Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 453
Filter
1.
Methods Mol Biol ; 2826: 117-129, 2024.
Article in English | MEDLINE | ID: mdl-39017889

ABSTRACT

Memory B cells are central to the establishment of immunological memory, providing long-term protection against specific pathogens and playing a vital role in the efficacy of vaccines. Understanding how memory B cell formation is disrupted during persistent infection is essential for new therapeutics. Lymphocytic choriomeningitis virus (LCMV) is an ideal model for investigating memory B cells in acute versus chronic infection. This protocol details techniques to isolate, enrich, and examine LCMV-specific memory B cells in both acute and chronic LCMV infection. Using an antigen tetramer enrichment system and flow cytometry, this method assesses low-frequency, polyclonal antigen-specific memory B cells.


Subject(s)
Antigens, Viral , Flow Cytometry , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Memory B Cells , Lymphocytic choriomeningitis virus/immunology , Animals , Mice , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Flow Cytometry/methods , Antigens, Viral/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Immunologic Memory , B-Lymphocytes/immunology , B-Lymphocytes/metabolism
3.
Viruses ; 16(5)2024 05 17.
Article in English | MEDLINE | ID: mdl-38793680

ABSTRACT

Immunotherapy with checkpoint inhibitors, albeit commonly used against tumors, is still at its infancy against chronic virus infections. It relies on the reinvigoration of exhausted T lymphocytes to eliminate virus-infected cells. Since T cell exhaustion is a physiological process to reduce immunopathology, the reinvigoration of these cells might be associated with an augmentation of pathological changes. To test this possibility, we here analyzed in the model system of chronic lymphocytic choriomeningitis virus (LCMV)-infected mice whether treatment with the checkpoint inhibitor anti-PD-L1 antibody would increase CD8 T cell-dependent fibrosis. We show that pre-existing spleen fibrosis did not worsen under conditions that increase CD8 T cell functionality and reduce virus loads suggesting that the CD8 T cell functionality increase remained below its pathogenicity threshold. These promising findings should further encourage immunotherapeutic trials against chronic virus infections.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Fibrosis , Immune Checkpoint Inhibitors , Immunotherapy , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Animals , Mice , Lymphocytic choriomeningitis virus/immunology , Immunotherapy/methods , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic Choriomeningitis/therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Viral Load , Spleen/immunology , Spleen/virology , Disease Models, Animal , Chronic Disease , Female
4.
Front Immunol ; 15: 1399989, 2024.
Article in English | MEDLINE | ID: mdl-38799448

ABSTRACT

Introduction: Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods: Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results: Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119 + P2ry12 + microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion: Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease.


Subject(s)
Brain , Microglia , Retina , Animals , Microglia/immunology , Microglia/metabolism , Mice , Retina/immunology , Retina/pathology , Brain/immunology , Brain/pathology , Brain/metabolism , Mice, Inbred C57BL , Lymphocytic choriomeningitis virus/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Single-Cell Analysis , CD8-Positive T-Lymphocytes/immunology , Transcriptome
5.
J Vis Exp ; (206)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38738889

ABSTRACT

Follicular Helper T (TFH) cells are perceived as an independent CD4+ T cell lineage that assists cognate B cells in producing high-affinity antibodies, thus establishing long-term humoral immunity. During acute viral infection, the fate commitment of virus-specific TFH cells is determined in the early infection phase, and investigations of the early-differentiated TFH cells are crucial in understanding T cell-dependent humoral immunity and optimizing vaccine design. In the study, using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection and the TCR-transgenic SMARTA (SM) mouse with CD4+ T cells specifically recognizing LCMV glycoprotein epitope I-AbGP66-77, we described procedures to access the early fate commitment of virus-specific TFH cells based on flow cytometry stainings. Furthermore, by exploiting retroviral transduction of SM CD4+ T cells, methods to manipulate gene expression in early-differentiated virus-specific TFH cells are also provided. Hence, these methods will help in studies exploring the mechanism(s) underlying the early commitment of virus-specific TFH cells.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Differentiation , Lymphocytic Choriomeningitis , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Flow Cytometry/methods , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Mice, Transgenic , T Follicular Helper Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology
6.
J Immunol ; 212(11): 1829-1842, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38619295

ABSTRACT

In response to acute infection, naive CD4+ T cells primarily differentiate into T helper 1 (Th1) or T follicular helper (Tfh) cells that play critical roles in orchestrating cellular or humoral arms of immunity, respectively. However, despite the well established role of T-bet and BCL-6 in driving Th1 and Tfh cell lineage commitment, respectively, whether additional transcriptional circuits also underlie the fate bifurcation of Th1 and Tfh cell subsets is not fully understood. In this article, we study how the transcriptional regulator Bhlhe40 dictates the Th1/Tfh differentiation axis in mice. CD4+ T cell-specific deletion of Bhlhe40 abrogates Th1 but augments Tfh differentiation. We also assessed an increase in germinal center B cells and Ab production, suggesting that deletion of Bhlhe40 in CD4+ T cells not only alters Tfh differentiation but also their capacity to provide help to B cells. To identify molecular mechanisms by which Bhlhe40 regulates Th1 versus Tfh lineage choice, we first performed epigenetic profiling in the virus specific Th1 and Tfh cells following LCMV infection, which revealed distinct promoter and enhancer activities between the two helper cell lineages. Furthermore, we identified that Bhlhe40 directly binds to cis-regulatory elements of Th1-related genes such as Tbx21 and Cxcr6 to activate their expression while simultaneously binding to regions of Tfh-related genes such as Bcl6 and Cxcr5 to repress their expression. Collectively, our data suggest that Bhlhe40 functions as a transcription activator to promote Th1 cell differentiation and a transcription repressor to suppress Tfh cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , T Follicular Helper Cells , Th1 Cells , Animals , Mice , Cell Differentiation/immunology , Cell Differentiation/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , T Follicular Helper Cells/immunology , Th1 Cells/immunology , Mice, Knockout , Mice, Inbred C57BL , B-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Germinal Center/immunology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Lymphocytic choriomeningitis virus/immunology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Homeodomain Proteins
7.
Cell Rep ; 43(5): 114156, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38687642

ABSTRACT

The maintenance of antigen-specific CD8+ T cells underlies the efficacy of vaccines and immunotherapies. Pathways contributing to CD8+ T cell loss are not completely understood. Uncovering the pathways underlying the limited persistence of CD8+ T cells would be of significant benefit for developing novel strategies of promoting T cell persistence. Here, we demonstrate that murine CD8+ T cells experience endoplasmic reticulum (ER) stress following activation and that the ER-associated degradation (ERAD) adapter Sel1L is induced in activated CD8+ T cells. Sel1L loss limits CD8+ T cell function and memory formation following acute viral infection. Mechanistically, Sel1L is required for optimal bioenergetics and c-Myc expression. Finally, we demonstrate that human CD8+ T cells experience ER stress upon activation and that ER stress is negatively associated with improved T cell functionality in T cell-redirecting therapies. Together, these results demonstrate that ER stress and ERAD are important regulators of T cell function and persistence.


Subject(s)
CD8-Positive T-Lymphocytes , Endoplasmic Reticulum Stress , Endoplasmic Reticulum-Associated Degradation , Immunologic Memory , Animals , Humans , Mice , Acute Disease , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Intracellular Signaling Peptides and Proteins , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic Choriomeningitis/pathology , Mice, Inbred C57BL , Proteins , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Male , Female
8.
Viruses ; 16(4)2024 04 19.
Article in English | MEDLINE | ID: mdl-38675975

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) and Lassa virus (LASV) share many genetic and biological features including subtle differences between pathogenic and apathogenic strains. Despite remarkable genetic similarity, the viscerotropic WE strain of LCMV causes a fatal LASV fever-like hepatitis in non-human primates (NHPs) while the mouse-adapted Armstrong (ARM) strain of LCMV is deeply attenuated in NHPs and can vaccinate against LCMV-WE challenge. Here, we demonstrate that internalization of WE is more sensitive to the depletion of membrane cholesterol than ARM infection while ARM infection is more reliant on endosomal acidification. LCMV-ARM induces robust NF-κB and interferon response factor (IRF) activation while LCMV-WE seems to avoid early innate sensing and failed to induce strong NF-κB and IRF responses in dual-reporter monocyte and epithelial cells. Toll-like receptor 2 (TLR-2) signaling appears to play a critical role in NF-κB activation and the silencing of TLR-2 shuts down IL-6 production in ARM but not in WE-infected cells. Pathogenic LCMV-WE infection is poorly recognized in early endosomes and failed to induce TLR-2/Mal-dependent pro-inflammatory cytokines. Following infection, Interleukin-1 receptor-associated kinase 1 (IRAK-1) expression is diminished in LCMV-ARM- but not LCMV-WE-infected cells, which indicates it is likely involved in the LCMV-ARM NF-κB activation. By confocal microscopy, ARM and WE strains have similar intracellular trafficking although LCMV-ARM infection appears to coincide with greater co-localization of early endosome marker EEA1 with TLR-2. Both strains co-localize with Rab-7, a late endosome marker, but the interaction with LCMV-WE seems to be more prolonged. These findings suggest that LCMV-ARM's intracellular trafficking pathway may facilitate interaction with innate immune sensors, which promotes the induction of effective innate and adaptive immune responses.


Subject(s)
Immunity, Innate , Lymphocytic choriomeningitis virus , Virus Internalization , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Lymphocytic choriomeningitis virus/physiology , Animals , Humans , Mice , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Endosomes/metabolism , NF-kappa B/metabolism , Signal Transduction , Cell Line , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Epithelial Cells/virology , Epithelial Cells/immunology
9.
Immunity ; 57(5): 1037-1055.e6, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38593796

ABSTRACT

Memory B cells (MBCs) are key providers of long-lived immunity against infectious disease, yet in chronic viral infection, they do not produce effective protection. How chronic viral infection disrupts MBC development and whether such changes are reversible remain unknown. Through single-cell (sc)ATAC-seq and scRNA-seq during acute versus chronic lymphocytic choriomeningitis viral infection, we identified a memory subset enriched for interferon (IFN)-stimulated genes (ISGs) during chronic infection that was distinct from the T-bet+ subset normally associated with chronic infection. Blockade of IFNAR-1 early in infection transformed the chromatin landscape of chronic MBCs, decreasing accessibility at ISG-inducing transcription factor binding motifs and inducing phenotypic changes in the dominating MBC subset, with a decrease in the ISG subset and an increase in CD11c+CD80+ cells. However, timing was critical, with MBCs resistant to intervention at 4 weeks post-infection. Together, our research identifies a key mechanism to instruct MBC identity during viral infection.


Subject(s)
Epigenesis, Genetic , Interferon Type I , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Memory B Cells , Animals , Interferon Type I/metabolism , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Mice , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/genetics , Immunologic Memory/immunology , Chronic Disease , B-Lymphocyte Subsets/immunology , Single-Cell Analysis
10.
Front Immunol ; 15: 1374617, 2024.
Article in English | MEDLINE | ID: mdl-38665911

ABSTRACT

Blindness or vision loss due to neuroretinal and photoreceptor degeneration affects millions of individuals worldwide. In numerous neurodegenerative diseases, including age-related macular degeneration, dysregulated immune response-mediated retinal degeneration has been found to play a critical role in the disease pathogenesis. To better understand the pathogenic mechanisms underlying the retinal degeneration, we used a mouse model of systemic immune activation where we infected mice with lymphocytic choriomeningitis virus (LCMV) clone 13. Here, we evaluated the effects of LCMV infection and present a comprehensive discovery-based proteomic investigation using tandem mass tag (TMT) labeling and high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS). Changes in protein regulation in the posterior part of the eye, neuroretina, and RPE/choroid were compared to those in the spleen as a secondary lymphoid organ and to the kidney as a non-lymphoid but encapsulated organ at 1, 8, and 28 weeks of infection. Using bioinformatic tools, we found several proteins responsible for maintaining normal tissue homeostasis to be differentially regulated in the neuroretina and the RPE/choroid during the degenerative process. Additionally, in the organs we observed, several important protein pathways contributing to cellular homeostasis and tissue development were perturbed and associated with LCMV-mediated inflammation, promoting disease progression. Our findings suggest that the response to a systemic chronic infection differs between the neuroretina and the RPE/choroid, and the processes induced by chronic systemic infection in the RPE/choroid are not unlike those induced in non-immune-privileged organs such as the kidney and spleen. Overall, our data provide detailed insight into several molecular mechanisms of neuroretinal degeneration and highlight various novel protein pathways that further suggest that the posterior part of the eye is not an isolated immunological entity despite the existence of neuroretinal immune privilege.


Subject(s)
Disease Models, Animal , Lymphocytic choriomeningitis virus , Proteomics , Retinal Degeneration , Animals , Mice , Proteomics/methods , Retinal Degeneration/immunology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Tandem Mass Spectrometry , Proteome , Retina/immunology , Retina/metabolism , Retina/pathology , Chromatography, Liquid , Choroid/immunology , Choroid/pathology , Choroid/metabolism
11.
Nature ; 627(8003): 399-406, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448581

ABSTRACT

Immune cells rely on transient physical interactions with other immune and non-immune populations to regulate their function1. To study these 'kiss-and-run' interactions directly in vivo, we previously developed LIPSTIC (labelling immune partnerships by SorTagging intercellular contacts)2, an approach that uses enzymatic transfer of a labelled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ T helper cells and antigen-presenting cells, however. Here we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the steady-state cellular partners of regulatory T cells and identify germinal centre-resident T follicular helper cells on the basis of their ability to interact cognately with germinal centre B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalogue of the immune populations that physically interact with intestinal epithelial cells at the steady state and profile the evolution of the interactome of lymphocytic choriomeningitis virus-specific CD8+ T cells in multiple organs following systemic infection. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.


Subject(s)
B-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Communication , Dendritic Cells , Epithelial Cells , T Follicular Helper Cells , T-Lymphocytes, Regulatory , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Ligands , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T Follicular Helper Cells/cytology , T Follicular Helper Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Germinal Center/cytology , Single-Cell Gene Expression Analysis , Epithelial Cells/cytology , Epithelial Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Organ Specificity
12.
Immunohorizons ; 6(12): 872-882, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36547389

ABSTRACT

CD8 cytotoxic T cells are a potent line of defense against invading pathogens. To aid in curtailing aberrant immune responses, the activation status of CD8 T cells is highly regulated. One mechanism in which CD8 T cell responses are dampened is via signaling through the immune-inhibitory receptor Programmed Cell Death Protein-1, encoded by Pdcd1. Pdcd1 expression is regulated through engagement of the TCR, as well as by signaling from extracellular cytokines. Understanding such pathways has influenced the development of numerous clinical treatments. In this study, we showed that signals from the cytokine IL-6 enhanced Pdcd1 expression when paired with TCR stimulation in murine CD8 T cells. Mechanistically, signals from IL-6 were propagated through activation of the transcription factor STAT3, resulting in IL-6-dependent binding of STAT3 to Pdcd1 cis-regulatory elements. Intriguingly, IL-6 stimulation overcame B Lymphocyte Maturation Protein 1-mediated epigenetic repression of Pdcd1, which resulted in a transcriptionally permissive landscape marked by heightened histone acetylation. Furthermore, in vivo-activated CD8 T cells derived from lymphocytic choriomeningitis virus infection required STAT3 for optimal Programmed Cell Death Protein-1 surface expression. Importantly, STAT3 was the only member of the STAT family present at Pdcd1 regulatory elements in lymphocytic choriomeningitis virus Ag-specific CD8 T cells. Collectively, these data define mechanisms by which the IL-6/STAT3 signaling axis can enhance and prolong Pdcd1 expression in murine CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-6 , Programmed Cell Death 1 Receptor , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Interleukin-6/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Lymphocytic choriomeningitis virus/immunology
13.
Virology ; 567: 77-86, 2022 02.
Article in English | MEDLINE | ID: mdl-35032866

ABSTRACT

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Subject(s)
Atrophy/virology , Interferon-alpha/immunology , Interferon-beta/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Thymocytes/virology , Thymus Gland/virology , Animals , Atrophy/genetics , Atrophy/immunology , Atrophy/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Female , Gene Expression Regulation , Humans , Immunologic Memory , Interferon-alpha/genetics , Interferon-beta/genetics , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Single-Cell Analysis , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology
15.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34882194

ABSTRACT

Pregnancy is a common immunization event, but the molecular mechanisms and immunological consequences provoked by pregnancy remain largely unknown. We used mouse models and human transplant registry data to reveal that pregnancy induced exhausted CD8 T cells (Preg-TEX), which associated with prolonged allograft survival. Maternal CD8 T cells shared features of exhaustion with CD8 T cells from cancer and chronic infection, including transcriptional down-regulation of ribosomal proteins and up-regulation of TOX and inhibitory receptors. Similar to other models of T cell exhaustion, NFAT-dependent elements of the exhaustion program were induced by fetal antigen in pregnancy, whereas NFAT-independent elements did not require fetal antigen. Despite using conserved molecular circuitry, Preg-TEX cells differed from TEX cells in chronic viral infection with respect to magnitude and dependency of T cell hypofunction on NFAT-independent signals. Altogether, these data reveal the molecular mechanisms and clinical consequences of maternal CD8 T cell hypofunction and identify pregnancy as a previously unappreciated context in which T cell exhaustion may occur.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Profiling/methods , Lymphocyte Activation/immunology , NFATC Transcription Factors/immunology , Adoptive Transfer , Animals , Antigens/immunology , CD8-Positive T-Lymphocytes/metabolism , Chlorocebus aethiops , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Kaplan-Meier Estimate , Lymphocyte Activation/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Pregnancy , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Skin Transplantation , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Vero Cells
16.
Cell Rep ; 37(9): 110064, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852223

ABSTRACT

CD4+ T cells have a remarkable potential to differentiate into diverse effector lineages following activation. Here, we probe the heterogeneity present among naive CD4+ T cells before encountering their cognate antigen to ask whether their effector potential is modulated by pre-existing transcriptional and chromatin landscape differences. Single-cell RNA sequencing shows that key drivers of variability are genes involved in T cell receptor (TCR) signaling. Using CD5 expression as a readout of the strength of tonic TCR interactions with self-peptide MHC, and sorting on the ends of this self-reactivity spectrum, we find that pre-existing transcriptional differences among naive CD4+ T cells impact follicular helper T (TFH) cell versus non-TFH effector lineage choice. Moreover, our data implicate TCR signal strength during thymic development in establishing differences in naive CD4+ T cell chromatin landscapes that ultimately shape their effector potential.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Chromatin/physiology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Female , Gene Expression Profiling , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Male , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism
17.
Cell Rep ; 37(9): 110061, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852228

ABSTRACT

Passive antibody therapy and vectored antibody gene delivery (VAGD) in particular offer an innovative approach to combat persistent viral diseases. Here, we exploit a small animal model to investigate synergies of VAGD with the host's endogenous immune defense for treating chronic viral infection. An adeno-associated virus (AAV) vector delivering the lymphocytic choriomeningitis virus (LCMV)-neutralizing antibody KL25 (AAV-KL25) establishes protective antibody titers for >200 days. When therapeutically administered to chronically infected immunocompetent wild-type mice, AAV-KL25 affords sustained viral load control. In contrast, viral mutational escape thwarts therapeutic AAV-KL25 effects when mice are unable to mount LCMV-specific antibody responses or lack CD8+ T cells. VAGD augments antiviral germinal center B cell and antibody-secreting cell responses and reduces inhibitory receptor expression on antiviral CD8+ T cells. These results indicate that VAGD fortifies host immune defense and synergizes with B cell and CD8 T cell responses to restore immune control of chronic viral infection.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, Immunoglobulin , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Germinal Center , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Viral Load
18.
Nat Immunol ; 22(12): 1524-1537, 2021 12.
Article in English | MEDLINE | ID: mdl-34795443

ABSTRACT

Inhibiting PD-1:PD-L1 signaling has transformed therapeutic immune restoration. CD4+ T cells sustain immunity in chronic infections and cancer, yet little is known about how PD-1 signaling modulates CD4+ helper T (TH) cell responses or the ability to restore CD4+ TH-mediated immunity by checkpoint blockade. We demonstrate that PD-1:PD-L1 specifically suppressed CD4+ TH1 cell amplification, prevents CD4+ TH1 cytokine production and abolishes CD4+ cytotoxic killing capacity during chronic infection in mice. Inhibiting PD-L1 rapidly restored these functions, while simultaneously amplifying and activating TH1-like T regulatory cells, demonstrating a system-wide CD4-TH1 recalibration. This effect coincided with decreased T cell antigen receptor signaling, and re-directed type I interferon (IFN) signaling networks towards dominant IFN-γ-mediated responses. Mechanistically, PD-L1 blockade specifically targeted defined populations with pre-established, but actively suppressed proliferative potential, with limited impact on minimally cycling TCF-1+ follicular helper T cells, despite high PD-1 expression. Thus, CD4+ T cells require unique differentiation and functional states to be targets of PD-L1-directed suppression and therapeutic restoration.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation/drug effects , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic choriomeningitis virus/immunology , Th1 Cells/drug effects , Adoptive Transfer , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Chronic Disease , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Female , Gene Regulatory Networks , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Transcriptome
19.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34709350

ABSTRACT

Chronic viral infections are associated with hematopoietic suppression, bone marrow (BM) failure, and hematopoietic stem cell (HSC) exhaustion. However, how persistent viral challenge and inflammatory responses target BM tissues and perturb hematopoietic competence remains poorly understood. Here, we combine functional analyses with advanced 3D microscopy to demonstrate that chronic infection with lymphocytic choriomeningitis virus leads to (1) long-lasting decimation of the BM stromal network of mesenchymal CXCL12-abundant reticular cells, (2) proinflammatory transcriptional remodeling of remaining components of this key niche subset, and (3) durable functional defects and decreased competitive fitness in HSCs. Mechanistically, BM immunopathology is elicited by virus-specific, activated CD8 T cells, which accumulate in the BM via interferon-dependent mechanisms. Combined antibody-mediated inhibition of type I and II IFN pathways completely preempts degeneration of CARc and protects HSCs from chronic dysfunction. Hence, viral infections and ensuing immune reactions durably impact BM homeostasis by persistently decreasing the competitive fitness of HSCs and disrupting essential stromal-derived, hematopoietic-supporting cues.


Subject(s)
Bone Marrow/virology , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Lymphocytic Choriomeningitis/pathology , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chronic Disease , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Interferons/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Mice, Inbred C57BL , Mice, Mutant Strains , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism
20.
Viruses ; 13(10)2021 09 29.
Article in English | MEDLINE | ID: mdl-34696381

ABSTRACT

Ever since the immune regulatory strains of lymphocytic choriomeningitis virus (LCMV), such as Clone 13, were isolated, LCMV infection of mice has served as a valuable model for the mechanistic study of viral immune suppression and virus persistence. The exhaustion of virus-specific T cells was demonstrated during LCMV infection, and the underlying mechanisms have been extensively investigated using LCMV infection in mouse models. In particular, the mechanism for gradual CD8+ T cell exhaustion at molecular and transcriptional levels has been investigated. These studies revealed crucial roles for inhibitory receptors, surface markers, regulatory cytokines, and transcription factors, including PD-1, PSGL-1, CXCR5, and TOX in the regulation of T cells. However, the action mode for CD4+ T cell suppression is largely unknown. Recently, sphingosine kinase 2 was proven to specifically repress CD4+ T cell proliferation and lead to LCMV persistence. As CD4+ T cell regulation was also known to be important for viral persistence, research to uncover the mechanism for CD4+ T cell repression could help us better understand how viruses launch and prolong their persistence. This review summarizes discoveries derived from the study of LCMV in regard to the mechanisms for T cell suppression and approaches for the termination of viral persistence with special emphasis on CD8+ T cells.


Subject(s)
Host Microbial Interactions/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Persistent Infection/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Humans , Lymphocyte Activation , Lymphocytic Choriomeningitis/virology , Mice , Persistent Infection/virology , T-Lymphocytes/classification
SELECTION OF CITATIONS
SEARCH DETAIL