1
|
He J, Chen D, Xiong W, Hou X, Quan Y, Yang M, Dong Z. Eomesodermin spatiotemporally orchestrates the early and late stages of NK cell development by targeting KLF2 and T-bet, respectively. Cell Mol Immunol 2024; 21:662-673. [PMID: 38740922 PMCID: PMC11214621 DOI: 10.1038/s41423-024-01164-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 04/07/2024] [Indexed: 05/16/2024] Open
Abstract
Eomesodermin (Eomes) is a critical factor in the development of natural killer (NK) cells, but its precise role in temporal and spatial coordination during this process remains unclear. Our study revealed that Eomes plays distinct roles during the early and late stages of NK cell development. Specifically, the early deletion of Eomes via the CD122-Cre transgene resulted in significant blockade at the progenitor stage due to the downregulation of KLF2, another important transcription factor. ChIP-seq revealed direct binding of Eomes to the conserved noncoding sequence (CNS) of Klf2. Utilizing the CHimeric IMmune Editing (CHIME) technique, we found that deletion of the CNS region of Klf2 via CRISPRi led to a reduction in the NK cell population and developmental arrest. Moreover, constitutive activation of this specific CNS region through CRISPRa significantly reversed the severe defects in NK cell development caused by Eomes deficiency. Conversely, Ncr1-Cre-mediated terminal deletion of Eomes expedited the transition of NK cell subsets from the CD27+CD11b+ phenotype to the CD27-CD11b+ phenotype. Late-stage deficiency of Eomes led to a significant increase in T-bet expression, which subsequently increased the expression of the transcription factor Zeb2. Genetic deletion of one allele of Tbx21, encoding T-bet, effectively reversed the aberrant differentiation of Eomes-deficient NK cells. In summary, we utilized two innovative genetic models to elucidate the intricate mechanisms underlying Eomes-mediated NK cell commitment and differentiation.
Collapse
Affiliation(s)
- Junming He
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Anhui, 230032, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Donglin Chen
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Wei Xiong
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Xinlei Hou
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Yuhe Quan
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China.
- The Biomedical Translational Research Institute. Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. School of Medicine. Jinan University, Guangzhou, 510632, China.
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Anhui, 230032, China.
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China.
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei, 230032, China.
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
2
|
Saadh MJ, Alfattah MA, Ismail AH, Saeed BA, Abbas HH, Elashmawy NF, Hashim GA, Ismail KS, Abo-Zaid MA, Waggiallah HA. The role of Interleukin-21 (IL-21) in allergic disorders: Biological insights and regulatory mechanisms. Int Immunopharmacol 2024; 134:111825. [PMID: 38723368 DOI: 10.1016/j.intimp.2024.111825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 06/03/2024]
Abstract
In recent decades, allergic diseases subsequent from an IgE-mediated response to specific allergens have become a progressively public chronic disease worldwide. They have shaped an important medical and socio-economic burden. A significant proportion of allergic disorders are branded via a form 2 immune response relating Th2 cells, type 2 natural lymphoid cells, mast cells and eosinophils. Interleukin-21 (IL-21) is a participant of the type-I cytokine family manufactured through numerous subsets of stimulated CD4+ T cells and uses controlling properties on a diversity of immune cells. Increasingly, experimental sign suggests a character for IL-21 in the pathogenesis of numerous allergic disorders. The purpose of this review is to discuss the biological properties of IL-21 and to summaries current developments in its role in the regulation of allergic disorders.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Mohammed A Alfattah
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Bashar Abdullah Saeed
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | | | - Nabila F Elashmawy
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ghassan A Hashim
- Department of Nursing, Al-Zahrawi University College, Karbala, Iraq
| | - Khatib Sayeed Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
3
|
Chen S, Huan X, Xu C, Luo S, Zhao C, Zhong H, Zheng X, Qiao K, Dong Y, Wang Y, Liu C, Huang H, Chen Y, Zou Z. Eomesodermin expression in CD4 +T-cells associated with disease progression in amyotrophic lateral sclerosis. CNS Neurosci Ther 2024; 30:e14503. [PMID: 37850654 PMCID: PMC11017423 DOI: 10.1111/cns.14503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
AIM To clarify the role of Eomesodermin (EOMES) to serve as a disease-relevant biomarker and the intracellular molecules underlying the immunophenotype shifting of CD4+T subsets in amyotrophic lateral sclerosis (ALS). METHODS The derivation and validation cohorts included a total of 148 ALS patients and 101 healthy controls (HCs). Clinical data and peripheral blood were collected. T-cell subsets and the EOMES expression were quantified using multicolor flow cytometry. Serum neurofilament light chain (NFL) was measured. In 1-year longitudinal follow-ups, the ALSFRS-R scores and primary endpoint events were further recorded in the ALS patients of the validation cohort. RESULTS In the derivation cohort, the CD4+EOMES+T-cell subsets were significantly increased (p < 0.001). EOMES+ subset was positively correlated with increased serum NFL levels in patients with onset longer than 12 months. In the validation cohort, the elevated CD4+EOMES+T-cell proportions and their association with NFL levels were also identified. The longitudinal study revealed that ALS patients with higher EOMES expression were associated with higher progression rates (p = .010) and worse prognosis (p = .003). CONCLUSIONS We demonstrated that increased CD4+EOMES+T-cell subsets in ALS were associated with disease progression and poor prognosis. Identifying these associations may contribute to a better understanding of the immunopathological mechanism of ALS.
Collapse
Affiliation(s)
- Sheng Chen
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhouChina
| | - Xiao Huan
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Chun‐Zuan Xu
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhouChina
| | - Su‐Shan Luo
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Chong‐Bo Zhao
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Hua‐Hua Zhong
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Xue‐Ying Zheng
- Department of Biostatistics, School of Public Health and Key Laboratory of Public Health SafetyFudan UniversityShanghaiChina
| | - Kai Qiao
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Yi Dong
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Ying Wang
- Department of PharmacyFudan University Huashan HospitalShanghaiChina
| | - Chang‐Yun Liu
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhouChina
| | - Hua‐Pin Huang
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhouChina
| | - Yan Chen
- Huashan Rare Disease Center and Department of NeurologyHuashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, Fudan UniversityShanghaiChina
| | - Zhang‐Yu Zou
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhouChina
| |
Collapse
|
4
|
Joulia E, Michieletto MF, Agesta A, Peillex C, Girault V, Le Dorze AL, Peroceschi R, Bucciarelli F, Szelechowski M, Chaubet A, Hakim N, Marrocco R, Lhuillier E, Lebeurrier M, Argüello RJ, Saoudi A, El Costa H, Adoue V, Walzer T, Sarry JE, Dejean AS. Eomes-dependent mitochondrial regulation promotes survival of pathogenic CD4+ T cells during inflammation. J Exp Med 2024; 221:e20230449. [PMID: 38189779 PMCID: PMC10772920 DOI: 10.1084/jem.20230449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/02/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
The mechanisms whereby Eomes controls tissue accumulation of T cells and strengthens inflammation remain ill-defined. Here, we show that Eomes deletion in antigen-specific CD4+ T cells is sufficient to protect against central nervous system (CNS) inflammation. While Eomes is dispensable for the initial priming of CD4+ T cells, it is required for long-term maintenance of CNS-infiltrating CD4+ T cells. We reveal that the impact of Eomes on effector CD4+ T cell longevity is associated with sustained expression of multiple genes involved in mitochondrial organization and functions. Accordingly, epigenetic studies demonstrate that Eomes supports mitochondrial function by direct binding to either metabolism-associated genes or mitochondrial transcriptional modulators. Besides, the significance of these findings was confirmed in CD4+ T cells from healthy donors and multiple sclerosis patients. Together, our data reveal a new mechanism by which Eomes promotes severity and chronicity of inflammation via the enhancement of CD4+ T cell mitochondrial functions and resistance to stress-induced cell death.
Collapse
Affiliation(s)
- Emeline Joulia
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Michaël F. Michieletto
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arantxa Agesta
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Cindy Peillex
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Virginie Girault
- Suivi Immunologique des Thérapeutiques Innovantes, Pôle de Biologie, Pontchaillou University Hospital, Rennes, France
- UMR1236, University of Rennes, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Bretagne, Rennes, France
| | - Anne-Louise Le Dorze
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Romain Peroceschi
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Florence Bucciarelli
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Marion Szelechowski
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Adeline Chaubet
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Nawad Hakim
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Rémi Marrocco
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Emeline Lhuillier
- GeT-Santé, Plateforme Génome et Transcriptome, GenoToul, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Manuel Lebeurrier
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Rafael J. Argüello
- Aix Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Abdelhadi Saoudi
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Hicham El Costa
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Veronique Adoue
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, Lyon, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Institut National de la Santé et de la Recherche Médicale, Toulouse, France
| | - Anne S. Dejean
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Institut National de la Santé et de la Recherche Médicale UMR1291, Centre National de la Recherche Scientifique UMR5051, Université Toulouse III, Toulouse, France
| |
Collapse
|
5
|
Narsale A, Almanza F, Tran T, Lam B, Seo D, Vu A, Long SA, Cooney L, Serti E, Davies JD. Th2 cell clonal expansion at diagnosis in human type 1 diabetes. Clin Immunol 2023; 257:109829. [PMID: 37907122 DOI: 10.1016/j.clim.2023.109829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
Soon after diagnosis with type 1 diabetes (T1D), many patients experience a period of partial remission. A longer partial remission is associated with a better response to treatment, but the mechanism is not known. The frequency of CD4+CD25+CD127hi (127-hi) cells, a cell subset with an anti-inflammatory Th2 bias, correlates positively with length of partial remission. The purpose of this study was to further characterize the nature of the Th2 bias in 127-hi cells. Single cell RNA sequencing paired with TCR sequencing of sorted 127-hi memory cells identifies clonally expanded Th2 clusters in 127-hi cells from T1D, but not from healthy donors. The Th2 clusters express GATA3, GATA3-AS1, PTGDR2, IL17RB, IL4R and IL9R. The existence of 127-hi Th2 cell clonal expansion in T1D suggests that disease factors may induce clonal expansion of 127-hi Th2 cells that prolong partial remission and delay disease progression.
Collapse
Affiliation(s)
- Aditi Narsale
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Francisco Almanza
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Theo Tran
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Breanna Lam
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - David Seo
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Alisa Vu
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - S Alice Long
- Benaroya Research Institute, 1201 9(th) Ave, Seattle, WA 98101, USA.
| | | | | | - Joanna D Davies
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| |
Collapse
|
6
|
Arra A, Lingel H, Pierau M, Brunner-Weinzierl MC. PD-1 limits differentiation and plasticity of Tc17 cells. Front Immunol 2023; 14:1104730. [PMID: 37205114 PMCID: PMC10186197 DOI: 10.3389/fimmu.2023.1104730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/05/2023] [Indexed: 05/21/2023] Open
Abstract
Blockade of surface co-inhibitory receptor programmed cell death-1 (PD-1; CD279) has been established as an important immunotherapeutic approach to treat malignancies. On a cellular level, PD-1 is demonstrated to be of particular importance in inhibiting differentiation and effector function of cytotoxic Tc1 cells (CTLs). Nevertheless, the role of PD-1 in modulating interleukin (IL)-17-producing CD8+ T-cells (Tc17 cells), which generally display suppressed cytotoxic nature, is not well understood. To evaluate the impact of PD-1 in Tc17 responses, we examined its functioning using different in vitro and in vivo models. Upon activation of CD8+ T-cells in Tc17 environment, we found that PD-1 was rapidly expressed on the surface of CD8+ T-cells and triggered a T-cell-internal mechanism that inhibited the expression of IL-17 and Tc17-supporting transcription factors pSTAT3 and RORγt. Expression of type17-polarising cytokine IL-21 and the receptor for IL-23 were also suppressed. Intriguingly, adoptively transferred, PD-1-/- Tc17 cells were highly efficient in rejection of established B16 melanoma in vivo and displayed Tc1 like characteristics ex vivo. When using IL-17A-eGFP reporter mice for in vitro fate tracking, IL-17A-eGFP expressing cells lacking PD-1 signaling upon re-stimulation with IL-12 quickly acquired Tc1 characteristics such as IFN-γ, and granzyme B expression, implicating lineage independent upregulation of CTL-characteristics that are needed for tumor control. In line with plasticity characteristics, absence of PD-1 signaling in Tc17 cells increased the expression of the stemness and persistence-associated molecules TCF1 and BCL6. Thus, PD-1 plays a central role in the specific suppression of Tc17 differentiation and its plasticity in relation to CTL-driven tumor rejection, which provides further explanation as to why the blockade of PD-1 is such an efficient therapeutic target for inducing tumor rejection.
Collapse
Affiliation(s)
- Aditya Arra
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Mandy Pierau
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Monika C. Brunner-Weinzierl
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
- *Correspondence: Monika C. Brunner-Weinzierl,
| |
Collapse
|
7
|
Transcriptome Profiling in the Hippocampi of Mice with Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2022; 23:ijms232314829. [PMID: 36499161 PMCID: PMC9738199 DOI: 10.3390/ijms232314829] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), approximates the key histopathological, clinical, and immunological features of MS. Hippocampal dysfunction in MS and EAE causes varying degrees of cognitive and emotional impairments and synaptic abnormalities. However, the molecular alterations underlying hippocampal dysfunctions in MS and EAE are still under investigation. The purpose of this study was to identify differentially expressed genes (DEGs) in the hippocampus of mice with EAE in order to ascertain potential genes associated with hippocampal dysfunction. Gene expression in the hippocampus was analyzed by RNA-sequencing and validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Gene expression analysis revealed 1202 DEGs; 1023 were upregulated and 179 were downregulated in the hippocampus of mice with EAE (p-value < 0.05 and fold change >1.5). Gene ontology (GO) analysis showed that the upregulated genes in the hippocampi of mice with EAE were associated with immune system processes, defense responses, immune responses, and regulation of immune responses, whereas the downregulated genes were related to learning or memory, behavior, and nervous system processes in the GO biological process. The expressions of hub genes from the search tool for the retrieval of interacting genes/proteins (STRING) analysis were validated by RT-qPCR. Additionally, gene set enrichment analysis showed that the upregulated genes in the hippocampus were associated with inflammatory responses: interferon-γ responses, allograft rejection, interferon-α responses, IL6_JAK_STAT3 signaling, inflammatory responses, complement, IL2_STAT5 signaling, TNF-α signaling via NF-κB, and apoptosis, whereas the downregulated genes were related to synaptic plasticity, dendritic development, and development of dendritic spine. This study characterized the transcriptome pattern in the hippocampi of mice with EAE and signaling pathways underpinning hippocampal dysfunction. However, further investigation is needed to determine the applicability of these findings from this rodent model to patients with MS. Collectively, these results indicate directions for further research to understand the mechanisms behind hippocampal dysfunction in EAE.
Collapse
|
8
|
Wei H, Li Y, Guo Z, Ma X, Li Y, Wei X, Han D, Zhang T, Chen X, Yan C, Zhou J, Pang Q, Wang P, Zhang W. Comparison of dynamic changes in the peripheral CD8 + T cells function and differentiation in ESCC patients treated with radiotherapy combined with anti-PD-1 antibody or concurrent chemoradiotherapy. Front Immunol 2022; 13:1060695. [PMID: 36479110 PMCID: PMC9720318 DOI: 10.3389/fimmu.2022.1060695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Objective The systematic immune status of cancer patients undergoing immunotherapy is little known. We prospectively identified the function and differentiation traits of peripheral CD8+ T cells based on our phase 1b clinical trial (NCT03222440) of radiotherapy combined with camrelizumab in patients with locally advanced esophageal squamous cell carcinoma (ESCC) and compared it with concurrent chemoradiotherapy (CCRT). Methods 19 and 18 patients were included in the cohort of radiotherapy plus camrelizumab and cohort of CCRT treatment. By using flow cytometry, we evaluated the expression levels of PD-1, Eomes, T-bet and IFN-γ (function), CD38 and HLA-DR (activation), and differentiation subsets classified according to the expression levels of CD45RA and CD62L in peripheral CD8+ T cells before and during treatment. Results Effective binding of anti-PD-1 antibody camrelizumab with PD-1 on CD8+ T cells was detected during treatment. Both two treatments elevated the expression levels of activation molecules CD38 and HLA-DR on CD8+ T cells. PD-1+CD8+ T cells had more activation features than PD-1-CD8+ T cells in two groups and the treatments did not alter these differences. The two treatments activated both PD-1+ and PD-1- CD8+ T cells. PD-1+CD8+ T cells had less Naïve and TEMRA but more Tcm and Tem than PD-1-CD8+ T cells in two groups and both two treatments changed the ratio of memory T cells in PD-1+ and PD-1- cells. RT plus camrelizumab treatment reduced Naïve T cells and TEMRA subsets both in PD-1+ and PD-1- CD8+ T cells while elevated Tcm subset in PD-1+CD8+ T cells and Tem subset in PD-1-CD8+ T cells. CCRT elevated Tcm subset and reduced TEMRA subset in PD-1-CD8+ T cells while did not change any subset in PD-1+CD8+ T cells. Furthermore, patients undergoing radiotherapy plus immunotherapy were found to obtain better prognosis than those receiving CCRT. Conclusions This study identified the dynamic changes of systematic immune status of patients undergoing treatment. The two treatments had similar activation effects on peripheral CD8+ T cells with different PD-1 properties but had different effects on their differentiation status. These results provided potential clues to the reasons underlying the difference in prognosis of the two treatments.
Collapse
Affiliation(s)
- Hui Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yanqi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zhoubo Guo
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoxue Ma
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yang Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoying Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Dong Han
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jiahuan Zhou
- Department of Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| |
Collapse
|
9
|
Kuehn J, Schleifenbaum S, Hendling M, Siebenhandl S, Krainer J, Fuehner S, Hellige A, Park C, Hinze C, Wittkowski H, Holzinger D, Thurner L, Weinhäusel A, Foell D, Kessel C. Aberrant Naive CD4-Positive T Cell Differentiation in Systemic Juvenile Idiopathic Arthritis Committed to B Cell Help. Arthritis Rheumatol 2022; 75:826-841. [PMID: 36409585 DOI: 10.1002/art.42409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Systemic juvenile idiopathic arthritis (JIA) features characteristics of autoinflammation and autoimmunity, culminating in chronic arthritis. In this study, we hypothesized that aberrant or incomplete polarization of T helper cells contributes to disease pathology. METHODS Cells or serum samples were obtained from healthy controls (n = 72) and systemic JIA patients (n = 171). Isolated naive T helper cells were cultured under Th1, Th17, and T follicular helper (Tfh) or T peripheral helper (Tph)-polarizing conditions and were partly cocultured with allogenic memory B cells. Cell samples were then analyzed for surface marker, transcription factor, and cytokine expression, as well as plasmablast generation. Serum samples were subjected to multiplexed bead and self-antigen arrays and enzyme-linked immunosorbent assays, and all data were compared to retrospective RNA profiling analyses. RESULTS Differentiation of systemic JIA-naive T helper cells toward Th1 cells resulted in low expression levels of interferon-γ (IFNγ) and eomesodermin, which was associated in part with disease duration. In contrast developing Th1 cells in patients with systemic JIA were found to produce elevated levels of interleukin-21 (IL-21), which negatively correlated with cellular expression of IFNγ and eomesodermin. In both in vitro and ex vivo analyses, IL-21 together with programmed cell death 1 (PD-1), inducible T cell costimulator (ICOS), and CXCR5 expression induced naive T helper cells from systemic JIA patients to polarize toward a Tfh/Tph cell phenotype. Retrospective analysis of whole-blood RNA-sequencing data demonstrated that Bcl-6, a master transcription factor in Tfh/Tph cell differentiation, was overexpressed specifically in patients with systemic JIA. Naive T helper cells from systemic JIA patients which were stimulated in vitro promoted B cellular plasmablast generation, and self-antigen array data indicated that IgG reactivity profiles of patients with systemic JIA differed from those of healthy controls. CONCLUSION In the pathogenesis of systemic JIA, skewing of naive T helper cell differentiation toward a Tfh/Tph cell phenotype may represent an echo of autoimmunity, which may indicate the mechanisms driving progression toward chronic destructive arthritis.
Collapse
Affiliation(s)
- Julia Kuehn
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Susanne Schleifenbaum
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Michaela Hendling
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Sandra Siebenhandl
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Julie Krainer
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Sabrina Fuehner
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Antje Hellige
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Carolin Park
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Claas Hinze
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Helmut Wittkowski
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Essen, Germany, and Department of Applied Health Sciences, University of Applied Sciences Bochum, Bochum, Germany
| | - Lorenz Thurner
- José Carreras Center for Immunology and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg, Germany
| | - Andreas Weinhäusel
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Dirk Foell
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Christoph Kessel
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| |
Collapse
|
10
|
Regulation of CD4 T Cell Responses by the Transcription Factor Eomesodermin. Biomolecules 2022; 12:biom12111549. [PMID: 36358898 PMCID: PMC9687629 DOI: 10.3390/biom12111549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Central to the impacts of CD4 T cells, both positive in settings of infectious disease and cancer and negative in the settings of autoimmunity and allergy, is their ability to differentiate into distinct effector subsets with specialized functions. The programming required to support such responses is largely dictated by lineage-specifying transcription factors, often called ‘master regulators’. However, it is increasingly clear that many aspects of CD4 T cell immunobiology that can determine the outcomes of disease states involve a broader transcriptional network. Eomesodermin (Eomes) is emerging as an important member of this class of transcription factors. While best studied in CD8 T cells and NK cells, an increasing body of work has focused on impacts of Eomes expression in CD4 T cell responses in an array of different settings. Here, we focus on the varied impacts reported in these studies that, together, indicate the potential of targeting Eomes expression in CD4 T cells as a strategy to improve a variety of clinical outcomes.
Collapse
|
11
|
Kim S, Boehme L, Nel L, Casian A, Sangle S, Nova-Lamperti E, Seitan V, Spencer J, Lavender P, D'Cruz DP, John S. Defective STAT5 Activation and Aberrant Expression of BCL6 in Naive CD4 T Cells Enhances Follicular Th Cell-like Differentiation in Patients with Granulomatosis with Polyangiitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:807-818. [PMID: 35039330 DOI: 10.4049/jimmunol.2001331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 11/25/2021] [Indexed: 12/21/2022]
Abstract
Granulomatosis with polyangiitis (GPA) is a potentially fatal small vessel vasculitis of unknown etiology, characterized by anti-neutrophil cytoplasmic autoantibodies, chronic inflammation, and granulomatous tissue damage. T cell dysregulation, comprising decreased regulatory T cell function and increased circulating effector memory follicular Th cells (TFH), is strongly associated with disease pathogenesis, but the mechanisms driving these observations are unknown. We undertook transcriptomic and functional analysis of naive CD4 T cells from patients with GPA to identify underlying functional defects that could manifest in the pathogenic profiles observed in GPA. Gene expression studies revealed a dysregulation of the IL-2 receptor β/JAK-STAT signaling pathway and higher expression of BCL6 and BCL6-regulated genes in GPA naive CD4 T cells. IL-2-induced STAT5 activation in GPA naive CD4 T cells was decreased, whereas STAT3 activation by IL-6 and IL-2 was unperturbed. Consistently, BCL6 expression was sustained following T cell activation of GPA naive CD4 T cells and in vitro TFH differentiation of these cells resulted in significant increases in the production TFH-related cytokines IL-21 and IL-6. Thus, naive CD4 T cells are dysregulated in patients with GPA, resulting from an imbalance in signaling equilibrium and transcriptional changes that drives the skewed pathogenic CD4 effector immune response in GPA.
Collapse
Affiliation(s)
- Sangmi Kim
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Lena Boehme
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Louise Nel
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; and
| | - Alina Casian
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; and
| | - Shirish Sangle
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; and
| | - Estefania Nova-Lamperti
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, University of Concepcion, Concepcion, Chile
| | - Vlad Seitan
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Jo Spencer
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Paul Lavender
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - David P D'Cruz
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; and
| | - Susan John
- School of Immunology & Microbial Sciences, King's College London, London, United Kingdom;
| |
Collapse
|
12
|
Campe J, Ullrich E. T Helper Cell Lineage-Defining Transcription Factors: Potent Targets for Specific GVHD Therapy? Front Immunol 2022; 12:806529. [PMID: 35069590 PMCID: PMC8766661 DOI: 10.3389/fimmu.2021.806529] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Allogenic hematopoietic stem cell transplantation (allo-HSCT) represents a potent and potentially curative treatment for many hematopoietic malignancies and hematologic disorders in adults and children. The donor-derived immunity, elicited by the stem cell transplant, can prevent disease relapse but is also responsible for the induction of graft-versus-host disease (GVHD). The pathophysiology of acute GVHD is not completely understood yet. In general, acute GVHD is driven by the inflammatory and cytotoxic effect of alloreactive donor T cells. Since several experimental approaches indicate that CD4 T cells play an important role in initiation and progression of acute GVHD, the contribution of the different CD4 T helper (Th) cell subtypes in the pathomechanism and regulation of the disease is a central point of current research. Th lineages derive from naïve CD4 T cell progenitors and lineage commitment is initiated by the surrounding cytokine milieu and subsequent changes in the transcription factor (TF) profile. Each T cell subtype has its own effector characteristics, immunologic function, and lineage specific cytokine profile, leading to the association with different immune responses and diseases. Acute GVHD is thought to be mainly driven by the Th1/Th17 axis, whereas Treg cells are attributed to attenuate GVHD effects. As the differentiation of each Th subset highly depends on the specific composition of activating and repressing TFs, these present a potent target to alter the Th cell landscape towards a GVHD-ameliorating direction, e.g. by inhibiting Th1 and Th17 differentiation. The finding, that targeting of Th1 and Th17 differentiation appears more effective for GVHD-prevention than a strategy to inhibit Th1 and Th17 cytokines supports this concept. In this review, we shed light on the current advances of potent TF inhibitors to alter Th cell differentiation and consecutively attenuate GVHD. We will focus especially on preclinical studies and outcomes of TF inhibition in murine GVHD models. Finally, we will point out the possible impact of a Th cell subset-specific immune modulation in context of GVHD.
Collapse
Affiliation(s)
- Julia Campe
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung (DKTK)), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Small Molecule Inhibitors Targeting Nuclear Factor κB Activation Markedly Reduce Expression of Interleukin-2, but Not Interferon-γ, Induced by Phorbol Esters and Calcium Ionophores. Int J Mol Sci 2021; 22:ijms222313098. [PMID: 34884902 PMCID: PMC8658103 DOI: 10.3390/ijms222313098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/24/2022] Open
Abstract
The T-box transcription factor Eomesodermin (Eomes) promotes the expression of interferon-γ (IFN-γ). We recently reported that the small molecule inhibitors, TPCA-1 and IKK-16, which target nuclear factor κB (NF-κB) activation, moderately reduced Eomes-dependent IFN-γ expression in mouse lymphoma BW5147 cells stimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin (IM). In the present study, we investigated the direct effects of NF-κB on IFN-γ expression in mouse lymphoma EL4 cells and primary effector T cells. Eomes strongly promoted IFN-γ expression and the binding of RelA and NFATc2 to the IFN-γ promoter when EL4 cells were stimulated with PMA and IM. Neither TPCA-1 nor IKK-16 reduced IFN-γ expression; however, they markedly decreased interleukin (IL)-2 expression in Eomes-transfected EL4 cells. Moreover, TPCA-1 markedly inhibited the binding of RelA, but not that of Eomes or NFATc2 to the IFN-γ promoter. In effector CD4+ and CD8+ T cells activated with anti-CD3 and anti-CD28 antibodies, IFN-γ expression induced by PMA and A23187 was not markedly decreased by TPCA-1 or IKK-16 under conditions where IL-2 expression was markedly reduced. Therefore, the present results revealed that NF-κB is dispensable for IFN-γ expression induced by PMA and calcium ionophores in EL4 cells expressing Eomes and primary effector T cells.
Collapse
|
14
|
Czarnowicki T, Kim HJ, Villani AP, Glickman J, Duca ED, Han J, Pavel AB, Lee BH, Rahman AH, Merad M, Krueger JG, Guttman‐Yassky E. High-dimensional analysis defines multicytokine T-cell subsets and supports a role for IL-21 in atopic dermatitis. Allergy 2021; 76:3080-3093. [PMID: 33818809 DOI: 10.1111/all.14845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Flow cytometry is a well-accepted approach for immune profiling; however, its value is restricted by the limited number of markers that can be analyzed simultaneously. Mass cytometry/CyTOF offers broad-scale immune characterization integrating large number of parameters. While partial blood phenotyping was reported in atopic dermatitis (AD), patients' comprehensive profiling, critical for leveraging new targeted treatments, is not available. IL-21 may be involved in inflammatory skin diseases but its role in AD is not well established. METHODS We studied T-cell polarization in the blood of 20 moderate-to-severe AD and 15 controls. Using CyTOF and an unsupervised analysis, we measured the frequencies and mean metal intensities of activated polar CD4+ /CD8+ T-cell subsets. Immunohistochemistry, immunofluorescence, and qRT-PCR were used to analyze skin samples. RESULTS Examining 24 surface, intracellular markers, and transcription factors, we identified six CD4+ and five CD8+ T-cell metaclusters. A CD4+ skin-homing IL-13+ monocytokine and a novel IL-13+ IL-21+ multicytokine metaclusters were increased in AD vs. controls (p < .01). While IL-13 signature characterized both clusters, levels were significantly higher in the IL-21+ group. Both clusters correlated with AD severity (r = 0.49, p = .029). Manual gating corroborated these results and identified additional multicytokine subsets in AD. Immunohistochemistry and immunofluorescence, validated by mRNA expression, displayed significantly increasedIL-21 counts and colocalization with IL-13/IL-4R in AD skin. CONCLUSION A multicytokine signature characterizes moderate-to-severe AD, possibly explaining partial therapeutic responses to one cytokine targeting, particularly in severe patients. Prominent IL-21 signature in blood and skin hints for a potential pathogenic role of IL-21 in AD.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Hyun Je Kim
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Axel P. Villani
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Jacob Glickman
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ester Del Duca
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Joseph Han
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ana B. Pavel
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Brian H. Lee
- Human Immune Monitoring Center Icahn School of Medicine at Mt. Sinai New York NY USA
| | - Adeeb H. Rahman
- Human Immune Monitoring Center Icahn School of Medicine at Mt. Sinai New York NY USA
- Department of Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai New York NY USA
| | - Miriam Merad
- Department of Oncological Sciences Icahn School of Medicine at Mount Sinai New York NY USA
- Icahn School of Medicine at Mount Sinai The Precision Immunology Institute New York NY USA
- Icahn School of Medicine at Mount Sinai The Tisch Cancer Institute New York NY USA
| | - James G. Krueger
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Emma Guttman‐Yassky
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| |
Collapse
|
15
|
Zhang J, Le Gras S, Pouxvielh K, Faure F, Fallone L, Kern N, Moreews M, Mathieu AL, Schneider R, Marliac Q, Jung M, Berton A, Hayek S, Vidalain PO, Marçais A, Dodard G, Dejean A, Brossay L, Ghavi-Helm Y, Walzer T. Sequential actions of EOMES and T-BET promote stepwise maturation of natural killer cells. Nat Commun 2021; 12:5446. [PMID: 34521844 PMCID: PMC8440589 DOI: 10.1038/s41467-021-25758-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/06/2021] [Indexed: 02/08/2023] Open
Abstract
EOMES and T-BET are related T-box transcription factors that control natural killer (NK) cell development. Here we demonstrate that EOMES and T-BET regulate largely distinct gene sets during this process. EOMES is dominantly expressed in immature NK cells and drives early lineage specification by inducing hallmark receptors and functions. By contrast, T-BET is dominant in mature NK cells, where it induces responsiveness to IL-12 and represses the cell cycle, likely through transcriptional repressors. Regardless, many genes with distinct functions are co-regulated by the two transcription factors. By generating two gene-modified mice facilitating chromatin immunoprecipitation of endogenous EOMES and T-BET, we show a strong overlap in their DNA binding targets, as well as extensive epigenetic changes during NK cell differentiation. Our data thus suggest that EOMES and T-BET may distinctly govern, via differential expression and co-factors recruitment, NK cell maturation by inserting partially overlapping epigenetic regulations.
Collapse
MESH Headings
- Animals
- Base Sequence
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- CD11b Antigen/genetics
- CD11b Antigen/immunology
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Cycle/immunology
- Cell Differentiation
- Cell Lineage/drug effects
- Cell Lineage/genetics
- Cell Lineage/immunology
- Epigenesis, Genetic/immunology
- Interleukin-12/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Promoter Regions, Genetic
- Protein Binding
- Spleen/cytology
- Spleen/immunology
- T-Box Domain Proteins/deficiency
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- Transcription, Genetic
- Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
Collapse
Affiliation(s)
- Jiang Zhang
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Stéphanie Le Gras
- IGBMC, CNRS UMR7104, Inserm U1258, Université de Strasbourg, Illkirch, France
- Plateforme GenomEast, infrastructure France Génomique, Illkirch, France
| | - Kevin Pouxvielh
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Fabrice Faure
- Institut NeuroMyoGène, INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard, Lyon 1, Lyon, France
| | - Lucie Fallone
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Nicolas Kern
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Marion Moreews
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Raphaël Schneider
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon Université Claude Bernard Lyon 1, 46 allée d'Italie, F-69364, Lyon, France
| | - Quentin Marliac
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Mathieu Jung
- IGBMC, CNRS UMR7104, Inserm U1258, Université de Strasbourg, Illkirch, France
- Plateforme GenomEast, infrastructure France Génomique, Illkirch, France
| | - Aurore Berton
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Simon Hayek
- Equipe Chimie et Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS UMR 8601, 75006, Paris, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Chimie et Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS UMR 8601, 75006, Paris, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Garvin Dodard
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Anne Dejean
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Yad Ghavi-Helm
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon Université Claude Bernard Lyon 1, 46 allée d'Italie, F-69364, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France.
| |
Collapse
|
16
|
Roessner PM, Llaó Cid L, Lupar E, Roider T, Bordas M, Schifflers C, Arseni L, Gaupel AC, Kilpert F, Krötschel M, Arnold SJ, Sellner L, Colomer D, Stilgenbauer S, Dietrich S, Lichter P, Izcue A, Seiffert M. EOMES and IL-10 regulate antitumor activity of T regulatory type 1 CD4 + T cells in chronic lymphocytic leukemia. Leukemia 2021; 35:2311-2324. [PMID: 33526861 PMCID: PMC8324479 DOI: 10.1038/s41375-021-01136-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 11/19/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
The transcription factor eomesodermin (EOMES) promotes interleukin (IL)-10 expression in CD4+ T cells, which has been linked to immunosuppressive and cytotoxic activities. We detected cytotoxic, programmed cell death protein-1 (PD-1) and EOMES co-expressing CD4+ T cells in lymph nodes (LNs) of patients with chronic lymphocytic leukemia (CLL) or diffuse large B-cell lymphoma. Transcriptome and flow cytometry analyses revealed that EOMES does not only drive IL-10 expression, but rather controls a unique transcriptional signature in CD4+ T cells, that is enriched in genes typical for T regulatory type 1 (TR1) cells. The TR1 cell identity of these CD4+ T cells was supported by their expression of interferon gamma and IL-10, as well as inhibitory receptors including PD-1. TR1 cells with cytotoxic capacity accumulate also in Eµ-TCL1 mice that develop CLL-like disease. Whereas wild-type CD4+ T cells control TCL1 leukemia development after adoptive transfer in leukopenic Rag2-/- mice, EOMES-deficient CD4+ T cells failed to do so. We further show that TR1 cell-mediated control of TCL1 leukemia requires IL-10 receptor (IL-10R) signaling, as Il10rb-deficient CD4+ T cells showed impaired antileukemia activity. Altogether, our data demonstrate that EOMES is indispensable for the development of IL-10-expressing, cytotoxic TR1 cells, which accumulate in LNs of CLL patients and control TCL1 leukemia in mice in an IL-10R-dependent manner.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- Cell Differentiation
- Female
- Gene Expression Regulation, Leukemic
- Humans
- Interferon-gamma
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/prevention & control
- Mice
- Mice, Inbred C57BL
- Prognosis
- Signal Transduction
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/immunology
- Transcriptome
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Philipp M Roessner
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Llaó Cid
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ekaterina Lupar
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Cellzome, Heidelberg, Germany
| | - Tobias Roider
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Marie Bordas
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Christoph Schifflers
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cell Biology Research Unit (URBC)-Namur Research Institute of Life Science (Narilis), University of Namur, Namur, Belgium
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lavinia Arseni
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ann-Christin Gaupel
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Kilpert
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Essen University Hospital, Institute of Human Genetics, Genome Informatics, Essen, Germany
| | - Marit Krötschel
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- BioMed X Institute, Heidelberg, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Leopold Sellner
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Dolors Colomer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hematopathology Unit, Hospital Clinic, CIBERONC, Barcelona, Spain
| | | | - Sascha Dietrich
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana Izcue
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, Freiburg, Germany
- Institute of Molecular Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Niu W, Xu Y, Zha X, Zeng J, Qiao S, Yang S, Zhang H, Tan L, Sun L, Pang G, Liu T, Zhao H, Zheng N, Zhang Y, Bai H. IL-21/IL-21R Signaling Aggravated Respiratory Inflammation Induced by Intracellular Bacteria through Regulation of CD4 + T Cell Subset Responses. THE JOURNAL OF IMMUNOLOGY 2021; 206:1586-1596. [PMID: 33608454 DOI: 10.4049/jimmunol.2001107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/19/2021] [Indexed: 12/16/2022]
Abstract
The IL-21/IL-21R interaction plays an important role in a variety of immune diseases; however, the roles and mechanisms in intracellular bacterial infection are not fully understood. In this study, we explored the effect of IL-21/IL-21R on chlamydial respiratory tract infection using a chlamydial respiratory infection model. The results showed that the mRNA expression of IL-21 and IL-21R was increased in Chlamydia muridarum-infected mice, which suggested that IL-21 and IL-21R were involved in host defense against C. muridarum lung infection. IL-21R-/- mice exhibited less body weight loss, a lower bacterial burden, and milder pathological changes in the lungs than wild-type (WT) mice during C. muridarum lung infection. The absolute number and activity of CD4+ T cells and the strength of Th1/Th17 responses in IL-21R-/- mice were significantly higher than those in WT mice after C. muridarum lung infection, but the Th2 response was weaker. Consistently, IL-21R-/- mice showed higher mRNA expression of Th1 transcription factors (T-bet/STAT4), IL-12p40, a Th17 transcription factor (STAT3), and IL-23. The mRNA expression of Th2 transcription factors (GATA3/STAT6), IL-4, IL-10, and TGF-β in IL-21R-/- mice was significantly lower than that in WT mice. Furthermore, the administration of recombinant mouse IL-21 aggravated chlamydial lung infection in C57BL/6 mice and reduced Th1 and Th17 responses following C. muridarum lung infection. These findings demonstrate that IL-21/IL-21R may aggravate chlamydial lung infection by inhibiting Th1 and Th17 responses.
Collapse
Affiliation(s)
- Wenhao Niu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yueyue Xu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Xiaoyu Zha
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Jiajia Zeng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Sai Qiao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Shuaini Yang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lu Tan
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lida Sun
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Gaoju Pang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Tengli Liu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Huili Zhao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Ningbo Zheng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yongci Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| |
Collapse
|
18
|
Ashida S, Ochi H, Hamatani M, Fujii C, Kimura K, Okada Y, Hashi Y, Kawamura K, Ueno H, Takahashi R, Mizuno T, Kondo T. Immune Skew of Circulating Follicular Helper T Cells Associates With Myasthenia Gravis Severity. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e945. [PMID: 33436376 PMCID: PMC8105905 DOI: 10.1212/nxi.0000000000000945] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/04/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To clarify functional alterations of follicular helper T cells (Tfh) in myasthenia gravis (MG) because Tfh play important roles in helping B cells generate antibody-producing cells. METHODS A total of 24 immunotherapy-naive patients with anti-acetylcholine receptor (AchR) antibody-positive MG and 18 age-matched healthy subjects (HS) were enrolled. Samples from 6 patients were available for posttreatment analysis. Subsets of circulating Tfh (cTfh) and B cells were identified by flow cytometry analysis of surface molecules. Cytokine production by isolated cTfh subsets from 5 patients with MG and 5 HS was measured in vitro. Analysis was performed to examine the correlation between the frequency of cTfh subsets and that of plasmablasts and between cTfh subsets and the quantitative MG score. RESULTS cTfh increased with elevated expression of inducible T-cell costimulator (ICOS) in patients with MG. cTfh shifted to Th2 and Th17 over Th1 in MG. ICOShighcTfh produced significantly higher levels of interleukin (IL)-21, IL-4, and IL-17A than ICOSlow cTfh only in patients with MG. The frequency of cTfh within CD4 T cells was more closely associated with disease severity than the serum anti-AchR antibody titer and frequency of plasmablasts within B cells. Abnormalities of cTfh were improved after immunotherapy in parallel with clinical improvement. CONCLUSIONS Alternation of cTfh is a key feature in the development of MG and may become a biomarker for disease severity and therapeutic efficacy. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that the level of cTfh is associated with disease severity in patients with MG.
Collapse
Affiliation(s)
- Shinji Ashida
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Hirofumi Ochi
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Mio Hamatani
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Chihiro Fujii
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Kimitoshi Kimura
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Yoichiro Okada
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Yuichiro Hashi
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Kazuyuki Kawamura
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Hideki Ueno
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Ryosuke Takahashi
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Toshiki Mizuno
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| | - Takayuki Kondo
- From the Department of Neurology (S.A., C.F., T.M.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; Department of Geriatric Medicine and Neurology (H.O.), Ehime University Graduate School of Medicine, Toon; Department of Neurology (M.H., R.T.), Kyoto University Graduate School of Medicine; Department of Neurology (M.H., Y.O., Y.H., T.K.), Kansai Medical University Medical Center, Osaka, Japan; Brigham and Women's Hospital (K. Kimura), Harvard Medical School, Boston, MA; Department of Neurology (K. Kawamura), National Hospital Organization Minami Kyoto Hospital; and Department of Immunology (H.U.), Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
19
|
Chen S, Zhang J, Yu WB, Zhuang JC, Xiao W, Wu ZY, Xiao BG. Eomesodermin in CD4 +T cells is essential for Ginkgolide K ameliorating disease progression in experimental autoimmune encephalomyelitis. Int J Biol Sci 2021; 17:50-61. [PMID: 33390832 PMCID: PMC7757039 DOI: 10.7150/ijbs.50041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/22/2020] [Indexed: 11/05/2022] Open
Abstract
Eomesodermin (Eomes), a transcription factor, could suppress the Th17 cell differentiation and proliferation through directly binding to the promoter zone of the Rorc and Il17a gene, meanwhile the expression of Eomes is suppressed when c-Jun directly binds to its promoter zone. Ginkgolide K (1,10-dihydroxy-3,14-didehydroginkgolide, GK) is a diterpene lactone isolated from the leaves of Ginkgo biloba. A previous study indicated that GK could decrease the level of phospho JNK (c-Jun N-terminal kinase). Here, we reported the therapeutic potential of Ginkgolide K (GK) treatment to ameliorate experimental autoimmune encephalomyelitis (EAE) disease progression. Methods: EAE was induced in both wildtype and CD4-Eomes conditional knockout mice. GK was injected intraperitoneally. Disease severity, inflammation, and tissue damage were assessed by clinical evaluation, flow cytometry of mononuclear cells (MNCs), and histopathological evaluation. Dual-luciferase reporter assays were performed to measure Eomes transcription activity in vitro. The potency of GK (IC50) was determined using JNK1 Kinase Enzyme System. Results: We revealed that GK could ameliorate EAE disease progression by the inhibition of the Th17 cells. Further mechanism studies demonstrated that the level of phospho JNK was decreased and the level of Eomes in CD4+T cells was dramatically increased. This therapeutic effect of GK was almost completely interrupted in CD4-Eomes conditional knockout mice. Conclusions: These results provided the therapeutic potential of GK treatment in EAE, and further suggested that Eomes expression in CD4+T cells might be essential in this process.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurology, Huashan Hospital, Fudan University
- Department of Neurology, Fujian Medical University Union Hospital
| | - Juan Zhang
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine
| | - Wen-Bo Yu
- Department of Neurology, Huashan Hospital, Fudan University
| | - Jing-Cong Zhuang
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University
| | - Wei Xiao
- Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine
| | - Bao-Guo Xiao
- Department of Neurology, Huashan Hospital, Fudan University
| |
Collapse
|
20
|
Eomesodermin promotes interaction of RelA and NFATc2 with the Ifng promoter and multiple conserved noncoding sequences across the Ifng locus in mouse lymphoma BW5147 cells. Immunol Lett 2020; 225:33-43. [DOI: 10.1016/j.imlet.2020.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/06/2020] [Accepted: 06/11/2020] [Indexed: 01/08/2023]
|
21
|
Gryzik S, Hoang Y, Lischke T, Mohr E, Venzke M, Kadner I, Poetzsch J, Groth D, Radbruch A, Hutloff A, Baumgrass R. Identification of a super-functional Tfh-like subpopulation in murine lupus by pattern perception. eLife 2020; 9:53226. [PMID: 32441253 PMCID: PMC7274784 DOI: 10.7554/elife.53226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/20/2020] [Indexed: 01/20/2023] Open
Abstract
Dysregulated cytokine expression by T cells plays a pivotal role in the pathogenesis of autoimmune diseases. However, the identification of the corresponding pathogenic subpopulations is a challenge, since a distinction between physiological variation and a new quality in the expression of protein markers requires combinatorial evaluation. Here, we were able to identify a super-functional follicular helper T cell (Tfh)-like subpopulation in lupus-prone NZBxW mice with our binning approach "pattern recognition of immune cells (PRI)". PRI uncovered a subpopulation of IL-21+ IFN-γhigh PD-1low CD40Lhigh CXCR5- Bcl-6- T cells specifically expanded in diseased mice. In addition, these cells express high levels of TNF-α and IL-2, and provide B cell help for IgG production in an IL-21 and CD40L dependent manner. This super-functional T cell subset might be a superior driver of autoimmune processes due to a polyfunctional and high cytokine expression combined with Tfh-like properties.
Collapse
Affiliation(s)
- Stefanie Gryzik
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Yen Hoang
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Timo Lischke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Elodie Mohr
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Melanie Venzke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Isabelle Kadner
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Josephine Poetzsch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | | | - Andreas Radbruch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,Charité, Campus Mitte, Berlin, Germany
| | - Andreas Hutloff
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Ria Baumgrass
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| |
Collapse
|
22
|
Inhibition of E protein activity facilitates the quiescence exit of naïve CD4+ T cells through modulating PI3K-AKT signaling and TCR microcluster formation. Cell Immunol 2020; 351:104065. [DOI: 10.1016/j.cellimm.2020.104065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 02/02/2020] [Accepted: 02/10/2020] [Indexed: 11/24/2022]
|
23
|
Preglej T, Hamminger P, Luu M, Bulat T, Andersen L, Göschl L, Stolz V, Rica R, Sandner L, Waltenberger D, Tschismarov R, Faux T, Boenke T, Laiho A, Elo LL, Sakaguchi S, Steiner G, Decker T, Bohle B, Visekruna A, Bock C, Strobl B, Seiser C, Boucheron N, Ellmeier W. Histone deacetylases 1 and 2 restrain CD4+ cytotoxic T lymphocyte differentiation. JCI Insight 2020; 5:133393. [PMID: 32102981 PMCID: PMC7101144 DOI: 10.1172/jci.insight.133393] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/24/2020] [Indexed: 12/11/2022] Open
Abstract
Some effector CD4+ T cell subsets display cytotoxic activity, thus breaking the functional dichotomy of CD4+ helper and CD8+ cytotoxic T lymphocytes. However, molecular mechanisms regulating CD4+ cytotoxic T lymphocyte (CD4+ CTL) differentiation are poorly understood. Here we show that levels of histone deacetylases 1 and 2 (HDAC1-HDAC2) are key determinants of CD4+ CTL differentiation. Deletions of both Hdac1 and 1 Hdac2 alleles (HDAC1cKO-HDAC2HET) in CD4+ T cells induced a T helper cytotoxic program that was controlled by IFN-γ-JAK1/2-STAT1 signaling. In vitro, activated HDAC1cKO-HDAC2HET CD4+ T cells acquired cytolytic activity and displayed enrichment of gene signatures characteristic of effector CD8+ T cells and human CD4+ CTLs. In vivo, murine cytomegalovirus-infected HDAC1cKO-HDAC2HET mice displayed a stronger induction of CD4+ CTL features compared with infected WT mice. Finally, murine and human CD4+ T cells treated with short-chain fatty acids, which are commensal-produced metabolites acting as HDAC inhibitors, upregulated CTL genes. Our data demonstrate that HDAC1-HDAC2 restrain CD4+ CTL differentiation. Thus, HDAC1-HDAC2 might be targets for the therapeutic induction of CD4+ CTLs.
Collapse
Affiliation(s)
- Teresa Preglej
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Maik Luu
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Tanja Bulat
- Institute of Animal Breeding and Genetics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Liisa Andersen
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lisa Göschl
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Valentina Stolz
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ramona Rica
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lisa Sandner
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Darina Waltenberger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Faux
- Medical Bioinformatics Centre, Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Thorina Boenke
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Asta Laiho
- Medical Bioinformatics Centre, Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Laura L. Elo
- Medical Bioinformatics Centre, Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Shinya Sakaguchi
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Günter Steiner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, and
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
IL-21 promotes allergic airway inflammation by driving apoptosis of FoxP3 + regulatory T cells. J Allergy Clin Immunol 2019; 143:2178-2189.e5. [PMID: 30654048 DOI: 10.1016/j.jaci.2018.11.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/05/2018] [Accepted: 11/30/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND IL-21 is a key player of adaptive immunity, with well-established roles in B-cell and cytotoxic T-cell responses. IL-21 has been implicated in promotion of effector CD4+ T cells and inhibition of forkhead box P3-positive regulatory T (Treg) cells, but the mechanism and functional relevance of these findings remain controversial. OBJECTIVE We sought to understand the mechanisms by which IL-21 controls effector CD4+ cell responses and Treg cell homeostasis. METHODS We used IL-21 receptor-deficient mice to study the effect of IL-21 on T-cell responses in models of asthma and colitis. We used mixed bone marrow chimeras and adoptive transfer of naive CD4+ T cells and Treg cells into lymphopenic mice to assess the cell-intrinsic effects of IL-21. Using various in vitro T-cell assays, we characterized the mechanism of IL-21-mediated inhibition of Treg cells. RESULTS We show that IL-21 production by TH2 and follicular helper T/ex-follicular helper T cells promotes asthma by inhibiting Treg cells. Il21r-/- mice displayed reduced generation of TH2 cells and increased generation of Treg cells. In mixed chimeras we demonstrate that IL-21 promotes TH2 responses indirectly through inhibition of Treg cells. Depleting Treg cells in Il21r-/- mice restored TH2 generation and eosinophilia. Furthermore, IL-21 inhibited Treg cell generation in mice with colitis. Using competitive transfer of Il21r+/+ and Il21r-/- CD4+ cells, we show that IL-21 directly inhibited expansion of differentiated Treg cells but was dispensable for TH1/TH17 effectors. We show that IL-21 sensitizes Treg cells to apoptosis by interfering with the expression of Bcl-2 family genes. CONCLUSION IL-21 directly promotes apoptosis of Treg cells and therefore indirectly sustains generation of inflammatory TH cells and related effector responses.
Collapse
|
25
|
Mazzoni A, Maggi L, Siracusa F, Ramazzotti M, Rossi MC, Santarlasci V, Montaini G, Capone M, Rossettini B, Palma R, Kruglov A, Chang H, Cimaz R, Maggi E, Romagnani S, Liotta F, Cosmi L, Annunziato F. Eomes
controls the development of Th17‐derived (non‐classic) Th1 cells during chronic inflammation. Eur J Immunol 2018; 49:79-95. [DOI: 10.1002/eji.201847677] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/20/2018] [Accepted: 08/20/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | | | - Matteo Ramazzotti
- Department of Biomedical Experimental and Clinical Sciences “Mario Serio” University of Florence Firenze Italy
| | - Maria Caterina Rossi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Veronica Santarlasci
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Gianni Montaini
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Beatrice Rossettini
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Raffaele Palma
- Diparimento di Medicina di Precisione Università della Campania Napoli Italy
- Institute of Protein Biochemistry CNR Napoli
| | | | | | - Rolando Cimaz
- Anna Meyer Children's Hospital and University of Florence Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
- Flow cytometry and Immunotherapy Diagnostic Center Azienda Ospedaliera Careggi Florence Italy
| |
Collapse
|
26
|
The RNA binding protein Sam68 controls T helper 1 differentiation and anti-mycobacterial response through modulation of miR-29. Cell Death Differ 2018; 26:1169-1180. [PMID: 30258098 DOI: 10.1038/s41418-018-0201-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/08/2018] [Accepted: 08/27/2018] [Indexed: 12/24/2022] Open
Abstract
Polarization of naive T cells into interferon (IFN)-γ-producing T helper 1 (Th1) cells is an essential event in the inflammatory response to pathogens. Herein, we identify the RNA binding protein Sam68 as a specific modulator of Th1 differentiation. Sam68-knockout (ko) naive T cells are strongly defective in IL-12-mediated Th1 polarization and express low levels of T-bet and Eomes. Consequently, Sam68-ko Th1 cells are significantly impaired in IFN-γ production. Moreover, we found that Sam68 is required for the induction of an inflammatory Th1 response during Mycobacterium bovis Bacillus Calmette-Guerin (BCG) infection, thus limiting bacterial dissemination in the lungs. Mechanistically, Sam68 directly binds to the microRNA miR-29, a negative regulator of Th1 response, and inhibits its expression during BCG infection. These findings uncover a novel post-transcriptional mechanism required for the Th1-mediated defense against intracellular pathogens and identify a new function for Sam68 in the regulation of the immune response.
Collapse
|
27
|
Zhang J, Marotel M, Fauteux-Daniel S, Mathieu AL, Viel S, Marçais A, Walzer T. T-bet and Eomes govern differentiation and function of mouse and human NK cells and ILC1. Eur J Immunol 2018; 48:738-750. [PMID: 29424438 DOI: 10.1002/eji.201747299] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/14/2017] [Accepted: 02/06/2018] [Indexed: 11/08/2022]
Abstract
T-bet and Eomes are T-box transcription factors that drive the differentiation and function of cytotoxic lymphocytes such as NK cells. Their DNA-binding domains are highly similar, suggesting redundant transcriptional activity. However, while these transcription factors have different patterns of expression, the phenotype of loss-of-function mouse models suggests that they play distinct roles in the development of NK cells and other innate lymphoid cells (ILCs). Recent technological advances using reporter mice and conditional knockouts were fundamental in defining the regulation and function of these factors at steady state and during pathological conditions such as various types of cancer or infection. Here, we review these recent developments, focusing on NK cells as prototypical cytotoxic lymphocytes and their development, and also discuss parallels between NK cells and T cells. We also examine the role of T-bet and Eomes in human NK cells and ILC1s. Considering divergent findings on mouse and human ILC1s, we propose that NK cells are defined by coexpression of T-bet and Eomes, while ILC1s express only one of these factors, either T-bet or Eomes, depending on the tissue or the species.
Collapse
Affiliation(s)
- Jiang Zhang
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France.,Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Marie Marotel
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Sébastien Fauteux-Daniel
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Sébastien Viel
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France.,Laboratoire d'Immunologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, 69495, Pierre-Bénite, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie-International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| |
Collapse
|
28
|
Tsukamoto H, Fujieda K, Senju S, Ikeda T, Oshiumi H, Nishimura Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci 2017; 109:523-530. [PMID: 29090850 PMCID: PMC5834784 DOI: 10.1111/cas.13433] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
Accompanied by the growing clinical applications of immunotherapy in the treatment of cancer patients, development of novel therapeutic approaches to reverse the immune-suppressive environment in cancer patients is eagerly anticipated, because the success of cancer immunotherapy is currently limited by immune-suppressive effects in tumor-bearing hosts. Interleukin (IL)-6, a pleotropic proinflammatory cytokine, participates in tumor cell-autonomous processes that are required for their survival and growth, and is therefore known as a poor prognostic factor in cancer patients. In addition, an emerging role of IL-6 in modulating multiple functions of immune cells including T cells, dendritic cells, and macrophages is responsible for the dysfunction of innate and adaptive immunity against tumors. Therefore, the IL-6-targeting approach is of value as a promising strategy for desensitization and prevention of immune-suppressive effects, and should be an effective treatment when combined with current immunotherapies. The aim of the present review is to discuss the immune-suppressive aspects of IL-6, notably with modification of T-cell functions in cancer patients, and their relationship to anti-tumor immune responses and cancer immunotherapy.
Collapse
Affiliation(s)
- Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Fujieda
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
29
|
Tsukamoto H, Fujieda K, Senju S, Ikeda T, Oshiumi H, Nishimura Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci 2017. [PMID: 29090850 DOI: 10.1111/cas.13433.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Accompanied by the growing clinical applications of immunotherapy in the treatment of cancer patients, development of novel therapeutic approaches to reverse the immune-suppressive environment in cancer patients is eagerly anticipated, because the success of cancer immunotherapy is currently limited by immune-suppressive effects in tumor-bearing hosts. Interleukin (IL)-6, a pleotropic proinflammatory cytokine, participates in tumor cell-autonomous processes that are required for their survival and growth, and is therefore known as a poor prognostic factor in cancer patients. In addition, an emerging role of IL-6 in modulating multiple functions of immune cells including T cells, dendritic cells, and macrophages is responsible for the dysfunction of innate and adaptive immunity against tumors. Therefore, the IL-6-targeting approach is of value as a promising strategy for desensitization and prevention of immune-suppressive effects, and should be an effective treatment when combined with current immunotherapies. The aim of the present review is to discuss the immune-suppressive aspects of IL-6, notably with modification of T-cell functions in cancer patients, and their relationship to anti-tumor immune responses and cancer immunotherapy.
Collapse
Affiliation(s)
- Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Fujieda
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
30
|
Göschl L, Preglej T, Hamminger P, Bonelli M, Andersen L, Boucheron N, Gülich AF, Müller L, Saferding V, Mufazalov IA, Hirahara K, Seiser C, Matthias P, Penz T, Schuster M, Bock C, Waisman A, Steiner G, Ellmeier W. A T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis. J Autoimmun 2017; 86:51-61. [PMID: 28964722 DOI: 10.1016/j.jaut.2017.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 10/18/2022]
Abstract
Multiple sclerosis (MS) is a human neurodegenerative disease characterized by the invasion of autoreactive T cells from the periphery into the CNS. Application of pan-histone deacetylase inhibitors (HDACi) ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model for MS, suggesting that HDACi might be a potential therapeutic strategy for MS. However, the function of individual HDAC members in the pathogenesis of EAE is not known. In this study we report that mice with a T cell-specific deletion of HDAC1 (using the Cd4-Cre deleter strain; HDAC1-cKO) were completely resistant to EAE despite the ability of HDAC1cKO CD4+ T cells to differentiate into Th17 cells. RNA sequencing revealed STAT1 as a prominent upstream regulator of differentially expressed genes in activated HDAC1-cKO CD4+ T cells and this was accompanied by a strong increase in phosphorylated STAT1 (pSTAT1). This suggests that HDAC1 controls STAT1 activity in activated CD4+ T cells. Increased pSTAT1 levels correlated with a reduced expression of the chemokine receptors Ccr4 and Ccr6, which are important for the migration of T cells into the CNS. Finally, EAE susceptibility was restored in WT:HDAC1-cKO mixed BM chimeric mice, indicating a cell-autonomous defect. Our data demonstrate a novel pathophysiological role for HDAC1 in EAE and provide evidence that selective inhibition of HDAC1 might be a promising strategy for the treatment of MS.
Collapse
Affiliation(s)
- Lisa Göschl
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Teresa Preglej
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Liisa Andersen
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nicole Boucheron
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexandra F Gülich
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Lena Müller
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Victoria Saferding
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; AMED-PRIME, AMED, Chiba 260-8670, Japan
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, 1090 Vienna, Austria
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Faculty of Sciences, University of Basel, 4031 Basel, Switzerland
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; Max Planck Institute for Informatics, 66123, Saarbrücken, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Günter Steiner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
31
|
Miyauchi K. Helper T Cell Responses to Respiratory Viruses in the Lung: Development, Virus Suppression, and Pathogenesis. Viral Immunol 2017. [DOI: 10.1089/vim.2017.0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kosuke Miyauchi
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| |
Collapse
|
32
|
Goswami R, Kaplan M. STAT Transcription Factors in T Cell Control of Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:123-180. [DOI: 10.1016/bs.ircmb.2016.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Heink S, Yogev N, Garbers C, Herwerth M, Aly L, Gasperi C, Husterer V, Croxford AL, Möller-Hackbarth K, Bartsch HS, Sotlar K, Krebs S, Regen T, Blum H, Hemmer B, Misgeld T, Wunderlich TF, Hidalgo J, Oukka M, Rose-John S, Schmidt-Supprian M, Waisman A, Korn T. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic T H17 cells. Nat Immunol 2016; 18:74-85. [PMID: 27893700 PMCID: PMC5164931 DOI: 10.1038/ni.3632] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 11/04/2016] [Indexed: 12/16/2022]
Abstract
The cellular sources of interleukin-6 (IL-6) that are relevant for the differentiation of TH17 cells remain unclear. Here, we used a novel strategy of IL-6 conditional deletion of distinct IL-6-producing cell types to show that Sirpα+ dendritic cells (DC) were essential for the generation of pathogenic TH17 cells. During the process of cognate interaction, Sirpα+ DCs trans-presented IL-6 to T cells using their own IL-6Rα. While ambient IL-6 was sufficient to suppress the induction of the transcription factor Foxp3 in T cells, IL-6 trans-presentation by DC-bound IL-6Rα (here defined as IL-6 cluster signaling) was required to prevent premature induction of IFN-γ in T cells and to generate pathogenic TH17 cells in vivo. These findings will guide therapeutic approaches for TH17-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Sylvia Heink
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Marina Herwerth
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany.,Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Lilian Aly
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | - Christiane Gasperi
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | - Veronika Husterer
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | - Andrew L Croxford
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Harald S Bartsch
- Institute of Pathology, Medical School, Ludwig-Maximilians-University, Munich, Germany
| | - Karl Sotlar
- Institute of Pathology, Medical School, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Krebs
- Gene Centre, Lafuga, Ludwig-Maximilians-University, Munich, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Helmut Blum
- Gene Centre, Lafuga, Ludwig-Maximilians-University, Munich, Germany
| | - Bernhard Hemmer
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Juan Hidalgo
- Department of Cellular Biology, Physiology, and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | - Mohamed Oukka
- Department of Immunology, University of Washington, Seattle, Washington, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Marc Schmidt-Supprian
- Department of Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
34
|
Stienne C, Michieletto MF, Benamar M, Carrié N, Bernard I, Nguyen XH, Lippi Y, Duguet F, Liblau RS, Hedrick SM, Saoudi A, Dejean AS. Foxo3 Transcription Factor Drives Pathogenic T Helper 1 Differentiation by Inducing the Expression of Eomes. Immunity 2016; 45:774-787. [PMID: 27742544 DOI: 10.1016/j.immuni.2016.09.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/21/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023]
Abstract
The transcription factor Foxo3 plays a crucial role in myeloid cell function but its role in lymphoid cells remains poorly defined. Here, we have shown that Foxo3 expression was increased after T cell receptor engagement and played a specific role in the polarization of CD4+ T cells toward pathogenic T helper 1 (Th1) cells producing interferon-γ (IFN-γ) and granulocyte monocyte colony stimulating factor (GM-CSF). Consequently, Foxo3-deficient mice exhibited reduced susceptibility to experimental autoimmune encephalomyelitis. At the molecular level, we identified Eomes as a direct target gene for Foxo3 in CD4+ T cells and we have shown that lentiviral-based overexpression of Eomes in Foxo3-deficient CD4+ T cells restored both IFN-γ and GM-CSF production. Thus, the Foxo3-Eomes pathway is central to achieve the complete specialized gene program required for pathogenic Th1 cell differentiation and development of neuroinflammation.
Collapse
Affiliation(s)
- Caroline Stienne
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Michaël F Michieletto
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Mehdi Benamar
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | | | - Isabelle Bernard
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Xuan-Hung Nguyen
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Yannick Lippi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse 31024, France
| | - Fanny Duguet
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Roland S Liblau
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Stephen M Hedrick
- Molecular Biology Section, Division of Biological Sciences and Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0377, USA
| | - Abdelhadi Saoudi
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France
| | - Anne S Dejean
- UMR Inserm, U1043, Toulouse 31300, France; UMR CNRS, U5282, Toulouse 31300, France; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse 31300, France.
| |
Collapse
|
35
|
Tian Y, Zajac AJ. IL-21 and T Cell Differentiation: Consider the Context. Trends Immunol 2016; 37:557-568. [PMID: 27389961 DOI: 10.1016/j.it.2016.06.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/27/2016] [Accepted: 06/01/2016] [Indexed: 12/18/2022]
Abstract
Accumulating studies demonstrate that IL-21 modulates the differentiation of various CD4 and CD8 T cell subsets and provide insights into the underlying cellular and molecular processes that are influenced by this cytokine. Intriguingly, the effects of IL-21 on T cells can be complex and vary depending on the experimental system used. We review our current understanding of the roles of IL-21 in the generation of phenotypically distinct CD4 and CD8 T cell populations and discuss the potential environmental cues, cellular factors, and molecular mediators that impact the actions of IL-21. We propose that IL-21 acts in a context-dependent manner to accentuate T cell subset development.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA
| | - Allan J Zajac
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA.
| |
Collapse
|
36
|
Krausgruber T, Schiering C, Adelmann K, Harrison OJ, Chomka A, Pearson C, Ahern PP, Shale M, Oukka M, Powrie F. T-bet is a key modulator of IL-23-driven pathogenic CD4(+) T cell responses in the intestine. Nat Commun 2016; 7:11627. [PMID: 27193261 PMCID: PMC4874038 DOI: 10.1038/ncomms11627] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/14/2016] [Indexed: 01/04/2023] Open
Abstract
IL-23 is a key driver of pathogenic Th17 cell responses. It has been suggested that the transcription factor T-bet is required to facilitate IL-23-driven pathogenic effector functions; however, the precise role of T-bet in intestinal T cell responses remains elusive. Here, we show that T-bet expression by T cells is not required for the induction of colitis or the differentiation of pathogenic Th17 cells but modifies qualitative features of the IL-23-driven colitogenic response by negatively regulating IL-23R expression. Consequently, absence of T-bet leads to unrestrained Th17 cell differentiation and activation characterized by high amounts of IL-17A and IL-22. The combined increase in IL-17A/IL-22 results in enhanced epithelial cell activation and inhibition of either IL-17A or IL-22 leads to disease amelioration. Our study identifies T-bet as a key modulator of IL-23-driven colitogenic responses in the intestine and has important implications for understanding of heterogeneity among inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Thomas Krausgruber
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
| | - Chris Schiering
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Krista Adelmann
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Oliver J. Harrison
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Agnieszka Chomka
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
| | - Claire Pearson
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
| | - Philip P. Ahern
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Matthew Shale
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Mohamed Oukka
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | - Fiona Powrie
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
| |
Collapse
|
37
|
Tavakolpour S. Interleukin 21 as a new possible player in pemphigus: Is it a suitable target? Int Immunopharmacol 2016; 34:139-145. [DOI: 10.1016/j.intimp.2016.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/15/2016] [Accepted: 02/19/2016] [Indexed: 11/26/2022]
|
38
|
Fukuoka N, Harada M, Nishida A, Ito Y, Shiota H, Kataoka T. Eomesodermin promotes interferon-γ expression and binds to multiple conserved noncoding sequences across the Ifng locus in mouse thymoma cell lines. Genes Cells 2016; 21:146-62. [PMID: 26749212 DOI: 10.1111/gtc.12328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 11/23/2015] [Indexed: 01/03/2023]
Abstract
The T-box transcription factors T-bet and eomesodermin (Eomes) have been shown to regulate the lineage-specific expression of interferon-γ (IFN-γ). However, in contrast to T-bet, the role of Eomes in the expression of IFN-γ remains unclear. In this study, we investigated the Eomes-dependent expression of IFN-γ in the mouse thymoma BW5147 and EL4 cells, which do not express T-bet or Eomes. The ectopic expression of Eomes induced BW5147 and EL4 cells to produce IFN-γ in response to phorbol 12-myristate 13-acetate (PMA) and ionomycin (IM). In BW5147 cells, Eomes augmented luciferase activity driven by the Ifng promoter encoding from -2500 to +113 bp; however, it was not increased by a stimulation with PMA and IM. A chromatin immunoprecipitation assay showed that Eomes bound to the Ifng promoter and conserved noncoding sequence (CNS) -22 kb across the Ifng locus with high efficacy in BW5147 cells. Moreover, Eomes increased permissive histone modifications in the Ifng promoter and multiple CNSs. The stimulation with PMA and IM greatly augmented Eomes binding to CNS-54, CNS-34, CNS+19 and CNS+30, which was inhibited by FK506. These results indicated that Eomes bound to the Ifng promoter and multiple CNSs in stimulation-dependent and stimulation-independent manners.
Collapse
Affiliation(s)
- Natsuki Fukuoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Misuzu Harada
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Ai Nishida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yuko Ito
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Hideki Shiota
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Takao Kataoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan.,Center for Biological Resources and Informatics, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| |
Collapse
|
39
|
Carpio VH, Opata MM, Montañez ME, Banerjee PP, Dent AL, Stephens R. IFN-γ and IL-21 Double Producing T Cells Are Bcl6-Independent and Survive into the Memory Phase in Plasmodium chabaudi Infection. PLoS One 2015; 10:e0144654. [PMID: 26646149 PMCID: PMC4672895 DOI: 10.1371/journal.pone.0144654] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/20/2015] [Indexed: 11/19/2022] Open
Abstract
CD4 T cells are required to fight malaria infection by promoting both phagocytic activity and B cell responses for parasite clearance. In Plasmodium chabaudi infection, one specific CD4 T cell subset generates anti-parasitic IFN-γ and the antibody-promoting cytokine, IL-21. To determine the lineage of these multifunctional T cells, we followed IFN-γ+ effector T cells (Teff) into the memory phase using Ifng-reporter mice. While Ifng+ Teff expanded, the level of the Th1 lineage-determining transcription factor T-bet only peaked briefly. Ifng+ Teff also co-express ICOS, the B cell area homing molecule CXCR5, and other Tfh lineage-associated molecules including Bcl6, the transcription factor required for germinal center (GC) T follicular helper cells (Tfh) differentiation. Because Bcl6 and T-bet co-localize to the nucleus of Ifng+ Teff, we hypothesized that Bcl6 controls the Tfh-like phenotype of Ifng+ Teff cells in P. chabaudi infection. We first transferred Bcl6-deficient T cells into wildtype hosts. Bcl6-deficient T cells did not develop into GC Tfh, but they still generated CXCR5+IFN-γ+IL-21+IL-10+ Teff, suggesting that this predominant population is not of the Tfh-lineage. IL-10 deficient mice, which have increased IFN-γ and T-bet expression, demonstrated expansion of both IFN-γ+IL-21+CXCR5+ cells and IFN-γ+ GC Tfh cells, suggesting a Th1 lineage for the former. In the memory phase, all Ifng+ T cells produced IL-21, but only a small percentage of highly proliferative Ifng+ T cells maintained a T-bethi phenotype. In chronic malaria infection, serum IFN-γ correlates with increased protection, and our observation suggests Ifng+ T cells are maintained by cellular division. In summary, we found that Ifng+ T cells are not strictly Tfh derived during malaria infection. T cells provide the host with a survival advantage when facing this well-equipped pathogen, therefore, understanding the lineage of pivotal T cell players will aid in the rational design of an effective malaria vaccine.
Collapse
Affiliation(s)
- Victor H. Carpio
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael M. Opata
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marelle E. Montañez
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Pinaki P. Banerjee
- Center for Human Immunobiology of Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander L. Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
40
|
Lupar E, Brack M, Garnier L, Laffont S, Rauch KS, Schachtrup K, Arnold SJ, Guéry JC, Izcue A. Eomesodermin Expression in CD4+ T Cells Restricts Peripheral Foxp3 Induction. THE JOURNAL OF IMMUNOLOGY 2015; 195:4742-52. [PMID: 26453746 DOI: 10.4049/jimmunol.1501159] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
CD4(+) T cells polarize into effector Th subsets characterized by signature transcription factors and cytokines. Although T-bet drives Th1 responses and represses the alternative Th2, Th17, and Foxp3(+) regulatory T cell fates, the role of the T-bet-related transcription factor eomesodermin (Eomes) in CD4(+) T cells is less well understood. In this study, we analyze the expression and effects of Eomes in mouse CD4(+) T lymphocytes. We find that Eomes is readily expressed in activated CD4(+) Th1 T cells in vivo. Eomes(+) CD4(+) T cells accumulated in old mice, under lymphopenic conditions in a T cell transfer model of colitis, and upon oral Ag administration. However, despite its expression, genetic deletion of Eomes in CD4(+) T cells did not impact on IFN-γ production nor increase Th2 or Th17 responses. In contrast, Eomes deficiency favored the accumulation of Foxp3(+) cells in old mice, after in vivo differentiation of Eomes-deficient naive CD4(+) T cells, and in response to oral Ag in a cell-intrinsic way. Enforced Eomes expression during in vitro regulatory T cell induction also reduced Foxp3 transcription. Likewise, bystander Eomes-deficient CD4(+) T cells were more efficient at protecting from experimental autoimmune encephalitis compared with wild-type CD4(+) T cells. This enhanced capacity of Eomes-deficient CD4(+) T cells to inhibit EAE in trans was associated with an enhanced frequency of Foxp3(+) cells. Our data identify a novel role for Eomes in CD4(+) T cells and indicate that Eomes expression may act by limiting Foxp3 induction, which may contribute to the association of EOMES to susceptibility to multiple sclerosis.
Collapse
Affiliation(s)
- Ekaterina Lupar
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany
| | - Maria Brack
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany
| | - Laure Garnier
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Sophie Laffont
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Katharina S Rauch
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany
| | - Sebastian J Arnold
- University Medical Centre, Renal Department, Centre for Clinical Research, D-79106 Freiburg, Germany; BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, D-79104 Freiburg, Germany; and Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, D-79102 Freiburg, Germany
| | - Jean-Charles Guéry
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Ana Izcue
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany;
| |
Collapse
|
41
|
Abstract
IL-21 is a type I cytokine essential for immune cell differentiation and function. Although IL-21 can activate several STAT family transcription factors, previous studies focused mainly on the role of STAT3 in IL-21 signaling. Here, we investigated the role of STAT1 and show that STAT1 and STAT3 have at least partially opposing roles in IL-21 signaling in CD4(+) T cells. IL-21 induced STAT1 phosphorylation, and this was augmented in Stat3-deficient CD4(+) T cells. RNA-Seq analysis of CD4(+) T cells from Stat1- and Stat3-deficient mice revealed that both STAT1 and STAT3 are critical for IL-21-mediated gene regulation. Expression of some genes, including Tbx21 and Ifng, was differentially regulated by STAT1 and STAT3. Moreover, opposing actions of STAT1 and STAT3 on IFN-γ expression in CD4(+) T cells were demonstrated in vivo during chronic lymphocytic choriomeningitis infection. Finally, IL-21-mediated induction of STAT1 phosphorylation, as well as IFNG and TBX21 expression, were higher in CD4(+) T cells from patients with autosomal dominant hyper-IgE syndrome, which is caused by STAT3 deficiency, as well as in cells from STAT1 gain-of-function patients. These data indicate an interplay between STAT1 and STAT3 in fine-tuning IL-21 actions.
Collapse
|
42
|
Lin SJ, Lu CH, Yan DC, Lee PT, Hsiao HS, Kuo ML. Expansion of regulatory T cells from umbilical cord blood and adult peripheral blood CD4(+)CD25 (+) T cells. Immunol Res 2015; 60:105-11. [PMID: 24515612 DOI: 10.1007/s12026-014-8488-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4(+)CD25(+) regulatory T cells (Treg), if properly expanded from umbilical cord blood (UCB), may provide a promising immunotherapeutic tool. Our previous data demonstrated that UCB CD4(+)CD25(+) T cells with 4-day stimulation have comparable phenotypes and suppressive function to that of adult peripheral blood (APB) CD4(+)CD25(+) T cells. We further examined whether 2-week culture would achieve higher expansion levels of Tregs. UCB CD4(+)CD25(+) T cells and their APB counterparts were stimulated with anti-CD3/anti-CD28 in the presence of IL-2 or IL-15 for 2 weeks. The cell proliferation and forkhead box P3 (FoxP3) expression were examined. The function of the expanded cells was then investigated by suppressive assay. IL-21 was applied to study whether it counteracts the function of UCB and APB CD4(+)CD25(+) T cells. The results indicate that UCB CD4(+)CD25(+) T cells expanded much better than their APB counterparts. IL-2 was superior to expand UCB and APB Tregs for 2 weeks than IL-15. FoxP3 expression which peaked on Day 10-14 was comparable. Most importantly, expanded UCB Tregs showed greater suppressive function in allogeneic mixed lymphocyte reaction. The addition of IL-21, however, counteracted the suppressive function of expanded UCB and APB Tregs. The results support using UCB as a source of Treg cells.
Collapse
Affiliation(s)
- Syh-Jae Lin
- Division of Asthma, Allergy, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
43
|
Lin PY, Jen HY, Chiang BL, Sheu F, Chuang YH. Interleukin-21 suppresses the differentiation and functions of T helper 2 cells. Immunology 2015; 144:668-76. [PMID: 25351608 DOI: 10.1111/imm.12419] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/03/2014] [Accepted: 10/24/2014] [Indexed: 12/19/2022] Open
Abstract
T helper type 2 (Th2) cells, which produce interleukin-4 (IL-4), IL-5 and IL-13, control immunity to all forms of allergic inflammatory responses. Interleukin-21 (IL-21) reduces allergic symptoms in murine models and inhibits IL-4-induced IgE secretion by B cells. However, whether or not IL-21 directly affects Th2 cells, which leads to reduced allergic symptoms, is unclear. In this study, we investigated the effects of IL-21 on the differentiation and effector functions of Th2 cells. We found that IL-21 reduced the number of differentiated Th2 cells and these Th2 cells showed a diminished Th2 cytokine production. Interleukin-21 suppressed Th2 cytokine production of already polarized Th2 cells by down-regulation of transcription factor GATA-3. It also induced apoptosis of Th2 cells with decreased anti-apoptotic factor Bcl-2. Intranasal administration of IL-21 at the beginning of ovalbumin (OVA) sensitization or before OVA challenge decreased Th2 cytokines in the bronchoalveolar lavage fluid of OVA/alum-immunized allergic mice. In addition, the inhibitory effects of IL-21 on Th2 effector functions can also be found in allergic patients. Our results demonstrate that IL-21 suppresses the development of Th2 cells and functions of polarized Th2 cells. Hence, the administration of IL-21 may be considered for use as a preventive and therapeutic approach when dealing with Th2-mediated allergic diseases.
Collapse
Affiliation(s)
- Pin-Yi Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
44
|
Fu W, Cheng Y, Zhang Y, Mo X, Li T, Liu Y, Wang P, Pan W, Chen Y, Xue Y, Ma D, Zhang Y, Han W. The Secreted Form of Transmembrane Protein 98 Promotes the Differentiation of T Helper 1 Cells. J Interferon Cytokine Res 2015; 35:720-33. [PMID: 25946230 DOI: 10.1089/jir.2014.0110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cytokines mediate the interaction of immune cells. Discovery of novel potential cytokines is of great value for both basic research and clinical application. In this study, we identified a novel immune-related molecule, transmembrane protein 98 (TMEM98), through a high-throughput screening platform for novel potential cytokines at a genome-wide level using the strategy of immunogenomics. So far, there is no characteristic and immune-related functional report about it. In this study, we demonstrate that TMEM98 exists as a type II transmembrane protein both in the ectopically and endogenously expressed systems. Interestingly, TMEM98 could also be secreted through exosomes. Moreover, the native secreted form of TMEM98 could be detected in the supernatants of activated human peripheral blood mononuclear cells and mouse CD4(+) T cells. Further expression profile analysis showed TMEM98 was upregulated during the activation and differentiation of T helper (Th) 1 cells. Function analysis showed that eukaryotic recombinant TMEM98 (rTMEM98) promoted the differentiation of Th1 cells under both antigen-nonspecific and antigen-specific Th1-skewing conditions. These findings were further confirmed in vivo as prokaryotic rTMEM98 administration significantly increased antigen-specific IFN-γ production and serum antigen-specific IgG2a in the methylated bovine serum albumin-induced delayed-type hypersensitivity model. Overall, these observations emphasize the characteristics and essential roles of TMEM98 for the first time and will be helpful in further understanding the development of Th1 cells.
Collapse
Affiliation(s)
- Weiwei Fu
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
- 3 Tsinghua University School of Medicine , Beijing, China
| | - Yingying Cheng
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Yanfei Zhang
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Xiaoning Mo
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Ting Li
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Yuanfeng Liu
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
| | - Pingzhang Wang
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Wen Pan
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Yingyu Chen
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Yintong Xue
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
| | - Dalong Ma
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| | - Yu Zhang
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
| | - Wenling Han
- 1 Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing, China
- 2 Peking University Center for Human Disease Genomics , Beijing, China
| |
Collapse
|
45
|
Global characterization of differential gene expression profiles in mouse Vγ1+ and Vγ4+ γδ T cells. PLoS One 2014; 9:e112964. [PMID: 25405356 PMCID: PMC4236085 DOI: 10.1371/journal.pone.0112964] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 10/16/2014] [Indexed: 11/19/2022] Open
Abstract
Peripheral γδ T cells in mice are classified into two major subpopulations, Vγ1+ and Vγ4+, based on the composition of T cell receptors. However, their intrinsic differences remain unclear. In this study, we analyzed gene expression profiles of the two subsets using Illumina HiSeq 2000 Sequencer. We identified 1995 transcripts related to the activation of Vγ1+ γδ T cells, and 2158 transcripts related to the activation of Vγ4+ γδ T cells. We identified 24 transcripts differentially expressed between the two subsets in resting condition, and 20 after PMA/Ionomycin treatment. We found that both cell types maintained phenotypes producing IFN-γ, TNF-α, TGF-β and IL-10. However, Vγ1+ γδ T cells produced more Th2 type cytokines, such as IL-4 and IL-5, while Vγ4+ γδ T cells preferentially produced IL-17. Our study provides a comprehensive gene expression profile of mouse peripheral Vγ1+ and Vγ4+ γδ T cells that describes the inherent differences between them.
Collapse
|
46
|
Ono C, Yu Z, Kasahara Y, Kikuchi Y, Ishii N, Tomita H. Fluorescently activated cell sorting followed by microarray profiling of helper T cell subtypes from human peripheral blood. PLoS One 2014; 9:e111405. [PMID: 25379667 PMCID: PMC4224392 DOI: 10.1371/journal.pone.0111405] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 10/02/2014] [Indexed: 12/31/2022] Open
Abstract
Background Peripheral blood samples have been subjected to comprehensive gene expression profiling to identify biomarkers for a wide range of diseases. However, blood samples include red blood cells, white blood cells, and platelets. White blood cells comprise polymorphonuclear leukocytes, monocytes, and various types of lymphocytes. Blood is not distinguishable, irrespective of whether the expression profiles reflect alterations in (a) gene expression patterns in each cell type or (b) the proportion of cell types in blood. CD4+ Th cells are classified into two functionally distinct subclasses, namely Th1 and Th2 cells, on the basis of the unique characteristics of their secreted cytokines and their roles in the immune system. Th1 and Th2 cells play an important role not only in the pathogenesis of human inflammatory, allergic, and autoimmune diseases, but also in diseases that are not considered to be immune or inflammatory disorders. However, analyses of minor cellular components such as CD4+ cell subpopulations have not been performed, partly because of the limited number of these cells in collected samples. Methodology/Principal Findings We describe fluorescently activated cell sorting followed by microarray (FACS–array) technology as a useful experimental strategy for characterizing the expression profiles of specific immune cells in the circulation. We performed reproducible gene expression profiling of Th1 and Th2, respectively. Our data suggest that this procedure provides reliable information on the gene expression profiles of certain small immune cell populations. Moreover, our data suggest that GZMK, GZMH, EOMES, IGFBP3, and STOM may be novel markers for distinguishing Th1 cells from Th2 cells, whereas IL17RB and CNTNAP1 can be Th2-specific markers. Conclusions/Significance Our approach may help in identifying aberrations and novel therapeutic or diagnostic targets for diseases that affect Th1 or Th2 responses and elucidating the involvement of a subpopulation of immune cells in some diseases.
Collapse
Affiliation(s)
- Chiaki Ono
- Department of Disaster Psychiatry, Internal Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Zhiqian Yu
- Department of Disaster Psychiatry, Internal Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshiyuki Kasahara
- Department of Disaster Psychiatry, Internal Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshie Kikuchi
- Department of Disaster Psychiatry, Internal Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Disaster Psychiatry, Internal Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- * E-mail:
| |
Collapse
|
47
|
Lorvik KB, Haabeth OAW, Clancy T, Bogen B, Corthay A. Molecular profiling of tumor-specific T H1 cells activated in vivo. Oncoimmunology 2014; 2:e24383. [PMID: 23762808 PMCID: PMC3667914 DOI: 10.4161/onci.24383] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/21/2022] Open
Abstract
The central role of tumor-specific TH1 cells in anticancer immune responses is becoming increasingly appreciated. However, little is known about how these cells are generated in vivo. Here, we used flow cytometry and gene expression microarrays to characterize the primary activation and TH1 differentiation of naïve tumor-specific CD4+ T cells in a mouse model of cancer immunosurveillance. We took advantage of T-cell receptor-transgenic mice in which CD4+ T cells recognize a tumor-specific antigen secreted by MHC class II-negative MOPC315 myeloma cells. Cancer cells were injected subcutaneously and T-cell activation was analyzed in draining lymph nodes and at the incipient tumor site 8 d later. Upon activation and migration to incipient tumor sites, tumor-specific CD4+ T cells exhibited the upregulation of 29 cell-surface molecules (CD2, CD5, CD11a, CD18, CD25, CD28, CD44, CD45, CD49d, CD51, CD54, CD69, CD71, CD83, CD86, CD90, CD95, CD102, CD122, CD153, CD166, CD200, CD249, CD254, CD274, CD279, Ly6C, MHC class I and CCR7) and the downregulation of five (CD27, CD31, CD45RB, CD62L and CD126). Activated CD4+ T cells produced interferon γ, a cytokine consistent with a TH1-polarized response, tumor necrosis factor α as well as interleukin (IL)-2, IL-3 and IL-10. The activation of naïve tumor-specific CD4+ T cells in draining lymph nodes resulted in the upregulation of 609 genes and the downregulation of 284 genes. The bioinformatic analysis of differentially expressed genes identified functional pathways related to tumor-specific TH1 cell activation. This study may represent a useful resource to guide the development of TH1-based immunotherapies against cancer.
Collapse
Affiliation(s)
- Kristina Berg Lorvik
- Centre for Immune Regulation; Department of Immunology; Oslo University Hospital Rikshospitalet and University of Oslo; Oslo, Norway
| | | | | | | | | |
Collapse
|
48
|
Kastirr I, Maglie S, Paroni M, Alfen JS, Nizzoli G, Sugliano E, Crosti MC, Moro M, Steckel B, Steinfelder S, Stölzel K, Romagnani C, Botti F, Caprioli F, Pagani M, Abrignani S, Geginat J. IL-21 Is a Central Memory T Cell–Associated Cytokine That Inhibits the Generation of Pathogenic Th1/17 Effector Cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:3322-31. [DOI: 10.4049/jimmunol.1400775] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
49
|
Pick J, Arra A, Lingel H, Hegel JK, Huber M, Nishanth G, Jorch G, Fischer KD, Schlüter D, Tedford K, Brunner-Weinzierl MC. CTLA-4 (CD152) enhances the Tc17 differentiation program. Eur J Immunol 2014; 44:2139-52. [PMID: 24723371 DOI: 10.1002/eji.201343497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 02/21/2014] [Accepted: 03/28/2014] [Indexed: 01/24/2023]
Abstract
Although CD8(+) T cells that produce IL-17 (Tc17 cells) have been linked to host defense, Tc17 cells show reduced cytotoxic activity, which is the characteristic function of CD8(+) T cells. Here, we show that CTLA-4 enhances the frequency of IL-17 in CD8(+) T cells, indicating that CTLA-4 (CD152) specifically promotes Tc17 differentiation. Simultaneous stimulation of CTLA-4(+/+) and CTLA-4(-/-) T cells in cocultures and agonistic CTLA-4 stimulation unambiguously revealed a cell-intrinsic mechanism for IL-17 control by CTLA-4. The quality of CTLA-4-induced Tc17 cells was tested in vivo, utilizing infection with the facultative intracellular bacterium Listeria monocytogenes (LM). Unlike CTLA-4(+/+) Tc17 cells, CTLA-4(-/-) were nearly as efficient as Tc1 CTLA-4(+/+) cells in LM clearance. Additionally, adoptively transferred CTLA-4(-/-) Tc17 cells expressed granzyme B after rechallenge, and produced Tc1 cytokines such as IFN-γ and TNF-α, which strongly correlate with bacterial clearance. CTLA-4(+/+) Tc17 cells demonstrated a high-quality Tc17 differentiation program ex vivo, which was also evident in isolated IL-17-secreting Tc17 cells, with CTLA-4-mediated enhanced upregulation of Tc17-related molecules such as IL-17A, RORγt, and IRF-4. Our results show that CTLA-4 promotes Tc17 differentiation that results in robust Tc17 responses. Its inactivation might therefore represent a central therapeutic target to enhance clearance of infection.
Collapse
Affiliation(s)
- Jonas Pick
- Department of Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tumes DJ, Onodera A, Suzuki A, Shinoda K, Endo Y, Iwamura C, Hosokawa H, Koseki H, Tokoyoda K, Suzuki Y, Motohashi S, Nakayama T. The polycomb protein Ezh2 regulates differentiation and plasticity of CD4(+) T helper type 1 and type 2 cells. Immunity 2014; 39:819-32. [PMID: 24238339 DOI: 10.1016/j.immuni.2013.09.012] [Citation(s) in RCA: 239] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 09/16/2013] [Indexed: 12/23/2022]
Abstract
After antigen encounter by CD4(+) T cells, polarizing cytokines induce the expression of master regulators that control differentiation. Inactivation of the histone methyltransferase Ezh2 was found to specifically enhance T helper 1 (Th1) and Th2 cell differentiation and plasticity. Ezh2 directly bound and facilitated correct expression of Tbx21 and Gata3 in differentiating Th1 and Th2 cells, accompanied by substantial trimethylation at lysine 27 of histone 3 (H3K27me3). In addition, Ezh2 deficiency resulted in spontaneous generation of discrete IFN-γ and Th2 cytokine-producing populations in nonpolarizing cultures, and under these conditions IFN-γ expression was largely dependent on enhanced expression of the transcription factor Eomesodermin. In vivo, loss of Ezh2 caused increased pathology in a model of allergic asthma and resulted in progressive accumulation of memory phenotype Th2 cells. This study establishes a functional link between Ezh2 and transcriptional regulation of lineage-specifying genes in terminally differentiated CD4(+) T cells.
Collapse
Affiliation(s)
- Damon J Tumes
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|