Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Immunol ; 18(12): 1342-1352, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058703

ABSTRACT

T cells reorganize their metabolic profiles after being activated, but the systemic metabolic effect of sustained activation of the immune system has remained unexplored. Here we report that augmented T cell responses in Pdcd1-/- mice, which lack the inhibitory receptor PD-1, induced a metabolic serum signature characterized by depletion of amino acids. We found that the depletion of amino acids in serum was due to the accumulation of amino acids in activated Pdcd1-/- T cells in the lymph nodes. A systemic decrease in tryptophan and tyrosine led to substantial deficiency in the neurotransmitters serotonin and dopamine in the brain, which resulted in behavioral changes dominated by anxiety-like behavior and exacerbated fear responses. Together these data indicate that excessive activation of T cells causes a systemic metabolomic shift with consequences that extend beyond the immune system.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Fear/physiology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , Amino Acids/blood , Animals , Brain/metabolism , Dopamine/deficiency , Interferon-gamma/blood , Kynurenine/blood , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Serotonin/deficiency , T-Lymphocytes/metabolism , Tryptophan/metabolism , Tyrosine/metabolism
2.
Mol Cell ; 77(5): 937-950.e6, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31926851

ABSTRACT

Targeted blockade of programmed cell death 1 (PD-1), an immune-checkpoint receptor that inhibits T cell activation, provides clinical benefits in various cancers. However, how PD-1 modulates gene expression in T cells remains enigmatic. Here we investigated how PD-1 affects transcriptome changes induced by T cell receptor (TCR) activation. Intriguingly, we identified a huge variance in PD-1 sensitivity among TCR-inducible genes. When we quantified the half maximal effective concentration (EC50) as the relationship between change in gene expression and TCR signal strength, we found that genes associated with survival and proliferation were efficiently expressed upon TCR activation and resistant to PD-1-mediated inhibition. Conversely, genes encoding cytokines and effector molecules were expressed less efficiently and sensitive to PD-1-mediated inhibition. We further demonstrated that transcription factor binding motifs and CpG frequency in the promoter region affect EC50 and thus the PD-1 sensitivity of genes. Our findings explain how PD-1, dependent on the TCR signal strength, calibrates cellular transcriptomes to shape functional properties of T cell populations.


Subject(s)
Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/metabolism , Transcriptome , Animals , Apoptosis , Binding Sites , Cell Proliferation , Coculture Techniques , CpG Islands , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Genes, T-Cell Receptor , HEK293 Cells , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Promoter Regions, Genetic , Signal Transduction , T-Lymphocytes/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
3.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34088845

ABSTRACT

CD8+ T cells play a central role in antitumor immune responses that kill cancer cells directly. In aged individuals, CD8+ T cell immunity is strongly suppressed, which is associated with cancer and other age-related diseases. The mechanism underlying this age-related decrease in immune function remains largely unknown. This study investigated the role of T cell function in age-related unresponsiveness to PD-1 blockade cancer therapy. We found inefficient generation of CD44lowCD62Llow CD8+ T cell subset (P4) in draining lymph nodes of tumor-bearing aged mice. In vitro stimulation of naive CD8+ T cells first generated P4 cells, followed by effector/memory T cells. The P4 cells contained a unique set of genes related to enzymes involved in one-carbon (1C) metabolism, which is critical to antigen-specific T cell activation and mitochondrial function. Consistent with this finding, 1C-metabolism-related gene expression and mitochondrial respiration were down-regulated in aged CD8+ T cells compared with young CD8+ T cells. In aged OVA-specific T cell receptor (TCR) transgenic mice, ZAP-70 was not activated, even after inoculation with OVA-expressing tumor cells. The attenuation of TCR signaling appeared to be due to elevated expression of CD45RB phosphatase in aged CD8+ T cells. Surprisingly, strong stimulation by nonself cell injection into aged PD-1-deficient mice restored normal levels of CD45RB and ameliorated the emergence of P4 cells and 1C metabolic enzyme expression in CD8+ T cells, and antitumor activity. These findings indicate that impaired induction of the P4 subset may be responsible for the age-related resistance to PD-1 blockade, which can be rescued by strong TCR stimulation.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Hyaluronan Receptors/immunology , L-Selectin/immunology , Neoplasms, Experimental/immunology , Aging/genetics , Animals , Cell Line, Tumor , Hyaluronan Receptors/genetics , L-Selectin/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/immunology
4.
Am J Pathol ; 191(6): 1077-1093, 2021 06.
Article in English | MEDLINE | ID: mdl-33705751

ABSTRACT

Programmed cell death protein (PD)-1 is a coinhibitory molecule that suppresses immune response and maintains immune homeostasis. Moreover, the PD-1 pathway blocks cancers from being attacked by immune cells. Anti-PD-1 antibody therapy such as nivolumab improves survival in cancer patients. However, the occurrence of autoimmune inflammatory disorders in various organs has been increasingly reported as an adverse effect of nivolumab. Of the disorders associated with nivolumab, Sicca syndrome occurs in 3% to 11% of cases and has unknown pathologic mechanisms. Whether the absence of the PD-1 pathway causes functional and morphologic disorders in lacrimal glands was determined by analyzing PD-1 gene-knockout (Pdcd1-/-) mice. Histopathologic analysis showed that Pdcd1-/- mice developed dacryoadenitis beginning at 3 to 4 months of age, and deteriorated with age. Flow-cytometric analysis confirmed that cells infiltrating the affected lacrimal glands consisted mainly of CD3+ T cells and only a small proportion of CD19+ B cells. Among infiltrating T cells, the CD4+ Th-cell subset consisted of Th1 cells producing interferon-γ in an early stage of dacryoadenitis in Pdcd1-/- mice. Experiments of lymphocyte transfer from Pdcd1-/- into irradiated wild-type mice confirmed that CD4+ T cells from Pdcd1-/- mice induced dacryoadenitis. These results indicate that PD-1 plays an important role in the prevention of autoimmune inflammatory disorders in lacrimal glands caused by activated CD4+ Th1 cells.


Subject(s)
Autoimmune Diseases/immunology , Dacryocystitis/immunology , Dacryocystitis/metabolism , Programmed Cell Death 1 Receptor/deficiency , Th1 Cells/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity/immunology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/immunology , Sjogren's Syndrome/immunology
5.
Int Immunol ; 33(11): 563-572, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34453440

ABSTRACT

Programmed cell death-1 (PD-1) is a co-inhibitory receptor that dampens immune responses upon interaction with PD-L1 and PD-L2. Although PD-1 expression on T cells is known to be activation-dependent, how cytokines modify its regulation is not fully resolved. Using polyclonal T-cell activation to study cytokine-dependent PD-1 regulation, we found that IL-2 inhibited transcriptional up-regulation of PD-1 despite the promotion of T-cell activation. The IL-2-mediated reduction in PD-1 expression augmented CD8+ T-cell activities against PD-L1-expressing target cells. To study the mechanism of PD-1 reduction, we focused on STAT5 activation in the IL-2 signaling pathway. Bioinformatic analysis suggested a novel conserved PD-1 promoter domain where NFAT and STAT5 can potentially compete with each other for binding. NFAT1 interaction with this domain revealed substantial potency in PD-1 transcription compared to STAT5A, and STAT5A overexpression could quench NFAT1-dependent PD-1 up-regulation in a sequence-specific manner. Chromatin immunoprecipitation analysis of activated T cells showed that IL-2 treatment significantly diminished the binding of NFAT1 and NFAT2 in the hypothesized competition site, while STAT5 binding to the same region was increased. These results raise the possibility that the competition of transcriptional factors might be involved in the fine-tuning of PD-1 expression by cytokines such as IL-2.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Programmed Cell Death 1 Receptor/immunology , STAT5 Transcription Factor/immunology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics
6.
Int Immunol ; 32(6): 397-405, 2020 05 30.
Article in English | MEDLINE | ID: mdl-32009163

ABSTRACT

Persistent exposure to tumor antigens results in exhausted tumor-infiltrating T cells (TILs) that express the immune checkpoint molecules, PD-1 and Tim3, and lack anti-tumor immunity. To examine the exhausted status of TILs in ovarian cancer, the potential for cytokine production, proliferation and cytotoxicity by purified PD-1+ Tim3+ CD8 TILs was assessed. The production of IFN-γ and TNF-α by PD-1+ Tim3+ CD8 TILs remained the same in an intracellular cytokine staining assay and was higher in a cytokine catch assay than that by PD-1- Tim3- and PD-1+ Tim3- CD8 TILs. %Ki67+ was higher in PD-1+ Tim3+ CD8 TILs than in PD-1- Tim3- CD8 TILs. However, patients with high PD-1+ Tim3+ CD8 TILs had a poor prognosis. The potential for cytotoxicity was then examined. %Perforin+ and %granzyme B+ were lower in PD-1+ Tim3+ CD8 TILs than in PD-1- Tim3- and PD-1+ Tim3- CD8 TILs. To observe the potential for direct cytotoxicity by T cells, a target cell line expressing membrane-bound anti-CD3scFv was newly established and a cytotoxic assay targeting these cells was performed. The cytotoxicity of PD-1+ Tim3+ CD8 TILs was significantly lower than that of PD-1- Tim3- and PD-1+ Tim3- CD8 TILs. Even though PD-1+ Tim3+ CD8 TILs in ovarian cancer showed a sustained potential for cytokine production and proliferation, cytotoxicity was markedly impaired, which may contribute to the poor prognosis of patients with ovarian cancer. Among the impaired functions of exhausted TILs, cytotoxicity may be an essential target for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Interferon-gamma/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Adult , Aged , Aged, 80 and over , Cell Proliferation , Female , Hepatitis A Virus Cellular Receptor 2/deficiency , Humans , Immunotherapy , Interferon-gamma/immunology , Middle Aged , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Programmed Cell Death 1 Receptor/deficiency
7.
Chem Res Toxicol ; 34(3): 849-856, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33617238

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) is an idiosyncratic drug reaction that is specific to an individual and can lead to liver failure and even death. The mechanism of IDILI remains poorly understood, but most IDILI appears to be immune-mediated. We have developed the first validated animal model by using a PD-1-/- mouse model in combination with anti-CTLA-4 to block immune checkpoints and impair immune tolerance. Treatment of these mice with drugs that cause IDILI in humans led to delayed-onset liver injury with characteristics similar to IDILI in humans. The current study investigates the effects of green tea extract, a weight-loss dietary supplement that has been reported to cause IDILI in humans. Green tea extracts contain a highly variable content of catechins including (-)-epigallocatechin gallate, the major catechin in green tea formulations. If the liver injury caused by green tea extract in humans is immune-mediated, it may occur in our impaired immune tolerance model. Female PD-1-/- mice treated with anti-CTLA-4 antibody and green tea extract (500 mg/kg), a dose that is considered a no-observed-adverse-effect level for liver in rodents, produced a delayed onset increase in serum alanine transaminase levels and an increase in hepatic CD8+ T cells. In contrast, the response in male PD-1-/- mice was less pronounced, and there was no evidence of liver injury in wild-type mice. These findings are consistent with the hypothesis that the IDILI caused by green tea extract is immune-mediated and is similar to IDILI caused by medications that are associated with IDILI.


Subject(s)
Catechin/pharmacology , Disease Models, Animal , Liver/drug effects , Plant Extracts/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tea/chemistry , Animals , Catechin/chemistry , Chemical and Drug Induced Liver Injury/immunology , Dose-Response Relationship, Drug , Female , Immune Tolerance/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plant Extracts/chemistry , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/immunology
8.
Am J Respir Cell Mol Biol ; 62(6): 767-782, 2020 06.
Article in English | MEDLINE | ID: mdl-32048861

ABSTRACT

Pneumocystis is an unusual, opportunistic fungal pathogen capable of causing Pneumocystis pneumonia (PCP) in immunocompromised hosts. Although PCP was discovered >100 years ago, its pathogenesis remains unclear. The inhibitory receptor PD-1 (programmed death 1), a negative regulator of activated T cells, has been reported to take part in tumor escape, immune tolerance, and infection immunity. In this study, we examined the role of the PD-1/PD-L1 (programmed death-ligand 1) pathway in patients with PCP and in mice. The expression levels of PD-1/PD-L1 in patients with PCP and in mice were measured by real-time PCR and flow cytometry. The effects of PD-1 deficiency are demonstrated using wild-type and PD-1-/- mice. Our data show that Pneumocystis infection promotes PD-1/PD-L1 expression; PD-1 deficiency enhances the phagocytic function of macrophages and the pulmonary T-helper cell type 1 (Th1)/Th17 response, which might contribute to Pneumocystis clearance; and PD-1 deficiency affects the polarization of macrophages. PCP mice treated with anti-PD-1 antibody showed improved pulmonary clearance of Pneumocystis. Collectively, our results demonstrate that the PD-1/PD-L1 pathway plays a role in regulating the innate and adaptive immune responses, suggesting that manipulation of this pathway may constitute an immunotherapeutic strategy for PCP.


Subject(s)
B7-H1 Antigen/physiology , Macrophage Activation/physiology , Pneumonia, Pneumocystis/immunology , Programmed Cell Death 1 Receptor/deficiency , Th1 Cells/immunology , Th17 Cells/immunology , Adaptive Immunity , Adult , Aged , Animals , Antibodies, Fungal/blood , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Female , Humans , Immunity, Innate , Immunocompromised Host , Immunotherapy , Lung/immunology , Lung/metabolism , Lung/microbiology , Macrophages/microbiology , Male , Mice , Mice, Knockout , Middle Aged , Nitric Oxide/metabolism , Opportunistic Infections/immunology , Pneumocystis/immunology , Pneumonia, Pneumocystis/genetics , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction
9.
J Neuroinflammation ; 15(1): 66, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29506535

ABSTRACT

BACKGROUND: Peripheral neuropathy is currently the most common neurological complication in HIV-infected individuals, occurring in 35-50% of patients undergoing combination anti-retroviral therapy. Data have shown that distal symmetric polyneuropathy develops in mice by 6 weeks following infection with the LP-BM5 retrovirus mixture. Previous work from our laboratory has demonstrated that glial cells modulate antiviral T-cell effector responses through the programmed death (PD)-1: PD-L1 pathway, thereby limiting the deleterious consequences of unrestrained neuroinflammation. METHODS: Using the MouseMet electronic von Frey system, we assessed hind-paw mechanical hypersensitivity in LP-BM5-infected wild-type (WT) and PD-1 KO animals. Using multi-color flow cytometry, we quantitatively assessed cellular infiltration and microglial activation. Using real-time RT-PCR, we assessed viral load, expression of IFN-γ, iNOS, and MHC class II. Using western blotting, we measured protein nitrosylation within the lumbar spinal cord (LSC) and dorsal root ganglion (DRG). Histochemical staining was performed to analyze the presence of CD3, ionized calcium binding adaptor molecule (Iba)-1, MHCII, nitrotyrosine, isolectin B4 (IB4) binding, and neurofilament 200 (NF200). Statistical analyses were carried out using graphpad prism. RESULTS: Hind-paw mechanical hypersensitivity observed in LP-BM5-infected animals was associated with significantly increased lymphocyte infiltration into the spinal cord and DRG. We also observed elevated expression of IFN-γ (in LSC and DRG) and MHC II (on resident microglia in LSC). We detected elevated levels of 3-nitrotyrosine within the LSC and DRG of LP-BM5-infected animals, an indicator of nitric oxide (NO)-induced protein damage. Moreover, we observed 3-nitrotyrosine in both small (IB4+) and large (NF200+) DRG sensory neurons. Additionally, infected PD-1 KO animals displayed significantly greater mechanical hypersensitivity than WT or uninfected mice at 4 weeks post-infection (p.i.). Accelerated onset of hind-paw hypersensitivity in PD-1 KO animals was associated with significantly increased infiltration of CD4+ and CD8+ T lymphocytes, macrophages, and microglial activation at early time points. Importantly, we also observed elevated levels of 3-nitrotyrosine and iNOS in infected PD-1 KO animals when compared with WT animals. CONCLUSIONS: Results reported here connect peripheral immune cell infiltration and reactive gliosis with nitrosative damage. These data may help elucidate how retroviral infection-induced neuroinflammatory networks contribute to nerve damage and neuropathic pain.


Subject(s)
Neuralgia/etiology , Retroviridae Infections/complications , Animals , Antigens, CD/metabolism , Disease Models, Animal , Ganglia, Spinal , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/metabolism , Leukocytes , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/pathology , Neuralgia/virology , Nitric Oxide Synthase Type II , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , RNA, Messenger , Retroviridae/pathogenicity , Spinal Cord/pathology
10.
Brain Behav Immun ; 73: 708-716, 2018 10.
Article in English | MEDLINE | ID: mdl-30086399

ABSTRACT

Innate immune activation is a major driver of neurodegenerative disease and immune regulatory pathways could be potential targets for therapeutic intervention. Recently, Programmed cell death-1 (PD-1) immune checkpoint inhibition has been proposed to mount an IFN-γ-dependent systemic immune response, leading to the recruitment of peripheral myeloid cells to the brain and neuropathological and functional improvements in mice with Alzheimer's disease-like ß-amyloid pathology. Here we investigate the impact of PD-1 deficiency on murine prion disease (ME7 strain), a model of chronic neurodegeneration. Although PD-1 was found to be increased in the brain of prion mice, the absence of PD-1 did not cause myeloid cell infiltration into the brain or major changes in the inflammatory profile. However, we observed a slight exacerbation of the behavioural phenotype of ME7 mice upon PD-1 deficiency. These results do not support the possibility of using immune checkpoint blockade as a therapeutic strategy in neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases/metabolism , Prion Diseases/metabolism , Programmed Cell Death 1 Receptor/deficiency , Alzheimer Disease/pathology , Animals , Brain/metabolism , Disease Models, Animal , Female , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Myeloid Cells/metabolism , Myeloid Cells/physiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Prion Diseases/pathology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/physiology
11.
Proc Natl Acad Sci U S A ; 112(21): 6682-7, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25964334

ABSTRACT

V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a negative immune-checkpoint protein that suppresses T-cell responses. To determine whether VISTA synergizes with another immune-checkpoint, programmed death 1 (PD-1), this study characterizes the immune responses in VISTA-deficient, PD-1-deficient (KO) mice and VISTA/PD-1 double KO mice. Chronic inflammation and spontaneous activation of T cells were observed in both single KO mice, demonstrating their nonredundancy. However, the VISTA/PD-1 double KO mice exhibited significantly higher levels of these phenotypes than the single KO mice. When bred onto the 2D2 T-cell receptor transgenic mice, which are predisposed to development of inflammatory autoimmune disease in the CNS, the level of disease penetrance was significantly enhanced in the double KO mice compared with in the single KO mice. Consistently, the magnitude of T-cell response toward foreign antigens was synergistically higher in the VISTA/PD-1 double KO mice. A combinatorial blockade using monoclonal antibodies specific for VISTA and PD-L1 achieved optimal tumor-clearing therapeutic efficacy. In conclusion, our study demonstrates the nonredundant role of VISTA that is distinct from the PD-1/PD-L1 pathway in controlling T-cell activation. These findings provide the rationale to concurrently target VISTA and PD-1 pathways for treating T-cell-regulated diseases such as cancer.


Subject(s)
Membrane Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens/administration & dosage , B7-H1 Antigen/deficiency , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Female , Immune Tolerance , Ligands , Lymphocyte Activation , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics
12.
J Neuroinflammation ; 14(1): 82, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28407741

ABSTRACT

BACKGROUND: Previous work from our laboratory has demonstrated that during acute viral brain infection, glial cells modulate antiviral T cell effector responses through the PD-1: PD-L1 pathway, thereby limiting the deleterious consequences of unrestrained neuroinflammation. Here, we evaluated the PD-1: PD-L1 pathway in development of brain-resident memory T cells (bTRM) following murine cytomegalovirus (MCMV) infection. METHODS: Flow cytometric analysis of immune cells was performed at 7, 14, and 30 days post-infection (dpi) to assess the shift of brain-infiltrating CD8+ T cell populations from short-lived effector cells (SLEC) to memory precursor effector cells (MPEC), as well as generation of bTRMs. RESULTS: In wild-type (WT) animals, we observed a switch in the phenotype of brain-infiltrating CD8+ T cell populations from KLRG1+ CD127- (SLEC) to KLRG1- CD127+ (MPEC) during transition from acute through chronic phases of infection. At 14 and 30 dpi, the majority of CD8+ T cells expressed CD127, a marker of memory cells. In contrast, fewer CD8+ T cells expressed CD127 within brains of infected, PD-L1 knockout (KO) animals. Notably, in WT mice, a large population of CD8+ T cells was phenotyped as CD103+ CD69+, markers of bTRM, and differences were observed in the numbers of these cells when compared to PD-L1 KOs. Immunohistochemical studies revealed that brain-resident CD103+ bTRM cells were localized to the parenchyma. Higher frequencies of CXCR3 were also observed among WT animals in contrast to PD-L1 KOs. CONCLUSIONS: Taken together, our results indicate that bTRMs are present within the CNS following viral infection and the PD-1: PD-L1 pathway plays a role in the generation of this brain-resident population.


Subject(s)
B7-H1 Antigen/deficiency , Brain/metabolism , CD8-Positive T-Lymphocytes/metabolism , Encephalitis, Viral/metabolism , Programmed Cell Death 1 Receptor/deficiency , Animals , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Encephalitis, Viral/immunology , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , NIH 3T3 Cells , Signal Transduction/physiology
13.
J Autoimmun ; 78: 101-110, 2017 03.
Article in English | MEDLINE | ID: mdl-28082047

ABSTRACT

CD8+ T cells have an emerging role in RA. Resent research indicates a causal relationship between the non-exhausted state of CD8+ T cells, defined by lost function of PD-1, and development of arthritis. We investigated how smoking contributes to the non-exhausted phenotype of CD8+ T cells and cause survivin release to serum. We compared serum survivin levels between smokers and non-smokers in 252 RA and 168 healthy subjects. Nicotine effects on CD8+ T cells were studied in peripheral blood of smoking women, bone marrow of nicotine treated mice and in sorted CD8 spleen cells in vitro using flow cytometry and quantitative PCR. Smoking increased the frequency of survivin release in serum of healthy women (OR 3.64, p = 0.025) and in RA patients (OR 1.98, p = 0.039). CD8+ T cells of smokers gained a non-exhausted PD-1 deficient phenotype. Expression of the cytotoxic marker CD107 correlated to survivin levels in serum. In the experimental setting, nicotine exposure led to an accumulation of non-exhausted PD-1-IL-7R+ CD8+ T cells in the bone marrow that is abundant with survivin producing cells. The production of the cytolytic protein perforin in bone marrow correlated to serum survivin levels. In vitro stimulation of nicotinic receptors on murine CD8+ T cells induced repressive transcription factors T-bet and Blimp-1 in support of the non-exhausted phenotype. We conclude that nicotine contributes to autoimmunity by supporting the non-exhausted state of CD8+ T cells resulting in the release of survivin. This presents a new mechanism by which smoking may contribute to the pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Lymphocyte Activation/immunology , Smoking , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Arthritis, Rheumatoid/genetics , Biomarkers , Bone Marrow/drug effects , Bone Marrow/metabolism , Case-Control Studies , Child , Child, Preschool , Disease Models, Animal , Female , Humans , Immunophenotyping , Inhibitor of Apoptosis Proteins/blood , Lymphocyte Activation/genetics , Male , Mice , Mice, Knockout , Middle Aged , Nicotine/pharmacology , Phenotype , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/metabolism , Survivin , T-Lymphocytes, Cytotoxic/drug effects , Young Adult
14.
J Immunol ; 194(5): 2289-99, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25624454

ABSTRACT

Despite the emergence of the programmed cell death 1 (PD-1):PD-1 ligand (PD-L) regulatory axis as a promising target for treating multiple human diseases, remarkably little is known about how this pathway regulates responses to extracellular bacterial infections. We found that PD-1(-/-) mice, as well as wild-type mice treated with a PD-1 blocking Ab, exhibited significantly increased survival against lethal Streptococcus pneumoniae infection following either priming with low-dose pneumococcal respiratory infection or S. pneumoniae-capsular polysaccharide immunization. Enhanced survival in mice with disrupted PD-1:PD-L interactions was explained by significantly increased proliferation, isotype switching, and IgG production by pneumococcal capsule-specific B cells. Both PD-L, B7-H1 and B7-DC, contributed to PD-1-mediated suppression of protective capsule-specific IgG. Importantly, PD-1 was induced on capsule-specific B cells and suppressed IgG production and protection against pneumococcal infection in a B cell-intrinsic manner. To our knowledge, these results provide the first demonstration of a physiologic role for B cell-intrinsic PD-1 expression in vivo. In summary, our study reveals that B cell-expressed PD-1 plays a central role in regulating protection against S. pneumoniae, and thereby represents a promising target for bolstering immunity to encapsulated bacteria.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Programmed Cell Death 1 Receptor/immunology , Animals , B-Lymphocytes/microbiology , B7-H1 Antigen/genetics , Gene Expression Regulation , Immunity, Humoral/drug effects , Immunization , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/mortality , Polysaccharides, Bacterial/administration & dosage , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Streptococcus pneumoniae/immunology , Survival Analysis
16.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G237-45, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288425

ABSTRACT

Recent studies suggest that coinhibitory receptors appear to be important in contributing sepsis-induced immunosuppression. Our laboratory reported that mice deficient in programmed cell death receptor (PD)-1 have increased bacterial clearance and improved survival in experimental sepsis induced by cecal ligation and puncture (CLP). In response to infection, the liver clears the blood of bacteria and produces cytokines. Kupffer cells, the resident macrophages in the liver, are strategically situated to perform the above functions. However, it is not known if PD-1 expression on Kupffer cells is altered by septic stimuli, let alone if PD-1 ligation contributes to the altered microbial handling seen. Here we report that PD-1 is significantly upregulated on Kupffer cells during sepsis. PD-1-deficient septic mouse Kupffer cells displayed markedly enhanced phagocytosis and restoration of the expression of major histocompatibility complex II and CD86, but reduced CD80 expression compared with septic wild-type (WT) mouse Kupffer cells. In response to ex vivo LPS stimulation, the cytokine productive capacity of Kupffer cells derived from PD-1-/- CLP mice exhibited a marked, albeit partial, restoration of the release of IL-6, IL-12, IL-1ß, monocyte chemoattractant protein-1, and IL-10 compared with septic WT mouse Kupffer cells. In addition, PD-1 gene deficiency decreased LPS-induced apoptosis of septic Kupffer cells, as indicated by decreased levels of cleaved caspase-3 and reduced terminal deoxynucleotidyl transferase dUTP nick end-labeling-positive cells. Exploring the signal pathways involved, we found that, after ex vivo LPS stimulation, septic PD-1-/- mouse Kupffer cells exhibited an increased Akt phosphorylation and a reduced p38 phosphorylation compared with septic WT mouse Kupffer cells. Together, these results indicate that PD-1 appears to play an important role in regulating the development of Kupffer cell dysfunction seen in sepsis.


Subject(s)
Coinfection/metabolism , Kupffer Cells/metabolism , Liver/metabolism , Programmed Cell Death 1 Receptor/metabolism , Sepsis/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cells, Cultured , Coinfection/genetics , Coinfection/microbiology , Cytokines/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Kupffer Cells/microbiology , Liver/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Phenotype , Phosphorylation , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/genetics , Sepsis/microbiology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Infect Immun ; 83(5): 2011-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25733520

ABSTRACT

Malaria infection treatment vaccine (ITV) is a promising strategy to induce homologous and heterologous protective immunity against the blood stage of the parasite. However, the underlying mechanism of protection remains largely unknown. Here, we found that a malaria-specific antibody (Ab) could mediate the protective immunity of ITV-immunized mice. Interestingly, PD-1 deficiency greatly elevated the levels of both malaria-specific total IgG and subclass IgG2a and enhanced the protective efficacy of ITV-immunized mice against the blood-stage challenge. A serum adoptive-transfer assay demonstrated that the increased Ab level contributed to the enhanced protective efficacy of the immunized PD-1-deficient mice. Further study showed that PD-1 deficiency could also promote the expansion of germinal center (GC) B cells and malaria parasite-specific TFH cells in the spleens of ITV-immunized mice. These results suggest that PD-1 deficiency improves the protective efficacy of ITV-immunized mice by promoting the generation of malaria parasite-specific Ab and the expansion of GC B cells. The results of this study provide new evidence to support the negative function of PD-1 on humoral immunity and will guide the design of a more effective malaria vaccine.


Subject(s)
Antibodies, Protozoan/blood , Malaria Vaccines/immunology , Malaria/therapy , Programmed Cell Death 1 Receptor/deficiency , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Cell Proliferation , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Mice, Inbred BALB C , Spleen/immunology , T-Lymphocytes/immunology
18.
Int Immunol ; 26(7): 407-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24648472

ABSTRACT

MiR-155 (-/-) mice are highly resistant to experimental autoimmune encephalomyelitis (EAE), while Pdcd1 (-/-) mice develop a more severe form of the disease. To determine the conflicting roles of these two molecules in the disease, we generated miR-155 (-/-) Pdcd1 (-/-) double knockout (DKO) mice. We found that ablation of programmed cell death protein 1 (PD-1) expression in miR-155-deficient mice restored the susceptibility to EAE. The increased severity of the disease in DKO mice was accompanied by an enhanced T-cell infiltration into the brain as well as an increased production of pro-inflammatory cytokines IFN-γ and IL-17. Furthermore, the major contribution of the DKO to EAE was T-cell intrinsic since adoptive transfer of CD4(+) T cells from DKO donors promoted the disease in lymphopenic recipients. These results define PD-1 deficiency in miR-155 (-/-) mice as a promoting factor of autoimmune inflammation by increasing antigen-driven T-cell expansion and infiltration.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , MicroRNAs/immunology , Programmed Cell Death 1 Receptor/immunology , Adoptive Transfer , Animals , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Movement , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-17/biosynthesis , Interleukin-17/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Severity of Illness Index , Signal Transduction , Spleen/immunology , Spleen/pathology
19.
J Immunol ; 188(10): 4876-84, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22491251

ABSTRACT

PD-1, a member of the CD28 family of immune regulatory molecules, is expressed on activated T cells, interacts with its ligands, PD-L1/B7-H1 and PD-L2/B7-DC, on other cells, and delivers inhibitory signals to the T cell. We studied the role of this pathway in modulating autoreactive T cell responses in two models of myocarditis. In a CD8(+) T cell-mediated adoptive transfer model, we found that compared with Pd1(+/+) CD8(+) T cells, Pd1(-/-) CD8(+) T cells cause enhanced disease, with increased inflammatory infiltrate, particularly rich in neutrophils. Additionally, we show enhanced proliferation in vivo and enhanced cytotoxic activity of PD-1-deficient T lymphocytes against myocardial endothelial cells in vitro. In experimental autoimmune myocarditis, a disease model dependent on CD4(+) T cells, we show that mice lacking PD-1 develop enhanced disease compared with wild-type mice. PD-1-deficient mice displayed increased inflammation, enhanced serum markers of myocardial damage, and an increased infiltration of inflammatory cells, including CD8(+) T cells. Together, these studies show that PD-1 plays an important role in limiting T cell responses in the heart.


Subject(s)
Muscle Cells/immunology , Muscle Cells/pathology , Myocarditis/immunology , Myocarditis/pathology , Programmed Cell Death 1 Receptor/physiology , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Myocarditis/genetics , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , T-Lymphocyte Subsets/pathology
20.
Zhonghua Yi Xue Za Zhi ; 94(30): 2377-81, 2014 Aug 13.
Article in Zh | MEDLINE | ID: mdl-25399984

ABSTRACT

OBJECTIVE: To explore the functional changes of T lymphocyte from PD-1⁻/⁻ mice and the effects on atherogenic immune responses. METHODS: PD-1⁻/⁻ mice were mated with ApoE⁻/⁻ mice to obtain PD-1⁻/⁻ApoE⁻/⁻mice. PD-1⁻/⁻ApoE⁻/⁻ mice were used as control. Both groups had a high-lipid diet for 12 weeks. Then the samples of aortic sinuses were harvested for oil-red and immunohistochemical stains. Samples of spleen and sera were obtained. Numbers of lymphocytes in spleen were evaluated by flow cytometry (FACS). And the cells of proliferation were also measured. The intracellular and serum cytokines were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS: After a 12-week high-lipid diet, aortic root revealed more lesions in PD-1⁻/⁻ApoE⁻/⁻ mice than in PD-1⁺/⁺ ApoE⁻/⁻ controls ((0.51 ± 0.08) vs (0.29 ± 0.06) mm², t = 2.36, P < 0.01)). There was a significant increase in CD3 T cells in the lesions of PD-1⁻/⁻ApoE⁻/⁻ mice ((154.88 ± 36.64)/mm² vs (59.38 ± 25.28)/mm², t = 2.14, P < 0.01). In PD-1⁻/⁻ApoE⁻/⁻ mice, the numbers of total cells ((8.59 ± 0.55)× 107) and cells in CD4⁺ and CD8⁺ T lymphocyte subsets ((1.53 ± 0.18)× 107 and (1.41 ± 0.15)×107), increased significantly than those in control mice ((6.29 ± 0.39)×107, (0.94 ± 0.10)×107 and (0.70 ± 0.09)× 107 respectively, t = 3.43, 2.88, 4.03, all P < 0.05). Same changes were detected in both CD3⁺ CD62L(-) and CD3⁺ CD25⁺ cells. PD-1 deficiency in both CD4⁺ and CD8⁺ T cells resulted in enhanced proliferation by either macrophages or dendritic cells as antigen presenting cells (APCs). The supernatant levels of interleukin-2 (IL-2) ((53.38 ± 5.94) pg/ml), interferon-γ (IFN-γ) ((114.50 ± 9.69) pg/ml) and tumor necrosis factor-α (TNF-α) ((326.00 ± 22.25) pg/ml ) from PD-1⁻/⁻ T cells co-cultured with CD3 were significantly higher than those of control T cells ((15.63 ± 2.23), (33.25 ± 4.16) and (64.50 ± 8.17) pg/ml respectively, t = 5.95, 7.70, 11.03, all P < 0.01). However, only the serum levels of TNF-α increased in PD-1⁻/⁻ApoE⁻/⁻mice compared with control. CONCLUSIONS: PD-1 deficiency in T cells increase markedly immune responses. And it is one of the mechanisms in atherogenesis.


Subject(s)
Programmed Cell Death 1 Receptor/metabolism , Animals , Aorta , Apolipoproteins E , Cytokines , Dendritic Cells , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Macrophages , Mice , Programmed Cell Death 1 Receptor/deficiency , Spleen , T-Lymphocyte Subsets , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL