Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 8(88): eadf8838, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37889984

ABSTRACT

In chronic infections and cancer, T cells are exposed to prolonged antigen stimulation, resulting in loss of function (or exhaustion) and impairment of effective immunological protection. Exhausted T cells are heterogeneous and include early progenitors (Tpex) and terminally exhausted cells (Tex). Here, we used bulk and single-cell transcriptomics to analyze expression of transposable elements (TEs) in subpopulations of mouse and human CD8+ tumor-infiltrating T lymphocytes (TILs). We show that in mice, members of the virus-like murine VL30 TE family (mostly intact, evolutionary young ERV1s) are strongly repressed in terminally exhausted CD8+ T cells in both tumor and viral models of exhaustion. Tpex expression of these VL30s, which are mainly intergenic and transcribed independently of their closest gene neighbors, was driven by Fli1, a transcription factor involved in progression from Tpex to Tex. Immune checkpoint blockade (ICB) in both mice and patients with cancer increased TE expression (including VL30 in mice), demonstrating that TEs may be applicable as ICB response biomarkers. We conclude that expression of TEs is tightly regulated in TILs during establishment of exhaustion and reprogramming by ICB. Analyses of TE expression on single cells and bulk populations open opportunities for understanding immune cell identity and heterogeneity, as well as for defining cellular gene expression signatures and disease biomarkers.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Mice , Humans , DNA Transposable Elements/genetics , T-Cell Exhaustion , Biomarkers
2.
Sci Immunol ; 8(80): eabm6359, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36735774

ABSTRACT

Although most characterized tumor antigens are encoded by canonical transcripts (such as differentiation or tumor-testis antigens) or mutations (both driver and passenger mutations), recent results have shown that noncanonical transcripts including long noncoding RNAs and transposable elements (TEs) can also encode tumor-specific neo-antigens. Here, we investigate the presentation and immunogenicity of tumor antigens derived from noncanonical mRNA splicing events between coding exons and TEs. Comparing human non-small cell lung cancer (NSCLC) and diverse healthy tissues, we identified a subset of splicing junctions that is both tumor specific and shared across patients. We used HLA-I peptidomics to identify peptides encoded by tumor-specific junctions in primary NSCLC samples and lung tumor cell lines. Recurrent junction-encoded peptides were immunogenic in vitro, and CD8+ T cells specific for junction-encoded epitopes were present in tumors and tumor-draining lymph nodes from patients with NSCLC. We conclude that noncanonical splicing junctions between exons and TEs represent a source of recurrent, immunogenic tumor-specific antigens in patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Male , Humans , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA Transposable Elements , CD8-Positive T-Lymphocytes/pathology , Neoplasm Recurrence, Local/genetics , Exons/genetics , Antigens, Neoplasm/genetics
3.
Sci Immunol ; 8(80): eabm6360, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36735776

ABSTRACT

Oncogenesis often implicates epigenetic alterations, including derepression of transposable elements (TEs) and defects in alternative splicing. Here, we explore the possibility that noncanonical splice junctions between exons and TEs represent a source of tumor-specific antigens. We show that mouse normal tissues and tumor cell lines express wide but distinct ranges of mRNA junctions between exons and TEs, some of which are tumor specific. Immunopeptidome analyses in tumor cell lines identified peptides derived from exon-TE splicing junctions associated to MHC-I molecules. Exon-TE junction-derived peptides were immunogenic in tumor-bearing mice. Both prophylactic and therapeutic vaccinations with junction-derived peptides delayed tumor growth in vivo. Inactivation of the TE-silencing histone 3-lysine 9 methyltransferase Setdb1 caused overexpression of new immunogenic junctions in tumor cells. Our results identify exon-TE splicing junctions as epigenetically controlled, immunogenic, and protective tumor antigens in mice, opening possibilities for tumor targeting and vaccination in patients with cancer.


Subject(s)
Antigens, Neoplasm , DNA Transposable Elements , Animals , Mice , DNA Transposable Elements/genetics , Antigens, Neoplasm/genetics , Exons/genetics , RNA, Messenger , Cell Line, Tumor
4.
Cell Rep ; 40(7): 111205, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977488

ABSTRACT

Despite its crucial role in initiation of cytotoxic immune responses, the molecular pathways underlying antigen cross-presentation remain incompletely understood. The mechanism of antigen exit from endocytic compartments into the cytosol is a long-standing matter of controversy, confronting two main models: transfer through specific channels/transporters or rupture of endocytic membranes and leakage of luminal content. By monitoring the occurrence of intracellular damage in conventional dendritic cells (cDCs), we show that cross-presenting cDC1s display more frequent endomembrane injuries and increased recruitment of endosomal sorting complex required for transport (ESCRT)-III, the main repair system for intracellular membranes, relative to cDC2s. Silencing of CHMP2a or CHMP4b, two effector subunits of ESCRT-III, enhances cytosolic antigen export and cross-presentation. This phenotype is partially reversed by chemical inhibition of RIPK3, suggesting that endocytic damage is related to basal activation of the necroptosis pathway. Membrane repair therefore proves crucial in containing antigen export to the cytosol and cross-presentation in cDCs.


Subject(s)
Cross-Priming , Endosomal Sorting Complexes Required for Transport , Antigen Presentation , Antigens/metabolism , Cytosol/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism
5.
Nature ; 584(7822): 533-534, 2020 08.
Article in English | MEDLINE | ID: mdl-32788699
7.
Proc Natl Acad Sci U S A ; 116(51): 25839-25849, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776254

ABSTRACT

Naive CD4+ T lymphocytes differentiate into different effector types, including helper and regulatory cells (Th and Treg, respectively). Heritable gene expression programs that define these effector types are established during differentiation, but little is known about the epigenetic mechanisms that install and maintain these programs. Here, we use mice defective for different components of heterochromatin-dependent gene silencing to investigate the epigenetic control of CD4+ T cell plasticity. We show that, upon T cell receptor (TCR) engagement, naive and regulatory T cells defective for TRIM28 (an epigenetic adaptor for histone binding modules) or for heterochromatin protein 1 ß and γ isoforms (HP1ß/γ, 2 histone-binding factors involved in gene silencing) fail to effectively signal through the PI3K-AKT-mTOR axis and switch to glycolysis. While differentiation of naive TRIM28-/- T cells into cytokine-producing effector T cells is impaired, resulting in reduced induction of autoimmune colitis, TRIM28-/- regulatory T cells also fail to expand in vivo and to suppress autoimmunity effectively. Using a combination of transcriptome and chromatin immunoprecipitation-sequencing (ChIP-seq) analyses for H3K9me3, H3K9Ac, and RNA polymerase II, we show that reduced effector differentiation correlates with impaired transcriptional silencing at distal regulatory regions of a defined set of Treg-associated genes, including, for example, NRP1 or Snai3. We conclude that TRIM28 and HP1ß/γ control metabolic reprograming through epigenetic silencing of a defined set of Treg-characteristic genes, thus allowing effective T cell expansion and differentiation into helper and regulatory phenotypes.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic/physiology , T-Lymphocytes/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Animals , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Plasticity/physiology , Cellular Reprogramming/genetics , Chromobox Protein Homolog 5 , Colon/pathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Silencing , Histones/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome , Tripartite Motif-Containing Protein 28/genetics
9.
Mol Immunol ; 101: 370-376, 2018 09.
Article in English | MEDLINE | ID: mdl-30055407

ABSTRACT

B cells are an integral part of the adaptive immune system. During an immune response, the actin cytoskeleton plays a central role in regulating B cell antigen uptake, polarization and presentation as well as B cell migration and interaction with T cells. Genetic defects affecting actin regulators can result in reduced B cell activation, limited antibody production and hence cause disease. In this review, we discuss molecular mechanisms of actin regulation and their involvement in antigen polarisation and presentation, as well as their role in influencing interactions between B and T cells. Improved understanding of these mechanisms is necessary for the development of new therapeutic options modulating humoral immune responses.


Subject(s)
Actins/metabolism , Antigen Presentation/immunology , B-Lymphocytes/immunology , Immunity, Humoral , T-Lymphocytes/immunology , Animals , Germinal Center/metabolism , Humans
10.
Cell Rep ; 24(3): 619-629, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021160

ABSTRACT

Wiskott-Aldrich syndrome protein (WASp) is a main cytoskeletal regulator in B cells. WASp-interacting protein (WIP) binds to and stabilizes WASp but also interacts with actin. Using mice with a mutated actin binding domain of WIP (WIPΔABD), we here investigated the role of WIP binding to actin during B cell activation. We found an altered differentiation of WIPΔABD B cells and diminished antibody affinity maturation after immunization. Mechanistically, WIPΔABD B cells showed impaired B cell receptor (BCR)-induced PI3K signaling and actin reorganization, likely caused by diminished CD81 expression and altered CD19 dynamics on the B cell surface. WIPΔABD B cells displayed reduced in vivo motility, concomitantly with impaired chemotaxis and defective F-actin polarization, HS1 phosphorylation, and polarization of HS1 to F-actin-rich structures after CXCL12 stimulation in vitro. We thus concluded that WIP binding to actin, independent of its binding to WASp, is critical for actin cytoskeleton plasticity in B cells.


Subject(s)
Actins/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Movement , Immunity, Humoral , Animals , Antibody Affinity , Antigens, CD/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cell Polarity , Chemotaxis , Cytoskeletal Proteins , Diffusion , Germinal Center/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
11.
Elife ; 72018 01 16.
Article in English | MEDLINE | ID: mdl-29337666

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an immune pathology associated with mutations in WAS protein (WASp) or in WASp interacting protein (WIP). Together with the small GTPase Cdc42 and other effectors, these proteins participate in the remodelling of the actin network downstream of BCR engagement. Here we show that mice lacking the adaptor protein ITSN2, a G-nucleotide exchange factor (GEF) for Cdc42 that also interacts with WASp and WIP, exhibited increased mortality during primary infection, incomplete protection after Flu vaccination, reduced germinal centre formation and impaired antibody responses to vaccination. These defects were found, at least in part, to be intrinsic to the B cell compartment. In vivo, ITSN2 deficient B cells show a reduction in the expression of SLAM, CD84 or ICOSL that correlates with a diminished ability to form long term conjugates with T cells, to proliferate in vivo, and to differentiate into germinal centre cells. In conclusion, our study not only revealed a key role for ITSN2 as an important regulator of adaptive immune-response during vaccination and viral infection but it is also likely to contribute to a better understanding of human immune pathologies.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , B-Lymphocytes/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Animals , Cell Adhesion , Cell Proliferation , Influenza Vaccines/administration & dosage , Mice , Survival Analysis
12.
Cell ; 172(3): 517-533.e20, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29249358

ABSTRACT

B cells constitute an essential line of defense from pathogenic infections through the generation of class-switched antibody-secreting cells (ASCs) in germinal centers. Although this process is known to be regulated by follicular helper T (TfH) cells, the mechanism by which B cells initially seed germinal center reactions remains elusive. We found that NKT cells, a population of innate-like T lymphocytes, are critical for the induction of B cell immunity upon viral infection. The positioning of NKT cells at the interfollicular areas of lymph nodes facilitates both their direct priming by resident macrophages and the localized delivery of innate signals to antigen-experienced B cells. Indeed, NKT cells secrete an early wave of IL-4 and constitute up to 70% of the total IL-4-producing cells during the initial stages of infection. Importantly, the requirement of this innate immunity arm appears to be evolutionarily conserved because early NKT and IL-4 gene signatures also positively correlate with the levels of neutralizing antibodies in Zika-virus-infected macaques. In conclusion, our data support a model wherein a pre-TfH wave of IL-4 secreted by interfollicular NKT cells triggers the seeding of germinal center cells and serves as an innate link between viral infection and B cell immunity.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Innate , Influenza, Human/immunology , Interleukin-4/genetics , Killer Cells, Natural/immunology , Zika Virus Infection/immunology , Animals , Chickens , Dogs , Germinal Center/cytology , Humans , Interleukin-4/metabolism , Macaca , Macrophages/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL
13.
J Immunol ; 199(5): 1682-1695, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28747344

ABSTRACT

Rho family GTPases regulate diverse cellular events, such as cell motility, polarity, and vesicle traffic. Although a wealth of data exists on the canonical Rho GTPases RhoA, Rac1, and Cdc42, several other family members remain poorly studied. In B cells, we recently demonstrated a critical role for Cdc42 in plasma cell differentiation. In this study, we focus on a close homolog of Cdc42, TC10 (also known as RhoQ), and investigate its physiological role in B cells. By generating a TC10-deficient mouse model, we show that despite reduced total B cell numbers, B cell development in these mice occurs normally through distinct developmental stages. Upon immunization, IgM levels were reduced and, upon viral infection, germinal center responses were defective in TC10-deficient mice. BCR signaling was mildly affected, whereas cell migration remained normal in TC10-deficient B cells. Furthermore, by generating a TC10/Cdc42 double knockout mouse model, we found that TC10 can compensate for the lack of Cdc42 in TLR-induced cell activation and proliferation, so the two proteins play partly redundant roles. Taken together, by combining in vivo and in vitro analysis using TC10-deficient mice, we define the poorly studied Rho GTPase TC10 as an immunomodulatory molecule playing a role in physiological B cell responses.


Subject(s)
B-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Vaccinia/immunology , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Germinal Center/immunology , Immunomodulation , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics
14.
Immunity ; 46(2): 163-164, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228271

ABSTRACT

Encounters between naive T lymphocytes and dendritic cells (DCs) bearing adequate co-stimulatory signals are rare. In this issue of Immunity, Brewitz et al. (2017) show that chemokines secreted by CD8+ T cells recruit myeloid and plasmacytoid DCs that in turn boost CD8+ T cell activation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Antigens , Chemokines , Lymphocyte Activation/immunology
15.
Science ; 355(6325): 641-647, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183981

ABSTRACT

Autophagy is important in a variety of cellular and pathophysiological situations; however, its role in immune responses remains elusive. Here, we show that among B cells, germinal center (GC) cells exhibited the highest rate of autophagy during viral infection. In contrast to mechanistic target of rapamycin complex 1-dependent canonical autophagy, GC B cell autophagy occurred predominantly through a noncanonical pathway. B cell stimulation was sufficient to down-regulate canonical autophagy transiently while triggering noncanonical autophagy. Genetic ablation of WD repeat domain, phosphoinositide-interacting protein 2 in B cells alone enhanced this noncanonical autophagy, resulting in changes of mitochondrial homeostasis and alterations in GC and antibody-secreting cells. Thus, B cell activation prompts a temporal switch from canonical to noncanonical autophagy that is important in controlling B cell differentiation and fate.


Subject(s)
Autophagy/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Virus Diseases/immunology , Animals , Down-Regulation , Germinal Center/immunology , Germinal Center/virology , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , WD40 Repeats/genetics
16.
Immunity ; 43(4): 660-73, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26453379

ABSTRACT

Humans with Wiskott-Aldrich syndrome display a progressive immunological disorder associated with compromised Wiskott-Aldrich Syndrome Interacting Protein (WIP) function. Mice deficient in WIP recapitulate such an immunodeficiency that has been attributed to T cell dysfunction; however, any contribution of B cells is as yet undefined. Here we have shown that WIP deficiency resulted in defects in B cell homing, chemotaxis, survival, and differentiation, ultimately leading to diminished germinal center formation and antibody production. Furthermore, in the absence of WIP, several receptors, namely the BCR, BAFFR, CXCR4, CXCR5, CD40, and TLR4, were impaired in promoting CD19 co-receptor activation and subsequent PI3 kinase (PI3K) signaling. The underlying mechanism was due to a distortion in the actin and tetraspanin networks that lead to altered CD19 cell surface dynamics. In conclusion, our findings suggest that, by regulating the cortical actin cytoskeleton, WIP influences the function of CD19 as a general hub for PI3K signaling.


Subject(s)
Antigens, CD19/physiology , B-Lymphocytes/immunology , Carrier Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology , Actin Cytoskeleton/ultrastructure , Actins/analysis , Animals , Antibody Formation , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/ultrastructure , Carrier Proteins/genetics , Cells, Cultured , Chemokines/pharmacology , Chemokines/physiology , Chemotaxis/drug effects , Cytoskeletal Proteins , Germinal Center/immunology , Germinal Center/pathology , Haptens , Hemocyanins/pharmacology , Lymphocyte Activation/drug effects , Lymphopoiesis , Membrane Proteins/immunology , Mice , Phosphorylation , Plasma Cells/immunology , Protein Processing, Post-Translational , Radiation Chimera , Receptors, Antigen, B-Cell/immunology , Receptors, Chemokine/physiology , Tetraspanins/analysis , Vaccinia/immunology , Vaccinia/pathology
17.
J Exp Med ; 212(1): 53-72, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25547673

ABSTRACT

The small Rho GTPase Cdc42, known to interact with Wiskott-Aldrich syndrome (WAS) protein, is an important regulator of actin remodeling. Here, we show that genetic ablation of Cdc42 exclusively in the B cell lineage is sufficient to render mice unable to mount antibody responses. Indeed Cdc42-deficient mice are incapable of forming germinal centers or generating plasma B cells upon either viral infection or immunization. Such severe immune deficiency is caused by multiple and profound B cell abnormalities, including early blocks during B cell development; impaired antigen-driven BCR signaling and actin remodeling; defective antigen presentation and in vivo interaction with T cells; and a severe B cell-intrinsic block in plasma cell differentiation. Thus, our study presents a new perspective on Cdc42 as key regulator of B cell physiology.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Immunity, Humoral/immunology , Orthomyxoviridae Infections/immunology , cdc42 GTP-Binding Protein/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Gene Expression/immunology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral/genetics , Influenza A virus/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL