Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Blood ; 136(6): 657-668, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32530039

ABSTRACT

Cytokine storm syndromes (CSS) are severe hyperinflammatory conditions characterized by excessive immune system activation leading to organ damage and death. Hemophagocytic lymphohistiocytosis (HLH), a disease often associated with inherited defects in cell-mediated cytotoxicity, serves as a prototypical CSS for which the 5-year survival is only 60%. Frontline therapy for HLH consists of the glucocorticoid dexamethasone (DEX) and the chemotherapeutic agent etoposide. Many patients, however, are refractory to this treatment or relapse after an initial response. Notably, many cytokines that are elevated in HLH activate the JAK/STAT pathway, and the JAK1/2 inhibitor ruxolitinib (RUX) has shown efficacy in murine HLH models and humans with refractory disease. We recently reported that cytokine-induced JAK/STAT signaling mediates DEX resistance in T cell acute lymphoblastic leukemia (T-ALL) cells, and that this could be effectively reversed by RUX. On the basis of these findings, we hypothesized that cytokine-mediated JAK/STAT signaling might similarly contribute to DEX resistance in HLH, and that RUX treatment would overcome this phenomenon. Using ex vivo assays, a murine model of HLH, and primary patient samples, we demonstrate that the hypercytokinemia of HLH reduces the apoptotic potential of CD8 T cells leading to relative DEX resistance. Upon exposure to RUX, this apoptotic potential is restored, thereby sensitizing CD8 T cells to DEX-induced apoptosis in vitro and significantly reducing tissue immunopathology and HLH disease manifestations in vivo. Our findings provide rationale for combining DEX and RUX to enhance the lymphotoxic effects of DEX and thus improve the outcomes for patients with HLH and related CSS.


Subject(s)
Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cytokine Release Syndrome/drug therapy , Dexamethasone/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Lymphohistiocytosis, Hemophagocytic/drug therapy , Pyrazoles/therapeutic use , Signal Transduction/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/physiopathology , Cytokines/physiology , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Disease Models, Animal , Drug Resistance/drug effects , Drug Therapy, Combination , Humans , Interleukin-2/pharmacology , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/pharmacology , Janus Kinases , Lymphocytic Choriomeningitis/complications , Lymphocytic Choriomeningitis/physiopathology , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/enzymology , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Mice, Inbred C57BL , Nitriles , Perforin/deficiency , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines , STAT5 Transcription Factor/physiology , Specific Pathogen-Free Organisms
2.
Nat Commun ; 11(1): 1632, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32242021

ABSTRACT

Co-stimulatory signals, cytokines and transcription factors regulate the balance between effector and memory cell differentiation during T cell activation. Here, we analyse the role of the TRAF2-/NCK-interacting kinase (TNIK), a signaling molecule downstream of the tumor necrosis factor superfamily receptors such as CD27, in the regulation of CD8+ T cell fate during acute infection with lymphocytic choriomeningitis virus. Priming of CD8+ T cells induces a TNIK-dependent nuclear translocation of ß-catenin with consecutive Wnt pathway activation. TNIK-deficiency during T cell activation results in enhanced differentiation towards effector cells, glycolysis and apoptosis. TNIK signaling enriches for memory precursors by favouring symmetric over asymmetric cell division. This enlarges the pool of memory CD8+ T cells and increases their capacity to expand after re-infection in serial re-transplantation experiments. These findings reveal that TNIK is an important regulator of effector and memory T cell differentiation and induces a population of stem cell-like memory T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Humans , Immunologic Memory , Lymphocyte Activation , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Wnt Signaling Pathway
3.
Nat Commun ; 8: 14003, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084302

ABSTRACT

T-cell proliferation is regulated by ubiquitination but the underlying molecular mechanism remains obscure. Here we report that Lys-48-linked ubiquitination of the transcription factor KLF4 mediated by the E3 ligase Mule promotes T-cell entry into S phase. Mule is elevated in T cells upon TCR engagement, and Mule deficiency in T cells blocks proliferation because KLF4 accumulates and drives upregulation of its transcriptional targets E2F2 and the cyclin-dependent kinase inhibitors p21 and p27. T-cell-specific Mule knockout (TMKO) mice develop exacerbated experimental autoimmune encephalomyelitis (EAE), show impaired generation of antigen-specific CD8+ T cells with reduced cytokine production, and fail to clear LCMV infections. Thus, Mule-mediated ubiquitination of the novel substrate KLF4 regulates T-cell proliferation, autoimmunity and antiviral immune responses in vivo.


Subject(s)
Cell Proliferation , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
Virology ; 490: 75-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26855329

ABSTRACT

Mcl-1, an anti-apoptotic member of Bcl-2 family maintains cell viability during clonal expansion of CD8 T cells, but the cell intrinsic role of Mcl-1 in contraction of effectors or the number of memory CD8 T cells is unknown. Mcl-1 levels decline during the contraction phase but rebound to high levels in memory CD8 T cells. Therefore, by overexpressing Mcl-1 in CD8 T cells we asked whether limiting levels of Mcl-1 promote contraction of effectors and constrain CD8 T-cell memory. Mcl-1 overexpression failed to affect CD8 T-cell expansion, contraction or the magnitude of CD8 T-cell memory. Strikingly, high Mcl-1 levels enhanced mTOR phosphorylation and augmented the differentiation of terminal effector cells and effector memory CD8 T cells to the detriment of poly-cytokine-producing central memory CD8 T cells. Taken together, these findings provided unexpected insights into the role of Mcl-1 in the differentiation of effector and memory CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Myeloid Cell Leukemia Sequence 1 Protein/immunology , Animals , Humans , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology
5.
Am J Pathol ; 185(11): 3025-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26506472

ABSTRACT

The immunoregulatory cytokine IL-10 suppresses T-cell immunity. The complementary question, whether IL-10 is also involved in limiting the collateral damage of vigorous T cell responses, has not been addressed in detail. Here, we report that the particularly strong virus-specific immune response during acute primary infection with the lymphocytic choriomeningitis virus (LCMV) in mice is significantly further increased in Il10-deficient mice, particularly regarding frequencies and cytotoxic activity of CD8(+) T cells. This increase results in exacerbating immunopathology in select organs, ranging from transient local swelling to an increased risk for mortality. Remarkably, LCMV-induced, T cell-mediated hepatitis is not affected by endogenous Il10. The alleviating effect of Il10 on LCMV-induced immunopathology was found to be operative in delayed-type hypersensitivity footpad-swelling reaction and in debilitating meningitis in mice of both the C57BL/6 and BALB/c strains. These strains are prototypic counterpoles for genetically imprinted type 1-biased versus type 2-biased T cell-mediated immune responses against various infectious pathogens. However, during acute LCMV infection, neither systemic cytokine patterns nor the impact of Il10 on LCMV-induced immunopathology differed conspicuously between these two strains of mice. This study documents a physiological role of Il10 in the regulation of a balanced T-cell response limiting immunopathological damage.


Subject(s)
Antiviral Agents/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Interleukin-10/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antiviral Agents/metabolism , CD8-Positive T-Lymphocytes/physiology , Cytokines/blood , Cytokines/immunology , Female , Hypersensitivity, Delayed , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytic Choriomeningitis/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
6.
Clin Ter ; 166(3): e197-202, 2015.
Article in English | MEDLINE | ID: mdl-26152632

ABSTRACT

IP-10 and its receptor CXCR3 play an important role in lymphocytic choriomeningitis (LCM). During the course of LCM in mice not only cells of the immune system but also central nervous system (CNS) resident cells are actively involved in both the pro- and the counter-inflammatory immune circuits. A central role for IP-10 in regulating the accumulation of effector T cells at sites of CNS inflammation, with no apparent compensatory effect of other CXCR3 ligands, has been shown. The producers of IP-10 are mainly resident cells of the CNS, and astrocytes are the dominant expressors in the neural parenchyma, not microglial cells or recruited bone marrow-derived cell types. These results are consistent with a mode suggesting a bidirectional interplay between resident cells of the CNS and the recruited virus-specific T cells with astrocytes as active participants in the local antiviral host response.


Subject(s)
Chemokine CXCL10/biosynthesis , Lymphocytic Choriomeningitis/physiopathology , Receptors, CXCR3/biosynthesis , Animals , Central Nervous System/physiopathology , Inflammation/physiopathology , Mice
7.
J Child Neurol ; 30(5): 644-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24736119

ABSTRACT

Assumed to be underreported and underrecognized, lymphocytic choriomeningitis presents as a febrile illness transmitted by the common house mouse, Mus musculus. Although asymptomatic or mild febrile illnesses are commonplace, meningitis and meningoencephalitis may develop after symptoms have seemed to improve. Neurologic sequelae are not typical but have been reported and can persist for months. We report a documented case of lymphocytic choriomeningitis in which a previously healthy 17-year-old girl experienced debilitating recurrent headaches and arthralgias for more than a year after discharge. Neuropsychological testing and visual changes were also documented. Further research is needed to estimate the prevalence of this infection, although it has been estimated that 5% of American adults have antibodies to lymphocytic choriomeningitis virus. Education and awareness of the medical community as well as the general public will be critical in prevention as well as advancing future treatment modalities of lymphocytic choriomeningitis virus.


Subject(s)
Lymphocytic Choriomeningitis/diagnosis , Lymphocytic Choriomeningitis/physiopathology , Adolescent , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Lymphocytic Choriomeningitis/drug therapy , Treatment Outcome
8.
PLoS One ; 9(5): e96442, 2014.
Article in English | MEDLINE | ID: mdl-24802239

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) can be transmitted through congenital infection, leading to persistent infection of numerous organ systems including the central nervous system (CNS). Adult mice persistently infected with LCMV (LCMV-cgPi mice) exhibit learning deficits, such as poor performance in spatial discrimination tests. Given that deficits in spatial learning have been linked to defects in adult neurogenesis, we investigated the impact of congenital LCMV infection on generation of neuroblasts from neural progenitor cells within neurogenic zones of adult mice. In LCMV-cgPi mice, QPCR and immunohistochemistry detected presence of LCMV glycoprotein-coding RNA and nucleoprotein in the hippocampal dentate gyrus and subventricular zone (SVZ), sites of neurogenesis that harbor populations of neuroblasts. Numbers of neuroblasts were reduced in LCMV-cgPi mice, as determined by IHC quantification, and analysis of BrdU incorporation by flow cytometry revealed lower numbers of BrdU-labeled neuroblasts. Additionally, TUNEL assays performed in situ showed increased numbers of apoptotic cells in the two neurogenic regions. Next, neurosphere cultures were infected in vitro with LCMV and differentiated to create a population of cells that consisted of both transit amplifying cells and neuroblasts. Immunocytochemical and TUNEL assays revealed increased numbers of TUNEL-positive cells that express nestin, suggesting that the drop in numbers of neuroblasts was due to a combination of impaired proliferation and apoptosis of progenitor cells. LCMV-cgPi mice exhibited transcriptional up-regulation several cytokines and chemokines, including gamma-interferon inducible chemokines CXCL9 and CXCL10. Chronic up-regulation of these chemokines can facilitate a pro-inflammatory niche that may contribute to defects in neurogenesis.


Subject(s)
Dentate Gyrus/virology , Lateral Ventricles/virology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Neurons/virology , Stem Cells/virology , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Dentate Gyrus/metabolism , Interferon-gamma/metabolism , Lateral Ventricles/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus , Mice , Neurogenesis/physiology , Neurons/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Transcription, Genetic/physiology , Up-Regulation/physiology
9.
Immunity ; 38(4): 782-91, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601687

ABSTRACT

Secondary lymphoid organ stromal cells comprise different subsets whose origins remain unknown. Herein, we exploit a genetic lineage-tracing approach to show that splenic fibroblastic reticular cells (FRCs), follicular dendritic cells (FDCs), marginal reticular cells (MRCs), and mural cells, but not endothelial cells, originate from embryonic mesenchymal progenitors of the Nkx2-5(+)Islet1(+) lineage. This lineage include embryonic mesenchymal cells with lymphoid tissue organizer (LTo) activity capable also of supporting ectopic lymphoid-like structures and a subset of resident spleen stromal cells that proliferate and regenerate the splenic stromal microenvironment following resolution of a viral infection. These findings identify progenitor cells that generate stromal diversity in spleen development and repair and suggest the existence of multipotent stromal progenitors in the adult spleen with regenerative capacity.


Subject(s)
Dendritic Cells, Follicular/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Spleen/pathology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Dendritic Cells, Follicular/pathology , Fibroblasts/pathology , Homeobox Protein Nkx-2.5 , Lymphocytic Choriomeningitis/physiopathology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Regeneration , Stromal Cells/metabolism , Stromal Cells/pathology
10.
Cell Death Differ ; 20(8): 998-1007, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23558951

ABSTRACT

Members of the Bcl-2 family have critical roles in regulating tissue homeostasis by modulating apoptosis. Anti-apoptotic molecules physically interact and restrain pro-apoptotic family members preventing the induction of cell death. However, the specificity of the functional interactions between pro- and anti-apoptotic Bcl-2 family members remains unclear. The pro-apoptotic Bcl-2 family member Bcl-2 interacting mediator of death (Bim) has a critical role in promoting the death of activated, effector T cells following viral infections. Although Bcl-2 is an important Bim antagonist in effector T cells, and Bcl-xL is not required for effector T-cell survival, the roles of other anti-apoptotic Bcl-2 family members remain unclear. Here, we investigated the role of myeloid cell leukemia sequence 1 (Mcl-1) in regulating effector T-cell responses in vivo. We found, at the peak of the response to lymphocytic choriomeningitis virus (LCMV) infection, that Mcl-1 expression was increased in activated CD4(+) and CD8(+) T cells. Retroviral overexpression of Mcl-1-protected activated T cells from death, whereas deletion of Mcl-1 during the course of infection led to a massive loss of LCMV-specific CD4(+) and CD8(+) T cells. Interestingly, the co-deletion of Bim failed to prevent the loss of Mcl-1-deficient T cells. Furthermore, lck-driven overexpression of a Bcl-xL transgene only partially rescued Mcl-1-deficient effector T cells suggesting a lack of redundancy between the family members. In contrast, additional loss of Bax and Bak completely rescued Mcl-1-deficient effector T-cell number and function, without enhancing T-cell proliferation. These data suggest that Mcl-1 is critical for promoting effector T-cell responses, but does so by combating pro-apoptotic molecules beyond Bim.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Myeloid Cell Leukemia Sequence 1 Protein/physiology , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/antagonists & inhibitors , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Survival/physiology , Disease Models, Animal , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/physiology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/deficiency , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology
11.
Immunol Invest ; 42(3): 204-20, 2013.
Article in English | MEDLINE | ID: mdl-23461613

ABSTRACT

During chronic viral infections, responses by virus-specific CD8(+) T cells become marginalized by the acquisition of functional defects and reduced cell numbers in a process defined as T cell exhaustion. Similarly, T cell tolerance to self-antigen is also characterized by impaired effector function and eventual deletion of self-reactive T cells. Induction of both tolerance and exhaustion involve many shared inhibitory mechanisms, thus similar therapeutic approaches have proven effective in these distinct environments. We previously demonstrated that tolerant self-reactive CD8(+) T cells expressing dual-T cell receptors (i.e., dual-TCR) could be rescued by immunization through a second TCR specific for a foreign antigen. These data revealed that T cell tolerance was regulated at the level of the self-reactive TCR. Here, dual-TCR CD8(+) T cells were used to examine if exhaustion during persistent viral infection could be rescued by an analogous strategy of immunization through a second TCR not involved in recognition of virus. In direct contrast to the rescue achievable in tolerant CD8(+) T cells, exhausted T cells were equally impaired through both TCR. These findings suggest that exhaustion is maintained by defects downstream of the virus-specific TCR, and establish that exhaustion and tolerance are distinctly regulated states of T cell dysfunction.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Receptors, Antigen, T-Cell/metabolism , Animals , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Humans , Immunization , Lymphocyte Activation , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics
12.
J Exp Med ; 209(13): 2485-99, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23230000

ABSTRACT

The inhibitory programmed death 1 (PD-1)-programmed death ligand 1 (PD-L1) pathway contributes to the functional down-regulation of T cell responses during persistent systemic and local virus infections. The blockade of PD-1-PD-L1-mediated inhibition is considered as a therapeutic approach to reinvigorate antiviral T cell responses. Yet previous studies reported that PD-L1-deficient mice develop fatal pathology during early systemic lymphocytic choriomeningitis virus (LCMV) infection, suggesting a host protective role of T cell down-regulation. As the exact mechanisms of pathology development remained unclear, we set out to delineate in detail the underlying pathogenesis. Mice deficient in PD-1-PD-L1 signaling or lacking PD-1 signaling in CD8 T cells succumbed to fatal CD8 T cell-mediated immunopathology early after systemic LCMV infection. In the absence of regulation via PD-1, CD8 T cells killed infected vascular endothelial cells via perforin-mediated cytolysis, thereby severely compromising vascular integrity. This resulted in systemic vascular leakage and a consequential collapse of the circulatory system. Our results indicate that the PD-1-PD-L1 pathway protects the vascular system from severe CD8 T cell-mediated damage during early systemic LCMV infection, highlighting a pivotal physiological role of T cell down-regulation and suggesting the potential development of immunopathological side effects when interfering with the PD-1-PD-L1 pathway during systemic virus infections.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Programmed Cell Death 1 Receptor/physiology , Shock/physiopathology , Animals , B7-H1 Antigen/deficiency , B7-H1 Antigen/genetics , B7-H1 Antigen/physiology , CD8-Positive T-Lymphocytes/immunology , Capillary Permeability/physiology , Disease Models, Animal , Endothelium, Vascular/pathology , H-2 Antigens/genetics , H-2 Antigens/physiology , Histocompatibility Antigen H-2D , Hypotension/etiology , Hypotension/physiopathology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/physiology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Pulmonary Edema/etiology , Pulmonary Edema/physiopathology , Shock/immunology , Shock/prevention & control , Signal Transduction
13.
J Neuroimmunol ; 226(1-2): 8-19, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20619904

ABSTRACT

Intracranial (i.c.) lymphocytic choriomeningitis virus (LCMV) infection of mice results in T cell-driven anorexia and weight loss, which is diminished in males compared to females. We investigated sex-specific effects on antigen-presenting cells (APCs) and T cells after i.c. LCMV infection. Numbers of LCMV-specific T cells, APC activation, and levels of inflammatory cytokines and chemokines in CSF were decreased in males compared to females. Orchidectomy enhanced these immune parameters in males, while dihydrotestosterone treatment of orchidectomized males and intact females decreased some of these parameters. These data suggest that qualitative and quantitative effects of androgens on APCs and T cells may contribute to the well-known, but poorly understood sex differences in immunity and autoimmunity.


Subject(s)
Androgens/pharmacology , Antigen-Presenting Cells/drug effects , Interferon-gamma/metabolism , Lymphocytic Choriomeningitis/pathology , T-Lymphocytes/drug effects , Aldosterone/analogs & derivatives , Aldosterone/therapeutic use , Androgens/therapeutic use , Animals , Brain/pathology , Brain/virology , Cytokines/metabolism , Eating/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Estradiol/blood , Female , Leukocyte Common Antigens/metabolism , Lymphocytic Choriomeningitis/blood , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Knockout , Myosin Heavy Chains/deficiency , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Orchiectomy/methods , Sex Factors , Statistics, Nonparametric , T-Lymphocytes/immunology , Testosterone/blood , Weight Loss/physiology
14.
J Immunol ; 184(9): 5151-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20368274

ABSTRACT

Although previous studies have demonstrated delayed viral clearance and blunted effector T cell responses in aged mice during infection, memory CD8 T cells and especially secondary responses have received less attention. In this study, we show that modest differences in the number of memory CD8 T cells formed in aged versus young animals were associated with altered memory CD8 T cell differentiation. Aged immune mice had increased morbidity and mortality upon secondary viral challenge, suggesting changes in T cell immunity. Indeed, virus-specific memory CD8 T cells from aged mice showed substantially reduced proliferative expansion upon secondary infection using multiple challenge models. In addition, this defect in recall capacity of aged memory CD8 T cells was cell-intrinsic and persisted upon adoptive transfer into young mice. Thus, the poor proliferative potential of memory T cells and altered memory CD8 T cell differentiation could underlie age-related defects in antiviral immunity.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Orthomyxoviridae/immunology , Adoptive Transfer , Aging/genetics , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Line , Cytotoxicity Tests, Immunologic , Dogs , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Genetic Predisposition to Disease , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/transplantation , T-Lymphocyte Subsets/virology , Vaccinia virus/genetics , Vaccinia virus/immunology
15.
J Neuroimmunol ; 194(1-2): 70-82, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18295350

ABSTRACT

In response to invading pathogens, Toll-like receptors (TLR) play a critical role in the initiation of the innate immune response, which can be either beneficial or detrimental to the host. In the present study, we demonstrated that central nervous system (CNS) glial cells are activated by Lymphocytic Choriomeningitis Virus (LCMV) in a TLR2-MyD88/Mal-dependent manner. Specifically, in response to LCMV, both astrocytes and microglial cells isolated from wild-type (WT) mice produced chemokines, such as MCP-1, RANTES and TNF-alpha. Similar responses occurred in TLR3 KO and TLR4 KO glial cells. In striking contrast, both astrocytes and microglial cells isolated from mice deficient in TLR2, MyD88, and Mal did not produce any of these chemokines. In addition, LCMV infection of glial cells induced up-regulation of TLR2, MHC class-I and II, CD40, CD86 in a MyD88-dependent manner. These results define a functional role for TLR signaling in viral infection-induced activation of CNS glial cells as well as for the immunopathology in the CNS.


Subject(s)
Astrocytes/virology , Lymphocytic choriomeningitis virus/physiology , Membrane Transport Proteins/physiology , Microglia/virology , Myelin Proteins/physiology , Myeloid Differentiation Factor 88/physiology , Proteolipids/physiology , Toll-Like Receptor 2/physiology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Chemokines/biosynthesis , Chemokines/genetics , Histocompatibility Antigens Class II/immunology , Lymphocytic Choriomeningitis/physiopathology , Membrane Transport Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Myelin Proteins/deficiency , Myelin and Lymphocyte-Associated Proteolipid Proteins , Myeloid Differentiation Factor 88/deficiency , Specific Pathogen-Free Organisms , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/physiology , Up-Regulation
16.
J Appl Physiol (1985) ; 98(2): 663-71, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15448122

ABSTRACT

Intense exercise to exhaustion leads to increased susceptibility and severity of infections. T cells play an essential role in control of viral infections. Whereas immune suppression is considered as a likely mechanism for exhaustive exercise-induced susceptibility to infection, we know little about viral-specific T-cell response following exhaustive exercise in young or old mice. In this study, one group of female young (10-12 wk) and old (22-24 mo) C57BL/6 mice was exposed to a single bout of intense exercise to exhaustion and immediately infected with lymphocytic choriomeningitis virus (LCMV). Eight days later, at the peak of expansion phase of T-cell response, we used tetramers of MHC class I molecules containing viral peptides to directly visualize antigen-specific CD8 T cells and a sensitive functional assay measuring interferon-gamma production at the single-cell level to quantitate the CD8 and CD4 T-cell response. To evaluate the impact of intense exercise during both the initiation and evolution of the expansion phase of the T-cell response, a second group of young and old mice continued their daily bouts of intense exercise to exhaustion over the next 8 days. Our data show that, in young mice, LCMV infection following exhaustive exercise leads to suppression of LCMV-specific CD8 and CD4 T-cell responses, and this suppression effect occurs at the initiation of the expansion phase of viral-specific T cells. However, in old mice, unlike young mice, exhaustive exercise does not cause suppression of LCMV-specific T-cell responses.


Subject(s)
Aging/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/immunology , Physical Endurance/immunology , Animals , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Female , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal/methods
17.
Diabetes ; 53(3): 591-6, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988242

ABSTRACT

Tissue-specific expression of Fas-ligand (Fas-L) can provide immune privilege by inducing apoptosis of "invading" lymphocytes expressing Fas. However, accelerated diabetes has been reported in transgenic mice expressing Fas-L in islets (RIP-Fas-L) as a result of Fas-dependent fratricide of beta-cells after transfer of diabetogenic clones. Here we studied whether Fas-L could protect islets from autoaggressive CD8 lymphocytes in a transgenic model of virally induced diabetes (RIP-LCMV-NP transgenic mice), in which the autoaggressive response is directed to a viral nucleoprotein (NP) expressed as a transgene in beta-cells. Indeed, disease incidence after viral (lymphocytic choriomeningitis virus [LCMV]) infection was reduced by approximately 30%, which was associated with a decrease of autoaggressive CD8 NP-specific lymphocytes in islets and pancreatic draining lymph nodes. However, surprisingly, a high degree (50%) of diabetes was seen in mice that expressed only Fas-L but not the viral transgene (NP) in beta-cells after infection with LCMV. This was due to induction of Fas on beta-cells after LCMV infection of the pancreas, resulting in Fas/Fas-L-mediated fratricide. Thus, although Fas-L can lend some immune privilege to islet cells, local virus-induced inflammation will induce Fas on beta-cells, leading to their mutual destruction if Fas-L is present. Expression of Fas-L therefore might not be protective in situations in which viral inflammation can be expected, resulting in Fas induction on the targeted cell itself.


Subject(s)
Islets of Langerhans/pathology , Lymphocytic Choriomeningitis/physiopathology , Membrane Glycoproteins/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Crosses, Genetic , Fas Ligand Protein , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Islets of Langerhans/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Spleen/immunology
18.
J Clin Invest ; 113(1): 74-84, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702111

ABSTRACT

Viruses can cause but can also prevent autoimmune disease. This dualism has certainly hampered attempts to establish a causal relationship between viral infections and type 1 diabetes (T1D). To develop a better mechanistic understanding of how viruses can influence the development of autoimmune disease, we exposed prediabetic mice to various viral infections. We used the well-established NOD and transgenic RIP-LCMV models of autoimmune diabetes. In both cases, infection with the lymphocytic choriomeningitis virus (LCMV) completely abrogated the diabetic process. Interestingly, such therapeutic viral infections resulted in a rapid recruitment of T lymphocytes from the islet infiltrate to the pancreatic draining lymph node, where increased apoptosis was occurring. In both models this was associated with a selective and extensive expression of the chemokine IP-10 (CXCL10), which predominantly attracts activated T lymphocytes, in the pancreatic draining lymph node, and in RIP-LCMV mice it depended on the viral antigenic load. In RIP-LCMV mice, blockade of TNF-alpha or IFN-gamma in vivo abolished the prevention of T1D. Thus, virally induced proinflammatory cytokines and chemokines can influence the ongoing autoaggressive process beneficially at the preclinical stage, if produced at the correct location, time, and levels.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/physiology , Prediabetic State/prevention & control , Aging , Animals , Diabetes Mellitus, Type 1/virology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Polymerase Chain Reaction , Prediabetic State/virology , Time Factors
19.
J Virol ; 77(13): 7393-400, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12805438

ABSTRACT

The human herpesvirus 8-encoded protein vMIP-II is a potent in vitro antagonist of many chemokine receptors believed to be associated with attraction of T cells with a type 1 cytokine profile. For the present report we have studied the in vivo potential of this viral chemokine antagonist to inhibit virus-induced T-cell-mediated inflammation. This was done by use of the well-established model system murine lymphocytic choriomeningitis virus infection. Mice were infected in the footpad, and the induced CD8(+) T-cell-dependent inflammation was evaluated in mice subjected to treatment with vMIP-II. We found that inflammation was markedly inhibited in mice treated during the efferent phase of the antiviral immune response. In vitro studies revealed that vMIP-II inhibited chemokine-induced migration of activated CD8(+) T cells, but not T-cell-target cell contact, granule exocytosis, or cytokine release. Consistent with these in vitro findings treatment with vMIP-II inhibited the adoptive transfer of a virus-specific delayed-type hypersensitivity response in vivo, but only when antigen-primed donor cells were transferred via the intravenous route and required to migrate actively, not when the cells were injected directly into the test site. In contrast to the marked inhibition of the effector phase, the presence of vMIP-II during the afferent phase of the immune response did not result in significant suppression of virus-induced inflammation. Taken together, these results indicate that chemokine-induced signals are pivotal in directing antiviral effector cells toward virus-infected organ sites and that vMIP-II is a potent inhibitor of type 1 T-cell-mediated inflammation.


Subject(s)
Chemokines/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data
20.
J Virol ; 77(8): 4911-27, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12663797

ABSTRACT

Chronic viral infections often result in ineffective CD8 T-cell responses due to functional exhaustion or physical deletion of virus-specific T cells. However, how persisting virus impacts various CD8 T-cell effector functions and influences other aspects of CD8 T-cell dynamics, such as immunodominance and tissue distribution, remains largely unknown. Using different strains of lymphocytic choriomeningitis virus (LCMV), we compared responses to the same CD8 T-cell epitopes during acute or chronic infection. Persistent infection led to a disruption of the normal immunodominance hierarchy of CD8 T-cell responses seen following acute infection and dramatically altered the tissue distribution of LCMV-specific CD8 T cells in lymphoid and nonlymphoid tissues. Most importantly, CD8 T-cell functional impairment occurred in a hierarchical fashion in chronically infected mice. Production of interleukin 2 and the ability to lyse target cells in vitro were the first functions compromised, followed by the ability to make tumor necrosis factor alpha, while gamma interferon production was most resistant to functional exhaustion. Antigen appeared to be the driving force for this loss of function, since a strong correlation existed between the viral load and the level of exhaustion. Further, epitopes presented at higher levels in vivo resulted in physical deletion, while those presented at lower levels induced functional exhaustion. A model is proposed in which antigen levels drive the hierarchical loss of different CD8 T-cell effector functions during chronic infection, leading to distinct stages of functional impairment and eventually to physical deletion of virus-specific T cells. These results have implications for the study of human chronic infections, where similar T-cell deletion and functional dysregulation has been observed.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Immunodominant Epitopes/immunology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/pathogenicity , Animals , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Epitopes, T-Lymphocyte/immunology , Female , Liver/immunology , Lung/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...