Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Immunol ; 208(5): 1057-1065, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35149531

ABSTRACT

T follicular regulatory (Tfr) cells are a subset of CD4+ T cells that express CXCR5 and migrate into germinal centers (GCs). They regulate GC reactions by communicating with T follicular helper (Tfh) and B cells. TNF inhibitors are used in inflammatory diseases; however, the generation of autoantibodies or anti-drug Abs sometimes causes problems. Because TNFR2 signaling is important for suppressive functions of regulatory T cells, we investigated the role of TNFR2 on human Tfr cells. Tfr cells stimulated with MR2-1 (an anti-TNFR2 agonistic Ab) were analyzed for cell proliferation, Foxp3 expression, and surface molecules. Tfh/B cell proliferation, IgM production, and differentiation in cocultures with MR2-1-stimulated Tfr cells were examined. Tfr cells express a high level of TNFR2. MR2-1 stimulation altered the gene expression profile of Tfr cells. Cell proliferation and Foxp3 expression of Tfr cells were enhanced by MR2-1. MR2-1-stimulated Tfr cells expressed ICOS and Programmed cell death protein 1 and significantly suppressed Tfh/B cell proliferation, IgM production, and B cell differentiation. TNFR2-stimulated Tfr cells retained the migration function according to the CXCL13 gradient. In conclusion, we showed that TNFR2-stiumulated Tfr cells can regulate Tfh and B cells. Aberrant antibody production during TNF inhibitor treatment might be, at least in part, associated with TNFR2 signaling inhibition in Tfr cells. In addition, expansion and maturation of Tfr cells via TNFR2 stimulation in vitro may be useful for a cell-based therapy in inflammatory and autoimmune diseases to control GC reactions.


Subject(s)
B-Lymphocytes/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/immunology , Autoimmune Diseases/therapy , B-Lymphocytes/cytology , B7-H1 Antigen/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Cell Proliferation , Chemokine CXCL13/metabolism , Forkhead Transcription Factors/biosynthesis , Gene Expression Profiling , Germinal Center/cytology , Humans , Immunoglobulin M/biosynthesis , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, CXCR5/metabolism , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors , Signal Transduction/immunology , Tumor Necrosis Factors/metabolism
2.
Pathol Oncol Res ; 26(1): 365-370, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30361907

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a highly malignant tumor, associated with poor patient prognoses, and high rates of morbidity and mortality. Currently, immune checkpoint therapy has brought new treatment strategy for NPC. The inducible T cell co-stimulator (ICOS) belongs to the B7-CD28 immunoglobulin superfamily, which is currently the subject of intense study due to great successes gained in treatment of different malignancies by disrupting their family members. However, the role of ICOS played in NPC remains poorly understood. Immunohistochemistry (IHC) was stained with the ICOS specific antibody and ICOS expression is decreased in patients with either lymphatic or distant metastasis and inversely associated with TNM stage of NPC patients. Importantly, high ICOS expression is significantly correlated with overall survival (OS) of NPC patients (N = 185, p < 0.001), and ICOS expression is also proved to be an independent prognostic factor by multivariate analysis. Surgical excised fresh NPC specimens (N = 185) were homogenized to analyze the specific cytokine expression by ELISA assay. ICOS expression level is associated with increased cytotoxic T lymphocyte number and high interferon IFNγ expression, the characteristics of Th1 cells. In addition, the correlation between the percentage of ICOS+ T cells in tumor tissue and survival was detected. Conclusively, expression of ICOS is associated with improved survival in NPC and percentage of ICOS+ cells acting as Th1 cells in primary tumor tissue may be a clinical biomarker for good prognosis of NPC patients.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Neoplasms/immunology , Th1 Cells/immunology , Adult , Aged , Biomarkers, Tumor/metabolism , Disease-Free Survival , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Neoplasms/mortality , Prognosis , Young Adult
3.
Nat Commun ; 9(1): 5344, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30559442

ABSTRACT

Foxp3-expressing CD4+ regulatory T (Treg) cells need to differentiate into effector Treg (eTreg) cells to maintain immune homeostasis. T-cell receptor (TCR)-dependent induction of the transcription factor IRF4 is essential for eTreg differentiation, but how IRF4 activity is regulated in Treg cells is still unclear. Here we show that the AP-1 transcription factor, JunB, is expressed in eTreg cells and promotes an IRF4-dependent transcription program. Mice lacking JunB in Treg cells develop multi-organ autoimmunity, concomitant with aberrant activation of T helper cells. JunB promotes expression of Treg effector molecules, such as ICOS and CTLA4, in BATF-dependent and BATF-independent manners, and is also required for homeostasis and suppressive functions of eTreg. Mechanistically, JunB facilitates the accumulation of IRF4 at a subset of IRF4 target sites, including those located near Icos and Ctla4. Thus, JunB is a critical regulator of IRF4-dependent Treg effector programs, highlighting important functions for AP-1 in Treg-mediated immune homeostasis.


Subject(s)
Interferon Regulatory Factors/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CTLA-4 Antigen/biosynthesis , Cell Differentiation/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/genetics
4.
Blood Adv ; 2(15): 1889-1900, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30087107

ABSTRACT

A better characterization of T-cell subsets in the microenvironment of classical Hodgkin lymphoma (cHL) would help to develop immunotherapies. Using multicolor flow cytometry, we identified in 6 of 43 cHL tissue samples a previously unrecognized subset of CD8 T cells coexpressing CXCR5 and inducible T-cell costimulator (ICOS) molecules (CD8CXCR5+ICOS+). These cells shared phenotypic features with follicular helper T (TFH) cells including low CCR7 expression together with high expression of B-cell lymphoma-6, programmed cell death 1, B and T lymphocyte attenuator, CD200, and OX40. They had deficient cytotoxicity, low interferon-γ secretion, and common functional properties with intratumoral CD4+ TFH cells, such as production of interleukin-4 (IL-4), IL-21, CXCL13, and capacity to sustain B cells. Gene profiling analysis showed a significant similarity between the signatures of CD8CXCR5+ICOS+ T cells and CD4+ TFH cells. Benign lymphadenitis tissues (n = 8) were devoid of CD8CXCR5+ICOS+ cells. Among the 35 B-cell lymphoma tissues analyzed, including follicular lymphomas (n = 13), diffuse large cell lymphomas (n = 12), marginal zone lymphomas (MZLs; n = 3), mantle cell lymphomas (n = 3), and chronic lymphocytic leukemias (n = 4), only 1 MZL sample contained CD8CXCR5+ICOS+ cells. Lymphoma tumors with CD8CXCR5+ICOS+ cells shared common histopathological features including residual germinal centers, and contained high amounts of activated CD8CXCR5-ICOS+ cells. These data demonstrate a CD8 T-cell differentiation pathway leading to the acquisition of some TFH similarities. They suggest a particular immunoediting process with global CD8 activation acting mainly, but not exclusively, in HL tumors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Hodgkin Disease/metabolism , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Neoplasm Proteins/biosynthesis , Receptors, CXCR5/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cytokines/metabolism , Female , Hodgkin Disease/pathology , Humans , Male
5.
Invest Ophthalmol Vis Sci ; 57(15): 6815-6823, 2016 12 01.
Article in English | MEDLINE | ID: mdl-28002569

ABSTRACT

Purpose: The interaction between the inducible costimulatory molecule (ICOS) and ICOS ligand (ICOSL) has been implicated in the differentiation and functions of T cells. The purpose of the present study was to determine the role of ICOS-ICOSL in the immune privilege of corneal allografts. Methods: Expression of ICOS and ICOSL mRNA from mouse eyes was assessed by RT-PCR. Corneas of C57BL/6 mice were orthotopically transplanted into the eyes of ICOS-/- BALB/c recipients and BALB/c wild-type (WT) recipients treated with anti-ICOSL mAb, and graft survival was assessed. A separate set of WT and ICOS-/- BALB/c mice received an anterior chamber injection of C57BL/6 splenocytes, and induction of allospecific anterior chamber-associated immune deviation (ACAID) was assessed. In vitro, cornea was incubated with T cells from WT and ICOS-/- BALB/c mice, and destruction of corneal endothelial cells (CECs) and the population of Foxp3+ CD25+ CD4+ T cells was assessed. Results: Inducible costimulatory molecule ligand mRNA was constitutively expressed in the cornea, iris-ciliary body, and retina. Allograft survival in ICOS-/- recipients and WT recipients treated with anti-ICOSL mAb was significantly shorter than in control recipients. Anterior chamber-associated immune deviation was induced less efficiently in ICOS-/- mice. Destruction of CECs by alloreactive ICOS-/- T cells was enhanced compared with WT T cells. After coincubation with allogeneic corneal tissue, the proportion of regulatory T cells was significantly greater among WT T cells than in ICOS-/- T cells. Conclusions: The expression of ICOSL in the cornea and the ICOS-mediated induction of Foxp3+ CD4+ regulatory T cells may contribute to successful corneal allograft survival.


Subject(s)
Cornea/metabolism , Corneal Transplantation , Gene Expression Regulation , Graft Survival/immunology , Immunity, Cellular , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Protein/genetics , Animals , Cornea/immunology , Cornea/pathology , Flow Cytometry , Graft Rejection/genetics , Graft Rejection/immunology , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
6.
Genet Mol Res ; 15(2)2016 May 20.
Article in English | MEDLINE | ID: mdl-27323062

ABSTRACT

As the most common cardiac disease, myocardial infarction is followed by hypertrophy of cardiac myocytes and reconstruction of ventricular structure. The up-regulation of a series of factors including metalloproteinases, inflammatory factors, and growth factors after primary infarction lead to the hypertrophy, apoptosis, necrosis, and fibroblast proliferation in cardiac muscle tissues. Recent studies have reported on the potency of small interfering RNA (siRNA) in treating cardiac diseases. We thus investigated the efficacy of inducible co-stimulatory molecule (ICOS)-specific siRNA silencing in myocardial hypertrophy in a cardiac infarction rat model. This cardiac infarction model was prepared by ligating the left anterior descending coronary artery. ICOS-siRNA treatment was administered in parallel with non-sense siRNA. After 18 days, the cross-sectional area of cardiac muscle tissues and the left ventricle weight index were measured, along with ICOS mRNA and protein expression levels, and pathological staining. Compared to those in the control groups, in myocardial infarcted rats, the application of ICOS-siRNA effectively decreased the left ventricle weight index, as well as the surface area of cardiac myocytes. Both mRNA and protein levels of ICOS were also significantly decreased. HE staining was consistent with these results. In conclusion, ICOS-targeted siRNA can effectively silence gene expression of ICOS, and provided satisfactory treatment efficacy for myocardial cell hypertrophy after infarction.


Subject(s)
Hypertrophy/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Myocardial Infarction/genetics , RNA, Small Interfering/administration & dosage , Animals , Gene Expression Regulation/drug effects , Genetic Therapy/methods , Heart Ventricles/pathology , Humans , Hypertrophy/pathology , Hypertrophy/therapy , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats
7.
Medicine (Baltimore) ; 94(27): e1125, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26166114

ABSTRACT

T follicular helper (Tfh) cells are important components in development of specific humoral immune responses; whether the number and biology of Tfh cells is impaired in HIV-1-infected children is not yet studied.The frequency, phenotype, and function of Tfh cells and B cells were determined in blood of HIV-1-infected children receiving antiretroviral therapy (ART) and age-matched controls. Flow cytometry was used to characterize the frequency of Tfh cells and B cell subsets. Cytokine expression was measured after in vitro activation of Tfh cells.A reduced frequency of memory Tfh cells (P < 0.001) was identified in HIV-1-infected children and, on these cells, a reduced expression of programmed death-1 (PD-1) and inducible T cell costimulator (ICOS) (P < 0.001 and P < 0.01). Upon activation, the capacity of Tfh cells to express IL-4, an important cytokine for B cell function, was impaired in HIV-1-infected children.B cell subpopulations in HIV-1-infected children displayed significant differences from the control group: the frequency of resting memory (RM) B cells was reduced (P < 0.01) whereas the frequency of exhausted memory B cells increased (P < 0.001). Interestingly, the decline of RM cells correlated with the reduction of memory Tfh cells (P = 0.02).Our study shows that function and phenotype of Tfh cells, pivotal cells for establishment of adaptive B cell responses, are impaired during HIV-1 infection in children. A consistent reduction of memory Tfh cells is associated with declined frequencies of RM B cells, creating a novel link between dysfunctional features of these cell types, major players in establishment of humoral immunity.


Subject(s)
Anti-Retroviral Agents/therapeutic use , B-Lymphocytes/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Child , Child, Preschool , Cytokines/biosynthesis , Female , Flow Cytometry , HIV-1 , Humans , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocyte Activation , Male , Phenotype , Programmed Cell Death 1 Receptor/biosynthesis
8.
J Immunol ; 195(3): 994-1005, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116502

ABSTRACT

The goal of an HIV vaccine is to generate robust and durable protective Ab. Vital to this goal is the induction of CD4(+) T follicular helper (TFH) cells. However, very little is known about the TFH response to HIV vaccination and its relative contribution to magnitude and quality of vaccine-elicited Ab titers. In this study, we investigated these questions in the context of a DNA/modified vaccinia virus Ankara SIV vaccine with and without gp140 boost in aluminum hydroxide in rhesus macaques. In addition, we determined the frequency of vaccine-induced CD4(+) T cells coexpressing chemokine receptor, CXCR5 (facilitates migration to B cell follicles) in blood and whether these responses were representative of lymph node TFH responses. We show that booster modified vaccinia virus Ankara immunization induced a distinct and transient accumulation of proliferating CXCR5(+) and CXCR5(-) CD4 T cells in blood at day 7 postimmunization, and the frequency of the former but not the latter correlated with TFH and B cell responses in germinal centers of the lymph node. Interestingly, gp140 boost induced a skewing toward CXCR3 expression on germinal center TFH cells, which was strongly associated with longevity, avidity, and neutralization potential of vaccine-elicited Ab response. However, CXCR3(+) cells preferentially expressed the HIV coreceptor CCR5, and vaccine-induced CXCR3(+)CXCR5(+) cells showed a moderate positive association with peak viremia following SIV251 infection. Taken together, our findings demonstrate that vaccine regimens that elicit CXCR3-biased TFH cell responses favor Ab persistence and avidity but may predispose to higher acute viremia in the event of breakthrough infections.


Subject(s)
SAIDS Vaccines/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Viremia/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Antibodies, Viral/blood , Glycoproteins/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymph Nodes/cytology , Lymph Nodes/immunology , Macaca mulatta , Male , Programmed Cell Death 1 Receptor/biosynthesis , Receptors, CCR5/biosynthesis , Receptors, CXCR3/biosynthesis , Receptors, CXCR5/biosynthesis , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccination/veterinary , Vaccines, DNA , Viral Load/immunology , Viremia/virology
9.
J Immunol ; 195(3): 944-52, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26109645

ABSTRACT

Regulatory T cells (Tregs), a subset of CD4(+) T cells, dramatically accumulate with age in humans and mice and contribute to age-related immune suppression. Recently, we showed that a majority of accumulating Tregs in aged mice expressed low levels of CD25, and their accrual is associated with declining levels of IL-2 in aged mice. In this study, we further investigated the origin of CD25(lo) Tregs in aged mice. First, aged Tregs had high expression of neuropilin-1 and Helios, and had a broad Vß repertoire. Next, we analyzed the gene expression profile of Tregs, naive T cells, and memory T cells in aged mice. We found that the gene expression profile of aged CD25(lo) Tregs were more related to young CD25(lo) Tregs than to either naive or memory T cells. Further, the gene expression profile of aged Tregs was consistent with recently described "effector" Tregs (eTregs). Additional analysis revealed that nearly all Tregs in aged mice were of an effector phenotype (CD44(hi)CD62L(lo)) and could be further characterized by high levels of ICOS and CD69. ICOS contributed to Treg maintenance in aged mice, because in vivo Ab blockade of ICOSL led to a loss of eTregs, and this loss was rescued in Bim-deficient mice. Further, serum levels of IL-6 increased with age and contributed to elevated expression of ICOS on aged Tregs. Finally, Treg accrual was significantly blunted in aged IL-6-deficient mice. Together, our data show a role for IL-6 in promoting eTreg accrual with age likely through maintenance of ICOS expression.


Subject(s)
Aging/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-6/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Apoptosis Regulatory Proteins/genetics , Base Sequence , Bcl-2-Like Protein 11 , Cell Death , Cell Survival , DNA-Binding Proteins/biosynthesis , Gene Expression Profiling , Hyaluronan Receptors/biosynthesis , Immunologic Memory/genetics , Immunologic Memory/immunology , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/blood , Interleukin-6/genetics , L-Selectin/biosynthesis , Lectins, C-Type/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilin-1/biosynthesis , Proto-Oncogene Proteins/genetics , Sequence Analysis, DNA , Transcription Factors/biosynthesis
10.
J Immunol ; 195(3): 832-40, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26071562

ABSTRACT

Follicular regulatory T cells (TFR) have been extensively characterized in mice and participate in germinal center responses by regulating the maturation of B cells and production of (auto)antibodies. We report that circulating TFR are phenotypically distinct from tonsil-derived TFR in humans. They have a lower expression of follicular markers, and display a memory phenotype and lack of high expression of B cell lymphoma 6 and ICOS. However, the suppressive function, expression of regulatory markers, and FOXP3 methylation status of blood TFR is comparable with tonsil-derived TFR. Moreover, we show that circulating TFR frequencies increase after influenza vaccination and correlate with anti-flu Ab responses, indicating a fully functional population. Multiple sclerosis (MS) was used as a model for autoimmune disease to investigate alterations in circulating TFR. MS patients had a significantly lower frequency of circulating TFR compared with healthy control subjects. Furthermore, the circulating TFR compartment of MS patients displayed an increased proportion of Th17-like TFR. Finally, TFR of MS patients had a strongly reduced suppressive function compared with healthy control subjects. We conclude that circulating TFR are a circulating memory population derived from lymphoid resident TFR, making them a valid alternative to investigate alterations in germinal center responses in the context of autoimmune diseases, and TFR impairment is prominent in MS.


Subject(s)
B-Cell Maturation Antigen/biosynthesis , Influenza Vaccines/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adult , Antibodies/blood , B-Lymphocytes/immunology , Female , Forkhead Transcription Factors/metabolism , Humans , Immunologic Memory/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Male , Methylation , Multiple Sclerosis/blood , Vaccination , Young Adult
11.
Immunity ; 43(1): 161-74, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26092469

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) and regulatory T (Treg) cells are systemically induced by helminth infection but also sustain metabolic homeostasis in adipose tissue and contribute to tissue repair during injury. Here we show that interleukin-33 (IL-33) mediates activation of ILC2s and Treg cells in resting adipose tissue, but also after helminth infection or treatment with IL-2. Unexpectedly, ILC2-intrinsic IL-33 activation was required for Treg cell accumulation in vivo and was independent of ILC2 type 2 cytokines but partially dependent on direct co-stimulatory interactions via ICOSL-ICOS. IFN-γ inhibited ILC2 activation and Treg cell accumulation by IL-33 in infected tissue, as well as adipose tissue, where repression increased with aging and high-fat diet-induced obesity. IL-33 and ILC2s are central mediators of type 2 immune responses that promote tissue and metabolic homeostasis, and IFN-γ suppresses this pathway, likely to promote inflammatory responses and divert metabolic resources necessary to protect the host.


Subject(s)
Interferon-gamma/immunology , Interleukins/immunology , Lymphocyte Activation/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Aging/immunology , Animals , Diet, High-Fat , Enzyme Activation/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/immunology , Interferon-gamma/pharmacology , Interleukin-2/pharmacology , Interleukin-33 , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/immunology , Lectins, C-Type , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Mice, Transgenic , Nippostrongylus/immunology , Obesity/immunology , Receptors, Immunologic/biosynthesis , Strongylida Infections/parasitology
12.
J Immunol ; 195(3): 1071-81, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26085685

ABSTRACT

The IL-6 cytokine family utilizes the common signal transduction molecule gp130, which can mediate a diverse range of outcomes. To clarify the role of gp130 signaling in vivo during acute viral infection, we infected Cd4-cre Il6st(fl/fl) mice, in which gp130 is conditionally ablated in T cells, with acute lymphocytic choriomeningitis virus. We found that by day 12, but not at day 8, after infection the number of virus-specific CD4(+) T cells was reduced in the absence of gp130, and this was sustained for up to 2 mo postinfection. Additionally, gp130-deficient T follicular helper cells had lower expression of Maf, IL-21, and ICOS, and this was accompanied by a reduction in the proportion of germinal center B cells and plasmablasts. Remarkably, at 2 mo postinfection the proportion of IgG2a/c(+) memory B cells and the systemic levels of lymphocytic choriomeningitis virus-specific IgG2 Abs were dramatically decreased, whereas there was a corresponding increase in IgG1(+) memory B cells and virus-specific IgG1 Abs. In the same animals gp130-deficient virus-specific CD8(+) T cells showed a reduced proportion of memory cells, which expressed lower levels of Tcf7, and displayed diminished recall responses on secondary infection. Mixed bone marrow chimeras revealed that the aforementioned gp130 effects on CD4(+) T cells were cell intrinsic. Overall, our data show that gp130 signaling in T cells influences the quantity and quality of long-lasting CD4(+) T cell responses as well as CD8(+) T cell- and Ab-mediated immunity after acute viral infection.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokine Receptor gp130/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/cytology , Cytokine Receptor gp130/genetics , Germinal Center/cytology , Germinal Center/immunology , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immunoglobulin G/immunology , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Interleukin-6/immunology , Interleukins/biosynthesis , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-maf/biosynthesis , Signal Transduction/immunology
13.
Genes Immun ; 16(2): 151-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25569260

ABSTRACT

Lymphocyte apoptosis is mainly induced by either death receptor-dependent activation of caspase-8 or mitochondria-dependent activation of caspase-9. Mutations in caspase-8 lead to autoimmunity/lymphoproliferation and immunodeficiency. This work describes a heterozygous H237P mutation in caspase-9 that can lead to similar disorders. H237P mutation was detected in two patients: Pt1 with autoimmunity/lymphoproliferation, severe hypogammaglobulinemia and Pt2 with mild hypogammaglobulinemia and Burkitt lymphoma. Their lymphocytes displayed defective caspase-9 activity and decreased apoptotic and activation responses. Transfection experiments showed that mutant caspase-9 display defective enzyme and proapoptotic activities and a dominant-negative effect on wild-type caspase-9. Ex vivo analysis of the patients' lymphocytes and in vitro transfection experiments showed that the expression of mutant caspase-9 correlated with a downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. Both patients carried a second inherited heterozygous mutation missing in the relatives carrying H237P: Pt1 in the transmembrane activator and CAML interactor (TACI) gene (S144X) and Pt2 in the perforin (PRF1) gene (N252S). Both mutations have been previously associated with immunodeficiencies in homozygosis or compound heterozygosis. Taken together, these data suggest that caspase-9 mutations may predispose to immunodeficiency by cooperating with other genetic factors, possibly by downregulating the expression of BAFFR and ICOS.


Subject(s)
B-Cell Activation Factor Receptor/biosynthesis , Caspase 9/genetics , Immunologic Deficiency Syndromes/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphoproliferative Disorders/genetics , Mutation , Adolescent , Adult , Apoptosis/genetics , Apoptosis/immunology , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , Caspase 9/immunology , Down-Regulation , HEK293 Cells , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Male , Pedigree
14.
J Immunol ; 193(8): 4010-20, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25217167

ABSTRACT

IL-33 is a recently characterized IL-1 family member that is proposed to function as an alarmin, or endogenous signal of cellular damage, as well as act as a pleiotropic cytokine. The ability of IL-33 to potentiate both Th1 and Th2 immunity supports its role in pathogen clearance and disease immunopathology. Yet, IL-33 restrains experimental colitis and transplant rejection by expanding regulatory T cells (Treg) via an undefined mechanism. We sought to determine the influence of IL-33 on hematopoietic cells that drives Treg expansion and underlies the therapeutic benefit of IL-33 administration. In this study, we identify a feedback loop in which conventional mouse CD11c(+) dendritic cells (DC) stimulated by IL-33 secrete IL-2 to selectively expand IL-33R(ST2(+))- suppressive CD4(+)Foxp3(+) Treg. Interestingly, this occurs in the absence of classical DC maturation, and DC-derived (innate) IL-2 increases ST2 expression on both DC and interacting Treg. ST2(+) Treg represent an activated subset of Foxp3(+) cells, demonstrated to be ICOS(high)CD44(high) compared with their ST2(-) counterparts. Furthermore, although studies have shown that IL-33-exposed DC promote Th2 responses, we reveal that ST2(+) DC are required for IL-33-mediated in vitro and in vivo Treg expansion. Thus, we have uncovered a relationship between IL-33 and innate IL-2 that promotes the selective expansion of ST2(+) Treg over non-Treg. These findings identify a novel regulatory pathway driven by IL-33 in immune cells that may be harnessed for therapeutic benefit or for robust expansion of Treg in vitro and in vivo.


Subject(s)
Dendritic Cells/drug effects , Interleukin-2/metabolism , Interleukins/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Forkhead Transcription Factors/biosynthesis , Hyaluronan Receptors/biosynthesis , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/biosynthesis , Signal Transduction/immunology , Th1 Cells/immunology , Th2 Cells/immunology
15.
J Cutan Pathol ; 40(12): 1006-13, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24274425

ABSTRACT

BACKGROUND: CD4+ small/medium-sized pleomorphic T-cell lymphoma (SMPTCL) is a controversial primary cutaneous lymphoma, in which the candidate neoplastic cells express a follicular T-helper phenotype. We describe 16 cases of SMPTCL and compare expression of PD-1, CXCL-13 and ICOS in these tumors with 40 dermatitis cases. METHODS: Histopathologic examination and immunocytochemistry were performed for 16 tumors and 40 assorted dermatitis cases. RESULTS: All but one patient presented with solitary lesions. Each biopsy revealed a dense nodular non-epitheliotropic infiltrate of atypical T-cells. Neoplastic cells were CD3+/CD4+/CD8(-)/CD30(-). Cutaneous recurrence occurred in one patient over a median follow up of 8 months (range 5-36). All tumors widely expressed PD-1 and ICOS to a lesser extent. CXCL-13 stained much fewer cells. Of the dermatitis cases, PD-1 (most numerous) and ICOS labeled lymphoid cells in all cases, albeit fewer than in the tumors, and CXCL-13 was negative in 32. A rosette pattern of PD-1 expression was identified in all the SMPTCL cases but not in dermatitis. CONCLUSIONS: There remains uncertainty about the appropriate nosological status of SMPTCL, which some authors consider to be a pseudolymphoma. However, this study suggests a significant difference in the prevalence and pattern of follicular T-helper cell markers between this tumor and lymphoid proliferations known to be reactive.


Subject(s)
Dermatitis , Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell, Cutaneous , Neoplasm Proteins/biosynthesis , Skin Neoplasms , T-Lymphocytes, Helper-Inducer , Adult , Aged , Chemokine CXCL13/biosynthesis , Dermatitis/metabolism , Dermatitis/pathology , Female , Humans , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Male , Middle Aged , Programmed Cell Death 1 Receptor/biosynthesis , Retrospective Studies , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
16.
BMC Immunol ; 14: 45, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24083425

ABSTRACT

BACKGROUND: The immune system consists of multiple preformed and more specific adaptive immune responses, which are all subject to both positive and negative regulation. Programmed cell death protein 1 (PD-1) is a cell surface ligand implicated in the induction of anergy, Inducible T-cell Costimulator (ICOS) plays a stimulatory role in the development of both CD4+ and CD8+ T-cells, Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) plays a role in inhibitory regulation of T-cell activity, and T cell immunoglobulin and mucin protein 3 (Tim-3) has been described as a negative regulatory molecule in CD4+ helper type 1 cells and CD8+ cytotoxic type 1 cells. Each of these ligands is induced with T-cell activation allowing greater opportunity to have a regulatory role. RESULTS: Flow cytometry was used to quantitate the expression of PD-1, ICOS, CTLA-4 and Tim-3 in human T-cells from geriatric and younger subjects both at baseline and after in vitro induction by mitogen. The magnitude of expression of the molecules increased significantly on activated blasts after mitogen stimulation compared to their baseline levels in resting cells. The increase in CTLA-4 expressing CD8+ T-cells was significantly higher after in vitro induction in older persons, while the increase in cells expressing Tim-3 and PD-1 was significantly reduced. In CD4+ T-cells, a greater increase in CTLA-4 expressing cells in older persons was the only difference between the age groups. CONCLUSIONS: We found several significant changes in the older individuals in regulatory elements of the adaptive immune system that occur particularly after immune activation. These differences could have ramifications to autoimmunity as well as immunology against infection and tumors.


Subject(s)
Aging/immunology , CTLA-4 Antigen/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Membrane Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Adult , Age Factors , Aged , Aged, 80 and over , Aging/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/biosynthesis , Cells, Cultured , Flow Cytometry , Hepatitis A Virus Cellular Receptor 2 , Humans , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes/metabolism , Young Adult
17.
Eur J Immunol ; 43(12): 3219-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24030473

ABSTRACT

CD4(+) T follicular helper (TFH) cells are central for generation of long-term B-cell immunity. A defining phenotypic attribute of TFH cells is the expression of the chemokine R CXCR5, and TFH cells are typically identified by co-expression of CXCR5 together with other markers such as PD-1, ICOS, and Bcl-6. Herein, we report high-level expression of the nutrient transporter folate R 4 (FR4) on TFH cells in acute viral infection. Distinct from the expression profile of conventional TFH markers, FR4 was highly expressed by naive CD4(+) T cells, was downregulated after activation and subsequently re-expressed on TFH cells. Furthermore, FR4 expression was maintained, albeit at lower levels, on memory TFH cells. Comparative gene expression profiling of FR4(hi) versus FR4(lo) Ag-specific CD4(+) effector T cells revealed a molecular signature consistent with TFH and TH1 subsets, respectively. Interestingly, genes involved in the purine metabolic pathway, including the ecto-enzyme CD73, were enriched in TFH cells compared with TH1 cells, and phenotypic analysis confirmed expression of CD73 on TFH cells. As there is now considerable interest in developing vaccines that would induce optimal TFH cell responses, the identification of two novel cell surface markers should be useful in characterization and identification of TFH cells following vaccination and infection.


Subject(s)
Gene Expression Regulation/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes, Helper-Inducer/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Acute Disease , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression Regulation/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR5/biosynthesis , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
18.
Am J Transplant ; 13(9): 2293-307, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23914930

ABSTRACT

Endogenous memory CD8 T cells infiltrate MHC-mismatched cardiac allografts within 12-24 h posttransplant in mice and are activated to proliferate and produce IFN-γ. To more accurately assess the graft injury directly imposed by these endogenous memory CD8 T cells, we took advantage of the ability of anti-LFA-1 mAb given to allograft recipients on days 3 and 4 posttransplant to inhibit the generation of primary effector T cells. When compared to grafts from IgG-treated recipients on day 7 posttransplant, allografts from anti-LFA-1 mAb-treated recipients had increased numbers of CD8 T cells but these grafts had marked decreases in expression levels of mRNA encoding effector mediators associated with graft injury and decreases in donor-reactive CD8 T cells producing IFN-γ. Despite this decreased activity within the allograft, CD8 T cells in allografts from recipients treated with anti-LFA-1 mAb continued to proliferate up to day 7 posttransplant and did not upregulate expression of the exhaustion marker LAG-3 but did have decreased expression of ICOS. These results indicate that endogenous memory CD8 T cells infiltrate and proliferate in cardiac allografts in mice but do not express sufficient levels of functions to mediate overt graft injury and acute rejection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Heart Transplantation , Transplantation Immunology , Allografts , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/drug effects , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphocyte Activation , Mice , Lymphocyte Activation Gene 3 Protein
19.
Mol Med Rep ; 7(4): 1197-202, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23426717

ABSTRACT

Previous studies have indicated that immune dysregulation is an important cause of HCV­mediated damage to the liver. Co­stimulation signals, including programmed cell death protein 1 (PD­1) and inducible T­cell co­stimulator (ICOS), have been demonstrated to be involved in the pathogenesis of HCV. The purpose of this study was to investigate the soluble PD­1 (sPD­1) and soluble ICOS (sICOS) serum levels in chronic HCV patients, and to elucidate the association of sPD­1 and sICOS levels with pathological injury of chronic HCV infection. Sixty­three patients with chronic HCV and 30 normal controls were recruited for this study. The serum concentration levels of sPD­1 and sICOS were measured by enzyme­linked immunosorbent assay, and the mRNA levels of PD­1 and ICOS were detected using real­time RT­PCR. The serum sPD­1 and sICOS levels were significantly elevated in the chronic HCV patient group compared with the normal control group. Furthermore, the relative mRNA expression levels of these proteins were also increased in chronic HCV patients. sPD­1 and sICOS serum levels were significantly correlated with anti­HCV antibody levels, but not with HCV RNA. Aberrant sPD­1 and sICOS serum levels may reflect the dysregulation of T­cell activation, and are associated with the pathological injury of chronic HCV infection.


Subject(s)
Hepatitis C, Chronic/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Liver/injuries , Programmed Cell Death 1 Receptor/biosynthesis , Adult , Apoptosis Regulatory Proteins/genetics , Female , Gene Expression Regulation , Hepacivirus/immunology , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/pathology , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Liver/immunology , Male , Middle Aged , Programmed Cell Death 1 Receptor/genetics , RNA, Messenger/biosynthesis
20.
Cancer Res ; 72(23): 6130-41, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23026134

ABSTRACT

Human breast tumors are infiltrated by memory CD4(+) T cells along with increased numbers of regulatory T cells (Treg) and plasmacytoid dendritic cells (pDC) that facilitate immune escape and correlate with poor prognosis. Here, we report that inducible costimulatory molecule (ICOS), a T cell costimulatory molecule of the CTLA4/PD1/CD28 family, is expressed mostly by tumor-associated Treg in primary breast tumors. A large proportion of these ICOS(+) Treg were Ki67(+) and this evident proliferative expansion was found to rely on interactions with tumor-associated pDC. Indeed, tumor-associated Treg highly expanded in presence of pDC but failed to proliferate under CD3/CD28 signal. In vitro experiments revealed that the addition of a neutralizing anti-ICOS antibody blocked pDC-induced Treg expansion and interleukin-10 secretion by memory CD4(+) T cells, establishing a pivotal role for ICOS in this process. Supporting these findings, the presence of ICOS(+) cells in clinical specimens of breast cancer correlated with a poor prognosis. Together, our results highlight an important relationship between Treg and pDC in breast tumors, and show that ICOS/ICOS-L interaction is a central event in immunosuppression of tumor-associated memory CD4(+) T cells. These findings strongly rationalize antibody-mediated ICOS blockade as a powerful clinical strategy to correct immune escape and promote therapeutic responses in breast cancer.


Subject(s)
Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Disease Progression , Female , Humans , Immunohistochemistry , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Activation , Retrospective Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...