Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
Immunity ; 54(2): 291-307.e7, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33450188

ABSTRACT

The role of innate immune cells in allergen immunotherapy that confers immune tolerance to the sensitizing allergen is unclear. Here, we report a role of interleukin-10-producing type 2 innate lymphoid cells (IL-10+ ILC2s) in modulating grass-pollen allergy. We demonstrate that KLRG1+ but not KLRG1- ILC2 produced IL-10 upon activation with IL-33 and retinoic acid. These cells attenuated Th responses and maintained epithelial cell integrity. IL-10+ KLRG1+ ILC2s were lower in patients with grass-pollen allergy when compared to healthy subjects. In a prospective, double-blind, placebo-controlled trial, we demonstrated that the competence of ILC2 to produce IL-10 was restored in patients who received grass-pollen sublingual immunotherapy. The underpinning mechanisms were associated with the modification of retinol metabolic pathway, cytokine-cytokine receptor interaction, and JAK-STAT signaling pathways in the ILCs. Altogether, our findings underscore the contribution of IL-10+ ILC2s in the disease-modifying effect by allergen immunotherapy.


Subject(s)
Interleukin-10/metabolism , Lymphocytes/immunology , Rhinitis, Allergic, Seasonal/immunology , Sublingual Immunotherapy/methods , Adult , Allergens/immunology , Double-Blind Method , Female , Humans , Immune Tolerance , Immunity, Innate , Janus Kinases/metabolism , Lectins, C-Type/metabolism , Male , Middle Aged , Placebo Effect , Poaceae/immunology , Pollen/immunology , Receptors, Immunologic/metabolism , Rhinitis, Allergic, Seasonal/therapy , STAT Transcription Factors/metabolism , Signal Transduction , Th2 Cells/immunology , Treatment Outcome , Vitamin A/metabolism , Young Adult
2.
Cell Commun Signal ; 22(1): 337, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898461

ABSTRACT

Killer cell lectin-like receptor G1 (KLRG1) is an immune checkpoint receptor expressed predominantly in NK and T-cell subsets that downregulates the activation and proliferation of immune cells and participates in cell-mediated immune responses. Accumulating evidence has demonstrated the importance of KLRG1 as a noteworthy disease marker and therapeutic target that can influence disease onset, progression, and prognosis. Blocking KLRG1 has been shown to effectively mitigate the effects of downregulation in various mouse tumor models, including solid tumors and hematologic malignancies. However, KLRG1 inhibitors have not yet been approved for human use, and the understanding of KLRG1 expression and its mechanism of action in various diseases remains incomplete. In this review, we explore alterations in the distribution, structure, and signaling pathways of KLRG1 in immune cells and summarize its expression patterns and roles in the development and progression of autoimmune diseases, infectious diseases, and cancers. Additionally, we discuss the potential applications of KLRG1 as a tool for tumor immunotherapy.


Subject(s)
Lectins, C-Type , Neoplasms , Receptors, Immunologic , Humans , Receptors, Immunologic/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/antagonists & inhibitors , Animals , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Biomarkers/metabolism , Signal Transduction , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/drug therapy , Immunotherapy
3.
Lupus ; 32(4): 549-559, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36876466

ABSTRACT

OBJECTIVES: Killer cell lectin-like receptor G 1 (KLRG1), a transmembrane receptor with inhibitory capacity expressed in human immune cells, emerged as a novel susceptibility gene for systemic lupus erythematosus (SLE). The aim of this study was to investigate the expression of KLRG1 in SLE patients compared to healthy controls (HC) on both NK and T cells and to evaluate its possible involvement in SLE pathogenesis. METHODS: Eighteen SLE patients and twelve healthy controls were enrolled. Peripheral blood mononuclear cells (PBMCs) from these patients were phenotypically characterized by immunofluorescence and flow cytometry. The effect of the hydroxychloroquine (HCQ) in vitro on KLRG1 expression and its signaling mediated functions in NK cells were analyzed. RESULTS: KLRG1 expression was significantly reduced on the analyzed immune cell populations in SLE patients compared to HC, especially on total NK cells. Moreover, expression of KLRG1 on total NK cells inversely correlated with the SLEDAI-2K. A direct association between KLRG1 expression on NK cells and patients' treatment with HCQ was observed. In vitro treatment with HCQ increased KLRG1 expression on NK cells. In HC, KLRG1+ NK cells showed reduced degranulation and IFNγ production, while in SLE patient, this inhibition occurred only for the IFNγ production. CONCLUSION: With this study we revealed a reduced expression and an impaired function of KLRG1 on NK cells in SLE patients. These results suggest a possible role of KLRG1 in the pathogenesis of SLE and as a novel biomarker of this disease.


Subject(s)
Hydroxychloroquine , Lupus Erythematosus, Systemic , Humans , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Leukocytes, Mononuclear/metabolism , Killer Cells, Natural , Flow Cytometry , Receptors, Immunologic/metabolism , Lectins, C-Type
4.
J Allergy Clin Immunol ; 149(1): 237-251.e12, 2022 01.
Article in English | MEDLINE | ID: mdl-33964300

ABSTRACT

BACKGROUND: Allergic asthma is more severe and frequent in women than in men. In male mice, androgens negatively control group 2 innate lymphoid cell (ILC2) development and function by yet unknown mechanisms. OBJECTIVES: We sought to investigate the impact of androgen on ILC2 homeostasis and IL-33-mediated inflammation in female lungs. We evaluated the role of androgen receptor (AR) signaling and the contribution of the putative inhibitory receptor killer cell lectin-like receptor G1 (KLRG1). METHODS: Subcutaneous pellets mimicking physiological levels of androgen were used to treat female mice together with mice expressing a reporter enzyme under the control of androgen response elements and mixed bone marrow chimeras to assess the cell-intrinsic role of AR activation within ILC2s. We generated KLRG1-deficient mice. RESULTS: We established that lung ILC2s express a functionally active AR that can be in vivo targeted with exogenous androgens to negatively control ILC2 homeostasis, proliferation, and function. Androgen signaling upregulated KLRG1 on ILC2s, which inhibited their proliferation on E-cadherin interaction. Despite evidence that KLRG1 impaired the competitive fitness of lung ILC2s during inflammation, KLRG1 deficiency neither alters in vivo ILC2 numbers and functions, nor did it lead to hyperactive ILC2s in either sexes. CONCLUSIONS: AR agonists can be used in vivo to inhibit ILC2 homeostatic numbers and ILC2-dependent lung inflammation through cell-intrinsic AR activation. Although androgen signals in ILC2s to upregulate KLRG1, we demonstrate that KLRG1 is dispensable for androgen-mediated inhibition of pulmonary ILC2s.


Subject(s)
Androgens/pharmacology , Lectins, C-Type/immunology , Lymphocytes/immunology , Pneumonia/immunology , Receptors, Immunologic/immunology , Testosterone/pharmacology , Animals , Female , Interleukin-33/immunology , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/pathology , Sex Characteristics , Signal Transduction
5.
BMC Immunol ; 23(1): 52, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36316644

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are the most dominant ILCs in heart tissue, and sex-related differences exist in mouse lung ILC2 phenotypes and functions; however, it is still unclear whether there are sex differences in heart ILC2s. RESULTS: Compared with age-matched wild-type (WT) male mice, 8-week-old but not 3-week-old WT female mice harbored an obviously greater percentage and number of heart ILC2s in homeostasis. However, the percentage of killer-cell lectin-like receptor G1 (Klrg1)- ILC2s was higher, but the Klrg1+ ILC2s were lower in female mice than in male mice in both heart tissues of 3- and 8-week-old mice. Eight-week-old Rag2-/- mice also showed sex differences similar to those of age-matched WT mice. Regarding surface marker expression, compared to age-matched male mice, WT female mice showed higher expression of CD90.2 and Ki67 and lower expression of Klrg1 and Sca-1 in heart total ILC2s. There was no sex difference in IL-4 and IL-5 secretion by male and female mouse heart ILC2s. Increased IL-33 mRNA levels within the heart tissues were also found in female mice compared with male mice. By reanalyzing published single-cell RNA sequencing data, we found 2 differentially expressed genes between female and male mouse heart ILC2s. Gene set variation analysis revealed that the glycine, serine and threonine metabolism pathway was upregulated in female heart ILC2s. Subcluster analysis revealed that one cluster of heart ILC2s with relatively lower expression of Semaphorin 4a and thioredoxin interacting protein but higher expression of hypoxia-inducible lipid droplet-associated. CONCLUSIONS: These results revealed greater numbers of ILC2s, higher expression of CD90.2, reduced Klrg1 and Sca-1 expression in the hearts of female mice than in male mice and no sex difference in IL-4 and IL-5 production in male and female mouse heart ILC2s. These sex differences in heart ILC2s might be due to the heterogeneity of IL-33 within the heart tissue.


Subject(s)
Heart , Immunity, Innate , Interleukin-33 , Lymphocytes , Sex Characteristics , Animals , Female , Male , Mice , Interleukin-33/genetics , Interleukin-33/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/metabolism , Lymphocytes/metabolism , Mice, Knockout , Thy-1 Antigens/metabolism
6.
Virol J ; 19(1): 68, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35413989

ABSTRACT

BACKGROUND: The onset and progression of cervical intraepithelial neoplasia (CIN) are closely associated with the persistent infection of high-risk HPV (especially type16), which is mainly caused by immune escape. Natural killer (NK) cells play an important role against virally infected cells and tumor cells through a fine balance of signals from multiple surface receptors. Overexpression of non-MHC-I specific inhibitory receptors TIGIT, KLRG1, Siglec-7, LAIR-1, and CD300a on NK cells correlates with cellular exhaustion and immune evasion, but these receptors have not been investigated in CIN. The aim of the present study was to examine the potential role of NK cell non-MHC-I specific inhibitory receptors expression in immune escape from HPV16(+)CIN patients. METHODS: The subset distribution, IFN-γ and TNF-α expression levels and immunophenotype of TIGIT, KLRG1, Siglec-7, LAIR-1, and CD300a of NK cells were investigated in peripheral blood mononuclear cell samples by flow cytometry from 82 women who were HPV16(+) with CIN grades 0, I, II-III or HPV(-) CIN 0. Immunohistochemistry was applied to detect the expression of ligands for NK receptors in the cervical tissues. HPV types were identified by PCR assays. RESULTS: The HPV16(+) subjects with high-grade lesions had an increased number of circulating peripheral blood CD56bright NK cells with reduced functionality and IFN-γ secretion. The expression levels of the inhibitory molecules TIGIT and KLRG1 on CD56bright NK cells increased in parallel with increasing CIN grade. In addition, TIGIT and KLRG1 related ligands, Poliovirus receptor (PVR), N-Cadherin and E-Cadherin expression level was also elevated with increasing CIN grade. CONCLUSIONS: Our results suggest that up-regulation of the inhibitory TIGIT, KLRG1 and their ligands may negatively regulate cervical CD56bright NK-mediated immunity to HPV16 and contribute to the progression of CIN. These results may facilitate the development of early-warning immune predictors and therapeutic strategies for HPV16(+) CIN based on the TIGIT and KLRG1 inhibitory pathways of NK cells.


Subject(s)
Papillomavirus Infections , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Female , Human papillomavirus 16 , Humans , Killer Cells, Natural , Lectins, C-Type/genetics , Leukocytes, Mononuclear/metabolism , Ligands , Papillomavirus Infections/pathology , Receptors, Immunologic/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins
7.
Immunol Invest ; 51(3): 670-687, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33401997

ABSTRACT

BACKGROUND: Killer cell lectin-like receptor G1 (KLRG1) and 2B4 play important roles in the immune regulation and immune tolerance to tumor cells by inhibiting T cell function. However, the clinical relevance of KLRG1 and 2B4 to cervical cancer remains to be understood. METHODS: We measured the frequency of KLRG1+ or 2B4+ cells in CD4+ or CD8 + T cells derived from peripheral blood or tumour biopsies in cervical cancer patients by flow cytometry. RESULTS: Compared with healthy controls, the level of KLRG1 and 2B4 on CD8 + T cells in the blood of the patients increased significantly (P = .0056 and .0441). KLRG1 level on CD8 + T cells and 2B4 level on CD4 + T cells in peripheral blood were significantly higher than in tumor tissues (P < .0001 and P = .0003). Higher KLRG1 level on blood-derived CD8 + T cells was observed in patients older than 54 years (P = .001) or tested to be HPV-negative (P = .026). Tumor-infiltrated CD8 + T cells demonstrated elevated KLRG1 level in patients having pelvic lymph node metastasis (P = .016). Increased 2B4 level on blood-derived CD8 + T cells was also observed in patients older than 54 years (P < .001). KLRG1 expression on both CD4 + T (P = .0158) and CD8 + T (P = .0187) cells in the peripheral blood increased after radiotherapy. CONCLUSION: KLRG1 level on T cells was related to age and HPV in patients with cervical cancer, while 2B4 level on T cells was related to age, underlying their roles in the host immune response to cervical cancer. Radiotherapy can improve the immune function of patients.


Subject(s)
Uterine Cervical Neoplasms , CD8-Positive T-Lymphocytes , Female , Humans , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family , T-Lymphocytes , Trans-Activators/metabolism , Uterine Cervical Neoplasms/metabolism
8.
BMC Cancer ; 21(1): 752, 2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34187403

ABSTRACT

BACKGROUND: As a marker of differentiation, Killer cell lectin like receptor G1 (KLRG1) plays an inhibitory role in human NK cells and T cells. However, its clinical role remains inexplicit. This work intended to investigate the predictive ability of KLRG1 on the efficacy of immune-checkpoint inhibitor in the treatment of lung adenocarcinoma (LUAD), as well as contribute to the possible molecular mechanisms of KLRG1 on LUAD development. METHODS: Using data from the Gene Expression Omnibus, the Cancer Genome Atlas and the Genotype-Tissue Expression, we compared the expression of KLRG1 and its related genes Bruton tyrosine kinase (BTK), C-C motif chemokine receptor 2 (CCR2), Scm polycomb group protein like 4 (SCML4) in LUAD and normal lung tissues. We also established stable LUAD cell lines with KLRG1 gene knockdown and investigated the effect of KLRG1 knockdown on tumor cell proliferation. We further studied the prognostic value of the four factors in terms of overall survival (OS) in LUAD. Using data from the Gene Expression Omnibus, we further investigated the expression of KLRG1 in the patients with different responses after immunotherapy. RESULTS: The expression of KLRG1, BTK, CCR2 and SCML4 was significantly downregulated in LUAD tissues compared to normal controls. Knockdown of KLRG1 promoted the proliferation of A549 and H1299 tumor cells. And low expression of these four factors was associated with unfavorable overall survival in patients with LUAD. Furthermore, low expression of KLRG1 also correlated with poor responses to immunotherapy in LUAD patients. CONCLUSION: Based on these findings, we inferred that KLRG1 had significant correlation with immunotherapy response. Meanwhile, KLRG1, BTK, CCR2 and SCML4 might serve as valuable prognostic biomarkers in LUAD.


Subject(s)
Adenocarcinoma of Lung/genetics , Gene Expression Profiling/methods , Immunotherapy/methods , Lectins, C-Type/metabolism , Lung Neoplasms/genetics , Receptors, Immunologic/metabolism , Adenocarcinoma of Lung/pathology , Cell Proliferation , Female , Humans , Lung Neoplasms/pathology , Male
9.
Fish Shellfish Immunol ; 113: 51-60, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33798718

ABSTRACT

Killer cell lectin-like receptor G subfamily 1 (KLRG1) is a receptor generally expressed on effector CD8+ T cells or NK cells at terminal differentiation stage, and it will be highly induced for lymphocyte cytotoxicity upon pathogen infection or lymphocyte activation. However, little is known about the character or function of KLRG1 in lower vertebrates. In present study, we reappraised a molecule that previously defined as KLRG1 in the genomic sequence of Nile tilapia Oreochromis niloticus, and identified it as an atypical KLRG1-like molecule (defined as On-KLRG1-L), and illustrated its potential function serving as a marker representing effector T lymphocytes of fish species. On-KLRG1-L consists of two C-type lectin-like domains (CTLDs) without transmembrane region, and the tertiary structure of the CTLD is highly alike to that in mouse KLRG1. As a CTLD-containing protein, the recombinant On-KLRG1-L could bind PGN and several microbes in vitro. On-KLRG1-L was widely expressed in immune-associated tissues, with the highest expression level in the gill. Once Nile tilapia is infected by Aeromonas hydrophila, mRNA level of On-KLRG1-L in spleen lymphocytes were significantly up-regulated on 5 days after infection. Meanwhile, On-KLRG1-L protein was also induced on 5 or 8 days after A. hydrophila infection. Furthermore, we found both mRNA and protein levels of On-KLRG1-L were dramatically enhanced within several hours after spleen lymphocytes were activated by T cell-specific mitogen PHA in vitro. More importantly, the ratio of On-KLRG1-L+ T cells was also augmented after PHA stimulation. The observations suggested that the KLRG1-like molecule from Nile tilapia participated in lymphocyte activation and anti-bacterial adaptive immune response, and could serve as an activation marker of T lymphocytes. Our study thus provided new evidences to understand lymphocyte-mediated adaptive immunity of teleost.


Subject(s)
Adaptive Immunity/genetics , Cichlids/immunology , Fish Diseases/immunology , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Biomarkers/metabolism , Fish Diseases/microbiology , Fish Proteins/genetics , Fish Proteins/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/veterinary , Lectins, C-Type/genetics , Protein Structure, Tertiary , Receptors, Immunologic/genetics , Sequence Alignment/veterinary
10.
Curr Rheumatol Rep ; 23(7): 56, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34212266

ABSTRACT

PURPOSE OF REVIEW: To review recent advances in immunopathology for idiopathic inflammatory myopathies, focusing on widely available immunohistochemical analyses. RECENT FINDINGS: Sarcoplasmic expression of myxovirus resistance protein A (MxA) is specifically observed in all types of dermatomyositis and informs that type I interferons are crucially involved in its pathogenesis. It is a more sensitive diagnostic marker than perifascicular atrophy. Diffuse tiny dots in the sarcoplasm highlighted by p62 immunostaining are characteristically seen in immune-mediated necrotizing myopathy. This feature is linked to a chaperone-assisted selective autophagy pathway. Myofiber invasion by highly differentiated T cells, a marker of which is KLRG1, is specific to inclusion body myositis and has a crucial role in its pathogenesis. The recent advances in immunopathology contribute to increased diagnostic accuracy and a better understanding of the underlying pathophysiology in different types of idiopathic inflammatory myopathies.


Subject(s)
Autoimmune Diseases , Dermatomyositis , Myositis, Inclusion Body , Myositis , Biomarkers , Humans
11.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807229

ABSTRACT

Cytomegalovirus (CMV) causes clinical issues primarily in immune-suppressed conditions. CMV-associated anterior uveitis (CMV-AU) is a notable new disease entity manifesting recurrent ocular inflammation in immunocompetent individuals. As patient demographics indicated contributions from genetic background and immunosenescence as possible underlying pathological mechanisms, we analyzed the immunogenetics of the cohort in conjunction with cell phenotypes to identify molecular signatures of CMV-AU. Among the immune cell types, natural killer (NK) cells are main responders against CMV. Therefore, we first characterized variants of polymorphic genes that encode differences in CMV-related human NK cell responses (Killer cell Immunoglobulin-like Receptors (KIR) and HLA class I) in 122 CMV-AU patients. The cases were then stratified according to their genetic features and NK cells were analyzed for human CMV-related markers (CD57, KLRG1, NKG2C) by flow cytometry. KIR3DL1 and HLA class I combinations encoding strong receptor-ligand interactions were present at substantially higher frequencies in CMV-AU. In these cases, NK cell profiling revealed expansion of the subset co-expressing CD57 and KLRG1, and together with KIR3DL1 and the CMV-recognizing NKG2C receptor. The findings imply that a mechanism of CMV-AU pathogenesis likely involves CMV-responding NK cells co-expressing CD57/KLRG1/NKG2C that develop on a genetic background of KIR3DL1/HLA-B allotypes encoding strong receptor-ligand interactions.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Uveitis, Anterior/metabolism , Adult , Aged , Aged, 80 and over , CD57 Antigens/genetics , CD57 Antigens/immunology , Cohort Studies , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Female , Genes, MHC Class I/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunocompromised Host/immunology , Immunocompromised Host/physiology , Killer Cells, Natural/physiology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, KIR/genetics , Transplantation, Homologous/adverse effects , Uveitis, Anterior/genetics , Uveitis, Anterior/virology
12.
Brain ; 142(9): 2590-2604, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31326977

ABSTRACT

Inclusion body myositis is a late onset treatment-refractory autoimmune disease of skeletal muscle associated with a blood autoantibody (anti-cN1A), an HLA autoimmune haplotype, and muscle pathology characterized by cytotoxic CD8+ T cell destruction of myofibres. Here, we report on translational studies of inclusion body myositis patient muscle compared with a diverse set of other muscle disease samples. Using available microarray data on 411 muscle samples from patients with inclusion body myositis (n = 40), other muscle diseases (n = 265), and without neuromuscular disease (normal, n = 106), we identified a signature of T-cell cytotoxicity in inclusion body myositis muscle coupled with a signature of highly differentiated CD8 T-cell effector memory and terminally differentiated effector cells. Further, we examined killer cell lectin-like receptor G1 (KLRG1) as a marker of this population of cells, demonstrated the correlation of KLRG1 gene expression with lymphocyte cytotoxicity across 28 870 human tissue samples, and identified the presence of KLRG1 on pathogenic inclusion body myositis muscle invading T cells and an increase in KLRG1 expressing T cells in inclusion body myositis blood. We examined inclusion body myositis muscle T-cell proliferation by Ki67 immunohistochemistry demonstrating that diseased muscle-invading T cells are minimally or non-proliferative, in accordance with known properties of highly differentiated or terminally differentiated T cells. We found low expression of KLRG1 on infection-protective human lymphoid tissue central memory T cells and autoimmune-protective human blood regulatory T cells. Targeting highly differentiated cytotoxic T cells could be a favourable approach to treatment of inclusion body myositis.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Lectins, C-Type/biosynthesis , Muscle, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , Receptors, Immunologic/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Humans , Lectins, C-Type/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myositis, Inclusion Body/immunology , Myositis, Inclusion Body/pathology , Receptors, Immunologic/immunology
13.
Eur J Immunol ; 47(8): 1354-1367, 2017 08.
Article in English | MEDLINE | ID: mdl-28612942

ABSTRACT

Designing CD8+ T-cell vaccines, which would provide protection against tumors is still considered a great challenge in immunotherapy. Here we show the robust potential of cytomegalovirus (CMV) vector expressing the NKG2D ligand RAE-1γ as CD8+ T cell-based vaccine against malignant tumors. Immunization with the CMV vector expressing RAE-1γ, delayed tumor growth or even provided complete protection against tumor challenge in both prophylactic and therapeutic settings. Moreover, a potent tumor control in mice vaccinated with this vector can be further enhanced by blocking the immune checkpoints TIGIT and PD-1. CMV vector expressing RAE-1γ potentiated expansion of KLRG1+ CD8+ T cells with enhanced effector properties. This vaccination was even more efficient in neonatal mice, resulting in the expansion and long-term maintenance of epitope-specific CD8+ T cells conferring robust resistance against tumor challenge. Our data show that immunomodulation of CD8+ T-cell responses promoted by herpesvirus expressing a ligand for NKG2D receptor can provide a powerful platform for the prevention and treatment of CD8+ T-cell sensitive tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cytomegalovirus/genetics , Membrane Proteins/genetics , Neoplasms/immunology , Animals , Animals, Newborn , Cytomegalovirus/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors , Humans , Immunomodulation , Immunotherapy/methods , Killer Cells, Natural/immunology , Lectins, C-Type , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Membrane Proteins/immunology , Mice , Neoplasms/prevention & control , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology
14.
Exerc Immunol Rev ; 24: 72-84, 2018.
Article in English | MEDLINE | ID: mdl-29461967

ABSTRACT

BACKGROUND/PURPOSE: Ageing has profound impact on the immune system, mainly on T-cells. However, it has been suggested that chronic exercise may delay immunosenescence. Master athletes represent an interesting sub-demographic group to test this theory since they maintain a high training frequency and load throughout life. The purpose of this study was to evaluate the effects of lifelong training on the senescence and mobilization of T lymphocytes in response to acute exercise. MATERIAL AND METHODS: Nineteen athletes who regularly participated in training and competitions for more than 20 years throughout their lives and a control group of 10 healthy individuals participated in this study. All subjects performed a progressive test to exhaustion on a cycle ergometer. Blood samples were obtained before (Pre), 10 min after the test (Post) and 1 h after the test (1h). Phenotypic study of peripheral blood T-cells was performed by flow cytometry. Genes of interest expression was done on T-cells purified by cell sorting. RESULTS: Master athletes had a lower percentage of senescent naïve, central memory and effector memory CD8+ T-cells and senescent naïve and effector memory CD4+ T-cells. Age had a positive effect on SLEC CD8+ T-cells and a negative effect on naïve CD8+ T-cells. VO2max positively correlated with the proportion of naïve CD4+ T-cells and negatively correlated with the percentage of total lymphocytes. No differences were founded for CD4+ and CD8+ T-cells and their subsets between master athletes and the control group at all times of measurement. No differences were observed in the CD45RA expressing effector memory cells (EMRA) for the various study conditions. The mRNA expression of the CCR7 gene for naïve CD8+ T-cells and the Fas-L gene for effector-terminal CD8+ T-cells was not different between masters and controls and did not change in response to the maximal protocol test. CONCLUSION: In conclusion, maintaining high levels of aerobic fitness during the natural course of aging may help prevent the accumulation of senescent T-cells.


Subject(s)
Athletes , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Exercise/physiology , Cell Separation , Female , Flow Cytometry , Humans , Immunologic Memory , Immunosenescence , Male , Middle Aged , Oxygen Consumption , T-Lymphocyte Subsets/cytology
15.
Immunol Rev ; 259(1): 103-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24712462

ABSTRACT

CD4(+) Forkhead box protein 3 (Foxp3)(+) regulatory T cells (Tregs) are the major cell type that mediates dominant tolerance in the periphery. Over the past decade, extensive study of Tregs has revealed that these cells express substantial heterogeneity to maintain tolerance and regulate immune responses. Tregs possess heterogeneity with respect to their origin and processes for development, functional activity, migratory pattern, and activation status. Some of the same environmental cues and molecular pathways utilized to generate specialized T-effector cells are also integrated by Tregs to colocalize and fine-tune suppressive mechanisms to optimally regulate and restrain distinctive self and antigen-specific T-cell responses. Here, we review our current understanding and significance of Treg heterogeneity in maintaining peripheral immune tolerance. We also highlight recent work from our laboratory that has studied the extent phenotypically distinct Treg subsets are related to each other and expand in an ordered fashion to give rise to highly activated short-lived Klrg1(+) suppressor cells to optimize immune regulation and maintain homeostasis of the Treg compartment.


Subject(s)
Self Tolerance/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cell Movement/immunology , Gene Expression , Humans , Immunologic Memory , Phenotype , Receptors, Immunologic/genetics , Self Tolerance/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
16.
Biochim Biophys Acta ; 1859(10): 1306-13, 2016 10.
Article in English | MEDLINE | ID: mdl-27424220

ABSTRACT

Genetic variations mapping to 3' untranslated regions (3'UTRs) may overlap with microRNA (miRNA) binding sites, therefore potentially interfering with translation inhibition or messenger RNA (mRNA) degradation. The aim of this study was to investigate whether single nucleotide polymorphisms (SNPs) located within the 3'UTRs of six candidate genes and predicted to interfere with miRNA ligation could account for disease-relevant differential mRNA levels. Focusing on pemphigus foliaceus (PF) - an autoimmune blistering skin condition with unique endemic patterns - we investigated whether nine 3'UTR SNPs from the CD1D, CTLA4, KLRD1, KLRG1, NKG7, and TNFSF13B genes differentially expressed in PF were disease-associated. The heterozygous genotype of the KLRG1 rs1805672 polymorphism was associated with increased predisposition to PF (A/G vs. A/A: P=0.038; OR=1.60), and a trend for augmented susceptibility was observed for carriers of the G allele (P=0.094; OR=1.44). In silico analyses suggested that rs1805672 G allele could disrupt binding of miR-584-5p, and indicated rs1805672 as an expression Quantitative Trait Locus (eQTL), with an effect on KLRG1 gene expression. Dual-luciferase assay showed that miR-584-5p mediated approximately 50% downregulation of the reporter gene's activity through the 3'UTR of KLRG1 harboring rs1805672 A allele (vs. miRNA-negative condition, P=0.006). This silencing relationship was lost after site-directed mutation to G allele (vs. miRNA-negative condition, P=0.391; vs. rs1805672 A allele, P=0.005). Collectively, these results suggest that a disease-associated SNP located within the 3'UTR of KLRG1 directly interferes with miR-584-5p binding, allowing for KLRG1 mRNA differential accumulation, which in turn may contribute to pathogenesis of autoimmune diseases, such as pemphigus.


Subject(s)
3' Untranslated Regions , Genetic Predisposition to Disease , Lectins, C-Type/genetics , MicroRNAs/genetics , Pemphigus/genetics , Polymorphism, Single Nucleotide , Trans-Activators/genetics , Alleles , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Base Sequence , Binding Sites , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Case-Control Studies , DNA Mutational Analysis , Gene Expression Regulation , Gene Frequency , Haplotypes , Humans , Lectins, C-Type/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/metabolism , Mutation , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Pemphigus/diagnosis , Pemphigus/metabolism , Pemphigus/pathology , Receptors, Immunologic , Trans-Activators/metabolism
17.
Immunology ; 152(1): 65-73, 2017 09.
Article in English | MEDLINE | ID: mdl-28437578

ABSTRACT

Immune homeostasis requires the tight, tissue-specific control of the different CD4+ Foxp3+ regulatory T (Treg) cell populations. The cadherin-binding inhibitory receptor killer cell lectin-like receptor G1 (KLRG1) is expressed by a subpopulation of Treg cells with GATA3+ effector phenotype. Although such Treg cells are important for the immune balance, especially in the gut, the role of KLRG1 in Treg cells has not been assessed. Using KLRG1 knockout mice, we found that KLRG1 deficiency does not affect Treg cell frequencies in spleen, mesenteric lymph nodes or intestine, or frequencies of GATA3+ Treg cells in the gut. KLRG1-deficient Treg cells were also protective in a T-cell transfer model of colitis. Hence, KLRG1 is not essential for the development or activity of the general Treg cell population. We then checked the effects of KLRG1 on Treg cell activation. In line with KLRG1's reported inhibitory activity, in vitro KLRG1 cross-linking dampened the Treg cell T-cell receptor response. Consistently, lack of KLRG1 on Treg cells conferred on them a competitive advantage in the gut, but not in lymphoid organs. Hence, although absence of KLRG1 is not enough to increase intestinal Treg cells in KLRG1 knockout mice, KLRG1 ligation reduces T-cell receptor signals and the competitive fitness of individual Treg cells in the intestine.


Subject(s)
Intestinal Mucosa/immunology , Lymphocyte Activation , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cells, Cultured , Colitis/immunology , Colitis/prevention & control , Disease Models, Animal , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Genotype , Immunity, Mucosal , Intestinal Mucosa/metabolism , Lectins, C-Type , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Time Factors
18.
J Autoimmun ; 78: 19-28, 2017 03.
Article in English | MEDLINE | ID: mdl-28129932

ABSTRACT

CXC Chemokine Receptor 3 (CXCR3) is functionally pleiotropic and not only plays an important role in chemotaxis, but also participates in T cell differentiation and may play a critical role in inducing and maintaining immune tolerance. These observations are particularly critical for autoimmune cholangitis in which CXCR3 positive T cells are found around the portal areas of both humans and mouse models of primary biliary cholangitis (PBC). Herein, we investigated the role of CXCR3 in the pathogenesis of autoimmune cholangitis. We have taken advantage of a unique CXCR3 knockout dnTGFßRII mouse to focus on the role of CXCR3, both by direct observation of its influence on the natural course of disease, as well as through adoptive transfer studies into Rag-/- mice. We report herein that not only do CXCR3 deficient mice develop an exacerbation of autoimmune cholangitis associated with an expanded effector memory T cell number, but also selective adoptive transfer of CXCR3 deficient CD8+ T cells induces autoimmune cholangitis. In addition, gene microarray analysis of CXCR3 deficient CD8+ T cells reveal an intense pro-inflammatory profile. Our data suggests that the altered gene profiles induced by CXCR3 deficiency promotes autoimmune cholangitis through pathogenic CD8+ T cells. These data have significance for human PBC and other autoimmune liver diseases in which therapeutic intervention might be directed to chemokines and/or their receptors.


Subject(s)
Autoimmunity/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, CXCR3/deficiency , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Immunologic Memory , Ligands , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Receptors, CXCR3/metabolism
19.
Cancer Immunol Immunother ; 65(4): 441-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850637

ABSTRACT

The CD57 antigen (alternatively HNK-1, LEU-7, or L2) is routinely used to identify terminally differentiated 'senescent' cells with reduced proliferative capacity and altered functional properties. In this article, we review current understanding of the attributes of CD57-expressing T-cells and NK cells in both health and disease and discuss how this marker can inform researchers about their likely functions in human blood and tissues in vivo. While CD57 expression on human lymphocytes indicates an inability to proliferate, these cells also display high cytotoxic potential, and CD57(pos) NK cells exhibit both memory-like features and potent effector functions. Accordingly, frequencies of CD57-expressing cells in blood and tissues have been correlated with clinical prognosis in chronic infections or various cancers and with human aging. Functional modulation of senescent CD57(pos) T-cells and mature CD57(pos) NK cells may therefore represent innovative strategies for protection against human immunological aging and/or various chronic diseases.


Subject(s)
CD57 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Arthritis, Rheumatoid , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Humans , Killer Cells, Natural/metabolism , Models, Immunological , Neoplasms/immunology , Neoplasms/metabolism
20.
Eur J Immunol ; 44(6): 1600-3, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24752778

ABSTRACT

The inhibitory C-type lectin-like immunoreceptor KLRG1 enables mature NK cells and differentiated T cells to sense cadherin-expressing cells by ligating "classical" cadherins. Upon engagement of the KLRG1 ectodomain, an inhibitory signal emanates from the cytoplasmic immunoreceptor-tyrosine-based inhibition motif (ITIM), dampening functional responses of these lymphocytes. Malignancy-associated loss of cadherins has been proposed to relieve KLRG1-mediated inhibition of cytotoxic lymphocytes and thereby to contribute to tumor surveillance by an alternate mode of "missing self-recognition". In this issue of the European Journal of Immunology, Schweier et al. [Eur. J. Immunol. 2014. 44: 1851-1856] propose another intriguing mechanism that may relieve KLRG1-mediated inhibition in the course of lymphocyte activation. Subsequent to identification of the transferrin receptor (TfR) as a component of a high molecular mass KLRG1 complex, they demonstrate that a fraction of mouse KLRG1 molecules undergoes disulfide-bonding with TfRs and colocalises with the latter at the cell surface. In functional terms, high levels of TfRs such as those found on activated lymphocytes were found to be associated with decreased KLRG1 inhibitory function, indicating that TfRs may sequester KLRG1 from interacting with cadherins. Hence, this unexpected liaison between KLRG1 and TfR may represent a regulatory link between metabolic activation and responses of lymphocytes.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , Receptors, Transferrin/immunology , Trans-Activators/immunology , Transferrin/immunology , Amino Acid Motifs , Animals , Cadherins/immunology , Humans , Mice , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL