Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 49(4): 678-694.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314757

ABSTRACT

CD8+ T cell exhaustion impedes control of chronic viral infection; yet how new T cell responses are mounted during chronic infection is unclear. Unlike T cells primed at the onset of infection that rapidly differentiate into effectors and exhaust, we demonstrate that virus-specific CD8+ T cells primed after establishment of chronic LCMV infection preferentially generate memory-like transcription factor TCF1+ cells that were transcriptionally and proteomically distinct, less exhausted, and more responsive to immunotherapy. Mechanistically, adaptations of antigen-presenting cells and diminished T cell signaling intensity promoted differentiation of the memory-like subset at the expense of rapid effector cell differentiation, which was now highly dependent on IL-21-mediated CD4+ T cell help for its functional generation. Chronic viral infection similarly redirected de novo differentiation of tumor-specific CD8+ T cells, ultimately preventing cancer control. Thus, targeting these T cell stimulatory pathways could enable strategies to control chronic infection, tumors, and enhance immunotherapeutic efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunity/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Chronic Disease , Gene Expression Profiling/methods , Immunity/genetics , Immunologic Memory/genetics , Immunotherapy , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Proteomics/methods , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T Cell Transcription Factor 1/metabolism
2.
Nat Immunol ; 14(6): 574-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584070

ABSTRACT

Chronic infection is difficult to overcome because of exhaustion or depletion of cytotoxic effector CD8(+) T cells (cytotoxic T lymphoytes (CTLs)). Here we report that signaling via Toll-like receptors (TLRs) induced intrahepatic aggregates of myeloid cells that enabled the population expansion of CTLs (iMATEs: 'intrahepatic myeloid-cell aggregates for T cell population expansion') without causing immunopathology. In the liver, CTL proliferation was restricted to iMATEs that were composed of inflammatory monocyte-derived CD11b(+) cells. Signaling via tumor-necrosis factor (TNF) caused iMATE formation that facilitated costimulation dependent on the receptor OX40 for expansion of the CTL population. The iMATEs arose during acute viral infection but were absent during chronic viral infection, yet they were still induced by TLR signaling. Such hepatic expansion of the CTL population controlled chronic viral infection of the liver after vaccination with DNA. Thus, iMATEs are dynamic structures that overcome regulatory cues that limit the population expansion of CTLs during chronic infection and can be used in new therapeutic vaccination strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Liver Diseases/immunology , Lymphocytic Choriomeningitis/immunology , Myeloid Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Animals, Newborn , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Host-Pathogen Interactions/immunology , Immunotherapy , Liver/immunology , Liver/metabolism , Liver/virology , Liver Diseases/therapy , Liver Diseases/virology , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Myeloid Cells/metabolism , Receptors, OX40/immunology , Receptors, OX40/metabolism , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
3.
Proc Natl Acad Sci U S A ; 117(10): 5420-5429, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094187

ABSTRACT

Chronic infection provokes alterations in inflammatory and suppressive pathways that potentially affect the function and integrity of multiple tissues, impacting both ongoing immune control and restorative immune therapies. Here we demonstrate that chronic lymphocytic choriomeningitis virus infection rapidly triggers severe thymic depletion, mediated by CD8 T cell-intrinsic type I interferon (IFN) and signal transducer and activator of transcription 2 (Stat2) signaling. Occurring temporal to T cell exhaustion, thymic cellularity reconstituted despite ongoing viral replication, with a rapid secondary thymic depletion following immune restoration by anti-programmed death-ligand 1 (PDL1) blockade. Therapeutic hematopoietic stem cell transplant (HSCT) during chronic infection generated new antiviral CD8 T cells, despite sustained virus replication in the thymus, indicating an impairment in negative selection. Consequently, low amounts of high-affinity self-reactive T cells also escaped the thymus following HSCT during chronic infection. Thus, by altering the stringency and partially impairing negative selection, the host generates new virus-specific T cells to replenish the fight against the chronic infection, but also has the potentially dangerous effect of enabling the escape of self-reactive T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Interferon Type I/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus , Thymus Gland/pathology , Thymus Gland/virology , Animals , Atrophy/virology , B7-H1 Antigen/antagonists & inhibitors , Chronic Disease , Hematopoietic Stem Cell Transplantation , Interferon Type I/genetics , Lymphocytic Choriomeningitis/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT2 Transcription Factor/metabolism , Signal Transduction , Virus Replication
4.
J Allergy Clin Immunol ; 142(3): 904-913.e3, 2018 09.
Article in English | MEDLINE | ID: mdl-29355678

ABSTRACT

BACKGROUND: Mutations in the perforin 1 (PRF1) gene account for up to 58% of familial hemophagocytic lymphohistiocytosis syndromes. The resulting defects in effector cell cytotoxicity lead to hypercytokinemia and hyperactivation with inflammation in various organs. OBJECTIVE: We sought to determine whether autologous gene-corrected T cells can restore cytotoxic function, reduce disease activity, and prevent hemophagocytic lymphohistiocytosis (HLH) symptoms in in vivo models. METHODS: We developed a gammaretroviral vector to transduce murine CD8 T cells in the Prf-/- mouse model. To verify functional correction of Prf-/- CD8 T cells in vivo, we used a lymphocytic choriomeningitis virus (LCMV) epitope-transfected murine lung carcinoma cell tumor model. Furthermore, we challenged gene-corrected and uncorrected mice with LCMV. One patient sample was transduced with a PRF1-encoding lentiviral vector to study restoration of cytotoxicity in human cells. RESULTS: We demonstrated efficient engraftment and functional reconstitution of cytotoxicity after intravenous administration of gene-corrected Prf-/- CD8 T cells into Prf-/- mice. In the tumor model infusion of Prf-/- gene-corrected CD8 T cells eliminated the tumor as efficiently as transplantation of wild-type CD8 T cells. Similarly, mice reconstituted with gene-corrected Prf-/- CD8 T cells displayed complete protection from the HLH phenotype after infection with LCMV. Patients' cells showed correction of cytotoxicity in human CD8 T cells after transduction. CONCLUSION: These data demonstrate the potential application of T-cell gene therapy in reconstituting cytotoxic function and protection against HLH in the setting of perforin deficiency.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Lymphocytic Choriomeningitis/therapy , Lymphohistiocytosis, Hemophagocytic/therapy , Perforin/genetics , Animals , Cell Line, Tumor , Child, Preschool , Genetic Therapy , Humans , Lymphocytic choriomeningitis virus , Male , Mice, Inbred C57BL , Mice, Knockout
5.
Transpl Infect Dis ; 20(6): e13013, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30325104

ABSTRACT

Solid organ transplant recipients (SOTR) are at increased risk for a wide variety of typical and atypical infections as a consequence of impaired cell mediated and humoral immunity. We report a case of meningoencephalitis in a renal transplant recipient caused by lymphocytic choriomeningitis virus (LCMV) acquired by exposure to mice excreta. The clinical course was complicated by the development of hydrocephalus, requiring a ventriculoperitoneal shunt. To our knowledge, this is the first reported case of LCMV infection in a SOTR that was not organ donor derived.


Subject(s)
Kidney Transplantation/adverse effects , Lymphocytic Choriomeningitis/transmission , Lymphocytic choriomeningitis virus/isolation & purification , Meningoencephalitis/transmission , Mice/virology , Adult , Animals , Feces/virology , Humans , Immunoglobulins, Intravenous/therapeutic use , Kidney Failure, Chronic/surgery , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Male , Meningoencephalitis/therapy , Meningoencephalitis/virology , Physical Therapy Modalities , Treatment Outcome
6.
J Immunol ; 193(10): 5033-43, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25281716

ABSTRACT

The costimulatory TNFR family member GITR can provide important survival signals for CD8 T cells. However, little is known about the regulation of this pathway during a chronic infection. In this study, we show that GITR ligand (GITRL) is maximally induced on APCs at day 2 post-lymphocytic choriomeningitis virus (LCMV) clone 13 infection, but is downregulated to below baseline levels by day 8 postinfection (p.i.), and remains so at the chronic stage of infection. At its peak, GITRL expression is highest on macrophages, with lower expression on conventional and plasmacytoid dendritic cells. GITR expression was highest on T regulatory cells but was also detected on Th1 and LCMV-specific CD8 T cells at day 8 p.i. and was maintained at low, but above baseline levels at the chronic stage of LCMV infection. As GITRL was limiting at the chronic stage of infection, we investigated the potential of therapeutic stimulation of GITR at this stage using agonistic anti-GITR Ab. Anti-GITR treatment at day 21 p.i. increased the frequency and number of LCMV-specific CD8 T cells, resulting in increased in vivo CTL activity and a concomitant decrease in viral load, despite the persistence of PD-1 expression. These effects of anti-GITR were CD8 T cell intrinsic, with no detectable effects on Th1 or T regulatory cells. In contrast to other TNFR agonists, such as anti-4-1BB, which can cause immune pathology, a single therapeutic dose of anti-GITR did not induce splenomegaly or increase serum alanine transaminase. These studies identify GITR as a promising therapeutic target for chronic infection.


Subject(s)
Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Tumor Necrosis Factors/agonists , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Gene Expression Regulation , Host-Pathogen Interactions , Immunotherapy , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology , Th1 Cells/immunology , Th1 Cells/pathology , Th1 Cells/virology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology
7.
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 , Animals , Female , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Disease Models, Animal , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Lymphocytic Choriomeningitis/complications , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Spleen/immunology , Spleen/virology , Viral Load
8.
J Exp Med ; 203(11): 2461-72, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17030951

ABSTRACT

A defining characteristic of persistent viral infections is the loss and functional inactivation of antiviral effector T cells, which prevents viral clearance. Interleukin-10 (IL-10) suppresses cellular immune responses by modulating the function of T cells and antigen-presenting cells. In this paper, we report that IL-10 production is drastically increased in mice persistently infected with lymphocytic choriomeningitis virus. In vivo blockade of the IL-10 receptor (IL-10R) with a neutralizing antibody resulted in rapid resolution of the persistent infection. IL-10 secretion was diminished and interferon gamma production by antiviral CD8+ T cells was enhanced. In persistently infected mice, CD8alpha+ dendritic cell (DC) numbers declined early after infection, whereas CD8alpha- DC numbers were not affected. CD8alpha- DCs supported IL-10 production and subsequent dampening of antiviral T cell responses. Therapeutic IL-10R blockade broke the cycle of IL-10-mediated immune suppression, preventing IL-10 priming by CD8alpha- DCs and enhancing antiviral responses and thereby resolving infection without causing immunopathology.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus , Receptors, Interleukin-10/antagonists & inhibitors , Receptors, Interleukin-10/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Chronic Disease , Immune Sera/administration & dosage , Interleukin-10/antagonists & inhibitors , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptors, Interleukin-10/biosynthesis
9.
Front Immunol ; 13: 908707, 2022.
Article in English | MEDLINE | ID: mdl-35958615

ABSTRACT

T cell-based therapies have been widely explored for the treatment of cancer and chronic infection, but B cell-based therapies have remained largely unexplored. To study the effect of B cell therapy, we adoptively transferred virus-specific B cells into mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). Adoptive transfer of virus-specific B cells resulted in increase in antibody titers and reduction of viral loads. Importantly, the efficacy of B cell therapy was partly dependent on antibody effector functions, and was improved by co-transferring virus-specific CD4 T cells. These findings provide a proof-of-concept that adoptive B cell therapy can be effective for the treatment of chronic infections, but provision of virus-specific CD4 T cells may be critical for optimal virus neutralization.


Subject(s)
Lymphocytic Choriomeningitis , Animals , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL
10.
Nat Med ; 5(12): 1370-4, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10581078

ABSTRACT

At present, little is known about the pathogenesis of acute virus-induced shock and pulmonary failure. A chief impediment in understanding the underlying disease mechanisms and developing treatment strategies has been the lack of a suitable animal model. This study describes a mouse model of virus-induced systemic shock and respiratory distress, and shows that blockade of the lymphotoxin beta receptor pathway reverses the disease.


Subject(s)
Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Respiratory Insufficiency/therapy , Shock, Septic/therapy , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Female , Humans , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/therapy , Lymphotoxin beta Receptor , Male , Mice , Mice, Inbred NZB , Respiratory Insufficiency/immunology , Respiratory Insufficiency/pathology , Shock, Septic/immunology , Shock, Septic/pathology , Signal Transduction , Time Factors
11.
Nat Med ; 9(5): 540-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12692546

ABSTRACT

Interleukin (IL)-2 is currently used to enhance T-cell immunity but can have both positive and negative effects on T cells. To determine whether these opposing results are due to IL-2 acting differently on T cells depending on their stage of differentiation, we examined the effects of IL-2 therapy during the expansion, contraction and memory phases of the T-cell response in lymphocytic choriomeningitis virus (LCMV)-infected mice. IL-2 treatment during the expansion phase was detrimental to the survival of rapidly dividing effector T cells. In contrast, IL-2 therapy was highly beneficial during the death phase, resulting in increased proliferation and survival of virus-specific T cells. IL-2 treatment also increased proliferation of resting memory T cells in mice that controlled the infection. Virus-specific T cells in chronically infected mice also responded to IL-2 resulting in decreased viral burden. Thus, timing of IL-2 administration and differentiation status of the T cell are critical parameters in designing IL-2 therapies.


Subject(s)
Interleukin-2/therapeutic use , Lymphocytic Choriomeningitis/therapy , T-Lymphocytes/drug effects , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Immunologic Memory/drug effects , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
12.
Brain ; 133(Pt 4): 1054-66, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20354003

ABSTRACT

Viral infection of the central nervous system can lead to disability and death. Yet the majority of viral infections with central nervous system involvement resolve with only mild clinical manifestations, if any. This is generally attributed to efficient elimination of the infection from the brain coverings, i.e. the meninges, ependyma and chorioplexus, which are the primary targets of haematogeneous viral spread. How the immune system is able to purge these structures from viral infection with only minimal detrimental effects is still poorly understood. In the present work we studied how an attenuated lymphocytic choriomeningitis virus can be cleared from the central nervous system in the absence of overt disease. We show that elimination of the virus from brain ependyma, but not from brain parenchyma, could be achieved by a T cell-dependent mechanism operating independently of major histocompatibility class I antigens and perforin. Considering that cytotoxic T lymphocyte-mediated cytotoxicity is a leading cause of viral immunopathology and tissue damage, our findings may explain why the most common viral intruders of the central nervous system rarely represent a serious threat to our health.


Subject(s)
Brain/immunology , Ependyma/immunology , Histocompatibility Antigens Class I/physiology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/therapy , Perforin/physiology , T-Lymphocytes/immunology , Animals , Brain/virology , Ependyma/pathology , Ependyma/virology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Vesicular stomatitis Indiana virus/immunology , Viral Load/immunology
13.
Front Immunol ; 12: 677824, 2021.
Article in English | MEDLINE | ID: mdl-34326837

ABSTRACT

Effective T cell differentiation during acute virus infections leads to the generation of effector T cells that mediate viral clearance, as well as memory T cells that confer protection against subsequent reinfection. While inhibitory immune checkpoints have been shown to promote T cell dysfunction during chronic virus infections and in tumors, their roles in fine tuning the differentiation and responses of effector and memory T cells are only just beginning to be appreciated. We previously identified PSGL-1 as a fundamental regulator of T cell exhaustion that sustains expression of several inhibitory receptors, including PD-1. We now show that PSGL-1 can restrict the magnitude of effector T cell responses and memory T cell development to acute LCMV virus infection by limiting survival, sustaining PD-1 expression, and reducing effector responses. After infection, PSGL-1-deficient effector T cells accumulated to a greater extent than wild type T cells, and preferentially generated memory precursor cells that displayed enhanced accumulation and functional capacity in response to TCR stimulation as persisting memory cells. Although, PSGL-1-deficient memory cells did not exhibit inherent greater sensitivity to cell death, they failed to respond to a homologous virus challenge after adoptive transfer into naïve hosts indicating an impaired capacity to generate memory effector T cell responses in the context of viral infection. These studies underscore the function of PSGL-1 as a key negative regulator of effector and memory T cell differentiation and suggest that PSGL-1 may limit excessive stimulation of memory T cells during acute viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/metabolism , Adoptive Transfer/methods , Animals , Cell Differentiation/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Treatment Outcome
14.
J Virol ; 83(17): 8905-15, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553326

ABSTRACT

Persistent viral infections are a major health concern worldwide. During persistent infection, overwhelming viral replication and the rapid loss of antiviral T-cell function can prevent immune-mediated clearance of the infection, and therapies to reanimate the immune response and purge persistent viruses have been largely unsuccessful. Adoptive immunotherapy using memory T cells is a highly successful therapeutic approach to eradicate a persistent viral infection. Understanding precisely how therapeutically administered memory T cells achieve clearance should improve our ability to terminate states of viral persistence in humans. Mice persistently infected from birth with lymphocytic choriomeningitis virus are tolerant to the pathogen at the T-cell level and thus provide an excellent model to evaluate immunotherapeutic regimens. Previously, we demonstrated that adoptively transferred memory T cells require recipient dendritic cells to effectively purge an established persistent viral infection. However, the mechanisms that reactivate and sustain memory T-cell responses during clearance of such an infection remain unclear. Here we establish that therapeutic memory T cells require CD80 and CD86 costimulatory signals to efficiently clear an established persistent viral infection in vivo. Early blockade of costimulatory pathways with CTLA-4-Fc decreased the secondary expansion of virus-specific CD8(+) and CD4(+) memory T cells as well as their ability to produce antiviral cytokines and purge the persistent infection. Late costimulation blockade also reduced virus-specific T-cell numbers, illustrating that sustained interactions with costimulatory molecules is required for efficient T-cell expansion. These findings indicate that antiviral memory T cells require costimulation to efficiently clear a persistent viral infection and that costimulatory pathways can be targeted to modulate the magnitude of an adoptive immunotherapeutic regimen.


Subject(s)
Adoptive Transfer , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Lymphocyte Activation/immunology , Mice
15.
J Immunol ; 181(2): 1161-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606669

ABSTRACT

Restrictions in the diversity of an adaptive immune repertoire can facilitate viral persistence. Because a host afflicted with an immune deficiency is not likely to purge a persistent infection using endogenous mechanisms, it is important to explore adoptive therapies to supplement the host with a functional immune defense. In this study, we describe a virus carrier state that results from introducing lymphocytic choriomeningitis virus (LCMV) into adult mice possessing a restricted T cell repertoire. On infection of these mice, LCMV establishes systemic persistence, and within the CNS the virus infects astrocytes (and later oligodendrocytes) rather than its traditional parenchymal target neurons. To determine whether LCMV could be purged from a novel target selection in the absence of an endogenous immune repertoire, we adoptively transferred virus-specific memory cells into adult carrier mice. The memory cells purged virus from the periphery as well as the CNS, but they induced fatalities not typically associated with adoptive immunotherapy. When the repertoire of the recipient mice was examined, a deficiency in natural regulatory T cells was noted. We therefore supplemented carrier mice with regulatory T cells and simultaneously performed adoptive immunotherapy. Cotransfer of regulatory T cells significantly reduced mortality while still permitting the antiviral memory cells to purge the persistent infection. These data indicate that regulatory T cells can be used therapeutically to lessen the pathogenicity of virus-specific immune cells in an immunodeficient host. We also propose that the novel carrier state described herein will facilitate the study of immunotherapeutic regimens.


Subject(s)
Central Nervous System/immunology , Immunologic Memory , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Animals , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/virology , Carrier State , Cell Line , Central Nervous System/cytology , Central Nervous System/virology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligodendroglia/cytology , Oligodendroglia/immunology , Oligodendroglia/virology , T-Lymphocytes, Regulatory/metabolism , Virus Replication
16.
Immunohorizons ; 4(4): 217-230, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332052

ABSTRACT

CD8+ T cell-mediated immunity is critical for host defense against viruses and requires mitochondria-mediated type I IFN (IFN-I) signaling for optimal protection. Cyclophilin D (CypD) is a mitochondrial matrix protein that modulates the mitochondrial permeability transition pore, but its role in IFN-I signaling and CD8+ T cell responses to viral infection has not been previously explored. In this study, we demonstrate that CypD plays a critical extrinsic role in the survival of Ag-specific CD8+ T cell following acute viral infection with lymphocytic choriomeningitis virus in mice. CypD deficiency resulted in reduced IFN-I and increased CD8+ T cell death, resulting in a reduced antiviral CD8+ T cell response. In addition, CypD deficiency was associated with an increase in pathogen burden at an early time-point following infection. Furthermore, our data demonstrate that transfer of wild-type macrophages (expressing CypD) to CypD-deficient mice can partially restore CD8+ T cell responses. These results establish that CypD plays an extrinsic role in regulating optimal effector CD8+ T cell responses to viral infection. Furthermore, this suggests that, under certain circumstances, inhibition of CypD function may have a detrimental impact on the host's ability to respond to viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Survival/genetics , Host Microbial Interactions/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Peptidyl-Prolyl Isomerase F/metabolism , Adoptive Transfer/methods , Animals , CD8 Antigens/metabolism , Peptidyl-Prolyl Isomerase F/genetics , Female , Host Microbial Interactions/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics
17.
Viruses ; 11(2)2019 01 27.
Article in English | MEDLINE | ID: mdl-30691215

ABSTRACT

Virology has played an essential role in deciphering many immunological phenomena, thus shaping our current understanding of the immune system. Animal models of viral infection and human viral infections were both important tools for immunological discoveries. This review discusses two immunological breakthroughs originally identified with the help of the lymphocytic choriomeningitis virus (LCMV) model; immunological restriction by major histocompatibility complex and immunotherapy using checkpoint blockade. In addition, we discuss related discoveries such as development of tetramers, viral escape mutation, and the phenomenon of T-cell exhaustion.


Subject(s)
Disease Models, Animal , Immunotherapy , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Major Histocompatibility Complex/immunology , Animals , Clinical Trials as Topic , Humans , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Mutation , T-Lymphocytes/immunology , T-Lymphocytes/pathology
18.
Front Immunol ; 10: 169, 2019.
Article in English | MEDLINE | ID: mdl-30814995

ABSTRACT

The long-term persistence of viral antigens drives virus-specific CD8 T cell exhaustion during chronic viral infection. Yet exhausted, CD8 T cells are still endowed with certain levels of effector function, by which they can keep viral replication in check in chronic infection. However, the regulatory factors involved in regulating the effector function of exhausted CD8 T cell are largely unknown. Using mouse model of chronic LCMV infection, we found that the deletion of transcription factor TCF-1 in LCMV-specific exhausted CD8 T cells led to the profound reduction in cytokine production and degranulation. Conversely, ectopic expression of TCF-1 or using agonist to activate TCF-1 activities promotes the effector function of exhausted CD8 T cells. Mechanistically, TCF-1 fuels the functionalities of exhausted CD8 T cells by promoting the expression of an array of key effector function-associated transcription regulators, including Foxo1, Zeb2, Id3, and Eomes. These results collectively indicate that targeting TCF-1 mediated transcriptional pathway may represent a promising immunotherapy strategy against chronic viral infections by reinvigorating the effector function of exhausted virus-specific CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , T Cell Transcription Factor 1/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Adoptive Transfer , Animals , Cell Survival , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , T Cell Transcription Factor 1/genetics , Transplantation Chimera , Viral Load , Virus Diseases/therapy
19.
Front Biosci ; 13: 4529-43, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508527

ABSTRACT

Viral infection of the central nervous system (CNS) can result in a multitude of responses including pathology, persistence or immune clearance. Lymphocytic choriomeningitis virus (LCMV) is a powerful model system to explore these potential outcomes of CNS infection due to the diversity of responses that can be achieved after viral inoculation. Several factors including tropism, timing, dose and variant of LCMV in combination with the development or suppression of the corresponding immune response dictates whether lethal meningitis, chronic infection or clearance of LCMV in the CNS will occur. Importantly, the functionality and positioning of the LCMV-specific CD8+ T cell response are critical in directing the subsequent outcome of CNS LCMV infection. Although a basic understanding of LCMV and immune interactions in the brain exists, the molecular machinery that shapes the balance between pathogenesis and clearance in the LCMV-infected CNS remains to be elucidated. This review covers the various outcomes of LCMV infection in the CNS and what is currently known about the impact of the virus itself versus the immune response in the development of disease or clearance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Acute Disease , Animals , Carrier State , Central Nervous System/immunology , Disease Models, Animal , Humans , Immunotherapy/methods , Infant, Newborn , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/therapy , Infant, Newborn, Diseases/virology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/genetics , Mental Disorders/etiology , Mice , T-Lymphocytes/immunology
20.
AIDS Res Hum Retroviruses ; 34(5): 430-438, 2018 05.
Article in English | MEDLINE | ID: mdl-29620933

ABSTRACT

Bone marrow transplantation is a treatment for various cancers and genetic diseases, and the only case of a cured HIV infection involved the use of this clinical procedure, highlighting the potential use of this therapy for curing many chronic diseases. However, little is known about how chronic viral infection influences lymphocyte reconstitution after bone marrow transplantation. To address this, we infected mice with chronic lymphocytic choriomeningitis virus, and performed bone marrow transplantation to assess lymphocyte reconstitution. Interestingly, we observed that adoptively transferred marrow cells exhibited preferential B cell differentiation in chronically infected mice. Moreover, donor marrow cells that were adoptively transferred into chronically infected mice differentiated into virus-specific CD8 T cells that were able to expand after PD-L1 blockade. Taken together, our data show that chronic viral infection induces a biased differentiation of bone marrow stem cells into B cells, and that exhausted virus-specific CD8 T cells generated de novo in this setting are rescuable by PD-1 blockade. These data contribute to the understanding of how chronic viral infection impacts lymphocyte reconstitution, and may provide valuable information to improve current hematopoietic transplantation regimens in chronically infected hosts.


Subject(s)
Bone Marrow Transplantation , Lymphocytes/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Disease Models, Animal , Female , Mice, Inbred C57BL , Stem Cells/physiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL