Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Immunol ; 17(4): 422-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950239

ABSTRACT

T cell responses are guided by cytokines that induce transcriptional regulators, which ultimately control differentiation of effector and memory T cells. However, it is unknown how the activities of these molecular regulators are coordinated and integrated during the differentiation process. Using genetic approaches and transcriptional profiling of antigen-specific CD8(+) T cells, we reveal a common program of effector differentiation that is regulated by IL-2 and IL-12 signaling and the combined activities of the transcriptional regulators Blimp-1 and T-bet. The loss of both T-bet and Blimp-1 leads to abrogated cytotoxic function and ectopic IL-17 production in CD8(+) T cells. Overall, our data reveal two major overlapping pathways of effector differentiation governed by the availability of Blimp-1 and T-bet and suggest a model for cytokine-induced transcriptional changes that combine, quantitatively and qualitatively, to promote robust effector CD8(+) T cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Interleukin-12/immunology , Interleukin-2/immunology , T-Box Domain Proteins/immunology , Transcription Factors/immunology , Animals , Arenaviridae Infections/immunology , Chromatin Immunoprecipitation , Cytokines/immunology , Flow Cytometry , Gene Expression Profiling , Influenza A Virus, H1N1 Subtype , Interleukin-17/immunology , Lymphocytic choriomeningitis virus , Mice , Orthomyxoviridae Infections/immunology , Positive Regulatory Domain I-Binding Factor 1 , Real-Time Polymerase Chain Reaction , STAT4 Transcription Factor/immunology , STAT5 Transcription Factor/immunology , Sequence Analysis, RNA , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 121(24): e2403054121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838017

ABSTRACT

Chronic Toxoplasma gondii infection induces brain-resident CD8+ T cells (bTr), but the protective functions and differentiation cues of these cells remain undefined. Here, we used a mouse model of latent infection by T. gondii leading to effective CD8+ T cell-mediated parasite control. Thanks to antibody depletion approaches, we found that peripheral circulating CD8+ T cells are dispensable for brain parasite control during chronic stage, indicating that CD8+ bTr are able to prevent brain parasite reactivation. We observed that the retention markers CD69, CD49a, and CD103 are sequentially acquired by brain parasite-specific CD8+ T cells throughout infection and that a majority of CD69/CD49a/CD103 triple-positive (TP) CD8+ T cells also express Hobit, a transcription factor associated with tissue residency. This TP subset develops in a CD4+ T cell-dependent manner and is associated with effective parasite control during chronic stage. Conditional invalidation of Transporter associated with Antigen Processing (TAP)-mediated major histocompatibility complex (MHC) class I presentation showed that presentation of parasite antigens by glutamatergic neurons and microglia regulates the differentiation of CD8+ bTr into TP cells. Single-cell transcriptomic analyses revealed that resistance to encephalitis is associated with the expansion of stem-like subsets of CD8+ bTr. In summary, parasite-specific brain-resident CD8+ T cells are a functionally heterogeneous compartment which autonomously ensure parasite control during T. gondii latent infection and which differentiation is shaped by neuronal and microglial MHC I presentation. A more detailed understanding of local T cell-mediated immune surveillance of this common parasite is needed for harnessing brain-resident CD8+ T cells in order to enhance control of chronic brain infections.


Subject(s)
Brain , CD8-Positive T-Lymphocytes , Cell Differentiation , Toxoplasma , Toxoplasmosis , Animals , CD8-Positive T-Lymphocytes/immunology , Toxoplasma/immunology , Mice , Brain/immunology , Brain/parasitology , Cell Differentiation/immunology , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Latent Infection/immunology , Latent Infection/parasitology , Antigens, CD/metabolism , Antigens, CD/immunology , Antigens, CD/genetics , Mice, Inbred C57BL , Female
3.
Immunity ; 43(6): 1101-11, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682984

ABSTRACT

Tissue-resident memory T (Trm) cells contribute to local immune protection in non-lymphoid tissues such as skin and mucosa, but little is known about their transcriptional regulation. Here we showed that CD8(+)CD103(+) Trm cells, independent of circulating memory T cells, were sufficient for protection against infection and described molecular elements that were crucial for their development in skin and lung. We demonstrated that the T-box transcription factors (TFs) Eomes and T-bet combined to control CD8(+)CD103(+) Trm cell formation, such that their coordinate downregulation was crucial for TGF-ß cytokine signaling. TGF-ß signaling, in turn, resulted in reciprocal T-box TF downregulation. However, whereas extinguishment of Eomes was necessary for CD8(+)CD103(+) Trm cell development, residual T-bet expression maintained cell surface interleukin-15 (IL-15) receptor ß-chain (CD122) expression and thus IL-15 responsiveness. These findings indicate that the T-box TFs control the two cytokines, TGF-ß and IL-15, which are pivotal for CD8(+)CD103(+) Trm cell development and survival.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-15/immunology , T-Box Domain Proteins/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Down-Regulation , Flow Cytometry , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology
4.
Nat Immunol ; 12(4): 304-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21378976

ABSTRACT

Regulatory T cells (T(reg) cells) are required for peripheral tolerance. Evidence indicates that T(reg) cells can adopt specialized differentiation programs in the periphery that are controlled by transcription factors usually associated with helper T cell differentiation. Here we demonstrate that expression of the transcription factor Blimp-1 defined a population of T(reg) cells that localized mainly to mucosal sites and produced IL-10. Blimp-1 was required for IL-10 production by these cells and for their tissue homeostasis. We provide evidence that the transcription factor IRF4, but not the transcription factor T-bet, was essential for Blimp-1 expression and for the differentiation of all effector T(reg) cells. Thus, our study defines a differentiation pathway that leads to the acquisition of T(reg) cell effector functions and requires both IRF4 and Blimp-1.


Subject(s)
Cell Differentiation/genetics , Interferon Regulatory Factors/genetics , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Positive Regulatory Domain I-Binding Factor 1 , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes, Regulatory/cytology , Transcription Factors/metabolism
5.
Brain ; 145(6): 2018-2030, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35552381

ABSTRACT

Narcolepsy with cataplexy or narcolepsy type 1 is a disabling chronic sleep disorder resulting from the destruction of orexinergic neurons in the hypothalamus. The tight association of narcolepsy with HLA-DQB1*06:02 strongly suggest an autoimmune origin to this disease. Furthermore, converging epidemiological studies have identified an increased incidence for narcolepsy in Europe following Pandemrix® vaccination against the 2009-2010 pandemic 'influenza' virus strain. The potential immunological link between the Pandemrix® vaccination and narcolepsy remains, however, unknown. Deciphering these mechanisms may reveal pathways potentially at play in most cases of narcolepsy. Here, we developed a mouse model allowing to track and study the T-cell response against 'influenza' virus haemagglutinin, which was selectively expressed in the orexinergic neurons as a new self-antigen. Pandemrix® vaccination in this mouse model resulted in hypothalamic inflammation and selective destruction of orexin-producing neurons. Further investigations on the relative contribution of T-cell subsets in this process revealed that haemagglutinin-specific CD4 T cells were necessary for the development of hypothalamic inflammation, but insufficient for killing orexinergic neurons. Conversely, haemagglutinin-specific CD8 T cells could not initiate inflammation but were the effectors of the destruction of orexinergic neurons. Additional studies revealed pathways potentially involved in the disease process. Notably, the interferon-γ pathway was proven essential, as interferon-γ-deficient CD8 T cells were unable to elicit the loss of orexinergic neurons. Our work demonstrates that an immunopathological process mimicking narcolepsy can be elicited by immune cross-reactivity between a vaccine antigen and a neuronal self-antigen. This process relies on a synergy between autoreactive CD4 and CD8 T cells for disease development. This work furthers our understanding of the mechanisms and pathways potentially involved in the development of a neurological side effect due to a vaccine and, likely, to narcolepsy in general.


Subject(s)
Autoimmunity , Influenza Vaccines , Narcolepsy , Animals , Autoantigens , Hemagglutinins , Inflammation/complications , Influenza Vaccines/adverse effects , Interferon-gamma , Mice , Narcolepsy/chemically induced , Neurons , Orexins , T-Lymphocytes/immunology , Vaccination/adverse effects
6.
Immunity ; 32(1): 7-9, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20152165

ABSTRACT

The differentiation of peripheral T lymphocytes depends on interactions between intrinsic and extrinsic factors. In this issue of Immunity, Pipkin et al. (2010) and Kalia et al. (2010) link differential interleukin-2 signaling and inflammation with the transcriptional events leading to the development of effector and memory cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-2/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Humans
7.
Blood ; 123(22): 3420-8, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24723679

ABSTRACT

Interleukin-10 (IL-10) is a key immunoregulatory cytokine that functions to prevent inflammatory and autoimmune diseases. Despite the critical role for IL-10 produced by effector CD8(+) T cells during pathogen infection and autoimmunity, the mechanisms regulating its production are poorly understood. We show that loss of the inhibitor of DNA binding 2 (Id2) in T cells resulted in aberrant IL-10 expression in vitro and in vivo during influenza virus infection and in a model of acute graft-versus-host disease (GVHD). Furthermore, IL-10 overproduction substantially reduced the immunopathology associated with GVHD. We demonstrate that Id2 acts to repress the E2A-mediated trans-activation of the Il10 locus. Collectively, our findings uncover a key regulatory role of Id2 during effector T cell differentiation necessary to limit IL-10 production by activated T cells and minimize their suppressive activity during the effector phase of disease control.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Inhibitor of Differentiation Protein 2/metabolism , Interleukin-10/genetics , T-Lymphocyte Subsets/metabolism , Transcriptional Activation , Animals , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation , Genetic Loci , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft vs Host Disease/mortality , Inhibitor of Differentiation Protein 2/deficiency , Inhibitor of Differentiation Protein 2/genetics , Interleukin-10/metabolism , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/mortality
8.
EMBO J ; 30(13): 2690-704, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21587207

ABSTRACT

Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α(+) and CD103(+) DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103(+) and CD8α(+) lineages. Notably, CD103(+) DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α(-) DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP(+) DCs. It did not restore cross-presenting capacity to Batf3(-/-) or CD103(-)Sirp-α(-)DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.


Subject(s)
Antigens, CD/metabolism , CD8 Antigens/metabolism , Cell Lineage/genetics , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/genetics , Integrin alpha Chains/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression/physiology , Genes, cdc/physiology , Inhibitor of Differentiation Protein 2/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological
9.
J Immunol ; 191(8): 4383-91, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24038093

ABSTRACT

Innate lymphocyte populations play a central role in conferring protective immunity at the mucosal frontier. In this study, we demonstrate that T cell factor 1 (TCF-1; encoded by Tcf7), a transcription factor also important for NK and T cell differentiation, is expressed by multiple innate lymphoid cell (ILC) subsets, including GATA3(+) nuocytes (ILC2) and NKp46(+) ILCs (ILC3), which confer protection against lung and intestinal inflammation. TCF-1 was intrinsically required for the differentiation of both ILC2 and NKp46(+) ILC3. Loss of TCF-1 expression impaired the capacity of these ILC subsets to produce IL-5, IL-13, and IL-22 and resulted in crippled responses to intestinal infection with Citrobacter rodentium. Furthermore, a reduction in T-bet expression required for Notch-2-dependent development of NKp46(+) ILC3 showed a dose-dependent reduction in TCF-1 expression. Collectively, our findings demonstrate an essential requirement for TCF-1 in ILC2 differentiation and reveal a link among Tcf7, Notch, and Tbx21 in NKp46(+) ILC3 development.


Subject(s)
Intestines/immunology , Killer Cells, Natural/metabolism , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, Ly/metabolism , Cell Differentiation/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 1-alpha , Inflammation/immunology , Inflammation/microbiology , Interleukin-13/biosynthesis , Interleukin-5/biosynthesis , Interleukins/biosynthesis , Intestines/microbiology , Lymphocyte Activation , Mice , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptor, Notch2/metabolism , T Cell Transcription Factor 1/genetics , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/immunology , Interleukin-22
10.
J Immunol ; 190(9): 4585-94, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536629

ABSTRACT

The transcription factor inhibitor of DNA binding (Id)2 modulates T cell fate decisions, but the molecular mechanism underpinning this regulation is unclear. In this study we show that loss of Id2 cripples effector differentiation and instead programs CD8(+) T cells to adopt a memory fate with increased Eomesodermin and Tcf7 expression. We demonstrate that Id2 restrains CD8(+) T cell memory differentiation by inhibiting E2A-mediated direct activation of Tcf7 and that Id2 expression level mirrors T cell memory recall capacity. As a result of the defective effector differentiation, Id2-deficient CD8(+) T cells fail to induce sufficient Tbx21 expression to generate short-lived effector CD8(+) T cells. Our findings reveal that the Id2/E2A axis orchestrates T cell differentiation through the induction or repression of downstream transcription factors essential for effector and memory T cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory/immunology , Inhibitor of Differentiation Protein 2/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Gene Expression/immunology , Hepatocyte Nuclear Factor 1-alpha , Immunologic Memory/genetics , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Mice , Mice, Inbred C57BL , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T Cell Transcription Factor 1/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism
11.
Neurol Neuroimmunol Neuroinflamm ; 11(6): e200306, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39231385

ABSTRACT

BACKGROUND AND OBJECTIVES: The rs763361 nonsynonymous variant in the CD226 gene, which results in a glycine-to-serine substitution at position 307 of the CD226 protein, has been implicated as a risk factor of various immune-mediated diseases, including multiple sclerosis (MS). Compelling evidence suggests that this allele may play a significant role in predisposing individuals to MS by decreasing the immune-regulatory capacity of Treg cells and increasing the proinflammatory potential of effector CD4 T cells. However, the impact of this CD226 gene variant on CD8 T-cell functions, a population that also plays a key role in MS, remains to be determined. METHODS: To study whether the CD226 risk variant affects human CD8 T-cell functions, we used CD8 T cells isolated from peripheral blood mononuclear cell of 16 age-matched healthy donors homozygous for either the protective or the risk allele of CD226. We characterized these CD8 T cells on T-cell receptor (TCR) stimulation using high-parametric flow cytometry and bulk RNAseq and through characterization of canonical signaling pathways and cytokine production. RESULTS: On TCR engagement, the phenotype of ex vivo CD8 T cells bearing the protective (CD226-307Gly) or the risk (CD226-307Ser) allele of CD226 was largely overlapping. However, the transcriptomic signature of CD8 T cells from the donors carrying the risk allele presented an enrichment in TCR, JAK/STAT, and IFNγ signaling. We next found that the CD226-307Ser risk allele leads to a selective increase in the phosphorylation of the mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 (ERK1/2) associated with enhanced phosphorylation of STAT4 and increased production of IFNγ. DISCUSSION: Our data suggest that the CD226-307Ser risk variant imposes immune dysregulation by increasing the pathways related to IFNγ signaling in CD8 T cells, thereby contributing to the risk of developing chronic inflammation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte , CD8-Positive T-Lymphocytes , Multiple Sclerosis , Humans , CD8-Positive T-Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Adult , Female , Male , Middle Aged
12.
J Immunol ; 186(7): 4059-66, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339359

ABSTRACT

Upon Ag encounter, naive T cells undergo extensive Ag-driven proliferation and can differentiate into effector cells. Up to 95% of these cells die leaving a small residual population of T cells that provide protective memory. In this study, we investigated the contribution of the BH3-only family protein Bid in the shutdown of T cell responses after acute and persistent infection. Influenza virus pathogenicity has been proposed to be mediated by a peptide encoded in the basic polymerase (PB1-RF2) acting through Bid. In our experiments, we found that after acute infection with influenza virus, mice lacking Bid had normal expansion and contraction of Ag-specific CD8(+) T cells. However, in chronic γ-herpesvirus infection, Bid-deficient virus-specific CD8(+) T cells expanded normally but failed to contract fully and were maintained at ∼2-fold higher levels. Previously, we have demonstrated that Bim plays a prominent role in T cell shutdown in persistent infection by cooperating with the death receptor Fas, which regulates apoptosis in response to repeated TCR signaling. Bid lies at the nexus of these two signaling pathways, thus we reasoned that Bid and Bim might cooperate in regulation of T cell shutdown in persistent infection. In this study, we observed that the combined loss of Bid and Bim synergistically enhanced the persistence of CD8(+) T cells during γ-herpesvirus infection. Thus, these data uncover a role for Bid in coordinating apoptotic signaling pathways to ensure appropriate shutdown of T cell immune responses in the setting of persistent Ag exposure.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/physiology , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Lymphocyte Cooperation/immunology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Bcl-2-Like Protein 11 , Cell Communication/immunology , Cell Death/immunology , Herpesviridae Infections/metabolism , Influenza A Virus, H3N2 Subtype/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Rhadinovirus/immunology , T-Lymphocyte Subsets/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Viral Load/immunology
13.
J Immunol ; 185(6): 3669-76, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20720208

ABSTRACT

A critical factor influencing the ability of the host to mount a robust immune response against a virus depends on the rapid recruitment of dendritic cells (DCs) presenting Ags. From the outset, this step sets the tempo for subsequent activation of virus-specific T cells. Despite this, how induction of the immune response might be modified by pathogens with the capacity to establish persistence is unclear. In this study, we have characterized the in vivo influence of murine gamma-herpesvirus K3-mediated interference with MHC class I in DCs that drive the initial adaptive immune response. We observed that gamma-herpesvirus could interfere with the very earliest phase of Ag presentation through K3 by directly targeting migratory and lymph node-resident DCs. These results show that a pathogen with the capacity to interfere with early Ag presentation can establish suboptimal conditions for rapid induction of the adaptive immune response and thus favor establishment of viral persistence.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Animals , Chronic Disease , Cross-Priming/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Herpesviridae Infections/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rhadinovirus/pathogenicity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Tumor Virus Infections/metabolism , Viral Interference/immunology , Viral Proteins/antagonists & inhibitors , Viral Proteins/biosynthesis
14.
Curr Opin Immunol ; 77: 102211, 2022 08.
Article in English | MEDLINE | ID: mdl-35644112

ABSTRACT

Tissue-resident memory T cells (TRM) represent a subset of antigen-experienced T cells that are constantly retained in a given tissue with limited trafficking through the circulation. These cells are characterized by expression of molecules enabling their tissue anchoring and downregulation of molecules promoting tissue egress. They reside at sites of previous antigen encounter and their number increases with age. TRM have been shown to provide rapid and efficient protection against tissue reinfection and TRM density correlates with efficient antitumor responses. Intriguingly, the density of CD8 TRM is increased in the central nervous system (CNS) of patients with neuroinflammatory diseases such as multiple sclerosis, or suffering from neurodegenerative diseases. In this review, we discuss current knowledge regarding the diversity of CNS-resident CD8 T cells and their role in CNS autoimmunity. Given their likely contribution to the protracted course of several inflammatory diseases of the CNS, their therapeutic targeting becomes an important challenge.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Antigens/metabolism , Central Nervous System , Crime , Humans
15.
Sci Transl Med ; 14(640): eabl6157, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417189

ABSTRACT

The mechanisms underlying the chronicity of autoimmune diseases of the central nervous system (CNS) are largely unknown. In particular, it is unclear whether tissue-resident memory T cells (TRM) contribute to lesion pathogenesis during chronic CNS autoimmunity. Here, we observed that a high frequency of brain-infiltrating CD8+ T cells exhibit a TRM-like phenotype in human autoimmune encephalitis. Using mouse models of neuronal autoimmunity and a combination of T single-cell transcriptomics, high-dimensional flow cytometry, and histopathology, we found that pathogenic CD8+ T cells behind the blood-brain barrier adopt a characteristic TRM differentiation program, and we revealed their phenotypic and functional heterogeneity. In the diseased CNS, autoreactive tissue-resident CD8+ T cells sustained focal neuroinflammation and progressive loss of neurons, independently of recirculating CD8+ T cells. Consistently, a large fraction of autoreactive tissue-resident CD8+ T cells exhibited proliferative potential as well as proinflammatory and cytotoxic properties. Persistence of tissue-resident CD8+ T cells in the CNS and their functional output, but not their initial differentiation, were crucially dependent on CD4+ T cells. Collectively, our results point to tissue-resident CD8+ T cells as essential drivers of chronic CNS autoimmunity and suggest that therapies targeting this compartmentalized autoreactive T cell subset might be effective for treating CNS autoimmune diseases.


Subject(s)
Autoimmune Diseases , CD8-Positive T-Lymphocytes , Animals , Autoimmune Diseases/pathology , Central Nervous System , Immunologic Memory , Mice , Neurons
16.
Cancer Immunol Res ; 9(7): 735-747, 2021 07.
Article in English | MEDLINE | ID: mdl-33906864

ABSTRACT

IL11 is a member of the IL6 family of cytokines and signals through its cognate receptor subunits, IL11RA and glycoprotein 130 (GP130), to elicit biological responses via the JAK/STAT signaling pathway. IL11 contributes to cancer progression by promoting the survival and proliferation of cancer cells, but the potential immunomodulatory properties of IL11 signaling during tumor development have thus far remained unexplored. Here, we have characterized a role for IL11 in regulating CD4+ T cell-mediated antitumor responses. Absence of IL11 signaling impaired tumor growth in a sporadic mouse model of colon cancer and syngeneic allograft models of colon cancer. Adoptive bone marrow transfer experiments and in vivo depletion studies demonstrated that the tumor-promoting activity of IL11 was mediated through its suppressive effect on host CD4+ T cells in the tumor microenvironment. Indeed, when compared with Il11ra-proficient CD4+ T cells associated with MC38 tumors, their Il11ra-deficient counterparts displayed elevated expression of mRNA encoding the antitumor mediators IFNγ and TNFα. Likewise, IL11 potently suppressed the production of proinflammatory cytokines (IFNγ, TNFα, IL6, and IL12p70) by CD4+ T cells in vitro, which we corroborated by RNAscope analysis of human colorectal cancers, where IL11RAhigh tumors showed less IFNG and CD4 expression than IL11RAlow tumors. Therefore, our results ascribe a tumor cell-extrinsic immunomodulatory role to IL11 during colon cancer development that could be amenable to an anticytokine-based therapy.See related Spotlight by van der Burg, p. 724.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Interleukin-11 Receptor alpha Subunit/metabolism , Interleukin-11/metabolism , Animals , CD4 Antigens/analysis , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colon/immunology , Colon/pathology , Colonic Neoplasms/pathology , Datasets as Topic , Disease Models, Animal , Gene Expression Profiling , Humans , Interferon-gamma/analysis , Interferon-gamma/metabolism , Interleukin-11 Receptor alpha Subunit/analysis , Interleukin-11 Receptor alpha Subunit/genetics , Mice , Mice, Knockout , Neoplasms, Bone Tissue , Receptors, Interleukin-11/metabolism , Tumor Microenvironment/immunology
17.
Handb Exp Pharmacol ; (188): 51-77, 2009.
Article in English | MEDLINE | ID: mdl-19031021

ABSTRACT

Antigen presenting cells (APCs) are recognized as key initiators of adaptive immunity, particularly to pathogens, by eliciting a rapid and potent immune attack on infected cells. Amongst APCs, dendritic cells (DCs) are specially equipped to initiate and regulate immune responses in a manner that depends on signals they receive from microbes and their cellular environment. To achieve this, they are equipped with highly efficient mechanisms that allow them to detect pathogens, to capture, process and present antigens, and to activate and guide the differentiation of T cells into effector and memory cells. DCs can no longer be considered as a homogeneous cell type performing a single function, but are heterogeneous both in phenotype, function and dependence on inflammatory stimuli for their formation and responsiveness. Recent studies of DC subtypes have highlighted the contrasting roles of different professional APCs in activating divergent arms of the immune response towards pathogens. In this review, we discuss the progress that has been made in dissecting the attributes of different DC subsets that migrate into, or reside permanently, within lymphoid tissues and their putative roles in the induction of the anti-viral immune response.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , Dendritic Cells/immunology , Virus Diseases/immunology , Animals , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology
18.
Nat Commun ; 10(1): 2735, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227713

ABSTRACT

The contribution of mast cells in the microenvironment of solid malignancies remains controversial. Here we functionally assess the impact of tumor-adjacent, submucosal mast cell accumulation in murine and human intestinal-type gastric cancer. We find that genetic ablation or therapeutic inactivation of mast cells suppresses accumulation of tumor-associated macrophages, reduces tumor cell proliferation and angiogenesis, and diminishes tumor burden. Mast cells are activated by interleukin (IL)-33, an alarmin produced by the tumor epithelium in response to the inflammatory cytokine IL-11, which is required for the growth of gastric cancers in mice. Accordingly, ablation of the cognate IL-33 receptor St2 limits tumor growth, and reduces mast cell-dependent production and release of the macrophage-attracting factors Csf2, Ccl3, and Il6. Conversely, genetic or therapeutic macrophage depletion reduces tumor burden without affecting mast cell abundance. Therefore, tumor-derived IL-33 sustains a mast cell and macrophage-dependent signaling cascade that is amenable for the treatment of gastric cancer.


Subject(s)
Interleukin-33/immunology , Macrophages/immunology , Mast Cells/immunology , Stomach Neoplasms/immunology , Aminopyridines/administration & dosage , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cromolyn Sodium/administration & dosage , Disease Models, Animal , Epithelium/immunology , Epithelium/pathology , Female , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Humans , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Male , Mice , Mice, Transgenic , Pyrroles/administration & dosage , Signal Transduction/drug effects , Signal Transduction/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis , Tumor Microenvironment/immunology
19.
Glia ; 56(15): 1625-36, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18551629

ABSTRACT

T cell mediated immunotherapies are proposed for many cancers including malignant astrocytoma. As such therapies become more potent, but not necessarily more tumor-specific, the risk of collateral autoimmune damage to normal tissue increases. Tumors of the brain present significant challenges in this respect, as autoimmune destruction of brain tissue could have severe consequences. To investigate local immune reactivity toward a tumor-associated antigen in the brain, transgenic mice were generated that express a defined antigen (CW3 170-179) in astroglial cells. The resulting six transgenic mouse lines expressed the transgenic self-antigen in cells of the gastrointestinal tract and CNS compartments, or in the CNS alone. By challenging transgenic mice with tumor cells that express CW3, self/tumor-specific immune responses were visualized within a normal polyclonal T cell repertoire. A large expansion of the endogenous CW3 170-179-specific CD8 T cell population was observed in nontransgenic mice after both subcutaneous and intracerebral implantation of tumor cells. In contrast, CW3 170-179-specific immune responses were not observed in transgenic mice that exhibited extracerebral transgene expression. Importantly, in certain groups of mice in which transgene expression was restricted to the CNS, antigen-specific immune responses occurred when tumor was implanted subcutaneously, but not intracerebrally. This local immune tolerance in the brain was induced via peripheral (extrathymic) rather than central (thymic) tolerance mechanisms. Thus, this study highlights the role of regional immune regulation in the prevention of autoimmunity in the brain, and the potential impact of these mechanisms for brain tumor immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Astrocytes/immunology , Astrocytoma/immunology , Brain Neoplasms/immunology , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Animals , Antigens, Neoplasm/genetics , Astrocytes/pathology , Astrocytoma/physiopathology , Autoantigens/genetics , Autoantigens/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/prevention & control , Autoimmunity/genetics , Autoimmunity/immunology , Brain/pathology , Brain/physiopathology , Brain Neoplasms/physiopathology , Brain Tissue Transplantation , Immunotherapy/adverse effects , Immunotherapy/methods , Mice , Mice, Transgenic , Tumor Cells, Cultured
20.
Cancer Immunol Res ; 6(4): 409-421, 2018 04.
Article in English | MEDLINE | ID: mdl-29463593

ABSTRACT

Interleukin 33 (IL33) is an inflammatory cytokine released during necrotic cell death. The epithelium and stroma of the intestine express large amounts of IL33 and its receptor St2. IL33 is therefore continuously released during homeostatic turnover of the intestinal mucosa. Although IL33 can prevent colon cancer associated with inflammatory colitis, the contribution of IL33 signaling to sporadic colon cancer remains unknown. Here, we utilized a mouse model of sporadic colon cancer to investigate the contribution of IL33 signaling to tumorigenesis in the absence of preexisting inflammation. We demonstrated that genetic ablation of St2 enhanced colon tumor development. Conversely, administration of recombinant IL33 reduced growth of colon cancer cell allografts. In reciprocal bone marrow chimeras, the concurrent loss of IL33 signaling within radioresistant nonhematopoietic, and the radiosensitive hematopoietic, compartments was associated with increased tumor burden. We detected St2 expression within the radioresistant mesenchymal cell compartment of the colon whose stimulation with IL33 induced expression of bona fide NF-κB target genes. Mechanistically, we discovered that St2 deficiency within the nonhematopoietic compartment coincided with increased abundance of regulatory T cells and suppression of an IFNγ gene expression signature, whereas IL33 administration triggered IFNγ expression by tumor allograft-infiltrating T cells. The decrease of this IFNγ gene expression signature was associated with more aggressive disease in human colon cancer patients, suggesting that lack of IL33 signaling impaired the generation of a potent IFNγ-mediated antitumor immune response. Collectively, our data reveal that IL33 functions as a tumor suppressor in sporadic colon cancer. Cancer Immunol Res; 6(4); 409-21. ©2018 AACR.


Subject(s)
Colonic Neoplasms/metabolism , Interferon-gamma/metabolism , Interleukin-33/metabolism , Signal Transduction , Allografts , Animals , Biomarkers , Biopsy , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression , Gene Expression Profiling , Interferon-gamma/genetics , Interleukin-33/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL