Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Comput Math Methods Med ; 2021: 7596343, 2021.
Article in English | MEDLINE | ID: mdl-34899969

ABSTRACT

BACKGROUND: N-Acetylcysteine (NAC) had exerted antioxidation and anti-inflammation effects on chronic obstructive pulmonary disease (COPD) patients. However, its effect in regulating interleukin- (IL-) 18 was not fully understood. This study was designed to evaluate the specific mechanism of NAC regulating IL-18. MATERIALS AND METHODS: A total of 112 COPD patients and 103 health individuals were recruited in the study. Cytokine level in patients' serum was measured by enzyme-linked immunosorbent assay (ELISA). A COPD mouse model was established by administration of lipopolysaccharide (LPS) and cigarette smoke. The expression of cytokines was measured by ELISA and flow cytometry. Inflammasome-related protein was measured by Western blot. RESULT: NAC could effectively improve the immune status of COPD patients as well as the COPD mouse model by downregulating proinflammation and inflammation cytokines including IL-1ß, interferon- (IFN-) γ, tumor necrosis factor- (TNF-) α, and IL-18. It also had the capability to suppress synthesis of IL-18 in macrophage to inhibit the secretion of IFN-γ from natural killer (NK) cells through influencing the inflammasome-related protein in macrophages. CONCLUSION: NAC could effectively inhibit the production of IL-18 by suppressing NLRP3 expression in macrophages to reduce the production of IFN-γ in NK cells.


Subject(s)
Acetylcysteine/pharmacology , Interferon-gamma/biosynthesis , Interleukin-18/biosynthesis , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Animals , Case-Control Studies , Computational Biology , Cytokines/blood , Disease Models, Animal , Female , Humans , Interleukin-18/antagonists & inhibitors , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pulmonary Disease, Chronic Obstructive/pathology
2.
Life Sci ; 285: 119974, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34560082

ABSTRACT

AIMS: Chemical renal toxicity is common and has limited therapeutic interventions. The NLRP3 inhibitor dapansutrile (DAPA) undergoes clinical phase II trials and it shows promising beneficial effects in various inflammatory diseases. The current study aims at evaluating the effect of DAPA on folic acid (FA) induced acute kidney injury (AKI) and its possible transition to chronic injury. MATERIALS AND METHODS: Two treatment protocols were studied depending on DAPA injection timing. A prophylactic protocol involving the injection of DAPA (0.2 mg/kg) daily for seven days before FA challenge and a therapeutic protocol where DAPA was injected after FA. Each protocol included four groups of rats: control group, DAPA group, FA group and DAPA+FA group. Serum creatinine, urea and uric acid were measured. Also, kidney injury, necrosis and fibrosis percentage in addition to infiltration of CD68 positive cells were evaluated. Activation markers of inflammasome and the expression of Ki-67 and LC-3 were measured. KEY FINDINGS: Results showed an improvement in renal tissue integrity and a significant decrease in kidney function biomarkers, caspase-1, IL-1ß and IL-18 by DAPA injection (p < 0.05). In addition, DAPA decreased the proliferation marker Ki-67 and the autophagic marker LC-3 (p < 0.01). SIGNIFICANCE: DAPA potentially alleviates FA induced nephrotoxicity through targeting inflammasome/caspase-1/IL axis. Moreover, it shows a regulatory effect on renal regeneration and autophagy.


Subject(s)
Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Caspase 1/metabolism , Folic Acid/toxicity , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Nitriles/therapeutic use , Acute Kidney Injury/chemically induced , Animals , Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Biomarkers , Cell Proliferation , Interleukin-18/antagonists & inhibitors , Interleukin-18/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Ki-67 Antigen/metabolism , Kidney/drug effects , Male , Nitriles/pharmacology , Rats , Rats, Sprague-Dawley
3.
Dis Markers ; 2021: 5586312, 2021.
Article in English | MEDLINE | ID: mdl-34367377

ABSTRACT

BACKGROUND: Obliterative bronchiolitis (OB) was a main cause of deterioration of long-term prognosis in lung transplant recipients after the first posttransplant year. Proinflammatory cytokine interleukin-18 (IL-18) strengthened both the natural immunity and acquired immunity and played an important role in organ transplantation. The roles of IL-18 in the occurrence, development, and drug treatment of OB remained unclear. METHODS: Small interfering RNA (siRNA) against mouse IL-18 (siRNA-IL-18) was used to silence IL-18 expression. Mouse heterotopic tracheal transplantation model was used to simulate OB. Recipient mice were divided into 5 groups (n = 12) according to donor mouse strains and drug treatment: isograft group, allograft group, allograft+tacrolimus group, allograft+azithromycin group, and allograft+tacrolimus+azithromycin group. The luminal obliteration rates were pathological evaluation. Expressions of cytokines and MMPs were detected by real-time PCR, western blot, and enzyme chain immunosorbent assay (ELISA). RESULTS: The luminal obliteration rates of IL-18 of the siRNA-IL-18 group were significantly lower than those of the negative control group (p < 0.0001) and the blank control group (p = 0.0002). mRNA expressions of IFN-γ, EMMPRIN, MMP-8, and MMP-9 of the siRNA-IL-18 group were significantly lower than those of the negative and blank control groups. No tracheal occlusion occurred in grafts of the isograft group. The rates of tracheal occlusion of the allograft group, allograft+tacrolimus group, allograft+azithromycin group, and allograft+tacrolimus+azithromycin group were 72.17 ± 4.66%, 40.33 ± 3.00%, 38.50 ± 2.08%, and 23.33 ± 3.24%, respectively. There were significant differences between the 4 groups (p < 0.001). Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-γ (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. CONCLUSIONS: IL-18 could be a novel molecular involved in the occurrence, development, and drug treatment of OB.


Subject(s)
Azithromycin/administration & dosage , Bronchiolitis Obliterans/drug therapy , Interleukin-18/genetics , Lung Transplantation/adverse effects , RNA, Small Interfering/administration & dosage , Tacrolimus/administration & dosage , Animals , Azithromycin/pharmacology , Basigin/genetics , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/genetics , Disease Models, Animal , Humans , Interferon-gamma/genetics , Interleukin-18/antagonists & inhibitors , Male , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 9/genetics , Mice , RNA, Small Interfering/pharmacology , Tacrolimus/pharmacology , Transplantation, Homologous/adverse effects , Treatment Outcome
4.
J Immunol Res ; 2021: 8998358, 2021.
Article in English | MEDLINE | ID: mdl-34239943

ABSTRACT

Increasing evidence indicates a pivotal role of macrophages in innate immunity, which contributes to the pathogenesis of adult-onset Still's disease (AOSD). Despite the available reviews that summarized the pathogenic role of proinflammatory cytokines in AOSD, a systematic approach focusing on the crucial role of macrophages in this disease is still lacking. This review summarizes the updated functions of macrophages in AOSD and their implication in clinical manifestations and therapeutics. We searched the MEDLINE database using the PubMed interface and reviewed the English-language literature as of 31 March 2021, from 1971 to 2021. We focus on the existing evidence on the pathogenic role of macrophages in AOSD and its implication in clinical characteristics and novel therapeutics. AOSD is an autoinflammatory disease mainly driven by the innate immune response. Among the innate immune responses, macrophage activation is a hallmark of AOSD pathogenesis. The pattern recognition receptors (PRRs) on macrophages recognize pathogen-associated molecular patterns and damage-associated molecular patterns and subsequently cause overproduction of proinflammatory cytokines and recruit adaptive immunity. Some biomarkers, such as ferritin and gasdermin D, reflecting macrophage activation were elevated and correlated with AOSD activity. Given that macrophage activation with the overproduction of proinflammatory cytokines plays a pathogenic role in AOSD, these inflammatory mediators would be the therapeutic targets. Accordingly, the inhibitors to interleukin- (IL-) 1, IL-6, and IL-18 have been shown to be effective in AOSD treatment. Gaining insights into the pathogenic role of macrophages in AOSD can aid in identifying disease biomarkers and therapeutic agents for this disease.


Subject(s)
Immunosuppressive Agents/pharmacology , Macrophage Activation/immunology , Macrophages/immunology , Still's Disease, Adult-Onset/immunology , Humans , Immunity, Innate/drug effects , Immunosuppressive Agents/therapeutic use , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Interleukin-18/antagonists & inhibitors , Interleukin-18/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Still's Disease, Adult-Onset/blood , Still's Disease, Adult-Onset/drug therapy
5.
J Surg Res ; 257: 468-476, 2021 01.
Article in English | MEDLINE | ID: mdl-32896815

ABSTRACT

BACKGROUND: Donation after circulatory death donors (DCD) can expand the donor pool for heart transplantation, which primarily depends on brain death donors. Ischemia and reperfusion injury are inherent to the DCD process. We hypothesize that pharmacologic inhibition of interleukin-1 (IL-1) and/or IL-18 is protective to DCD hearts. MATERIALS AND METHODS: Following clinical protocol, in-situ ischemia time in control beating-heart donor (CBD) and DCD groups was less than 5 and 40 min, respectively. Wild type (WT) C57Bl6/j, IL-1 receptor type I knockout (IL-1RI-KO), and IL-18 KO mice were used. Hearts were reanimated for 90 min on a Langendorff system with Krebs-Henseleit buffer at 37°C, to assess physiologic parameters. Recombinant IL-1 receptor antagonist (IL-1Ra) and/or IL-18 binding protein (IL-18BP) were added to the Krebs-Henseleit buffer to inhibit IL-1 and/or the IL-18 signaling, respectively. RESULTS: Developed pressure and ± dP/dt were significantly impaired in the DCD-WT group compared to CBD-WT (P ≤ 0.05). Troponin release was higher in DCD-WT groups. Functional parameters were preserved, and troponin release was significantly less in the DCD knockout groups. Heart function was improved in DCD groups treated with IL-1Ra or IL-18BP compared to the DCD-WT group. CONCLUSIONS: Heart function was significantly impaired in the DCD-WT group compared to CBD-WT. Genetic deletion or pharmacologic blockade of IL-1 or IL-18 was protective to DCD hearts.


Subject(s)
Intercellular Signaling Peptides and Proteins/therapeutic use , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Myocardial Reperfusion Injury/prevention & control , Tissue and Organ Procurement , Animals , Death , Drug Evaluation, Preclinical , Heart/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-18/antagonists & inhibitors , Interleukin-18/genetics , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Male , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Random Allocation
6.
J Cell Physiol ; 236(3): 1638-1657, 2021 03.
Article in English | MEDLINE | ID: mdl-32794180

ABSTRACT

Interleukin (IL)-18 is a pro-inflammatory cytokine belonging to the IL-1 family, first identified for its interferon-γ-inducing properties. IL-18 regulates both T helper (Th) 1 and Th2 responses. It acts synergistically with IL-12 in the Th1 paradigm, whereas with IL-2 and without IL-12 it can induce Th2 cytokine production from cluster of differentation (CD)4+ T cells, natural killer (NK cells, NKT cells, as well as from Th1 cells. IL-18 also plays a role in the hemophagocytic lymphohistiocytosis, a life-threatening condition characterized by a cytokine storm that can be secondary to infections. IL-18-mediated inflammation was largely studied in animal models of bacterial, viral, parasitic, and fungal infections. These studies highlight the contribution of either IL-18 overproduction by the host or overresponsiveness of the host to IL-18 causing an exaggerated inflammatory burden and leading to tissue injury. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19). The damage in the later phase of the disease appears to be driven by a cytokine storm, including interleukin IL-1 family members and secondary cytokines like IL-6. IL-18 may participate in this hyperinflammation, as it was previously found to be able to cause injury in the lung tissue of infected animals. IL-18 blockade has become an appealing therapeutic target and has been tested in some IL-18-mediated rheumatic diseases and infantile-onset macrophage activation syndrome. Given its role in regulating the immune response to infections, IL-18 blockade might represent a therapeutic option for COVID-19, although further studies are warranted to investigate more in detail the exact role of IL-18 in SARS-CoV-2 infection.


Subject(s)
COVID-19/immunology , Interleukin-18/immunology , Animals , Humans , Inflammation/immunology , Inflammation/virology , Interleukin-18/antagonists & inhibitors , SARS-CoV-2/immunology
7.
Ann Intern Med ; 173(7): 509-515, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32744862

ABSTRACT

BACKGROUND: Osteoarthritis is a common inflammatory disorder with no disease-modifying therapies. Whether inhibition of interleukin-1ß (IL-1ß) can reduce the consequences of large joint osteoarthritis is unclear. OBJECTIVE: To determine whether IL-1ß inhibition with canakinumab reduces incident total hip or knee replacement (THR/TKR). DESIGN: Exploratory analysis of a randomized trial. (ClinicalTrials.gov: NCT01327846). SETTING: 1091 clinical sites in 39 countries. PARTICIPANTS: 10 061 CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcomes Study) participants. INTERVENTION: Random allocation to placebo or canakinumab (50, 150, or 300 mg) subcutaneously once every 3 months. MEASUREMENTS: The primary and secondary outcomes were time to first incident THR/TKR and time to first occurrence of an osteoarthritis-related adverse event (AE). Data were obtained through blinded ascertainment of trial clinical and safety databases. RESULTS: Median follow-up was 3.7 years. For the individual canakinumab dose groups, compared with placebo, hazard ratios (HRs) for incident THR/TKR during follow-up were 0.60 (95% CI, 0.38 to 0.95) for the 50-mg group, 0.53 (CI, 0.33 to 0.84) for the 150-mg group, and 0.60 (CI, 0.38 to 0.93) for the 300-mg group. Thus, in the pooled canakinumab groups, compared with the placebo group, incidence rates for THR/TKR were 0.31 and 0.54 events per 100 person-years (HR, 0.58 [CI, 0.42 to 0.80]; P = 0.001), respectively. The HR for the secondary end point of osteoarthritis-related AEs was 0.73 (CI, 0.61 to 0.87). Similar findings were observed in analyses restricted to participants with a history of osteoarthritis. LIMITATION: Because the parent trial was not designed to examine the efficacy of IL-1ß inhibitors in osteoarthritis, information on structural joint outcomes was not collected. CONCLUSION: Findings from this exploratory analysis of a randomized controlled trial support further investigation of IL-1ß inhibition for treatment of large joint osteoarthritis. PRIMARY FUNDING SOURCE: Novartis Pharmaceuticals.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Arthroplasty, Replacement, Hip/statistics & numerical data , Arthroplasty, Replacement, Knee/statistics & numerical data , Interleukin-18/antagonists & inhibitors , Osteoarthritis, Hip/drug therapy , Osteoarthritis, Knee/drug therapy , Double-Blind Method , Female , Humans , Incidence , Male , Middle Aged
8.
Nat Immunol ; 21(6): 626-635, 2020 06.
Article in English | MEDLINE | ID: mdl-32424362

ABSTRACT

The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Enterocolitis/etiology , Enterocolitis/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptors, Cell Surface/metabolism , Animals , Citrobacter rodentium , Deubiquitinating Enzyme CYLD/genetics , Disease Models, Animal , Disease Susceptibility , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Enterocolitis/pathology , Gene Expression , Humans , Interleukin-18/antagonists & inhibitors , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Protein Binding/immunology , Ubiquitination
9.
Eur Rev Med Pharmacol Sci ; 24(7): 3915-3922, 2020 04.
Article in English | MEDLINE | ID: mdl-32329867

ABSTRACT

OBJECTIVE: To explore the protective effect of remifentanil against myocardial ischemia-reperfusion injury (MIRI) in rats and its mechanism. MATERIALS AND METHODS: The rat models of IRI were established and randomly divided into 1) sham-operation group (S group), 2) IRI rat model group (M group), 3) low-dose remifentanil group (R-L group), 4) moderate-dose remifentanil group (R-M group), and 5) high-dose remifentanil group (R-H group). The rats in R-L group, R-M group, and R-H group were administrated with remifentanil at 0.4 µg/kg/min, 2 µg/kg/min, and 10 µg/kg/min, respectively. The activity of creatine kinase-MB (CK-MB), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) in myocardial cells was detected using the automatic biochemical analyzer, and the apoptosis rate of myocardial cells was detected via terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Moreover, the messenger ribonucleic acid (mRNA) and protein levels of related cytokines in myocardial cells were determined through quantitative Polymerase Chain Reaction (qPCR) and Western blotting, and the content of interleukin-1ß (IL-1Symbol ) and IL-18 in peripheral blood was detected via enzyme-linked immunosorbent assay (ELISA). RESULTS: Remifentanil at different concentrations could protect myocardium from IRI, and remifentanil at 2 µg/kg/min and 10 µg/kg/min could significantly down-regulate the myocardial enzyme indexes in IRI myocardial cells (p<0.01). Besides, remifentanil reduced the mRNA expressions of IL-18, INF-γ, TNF-ß, and IL-1ß (p<0.01), significantly decreased the protein expression of IL-18, and raised the protein expression of IL-18BP, thereby improving myocardial pathological damage. CONCLUSIONS: The protective mechanism of remifentanil on the myocardium of MIRI rats may be related to the inhibition on the IL-18 signaling pathway.


Subject(s)
Interleukin-18/antagonists & inhibitors , Myocardial Reperfusion Injury/drug therapy , Remifentanil/pharmacology , Signal Transduction/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Interleukin-18/metabolism , Male , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Sprague-Dawley , Remifentanil/administration & dosage
10.
Arch Biochem Biophys ; 672: 108052, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31351069

ABSTRACT

Vascular disease is one of the most significant threats to the lives of patients suffering from diabetes, and chronic exposure of vascular endothelial cells to high glucose has been shown to significantly contribute to the process of endothelial cell dysfunction, one of the earliest events in diabetes-associated vascular disease. Nucleotide oligomerization domain (NOD)-like receptor pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in initiating the inflammatory process by facilitating the production of interleukin-1ß (IL-1ß) and IL-18. ASC and caspase 1 are also implicated in NLRP3 inflammasome-mediated chronic inflammation. While under normal conditions, a balance exists between oxidants and antioxidants, exposure to high glucose significantly increases the production of ROS, which is enhanced by NOX4 expression. In the present study, we explored the role of orexin A, an endogenous peptide produced in the hypothalamus, in high glucose-induced activation of the NLRP3 inflammasome, oxidative stress, and expression of several key cytokines. Our findings demonstrate that orexin A exerts potent antioxidant effects in human aortic endothelial cells exposed to high glucose by inhibiting mitochondrial ROS and expression of NOX4 at both the mRNA and protein levels as revealed by MitoSOX staining, real-time PCR, and Western blot analysis. We also show that orexin A inhibits high glucose-induced expression of TxNIP, which is crucial to the activation of the NLRP3 inflammasome, as well as that of HMGB1. We confirmed via real-time PCR and Western blot analysis that orexin A suppressed the production of the inflammatory cytokines IL-1ß and IL-18. Additionally, through SIRT1 knockdown siRNA experimentation, we confirmed that SIRT1 knockdown abolishes the effects of orexin A described above, thereby indicating a critical role of SIRT in the capacity of orexin A to ameliorate high glucose-induced oxidative stress and activation of NLRP3 inflammasome.


Subject(s)
Antioxidants/pharmacology , Glucose/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Orexins/pharmacology , Carrier Proteins/antagonists & inhibitors , Endothelial Cells , Gene Knockdown Techniques , HMGB1 Protein/antagonists & inhibitors , Humans , Inflammasomes/drug effects , Interleukin-18/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , NADPH Oxidase 4/antagonists & inhibitors , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
11.
Circ Res ; 124(3): 437-450, 2019 02.
Article in English | MEDLINE | ID: mdl-30702995

ABSTRACT

The recognition that atherosclerosis is a complex chronic inflammatory disorder mediated through both adaptive and innate immunity has led to the hypothesis that anticytokine therapies targeting specific IL (interleukin) signaling pathways could serve as powerful adjuncts to lipid lowering in the prevention and treatment of cardiovascular disease. Cytokines involved in human atherosclerosis can be broadly classified as proinflammatory and proatherogenic (such as IL-1, IL-6, and TNF [tumor necrosis factor]) or as anti-inflammatory and antiatherogenic (such as IL-10 and IL-1rA). The recent CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study) has shown that specific targeting of IL-1ß can significantly reduce cardiovascular event rates without lipid or blood pressure lowering. In CANTOS, the magnitude of benefit of this cytokine-targeted approach to atherosclerosis treatment was associated to the magnitude of reduction of the central signaling cytokine IL-6 and the downstream clinical biomarker high-sensitivity CRP (C-reactive protein). By contrast, in the recent CIRT (Cardiovascular Inflammation Reduction Trial), low-dose methotrexate neither reduced IL-1ß, IL-6, or high-sensitivity CRP nor lowered cardiovascular event rates. Taken together, these 2 contemporary trials provide proof of principle that focused cytokine inhibition, not broad-spectrum anti-inflammatory therapy, is likely to be crucial for atheroprotection. This review provides an overview of cytokines in atherosclerosis, the potential benefits and risks associated with targeted anticytokine therapies, and a look to the future of clinical practices addressing residual inflammatory risk.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Atherosclerosis/drug therapy , C-Reactive Protein/antagonists & inhibitors , Cardiovascular Diseases/drug therapy , Interleukin-1beta/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Atherosclerosis/complications , CD40 Antigens/antagonists & inhibitors , CD40 Ligand/antagonists & inhibitors , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Hematopoiesis , Humans , Inflammasomes/antagonists & inhibitors , Interleukin-18/antagonists & inhibitors , Methotrexate/therapeutic use , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Randomized Controlled Trials as Topic , Renal Insufficiency, Chronic/complications , Signal Transduction/drug effects , Stroke/prevention & control , Thrombosis/etiology , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors
12.
Int J Mol Sci ; 20(3)2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30717382

ABSTRACT

Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4⁺ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.


Subject(s)
Disease Susceptibility , Interleukin-18/genetics , Interleukin-18/metabolism , Animals , Gene Expression Regulation , Humans , Immune System/immunology , Immune System/metabolism , Interleukin-18/antagonists & inhibitors , Interleukin-33/genetics , Interleukin-33/metabolism , Molecular Targeted Therapy , Protein Binding , Receptors, Interleukin-18/metabolism , Signal Transduction
13.
Cytokine ; 118: 8-18, 2019 06.
Article in English | MEDLINE | ID: mdl-29396054

ABSTRACT

Chronic inflammation is recognized as a key tumor-promoting factor in a number of epithelial cancers, including gastric cancer (GC). The production of pro-inflammatory cytokines in the tumor microenvironment by both the innate and the adaptive immune response can activate signaling pathways that are associated with increased cell survival and proliferation of cancer cells. Among the cytokines that have most commonly been linked to inflammation-associated cancers, are the Th17 cell-associated cytokines IL-17A, IL-23, IL-22, and the IL-1 family members IL-1ß and IL-18. However, whether their contribution to inflammation-associated cancers is universal, or specific to individual types of cancers, remains to be elucidated. This review will explore our current understanding of the known roles of these cytokines in gastritis and discuss how their therapeutic inhibition may be useful for GC.


Subject(s)
Antineoplastic Agents/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Animals , Humans , Inflammation/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
J Cell Physiol ; 234(5): 5674-5682, 2019 05.
Article in English | MEDLINE | ID: mdl-30417374

ABSTRACT

The inflammatory response has an important role in the pathophysiology of diabetic nephropathy that is contributed to by inflammatory mediators such as interleukin-1 (IL-1), IL-6, IL-18, tumor necrosis factor-α, and macrophage chemotactic protein-1; however, the role of IL-18 seems to be more specific than other cytokines in the inflammatory process. IL-18 is expressed in renal tissue and is upregulated by several stimuli including hyperglycemia. The expression/urinary level of IL-18 is positively correlated with the progression of diabetic nephropathy and the urinary albumin excretion rate. In this review, we have focused on the molecular pathways modulating the relationship between IL-18 and diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Kidney/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Blood Glucose/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/immunology , Disease Progression , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/immunology , Kidney/drug effects , Kidney/immunology , Signal Transduction
15.
Biomed Pharmacother ; 106: 879-889, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119258

ABSTRACT

Acute kidney injury induced by ischemia-reperfusion injury (IRI) is a high risk factor in the progression towards chronic kidney disease, which is featured by renal interstitial fibrosis. Interleukin (IL)-18 is produced by T cells and macrophages and has been involved in the pathophysiology of IRI. However, the role of IL-18 in IRI-induced renal fibrosis is poorly understood. In the present study, we showed that interleukin (IL)-18 was significantly up-regulated after IRI stress. Mice treated with IL-18 Bp, a natural inhibitor of IL-18, presented less severe fibrotic response in the kidneys following IRI compared with vehicle-treated mice. Inhibition of IL-18 decreased myofibroblasts formation in the kidneys in response to IRI, which was associated with reduction of fibronectin and collagenⅠproteins. Moreover, inhibition of IL-18 impaired infiltration of CD3+ T cells and F4/80+ macrophages in the kidneys of mice after IRI. Treatment with IL-18 Bp reduces the levels of profibrotic molecules in the kidneys of mice following IRI. Finally, administration of IL-18 Bp impedes the transition of M2 macrophages to myofibroblasts and suppressed the accumulation of bone marrow-derived M2 macrophages. Adoptive transfer of M2 macrophages abolished the anti-fibrotic effect of IL-18 Bp. In summary, our results suggest that IL-18 plays an important role in the progression of IRI-induced renal fibrosis via modulating inflammation cells infiltration, the expression of inflammatory cytokines and chemokines, and the transition of bone marrow-derived M2 macrophages to myofibroblasts.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Interleukin-18/antagonists & inhibitors , Kidney/drug effects , Macrophages/drug effects , Myofibroblasts/drug effects , Reperfusion Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Adoptive Transfer , Animals , Cell Transdifferentiation/drug effects , Chemotaxis, Leukocyte/drug effects , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Macrophages/pathology , Macrophages/transplantation , Male , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
16.
J Physiol Biochem ; 74(4): 503-510, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30019185

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease. NLRP3 inflammasome activation has been widely studied in the pathogenesis of NAFLD. Cathepsin B (CTSB) is a ubiquitous cysteine cathepsin, and the role of CTSB in the progression and development of NAFLD has received extensive concern. However, the exact roles of CTSB in the NAFLD development and NLRP3 inflammasome activation are yet to be evaluated. In the present study, we used methionine choline-deficient (MCD) diet to establish mice NASH model. CTSB inhibitor (CA-074) was used to suppress the expression of CSTB. Expressions of CTSB and caspase-1 were evaluated by immunohistochemical staining. Serum IL-1ß and IL-18 levels were also determined. Palmitic acid was used to stimulate Kupffer cells (KCs), and protein expressions of CTSB, NLRP3, ASC (apoptosis-associated speck-like protein containing CARD), and caspase-1 in KCs were detected. The levels of IL-1ß and IL-18 in the supernatant of KCs were evaluated by enzyme-linked immunosorbent assay (ELISA). Our results showed that CTSB inhibition improved the liver function and reduced hepatic inflammation and ballooning, and the levels of pro-inflammatory cytokines IL-1ß and IL-18 were decreased. The expressions of CTSB and caspase-1 in liver tissues were increased in the NASH group. In in vitro experiments, PA stimulation could increase the expressions of CTSB and NLRP3 inflammasome in KCs, and CTSB inhibition downregulated the expression of NLRP3 inflammasome in KCs, when challenged by PA. Moreover, CTSB inhibition effectively suppressed the expression and activity of caspase-1 and subsequently secretions of IL-1ß and IL-18. Collectively, these results suggest that CTSB inhibition limits NLRP3 inflammasome-dependent NASH formation through regulating the expression and activity of caspase-1, thus providing a novel anti-inflammatory signal pathway for the therapy of NAFLD.


Subject(s)
Caspase 1/metabolism , Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Dipeptides/therapeutic use , Disease Models, Animal , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Caspase 1/chemistry , Cathepsin B/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/administration & dosage , Dipeptides/administration & dosage , Enzyme Activation/drug effects , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Injections, Intraperitoneal , Interleukin-18/antagonists & inhibitors , Interleukin-18/blood , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Kupffer Cells/drug effects , Kupffer Cells/immunology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/immunology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Random Allocation
17.
J Biochem Mol Toxicol ; 32(9): e22189, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29992668

ABSTRACT

Acute renal failure is one of the most frequent effects observed after taking medicine. Such situations have been tardily discovered, given that existing methods for assessing toxicity are not predictive. In this light, the present work evaluated the effects of gentamicin, a form of nephrotoxic drug, on HK-2 and HEK-293 cells. By using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and flow cytometry, both cells demonstrated that cytotoxicity occurs in a dose-dependent manner through the processes of apoptosis and cell necrosis. Gene expression analysis showed a relative increase of expression for genes related to cell processes and classic biomarkers, such as TP53, CASP3, CASP8, CASP9, ICAM-1, EXOC3, KIM-1, and CST3. A decrease in expression for genes BCL2L1 and EGF was observed. This study, therefore, indicates that, when the methods are used together, gene expression analysis is able to evaluate the nephrotoxic potential of a substance.


Subject(s)
Anti-Bacterial Agents/adverse effects , Apoptosis/drug effects , Gene Expression Regulation/drug effects , Gentamicins/adverse effects , Kidney/drug effects , Protein Synthesis Inhibitors/adverse effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animal Use Alternatives , Biomarkers, Pharmacological/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Cystatin C/agonists , Cystatin C/genetics , Cystatin C/metabolism , Drug Evaluation, Preclinical/methods , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Flow Cytometry , Gene Expression Profiling , Hepatitis A Virus Cellular Receptor 1/agonists , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-18/antagonists & inhibitors , Interleukin-18/genetics , Interleukin-18/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Necrosis
18.
Biochem Pharmacol ; 155: 150-161, 2018 09.
Article in English | MEDLINE | ID: mdl-29963998

ABSTRACT

Interleukin (IL)-1 and IL-18 belong to the IL-1 family of ligands, and their receptors are members of the IL-1 receptor family. Both cytokines drive an extensive range of pro-inflammatory networks in many cell types using common signal transduction cascades. Anyway, differences in signaling pathways exist. With this aim in mind, we investigated by using transgenic mice the mechanisms through the simultaneous deficiency of both IL-1ß and IL-18 could be more protective compared to blocking the single cytokine IL-1ß or IL-18 during colitis. Colitis was provoked in mice by instillation of dinitrobenzene sulfonic acid (DNBS) in the colon. The results indicated that single knockout (KO) mice of IL-1ß or IL-18, and double KO mice of both IL-1ß and IL-18 were hyporesponsive to DNBS-induced colitis compared to wild type (WT) mice, in which double KO were less sensitive than single KO mice. Moreover, treatment with Anakinra (IL-1R antagonist) also ameliorated colitis, in views of macroscopic and histological alteration, infiltration of neutrophils or Th1 cells, oxidative and nitrosative stress. Anakinra more significantly reduced cyclooxygenase (COX-2) and nuclear factor (NF-κB) levels as well as IKB-α degradation compared to blocking IL-18. On the contrary, the absence of IL-18 reduced p-ERK and p-p38 mitogen-activated protein kinase (MAPKs) in a more significant way compared to blocking IL-1ß. Thus, the double KO increased the protective effects against colon inflammation maybe because different converging inflammatory pathways are being inhibited. In conclusion, the blocking of both IL-1ß and IL-18 function may be advantageous in the treatment of IBD or inflammatory diseases.


Subject(s)
Colitis/chemically induced , Colitis/metabolism , Dinitrofluorobenzene/analogs & derivatives , Drug Delivery Systems/methods , Interleukin-18/deficiency , Interleukin-1beta/deficiency , Animals , Colitis/drug therapy , Dinitrofluorobenzene/toxicity , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin-18/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Inflammopharmacology ; 26(3): 725-735, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29600486

ABSTRACT

BACKGROUND AND AIMS: Behçet's disease (BD) is a chronic multisystemic inflammatory disease with complex etiopathogenesis. Th1-proinflammatory cytokines seem to be involved in its pathogenesis. Our current study aims to evaluate interleukin-18 (IL-18) and nitric oxide (NO) involvement in the development of different clinical manifestations of BD as well as to investigate the corticosteroid therapy effect on this production in Algerian patients. METHODS: For this purpose, we evaluated in vivo and ex vivo IL-18, interferon-γ (IFN-γ) levels using ELISA and NO production by the Griess' method in naïve-active and corticosteroid-treated BD patients with different clinical manifestations. Additionally, we assessed CD40/CD40L expression by flow cytometrics assay in these groups of patients. RESULTS AND DISCUSSION: Our results indicate that IL-18 and nitrite levels were higher in naïve-active BD patients. Interestingly, this high production differed according to the clinical manifestations and was associated with an increased risk of mucocutaneous and vascular involvement. Concerning corticosteroid treated-active BD patients, no difference was observed in this production between each clinical subgroup. However, IFN-γ levels increased in all categories of active patients. Interestingly, corticosteroid therapy reduced significantly these inflammatory mediators regardless of the clinical manifestations studied. In addition, the CD40/CD40L expression differed according to the clinical presentations. CONCLUSION: Collectively, our results suggest that concomitant high production of IL-18 and NO in naïve-active BD patients is related to an increased risk of mucocutaneous lesions and vascular involvement. Moreover, the relationship between these two inflammatory markers could constitute a predictable tool of BD clinical presentations and an early factor of therapy efficiency.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Behcet Syndrome/blood , Behcet Syndrome/drug therapy , Interleukin-18/blood , Nitric Oxide/blood , Adrenal Cortex Hormones/pharmacology , Adult , Algeria/epidemiology , Behcet Syndrome/epidemiology , Biomarkers/blood , Female , Humans , Interleukin-18/antagonists & inhibitors , Male , Middle Aged , Nitric Oxide/antagonists & inhibitors
20.
Front Immunol ; 9: 161, 2018.
Article in English | MEDLINE | ID: mdl-29472923

ABSTRACT

Acetaminophen [paracetamol, N-acetyl-p-aminophenol (APAP)]-induced acute liver injury (ALI) not only remains a persistent clinical challenge but likewise stands out as well-characterized paradigmatic model of drug-induced liver damage. APAP intoxication associates with robust hepatic necroinflammation the role of which remains elusive with pathogenic but also pro-regenerative/-resolving functions being ascribed to leukocyte activation. Here, we shine a light on and put forward a unique role of the interleukin (IL)-1 family member IL-18 in experimental APAP-induced ALI. Indeed, amelioration of disease as previously observed in IL-18-deficient mice was further substantiated herein by application of the IL-18 opponent IL-18-binding protein (IL-18BPd:Fc) to wild-type mice. Data altogether emphasize crucial pathological action of this cytokine in APAP toxicity. Adding recombinant IL-22 to IL-18BPd:Fc further enhanced protection from liver injury. In contrast to IL-18, the role of prototypic pro-inflammatory IL-1 and tumor necrosis factor-α is controversially discussed with lack of effects or even protective action being repeatedly reported. A prominent detrimental function for IL-18 in APAP-induced ALI as proposed herein should relate to its pivotal role for hepatic expression of interferon-γ and Fas ligand, both of which aggravate APAP toxicity. As IL-18 serum levels increase in patients after APAP overdosing, targeting IL-18 may evolve as novel therapeutic option in those hard-to-treat patients where standard therapy with N-acetylcysteine is unsuccessful. Being a paradigmatic experimental model of ALI, current knowledge on ill-fated properties of IL-18 in APAP intoxication likewise emphasizes the potential of this cytokine to serve as therapeutic target in other entities of inflammatory liver diseases.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/immunology , Interleukin-18/immunology , Liver/drug effects , Animals , Fas Ligand Protein/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Inflammation Mediators , Intercellular Signaling Peptides and Proteins/administration & dosage , Interferon-gamma/metabolism , Interleukin-18/antagonists & inhibitors , Interleukins/administration & dosage , Liver/immunology , Liver/pathology , Mice , Mice, Inbred C57BL , Necrosis/chemically induced , Necrosis/drug therapy , Necrosis/immunology , Recombinant Proteins/administration & dosage , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL