Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Nat Commun ; 15(1): 4227, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762592

ABSTRACT

Multisystem inflammatory syndrome in children is a post-infectious presentation SARS-CoV-2 associated with expansion of the T cell receptor Vß21.3+ T-cell subgroup. Here we apply muti-single cell omics to compare the inflammatory process in children with acute respiratory COVID-19 and those presenting with non SARS-CoV-2 infections in children. Here we show that in Multi-Inflammatory Syndrome in Children (MIS-C), the natural killer cell and monocyte population demonstrate heightened CD95 (Fas) and Interleuking 18 receptor expression. Additionally, TCR Vß21.3+ CD4+ T-cells exhibit skewed differentiation towards T helper 1, 17 and regulatory T cells, with increased expression of the co-stimulation receptors ICOS, CD28 and interleukin 18 receptor. We observe no functional evidence for NLRP3 inflammasome pathway overactivation, though MIS-C monocytes show elevated active caspase 8. This, coupled with raised IL18 mRNA expression in CD16- NK cells on single cell RNA sequencing analysis, suggests interleukin 18 and CD95 signalling may trigger activation of TCR Vß21.3+ T-cells in MIS-C, driven by increased IL-18 production from activated monocytes and CD16- Natural Killer cells.


Subject(s)
COVID-19 , Interleukin-18 , Killer Cells, Natural , Monocytes , Signal Transduction , Systemic Inflammatory Response Syndrome , fas Receptor , Humans , Interleukin-18/metabolism , Child , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , fas Receptor/metabolism , fas Receptor/genetics , Monocytes/immunology , Monocytes/metabolism , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/metabolism , COVID-19/immunology , COVID-19/virology , COVID-19/metabolism , COVID-19/complications , Inflammasomes/metabolism , Inflammasomes/immunology , SARS-CoV-2/immunology , Adolescent , Male , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Female , Child, Preschool , Single-Cell Analysis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD28 Antigens/metabolism , Lymphocyte Activation/immunology , Receptors, Interleukin-18/metabolism , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology
2.
Clin Investig Arterioscler ; 36(4): 210-217, 2024.
Article in English, Spanish | MEDLINE | ID: mdl-38216380

ABSTRACT

BACKGROUND: Atherosclerosis is an inflammatory disease. Interleukin 18 (IL-18) is an inflammatory molecule that has been linked to the development of atherosclerosis and cardiovascular disease. OBJECTIVE: To evaluate the possible relationship between plasma levels of IL-18 and the presence of atherosclerosis evaluated at the carotid level, as well as to analyze the possible modulation by different polymorphisms in a Mediterranean population. MATERIAL AND METHODS: Seven hundred and forty-six individuals from the metropolitan area of Valencia were included, recruited over a period of 2 years. Hydrocarbon and lipid metabolism parameters were determined using standard methodology and IL-18 using ELISA. In addition, carotid ultrasound was performed and the genotype of four SNPs related to the IL-18 signaling pathway was analyzed. RESULTS: Patients with higher plasma levels of IL-18 had other associated cardiovascular risk factors. Elevated IL-18 levels were significantly associated with higher carotid IMT and the presence of atheromatous plaques. The genotype with the A allele of the SNP rs2287037 was associated with a higher prevalence of carotid atheromatous plaque. On the contrary, the genotype with the C allele of the SNP rs2293224 was associated with a lower prevalence of atheromatous plaque. CONCLUSIONS: High levels of IL-18 were significantly associated with a higher carotid IMT and the presence of atheromatous plaques, which appear to be influenced by genetic factors, as evidenced by associations between SNPs in the IL-18 receptor gene and the presence of atheroma plaque.


Subject(s)
Carotid Artery Diseases , Carotid Intima-Media Thickness , Genotype , Interleukin-18 , Plaque, Atherosclerotic , Polymorphism, Single Nucleotide , Aged , Female , Humans , Male , Middle Aged , Alleles , Atherosclerosis/genetics , Carotid Artery Diseases/genetics , Enzyme-Linked Immunosorbent Assay , Heart Disease Risk Factors , Interleukin-18/genetics , Plaque, Atherosclerotic/genetics , Receptors, Interleukin-18/genetics , Risk Factors , Spain
3.
J Biol Chem ; 296: 100630, 2021.
Article in English | MEDLINE | ID: mdl-33823154

ABSTRACT

Unchecked inflammation can result in severe diseases with high mortality, such as macrophage activation syndrome (MAS). MAS and associated cytokine storms have been observed in COVID-19 patients exhibiting systemic hyperinflammation. Interleukin-18 (IL-18), a proinflammatory cytokine belonging to the IL-1 family, is elevated in both MAS and COVID-19 patients, and its level is known to correlate with the severity of COVID-19 symptoms. IL-18 binds its specific receptor IL-1 receptor 5 (IL-1R5, also known as IL-18 receptor alpha chain), leading to the recruitment of the coreceptor, IL-1 receptor 7 (IL-1R7, also known as IL-18 receptor beta chain). This heterotrimeric complex then initiates downstream signaling, resulting in systemic and local inflammation. Here, we developed a novel humanized monoclonal anti-IL-1R7 antibody to specifically block the activity of IL-18 and its inflammatory signaling. We characterized the function of this antibody in human cell lines, in freshly obtained peripheral blood mononuclear cells (PBMCs) and in human whole blood cultures. We found that the anti-IL-1R7 antibody significantly suppressed IL-18-mediated NFκB activation, reduced IL-18-stimulated IFNγ and IL-6 production in human cell lines, and reduced IL-18-induced IFNγ, IL-6, and TNFα production in PBMCs. Moreover, the anti-IL-1R7 antibody significantly inhibited LPS- and Candida albicans-induced IFNγ production in PBMCs, as well as LPS-induced IFNγ production in whole blood cultures. Our data suggest that blocking IL-1R7 could represent a potential therapeutic strategy to specifically modulate IL-18 signaling and may warrant further investigation into its clinical potential for treating IL-18-mediated diseases, including MAS and COVID-19.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Immunologic Factors/pharmacology , Interleukin-18/genetics , Receptors, Interleukin-18/genetics , Anti-Inflammatory Agents/metabolism , Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/biosynthesis , Candida albicans/growth & development , Candida albicans/pathogenicity , Gene Expression Regulation , HEK293 Cells , Humans , Immunologic Factors/biosynthesis , Inflammation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-18/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophage Activation Syndrome/drug therapy , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , Receptors, Interleukin-18/antagonists & inhibitors , Receptors, Interleukin-18/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , COVID-19 Drug Treatment
4.
Int J Mol Sci ; 21(22)2020 Nov 14.
Article in English | MEDLINE | ID: mdl-33202693

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is rising in prevalence, and a better pathophysiologic understanding of the transition to its inflammatory phenotype (NASH) is key to the development of effective therapies. To evaluate the contribution of the NLRP3 inflammasome and its downstream effectors IL-1 and IL-18 in this process, we applied the true-to-life "American lifestyle-induced obesity syndrome" (ALiOS) diet mouse model. Development of obesity, fatty liver and liver damage was investigated in mice fed for 24 weeks according to the ALiOS protocol. Lipidomic changes in mouse livers were compared to human NAFLD samples. Receptor knockout mice for IL-1 and IL-18 were used to dissect the impact of downstream signals of inflammasome activity on the development of NAFLD. The ALiOS diet induced obesity and liver steatosis. The lipidomic changes closely mimicked changes in human NAFLD. A pro-inflammatory gene expression pattern in liver tissue and increased serum liver transaminases indicated early liver damage in the absence of histological evidence of NASH. Mechanistically, Il-18r-/-- but not Il-1r-/- mice were protected from early liver damage, possibly due to silencing of the pro-inflammatory gene expression pattern. Our study identified NLRP3 activation and IL-18R-dependent signaling as potential modulators of early liver damage in NAFLD, preceding development of histologic NASH.


Subject(s)
Interleukin-18/metabolism , Interleukin-1/metabolism , Liver/injuries , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Animals , Interleukin-1/genetics , Interleukin-18/genetics , Liver/pathology , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism
5.
Sci Rep ; 9(1): 9386, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253830

ABSTRACT

Support from human genetics increases the probability of success in drug development. However, few examples exist of successful genomically-driven drug repositioning. Given that a Mendelian form of severe enterocolitis is due to up-regulation of the interleukin-18 (IL18) signaling pathway, and pharmacologic inhibition of IL18 has been shown to reverse this enterocolitis, we undertook a Mendelian randomization study to test the causal effect of elevated IL18 levels on inflammatory bowel disease susceptibility (IBD) in 12,882 cases and 21,770 controls. Mendelian randomization is an established method to assess the role of biomarkers in disease etiology in a manner that minimizes confounding and prevents reverse causation. Using three SNPs that explained almost 7% of the variance in IL18 level, we found that each genetically predicted standard deviation increase in IL18 was associated with an increase in IBD susceptibility (odds ratio = 1.22, 95% CI = 1.11-1.34, P-value = 6 × 10-5). This association was further validated in 25,042 IBD cases and 34,915 controls (odds ratio = 1.13, 95% CI = 1.05-1.20). Recently, an anti-IL18 monoclonal antibody, which decreased free IL18 levels, was found to be safe, yet ineffective in a phase II trial for type 2 diabetes. Taken together, these genomic findings implicated IBD as an alternative indication for anti-IL18 therapy, which should be tested in randomized controlled trials.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Drug Repositioning , Inflammatory Bowel Diseases/drug therapy , Interleukin-18/therapeutic use , Alleles , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Biomarkers , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Interleukin-18/blood , Mendelian Randomization Analysis , Odds Ratio , Polymorphism, Single Nucleotide , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism , Severity of Illness Index , Treatment Outcome
6.
Br J Pharmacol ; 176(12): 2015-2027, 2019 06.
Article in English | MEDLINE | ID: mdl-29774543

ABSTRACT

BACKGROUND AND PURPOSE: Monocytes play a critical role in hypertension. The purpose of our study was to use an unbiased approach to determine whether hypertensive individuals on conventional therapy exhibit an altered monocyte gene expression profile and to perform validation studies of selected genes to identify novel therapeutic targets for hypertension. EXPERIMENTAL APPROACH: Next generation RNA sequencing identified differentially expressed genes in a small discovery cohort of normotensive and hypertensive individuals. Several of these genes were further investigated for association with hypertension in multiple validation cohorts using qRT-PCR, regression analysis, phenome-wide association study and case-control analysis of a missense polymorphism. KEY RESULTS: We identified 60 genes that were significantly differentially expressed in hypertensive monocytes, many of which are related to IL-1ß. Uni- and multivariate regression analyses of the expression of these genes with mean arterial pressure (MAP) revealed four genes that significantly correlated with MAP in normotensive and/or hypertensive individuals. Of these, lactoferrin (LTF), peptidoglycan recognition protein 1 and IL-18 receptor accessory protein (IL18RAP) remained significantly elevated in peripheral monocytes of hypertensive individuals in a separate validation cohort. Interestingly, IL18RAP expression associated with MAP in a cohort of African Americans. Furthermore, homozygosity for a missense single nucleotide polymorphism in LTF that decreases antimicrobial function and increases protein levels (rs1126478) was over-represented in patients with hypertension relative to controls (odds ratio 1.16). CONCLUSIONS AND IMPLICATIONS: These data demonstrate that monocytes exhibit enhanced pro-inflammatory gene expression in hypertensive individuals and identify IL18RAP and LTF as potential novel mediators of human hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.


Subject(s)
Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Lactoferrin/pharmacology , Monocytes/metabolism , Receptors, Interleukin-18/genetics , Black or African American , Blood Pressure/drug effects , Blood Pressure/immunology , Case-Control Studies , Gene Expression Profiling , Humans , Hypertension/immunology , Multivariate Analysis , Receptors, Interleukin-18/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
7.
Mucosal Immunol ; 11(6): 1705-1715, 2018 11.
Article in English | MEDLINE | ID: mdl-30087443

ABSTRACT

This report deals with the possible mechanism by which IL-18 can contribute to the control and resolution of inflammatory lesions in the cornea caused by herpes simplex virus infection. Our results demonstrate that the expression of the IL-18R by both regulatory T cells (Treg) and effector T cells was a pivotal event that influenced lesion pathogenesis. The engagement of IL-18R on Treg with its cytokine ligand resulted in Amphiregulin expression a molecule associated with tissue repair. In support of this scheme of events, lesion severity became more severe in animals unable to express the IL-18R because of gene knockout and was reduced in severity when IL-18 was overexpressed in the cornea. These changes in lesion severity correlated with the frequency and number of both Treg and Teff that expressed Amphiregulin. Additional experiments indicated that IL-12 and IL-18 acted synergistically to enhance Amphiregulin expression in Treg, an event partly dependent on P38 MAPK activity. Finally, sub-conjunctival administration of Amphiregulin resulted in resolution of both developing and developed lesions. Thus, overall our results imply that IL-18 may participate in controlling the severity of SK and contribute to tissue repair by converting both Treg and effector T cells into those that produce Amphiregulin.


Subject(s)
Amphiregulin/metabolism , Cornea/immunology , Herpes Simplex/immunology , Keratitis, Herpetic/immunology , Receptors, Interleukin-18/physiology , Simplexvirus/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Amphiregulin/genetics , Animals , Female , Interleukin-12/metabolism , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-18/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Metab ; 12: 89-97, 2018 06.
Article in English | MEDLINE | ID: mdl-29699928

ABSTRACT

OBJECTIVE: Interleukin (IL)-18 plays a crucial role in maintaining metabolic homeostasis and levels of this cytokine are influenced by gender, age, and sex hormones. The role of gender on IL-18 signaling, however, is unclear. We hypothesized that the presence of female sex hormone could preserve the metabolic phenotype of the IL-18R-/- animals. METHODS: We studied female mice with a global deletion of the α isoform of the IL-18 receptor (IL-18R-/-) and littermates control. Three studies were done: 1) animals fed a high fat diet (HFD) for 16 weeks; 2) animals fed chow diet for 72 weeks and 3) animals (3 weeks-old) randomized to either bilateral ovariectomy (OVX) or control surgery (SHAM) and followed for 16 weeks. RESULTS: Female IL-18R-/- mice gained less weight and maintained glucose homeostasis on a chow diet compared with HFD, but no differences between genotypes were observed. The maintenance of body weight and glucose homeostasis in IL-18R-/- mice was lost with aging. By 72 weeks of age, IL-18R-/- mice became heavier compared with WT mice due to an increase in both visceral and subcutaneous adiposity and displayed glucose intolerance. OVX did not affect body weight in IL-18R-/- mice but exacerbated glucose intolerance and impaired liver insulin signaling when compared with SHAM mice. CONCLUSIONS: Female mice harboring a global deletion of the IL-18R, only present the same phenotype as reported in male IL-18R-/- mice if they are aged or have undergone OVX, in which circulating estrogen is likely to be blunted. The role of estrogen signaling in the protection against altered metabolic homeostasis in IL-18R-/- mice appears to be mediated by liver insulin signaling. We therefore suggest that the metabolic effects mediated by loss of IL-18 signaling are only present in a female sex hormone free environment.


Subject(s)
Estrogens/metabolism , Interleukin-18/metabolism , Obesity/metabolism , Signal Transduction , Adiposity , Animals , Diet, High-Fat/adverse effects , Estrogens/deficiency , Female , Glucose/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/genetics , Ovariectomy/adverse effects , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism
9.
PLoS Pathog ; 13(12): e1006782, 2017 12.
Article in English | MEDLINE | ID: mdl-29281733

ABSTRACT

The efficacies of many new T cell vaccines rely on generating large populations of long-lived pathogen-specific effector memory CD8 T cells. However, it is now increasingly recognized that prior infection history impacts on the host immune response. Additionally, the order in which these infections are acquired could have a major effect. Exploiting the ability to generate large sustained effector memory (i.e. inflationary) T cell populations from murine cytomegalovirus (MCMV) and human Adenovirus-subtype (AdHu5) 5-beta-galactosidase (Ad-lacZ) vector, the impact of new infections on pre-existing memory and the capacity of the host's memory compartment to accommodate multiple inflationary populations from unrelated pathogens was investigated in a murine model. Simultaneous and sequential infections, first with MCMV followed by Ad-lacZ, generated inflationary populations towards both viruses with similar kinetics and magnitude to mono-infected groups. However, in Ad-lacZ immune mice, subsequent acute MCMV infection led to a rapid decline of the pre-existing Ad-LacZ-specific inflating population, associated with bystander activation of Fas-dependent apoptotic pathways. However, responses were maintained long-term and boosting with Ad-lacZ led to rapid re-expansion of the inflating population. These data indicate firstly that multiple specificities of inflating memory cells can be acquired at different times and stably co-exist. Some acute infections may also deplete pre-existing memory populations, thus revealing the importance of the order of infection acquisition. Importantly, immunization with an AdHu5 vector did not alter the size of the pre-existing memory. These phenomena are relevant to the development of adenoviral vectors as novel vaccination strategies for diverse infections and cancers. (241 words).


Subject(s)
Adenoviruses, Human/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Muromegalovirus/immunology , Viral Vaccines/immunology , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/genetics , Adenoviruses, Human/pathogenicity , Animals , Coinfection/immunology , Coinfection/prevention & control , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Host-Pathogen Interactions/immunology , Humans , Lac Operon , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Muromegalovirus/genetics , Muromegalovirus/pathogenicity , Receptors, Interleukin-18/deficiency , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology , Viral Vaccines/genetics
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(4): 526-530, 2017 Apr.
Article in Chinese | MEDLINE | ID: mdl-28395725

ABSTRACT

Objective To investigate the changes of interleukin-18 (IL-18), interleukin 18 binding protein (IL-18BP) and interleukin 18 receptor (IL-18R) expressions in eosinophil-enriched cells from asthmatic patients, and study the correlations among them. Methods Peripheral venous blood from asthmatic patients and normal subjects were collected and stimulated with the extracts of Artemisia pollen, dust mite, and Platanus pollen. The expressions of IL-18, IL-18BP and IL-18R in eosinophil-enriched blood cells were detected by flow cytometry, and the correlations among them were analyzed. Results The proportion of IL-18+ cells in eosinophil-enriched cells from asthmatic patients increased 15-fold compared with normal subjects. The proportion of IL-18+ cells increased about 1.3-fold and 1.5-fold in eosinophil-enriched cells from asthmatic patients whose blood was stimulated with dust mite and Platanus pollen extracts, respectively. The average fluorescence intensity (MFI) of IL-18BP+ cells increased about 1.5-fold and the proportion of IL-18R+ cells increased about 2-fold in eosinophil-enriched cells from asthmatic patients' blood stimulated with Platanus pollen extracts. In addition, the expressions of IL-18BP+ cells and IL-18R+ cells showed a positive correlation (r=0.639) in eosinophil-enriched cells from asthmatic patients stimulated with allergens. However, the expressions of IL-18, IL-18BP and IL-18R in eosinophil-enriched cells did not obviously change in normal subjects. Conclusion IL-18, IL-18R and IL-18BP expressed in eosinophils may be involved in the inflammatory reaction of asthma.


Subject(s)
Asthma/immunology , Eosinophils/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-18/blood , Receptors, Interleukin-18/immunology , Adolescent , Adult , Aged , Asthma/blood , Asthma/genetics , Case-Control Studies , Female , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-18/immunology , Male , Middle Aged , Receptors, Interleukin-18/blood , Receptors, Interleukin-18/genetics , Young Adult
11.
J Endocrinol Invest ; 40(4): 447-454, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27914037

ABSTRACT

PURPOSE: There is no information available about the IL-18 receptor in ovarian follicles, so the present study attempts to demonstrate the expression of IL-18 and its receptor in human granulosa cells (GCs). METHODS: To evaluate the concentration of IL-18 in serum and follicular fluid (FF), we collected serum and FF from 102 women undergoing oocyte retrieval. Also, to detect expression of IL-18 and its receptor by luteinized GCs, these cells were pooled six times from a total of twenty individual patients with 5-16 follicles each. The IL-18 concentration was determined by ELISA and the expression of IL-18 and its receptor by immunocytochemistry and reverse transcription polymerase chain reaction. RESULTS: Our results showed that the median IL-18 concentration in serum, 159.27 pg/ml (IQR 121.41-210.1), was significantly higher than in FF, 142.1 pg/ml (IQR 95.7-176.5), p < 0.001. Moreover, we found that IL-18 and its receptor are expressed by GCs. CONCLUSION: The presence of IL-18 in FF and the expression of IL-18 and its receptor by GCs suggest an important role for this cytokine in ovarian function.


Subject(s)
Follicular Fluid/metabolism , Granulosa Cells/metabolism , Interleukin-18/metabolism , Ovarian Follicle/metabolism , RNA, Messenger/genetics , Receptors, Interleukin-18/metabolism , Adult , Cells, Cultured , Female , Fertilization in Vitro , Follicular Fluid/cytology , Granulosa Cells/cytology , Humans , Interleukin-18/genetics , Ovarian Follicle/cytology , Receptors, Interleukin-18/genetics , Young Adult
12.
J Immunol ; 198(1): 270-278, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27881706

ABSTRACT

The IL-1 families of ligands and receptors exhibit similarity of coding sequences, protein structures, and chromosomal positions, suggesting that they have arisen via duplication of ancestral genes. Within these families there is selectivity in ligand-receptor interactions as well as promiscuity. IL-18 and its receptor are members of these families. IL-18 is recognized as binding to the protein products of the IL18R1 and IL18RAP genes, and with high affinity to a separate IL-18 binding protein (IL-18BP). However, IL-18BP is anomalous, as it exhibits little resemblance to IL-18R proteins. Additionally, IL-18 is produced in the brain in medial habenula neurons, which project IL-18-containing axons to the interpeduncular nucleus. However, there is a lack of focal IL-18R expression in their terminal field. Given these anomalies, we hypothesized that another receptor for IL-18 may exist, and that IL18BP is evolutionarily related to this receptor. We examined Ensembl and National Center for Biotechnology Information databases to identify available IL18BP records (n = 86 species) and show through bioinformatics approaches that across mammalian species with IL18BP genes, IL-18BP is consistently most similar to IL-1R9 (IL-1R accessory protein-like 2), another member of the IL-1R family. IL-1R9 and the related IL-1R8, but not other IL-1R family members, exhibit an amino acid sequence similar to binding site A of human and viral IL-18BPs. Conserved intron/exon boundaries, protein structure, and key binding site amino acids suggest that IL18BP and IL1R9 are evolutionarily related, and that IL-1R9 and IL-1R8 may bind IL-18.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Receptors, Interleukin-18/genetics , Receptors, Interleukin-1/genetics , Amino Acid Sequence , Animals , Base Sequence , Computational Biology , Evolution, Molecular , Humans , Sequence Homology
13.
Proc Natl Acad Sci U S A ; 113(36): 10139-44, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27551096

ABSTRACT

Natural killer (NK) cells are known to be activated by Th1-type cytokines, such as IL-2, -12, or -18, and they secrete a large amount of IFN-γ that accelerates Th1-type responses. However, the roles of NK cells in Th2-type responses have remained unclear. Because IL-4 acts as an initiator of Th2-type responses, we examined the characteristics of NK cells in mice overexpressing IL-4. In this study, we report that IL-4 overexpression induces distinctive characteristics of NK cells (B220(high)/CD11b(low)/IL-18Rα(low)), which are different from mature conventional NK (cNK) cells (B220(low)/CD11b(high)/IL-18Rα(high)). IL-4 overexpression induces proliferation of tissue-resident macrophages, which contributes to NK cell proliferation via production of IL-15. These IL-4-induced NK cells (IL4-NK cells) produce higher levels of IFN-γ, IL-10, and GM-CSF, and exhibit high cytotoxicity compared with cNK cells. Furthermore, incubation of cNK cells with IL-15 and IL-4 alters their phenotype to that similar to IL4-NK cells. Finally, parasitic infection, which typically causes strong Th2-type responses, induces the development of NK cells with characteristics similar to IL4-NK cells. These IL4-NK-like cells do not develop in IL-4Rα KO mice by parasitic infection. Collectively, these results suggest a novel role of IL-4 in immune responses through the induction of the unique NK cells.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-15/immunology , Interleukin-4/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Strongylida Infections/immunology , Animals , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Proliferation , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-15/genetics , Interleukin-15/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/parasitology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/immunology , Nippostrongylus/pathogenicity , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/immunology , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology
14.
PLoS One ; 11(8): e0160321, 2016.
Article in English | MEDLINE | ID: mdl-27483370

ABSTRACT

In recent years, cytokine-mediated therapy has emerged as further advance alternative in cancer therapy. Interleukin-18 (IL-18) has exhibited interesting anti-cancer properties especially when combined with IL-12. We engineered IL-18 in order to improve its activity using single point mutagenesis. IL-18 mutants were constructed according to binding residues and polarity which we tried to increase polarity in M33Q and M60Q, enhanced cationicity in E6K, and flexibility in T63A. All IL-18 proteins were expressed in Pichia pastoris, purified, and then measured the activity by treating with the NK-92MI cell line to evaluate interferon-γ (IFN-γ) stimulation. The E6K and T63A mutant forms showed higher activity with respect to native proteins at the concentration of 200 ng mL-1 by inducing the expression of IFN-γ, about factors of 9 and 4, respectively. Meanwhile, M33Q and M60Q had no significant activity to induce IFN-γ. Interestingly, the combination of E6K and T63A mutations could synergize the induction activity of IL-18 to be 16 times at 200 ng mL-1. Furthermore, molecular dynamics studies have elucidated the effect due to mutation on conformation of the binding site of IL-18. The results turn out that E6K provides structural perseverance against mutation, while M33Q and M60Q promote vivid overall change in protein conformation, especially at the binding site. For T63A, mutation yields small difference in structure but clearly increases structural flexibility. However, a small structural change was observed when T63A was combined with E6K. Our research resulted in a novel version of IL-18 which could be a new key candidate for cytokine-mediated therapy.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-18/chemistry , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Receptors, Interleukin-18/chemistry , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cloning, Molecular , Gene Expression , Humans , Interferon-gamma/metabolism , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-18/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Kinetics , Models, Molecular , Molecular Weight , Pichia/genetics , Pichia/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Engineering , Protein Interaction Domains and Motifs , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Structure-Activity Relationship , Substrate Specificity
15.
Cancer Res ; 76(8): 2394-405, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26893476

ABSTRACT

Chronic inflammation in liver tissue is an underlying cause of hepatocellular carcinoma. High levels of inflammatory cytokine IL18 in the circulation of patients with hepatocellular carcinoma correlates with poor prognosis. However, conflicting results have been reported for IL18 in hepatocellular carcinoma development and progression. In this study, we used tissue specimens from hepatocellular carcinoma patients and clinically relevant mouse models of hepatocellular carcinoma to evaluate IL18 expression and function. In a mouse model of liver fibrosis that recapitulates a tumor-promoting microenvironment, global deletion of the IL18 receptor IL18R1 enhanced tumor growth and burden. Similarly, in a carcinogen-induced model of liver tumorigenesis, IL18R1 deletion increased tumor burden. Mechanistically, we found that IL18 exerted inflammation-dependent tumor-suppressive effects largely by promoting the differentiation, activity, and survival of tumor-infiltrating T cells. Finally, differences in the expression of IL18 in tumor tissue versus nontumor tissue were more predictive of patient outcome than overall tissue expression. Taken together, our findings resolve a long-standing contradiction regarding a tumor-suppressive role for IL18 in established hepatocellular carcinoma and provide a mechanistic explanation for the complex relationship between its expression pattern and hepatocellular carcinoma prognosis. Cancer Res; 76(8); 2394-405. ©2016 AACR.


Subject(s)
Carcinoma, Hepatocellular/pathology , Interleukin-18/metabolism , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Signal Transduction , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Mice , Mice, Knockout , Prognosis , Receptors, Interleukin-18/genetics
16.
Mol Cell Neurosci ; 71: 114-24, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26763728

ABSTRACT

Currently, the low efficacy of antinociceptive drugs for the treatment of neuropathic pain is a major therapeutic problem. Here, we show the potential role of interleukin (IL)-18 signaling in this phenomenon. IL-18 is an important molecule that performs various crucial functions, including the alteration of nociceptive transmission in response to neuropathic pain. We have studied the changes in the mRNA and protein levels (qRT-PCR and Western blot analysis, respectively) of IL-18, IL-18-binding protein (IL-18BP) and the IL-18 receptor (IL-18R) over time in rats following chronic constriction injury (CCI) of the sciatic nerve. Our study demonstrated that the spinal levels of IL-18BP were slightly downregulated at days 7 and 14 in the rats subjected to CCI. In contrast, the IL-18 and IL-18R mRNA expression and protein levels were elevated in the ipsilateral spinal cord on days 2, 7 and 14. Moreover, in rats exposed to a single intrathecal administration of IL-18BP (50 and 100 ng) 7 or 14 days following CCI, symptoms of neuropathic pain were attenuated, and the analgesia pursuant to morphine and buprenorphine (0.5 and 2.5 µg) was enhanced. In summary, the restoration of the analgesic activity of morphine and buprenorphine via the blockade of IL-18 signaling suggests that increased IL-18 pathway may account for the decreased analgesic efficacy of opioids for neuropathic pain.


Subject(s)
Analgesics, Opioid/pharmacology , Buprenorphine/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-18/metabolism , Morphine/pharmacology , Neuralgia/metabolism , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Buprenorphine/administration & dosage , Buprenorphine/therapeutic use , Cells, Cultured , Drug Synergism , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/genetics , Male , Morphine/administration & dosage , Morphine/therapeutic use , Neuralgia/drug therapy , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism
17.
Biol Pharm Bull ; 39(1): 138-42, 2016.
Article in English | MEDLINE | ID: mdl-26725437

ABSTRACT

Previous studies showed an increased prevalence of cataracts in postmenopausal women. In this study, we investigated changes in the levels of calcium ion (Ca(2+)) and interleukin (IL)-18, which are factors in cataract development, in the lenses of ovariectomized (OVX) rats, a model of postmenopausal woman. Although the Ca(2+) content in the blood of OVX rats increased 1 month after ovariectomy and subsequently decreased, the Ca(2+) content in the lenses was unchanged in OVX rats 1-3 months after ovariectomy. The Ca(2+)-ATPase activity in the lenses of OVX rats peaked 1 month after ovariectomy, and the behavior of Ca(2+)-ATPase activity in lenses of OVX rats was similar to that of the Ca(2+) concentration in the blood. It is possible that hypercalcemia increases the Ca(2+) inflow into the lens; however, the enhanced Ca(2+)-ATPase activity prevents the Ca(2+) level from rising. On the other hand, we found that the levels of both IL-18 and interferon (IFN)-γ in the lenses of OVX rats were significantly increased as compared with the lenses of sham (control) rats during the period 1-3 months after surgery. These results suggest that the expression of IFN-γ via IL-18 in the lenses of OVX rats is induced by ovariectomy, and that excessive IL-18 and IFN-γ production in the lenses may be related to cataract development in postmenopausal women. These findings support those of previous studies that assessed lens opacification in postmenopausal women.


Subject(s)
Gene Expression Regulation/physiology , Interleukin-18/metabolism , Lens, Crystalline/metabolism , Ovariectomy , Animals , Bone Density , Calcium/blood , Calcium/metabolism , Female , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-18/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism
18.
Sci Rep ; 5: 18533, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26688048

ABSTRACT

Pattern recognition receptors (PRRs) and cytokine receptors are key players in the initiation of immune responses to infection. PRRs detecting viral RNA, such as toll like receptor (TLR)-3, -7/8, and RIG-I like receptors (RLRs; RIG-I and MDA-5), as well as cytokine receptors such as interleukin 1 receptor (IL-1R), have been implicated in responses to RNA viruses that infect the airways. The latter includes respiratory syncytial virus (RSV), a human pathogen that can cause severe lower respiratory tract infections, especially in infants. To evaluate the collective contribution of PRRs and IL-1R signalling to RSV immunity, we generated Myd88/Trif/Mavs(-/-) mice that are deficient in signalling by all TLRs, RLRs and IL-1R, as well as other cytokine receptors such as IL-18 receptor. Early production of pro-inflammatory mediators and lung infiltration by immune cells were completely abrogated in infected Myd88/Trif/Mavs(-/-) mice. However, RSV-specific CD8(+) T cells were elicited and recruited into the lungs and airways. Consistent with these findings, Myd88/Trif/Mavs(-/-) mice survived RSV infection but displayed higher viral load and weight loss. These data highlight an unappreciated level of redundancy in pathways that couple innate virus sensing to adaptive immunity, providing the host with remarkable resilience to infection.


Subject(s)
Infections/genetics , Receptors, Interleukin-1 Type II/genetics , Receptors, Interleukin-18/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Tract Infections/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Humans , Infections/immunology , Infections/virology , Mice , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Receptors, Interleukin-18/immunology , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Load
19.
Cytokine ; 74(2): 327-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26009021

ABSTRACT

We evaluated the role of IL-18 during Leishmania amazonensis infection in C57BL/6 mice, using IL-18KO mice. We showed that IL-18 is involved in susceptibility to L. amazonensis, since IL-18KO mice presented reduced lesions and parasite loads. Because macrophages are the host cells of the parasite, we investigated if macrophages were involved in IL-18-mediated susceptibility to L. amazonensis. We showed that macrophages obtained from WT or IL-18KO responded similarly to L. amazonensis infection. Moreover, we showed that C57BL/6 macrophages do not respond to IL-18, since they do not express IL-18R. Therefore, macrophages are not involved in IL-18-mediated susceptibility to L. amazonensis.


Subject(s)
Interleukin-18/immunology , Leishmania/immunology , Leishmaniasis/immunology , Animals , Disease Susceptibility , Interleukin-18/genetics , Leishmaniasis/genetics , Leishmaniasis/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology
20.
Immunol Cell Biol ; 93(10): 849-57, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25801352

ABSTRACT

Elevated levels of interleukin (IL)-18 have been reported in a number of allergic diseases. We recently reported that IL-18 in the blood and IL-18Rα mRNA in the oesophagus are induced during human eosinophilic oesophagitis (EoE). Additionally, we earlier showed that invariant natural killer T (iNKT) cells are critical to EoE pathogenesis; however, the mechanism of iNKT cell activation in EoE is not well understood. Therefore, the current study focused on the hypothesis that allergen-induced IL-18 may have an important role in iNKT cell-mediated EoE pathogenesis. We first validated the human EoE findings of IL-18 in experimental EoE by examining blood levels of IL-18 and oesophageal IL-18Rα mRNA levels in aeroallergen- and food allergen-induced experimental mouse models of EoE. We demonstrate that blood IL-18 protein and oesophageal IL-18Rα mRNA are induced in the mouse model of EoE and that IL-18Rα is expressed by iNKT cells in the oesophagus. Intranasal delivery of rIL-18 induced both mast cells and eosinophilic inflammation in the oesophagus in a time- and dose-dependent manner. To establish the significance of IL-18 in EoE pathogenesis, we examined DOX-inducible rtTA-CC10-IL-18 bitransgenic mice that induce IL-18 protein expression in the oesophagus. Our analysis indicated that induction of IL-18 in these mice resulted in the development of many of the characteristics of EoE, including oesophageal intraepithelial eosinophilia, increased mast cells, oesophageal remodelling and fibrosis. The current study provides evidence that IL-18 may induce iNKT cell activation to release the eosinophil-activating cytokine IL-5, as IL-5-deficient mice and iNKT cell-deficient (CD1d null) mice do not induce EoE in response to intranasal IL-18 challenge. Taken together, these findings provide evidence that allergen-induced IL-18 has a significant role in promoting IL-5- and iNKT-dependent EoE pathogenesis.


Subject(s)
Allergens/immunology , Eosinophilic Esophagitis/immunology , Eosinophils/immunology , Hypersensitivity/immunology , Interleukin-18/metabolism , Mast Cells/immunology , Natural Killer T-Cells/immunology , Animals , Disease Models, Animal , Fibrosis , Humans , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-5/genetics , Interleukin-5/metabolism , Mice , Mice, Transgenic , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...