus is an important primary lymphoid organ for T cell development. After T-lineage commitment, the early thymic progenitors (ETPs) develop into CD4-CD8- (DN), CD4+CD8+ (DP) and further CD4+ SP or CD8+ SP T cells. Under the help of thymic epithelial cells (TEC), dendritic cell (DC), macrophage, and B cells, ETPs undergo proliferation, T cell receptor (TCR) rearrangement, β-selection, positive selection, and negative selection, and thus leading to the generation of T cells that are diverse repertoire immunocompetent but not self-reactive. Additionally, some self-reactive thymocytes give rise to Treg under the help of TEC and DC. The regulation of T cell development is complicated. As a post-translational modification, ubiquitination regulates signal transduction in diverse biological processes. Ubiquitination functions in T cell development through regulating key signal pathway or maturation and function of related cells. In this review, the regulation of T cell development by ubiquitination is summarized and discussed.
Collapse
Affiliation(s)
- Zhengcan Peng
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
5
|
Hasanoor Reja AH, De A, Chakraborty D, Ahmed SS, Sarda A. A Cross-Sectional Study to Evaluate the Alteration of Cytokine Expression and Activation of Inflammatory Pathway in Response to NOD1 and NOD2 Signal in Leprosy. Indian J Dermatol 2023; 68:724. [PMID: 38371539 PMCID: PMC10868971 DOI: 10.4103/ijd.ijd_386_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024] Open
Abstract
Introduction Leprae bacilli are identified as foreign by pattern recognition receptors (PRRs) present in the microbes but absent in the host. The Nucleotide oligomerization domain (NOD)-like receptor (NLR) family comprises the nucleotide-binding oligomerisation domain (NOD1 and NOD2) proteins, which are two well-known PRRs. The objectives of this study were to study the expression of cytoplasmic NOD1 and NOD2 in the pathogenesis of leprosy and the serum level of expressed cytokines and to measure the messenger Ribonucleic Acid (mRNA) expression. Methods Clinically suspected Hansen's patients were analysed for 4 years. Newly diagnosed leprosy patients were considered leprosy disease control (LDC). The cases with active or new lesions and an increase in Bacteriological index (BI) by at least 2 + after 12 months of completion of Multidrug therapy (MDT) were considered leprosy disease relapse (LDR) cases. Age- and sex-matched healthy individuals served as our control group (healthy control (HC)). enzyme-linked immunosorbent assay (ELISA) was performed to measure the concentration of five human cytokines in serum, including three pro-inflammatory cytokines (Tumor necrosis factor (TNF)-α, Interferon gamma (IFN-γ) and IL-6), one anti-inflammatory cytokine (IL-10) and one chemokine (IL-8). Quantitative expression of receptor genes (NOD1 and NOD2) and cytokine genes (TNF-α, IFN-γ, IL-6, IL-10 and IL-8) was evaluated by quantitative real-time polymerase chain reaction (PCR) (qRT-PCR). We studied NOD1 and NOD2 expression in the tissues through fluorescence immunohistochemistry. Differential NLR intracellular expression on peripheral blood monocytes (PBMs) and their response to stimulation with specific ligands (lipopolysaccharide (LPS) and muramyl dipeptide (MDP)) were studied. Results A significant difference in the expression of the NOD1 gene was observed in unstimulated monocytes of the LDC and LDR cases when compared to HC. The NOD2 transcript level was significantly higher in stimulated monocytes from LDC and LDR patients than in similarly stimulated cells from HC. The LDC patients had a significantly higher level of pro-inflammatory cytokines as compared to the HC. Conclusion In conclusion, this study has demonstrated the expression of both cytokines and chemokines in response to NLR activation in the skin of leprosy patients.
Collapse
Affiliation(s)
| | - Abhishek De
- Department of Dermatology, Calcutta National Medical College, Kolkata, West Bengal, India
| | - Disha Chakraborty
- Department of Dermatology, Calcutta National Medical College, Kolkata, West Bengal, India
| | - Sk. S. Ahmed
- Department of Dermatology, Calcutta National Medical College, Kolkata, West Bengal, India
| | - Aarti Sarda
- Department of Dermatology, Wizderm Skin and Hair Specialty Clinic, Kolkata, West Bengal, India
| |
Collapse
|
6
|
Notch Signaling in Acute Inflammation and Sepsis. Int J Mol Sci 2023; 24:ijms24043458. [PMID: 36834869 PMCID: PMC9967996 DOI: 10.3390/ijms24043458] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Notch signaling, a highly conserved pathway in mammals, is crucial for differentiation and homeostasis of immune cells. Besides, this pathway is also directly involved in the transmission of immune signals. Notch signaling per se does not have a clear pro- or anti-inflammatory effect, but rather its impact is highly dependent on the immune cell type and the cellular environment, modulating several inflammatory conditions including sepsis, and therefore significantly impacts the course of disease. In this review, we will discuss the contribution of Notch signaling on the clinical picture of systemic inflammatory diseases, especially sepsis. Specifically, we will review its role during immune cell development and its contribution to the modulation of organ-specific immune responses. Finally, we will evaluate to what extent manipulation of the Notch signaling pathway could be a future therapeutic strategy.
Collapse
|
7
|
Calcagno G, Ouzren N, Kaminski S, Ghislin S, Frippiat JP. Chronic Hypergravity Induces a Modification of Histone H3 Lysine 27 Trimethylation at TCRβ Locus in Murine Thymocytes. Int J Mol Sci 2022; 23:7133. [PMID: 35806138 PMCID: PMC9267123 DOI: 10.3390/ijms23137133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Gravity changes are major stressors encountered during spaceflight that affect the immune system. We previously evidenced that hypergravity exposure during gestation affects the TCRβ repertoire of newborn pups. To identify the mechanisms underlying this observation, we studied post-translational histone modifications. We first showed that among the four studied post-translational histone H3 modifications, only lysine 27 trimethylation (H3K27me3) is downregulated in the thymus of mice exposed to 2× g for 21 days. We then asked whether the TCRβ locus chromatin structure is altered by hypergravity exposure. ChIP studies performed on four Vβ segments of the murine double-negative SCIET27 thymic cell line, which corresponds to the last maturation stage before V(D)J recombination, revealed increases in H3K27me3 after 2× g exposure. Finally, we evaluated the implication for the EZH2 methyltransferase in the regulation of the H3K27me3 level at these Vβ segments by treating SCIET27 cells with the GSK126-specific inhibitor. These experiments showed that the downregulation of H3K27me3 contributes to the regulation of the Vβ germline transcript expression that precedes V(D)J recombination. These data show that modifications of H3K27me3 at the TCRβ locus likely contribute to an explanation of why the TCR repertoire is affected by gravity changes and imply, for the first time, EZH2 in the regulation of the TCRβ locus chromatin structure.
Collapse
|
8
|
Link between the EZH2 noncanonical pathway and microtubule organization center polarization during early T lymphopoiesis. Sci Rep 2022; 12:3655. [PMID: 35256668 PMCID: PMC8901749 DOI: 10.1038/s41598-022-07684-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/22/2022] [Indexed: 12/11/2022] Open
Abstract
EZH2 plays an essential role at the β-selection checkpoint of T lymphopoiesis by regulating histone H3 lysine 27 trimethylation (H3K27me3) via its canonical mode of action. Increasing data suggest that EZH2 could also regulate other cellular functions, such as cytoskeletal reorganization, via its noncanonical pathway. Consequently, we investigated whether the EZH2 noncanonical pathway could be involved in early T-cell maturation, which requires cell polarization. We observed that EZH2 localization is tightly regulated during the early stages of T-cell development and that EZH2 relocalizes in the nucleus of double-negative thymocytes enduring TCRβ recombination and β-selection processes. Furthermore, we observed that EZH2 and EED, but not Suz12, colocalize with the microtubule organization center (MTOC), which might prevent its inappropriate polarization in double negative cells. In accordance with these results, we evidenced the existence of direct or indirect interaction between EED and α-tubulin. Taken together, these results suggest that the EZH2 noncanonical pathway, in association with EED, is involved in the early stages of T-cell maturation.
Collapse
|
9
|
Giang N, Mars M, Moreau M, Mejia JE, Bouchaud G, Magnan A, Michelet M, Ronsin B, Murphy GG, Striessnig J, Guéry J, Pelletier L, Savignac M. Separation of the Ca V 1.2-Ca V 1.3 calcium channel duo prevents type 2 allergic airway inflammation. Allergy 2022; 77:525-539. [PMID: 34181765 DOI: 10.1111/all.14993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/16/2021] [Accepted: 05/16/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Voltage-gated calcium (Cav 1) channels contribute to T-lymphocyte activation. Cav 1.2 and Cav 1.3 channels are expressed in Th2 cells but their respective roles are unknown, which is investigated herein. METHODS We generated mice deleted for Cav 1.2 in T cells or Cav 1.3 and analyzed TCR-driven signaling. In this line, we developed original fast calcium imaging to measure early elementary calcium events (ECE). We also tested the impact of Cav 1.2 or Cav 1.3 deletion in models of type 2 airway inflammation. Finally, we checked whether the expression of both Cav 1.2 and Cav 1.3 in T cells from asthmatic children correlates with Th2-cytokine expression. RESULTS We demonstrated non-redundant and synergistic functions of Cav 1.2 and Cav 1.3 in Th2 cells. Indeed, the deficiency of only one channel in Th2 cells triggers TCR-driven hyporesponsiveness with weakened tyrosine phosphorylation profile, a strong decrease in initial ECE and subsequent reduction in the global calcium response. Moreover, Cav 1.3 has a particular role in calcium homeostasis. In accordance with the singular roles of Cav 1.2 and Cav 1.3 in Th2 cells, deficiency in either one of these channels was sufficient to inhibit cardinal features of type 2 airway inflammation. Furthermore, Cav 1.2 and Cav 1.3 must be co-expressed within the same CD4+ T cell to trigger allergic airway inflammation. Accordingly with the concerted roles of Cav 1.2 and Cav 1.3, the expression of both channels by activated CD4+ T cells from asthmatic children was associated with increased Th2-cytokine transcription. CONCLUSIONS Thus, Cav 1.2 and Cav 1.3 act as a duo, and targeting only one of these channels would be efficient in allergy treatment.
Collapse
Affiliation(s)
- Nicolas Giang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Marion Mars
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Marc Moreau
- Centre de Biologie du Développement Centre de Biologie Intégrative Université de ToulouseCNRSUniversité Paul Sabatier III Toulouse France
| | - Jose E. Mejia
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | | | - Antoine Magnan
- Institut du Thorax INSERM CNRSUniversité de Nantes Nantes France
- Service de Pneumologie Centre Hospitalier Universitaire de Nantes Nantes France
| | - Marine Michelet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
- Pediatric Pneumology and Allergology Unit Hôpital des EnfantsCentre Hospitalier Universitaire Toulouse Toulouse France
- Unité de Recherche Clinique Pédiatrique/module plurithématique pédiatrique du CIC Toulouse France
| | - Brice Ronsin
- Centre de Biologie du Développement Centre de Biologie Intégrative Université de ToulouseCNRSUniversité Paul Sabatier III Toulouse France
| | - Geoffrey G. Murphy
- Molecular and Behavioral Neuroscience Institute University of Michigan Ann Arbor MI USA
| | - Joerg Striessnig
- Department of Pharmacology and Toxicology Institute of Pharmacy Center for Molecular Biosciences University of Innsbruck Innsbruck Austria
| | - Jean‐Charles Guéry
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Lucette Pelletier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Magali Savignac
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| |
Collapse
|
10
|
Allam AH, Charnley M, Pham K, Russell SM. Developing T cells form an immunological synapse for passage through the β-selection checkpoint. J Cell Biol 2021; 220:e201908108. [PMID: 33464309 PMCID: PMC7814350 DOI: 10.1083/jcb.201908108] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/22/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
The β-selection checkpoint of T cell development tests whether the cell has recombined its genomic DNA to produce a functional T cell receptor β (TCRβ). Passage through the β-selection checkpoint requires the nascent TCRβ protein to mediate signaling through a pre-TCR complex. In this study, we show that developing T cells at the β-selection checkpoint establish an immunological synapse in in vitro and in situ, resembling that of the mature T cell. The immunological synapse is dependent on two key signaling pathways known to be critical for the transition beyond the β-selection checkpoint, Notch and CXCR4 signaling. In vitro and in situ analyses indicate that the immunological synapse promotes passage through the β-selection checkpoint. Collectively, these data indicate that developing T cells regulate pre-TCR signaling through the formation of an immunological synapse. This signaling platform integrates cues from Notch, CXCR4, and MHC on the thymic stromal cell to allow transition beyond the β-selection checkpoint.
Collapse
Affiliation(s)
- Amr H. Allam
- Optical Sciences Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Mirren Charnley
- Optical Sciences Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Kim Pham
- Optical Sciences Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Department of Pathology, The University of Melbourne, Australia
| | - Sarah M. Russell
- Optical Sciences Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Department of Pathology, The University of Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Australia
| |
Collapse
|
11
|
Vaeth M, Kahlfuss S, Feske S. CRAC Channels and Calcium Signaling in T Cell-Mediated Immunity. Trends Immunol 2020; 41:878-901. [PMID: 32711944 DOI: 10.1016/j.it.2020.06.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022]
Abstract
Calcium (Ca2+) signals play fundamental roles in immune cell function. The main sources of Ca2+ influx in mammalian lymphocytes following antigen receptor stimulation are Ca2+ release-activated Ca2+ (CRAC) channels. These are formed by ORAI proteins in the plasma membrane and are activated by stromal interaction molecules (STIM) located in the endoplasmic reticulum (ER). Human loss-of-function (LOF) mutations in ORAI1 and STIM1 that abolish Ca2+ influx cause a unique disease syndrome called CRAC channelopathy that is characterized by immunodeficiency autoimmunity and non-immunological symptoms. Studies in mice lacking Stim and Orai genes have illuminated many cellular and molecular mechanisms by which these molecules control lymphocyte function. CRAC channels are required for the differentiation and function of several T lymphocyte subsets that provide immunity to infection, mediate inflammation and prevent autoimmunity. This review examines new insights into how CRAC channels control T cell-mediated immunity.
Collapse
Affiliation(s)
- Martin Vaeth
- Institute of Systems Immunology, Julius-Maximilians University of Würzburg, Würzburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Peters C, Kouakanou L, Kabelitz D. A comparative view on vitamin C effects on αβ- versus γδ T-cell activation and differentiation. J Leukoc Biol 2020; 107:1009-1022. [PMID: 32034803 DOI: 10.1002/jlb.1mr1219-245r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/06/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
Vitamin C (VitC) is an essential vitamin that needs to be provided through exogenous sources. It is a potent anti-oxidant, and an essential cofactor for many enzymes including a group of enzymes that modulate epigenetic regulation of gene expression. Moreover, VitC has a significant influence on T-cell differentiation, and can directly interfere with T-cell signaling. Conventional CD4 and CD8 T cells express the αβ TCR and recognize peptide antigens in the context of MHC presentation. The numerically small population of γδ T cells recognizes antigens in an MHC-independent manner. γδ T cells kill a broad variety of malignant cells, and because of their unique features, are interesting candidates for cancer immunotherapy. In this review, we summarize what is known about the influence of VitC on T-cell activation and differentiation with a special focus on γδ T cells. The known mechanisms of action of VitC on αβ T cells are discussed and extrapolated to the effects observed on γδ T-cell activation and differentiation. Overall, VitC enhances proliferation and effector functions of γδ T cells and thus may help to increase the efficacy of γδ T cells applied as cancer immunotherapy in adoptive cell transfer.
Collapse
Affiliation(s)
- Christian Peters
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Léonce Kouakanou
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
13
|
Abstract
Metals are essential components in all forms of life required for the function of nearly half of all enzymes and are critically involved in virtually all fundamental biological processes. Especially, the transition metals iron (Fe), zinc (Zn), manganese (Mn), nickel (Ni), copper (Cu) and cobalt (Co) are crucial micronutrients known to play vital roles in metabolism as well due to their unique redox properties. Metals carry out three major functions within metalloproteins: to provide structural support, to serve as enzymatic cofactors, and to mediate electron transportation. Metal ions are also involved in the immune system from metal allergies to nutritional immunity. Within the past decade, much attention has been drawn to the roles of metal ions in the immune system, since increasing evidence has mounted to suggest that metals are critically implicated in regulating both the innate immune sensing of and the host defense against invading pathogens. The importance of ions in immunity is also evidenced by the identification of various immunodeficiencies in patients with mutations in ion channels and transporters. In addition, cancer immunotherapy has recently been conclusively demonstrated to be effective and important for future tumor treatment, although only a small percentage of cancer patients respond to immunotherapy because of inadequate immune activation. Importantly, metal ion-activated immunotherapy is becoming an effective and potential way in tumor therapy for better clinical application. Nevertheless, we are still in a primary stage of discovering the diverse immunological functions of ions and mechanistically understanding the roles of these ions in immune regulation. This review summarizes recent advances in the understanding of metal-controlled immunity. Particular emphasis is put on the mechanisms of innate immune stimulation and T cell activation by the essential metal ions like calcium (Ca2+), zinc (Zn2+), manganese (Mn2+), iron (Fe2+/Fe3+), and potassium (K+), followed by a few unessential metals, in order to draw a general diagram of metalloimmunology.
Collapse
Affiliation(s)
- Chenguang Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rui Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xiaoming Wei
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Mengze Lv
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
14
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 PMCID: PMC6669380 DOI: 10.3389/fimmu.2019.01704] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 01/07/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
An integrated transcriptional switch at the β-selection checkpoint determines T cell survival, development and leukaemogenesis. Biochem Soc Trans 2019; 47:1077-1089. [DOI: 10.1042/bst20180414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023]
Abstract
Abstract
In T cell development, a pivotal decision-making stage, termed β-selection, integrates a TCRβ checkpoint to coordinate survival, proliferation and differentiation to an αβ T cell. Here, we review how transcriptional regulation coordinates fate determination in early T cell development to enable β-selection. Errors in this transcription control can trigger T cell acute lymphoblastic leukaemia. We describe how the β-selection checkpoint goes awry in leukaemic transformation.
Collapse
|
16
|
Saadi W, Kermezli Y, Dao LTM, Mathieu E, Santiago-Algarra D, Manosalva I, Torres M, Belhocine M, Pradel L, Loriod B, Aribi M, Puthier D, Spicuglia S. A critical regulator of Bcl2 revealed by systematic transcript discovery of lncRNAs associated with T-cell differentiation. Sci Rep 2019; 9:4707. [PMID: 30886319 PMCID: PMC6423290 DOI: 10.1038/s41598-019-41247-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Normal T-cell differentiation requires a complex regulatory network which supports a series of maturation steps, including lineage commitment, T-cell receptor (TCR) gene rearrangement, and thymic positive and negative selection. However, the underlying molecular mechanisms are difficult to assess due to limited T-cell models. Here we explore the use of the pro-T-cell line P5424 to study early T-cell differentiation. Stimulation of P5424 cells by the calcium ionophore ionomycin together with PMA resulted in gene regulation of T-cell differentiation and activation markers, partially mimicking the CD4-CD8- double negative (DN) to double positive (DP) transition and some aspects of subsequent T-cell maturation and activation. Global analysis of gene expression, along with kinetic experiments, revealed a significant association between the dynamic expression of coding genes and neighbor lncRNAs including many newly-discovered transcripts, thus suggesting potential co-regulation. CRISPR/Cas9-mediated genetic deletion of Robnr, an inducible lncRNA located downstream of the anti-apoptotic gene Bcl2, demonstrated a critical role of the Robnr locus in the induction of Bcl2. Thus, the pro-T-cell line P5424 is a powerful model system to characterize regulatory networks involved in early T-cell differentiation and maturation.
Collapse
Affiliation(s)
- Wiam Saadi
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Yasmina Kermezli
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Lan T M Dao
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Vinmec Research Institute of Stem cell and Gene technology (VRISG), Hanoi, Vietnam
| | - Evelyne Mathieu
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - David Santiago-Algarra
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Iris Manosalva
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Magali Torres
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Mohamed Belhocine
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Molecular Biology and Genetics Laboratory, Dubai, United Arab Emirates
| | - Lydie Pradel
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Beatrice Loriod
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Denis Puthier
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| |
Collapse
|
17
|
Yang K, Blanco DB, Chen X, Dash P, Neale G, Rosencrance C, Easton J, Chen W, Cheng C, Dhungana Y, Kc A, Awad W, Guo XZJ, Thomas PG, Chi H. Metabolic signaling directs the reciprocal lineage decisions of αβ and γδ T cells. Sci Immunol 2019; 3:3/25/eaas9818. [PMID: 29980617 DOI: 10.1126/sciimmunol.aas9818] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/27/2018] [Indexed: 01/07/2023]
Abstract
The interaction between extrinsic factors and intrinsic signal strength governs thymocyte development, but the mechanisms linking them remain elusive. We report that mechanistic target of rapamycin complex 1 (mTORC1) couples microenvironmental cues with metabolic programs to orchestrate the reciprocal development of two fundamentally distinct T cell lineages, the αβ and γδ T cells. Developing thymocytes dynamically engage metabolic programs including glycolysis and oxidative phosphorylation, as well as mTORC1 signaling. Loss of RAPTOR-mediated mTORC1 activity impairs the development of αβ T cells but promotes γδ T cell generation, associated with disrupted metabolic remodeling of oxidative and glycolytic metabolism. Mechanistically, we identify mTORC1-dependent control of reactive oxygen species production as a key metabolic signal in mediating αβ and γδ T cell development, and perturbation of redox homeostasis impinges upon thymocyte fate decisions and mTORC1-associated phenotypes. Furthermore, single-cell RNA sequencing and genetic dissection reveal that mTORC1 links developmental signals from T cell receptors and NOTCH to coordinate metabolic activity and signal strength. Our results establish mTORC1-driven metabolic signaling as a decisive factor for reciprocal αβ and γδ T cell development and provide insight into metabolic control of cell signaling and fate decisions.
Collapse
Affiliation(s)
- Kai Yang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Daniel Bastardo Blanco
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Pradyot Dash
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Celeste Rosencrance
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenan Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Changde Cheng
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yogesh Dhungana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anil Kc
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Walid Awad
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xi-Zhi J Guo
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
18
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 DOI: 10.3389/fimmu.2019.01704/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 05/22/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
19
|
Ferrandino F, Grazioli P, Bellavia D, Campese AF, Screpanti I, Felli MP. Notch and NF-κB: Coach and Players of Regulatory T-Cell Response in Cancer. Front Immunol 2018; 9:2165. [PMID: 30364244 PMCID: PMC6193072 DOI: 10.3389/fimmu.2018.02165] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
The Notch signaling pathway plays multiple roles in driving T-cell fate decisions, proliferation, and aberrant growth. NF-κB is a cell-context key player interconnected with Notch signaling either in physiological or in pathological conditions. This review focuses on how the multilayered crosstalk between different Notches and NF-κB subunits may converge on Foxp3 gene regulation and orchestrate CD4+ regulatory T (Treg) cell function, particularly in a tumor microenvironment. Notably, Treg cells may play a pivotal role in the inhibition of antitumor immune responses, possibly promoting tumor growth. A future challenge is represented by further dissection of both Notch and NF-κB pathways and consequences of their intersection in tumor-associated Treg biology. This may shed light on the molecular mechanisms regulating Treg cell expansion and migration to peripheral lymphoid organs thought to facilitate tumor development and still to be explored. In so doing, new opportunities for combined and/or more selective therapeutic approaches to improve anticancer immunity may be found.
Collapse
Affiliation(s)
| | - Paola Grazioli
- Department of Experimental Medicine, La Sapienza University, Rome, Italy
| | - Diana Bellavia
- Department of Molecular Medicine, La Sapienza University, Rome, Italy
| | | | | | - Maria Pia Felli
- Department of Experimental Medicine, La Sapienza University, Rome, Italy
| |
Collapse
|
20
|
Bellavia D, Checquolo S, Palermo R, Screpanti I. The Notch3 Receptor and Its Intracellular Signaling-Dependent Oncogenic Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:205-222. [PMID: 30030828 DOI: 10.1007/978-3-319-89512-3_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During evolution, gene duplication of the Notch receptor suggests a progressive functional diversification. The Notch3 receptor displays a number of structural differences with respect to Notch1 and Notch2, most of which have been reported in the transmembrane and in the intracellular regions, mainly localized in the negative regulatory region (NRR) and trans-activation domain (TAD). Targeted deletion of Notch3 does not result in embryonic lethality, which is in line with its highly restricted tissue expression pattern. Importantly, deregulated Notch3 expression and/or activation, often results in disrupted cell differentiation and/or pathological development, most notably in oncogenesis in different cell contexts. Mechanistically this is due to Notch3-related genetic alterations or epigenetic or posttranslational control mechanisms. In this chapter we discuss the possible relationships between the structural differences and the pathological role of Notch3 in the control of mouse and human cancers. In future, targeting the unique features of Notch3-oncogenic mechanisms could be exploited to develop anticancer therapeutics.
Collapse
Affiliation(s)
- Diana Bellavia
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Rocco Palermo
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
21
|
Guan S, Yu X, Fang B, Huang Y, Xu L, Lu J. The toxicity of 3-monochloro-1,2-propanediol (+) to activated T cells in mice. FOOD AGR IMMUNOL 2017. [DOI: 10.1080/09540105.2017.1309357] [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
Affiliation(s)
- Shuang Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Xin Yu
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Baochen Fang
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Yixuan Huang
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Linli Xu
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
22
|
Guan S, Huang Y, Feng Z, Xu L, Jin Y, Lu J. The toxic effects of benzo[a]pyrene on activated mouse T cells in vitro. Immunopharmacol Immunotoxicol 2017; 39:117-123. [DOI: 10.1080/08923973.2017.1299173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Shuang Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- College of Food Science and Engineering, Jilin University, Changchun, People’s Republic of China
| | - Yixuan Huang
- College of Food Science and Engineering, Jilin University, Changchun, People’s Republic of China
| | - Zhe Feng
- College of Food Science and Engineering, Jilin University, Changchun, People’s Republic of China
| | - Linli Xu
- College of Food Science and Engineering, Jilin University, Changchun, People’s Republic of China
| | - Yingli Jin
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Jing Lu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- College of Food Science and Engineering, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
23
|
Abstract
In human T cell development, the mechanisms that regulate cell fate decisions after TCRβ expression remain unclear. We defined the stages of T cell development that flank TCRβ expression and found distinct patterns of human T cell development. In half the subjects, T cell development progressed from the CD4(-)CD8(-) double-negative stage to the CD4(+)CD8(+) double-positive (DP) stage through an immature single-positive (ISP) CD4(+) intermediate. However, in some patients, CD4 and CD8 were expressed simultaneously and the ISP population was small. In each group of patients, CD3(-) ISP and DP thymocytes were subdivided into ISP1, ISP2, DP1, DP2, DP3, DP4, and DP5 developmental stages according to their expression of CD28, CD44, CD1a, CD7, CD45RO, and CD38. The ISP2, DP2, and DP3 thymocyte populations proliferated more robustly than ISP1 and DP1 and expressed markers consistent with TCRβ expression. After the DP3 stage, proliferation returned to baseline levels. We then analyzed protein levels of Ikaros, Helios, and Aiolos, the three Ikaros family members most abundantly expressed in human thymocytes. Ikaros and Helios expression increased transiently at the ISP2, DP2, and DP3 populations. Aiolos expression also increased at the ISP2, DP2, and DP3 stages, but its expression remained elevated throughout the DP4 and DP5 stages. In summary, we propose a model of human T cell development that reflects the asynchronous nature of TCRβ expression and we define the subpopulations of thymocytes that are highly proliferative and express Ikaros family members.
Collapse
|
24
|
Fu G, Yu M, Chen Y, Zheng Y, Zhu W, Newman DK, Wang D, Wen R. Phospholipase Cγ1 is required for pre-TCR signal transduction and pre-T cell development. Eur J Immunol 2016; 47:74-83. [PMID: 27759161 DOI: 10.1002/eji.201646522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/14/2016] [Accepted: 10/17/2016] [Indexed: 12/23/2022]
Abstract
Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.
Collapse
Affiliation(s)
- Guoping Fu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Mei Yu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yuhong Chen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yongwei Zheng
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Wen Zhu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Interdisciplinary Program in Biomedical Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Debra K Newman
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Demin Wang
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Renren Wen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
Sahni H, Ross S, Barbarulo A, Solanki A, Lau CI, Furmanski A, Saldaña JI, Ono M, Hubank M, Barenco M, Crompton T. A genome wide transcriptional model of the complex response to pre-TCR signalling during thymocyte differentiation. Oncotarget 2016; 6:28646-60. [PMID: 26415229 PMCID: PMC4745683 DOI: 10.18632/oncotarget.5796] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/08/2015] [Indexed: 01/19/2023] Open
Abstract
Developing thymocytes require pre-TCR signalling to differentiate from CD4-CD8- double negative to CD4+CD8+ double positive cell. Here we followed the transcriptional response to pre-TCR signalling in a synchronised population of differentiating double negative thymocytes. This time series analysis revealed a complex transcriptional response, in which thousands of genes were up and down-regulated before changes in cell surface phenotype were detected. Genome-wide measurement of RNA degradation of individual genes showed great heterogeneity in the rate of degradation between different genes. We therefore used time course expression and degradation data and a genome wide transcriptional modelling (GWTM) strategy to model the transcriptional response of genes up-regulated on pre-TCR signal transduction. This analysis revealed five major temporally distinct transcriptional activities that up regulate transcription through time, whereas down-regulation of expression occurred in three waves. Our model thus placed known regulators in a temporal perspective, and in addition identified novel candidate regulators of thymocyte differentiation.
Collapse
Affiliation(s)
- Hemant Sahni
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Susan Ross
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Anisha Solanki
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ching-In Lau
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Anna Furmanski
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Masahiro Ono
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mike Hubank
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Martino Barenco
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Tessa Crompton
- Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
26
|
Luo T, Ji WJ, Yuan F, Guo ZZ, Li YX, Dong Y, Ma YQ, Zhou X, Li YM. Th17/Treg Imbalance Induced by Dietary Salt Variation Indicates Inflammation of Target Organs in Humans. Sci Rep 2016; 6:26767. [PMID: 27353721 PMCID: PMC4926124 DOI: 10.1038/srep26767] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/09/2016] [Indexed: 01/18/2023] Open
Abstract
The functions of T helper 17 (Th17) and regulatory T (Treg) cells are tightly orchestrated through independent differentiation pathways that are involved in the secretion of pro- and anti-inflammatory cytokines induced by high-salt dietary. However, the role of imbalanced Th17/Treg ratio implicated in inflammation and target organ damage remains elusive. Here, by flow cytometry analysis, we demonstrated that switching to a high-salt diet resulted in decreased Th17 cells and reciprocally increased Treg cells, leading to a decreased Th17/Treg ratio. Meanwhile, Th17-related pathway was down-regulated after one day of high salt loading, with the increase in high salt loading as shown by microarray and RT-PCR. Subsequently, blood oxygen level-dependent magnetic resonance imaging (BOLD-MRI) observed hypoxia in the renal medulla (increased R2* signal) during high-salt loading, which was regressed to its baseline level in a step-down fashion during low-salt feeding. The flow-mediated vasodilatation (FMD) of the branchial artery was significantly higher on the first day of high salt loading. Collectively, these observations indicate that a short-term increase in dietary salt intake could induce reciprocal switches in Th17/Treg ratio and related cytokines, which might be the underlying cellular mechanism of high-salt dietary induced end organ inflammation and potential atherosclerotic risk.
Collapse
Affiliation(s)
- Tao Luo
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China.,Department of Cardiology, No. 254 Hospital of PLA, Tianjin, P.R. China
| | - Wen-Jie Ji
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Fei Yuan
- MRI Department, Pingjin Hospital, Tianjin, P.R. China
| | - Zhao-Zeng Guo
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yun-Xiao Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yan Dong
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yong-Qiang Ma
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| |
Collapse
|
27
|
A threshold level of NFATc1 activity facilitates thymocyte differentiation and opposes notch-driven leukaemia development. Nat Commun 2016; 7:11841. [PMID: 27312418 PMCID: PMC4915031 DOI: 10.1038/ncomms11841] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022] Open
Abstract
NFATc1 plays a critical role in double-negative thymocyte survival and differentiation. However, the signals that regulate Nfatc1 expression are incompletely characterized. Here we show a developmental stage-specific differential expression pattern of Nfatc1 driven by the distal (P1) or proximal (P2) promoters in thymocytes. Whereas, preTCR-negative thymocytes exhibit only P2 promoter-derived Nfatc1β expression, preTCR-positive thymocytes express both Nfatc1β and P1 promoter-derived Nfatc1α transcripts. Inducing NFATc1α activity from P1 promoter in preTCR-negative thymocytes, in addition to the NFATc1β from P2 promoter impairs thymocyte development resulting in severe T-cell lymphopenia. In addition, we show that NFATc1 activity suppresses the B-lineage potential of immature thymocytes, and consolidates their differentiation to T cells. Further, in the pTCR-positive DN3 cells, a threshold level of NFATc1 activity is vital in facilitating T-cell differentiation and to prevent Notch3-induced T-acute lymphoblastic leukaemia. Altogether, our results show NFATc1 activity is crucial in determining the T-cell fate of thymocytes. NFATc1 orchestrates thymocyte development. Here the authors show that NFATc1 expression is regulated by distinct promoters during thymocyte differentiation, and by conditional deletion of individual promoters in mice they define their specific roles in the control of T-cell development by NFATc1.
Collapse
|
28
|
Staal FJT, Chhatta A, Mikkers H. Caught in a Wnt storm: Complexities of Wnt signaling in hematopoiesis. Exp Hematol 2016; 44:451-7. [PMID: 27016274 DOI: 10.1016/j.exphem.2016.03.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/11/2016] [Accepted: 03/15/2016] [Indexed: 01/10/2023]
Abstract
The Wnt signaling pathway is an evolutionary conserved pathway that is involved in the development of almost every organ system in the body and provides self-renewal signals for most, if not all, adult stem cell systems. In recent years, this pathway has been studied by various research groups working on hematopoietic stem cells, resulting in contradicting conclusions. Here, we discuss and interpret the results of these studies and propose that Wnt dosage, the source of hematopoietic stem cells, and interactions with other pathways explain these disparate results.
Collapse
Affiliation(s)
- Frank J T Staal
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Amiet Chhatta
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Harald Mikkers
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Jha A, Singh AK, Weissgerber P, Freichel M, Flockerzi V, Flavell RA, Jha MK. Essential roles for Cavβ2 and Cav1 channels in thymocyte development and T cell homeostasis. Sci Signal 2015; 8:ra103. [DOI: 10.1126/scisignal.aac7538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
30
|
Fernandes MT, Ghezzo MN, Silveira AB, Kalathur RK, Póvoa V, Ribeiro AR, Brandalise SR, Dejardin E, Alves NL, Ghysdael J, Barata JT, Yunes JA, dos Santos NR. Lymphotoxin-β receptor in microenvironmental cells promotes the development of T-cell acute lymphoblastic leukaemia with cortical/mature immunophenotype. Br J Haematol 2015; 171:736-51. [PMID: 26456771 DOI: 10.1111/bjh.13760] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/29/2015] [Indexed: 01/21/2023]
Abstract
Lymphotoxin-mediated activation of the lymphotoxin-β receptor (LTβR; LTBR) has been implicated in cancer, but its role in T-cell acute lymphoblastic leukaemia (T-ALL) has remained elusive. Here we show that the genes encoding lymphotoxin (LT)-α and LTβ (LTA, LTB) are expressed in T-ALL patient samples, mostly of the TAL/LMO molecular subtype, and in the TEL-JAK2 transgenic mouse model of cortical/mature T-ALL (Lta, Ltb). In these mice, expression of Lta and Ltb is elevated in early stage T-ALL. Surface LTα1 β2 protein is expressed in primary mouse T-ALL cells, but only in the absence of microenvironmental LTβR interaction. Indeed, surface LT expression is suppressed in leukaemic cells contacting Ltbr-expressing but not Ltbr-deficient stromal cells, both in vitro and in vivo, thus indicating that dynamic surface LT expression in leukaemic cells depends on interaction with its receptor. Supporting the notion that LT signalling plays a role in T-ALL, inactivation of Ltbr results in a significant delay in TEL-JAK2-induced leukaemia onset. Moreover, young asymptomatic TEL-JAK2;Ltbr(-/-) mice present markedly less leukaemic thymocytes than age-matched TEL-JAK2;Ltbr(+/+) mice and interference with LTβR function at this early stage delayed T-ALL development. We conclude that LT expression by T-ALL cells activates LTβR signalling in thymic stromal cells, thus promoting leukaemogenesis.
Collapse
Affiliation(s)
- Mónica T Fernandes
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | - Marinella N Ghezzo
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | | | - Ravi K Kalathur
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Vanda Póvoa
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Ana R Ribeiro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Institute for Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | | | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, University of Liège, Liège, Belgium
| | - Nuno L Alves
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Jacques Ghysdael
- Institut Curie-Centre de Recherche, Centre Universitaire, Orsay, France.,CNRS UMR3306, Centre Universitaire, Orsay, France.,INSERM U1005, Centre Universitaire, Orsay, France
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - José Andres Yunes
- Centro Infantil Boldrini, Campinas, SP, Brazil.,Department of Paediatrics, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Nuno R dos Santos
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| |
Collapse
|
31
|
Notch3/Jagged1 circuitry reinforces notch signaling and sustains T-ALL. Neoplasia 2015; 16:1007-17. [PMID: 25499214 PMCID: PMC4309263 DOI: 10.1016/j.neo.2014.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/01/2014] [Accepted: 10/06/2014] [Indexed: 11/23/2022] Open
Abstract
Deregulated Notch signaling has been extensively linked to T-cell acute lymphoblastic leukemia (T-ALL). Here, we show a direct relationship between Notch3 receptor and Jagged1 ligand in human cell lines and in a mouse model of T-ALL. We provide evidence that Notch-specific ligand Jagged1 is a new Notch3 signaling target gene. This essential event justifies an aberrant Notch3/Jagged1 cis-expression inside the same cell. Moreover, we demonstrate in Notch3-IC–overexpressing T lymphoma cells that Jagged1 undergoes a raft-associated constitutive processing. The proteolytic cleavage allows the Jagged1 intracellular domain to empower Notch signaling activity and to increase the transcriptional activation of Jagged1 itself (autocrine effect). On the other hand, the release of the soluble Jagged1 extracellular domain has a positive impact on activating Notch signaling in adjacent cells (paracrine effect), finally giving rise to a Notch3/Jagged1 auto-sustaining loop that supports the survival, proliferation, and invasion of lymphoma cells and contributes to the development and progression of Notch-dependent T-ALL. These observations are also supported by a study conducted on a cohort of patients in which Jagged1 expression is associated to adverse prognosis.
Collapse
|
32
|
López-Rodríguez C, Aramburu J, Berga-Bolaños R. Transcription factors and target genes of pre-TCR signaling. Cell Mol Life Sci 2015; 72:2305-21. [PMID: 25702312 PMCID: PMC11113633 DOI: 10.1007/s00018-015-1864-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/22/2015] [Accepted: 02/16/2015] [Indexed: 11/27/2022]
Abstract
Almost 30 years ago pioneering work by the laboratories of Harald von Boehmer and Susumo Tonegawa provided the first indications that developing thymocytes could assemble a functional TCRβ chain-containing receptor complex, the pre-TCR, before TCRα expression. The discovery and study of the pre-TCR complex revealed paradigms of signaling pathways in control of cell survival and proliferation, and culminated in the recognition of the multifunctional nature of this receptor. As a receptor integrated in a dynamic developmental process, the pre-TCR must be viewed not only in the light of the biological outcomes it promotes, but also in context with those molecular processes that drive its expression in thymocytes. This review article focuses on transcription factors and target genes activated by the pre-TCR to drive its different outcomes.
Collapse
Affiliation(s)
- Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences and Barcelona Biomedical Research Park, Universitat Pompeu Fabra, C/Doctor Aiguader Nº88, 08003, Barcelona, Barcelona, Spain,
| | | | | |
Collapse
|
33
|
Induced transcription and stability of CELF2 mRNA drives widespread alternative splicing during T-cell signaling. Proc Natl Acad Sci U S A 2015; 112:E2139-48. [PMID: 25870297 DOI: 10.1073/pnas.1423695112] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies in several cell types have highlighted dramatic and diverse changes in mRNA processing that occur upon cellular stimulation. However, the mechanisms and pathways that lead to regulated changes in mRNA processing remain poorly understood. Here we demonstrate that expression of the splicing factor CELF2 (CUGBP, Elav-like family member 2) is regulated in response to T-cell signaling through combined increases in transcription and mRNA stability. Transcriptional induction occurs within 6 h of stimulation and is dependent on activation of NF-κB. Subsequently, there is an increase in the stability of the CELF2 mRNA that correlates with a change in CELF2 3'UTR length and contributes to the total signal-induced enhancement of CELF2 expression. Importantly, we uncover dozens of splicing events in cultured T cells whose changes upon stimulation are dependent on CELF2 expression, and provide evidence that CELF2 controls a similar proportion of splicing events during human thymic T-cell development. Taken together, these findings expand the physiologic impact of CELF2 beyond that previously documented in developing neuronal and muscle cells to T-cell development and function, identify unappreciated instances of alternative splicing in the human thymus, and uncover novel mechanisms for CELF2 regulation that may broadly impact CELF2 expression across diverse cell types.
Collapse
|
34
|
Abstract
Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, California 95616
| | - Edward Y. Skolnik
- Division of Nephrology, New York University School of Medicine, New York, NY 10016
- Department of Molecular Pathogenesis, New York University School of Medicine, New York, NY 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
35
|
Martin-Blanco NM, Checquolo S, Del Gaudio F, Palermo R, Franciosa G, Di Marcotullio L, Gulino A, Canelles M, Screpanti I. Numb-dependent integration of pre-TCR and p53 function in T-cell precursor development. Cell Death Dis 2014; 5:e1472. [PMID: 25321479 PMCID: PMC4237259 DOI: 10.1038/cddis.2014.438] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/30/2014] [Accepted: 09/02/2014] [Indexed: 11/09/2022]
Abstract
Numb asymmetrically segregates at mitosis to control cell fate choices during development. Numb inheritance specifies progenitor over differentiated cell fates, and, paradoxically, also promotes neuronal differentiation, thus indicating that the role of Numb may change during development. Here we report that Numb nuclear localization is restricted to early thymocyte precursors, whereas timed appearance of pre-T-cell receptor (pre-TCR) and activation of protein kinase Cθ promote phosphorylation-dependent Numb nuclear exclusion. Notably, nuclear localization of Numb in early thymocyte precursors favors p53 nuclear stabilization, whereas pre-TCR-dependent Numb nuclear exclusion promotes the p53 downmodulation essential for further differentiation. Accordingly, the persistence of Numb in the nucleus impairs the differentiation and promotes precursor cell death. This study reveals a novel regulatory mechanism for Numb function based on its nucleus–cytosol shuttling, coupling the different roles of Numb with different stages of T-cell development.
Collapse
Affiliation(s)
- N M Martin-Blanco
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - S Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University, Latina 04100, Italy
| | - F Del Gaudio
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - R Palermo
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - G Franciosa
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - L Di Marcotullio
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - A Gulino
- 1] Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy [2] Neuromed Institute, Pozzilli 86007, Italy
| | - M Canelles
- Institute of Parasitology and Biomedicine 'Lopez Neyra', Spanish National Research Council, Granada 18100, Spain
| | - I Screpanti
- 1] Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy [2] Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy [3] Institut Pasteur-Foundation Cenci Bolognetti, Sapienza University, Rome 00161, Italy
| |
Collapse
|
36
|
Loss of IP3R-dependent Ca2+ signalling in thymocytes leads to aberrant development and acute lymphoblastic leukemia. Nat Commun 2014; 5:4814. [PMID: 25215520 DOI: 10.1038/ncomms5814] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/25/2014] [Indexed: 01/28/2023] Open
Abstract
Calcium ions (Ca(2+)) function as universal second messengers in eukaryotic cells, including immune cells. Ca(2+) is crucial for peripheral T-lymphocyte activation and effector functions, and influences thymocyte selection and motility in the developing thymus. However, the role of Ca(2+) signalling in early T-lymphocyte development is not well understood. Here we show that the inositol triphosphate receptors (IP3Rs) Ca(2+) ion channels are required for proliferation, survival and developmental progression of T-lymphocyte precursors. Our studies indicate that signalling via IP3Rs represses Sox13, an antagonist of the developmentally important transcription factor Tcf-1. In the absence of IP3R-mediated Ca(2+) signalling, repression of key Notch transcriptional targets--including Hes1--fail to occur in post β-selection thymocytes, and mice develop aggressive T-cell malignancies that resemble human T-cell acute lymphoblastic leukemia (T-ALL). These data indicate that IP3R-mediated Ca(2+) signalling reinforces Tcf-1 activity to both ensure normal development and prevent thymocyte neoplasia.
Collapse
|
37
|
Buckley MW, Arandjelovic S, Trampont PC, Kim TS, Braciale TJ, Ravichandran KS. Unexpected phenotype of mice lacking Shcbp1, a protein induced during T cell proliferation. PLoS One 2014; 9:e105576. [PMID: 25153088 PMCID: PMC4143286 DOI: 10.1371/journal.pone.0105576] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/21/2014] [Indexed: 11/25/2022] Open
Abstract
T cell development and activation are highly regulated processes, and their proper execution is important for a competent immune system. Shc SH2-domain binding protein-1 (Shcbp1) is an evolutionarily conserved protein that binds to the adaptor protein ShcA. Studies in Drosophila and in cell lines have strongly linked Shcbp1 to cell proliferation, embryonic development, growth factor signaling, and tumorigenesis. Here we show that Shcbp1 expression is strikingly upregulated during the β-selection checkpoint in thymocytes, and that its expression tightly correlates with proliferative stages of T cell development. To evaluate the role for Shcbp1 during thymic selection and T cell function in vivo, we generated mice with global and conditional deletion of Shcbp1. Surprisingly, the loss of Shcbp1 expression did not have an obvious effect during T cell development. However, in a mouse model of experimental autoimmune encephalomyelitis (EAE), which depends on CD4+ T cell function and mimics multiple features of the human disease multiple sclerosis, Shcbp1 deficient mice had reduced disease severity and improved survival, and this effect was T cell intrinsic. These data suggest that despite the striking upregulation of Shcbp1 during T cell proliferation, loss of Shcbp1 does not directly affect T cell development, but regulates CD4+ T cell effector function in vivo.
Collapse
Affiliation(s)
- Monica W. Buckley
- Department of Microbiology, Immunology, Cancer biology, University of Virginia, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sanja Arandjelovic
- Department of Microbiology, Immunology, Cancer biology, University of Virginia, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Paul C. Trampont
- Department of Microbiology, Immunology, Cancer biology, University of Virginia, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Taeg S. Kim
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas J. Braciale
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kodi S. Ravichandran
- Department of Microbiology, Immunology, Cancer biology, University of Virginia, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
38
|
van der Weerd K, van Hagen PM, Schrijver B, Heuvelmans SJWM, Hofland LJ, Swagemakers SMA, Bogers AJJC, Dik WA, Visser TJ, van Dongen JJM, van der Lelij AJ, Staal FJT. Thyrotropin acts as a T-cell developmental factor in mice and humans. Thyroid 2014; 24:1051-61. [PMID: 24635198 DOI: 10.1089/thy.2013.0396] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Using gene expression profiling, we detected differential thyrotropin receptor (TSH-R) expression during human T-cell development in the thymus. This expression pattern indicated a potential role for the TSH-R within the thymus, independent of its function in the thyroid gland. Here, we demonstrate that TSH-R expression is thymus-specific within the immune system. TSH was able to bind and activate the TSH-R present on thymocytes, thereby activating calcium signaling and cyclic adenosine monophosphate signaling pathways. Mice lacking functional TSH-R expression (hyt/hyt mice) were shown to have lower frequencies of DP and SP thymocytes compared to their heterozygous littermates. Moreover, addition of TSH to co-cultures of human thymocytes enhanced T-cell development. Thus, TSH acts as a previously unrecognized growth factor for developing T cells, with potential clinical use to enhance thymic output and thereby the functional T-cell repertoire in the periphery. The direct effects of TSH on thymocytes may also explain the thus far enigmatic thymic hyperplasia in Graves' disease.
Collapse
Affiliation(s)
- Kim van der Weerd
- 1 Department of Immunology, Erasmus University Medical Center , Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Immunosuppressive activity of daphnetin, one of coumarin derivatives, is mediated through suppression of NF-κB and NFAT signaling pathways in mouse T cells. PLoS One 2014; 9:e96502. [PMID: 24800925 PMCID: PMC4011761 DOI: 10.1371/journal.pone.0096502] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 04/09/2014] [Indexed: 11/19/2022] Open
Abstract
Daphnetin, a plant-derived dihydroxylated derivative of coumarin, is an effective compound extracted from a plant called Daphne Korean Nakai. Coumarin derivates were known for their antithrombotic, anti-inflammatory, and antioxidant activities. The present study was aimed to determine the immunosuppressive effects and the underlying mechanisms of daphnetin on concanavalin A (ConA) induced T lymphocytes in mice. We showed that, in vitro, daphnetin suppressed ConA-induced splenocyte proliferation, influenced production of the cytokines and inhibited cell cycle progression through the G0/G1 transition. The data also revealed that daphnetin could down-regulate activation of ConA induced NF-κB and NFAT signal transduction pathways in mouse T lymphocyte. In vivo, daphnetin treatment significantly inhibited the 2, 4- dinitrofluorobenzene (DNFB) -induced delayed type hypersensitivity (DTH) reactions in mice. Collectively, daphnetin had strong immunosuppressive activity both in vitro and in vivo, suggesting a potential role for daphnetin as an immunosuppressive agent, and established the groundwork for further research on daphnetin.
Collapse
|
40
|
Pontin is required for pre-TCR signaling at the β-selection checkpoint in T cell development. Biochem Biophys Res Commun 2014; 447:44-50. [PMID: 24680824 DOI: 10.1016/j.bbrc.2014.03.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/19/2014] [Indexed: 11/24/2022]
Abstract
Pontin is a chromatin remodeling factor that possesses both ATPase and DNA helicase activities. Based on high expression in lymphoid tissues, we examined whether Pontin has a T cell-specific function. We generated Pontin(f/f);Lck-Cre mice, in which Pontin can be conditionally deleted in T cells and then explored T cell-specific function of Pontin in vivo. Here, we show that specific abrogation of Pontin expression in T cells almost completely blocked development of αβ T cells at the β-selection checkpoint by inducing cell apoptosis indicating that Pontin is essential for early T cell development. Pontin-deficient thymocytes show a comparable expression level of T cell receptor (TCR)β chain, but have enhanced activation of p53 and Notch signaling compared to wild-type thymocytes. Intriguingly, the developmental block of αβ T cells can be partially rescued by loss of p53. Together, our data demonstrate a novel role of Pontin as a crucial regulator in pre-TCR signaling during T cell development.
Collapse
|
41
|
Gerondakis S, Fulford TS, Messina NL, Grumont RJ. NF-κB control of T cell development. Nat Immunol 2014; 15:15-25. [PMID: 24352326 DOI: 10.1038/ni.2785] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 11/12/2013] [Indexed: 12/12/2022]
Abstract
The NF-κB signal transduction pathway is best known as a major regulator of innate and adaptive immune responses, yet there is a growing appreciation of its importance in immune cell development, particularly of T lineage cells. In this Review, we discuss how the temporal regulation of NF-κB controls the stepwise differentiation and antigen-dependent selection of conventional and specialized subsets of T cells in response to T cell receptor and costimulatory, cytokine and growth factor signals.
Collapse
Affiliation(s)
- Steve Gerondakis
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Thomas S Fulford
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Nicole L Messina
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Raelene J Grumont
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
NFAT5 induction by the pre-T-cell receptor serves as a selective survival signal in T-lymphocyte development. Proc Natl Acad Sci U S A 2013; 110:16091-6. [PMID: 24043824 DOI: 10.1073/pnas.1215934110] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The Rel-like transcription factors nuclear factor kappa B (NF-κB) and the calcineurin-dependent nuclear factor of activated T cells (NFATc) control specific points of thymocyte maturation. Thymocytes also express a distinct member of the Rel family, the calcineurin-independent, osmostress response regulator NFAT5. Here we show that IKKβ regulates the expression of NFAT5 in thymocytes, which in turn contributes to the survival of T-cell receptor αβ thymocytes and the transition from the β-selection checkpoint to the double-positive stage in an osmostress-independent manner. NFAT5-deficient thymocytes had normal expression and proximal signaling of the pre-T-cell receptor but exhibited a partial defect in β-chain allelic exclusion and increased apoptosis. Further analysis showed that NFAT5 regulated the expression of the prosurvival factors A1 and Bcl2 and attenuated the proapoptotic p53/Noxa axis. These findings position NFAT5 as a target of the IKKβ/NF-κB pathway in thymocytes and as a downstream effector of the prosurvival role of the pre-T-cell receptor.
Collapse
|
43
|
Trčka F, Šmarda J, Knopfová L, Kuziaková K, Beneš P. Nuclear factor of activated T-cells 1 increases sensitivity of v-myb transformed monoblasts to all-trans retinoic acid. Cell Signal 2013; 25:1546-55. [PMID: 23571271 DOI: 10.1016/j.cellsig.2013.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/12/2013] [Accepted: 03/26/2013] [Indexed: 11/28/2022]
Abstract
Nuclear factors of activated T-cells (NFATs) are important regulators of the cytokine gene expression in activated T-cells. In the last decade, NFATs have been shown to regulate cell cycle, differentiation and apoptosis in cells of various origins revealing their importance for cell homeostasis. In this study, we investigated the effects of NFAT1 on proliferation and differentiation of v-myb-transformed BM2 monoblasts. In contrast to many other leukemic cell lines, BM2 cells do not respond to retinoic acid. However, once overexpressing NFAT1, they became sensitive to all-trans retinoic acid (ATRA). The ATRA-treated BM2NFAT1 cells differentiated along monocyte/macrophage pathway as evidenced by changes in cell morphology, adherence, phagocytic and non-specific esterase activities, reactive oxygen species production, and vimentin expression. Furthermore, overexpressed NFAT1 either alone or in combination with the ATRA-driven signalling pathway deregulated cyclin A and retinoic acid receptor proteins in BM2 cells. Data presented in this study indicate that the NFAT1 and ATRA signalling pathways synergize in control of proliferation and differentiation of BM2 monoblasts.
Collapse
Affiliation(s)
- Filip Trčka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| | | | | | | | | |
Collapse
|
44
|
An alternative NFAT-activation pathway mediated by IL-7 is critical for early thymocyte development. Nat Immunol 2012; 14:127-35. [DOI: 10.1038/ni.2507] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/26/2012] [Indexed: 01/20/2023]
|
45
|
|
46
|
Patel ES, Chang LJ. Synergistic effects of interleukin-7 and pre-T cell receptor signaling in human T cell development. J Biol Chem 2012; 287:33826-35. [PMID: 22859301 DOI: 10.1074/jbc.m112.380113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The role of IL-7 in pre-T cell receptor (TCR) signaling during human T cell development is poorly understood. To study this, we engineered Molt3, a T cell progenitor T-acute lymphoblastic leukemia cell line, using lentiviral IL-7 receptor α (IL-7Rα) to serve as a model system. IL-7 promoted pre-TCR activation in IL-7Rα(hi) Molt3 as illustrated by CD25 up-regulation after anti-CD3 stimulation. Anti-CD3 treatment activated Akt and Erk1/2 signaling pathways as proven using specific inhibitors, and IL-7 further enhanced both signaling pathways. The close association of IL-7Rα with CD3ζ in the pre-TCR complex was illustrated through live imaging confocal fluorescence microscopy. These results demonstrate a direct and cooperative role of IL-7 in pre-TCR signaling.
Collapse
Affiliation(s)
- Ekta S Patel
- Department of Molecular Genetics, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | | |
Collapse
|
47
|
Abstract
The stromal interaction molecules STIM1 and STIM2 are Ca2+ sensors, mostly located in the endoplasmic reticulum, that detect changes in the intraluminal Ca2+ concentration and communicate this information to plasma membrane store-operated channels, including members of the Orai family, thus mediating store-operated Ca2+ entry (SOCE). Orai and STIM proteins are almost ubiquitously expressed in human cells, where SOCE has been reported to play a relevant functional role. The phenotype of patients bearing mutations in STIM and Orai proteins, together with models of STIM or Orai deficiency in mice, as well as other organisms such as Drosophila melanogaster, have provided compelling evidence on the relevant role of these proteins in cellular physiology and pathology. Orai1-deficient patients suffer from severe immunodeficiency, congenital myopathy, chronic pulmonary disease, anhydrotic ectodermal dysplasia and defective dental enamel calcification. STIM1-deficient patients showed similar abnormalities, as well as autoimmune disorders. This review summarizes the current evidence that identifies and explains diseases induced by disturbances in SOCE due to deficiencies or mutations in Orai and STIM proteins.
Collapse
Affiliation(s)
- A Berna-Erro
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | | | | |
Collapse
|
48
|
del Blanco B, García-Mariscal A, Wiest DL, Hernández-Munain C. Tcra enhancer activation by inducible transcription factors downstream of pre-TCR signaling. THE JOURNAL OF IMMUNOLOGY 2012; 188:3278-93. [PMID: 22357628 DOI: 10.4049/jimmunol.1100271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Tcra enhancer (Eα) is essential for pre-TCR-mediated activation of germline transcription and V(D)J recombination. Eα is considered an archetypical enhanceosome that acts through the functional synergy and cooperative binding of multiple transcription factors. Based on dimethylsulfate genomic footprinting experiments, there has been a long-standing paradox regarding Eα activation in the absence of differences in enhancer occupancy. Our data provide the molecular mechanism of Eα activation and an explanation of this paradox. We found that germline transcriptional activation of Tcra is dependent on constant phospholipase Cγ, as well as calcineurin- and MAPK/ERK-mediated signaling, indicating that inducible transcription factors are crucially involved. NFAT, AP-1, and early growth response factor 1, together with CREB-binding protein/p300 coactivators, bind to Eα as part of an active enhanceosome assembled during pre-TCR signaling. We favor a scenario in which the binding of lymphoid-restricted and constitutive transcription factors to Eα prior to its activation forms a regulatory scaffold to recruit factors induced by pre-TCR signaling. Thus, the combinatorial assembly of tissue- and signal-specific transcription factors dictates the Eα function. This mechanism for enhancer activation may represent a general paradigm in tissue-restricted and stimulus-responsive gene regulation.
Collapse
Affiliation(s)
- Beatriz del Blanco
- Departamento de Biología Celular e Inmunología, Instituto de Parasitología y Biomedicina López-Neyra (IPBLN-CSIC), Consejo Superior de Investigaciones Científicas, 18100-Armilla, Granada, Spain
| | | | | | | |
Collapse
|
49
|
Abstract
Mutations in genes encoding the calcium-release activated calcium (CRAC) channel abolish calcium influx in cells of the immune system and cause severe congenital immunodeficiency. Patients with autosomal recessive mutations in the CRAC channel gene ORAI1, its activator stromal interaction molecule 1 (STIM1), and mice with targeted deletion of Orai1, Stim1, and Stim2 genes reveal important roles for CRAC channels in adaptive and innate immune responses to infection and in autoimmunity. Because CRAC channels have important functions outside the immune system, deficiency of either ORAI1 or STIM1 is associated with a unique clinical phenotype. This review will give an overview of CRAC channel function in the immune system, examine the consequences of CRAC channel deficiency for immunity in human patients and mice, and discuss genetic defects in immunoreceptor-associated signaling molecules that compromise calcium influx and cause immunodeficiency.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University Langone Medical Center, New York, USA.
| |
Collapse
|
50
|
Shaw PJ, Feske S. Physiological and pathophysiological functions of SOCE in the immune system. Front Biosci (Elite Ed) 2012. [PMID: 22202035 DOI: 10.2741/540] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcium signals play a critical role in many cell-type specific effector functions during innate and adaptive immune responses. The predominant mechanism to raise intracellular (Ca²⁺) used by most immune cells is store-operated Ca²⁺ entry (SOCE), whereby the depletion of endoplasmic reticulum (ER) Ca²⁺ stores triggers the influx of extracellular Ca²⁺. SOCE in immune cells is mediated by the highly Ca²⁺ selective Ca²⁺-release-activated Ca²⁺ (CRAC) channel, encoded by ORAI1, ORAI2 and ORAI3 genes. ORAI proteins are activated by stromal interaction molecules (STIM) 1 and 2, which act as sensors of ER Ca²⁺ store depletion. The importance of SOCE mediated by STIM and ORAI proteins for immune function is evident from the immunodeficiency and autoimmunity in patients with mutations in STIM1 and ORAI1 genes. These patients and studies in gene-targeted mice have revealed an essential role for ORAI/STIM proteins in the function of several immune cells. This review focuses on recent advances made towards understanding the role of SOCE in immune cells with an emphasis on the immune dysregulation that results from defects in SOCE in human patients and transgenic mice.
Collapse
Affiliation(s)
- Patrick J Shaw
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA
| | | |
Collapse
|