Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Joint Bone Spine ; 91(3): 105698, 2024 May.
Article in English | MEDLINE | ID: mdl-38309518

ABSTRACT

OBJECTIVE: Hyperuricaemia is necessary for gout. High urate concentrations have been linked to inflammation in mononuclear cells. Here, we explore the role of the suppressor of cytokine signaling 3 (SOCS3) in urate-induced inflammation. METHODS: Peripheral blood mononuclear cells (PBMCs) from gout patients, hyperuricemic and normouricemic individuals were cultured for 24h with varying concentrations of soluble urate, followed by 24h restimulation with lipopolysaccharides (LPS)±monosodium urate (MSU) crystals. Transcriptomic profiling was performed using RNA-Sequencing. DNA methylation was assessed using Illumina Infinium® MethylationEPIC BeadChip system (EPIC array). Phosphorylation of signal transducer and activator of transcription 3 (STAT3) was determined by flow cytometry. Cytokine responses were also assessed in PBMCs from patients with JAK2 V617F tyrosine kinase mutation. RESULTS: PBMCs pre-treated with urate produced more interleukin-1beta (IL-1ß) and interleukin-6 (IL-6) and less interleukin-1 receptor anatagonist (IL-1Ra) after LPS simulation. In vitro, urate treatment enhanced SOCS3 expression in control monocytes but no DNA methylation changes were observed at the SOCS3 gene. A dose-dependent reduction in phosphorylated STAT3 concomitant with a decrease in IL-1Ra was observed with increasing concentrations of urate. PBMCs with constitutively activated STAT3 (JAK2 V617F mutation) could not be primed by urate. CONCLUSION: In vitro, urate exposure increased SOCS3 expression, while urate priming, and subsequent stimulation resulted in decreased STAT3 phosphorylation and IL-1Ra production. There was no evidence that DNA methylation constitutes a regulatory mechanism of SOCS3. Elevated SOCS3 and reduced pSTAT3 could play a role in urate-induced hyperinflammation since urate priming had no effect in PBMCs from patients with constitutively activated STAT3.


Subject(s)
Cytokines , Gout , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Uric Acid , Humans , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Uric Acid/pharmacology , STAT3 Transcription Factor/metabolism , Cytokines/metabolism , Gout/genetics , Gout/metabolism , Cells, Cultured , Male , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Hyperuricemia/metabolism , Female , Middle Aged , DNA Methylation , Janus Kinase 2/metabolism
2.
Cytokine ; 175: 156502, 2024 03.
Article in English | MEDLINE | ID: mdl-38237388

ABSTRACT

BACKGROUND: Hyperuricemia has been shown to be an inducer of pro-inflammatory mediators by human primary monocytes. To study the deleterious effects of hyperuricemia, a reliable and stable in vitro model using soluble urate is needed. One recent report showed different urate-dissolving methods resulted in either pro-inflammatory or anti-inflammatory properties. The aim of this study was to compare the effect of two methods of dissolving urate on both primary human peripheral blood mononuclear cells (PBMCs) and THP-1 cells. The two methods tested were 'pre-warming' and 'dissolving with NaOH'. METHODS: Primary human PBMCs and THP-1 cells were exposed to urate solutions, prepared using the two methodologies: pre-warming and dissolving with NaOH. Afterwards, cells were stimulated with various stimuli, followed by the measurement of the inflammatory mediators IL-1ß, IL-6, IL-1Ra, TNF, IL-8, and MCP-1. RESULTS: In PBMCs, we observed an overall pro-inflammatory effect of urate, both in the pre-warming and the NaOH dissolving method. A similar pro-inflammatory effect was seen in THP-1 cells for both dissolving methods after restimulation. However, THP-1 cells exhibited pro-inflammatory profile with exposure to urate alone without restimulation. We did not find MSU crystals in our cellular assays. CONCLUSIONS: Overall, the urate dissolving methods do not have critical impact on its inflammatory properties. Soluble urate prepared using either of the two methods showed mostly pro-inflammatory effects on human primary PBMCs and monocytic cell line THP-1. However, human primary PBMCs and the THP-1 differ in their response to soluble urate without restimulation.


Subject(s)
Hyperuricemia , Uric Acid , Humans , Uric Acid/pharmacology , Uric Acid/metabolism , Hyperuricemia/metabolism , Leukocytes, Mononuclear/metabolism , Sodium Hydroxide/metabolism , Sodium Hydroxide/pharmacology , Monocytes , Inflammation Mediators/metabolism
3.
Int J Mol Sci ; 24(19)2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37833885

ABSTRACT

Chronic immune activation in systemic sclerosis is supported by the production of a plethora of cytokines with proven regulatory activities of the immune responses. This study aimed to explore PBMCs' cytokine profiles in SSc patients versus controls, as well as to investigate the balance between pro- and anti-inflammatory cytokines in association with disease duration. PBMCs were isolated from 18 SSc patients and 17 controls and further subjected to in vitro stimulation with lipopolysaccharide and heat-killed Candida albicans. Cytokine production was measured after 24 h and 7 days, respectively, using ELISA kits for interleukin (IL)-1ß, IL-1 receptor antagonist (IL-1Ra), IL-6, tumor necrosis factor (TNF), IL-10, IL-17, and interferon-gamma (IFN-gamma). IL-1 ß, IL-6, and TNF levels were increased in SSc patients compared with healthy volunteers irrespective of the stimulus used. IL-1Ra and Il-17 concentrations were not statistically different between groups, even though a trend toward higher levels in patients compared with their matched controls was also observed. Most cytokines demonstrated a stable course with disease progression, except for IL-10 levels, which declined over time. In conclusion, the results of this pilot study reveal that in patients with SSc a persistently enhanced immune response is established and maintained regardless of stimulus or disease duration.


Subject(s)
Leukocytes, Mononuclear , Scleroderma, Systemic , Humans , Interleukin-10 , Interleukin-17/pharmacology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-6/pharmacology , Pilot Projects , Cytokines , Tumor Necrosis Factor-alpha/pharmacology , Immunity
4.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37108648

ABSTRACT

Autoimmune hepatitis (AIH) is characterized by immune-mediated hepatocyte injury resulting in the destruction of liver cells, causing inflammation, liver failure, and fibrosis. Pediatric (AIH) is an autoimmune inflammatory disease that usually requires immunosuppression for an extended period. Frequent relapses after treatment discontinuation demonstrate that current therapies do not control intrahepatic immune processes. This study describes targeted proteomic profiling data in patients with AIH and controls. A total of 92 inflammatory and 92 cardiometabolic plasma markers were assessed for (i) pediatric AIH versus controls, (ii) AIH type 1 versus type 2, (iii) AIH and AIH-autoimmune sclerosing cholangitis overlapping syndrome and (iv) correlations with circulating vitamin D levels in AIH. A total of 16 proteins showed a nominally significant differential abundance in pediatric patients with AIH compared to controls. No clustering of AIH subphenotypes based on all protein data was observed, and no significant correlation of vitamin D levels was observed for the identified proteins. The proteins that showed variable expression include CA1, CA3, GAS6, FCGR2A, 4E-BP1 and CCL19, which may serve as potential biomarkers for patients with AIH. CX3CL1, CXCL10, CCL23, CSF1 and CCL19 showed homology to one another and may be coexpressed in AIH. CXCL10 seems to be the central intermediary link for the listed proteins. These proteins were involved in relevant mechanistic pathways for liver diseases and immune processes in AIH pathogenesis. This is the first report on the proteomic profile of pediatric AIH. The identified markers could potentially lead to new diagnostic and therapeutic tools. Nevertheless, considering the complex pathogenesis of AIH, more extensive studies are warranted to replicate and validate the present study's findings.


Subject(s)
Cholangitis, Sclerosing , Hepatitis, Autoimmune , Liver Diseases , Humans , Child , Hepatitis, Autoimmune/diagnosis , Proteomics , Cholangitis, Sclerosing/therapy , Biomarkers , Vitamin D
5.
Osteoarthritis Cartilage ; 31(8): 1022-1034, 2023 08.
Article in English | MEDLINE | ID: mdl-37105395

ABSTRACT

OBJECTIVE: Basic calcium phosphate (BCP) crystals can activate the NLRP3 inflammasome and are potentially involved in the pathogenesis of osteoarthritis (OA). In order to elucidate relevant inflammatory mechanisms in OA, we used a functional genomics approach to assess genetic variation influencing BCP crystal-induced cytokine production. METHOD: Peripheral blood mononuclear cells (PBMCs) were isolated from healthy volunteers who were previously genotyped and stimulated with BCP crystals and/or lipopolysaccharide (LPS) after which cytokines release was assessed. Cytokine quantitative trait locus (cQTL) mapping was performed. For in vitro validation of the cQTL located in anoctamin 3 (ANO3), PBMCs were incubated with Tamoxifen and Benzbromarone prior to stimulation. Additionally, we performed co-localisation analysis of our top cQTLs with the most recent OA meta-analysis of genome-wide association studies (GWAS). RESULTS: We observed that BCP crystals and LPS synergistically induce IL-1ß in human PBMCs. cQTL analysis revealed several suggestive loci influencing cytokine release upon stimulation, among which are quantitative trait locus annotated to ANO3 and GLIS3. As functional validation, anoctamin inhibitors reduced IL-1ß release in PBMCs after stimulation. Co-localisation analysis showed that the GLIS3 locus was shared between LPS/BCP crystal-induced IL-1ß and genetic association with Knee OA. CONCLUSIONS: We identified and functionally validated a new locus, ANO3, associated with LPS/BCP crystal-induced inflammation in PBMCs. Moreover, the cQTL in the GLIS3 locus co-localises with the previously found locus associated with Knee OA, suggesting that this Knee OA locus might be explained through an inflammatory mechanism. These results form a basis for further exploration of inflammatory mechanisms in OA.


Subject(s)
Osteoarthritis, Knee , Quantitative Trait Loci , Humans , Toll-Like Receptor 4/genetics , Leukocytes, Mononuclear , Genome-Wide Association Study , Lipopolysaccharides , Calcium Phosphates/pharmacology , Inflammation/genetics , Genomics , Anoctamins
6.
Arthritis Res Ther ; 25(1): 30, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36850003

ABSTRACT

BACKGROUND: Soluble urate leads to a pro-inflammatory phenotype in human monocytes characterized by increased production of IL-1ß and downregulation of IL-1 receptor antagonist, the mechanism of which remains to be fully elucidated. Previous transcriptomic data identified differential expression of genes in the transforming growth factor (TGF)-ß pathway in monocytes exposed to urate in vitro. In this study, we explore the role of TGF-ß in urate-induced hyperinflammation in peripheral blood mononuclear cells (PBMCs). METHODS: TGF-ß mRNA in unstimulated PBMCs and protein levels in plasma were measured in individuals with normouricemia, hyperuricemia and gout. For in vitro validation, PBMCs of healthy volunteers were isolated and treated with a dose ranging concentration of urate for assessment of mRNA and pSMAD2. Urate and TGF-ß priming experiments were performed with three inhibitors of TGF-ß signalling: SB-505124, 5Z-7-oxozeaenol and a blocking antibody against TGF-ß receptor II. RESULTS: TGF-ß mRNA levels were elevated in gout patients compared to healthy controls. TGF-ß-LAP levels in serum were significantly higher in individuals with hyperuricemia compared to controls. In both cases, TGF-ß correlated positively to serum urate levels. In vitro, urate exposure of PBMCs did not directly induce TGF-ß but did enhance SMAD2 phosphorylation. The urate-induced pro-inflammatory phenotype of monocytes was partly reversed by blocking TGF-ß. CONCLUSIONS: TGF-ß is elevated in individuals with hyperuricemia and correlated to serum urate concentrations. In addition, the urate-induced pro-inflammatory phenotype in human monocytes is mediated by TGF-ß signalling. Future studies are warranted to explore the intracellular pathways involved and to assess the clinical significance of urate-TGF-ß relation.


Subject(s)
Gout , Hyperuricemia , Humans , Gout/genetics , Leukocytes , Leukocytes, Mononuclear , Uric Acid/pharmacology , Transforming Growth Factor beta/genetics
7.
Nat Rev Rheumatol ; 16(2): 75-86, 2020 02.
Article in English | MEDLINE | ID: mdl-31822862

ABSTRACT

Asymptomatic hyperuricaemia affects ~20% of the general population in the USA, with variable rates in other countries. Historically, asymptomatic hyperuricaemia was considered a benign laboratory finding with little clinical importance in the absence of gout or kidney stones. Yet, increasing evidence suggests that asymptomatic hyperuricaemia can predict the development of hypertension, obesity, diabetes mellitus and chronic kidney disease and might contribute to disease by stimulating inflammation. Although urate has been classically viewed as an antioxidant with beneficial effects, new data suggest that both crystalline and soluble urate activate various pro-inflammatory pathways. This Review summarizes what is known about the role of urate in the inflammatory response. Further research is needed to define the role of asymptomatic hyperuricaemia in these pro-inflammatory pathways.


Subject(s)
Biomarkers/metabolism , Hyperuricemia/immunology , Immunity, Innate , Asymptomatic Diseases , Cytokines/metabolism , Humans , Hyperuricemia/metabolism , Uric Acid/metabolism
8.
Proc Natl Acad Sci U S A ; 114(21): 5485-5490, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28484006

ABSTRACT

Metabolic triggers are important inducers of the inflammatory processes in gout. Whereas the high serum urate levels observed in patients with gout predispose them to the formation of monosodium urate (MSU) crystals, soluble urate also primes for inflammatory signals in cells responding to gout-related stimuli, but also in other common metabolic diseases. In this study, we investigated the mechanisms through which uric acid selectively lowers human blood monocyte production of the natural inhibitor IL-1 receptor antagonist (IL-1Ra) and shifts production toward the highly inflammatory IL-1ß. Monocytes from healthy volunteers were first primed with uric acid for 24 h and then subjected to stimulation with lipopolysaccharide (LPS) in the presence or absence of MSU. Transcriptomic analysis revealed broad inflammatory pathways associated with uric acid priming, with NF-κB and mammalian target of rapamycin (mTOR) signaling strongly increased. Functional validation did not identify NF-κB or AMP-activated protein kinase phosphorylation, but uric acid priming induced phosphorylation of AKT and proline-rich AKT substrate 40 kDa (PRAS 40), which in turn activated mTOR. Subsequently, Western blot for the autophagic structure LC3-I and LC3-II (microtubule-associated protein 1A/1B-light chain 3) fractions, as well as fluorescence microscopy of LC3-GFP-overexpressing HeLa cells, revealed lower autophagic activity in cells exposed to uric acid compared with control conditions. Interestingly, reactive oxygen species production was diminished by uric acid priming. Thus, the Akt-PRAS40 pathway is activated by uric acid, which inhibits autophagy and recapitulates the uric acid-induced proinflammatory cytokine phenotype.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Monocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Uric Acid/metabolism , Animals , Disease Models, Animal , Gout/metabolism , Granulomatous Disease, Chronic/metabolism , HeLa Cells , Humans , Hyperuricemia/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Mice , Reactive Oxygen Species/metabolism , Transcriptome , Urate Oxidase/antagonists & inhibitors
9.
Curr Opin Rheumatol ; 29(2): 163-170, 2017 03.
Article in English | MEDLINE | ID: mdl-27941389

ABSTRACT

PURPOSE OF REVIEW: Gout is a common debilitating form of arthritis and despite our extensive knowledge on the pathogenesis its prevalence is still rising quickly. In the current review, we provide a concise overview of recent discoveries in factors tuning the inflammatory response to soluble uric acid and monosodium urate crystals. RECENT FINDINGS: It appears that soluble uric acid has a much larger role to play than just being a risk factor for gout. It may have widespread consequences for systemic inflammation and the development of metabolic syndrome. Additionally, a specific gout-related gut microbiome might not only provide us with a new diagnostic tool, but also highlights possible new therapeutic targets. Furthermore, several recent publications further elucidated the roles of mitochondrial dysfunction, production of reactive oxygen species, autophagy, and AMP-dependent protein kinase in monosodium urate-induced NLRP3 inflammasome activation. Finally, neutrophils have been shown to be involved in both the promotion and resolution of gouty inflammation. A new alpha-1-antitrypsin fusion protein may limit the proinflammatory effects of neutrophil-derived serine proteases. SUMMARY: Together, these studies provide us with many new insights in the pathogenesis of gout, important new treatment targets, and a rationale to further study the role of soluble uric acid in inflammatory diseases.


Subject(s)
Arthritis, Gouty/immunology , Gastrointestinal Microbiome/immunology , Hyperuricemia/immunology , Uric Acid/immunology , Autophagy/immunology , Gout/immunology , Humans , Inflammasomes/immunology , Inflammation , Mitochondria/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/immunology , Oxidative Stress , Phosphotransferases (Phosphate Group Acceptor)/immunology , Reactive Oxygen Species/immunology
10.
Ann Rheum Dis ; 75(3): 593-600, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25589513

ABSTRACT

OBJECTIVES: Acute gouty arthritis is caused by endogenously formed monosodium urate (MSU) crystals, which are potent activators of the NLRP3 inflammasome. However, to induce the release of active interleukin (IL)-1ß, an additional stimulus is needed. Saturated long-chain free fatty acids (FFAs) can provide such a signal and stimulate transcription of pro-IL-1ß. In contrast, the short-chain fatty acid butyrate possesses anti-inflammatory effects. One of the mechanisms involved is inhibition of histone deacetylases (HDACs). Here, we explored the effects of butyrate on MSU+FFA-induced cytokine production and its inhibition of specific HDACs. METHODS: Freshly isolated peripheral blood mononuclear cells (PBMCs) from healthy donors were stimulated with MSU and palmitic acid (C16.0) in the presence or absence of butyrate or a synthetic HDAC inhibitor. Cytokine responses were measured with ELISA and quantitative PCR. HDAC activity was measured with fluorimetric assays. RESULTS: Butyrate decreased C16.0+MSU-induced production of IL-1ß, IL-6, IL-8 and IL-1ß mRNA in PBMCs from healthy donors. Similar results were obtained in PBMCs isolated from patients with gout. Butyrate specifically inhibited class I HDACs. The HDAC inhibitor, panobinostat and the potent HDAC inhibitor, ITF-B, also decreased ex vivo C16.0+MSU-induced IL-1ß production. CONCLUSIONS: In agreement with the reported low inhibitory potency of butyrate, a high concentration was needed for cytokine suppression, whereas synthetic HDAC inhibitors showed potent anti-inflammatory effects at nanomolar concentrations. These novel HDAC inhibitors could be effective in the treatment of acute gout. Moreover, the use of specific HDAC inhibitors could even improve the efficacy and reduce any potential adverse effects.


Subject(s)
Antioxidants/pharmacology , Arthritis, Gouty , Butyrates/pharmacology , Cytokines/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Palmitic Acid/pharmacology , RNA, Messenger/drug effects , Uric Acid/pharmacology , Adult , Carbamates/pharmacology , Crystallization , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Histone Deacetylases , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Leukocytes, Mononuclear , Male , Middle Aged , Panobinostat , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Ann Rheum Dis ; 75(6): 1219-27, 2016 06.
Article in English | MEDLINE | ID: mdl-26174021

ABSTRACT

OBJECTIVES: In the present study, we generated a new protein, recombinant human alpha-1-anti-trypsin (AAT)-IgG1 Fc fusion protein (AAT-Fc), and evaluated its properties to suppress inflammation and interleukin (IL)-1ß in a mouse model of gouty arthritis. METHODS: A combination of monosodium urate (MSU) crystals and the fatty acid C16.0 (MSU/C16.0) was injected intra-articularly into the knee to induce gouty arthritis. Joint swelling, synovial cytokine production and histopathology were determined after 4 h. AAT-Fc was evaluated for inhibition of MSU/C16.0-induced IL-1ß release from human blood monocytes and for inhibition of extracellular IL-1ß precursor processing. RESULTS: AAT-Fc markedly suppressed MSU/C16.0-induced joint inflammation by 85-91% (p<0.001). Ex vivo production of IL-1ß and IL-6 from cultured synovia were similarly reduced (63% and 65%, respectively). The efficacy of 2.0 mg/kg AAT-Fc in reducing inflammation was comparable to 80 mg/kg of plasma-derived AAT. Injection of AAT-Fc into mice increased circulating levels of endogenous IL-1 receptor antagonist by fourfold. We also observed that joint swelling was reduced by 80%, cellular infiltration by 95% and synovial production of IL-1ß by 60% in transgenic mice expressing low levels of human AAT. In vitro, AAT-Fc reduced MSU/C16.0-induced release of IL-1ß from human blood monocytes and inhibited proteinase-3-mediated extracellular processing of the IL-1ß precursor into active IL-1ß. CONCLUSIONS: A single low dose of AAT-Fc is highly effective in reducing joint inflammation in this model of acute gouty arthritis. Considering the long-term safety of plasma-derived AAT use in humans, subcutaneous AAT-Fc emerges as a promising therapy for gout attacks.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Gouty/drug therapy , Gout Suppressants/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin-1beta/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , alpha 1-Antitrypsin/therapeutic use , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Gouty/immunology , Arthritis, Gouty/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Gout Suppressants/administration & dosage , Gout Suppressants/pharmacology , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/pharmacology , Injections, Intra-Articular , Injections, Intraperitoneal , Interleukin-1beta/metabolism , Lipopolysaccharide Receptors/analysis , Male , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/drug effects , Monocytes/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , alpha 1-Antitrypsin/administration & dosage , alpha 1-Antitrypsin/pharmacology
12.
Clin Rheumatol ; 33(9): 1341-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24760192

ABSTRACT

Monosodium urate (MSU) monohydrate crystals synergize with various toll-like receptor (TLR) ligands to induce interleukin-(IL)-1ß production. Data are shown from a young male with mitochondriopathy in Kearns-Sayre syndrome (KSS) who developed gout and underwent urate-lowering therapy (ULT) versus a group of common gout patients. Peripheral blood mononuclear cells (PBMCs) are exposed in vitro to MSU crystals in the presence/absence of TLR2 ligands palmitic acid (C16:0) or palmitoyl-3-cysteine (Pam3Cys); proinflammatory cytokine production (IL-1ß, IL-6, IL-8) is assessed by specific ELISA's. MSU crystals alone failed to induce IL-1beta, IL-6, or IL-8 in both the KSS patient and gout controls. A strong synergy between MSU crystals and C16:0 or Pam3Cys for induction of IL-1 beta/IL-6 is found in gout patients, but in gout with KSS, we found even more response than in control gout patients. Pam3Cys exposure reveals an enhanced response in cells originating from the KSS patient, indicating a high producer phenotype in response to TLR2 stimulation. During ULT, serum urate levels dropped in the KSS case. The hyperresponse of TLR2 may be secondary to the high serum urate concentration of 0.92 mmol/l that was initially found in circulation in vivo. Within a 6-month period, the serum urate concentration dropped, and the in vitro stimulation tests improved but did not fully normalize yet. The ex vivo cytokine production in gout patients is promising a novel gout test; PBMCs' responses in the mitochondriopathic gout patient is enhanced when compared with common gout patients, indicating a supersensitive gout patient profile. The non-inflammatory presentation in the KSS case with bulky gout is due to less inflammatory MSU crystals, i.e., specific crystal stereochemical/conformational properties. For developing gout attacks, the serum urate level and specific crystal properties both are of importance. Key Messages 1. Ex vivo cell tests are promising to serve as a novel gout lab test for screening purposes. 2. Ex vivo cellular responses are reduced following intraarticular glucocorticoid injection and/or urate-lowering therapy. 3. Crystal conformation properties play a role in the inflammatory in vivo and ex vivo response in gout. 4. A young male with Kearns-Sayre syndrome is described with less pronounced inflammatory PMBC responses to his own MSU crystals which explains the advanced stage of urate accumulation in this individual.


Subject(s)
Cytokines/biosynthesis , Gout/diagnosis , Kearns-Sayre Syndrome/diagnosis , Leukocytes, Mononuclear/metabolism , Adult , Gout/complications , Gout/metabolism , Humans , Kearns-Sayre Syndrome/complications , Kearns-Sayre Syndrome/metabolism , Male , Uric Acid/metabolism
13.
Arthritis Res Ther ; 14(4): R158, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22762240

ABSTRACT

INTRODUCTION: Monosodium urate monohydrate (MSU) crystals synergize with various toll-like receptor (TLR) ligands to induce cytokine production via activation of the NOD-like receptor (NLR) family, pyrin domain-containing 3 (NLPR3) inflammasome. This has been demonstrated in vitro using human cell lines or monocytes of healthy volunteers. In the present study, we have investigated the effect of MSU crystals and of their combination with TLR ligands in peripheral blood mononuclear cells (PBMC) of patients with gout. METHODS: PBMCs from 18 patients with primary gout and 12 healthy donors were exposed to MSU crystals in the presence or absence of saturated fatty acid C18:0 (free fatty acid, TLR2 ligand), palmitoyl-3-cystein (Pam3Cys, TLR1/2 ligand) and fibroblast stimulating factor-1 (FSL-1, TLR 2/6 ligand). Production of IL-1ß, IL-6, IL-8, IL-17 and tumor necrosis factor alpha (TNFα) was determined by ELISA. mRNA transcripts of IL-1ß were measured by real-time PCR. RESULTS: MSU crystals alone failed to induce IL-1ß, IL-6 or TNFα in both patients and control groups, but a stronger synergy between MSU/Pam3Cys and MSU/C18:0 for the induction of IL-1ß was found in patients with gout compared to healthy controls. IL-6, but not IL-8, followed the kinetics of IL-1ß. No production of the neutrophil-recruiting IL-17 was detectable after stimulation of the patients' PBMCs with MSU in both the presence or absence of TLR ligands. No change of gene transcripts of IL-1ß after stimulation with MSU and Pam3Cys or with MSU and C18:0 was found. A positive correlation was found between synergy in IL-1ß production from PBMCs of patients between C18:0 and MSU crystals, as well as the annual number of attacks of acute gouty arthritis (rs: +0.649, P: 0.022). CONCLUSIONS: The synergy between MSU crystals and TLR-2 ligands is more prominent in patients with gout than in controls. This is likely mediated by the enhanced maturation of pro-IL-1ß into IL-1ß.


Subject(s)
Gout/metabolism , Interleukin-1beta/biosynthesis , Leukocytes, Mononuclear/metabolism , Toll-Like Receptor 2/biosynthesis , Uric Acid/pharmacology , Adult , Aged , Crystallization , Female , Humans , Leukocytes, Mononuclear/drug effects , Ligands , Male , Middle Aged , Toll-Like Receptor 2/agonists
14.
J Gastrointestin Liver Dis ; 21(2): 177-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22720307

ABSTRACT

BACKGROUND AND AIMS: HFE-associated haemochromatosis is one of the most frequent autosomal recessive disorders in the Caucasian population. Although most of the cases are homozygous individuals for the C282Y mutation, another two mutations, H63D and S65C, have been reported to be associated with milder forms of the disease. This study was a first attempt to evaluate the distribution of these HFE gene mutations in the Transylvania region. METHODS: Two-hundred and twenty-five healthy, unrelated volunteers originating from the Transylvania region, Romania, were screened for the HFE gene C282Y, H63D and S65C mutations, using molecular genetics assays (Polymerase Chain Reaction-Restriction Fragments Length Polymorphism). RESULTS: For the C282Y mutation, 7 heterozygotes (3.1%) were found, but no homozygous individual. In the case of the H63D mutation, 40 heterozygotes (17.8%) and 4 homozygotes (1.75%) for the mutant allele were evidenced. We found a compound heterozygous genotype (C282Y/H63D) in one individual (0.45%). Thus, the allele frequencies of the C282Y and H63D were 1.75% and 10.9%, respectively. Three individuals (1.3%) were found to harbour the S65C mutation in a heterozygous state, but none in a homozygous state: the allele frequency of the mutant allele was 0.75%. CONCLUSIONS: The distribution of the HFE gene C282Y, H63D and S65C mutations found in our group matches the tendencies observed in other European countries: a decreasing gradient from Northern to Southern Europe for the C282Y mutation; high frequency for the H63D mutation, and low frequency for the S65C mutation in most of the countries.


Subject(s)
Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Mutation , Adolescent , Adult , Female , Gene Frequency , Genotype , Hemochromatosis/epidemiology , Hemochromatosis Protein , Heterozygote , Humans , Male , Middle Aged , Polymorphism, Restriction Fragment Length , Romania/epidemiology , Young Adult
16.
Gene ; 494(1): 109-11, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22200569

ABSTRACT

Toll-like receptors (TLRs) are critical components of the pathogen recognition by the host innate immune system. Recently it has been shown that TLR1 is under evolutionary pressure in Europeans. This involves the positive selection of the nonsynonymous TLR1 1805G variant in Europeans, although this is associated with poor TLR1 response and unfavorable prognosis in various infections. In terms of natural selection, differential fertility is another mechanism, independent of infection susceptibility, that may explain the polymorphism pattern observed for TLR1. To test this hypothesis, we assessed the correlation of two TLR1 SNPs (T1805G and G239C) with spontaneous pregnancy loss in a case-control study that included 132 spontaneous pregnancy loss patients and 142 control volunteers. Similar allele frequencies of T1805G were observed between cases and controls, but GG genotype tended to be associated with pregnancy loss (OR 1.91; 95%CI 1.03, 3.53). No differences were observed for the TLR1 G239C SNP. Our findings showed slight differences in the distribution of T1805G variants in women with pregnancy loss, but these were not indicative of a protective effect of the TLR1 1805G allele for this fertility disorder. Although our hypothesis was not proven, potential effects of TLR1 polymorphisms on pregnancy outcome have been suggested, and future studies in larger cohorts are warranted.


Subject(s)
Abortion, Spontaneous/genetics , Polymorphism, Single Nucleotide , Toll-Like Receptor 1/genetics , Adult , Case-Control Studies , Female , Gene Frequency , Genotype , Humans , Pregnancy , White People/genetics
17.
PLoS One ; 6(4): e18666, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21490934

ABSTRACT

Autophagy is a cell housekeeping mechanism that has recently received attention in relation to its effects on the immune response. Genetic studies have identified candidate loci for Crohn's disease susceptibility among autophagy genes, while experiments in murine macrophages from ATG16L1 deficient mice have shown that disruption of autophagy increases processing of IL-1ß and IL-18 through an inflammasome-dependent manner. Using complementary approaches either inducing or inhibiting autophagy, we describe modulatory effects of autophagy on proinflammatory cytokine production in human cells. Inhibition of basal autophagy in human peripheral blood mononuclear cells (PBMCs) significantly enhances IL-1ß after stimulation with TLR2 or TLR4 ligands, while at the same time reducing the production of TNFα. In line with this, induction of autophagy by starvation inhibited IL-1ß production. These effects of autophagy were not exerted at the processing step, as inflammasome activation was not influenced. In contrast, the effect of autophagy on cytokine production was on transcription level, and possibly involving the inhibition of p38 mitogen activated protein kinase (MAPK) phosphorylation. In conclusion, autophagy modulates the secretion of proinflammatory cytokines in human cells through an inflammasome-independent pathway, and this is a novel mechanism that may be targeted in inflammatory diseases.


Subject(s)
Autophagy/drug effects , Cytokines/metabolism , Inflammasomes/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Gut ; 60(9): 1229-35, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21406388

ABSTRACT

OBJECTIVE: Autophagy has recently been shown to modulate the production of pro-inflammatory cytokine production and to contribute to antigen processing and presentation through the major histocompatibility complex. Genetic variation in the autophagy gene ATG16L1 has been recently implicated in Crohn's disease pathogenesis. The mechanisms underlying this association are not yet known, although experimental models suggest an inhibitory effect of autophagy on interleukin 1ß (IL-1ß) responses. Here, the effect of ATG16L1 genetic variation on cytokine responses has been assessed in humans. DESIGN AND SETTING: Peripheral blood mononuclear cells from healthy individuals and patients with Crohn's disease with different ATG16L1 genotypes were stimulated with ligands for Toll-like receptor 2 (TLR2), TLR4 and nucleotide-binding oligomerisation domain 2 (NOD2), with or without the autophagy inhibitor 3-methyladenine. Induction of cytokine production and related factors were measured at the mRNA and protein level. Furthermore, protein levels of ATG16L1 were assessed by western blot. RESULTS: The present study demonstrates that cells isolated from individuals bearing the ATG16L1 Thr300Ala risk variant, which is shown to affect ATG16L1 protein expression upon NOD2 stimulation, display increased production of the pro-inflammatory cytokines IL-1ß and IL-6, specifically after stimulation with NOD2 ligands. In contrast, no differences were found when cells were stimulated with TLR2 or TLR4 agonists. These findings were confirmed in two independent cohorts of volunteers and in a group of patients with Crohn's disease. The increased production could be ascribed to increased mRNA expression, while processing of pro-IL-1ß by caspase-1 activation was not affected. The effect of the ATG16L1 polymorphism was abrogated when autophagy was blocked. CONCLUSIONS: The present study is the first to link the ATG16L1 polymorphism with an excessive production of IL-1ß and IL-6 in humans, which may explain the effects of this polymorphism on the inflammatory process in Crohn's disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Cytokines/biosynthesis , Inflammation Mediators/metabolism , Nod2 Signaling Adaptor Protein/physiology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/drug effects , Autophagy/genetics , Autophagy/immunology , Autophagy-Related Proteins , Carrier Proteins/biosynthesis , Caspase 1/metabolism , Cells, Cultured , Crohn Disease/immunology , Enzyme Activation/immunology , Gene Expression , Genetic Predisposition to Disease , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-6/biosynthesis , Leukocytes, Mononuclear/immunology , Polymorphism, Genetic , RNA, Messenger/genetics
19.
J Matern Fetal Neonatal Med ; 24(1): 189-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20334533

ABSTRACT

OBJECTIVE: Our aim was to evaluate the possible association between recurrent spontaneous abortions (RSA) and the c.1958 G>A SNP in the MTHFD1 gene encoding a trifunctional enzyme involved in DNA synthesis and folate metabolism. METHODS: By the means of PCR-RFLP we genotyped 131 women with a history of at least two consecutive spontaneous abortions and a matched number of controls. RESULTS: Our findings show an allele frequency of 44.3% of the A allele and 55.7% of the G allele in patients and 42.4% of the A allele and 57.6% of the G allele in controls. CONCLUSIONS: No major difference between cases and controls was revealed, therefore, it is unlikely that this SNP plays a major role in RSA.


Subject(s)
Abortion, Habitual/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Case-Control Studies , Female , Humans , Minor Histocompatibility Antigens , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL