1
|
Ni G, Liu X, Li H, Fogarty CE, Chen S, Zhang P, Liu Y, Wu X, Wei MQ, Chen G, Zhang P, Wang T. Topical Application of Temperature-Sensitive Gel Containing Caerin 1.1 and 1.9 Peptides on TC-1 Tumour-Bearing Mice Induced High-Level Immune Response in the Tumour Microenvironment. Front Oncol 2021; 11:754770. [PMID: 34858827 PMCID: PMC8632150 DOI: 10.3389/fonc.2021.754770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2021] [Indexed: 01/22/2023] Open
Abstract
The development of topical cream drugs that increase the immune activation of tumour-infiltrating lymphocytes against tumour and chronic viral infection-associated lesions is of great immunotherapeutic significance. This study demonstrates that the topical application of a temperature-sensitive gel containing caerin 1.1 and 1.9 peptides reduces nearly 50% of the tumour weight of HPV16 E6/E7-transformed TC-1 tumour-bearing mice via improving the tumour microenvironment. Confocal microscopy confirms the time-dependent penetration of caerin 1.9 through the epidermal layer of the ear skin structure of mice. Single-cell transcriptomic analysis shows that the caerin 1.1/1.9 gel expands the populations with high immune activation level and largely stimulates the pro-inflammatory activity of NK and dendritic cells. Closely associated with INFα response, Cebpb seems to play a key role in altering the function of all Arg1hi macrophages in the caerin group. In addition, the caerin gel treatment recruits almost two-fold more activated CD8+ T cells to the TME, relative to the untreated tumour, which shows a synergistic effect derived from the regulation of S1pr1, Ccr7, Ms4a4b and Gimap family expression. The TMT10plex-labelling proteomic quantification further demonstrates the activation of interferon-alpha/beta secretion and response to cytokine stimulus by the caerin gel, while the protein contents of several key regulators were elevated by more than 30%, such as Cd5l, Gzma, Ifit1, Irf9 and Stat1. Computational integration of the proteome with the single-cell transcriptome consistently suggested greater activation of NK and T cells with the topical application of caerin peptide gel.
Collapse
Affiliation(s)
- Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China.,Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Xiaosong Liu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China.,Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Hejie Li
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Conor E Fogarty
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Shu Chen
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Pingping Zhang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ying Liu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Xiaolian Wu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ming Q Wei
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Guoqiang Chen
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ping Zhang
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Tianfang Wang
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| |
Collapse
|
2
|
Qin R, Zhao C, Wang CJ, Xu W, Zhao JY, Lin Y, Yuan YY, Lin PC, Li Y, Zhao S, Huang Y. Tryptophan potentiates CD8 + T cells against cancer cells by TRIP12 tryptophanylation and surface PD-1 downregulation. J Immunother Cancer 2021; 9:jitc-2021-002840. [PMID: 34326168 PMCID: PMC8323461 DOI: 10.1136/jitc-2021-002840] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Tryptophan catabolites suppress immunity. Therefore, blocking tryptophan catabolism with indoleamine 2,3-dioxygenase (IDO) inhibitors is pursued as an anticancer strategy. METHODS The intracellular level of tryptophan and kynurenine was detected by mass spectrum analysis. The effect of tryptophan and IDO inhibitors on cell surface programmed cell death protein 1 (PD-1) level were measured by flow cytometry. A set of biochemical analyses were used to figure out the underlying mechanism. In vitro co-culture system, syngeneic mouse models, immunofluorescent staining, and flow cytometry analysis were employed to investigate the role of tryptophan and IDO inhibitor in regulating the cytotoxicity of CD8+ T cells. RESULTS Here, we reported that IDO inhibitors activated CD8+ T cells also by accumulating tryptophan that downregulated PD-1. Tryptophan and IDO inhibitors administration, both increased intracellular tryptophan, and tryptophanyl-tRNA synthetase (WARS) overexpression decreased Jurkat and mice CD8+ T cell surface PD-1. Mechanistically, WARS tryptophanylated lysine 1136 of and activated E3 ligase TRIP12 to degrade NFATc1, a PD-1 transcription activator. SIRT1 de-tryptophanylated TRIP12 and reversed the effects of tryptophan and WARS on PD-1. Tryptophan or IDO inhibitors potentiated CD8+ T cells to induce apoptosis of co-cultured cancer cells, increased cancer-infiltrating CD8+ T cells and slowed down tumor growth of lung cancer in mice. CONCLUSIONS Our results revealed the immune-activating efficacy of tryptophan, and suggested tryptophan supplemental may benefit IDO inhibitors and PD-1 blockade during anticancer treatments.
Collapse
Affiliation(s)
- Rui Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Chen Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Chen-Ji Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Wei Xu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Metabolic Remodeling, Institute of Metabolism and Integrative Biology and Institutes of Biomedical Sciences, Shanghai, China.,Department of Cardiology, Children's Hospital of Fudan University, Shanghai, China
| | - Jian-Yuan Zhao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Metabolic Remodeling, Institute of Metabolism and Integrative Biology and Institutes of Biomedical Sciences, Shanghai, China.,Department of Cardiology, Children's Hospital of Fudan University, Shanghai, China
| | - Yan Lin
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Metabolic Remodeling, Institute of Metabolism and Integrative Biology and Institutes of Biomedical Sciences, Shanghai, China.,Department of Cardiology, Children's Hospital of Fudan University, Shanghai, China
| | - Yi-Yuan Yuan
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Metabolic Remodeling, Institute of Metabolism and Integrative Biology and Institutes of Biomedical Sciences, Shanghai, China.,Department of Cardiology, Children's Hospital of Fudan University, Shanghai, China
| | - Peng-Cheng Lin
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining, China
| | - Yao Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Shimin Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China .,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Metabolic Remodeling, Institute of Metabolism and Integrative Biology and Institutes of Biomedical Sciences, Shanghai, China
| | - Yan Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
3
|
Mastio J, Saeed MB, Wurzer H, Krecke M, Westerberg LS, Thomas C. Higher Incidence of B Cell Malignancies in Primary Immunodeficiencies: A Combination of Intrinsic Genomic Instability and Exocytosis Defects at the Immunological Synapse. Front Immunol 2020; 11:581119. [PMID: 33240268 PMCID: PMC7680899 DOI: 10.3389/fimmu.2020.581119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.
Collapse
Affiliation(s)
- Jérôme Mastio
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Mezida B Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Wurzer
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Max Krecke
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Clément Thomas
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| |
Collapse
|
4
|
LRCH1 deficiency enhances LAT signalosome formation and CD8 + T cell responses against tumors and pathogens. Proc Natl Acad Sci U S A 2020; 117:19388-19398. [PMID: 32727906 DOI: 10.1073/pnas.2000970117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CD8+ T cells play pivotal roles in eradicating pathogens and tumor cells. T cell receptor (TCR) signaling is vital for the optimal activation of CD8+ T cells. Upon TCR engagement, the transmembrane adapter protein LAT (linker for activation of T cells) recruits other key signaling molecules and forms the "LAT signalosome" for downstream signal transduction. However, little is known about which functional partners could restrain the formation of the LAT signalosome and inhibit CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity. Here we have demonstrated that LRCH1 (leucine-rich repeats and calponin homology domain containing 1) directly binds LAT, reduces LAT phosphorylation and interaction with GRB2, and also promotes the endocytosis of LAT. Lrch1 -/- mice display better protection against influenza virus and Listeria infection, with enhanced CD8+ T cell proliferation and cytotoxicity. Adoptive transfer of Lrch1 -/- CD8+ CTLs leads to increased B16-MO5 tumor clearance in vivo. Furthermore, knockout of LRCH1 in human chimeric antigen receptor (CAR) T cells that recognize the liver tumor-associated antigen glypican-3 could improve CAR T cell migration and proliferation in vitro. These findings suggest LRCH1 as a potential translational target to improve T cell immunotherapy against infection and tumors.
Collapse
|
5
|
Various Stages of Immune Synapse Formation Are Differently Dependent on the Strength of the TCR Stimulus. Int J Mol Sci 2020; 21:ijms21072475. [PMID: 32252488 PMCID: PMC7177831 DOI: 10.3390/ijms21072475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) are key players of the adaptive immune system that target tumors and infected cells. A central step to that is the formation of a cell–cell contact zone between the CTL and its target called an immune synapse (IS). Here, we investigate the influence of the initial T cell receptor (TCR) trigger of a cytolytic IS on the distinct steps leading to cytotoxic granule (CG) exocytosis. We stimulated primary CTLs from mouse using lipid bilayers with varying anti-CD3 but constant ICAM concentrations. We fluorescently labeled molecular markers of distinct IS zones such as actin, CD3, granzyme B, and Synaptobrevin2 in CTLs and imaged cytolytic IS formation by total internal reflection fluorescence microscopy (TIRFM). We found that an intermediate anti-CD3 concentration of 10 µg/mL induces the fastest adhesion of CTLs to the bilayers and results in maximal CG fusion efficiency. The latency of actin ring formation, dwell time, and maximum surface area at the IS exhibit different dependencies on the stimulatory anti-CD3 concentrations. The number and surface area of CD3 clusters at the IS seem to show a different dependency to the TCR trigger when compared to their dwell time. Finally, the mode of full CG exocytosis appears to be independent of the TCR trigger.
Collapse
|
6
|
Li X, Liu Z, Mi M, Zhang C, Xiao Y, Liu X, Wu G, Zhang L. Identification of hub genes and key pathways associated with angioimmunoblastic T-cell lymphoma using weighted gene co-expression network analysis. Cancer Manag Res 2019; 11:5209-5220. [PMID: 31239775 PMCID: PMC6559227 DOI: 10.2147/cmar.s185030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 05/11/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive subtype of peripheral T-cell lymphoma (PTCL) that has a poor 5-year overall survival rate due to its lack of precise therapeutic targets. Identifying potential prognostic markers of AITL may provide information regarding the development of precision medicine. Methods: RNA sequence data from PTCL and patient clinic traits were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed gene (DEG) analysis and weighted gene co-expression network analysis (WGCNA) were performed to identify DEGs between the different PTCL subtypes and investigate the relationship underlying co-expression modules and clinic traits. Gene ontology (GO) and protein-protein interaction (PPI) network analyses based on DAVID and the STRING website, respectively, were utilized to deeply excavate hub genes. Results: After removing the outliers from the GSE65823, GSE58445, GSE19069, and GSE6338 datasets using the results from an unsupervised cluster heatmap, 50 AITL samples and 55 anaplastic large cell lymphoma (ALCL) samples were screened. A total of 677 upregulated DEGs and 237 downregulated DEGs were identified in AITL and used to construct a PPI network complex. Using WGCNA, 12 identified co-expression modules were constructed from the 5468 genes with the top 10% of variance, and 192 genes from the Turquoise and Brown modules were with a Gene Significance (GS) cut-off threshold >0.6. Eleven hub genes (CDH1, LAT, LPAR1, CXCL13, CD27, ICAM2, CD3E, CCL19, CTLA-4, CXCR5, and C3) were identified. Only CTLA-4 overexpressed was found to be a poor prognostic factor according to survival analysis. Gene set enrichment analysis (GSEA) identified and validated the intersection of key pathways (T cell receptor, primary immunodeficiency, and chemokine signaling pathways). Conclusion: Our findings provide the framework for the identification of AITL co-expression gene modules and identify key pathways and driving genes that may be novel treatment targets and helpful for the development of a prognostic evaluation index.
Collapse
Affiliation(s)
- Xiaoqian Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zijian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mi Mi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Caijiao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinxiu Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Liling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
7
|
Kabanova A, Zurli V, Baldari CT. Signals Controlling Lytic Granule Polarization at the Cytotoxic Immune Synapse. Front Immunol 2018. [PMID: 29515593 PMCID: PMC5826174 DOI: 10.3389/fimmu.2018.00307] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytotoxic immunity relies on specialized effector T cells, the cytotoxic T cells, which are endowed with specialized cytolytic machinery that permits them to induce death of their targets. Upon recognition of a target cell, cytotoxic T cells form a lytic immune synapse and by docking the microtubule-organizing center at the synaptic membrane get prepared to deliver a lethal hit of enzymes contained in lytic granules. New insights suggest that the directionality of lytic granule trafficking along the microtubules represents a fine means to tune the functional outcome of the encounter between a T cell and its target. Thus, mechanisms regulating the directionality of granule transport may have a major impact in settings characterized by evasion from the cytotoxic response, such as chronic infection and cancer. Here, we review our current knowledge on the signaling pathways implicated in the polarized trafficking at the immune synapse of cytotoxic T cells, complementing it with information on the regulation of this process in natural killer cells. Furthermore, we highlight some of the parameters which we consider critical in studying the polarized trafficking of lytic granules, including the use of freshly isolated cytotoxic T cells, and discuss some of the major open questions.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Vanessa Zurli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
8
|
Myers DR, Lau T, Markegard E, Lim HW, Kasler H, Zhu M, Barczak A, Huizar JP, Zikherman J, Erle DJ, Zhang W, Verdin E, Roose JP. Tonic LAT-HDAC7 Signals Sustain Nur77 and Irf4 Expression to Tune Naive CD4 T Cells. Cell Rep 2017; 19:1558-1571. [PMID: 28538176 PMCID: PMC5587137 DOI: 10.1016/j.celrep.2017.04.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 04/05/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022] Open
Abstract
CD4+ T cells differentiate into T helper cell subsets in feedforward manners with synergistic signals from the T cell receptor (TCR), cytokines, and lineage-specific transcription factors. Naive CD4+ T cells avoid spontaneous engagement of feedforward mechanisms but retain a prepared state. T cells lacking the adaptor molecule LAT demonstrate impaired TCR-induced signals yet cause a spontaneous lymphoproliferative T helper 2 (TH2) cell syndrome in mice. Thus, LAT constitutes an unexplained maintenance cue. Here, we demonstrate that tonic signals through LAT constitutively export the repressor HDAC7 from the nucleus of CD4+ T cells. Without such tonic signals, HDAC7 target genes Nur77 and Irf4 are repressed. We reveal that Nur77 suppresses CD4+ T cell proliferation and uncover a suppressive role for Irf4 in TH2 polarization; halving Irf4 gene-dosage leads to increases in GATA3+ and IL-4+ cells. Our studies reveal that naive CD4+ T cells are dynamically tuned by tonic LAT-HDAC7 signals.
Collapse
Affiliation(s)
- Darienne R Myers
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tannia Lau
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Evan Markegard
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hyung W Lim
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Herbert Kasler
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Minghua Zhu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrea Barczak
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John P Huizar
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David J Erle
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Weiguo Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Eric Verdin
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
9
|
SCIMP is a transmembrane non-TIR TLR adaptor that promotes proinflammatory cytokine production from macrophages. Nat Commun 2017; 8:14133. [PMID: 28098138 PMCID: PMC5253658 DOI: 10.1038/ncomms14133] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/02/2016] [Indexed: 01/04/2023] Open
Abstract
Danger signals activate Toll-like receptors (TLRs), thereby initiating inflammatory responses. Canonical TLR signalling, via Toll/Interleukin-1 receptor domain (TIR)-containing adaptors and proinflammatory transcription factors such as NF-κB, occurs in many cell types; however, additional mechanisms are required for specificity of inflammatory responses in innate immune cells. Here we show that SCIMP, an immune-restricted, transmembrane adaptor protein (TRAP), promotes selective proinflammatory cytokine responses by direct modulation of TLR4. SCIMP is a non-TIR-containing adaptor, binding directly to the TLR4-TIR domain in response to lipopolysaccharide. In macrophages, SCIMP is constitutively associated with the Lyn tyrosine kinase, is required for tyrosine phosphorylation of TLR4, and facilitates TLR-inducible production of the proinflammatory cytokines IL-6 and IL-12p40. Point mutations in SCIMP abrogating TLR4 binding also prevent SCIMP-mediated cytokine production. SCIMP is, therefore, an immune-specific TLR adaptor that shapes host defence and inflammation. Toll-like receptors engage TIR domain-containing adaptors to control proinflammatory gene expression in response to pathogens and tissue damage. Here the authors show that the non-TIR domain-containing transmembrane protein SCIMP is a previously unrecognized TLR adaptor expressed by macrophages.
Collapse
|
10
|
Danzaki K, Kanayama M, Alcazar O, Shinohara ML. Osteopontin has a protective role in prostate tumor development in mice. Eur J Immunol 2016; 46:2669-2678. [PMID: 27601131 DOI: 10.1002/eji.201646391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/12/2016] [Accepted: 09/02/2016] [Indexed: 12/25/2022]
Abstract
Osteopontin (OPN) is a protein, generally considered to play a pro-tumorigenic role, whereas several reports have demonstrated the anti-tumorigenic function of OPN during tumor development. These opposing anti- and pro-tumorigenic functions are not fully understood. Here, we report that host-derived OPN plays an anti-tumorigenic role in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model and a TRAMP tumor transplant model. Tumor suppression mediated by OPN in Rag2-/- mice suggests that OPN is dispensable in the adaptive immune response. We found that host-derived OPN enhanced infiltration of natural killer (NK) cells into TRAMP tumors. The requirement of OPN in NK cell migration towards TRAMP cells was confirmed by an ex vivo cell migration assay. In contrast to TRAMP cells, in vivo B16 tumor development was not inhibited by OPN, and B16 tumors did not show OPN-mediated cell recruitment. It is possible that low levels of chemokine expression by B16 cells do not allow OPN to enhance immune cell recruitment. In addition to demonstrating the anti-tumorigenic role of OPN in TRAMP tumor development, this study also suggests that the contribution of OPN to tumor development depends on the type of tumor as well as the source and isoform of OPN.
Collapse
Affiliation(s)
- Keiko Danzaki
- Department of Immunology, Duke University Medical School, Durham, NC, 27710, USA
| | - Masashi Kanayama
- Department of Immunology, Duke University Medical School, Durham, NC, 27710, USA
| | - Oscar Alcazar
- Department of Immunology, Duke University Medical School, Durham, NC, 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical School, Durham, NC, 27710, USA. .,Department of Molecular Genetics and Microbiology, Duke University Medical School, Durham, NC, 27710, USA.
| |
Collapse
|
11
|
Sand K, Theorell J, Bruserud Ø, Bryceson YT, Kittang AO. Reduced potency of cytotoxic T lymphocytes from patients with high-risk myelodysplastic syndromes. Cancer Immunol Immunother 2016; 65:1135-47. [PMID: 27481108 PMCID: PMC11029614 DOI: 10.1007/s00262-016-1865-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 07/01/2016] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Myelodysplastic syndromes (MDS) are a group of clonal bone marrow disorders, with dysplasia, cytopenias and increased risk of progression to acute myeloid leukemia. A dysregulated immune system precipitates MDS, and to gain insights into the relevance of cytotoxic T lymphocyte (CTL) in this process, we examined the frequency and function of CX3CR1- and CD57-positive T lymphocytes from MDS patients. MATERIALS AND METHODS Peripheral blood and/or bone marrow samples from 31 MDS patients and 12 healthy controls were examined by flow cytometry. Expression of cytotoxic granule constituents, immunological co-receptors, adhesion molecules and markers of activation were quantified on unstimulated lymphocytes. Degranulation, cytotoxicity and conjugate formation with target cells following co-culture of CTL with target cell lines or autologous bone marrow-derived CD34(+) cells were quantified by flow cytometry. RESULTS CX3CR1 expression was increased in bone marrow from high-risk MDS patients compared to healthy controls. Expression of CD57 and CX3CR1 was closely correlated, identifying a CTL subset with high cytotoxic capacity. In vitro, TCR-induced redirected cytotoxicity was markedly decreased for high-risk MDS patients compared to controls. CTL from MDS patients with the lowest target cell cytotoxicity had reduced expression of adhesion molecules and formed fewer conjugates with target cells. DISCUSSION Although phenotypically defined CTL numbers were increased in the bone marrow of MDS patients, we found that CTL from high-risk MDS patients exhibited a lower TCR-induced redirected cytotoxic capacity. Thus, decreased T cell cytotoxicity seems related to reduced adhesion to target cells and may contribute to impaired anti-leukemic immune surveillance in MDS.
Collapse
Affiliation(s)
- Kristoffer Sand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jakob Theorell
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Yenan T Bryceson
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Astrid Olsnes Kittang
- Department of Clinical Science, University of Bergen, Bergen, Norway.
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway.
| |
Collapse
|
12
|
Keller B, Zaidman I, Yousefi OS, Hershkovitz D, Stein J, Unger S, Schachtrup K, Sigvardsson M, Kuperman AA, Shaag A, Schamel WW, Elpeleg O, Warnatz K, Stepensky P. Early onset combined immunodeficiency and autoimmunity in patients with loss-of-function mutation in LAT. J Exp Med 2016; 213:1185-99. [PMID: 27242165 PMCID: PMC4925012 DOI: 10.1084/jem.20151110] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 05/04/2016] [Indexed: 12/13/2022] Open
Abstract
Keller et al. describe for the first time human LAT deficiency, which causes severe immune dysregulation with autoimmunity, lymphoproliferation, and progressive immunodeficiency. The adapter protein linker for activation of T cells (LAT) is a critical signaling hub connecting T cell antigen receptor triggering to downstream T cell responses. In this study, we describe the first kindred with defective LAT signaling caused by a homozygous mutation in exon 5, leading to a premature stop codon deleting most of the cytoplasmic tail of LAT, including the critical tyrosine residues for signal propagation. The three patients presented from early childhood with combined immunodeficiency and severe autoimmune disease. Unlike in the mouse counterpart, reduced numbers of T cells were present in the patients. Despite the reported nonredundant role of LAT in Ca2+ mobilization, residual T cells were able to induce Ca2+ influx and nuclear factor (NF) κB signaling, whereas extracellular signal-regulated kinase (ERK) signaling was completely abolished. This is the first report of a LAT-related disease in humans, manifesting by a progressive combined immune deficiency with severe autoimmune disease.
Collapse
Affiliation(s)
- Baerbel Keller
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany
| | - Irina Zaidman
- Department of Pediatric Hematology Oncology, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa 3109601, Israel
| | - O Sascha Yousefi
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Dov Hershkovitz
- Department of Pathology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Jerry Stein
- Department of Pediatric Hematology Oncology and Bone Marrow Transplantation Unit, Schneider Children's Medical Center of Israel, Petah-Tikva 49202, Israel
| | - Susanne Unger
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany
| | - Mikael Sigvardsson
- Department of Clinical and Experimental Medicine, Experimental Hematopoiesis Unit, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Amir A Kuperman
- Blood Coagulation Service and Pediatric Hematology Clinic, Galilee Medical Center, Nahariya 22100, Israel Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 5290002, Israel
| | - Avraham Shaag
- Monique and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel
| | - Wolfgang W Schamel
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency (CCI), University Medical Center and University of Freiburg, 79106 Freiburg, Germany
| | - Polina Stepensky
- Monique and Jacques Roboh Department of Genetic Research, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel Department of Pediatric Hematology-Oncology and Bone Marrow Transplantation, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel
| |
Collapse
|
13
|
Zhang B, Wu J, Jiao Y, Bock C, Dai M, Chen B, Chao N, Zhang W, Zhuang Y. Differential Requirements of TCR Signaling in Homeostatic Maintenance and Function of Dendritic Epidermal T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:4282-91. [PMID: 26408667 DOI: 10.4049/jimmunol.1501220] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/01/2015] [Indexed: 01/17/2023]
Abstract
Dendritic epidermal T cells (DETCs) are generated exclusively in the fetal thymus and maintained in the skin epithelium throughout postnatal life of the mouse. DETCs have restricted antigenic specificity as a result of their exclusive usage of a canonical TCR. Although the importance of the TCR in DETC development has been well established, the exact role of TCR signaling in DETC homeostasis and function remains incompletely defined. In this study, we investigated TCR signaling in fully matured DETCs by lineage-restricted deletion of the Lat gene, an essential signaling molecule downstream of the TCR. We found that Lat deletion impaired TCR-dependent cytokine gene activation and the ability of DETCs to undergo proliferative expansion. However, linker for activation of T cells-deficient DETCs were able to maintain long-term population homeostasis, although with a reduced proliferation rate. Mice with Lat deletion in DETCs exhibited delayed wound healing accompanied by impaired clonal expansion within the wound area. Our study revealed differential requirements for TCR signaling in homeostatic maintenance of DETCs and in their effector function during wound healing.
Collapse
Affiliation(s)
- Baojun Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Jianxuan Wu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Yiqun Jiao
- Department of Medicine, Duke University Medical Center, Durham, NC 27710; and
| | - Cheryl Bock
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710
| | - Meifang Dai
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Benny Chen
- Department of Medicine, Duke University Medical Center, Durham, NC 27710; and
| | - Nelson Chao
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; Department of Medicine, Duke University Medical Center, Durham, NC 27710; and Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710
| | - Weiguo Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710
| | - Yuan Zhuang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
14
|
Malissen B, Grégoire C, Malissen M, Roncagalli R. Integrative biology of T cell activation. Nat Immunol 2014; 15:790-7. [PMID: 25137453 DOI: 10.1038/ni.2959] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 07/10/2014] [Indexed: 12/11/2022]
Abstract
The activation of T cells mediated by the T cell antigen receptor (TCR) requires the interaction of dozens of proteins, and its malfunction has pathological consequences. Our major focus is on new developments in the systems-level understanding of the TCR signal-transduction network. To make sense of the formidable complexity of this network, we argue that 'fine-grained' methods are needed to assess the relationships among a few components that interact on a nanometric scale, and those should be integrated with high-throughput '-omic' approaches that simultaneously capture large numbers of parameters. We illustrate the utility of this integrative approach with the transmembrane signaling protein Lat, which is a key signaling hub of the TCR signal-transduction network, as a connecting thread.
Collapse
Affiliation(s)
- Bernard Malissen
- 1] Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France. [2] INSERM U1104, Marseille, France. [3] CNRS UMR7280, Marseille, France. [4] Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France. [5] INSERM US012, Marseille, France. [6] CNRS UMS3367, Marseille, France
| | - Claude Grégoire
- 1] Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France. [2] INSERM U1104, Marseille, France. [3] CNRS UMR7280, Marseille, France
| | - Marie Malissen
- 1] Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France. [2] INSERM U1104, Marseille, France. [3] CNRS UMR7280, Marseille, France. [4] Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France. [5] INSERM US012, Marseille, France. [6] CNRS UMS3367, Marseille, France
| | - Romain Roncagalli
- 1] Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France. [2] INSERM U1104, Marseille, France. [3] CNRS UMR7280, Marseille, France
| |
Collapse
|
15
|
Sheng W, Liu C, Fu R, Wang H, Qu W, Ruan E, Wang G, Liu H, Wu Y, Song J, Xing L, Guan J, Li L, Liu H, Shao Z. Abnormalities of quantities and functions of linker for activations of T cells in severe aplastic anemia. Eur J Haematol 2014; 93:214-23. [PMID: 24673455 DOI: 10.1111/ejh.12327] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Severe aplastic anemia (SAA) is a rare immune-regulated disease characterized by severe pancytopenia and bone marrow failure, caused by destruction of hematopoietic cells by the activated T lymphocytes. Linker for activation of T cells (LAT), a transmembrane adaptor protein, plays a key role in T-cell and mast cell functions. However, it remains unclear how LAT may change in patients with SAA. This study aims at understanding the role of lymphocyte LAT in SAA. METHODS The expression of LAT, related signaling molecules, and T-cell effector molecules was determined by flow cytometry. LAT mRNA was evaluated by quantitative real-time PCR. Cytokine production by cultured T cells was determined by ELISA. RESULTS Patients with SAA had an increased levels of LAT and both total phosphorylated LAT and of the related molecule (ZAP-70) in circulating T cells compared with normal controls. In patients with SAA, the expression of LAT was positively associated with the expression of perforin and granzyme B in CD8(+) T cells. Inhibition of LAT expression in T cells from patients with SAA decreased the activation of the CD4(+) and CD8(+) T-cell subsets. Overexpression of LAT in T cells from normal controls increased the activation of CD4(+) and CD8(+) T-cell subsets with increased apoptosis of K562 cells in coculture. CONCLUSIONS Our findings demonstrate that dysregulation of LAT expression and activation may contribute to over-function of T cells, imbalance of Th1/Th2 subsets and thus lead to hematopoiesis failure in SAA. Immunosuppressive therapy dramatically reduced the expression of LAT making it an attractive therapeutic target in SAA.
Collapse
Affiliation(s)
- Weiwei Sheng
- The Department of Hematology, The General Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Roncagalli R, Hauri S, Fiore F, Liang Y, Chen Z, Sansoni A, Kanduri K, Joly R, Malzac A, Lähdesmäki H, Lahesmaa R, Yamasaki S, Saito T, Malissen M, Aebersold R, Gstaiger M, Malissen B. Quantitative proteomics analysis of signalosome dynamics in primary T cells identifies the surface receptor CD6 as a Lat adaptor-independent TCR signaling hub. Nat Immunol 2014; 15:384-392. [PMID: 24584089 DOI: 10.1038/ni.2843] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/03/2014] [Indexed: 02/08/2023]
Abstract
T cell antigen receptor (TCR)-mediated activation of T cells requires the interaction of dozens of proteins. Here we used quantitative mass spectrometry and activated primary CD4(+) T cells from mice in which a tag for affinity purification was knocked into several genes to determine the composition and dynamics of multiprotein complexes that formed around the kinase Zap70 and the adaptors Lat and SLP-76. Most of the 112 high-confidence time-resolved protein interactions we observed were previously unknown. The surface receptor CD6 was able to initiate its own signaling pathway by recruiting SLP-76 and the guanine nucleotide-exchange factor Vav1 regardless of the presence of Lat. Our findings provide a more complete model of TCR signaling in which CD6 constitutes a signaling hub that contributes to the diversification of TCR signaling.
Collapse
Affiliation(s)
- Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Simon Hauri
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Competence Center for Systems Physiology and Metabolic Diseases, ETH Zurich, Switzerland
| | - Fréderic Fiore
- Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| | - Yinming Liang
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Zhi Chen
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Amandine Sansoni
- Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| | - Kartiek Kanduri
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Rachel Joly
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Aurélie Malzac
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Harri Lähdesmäki
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland.,Department of Information and Computer Science, Aalto University, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Sho Yamasaki
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takashi Saito
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Marie Malissen
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Competence Center for Systems Physiology and Metabolic Diseases, ETH Zurich, Switzerland
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France.,Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| |
Collapse
|
17
|
Clayton KL, Haaland MS, Douglas-Vail MB, Mujib S, Chew GM, Ndhlovu LC, Ostrowski MA. T cell Ig and mucin domain-containing protein 3 is recruited to the immune synapse, disrupts stable synapse formation, and associates with receptor phosphatases. THE JOURNAL OF IMMUNOLOGY 2013; 192:782-91. [PMID: 24337741 DOI: 10.4049/jimmunol.1302663] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD8(+) CTLs are adept at killing virally infected cells and cancer cells and releasing cytokines (e.g., IFN-γ) to aid this response. However, during cancer and chronic viral infections, such as with HIV, this CTL response is progressively impaired due to a process called T cell exhaustion. Previous work has shown that the glycoprotein T cell Ig and mucin domain-containing protein 3 (Tim-3) plays a functional role in establishing T cell exhaustion. Tim-3 is highly upregulated on virus and tumor Ag-specific CD8(+) T cells, and antagonizing Tim-3 helps restore function of CD8(+) T cells. However, very little is known of how Tim-3 signals in CTLs. In this study, we assessed the role of Tim-3 at the immunological synapse as well as its interaction with proximal TCR signaling molecules in primary human CD8(+) T cells. Tim-3 was found within CD8(+) T cell lipid rafts at the immunological synapse. Blocking Tim-3 resulted in a significantly greater number of stable synapses being formed between Tim-3(hi)CD8(+) T cells and target cells, suggesting that Tim-3 plays a functional role in synapse formation. Further, we confirmed that Tim-3 interacts with Lck, but not the phospho-active form of Lck. Finally, Tim-3 colocalizes with receptor phosphatases CD45 and CD148, an interaction that is enhanced in the presence of the Tim-3 ligand, galectin-9. Thus, Tim-3 interacts with multiple signaling molecules at the immunological synapse, and characterizing these interactions could aid in the development of therapeutics to restore Tim-3-mediated immune dysfunction.
Collapse
Affiliation(s)
- Kiera L Clayton
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
It has been over 30 years since the reorganization of both the microtubule network and a 'peculiar actin polarization' was reported at the contact area of cytotoxic T lymphocytes interacting with target cells. Since that time, hundreds of studies have been published in an effort to elucidate the structure and function of the microtubule network and the actin cytoskeleton in T-cell activation, migration, and effector function at the interface between a T cell and its cognate antigen-presenting cell or target cell. This interface has become known as the immunological synapse, and this review examines some of the roles played by the cytoskeleton at the synapse.
Collapse
Affiliation(s)
- Alex T Ritter
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| | - Karen L Angus
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| |
Collapse
|
19
|
Ou-Yang CW, Zhu M, Sullivan SA, Fuller DM, Zhang W. The requirement of linker for activation of T cells in the primary and memory responses of CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:2938-47. [PMID: 23401587 DOI: 10.4049/jimmunol.1203163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Linker for activation of T cells (LAT) is a transmembrane adaptor protein that links TCR engagement to downstream signaling events. Although it is clear that LAT is essential in thymocyte development and initiation of T cell activation, its function during T cell expansion, contraction, and memory formation remains unknown. To study the role of TCR-mediated signaling in CD8 T cells during the course of pathogen infection, we used an inducible mouse model to delete LAT in Ag-specific CD8 T cells at different stages of Listeria infection and analyzed the effect of deletion on T cell responses. Our data showed that LAT is important for maintaining CD8 T cell expansion during the priming phase; however, it is not required for CD8 T cell contraction and memory maintenance. Moreover, LAT deficiency accelerates memory differentiation during the effector-to-memory transition, leading to a higher frequency of KLRG1(low)IL-7R(high)CD62L(high) memory T cells. Nonetheless, these LAT-deficient memory T cells were unable to proliferate or produce cytokines upon secondary infection. Our data demonstrated that, although TCR-mediated signaling is dispensable for contraction and memory maintenance, it regulates CD8 T cell memory differentiation and is essential for the memory response against pathogens.
Collapse
Affiliation(s)
- Chih-wen Ou-Yang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
20
|
Bartelt RR, Houtman JCD. The adaptor protein LAT serves as an integration node for signaling pathways that drive T cell activation. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2012; 5:101-10. [PMID: 23150273 DOI: 10.1002/wsbm.1194] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
T cells are essential for the adaptive immune response to pathogens. However, dysfunctional T cell activity has been implicated in numerous diseases, including the failure of organ transplants, allergic reactions, asthma, autoimmune disorders, and coronary artery disease. T cell responses to pathogens require the induction of the primary activating receptor, the T cell receptor (TCR), along with other costimulatory and adhesion receptors. Signal transduction pathways activated downstream of these receptors drive T cell responses required for the immune response and disease progression. A key question in our understanding of the mechanism of T cell activation is how signaling pathways emanating from multiple receptors integrate together to alter T cell effector functions. One integration node for intracellular signaling is the membrane-associated adaptor protein linker for the activation of T cells or LAT. Upon stimulation of the TCR and other receptors, LAT is phosphorylated at several tyrosines residues on its cytoplasmic tail. This leads to the binding of SH2 domain-containing proteins and their associated molecules and the formation of large multiprotein complexes. These dynamic and highly regulated signaling complexes facilitate the production of second messengers, activate downstream pathways, induce actin cytoskeleton polymerization, and stimulate the activity of multiple transcription factors. Thus, signaling pathways from several receptors feed into LAT, which then integrates this information and selectively induces pathways critical for T cell activation and the adaptive immune response.
Collapse
|