Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 25(2): 256-267, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38172258

ABSTRACT

The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Animals , Humans , Mice , Alarmins , Antiviral Agents , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , T-Lymphocyte Subsets/metabolism
2.
Cell ; 175(2): 458-471.e19, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173917

ABSTRACT

Inflammatory disorders of the CNS are frequently accompanied by synaptic loss, which is thought to involve phagocytic microglia and complement components. However, the mechanisms accounting for aberrant synaptic connectivity in the context of CD8+ T cell-driven neuronal damage are poorly understood. Here, we profiled the neuronal translatome in a murine model of encephalitis caused by CD8+ T cells targeting antigenic neurons. Neuronal STAT1 signaling and downstream CCL2 expression were essential for apposition of phagocytes, ensuing synaptic loss and neurological disease. Analogous observations were made in the brains of Rasmussen's encephalitis patients. In this devastating CD8+ T cell-driven autoimmune disease, neuronal STAT1 phosphorylation and CCL2 expression co-clustered with infiltrating CD8+ T cells as well as phagocytes. Taken together, our findings uncover an active role of neurons in coordinating phagocyte-mediated synaptic loss and highlight neuronal STAT1 and CCL2 as critical steps in this process that are amenable to pharmacological interventions.


Subject(s)
Neurons/metabolism , Phagocytosis/physiology , Synapses/physiology , Animals , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Encephalitis/physiopathology , Female , Humans , Inflammation/immunology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nervous System Diseases/metabolism , Neurons/physiology , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/immunology , Phosphorylation , STAT1 Transcription Factor/physiology , Transcriptome/genetics
3.
Immunity ; 56(4): 813-828.e10, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36809763

ABSTRACT

T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-33 , Lymphocytic Choriomeningitis , Animals , Mice , Alarmins/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Persistent Infection , T Cell Transcription Factor 1/metabolism
4.
Immunity ; 48(5): 937-950.e8, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768177

ABSTRACT

Infections are thought to trigger CD8+ cytotoxic T lymphocyte (CTL) responses during autoimmunity. However, the transcriptional programs governing the tissue-destructive potential of CTLs remain poorly defined. In a model of central nervous system (CNS) inflammation, we found that infection with lymphocytic choriomeningitis virus (LCMV), but not Listeria monocytogenes (Lm), drove autoimmunity. The DNA-binding factor TOX was induced in CTLs during LCMV infection and was essential for their encephalitogenic properties, and its expression was inhibited by interleukin-12 during Lm infection. TOX repressed the activity of several transcription factors (including Id2, TCF-1, and Notch) that are known to drive CTL differentiation. TOX also reduced immune checkpoint sensitivity by restraining the expression of the inhibitory checkpoint receptor CD244 on the surface of CTLs, leading to increased CTL-mediated damage in the CNS. Our results identify TOX as a transcriptional regulator of tissue-destructive CTLs in autoimmunity, offering a potential mechanistic link to microbial triggers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeodomain Proteins/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adult , Aged , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
Proc Natl Acad Sci U S A ; 121(11): e2318657121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38446855

ABSTRACT

Viral mimicry of host cell structures has been postulated to curtail the B cell receptor (BCR) repertoire against persisting viruses through tolerance mechanisms. This concept awaits, however, experimental testing in a setting of natural virus-host relationship. We engineered mouse models expressing a monoclonal BCR specific for the envelope glycoprotein of lymphocytic choriomeningitis virus (LCMV), a naturally persisting mouse pathogen. When the heavy chain of the LCMV-neutralizing antibody KL25 was paired with its unmutated ancestor light chain, most B cells underwent receptor editing, a behavior reminiscent of autoreactive clones. In contrast, monoclonal B cells expressing the same heavy chain in conjunction with the hypermutated KL25 light chain did not undergo receptor editing but exhibited low levels of surface IgM, suggesting that light chain hypermutation had lessened KL25 autoreactivity. Upon viral challenge, these IgMlow cells were not anergic but up-regulated IgM, participated in germinal center reactions, produced antiviral antibodies, and underwent immunoglobulin class switch as well as further affinity maturation. These studies on a persisting virus in its natural host species suggest that central tolerance mechanisms prune the protective antiviral B cell repertoire.


Subject(s)
B-Lymphocytes , Central Tolerance , Animals , Mice , Antibodies, Viral , Lymphocytic choriomeningitis virus , Antiviral Agents , Immunoglobulin M
6.
Nature ; 574(7777): 200-205, 2019 10.
Article in English | MEDLINE | ID: mdl-31582858

ABSTRACT

The responses of CD8+ T cells to hepatotropic viruses such as hepatitis B range from dysfunction to differentiation into effector cells, but the mechanisms that underlie these distinct outcomes remain poorly understood. Here we show that priming by Kupffer cells, which are not natural targets of hepatitis B, leads to differentiation of CD8+ T cells into effector cells that form dense, extravascular clusters of immotile cells scattered throughout the liver. By contrast, priming by hepatocytes, which are natural targets of hepatitis B, leads to local activation and proliferation of CD8+ T cells but not to differentiation into effector cells; these cells form loose, intravascular clusters of motile cells that coalesce around portal tracts. Transcriptomic and chromatin accessibility analyses reveal unique features of these dysfunctional CD8+ T cells, with limited overlap with those of exhausted or tolerant T cells; accordingly, CD8+ T cells primed by hepatocytes cannot be rescued by treatment with anti-PD-L1, but instead respond to IL-2. These findings suggest immunotherapeutic strategies against chronic hepatitis B infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Hepatitis B virus/immunology , Hepatocytes/immunology , Hepatocytes/virology , Animals , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chromatin/metabolism , Female , Hepatitis B/drug therapy , Hepatitis B/immunology , Hepatitis B/virology , Humans , Immune Tolerance , Interleukin-2/immunology , Interleukin-2/therapeutic use , Kupffer Cells/immunology , Lymphocyte Activation , Male , Mice , Transcriptome/genetics
7.
Proc Natl Acad Sci U S A ; 119(25): e2202059119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35714289

ABSTRACT

The bacterial genus Bartonella comprises numerous emerging pathogens that cause a broad spectrum of disease manifestations in humans. The targets and mechanisms of the anti-Bartonella immune defense are ill-defined and bacterial immune evasion strategies remain elusive. We found that experimentally infected mice resolved Bartonella infection by mounting antibody responses that neutralized the bacteria, preventing their attachment to erythrocytes and suppressing bacteremia independent of complement or Fc receptors. Bartonella-neutralizing antibody responses were rapidly induced and depended on CD40 signaling but not on affinity maturation. We cloned neutralizing monoclonal antibodies (mAbs) and by mass spectrometry identified the bacterial autotransporter CFA (CAMP-like factor autotransporter) as a neutralizing antibody target. Vaccination against CFA suppressed Bartonella bacteremia, validating CFA as a protective antigen. We mapped Bartonella-neutralizing mAb binding to a domain in CFA that we found is hypervariable in both human and mouse pathogenic strains, indicating mutational antibody evasion at the Bartonella subspecies level. These insights into Bartonella immunity and immune evasion provide a conceptual framework for vaccine development, identifying important challenges in this endeavor.


Subject(s)
Antibodies, Neutralizing , Antigens, Bacterial , Bacteremia , Bartonella Infections , Bartonella , Type V Secretion Systems , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/prevention & control , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Bartonella/genetics , Bartonella/immunology , Bartonella Infections/immunology , Bartonella Infections/microbiology , Bartonella Infections/prevention & control , Cloning, Molecular , Immune Evasion , Mice , Type V Secretion Systems/immunology , Vaccination
8.
Eur J Immunol ; 53(8): e2350402, 2023 08.
Article in English | MEDLINE | ID: mdl-37179469

ABSTRACT

Previous studies that used peptide-MHC (pMHC) tetramers (tet) to identify self-specific T cells have questioned the effectiveness of thymic-negative selection. Here, we used pMHCI tet to enumerate CD8 T cells specific for the immunodominant gp33 epitope of lymphocytic choriomeningitis virus glycoprotein (GP) in mice transgenically engineered to express high levels of GP as a self-antigen in the thymus. In GP-transgenic mice (GP+ ), monoclonal P14 TCR+ CD8 T cells that express a GP-specific TCR could not be detected by gp33/Db -tet staining, indicative of their complete intrathymic deletion. By contrast, in the same GP+ mice, substantial numbers of polyclonal CD8 T cells identifiable by gp33/Db -tet were present. The gp33-tet staining profiles of polyclonal T cells from GP+ and GP-negative (GP- ) mice were overlapping, but mean fluorescence intensities were ∼15% lower in cells from GP+ mice. Remarkably, the gp33-tet+ T cells in GP+ mice failed to clonally expand after lymphocytic choriomeningitis virus infection, whereas those of GP- mice did so. In Nur77GFP -reporter mice, dose-dependent responses to gp33 peptide-induced TCR stimulation revealed that gp33-tet+ T cells with high ligand sensitivity are lacking in GP+ mice. Hence, pMHCI tet staining identifies self-specific CD8 T cells but tends to overestimate the number of truly self-reactive cells.


Subject(s)
Antigens, Viral , Viral Proteins , Mice , Animals , Receptors, Antigen, T-Cell/genetics , CD8-Positive T-Lymphocytes , Mice, Transgenic , Glycoproteins , Lymphocytic choriomeningitis virus , Peptides , Mice, Inbred C57BL
9.
PLoS Pathog ; 18(5): e1010489, 2022 05.
Article in English | MEDLINE | ID: mdl-35580143

ABSTRACT

Vertical transmission of Bartonella infection has been reported for several mammalian species including mice and humans. Accordingly, it is commonly held that acquired immunological tolerance contributes critically to the high prevalence of Bartonellae in wild-ranging rodent populations. Here we studied an experimental model of Bartonella infection in mice to assess the impact of maternal and newborn immune defense on vertical transmission and bacterial persistence in the offspring, respectively. Congenital infection was frequently observed in B cell-deficient mothers but not in immunocompetent dams, which correlated with a rapid onset of an antibacterial antibody response in infected WT animals. Intriguingly, B cell-deficient offspring with congenital infection exhibited long-term bacteremia whereas B cell-sufficient offspring cleared bacteremia within a few weeks after birth. Clearance of congenital Bartonella infection resulted in immunity against bacterial rechallenge, with the animals mounting Bartonella-neutralizing antibody responses of normal magnitude. These observations reveal a key role for humoral immune defense by the mother and offspring in preventing and eliminating vertical transmission. Moreover, congenital Bartonella infection does not induce humoral immune tolerance but results in anti-bacterial immunity, questioning the contribution of neonatal tolerance to Bartonella prevalence in wild-ranging rodents.


Subject(s)
Bacteremia , Bartonella Infections , Bartonella , Animals , Bacteremia/microbiology , Female , Infectious Disease Transmission, Vertical , Mammals , Mice , Placenta , Pregnancy
10.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Article in English | MEDLINE | ID: mdl-34772811

ABSTRACT

Chronic viral infections subvert protective B cell immunity. An early type I interferon (IFN-I)-driven bias to short-lived plasmablast differentiation leads to clonal deletion, so-called "decimation," of antiviral memory B cells. Therefore, prophylactic countermeasures against decimation remain an unmet need. We show that vaccination-induced CD4 T cells prevented the decimation of naïve and memory B cells in chronically lymphocytic choriomeningitis virus (LCMV)-infected mice. Although these B cell responses were largely T independent when IFN-I was blocked, preexisting T help assured their sustainability under conditions of IFN-I-driven inflammation by instructing a germinal center B cell transcriptional program. Prevention of decimation depended on T cell-intrinsic Bcl6 and Tfh progeny formation. Antigen presentation by B cells, interactions with antigen-specific T helper cells, and costimulation by CD40 and ICOS were also required. Importantly, B cell-mediated virus control averted Th1-driven immunopathology in LCMV-challenged animals with preexisting CD4 T cell immunity. Our findings show that vaccination-induced Tfh cells represent a cornerstone of effective B cell immunity to chronic virus challenge, pointing the way toward more effective B cell-based vaccination against persistent viral diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Persistent Infection/immunology , Vaccines/immunology , Virus Diseases/immunology , Animals , Antibodies, Viral/immunology , Antigen Presentation/immunology , Antiviral Agents/immunology , Cells, Cultured , Germinal Center/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Mice , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Vaccination/methods
12.
J Infect Dis ; 225(8): 1399-1410, 2022 04 19.
Article in English | MEDLINE | ID: mdl-32313928

ABSTRACT

BACKGROUND: A vaccine (HB-101) consisting of 2 nonreplicating lymphocytic choriomeningitis virus (LCMV) vectors expressing the human cytomegalovirus antigens glycoprotein B (gB) and the 65-kD phosphoprotein (pp65), respectively, is in development to prevent cytomegalovirus infection. METHODS: HB-101 was tested in cytomegalovirus-naive, healthy adults in a randomized, double-blind, placebo-controlled, dose-escalation Phase I trial. Fifty-four subjects received low, medium, or high dose of HB-101 or placebo by intramuscular administration at Month 0, 1, and 3. Safety and immunogenicity were the respective primary and secondary endpoints. Subjects were followed for 12 months after the initial immunization. RESULTS: Vaccination was associated with transient mild to moderate adverse events. HB-101 administration induced dose-dependent gB- and pp65-specific cellular responses, dominated by pp65-specific CD8 T cells, a high fraction of which were polyfunctional. Two administrations were sufficient to elicit dose-dependent gB-binding and cytomegalovirus-neutralizing antibodies (Abs). Cytomegalovirus-specific immune responses were boosted after each administration. Only 1 of 42 vaccine recipients mounted a transient LCMV vector-neutralizing Ab response. CONCLUSIONS: HB-101 was well tolerated and induced cytomegalovirus-specific polyfunctional CD8 T-cell and neutralizing Ab responses in the majority of subjects. Lack of vector-neutralizing Ab responses should facilitate booster vaccinations. These results justify further clinical evaluation of this vaccine candidate.


Subject(s)
Cytomegalovirus Vaccines , Vaccines , Adult , Antibodies, Neutralizing , Antibodies, Viral , Cytomegalovirus/genetics , Humans , Immunization, Secondary , Lymphocytic choriomeningitis virus/genetics
13.
Eur J Immunol ; 51(1): 76-90, 2021 01.
Article in English | MEDLINE | ID: mdl-32700362

ABSTRACT

Upon viral infection, stressed or damaged cells can release alarmins like IL-33 that act as endogenous danger signals alerting innate and adaptive immune cells. IL-33 coming from nonhematopoietic cells has been identified as important factor triggering the expansion of antiviral CD8+ T cells. In LN the critical cellular source of IL-33 is unknown, as is its potential cell-intrinsic function as a chromatin-associated factor. Using IL-33-GFP reporter mice, we identify fibroblastic reticular cells (FRC) and lymphatic endothelial cells (LEC) as the main IL-33 source. In homeostasis, IL-33 is dispensable as a transcriptional regulator in FRC, indicating it functions mainly as released cytokine. Early during infection with lymphocytic choriomeningitis virus (LCMV) clone 13, both FRC and LEC lose IL-33 protein expression suggesting cytokine release, correlating timewise with IL-33 receptor expression by reactive CD8+ T cells and their greatly augmented expansion in WT versus ll33-/- mice. Using mice lacking IL-33 selectively in FRC versus LEC, we identify FRC as key IL-33 source driving acute and chronic antiviral T-cell responses. Collectively, these findings show that LN T-zone FRC not only regulate the homeostasis of naïve T cells but also their expansion and differentiation several days into an antiviral response.


Subject(s)
Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Acute Disease , Adaptive Immunity , Animals , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Endothelial Cells/immunology , Fibroblasts/immunology , Homeostasis , Humans , Immunity, Innate , Interleukin-33/deficiency , Interleukin-33/genetics , Lymph Nodes/immunology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological
14.
Int J Mol Sci ; 23(5)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35269842

ABSTRACT

Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Animals , Antigens, Bacterial , BCG Vaccine , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Lymphocytic choriomeningitis virus/genetics , Mice , Mycobacterium tuberculosis/genetics
15.
Eur J Immunol ; 49(4): 626-637, 2019 04.
Article in English | MEDLINE | ID: mdl-30636035

ABSTRACT

Infection of C57BL/6 mice with lymphocytic choriomeningitis virus (LCMV) strain Armstrong (Arm) induces an acute infection with rapid virus clearance by CD8+ T cells independently of CD4+ T cell help. Residual viral antigen may, however, persist for a prolonged time. Here, we demonstrate that mice that had been transiently depleted of CD4+ T cells during acute LCMV Arm infection generated high levels of virus-specific IgG antibodies (Ab) after viral clearance. Robust induction of LCMV-specific IgG after transient CD4+ T cell depletion was dependent on Fcγ receptors but not on the complement receptors CD21/CD35. In contrast to the potent production of LCMV-specific IgG, the generation of LCMV-specific isotype-switched memory B cells after transient CD4+ T cell depletion was considerably reduced. Moreover, mice depleted of CD4+ T cells during acute infection were strongly impaired in generating a secondary LCMV-specific B cell response upon LCMV rechallenge. In conclusion, our data indicate that LCMV antigen depots after viral clearance were capable of inducing high levels of virus-specific IgG. They failed, however, to induce robust virus-specific B cell memory revealing a previously unappreciated dichotomy of specific Ab production and memory cell formation after priming with residual antigen.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Biomarkers , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunologic Memory , Immunophenotyping , Lymphocyte Depletion , Lymphocytic Choriomeningitis/virology , Mice , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology
16.
Proc Natl Acad Sci U S A ; 114(27): 7031-7036, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28630325

ABSTRACT

Transmission of hemorrhagic fever New World arenaviruses from their rodent reservoirs to human populations poses substantial public health and economic dangers. These zoonotic events are enabled by the specific interaction between the New World arenaviral attachment glycoprotein, GP1, and cell surface human transferrin receptor (hTfR1). Here, we present the structural basis for how a mouse-derived neutralizing antibody (nAb), OD01, disrupts this interaction by targeting the receptor-binding surface of the GP1 glycoprotein from Junín virus (JUNV), a hemorrhagic fever arenavirus endemic in central Argentina. Comparison of our structure with that of a previously reported nAb complex (JUNV GP1-GD01) reveals largely overlapping epitopes but highly distinct antibody-binding modes. Despite differences in GP1 recognition, we find that both antibodies present a key tyrosine residue, albeit on different chains, that inserts into a central pocket on JUNV GP1 and effectively mimics the contacts made by the host TfR1. These data provide a molecular-level description of how antibodies derived from different germline origins arrive at equivalent immunological solutions to virus neutralization.


Subject(s)
Antibodies, Neutralizing/immunology , Hemorrhagic Fever, American/immunology , Neutralization Tests , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Binding Sites , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Glycoproteins/chemistry , HEK293 Cells , Humans , Immune System , Junin virus , Protein Binding , Recombinant Proteins/immunology , Viral Envelope Proteins/chemistry
17.
PLoS Pathog ; 13(12): e1006758, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29261807

ABSTRACT

RNA-dependent RNA polymerases (RdRps) play a key role in the life cycle of RNA viruses and impact their immunobiology. The arenavirus lymphocytic choriomeningitis virus (LCMV) strain Clone 13 provides a benchmark model for studying chronic infection. A major genetic determinant for its ability to persist maps to a single amino acid exchange in the viral L protein, which exhibits RdRp activity, yet its functional consequences remain elusive. To unravel the L protein interactions with the host proteome, we engineered infectious L protein-tagged LCMV virions by reverse genetics. A subsequent mass-spectrometric analysis of L protein pulldowns from infected human cells revealed a comprehensive network of interacting host proteins. The obtained LCMV L protein interactome was bioinformatically integrated with known host protein interactors of RdRps from other RNA viruses, emphasizing interconnected modules of human proteins. Functional characterization of selected interactors highlighted proviral (DDX3X) as well as antiviral (NKRF, TRIM21) host factors. To corroborate these findings, we infected Trim21-/- mice with LCMV and found impaired virus control in chronic infection. These results provide insights into the complex interactions of the arenavirus LCMV and other viral RdRps with the host proteome and contribute to a better molecular understanding of how chronic viruses interact with their host.


Subject(s)
DEAD-box RNA Helicases/metabolism , Lymphocytic choriomeningitis virus/enzymology , Models, Molecular , RNA-Dependent RNA Polymerase/metabolism , Repressor Proteins/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Animals , CRISPR-Cas Systems , Computational Biology , Crosses, Genetic , DEAD-box RNA Helicases/chemistry , Female , HEK293 Cells , Humans , Immunoprecipitation , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/veterinary , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Specific Pathogen-Free Organisms , Viral Proteins/chemistry , Viral Proteins/genetics
18.
Immunity ; 32(1): 116-28, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20079668

ABSTRACT

Current T cell differentiation models invoke separate T helper 2 (Th2) and Th1 cell lineages governed by the lineage-specifying transcription factors GATA-3 and T-bet. However, knowledge on the plasticity of Th2 cell lineage commitment is limited. Here we show that infection with Th1 cell-promoting lymphocytic choriomeningitis virus (LCMV) reprogrammed otherwise stably committed GATA-3(+) Th2 cells to adopt a GATA-3(+)T-bet(+) and interleukin-4(+)interferon-gamma(+) "Th2+1" phenotype that was maintained in vivo for months. Th2 cell reprogramming required T cell receptor stimulation, concerted type I and type II interferon and interleukin-12 signals, and T-bet. LCMV-triggered T-bet induction in adoptively transferred virus-specific Th2 cells was crucial to prevent viral persistence and fatal immunopathology. Thus, functional reprogramming of unfavorably differentiated Th2 cells may facilitate the establishment of protective immune responses. Stable coexpression of GATA-3 and T-bet provides a molecular concept for the long-term coexistence of Th2 and Th1 cell lineage characteristics in single memory T cells.


Subject(s)
Cell Differentiation/immunology , Interferon-gamma/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/cytology , Th1 Cells/cytology , Th2 Cells/cytology , Adoptive Transfer , Animals , Cell Separation , Cytokines/immunology , Flow Cytometry , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Interferon-gamma/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Mol Ther ; 25(11): 2533-2545, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28803862

ABSTRACT

Successful vaccination against cancer and infectious diseases relies on the induction of adaptive immune responses that induce high-titer antibodies or potent cytoxic T cell responses. In contrast to humoral vaccines, the amplification of cellular immune responses is often hampered by anti-vector immunity that either pre-exists or develops after repeated homologous vaccination. Replication-defective lymphocytic choriomeningitis virus (LCMV) vectors represent a novel generation of vaccination vectors that induce potent immune responses while escaping recognition by neutralizing antibodies. Here, we characterize the CD8 T cell immune response induced by replication-defective recombinant LCMV (rLCMV) vectors with regard to expansion kinetics, trafficking, phenotype, and function and we perform head-to-head comparisons of the novel rLCMV vectors with established vectors derived from adenovirus, vaccinia virus, or Listeria monocytogenes. Our results demonstrate that replication-deficient rLCMV vectors are safe and ideally suited for both homologous and heterologous vaccination regimens to achieve optimal amplification of CD8 T cell immune responses in vivo.


Subject(s)
Genetic Vectors/immunology , Immunity, Cellular , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination/methods , Adenoviridae/genetics , Adenoviridae/immunology , Adoptive Transfer , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Immunologic Memory , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/transplantation , Vaccinia virus/genetics , Vaccinia virus/immunology
20.
Proc Natl Acad Sci U S A ; 112(13): 4056-61, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25829541

ABSTRACT

During infection, the release of damage-associated molecular patterns, so-called "alarmins," orchestrates the immune response. The alarmin IL-33 plays a role in a wide range of pathologies. Upon release, IL-33 signals through its receptor ST2, which reportedly is expressed only on CD4(+) T cells of the Th2 and regulatory subsets. Here we show that Th1 effector cells also express ST2 upon differentiation in vitro and in vivo during lymphocytic choriomeningitis virus (LCMV) infection. The expression of ST2 on Th1 cells was transient, in contrast to constitutive ST2 expression on Th2 cells, and marked highly activated effector cells. ST2 expression on virus-specific Th1 cells depended on the Th1-associated transcription factors T-bet and STAT4. ST2 deficiency resulted in a T-cell-intrinsic impairment of LCMV-specific Th1 effector responses in both mixed bone marrow-chimeric mice and adoptive cell transfer experiments. ST2-deficient virus-specific CD4(+) T cells showed impaired expansion, Th1 effector differentiation, and antiviral cytokine production. Consequently, these cells mediated little virus-induced immunopathology. Thus, IL-33 acts as a critical and direct cofactor to drive antiviral Th1 effector cell activation, with implications for vaccination strategies and immunotherapeutic approaches.


Subject(s)
Arenaviridae Infections/immunology , Gene Expression Regulation , Interleukins/immunology , STAT4 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Separation , Flow Cytometry , Gene Expression Profiling , Hyaluronan Receptors/metabolism , Interferon-gamma/metabolism , Interleukin-33 , L-Selectin/metabolism , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , T-Lymphocytes/cytology , Th1 Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL