Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 114
Filtrer
1.
Int J Mol Sci ; 24(3)2023 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-36768186

RÉSUMÉ

Monocyte chemoattractant protein-1 (MCP-1/CCL2) is a potent chemotactic agent for monocytes, primarily produced by macrophages and endothelial cells. Significantly elevated levels of MCP-1/CCL2 were found in synovial fluids of patients with rheumatoid arthritis (RA), compared to osteoarthritis or other arthritis patients. Several studies suggested an important role for MCP-1 in the massive inflammation at the damaged joint, in part due to its chemotactic and angiogenic effects. It is a known fact that the post-translational modifications (PTMs) of proteins have a significant impact on their properties. In mammals, arginine residues within proteins can be converted into citrulline by peptidylarginine deiminase (PAD) enzymes. Anti-citrullinated protein antibodies (ACPA), recognizing these PTMs, have become a hallmark for rheumatoid arthritis (RA) and other autoimmune diseases and are important in diagnostics and prognosis. In previous studies, we found that citrullination converts the neutrophil attracting chemokine neutrophil-activating peptide 78 (ENA-78) into a potent macrophage chemoattractant. Here we report that both commercially available and recombinant bacterially produced MCP-1/CCL2 are rapidly (partially) degraded upon in vitro citrullination. However, properly glycosylated MCP-1/CCL2 produced by mammalian cells is protected against degradation during efficient citrullination. Site-directed mutagenesis of the potential glycosylation site at the asparagine-14 residue within human MCP-1 revealed lower expression levels in mammalian expression systems. The glycosylation-mediated recombinant chemokine stabilization allows the production of citrullinated MCP-1/CCL2, which can be effectively used to calibrate crucial assays, such as modified ELISAs.


Sujet(s)
Polyarthrite rhumatoïde , Chimiokine CCL2 , Animaux , Humains , Chimiokine CCL2/métabolisme , Glycosylation , Cellules endothéliales/métabolisme , Polyarthrite rhumatoïde/métabolisme , Protéines/métabolisme , Mammifères/métabolisme , Citrulline/métabolisme
3.
Adv Ther ; 39(10): 4723-4741, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-35962234

RÉSUMÉ

INTRODUCTION: To compare the mortality of hospitalized patients with COVID-19 between those that required supplemental oxygen and received dexamethasone with a comparable set of patients who did not receive dexamethasone. METHODS: We utilized the Premier Health Database to identify hospitalized adult patients with COVID-19 from July 1, 2020-January 31, 2021. Index date was when patients first initiated oxygen therapy. The primary endpoint was in-hospital mortality for patients receiving dexamethasone versus those not receiving dexamethasone 1-day pre- to 1-day post-index period. Secondary endpoints included 28-day mortality, time to in-hospital mortality, progression to invasive mechanical ventilation or death, time to discharge, and proportion discharged alive by day 28. Twenty-three models using weighting, matching, stratification, and regression were deployed through the concept of frequentist model average (FMA) to estimate the effect of dexamethasone on all-cause mortality up to the 28-day hospitalization period. RESULTS: A total of 1,208,881 patients with COVID-19 were screened; as an inpatient 255,216 used oxygen, and 251,536 were included in the analysis. In the dexamethasone group, odds of in-hospital mortality were higher than those of the comparator (FMA: odds ratio [OR] 1.15, 95% CI 1.08, 1.22). Using a best fit model, OR for in-hospital mortality was non-significant for the dexamethasone group compared with the comparator (OR 1.02, 95% CI 0.92, 1.14). Dexamethasone treatment was associated with poorer outcomes versus the comparator group across the majority of secondary endpoints, except for number of days in hospital, which was lower in the dexamethasone group versus the comparator group (mean difference - 2.14, 95% CI - 2.43, - 1.47). CONCLUSIONS: Hospitalized adult patients with COVID-19 who required supplemental oxygen and received dexamethasone did not have a survival benefit versus similar patients not receiving dexamethasone. The dexamethasone group was not associated with favorable responses for outcomes such as progression to death or mechanical ventilation and time to in-hospital death.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Adulte , Dexaméthasone/usage thérapeutique , Mortalité hospitalière , Humains , Patients hospitalisés , Oxygène , SARS-CoV-2 , États-Unis
4.
Ann Rheum Dis ; 2022 May 24.
Article de Anglais | MEDLINE | ID: mdl-35609978

RÉSUMÉ

OBJECTIVES: To elucidate the mechanism of action of baricitinib, a Janus kinase (JAK) 1/2 inhibitor, and describe immunological pathways related to disease activity in adults with systemic lupus erythematosus (SLE) receiving standard background therapy in a phase II trial. METHODS: Patients with SLE were treated with baricitinib 2 mg or 4 mg in a phase II randomised, placebo-controlled study. Sera from 239 patients (baricitinib 2 mg: n=88; baricitinib 4 mg: n=82; placebo: n=69) and 49 healthy controls (HCs) were collected at baseline and week 12 and analysed using a proximity extension assay (Target 96 Inflammation Panel (Olink)). Interferon (IFN) scores were determined using an mRNA panel. Analytes were compared in patients with SLE versus HCs and in changes from baseline at week 12 between baricitinib 2 mg, 4 mg and placebo groups using a restricted maximum likelihood-based mixed models for repeated measures. Spearman correlations were computed for analytes and clinical measurements. RESULTS: At baseline, SLE sera had strong cytokine dysregulation relative to HC sera. C-C motif chemokine ligand (CCL) 19, C-X-C motif chemokine ligand (CXCL) 10, tumour necrosis factor alpha (TNF-α), TNF receptor superfamily member (TNFRSF)9/CD137, PD-L1, IL-6 and IL-12ß were significantly reduced in patients treated with baricitinib 4 mg versus placebo at week 12. Inflammatory biomarkers indicated correlations/associations with type I IFN (CCL19, CXCL10, TNF-α and PD-L1), anti-double stranded DNA (dsDNA) (TNF-α, CXCL10) and Systemic Lupus Erythematosus Disease Activity Index-2000, tender and swollen joint count and worst joint pain (CCL19, IL-6 and TNFRSF9/CD137). CONCLUSION: These results suggest that baricitinib 4 mg downregulated key cytokines that are upregulated in patients with SLE and may play a role in a multitargeted mechanism beyond the IFN signature although clinical relevance remains to be further delineated. TRIAL REGISTRATION NUMBER: NCT02708095.

5.
Arthritis Res Ther ; 22(1): 235, 2020 10 12.
Article de Anglais | MEDLINE | ID: mdl-33046136

RÉSUMÉ

BACKGROUND: Tissue released blood-based biomarkers can provide insight into drug mode of action and response. To understand the changes in extracellular matrix turnover, we analyzed biomarkers associated with joint tissue turnover from a phase 3, randomized, placebo-controlled study of baricitinib in patients with active rheumatoid arthritis (RA). METHODS: Serum biomarkers associated with synovial inflammation (C1M, C3M, and C4M), cartilage degradation (C2M), bone resorption (CTX-I), and bone formation (osteocalcin) were analyzed at baseline, and weeks 4 and 12, from a subgroup of patients (n = 240) randomized to placebo or 2-mg or 4-mg baricitinib (RA-BUILD, NCT01721057). Mixed-model repeated measure was used to identify biomarkers altered by baricitinib. The relationship between changes in biomarkers and clinical measures was evaluated using correlation analysis. RESULTS: Treatment arms were well balanced for baseline biomarkers, demographics, and disease activity. At week 4, baricitinib 4-mg significantly reduced C1M from baseline by 21% compared to placebo (p < 0.01); suppression was sustained at week 12 (27%, p < 0.001). Baricitinib 4-mg reduced C3M and C4M at week 4 by 14% and 12% compared to placebo, respectively (p < 0.001); they remained reduced by 16% and 11% at week 12 (p < 0.001). In a pooled analysis including all treatment arms, patients with the largest reduction (upper 25% quartile) in C1M, C3M, and C4M by week 12 had significantly greater clinical improvement in the Simplified Disease Activity Index at week 12 compared to patients with the smallest reduction (lowest 25% quartile). CONCLUSION: Baricitinib treatment resulted in reduced circulating biomarkers associated with joint tissue destruction as well as concomitant RA clinical improvement. TRIAL REGISTRATION: ClinicalTrials.gov NCT01721057 ; date of registration: November 1, 2012.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Antirhumatismaux/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Azétidines , Marqueurs biologiques , Humains , Janus kinase 1 , Méthotrexate/usage thérapeutique , Purines , Pyrazoles , Sulfonamides
6.
Arthritis Res Ther ; 20(1): 31, 2018 02 12.
Article de Anglais | MEDLINE | ID: mdl-29433546

RÉSUMÉ

BACKGROUND: Galectin-9 (Gal-9) is a mammalian lectin secreted by endothelial cells that is highly expressed in rheumatoid arthritis synovial tissues and synovial fluid. Roles have been proposed for galectins in the regulation of inflammation and angiogenesis. Therefore, we examined the contribution of Gal-9 to angiogenesis and inflammation in arthritis. METHODS: To determine the role of Gal-9 in angiogenesis, we performed human dermal microvascular endothelial cell (HMVEC) chemotaxis, Matrigel tube formation, and mouse Matrigel plug angiogenesis assays. We also examined the role of signaling molecules in Gal-9-induced angiogenesis by using signaling inhibitors and small interfering RNA (siRNA). We performed monocyte (MN) migration assays in a modified Boyden chamber and assessed the arthritogenicity of Gal-9 by injecting Gal-9 into mouse knees. RESULTS: Gal-9 significantly increased HMVEC migration, which was decreased by inhibitors of extracellular signal-regulating kinases 1/2 (Erk1/2), p38, Janus kinase (Jnk), and phosphatidylinositol 3-kinase. Gal-9 HMVEC-induced tube formation was reduced by Erk1/2, p38, and Jnk inhibitors, and this was confirmed by siRNA knockdown. In mouse Matrigel plug assays, plugs containing Gal-9 induced significantly higher angiogenesis, which was attenuated by a Jnk inhibitor. Gal-9 also induced MN migration, and there was a marked increase in MN ingress when C57BL/6 mouse knees were injected with Gal-9 compared with the control, pointing to a proinflammatory role for Gal-9. CONCLUSIONS: Gal-9 mediates angiogenesis, increases MN migration in vitro, and induces acute inflammatory arthritis in mice, suggesting a novel role for Gal-9 in angiogenesis, joint inflammation, and possibly other inflammatory diseases.


Sujet(s)
Polyarthrite rhumatoïde/métabolisme , Galectines/métabolisme , Inflammation/métabolisme , Néovascularisation pathologique/métabolisme , Animaux , Polyarthrite rhumatoïde/génétique , Mouvement cellulaire , Cellules cultivées , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Femelle , Galectines/génétique , Humains , Inflammation/génétique , Souris de lignée C57BL , Monocytes/cytologie , Monocytes/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation physiologique , Interférence par ARN
7.
Nat Commun ; 8: 14252, 2017 02 06.
Article de Anglais | MEDLINE | ID: mdl-28165452

RÉSUMÉ

Novel therapeutics are required for improving the management of chronic inflammatory diseases. Aptamers are single-stranded RNA or DNA molecules that have recently shown utility in a clinical setting, as they can specifically neutralize biomedically relevant proteins, particularly cell surface and extracellular proteins. The nuclear chromatin protein DEK is a secreted chemoattractant that is abundant in the synovia of patients with juvenile idiopathic arthritis (JIA). Here, we show that DEK is crucial to the development of arthritis in mouse models, thus making it an appropriate target for aptamer-based therapy. Genetic depletion of DEK or treatment with DEK-targeted aptamers significantly reduces joint inflammation in vivo and greatly impairs the ability of neutrophils to form neutrophil extracellular traps (NETs). DEK is detected in spontaneously forming NETs from JIA patient synovial neutrophils, and DEK-targeted aptamers reduce NET formation. DEK is thus key to joint inflammation, and anti-DEK aptamers hold promise for the treatment of JIA and other types of arthritis.


Sujet(s)
Aptamères nucléotidiques/usage thérapeutique , Arthrite juvénile/thérapie , Facteurs chimiotactiques/antagonistes et inhibiteurs , Protéines chromosomiques nonhistones/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/génétique , Pièges extracellulaires/immunologie , Protéines oncogènes/antagonistes et inhibiteurs , Protéines oncogènes/génétique , Protéines liant le poly-adp-ribose/antagonistes et inhibiteurs , Protéines liant le poly-adp-ribose/génétique , Adulte , Animaux , Arthrite juvénile/immunologie , Facteurs chimiotactiques/génétique , Facteurs chimiotactiques/immunologie , Facteurs chimiotactiques/métabolisme , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/immunologie , Protéines chromosomiques nonhistones/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/métabolisme , Modèles animaux de maladie humaine , Pièges extracellulaires/métabolisme , Femelle , Volontaires sains , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Granulocytes neutrophiles/immunologie , Protéines oncogènes/immunologie , Protéines oncogènes/métabolisme , Protéines liant le poly-adp-ribose/immunologie , Protéines liant le poly-adp-ribose/métabolisme , Culture de cellules primaires , Synovie/composition chimique , Synovie/cytologie , Synovie/immunologie , Zymosan/immunologie
8.
Cell Rep ; 17(10): 2620-2631, 2016 12 06.
Article de Anglais | MEDLINE | ID: mdl-27926866

RÉSUMÉ

Approximately 50% of prostate cancers are associated with gene fusions of the androgen-regulated gene TMPRSS2 to the oncogenic erythroblast transformation-specific (ETS) transcription factor ERG. The three-dimensional proximity of TMPRSS2 and ERG genes, in combination with DNA breaks, facilitates the formation of TMPRSS2-ERG gene fusions. However, the origins of DNA breaks that underlie gene fusion formation in prostate cancers are far from clear. We demonstrate a role for inflammation-induced oxidative stress in the formation of DNA breaks leading to recurrent TMPRSS2-ERG gene fusions. The transcriptional status and epigenetic features of the target genes influence this effect. Importantly, inflammation-induced de novo genomic rearrangements are blocked by homologous recombination (HR) and promoted by non-homologous end-joining (NHEJ) pathways. In conjunction with the association of proliferative inflammatory atrophy (PIA) with human prostate cancer, our results support a working model in which recurrent genomic rearrangements induced by inflammatory stimuli lead to the development of prostate cancer.


Sujet(s)
Inflammation/génétique , Protéines de fusion oncogènes/génétique , Tumeurs de la prostate/génétique , Serine endopeptidases/génétique , Androgènes/génétique , Lignée cellulaire tumorale , Cassures de l'ADN , Réparation de l'ADN par jonction d'extrémités/génétique , Protéines de liaison à l'ADN/génétique , Humains , Inflammation/complications , Inflammation/anatomopathologie , Mâle , Stress oxydatif/génétique , Tumeurs de la prostate/complications , Tumeurs de la prostate/anatomopathologie , Régulateur transcriptionnel ERG/génétique
9.
N Engl J Med ; 374(13): 1243-52, 2016 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-27028914

RÉSUMÉ

BACKGROUND: In phase 2 studies, baricitinib, an oral Janus kinase 1 and 2 inhibitor, reduced disease activity in patients with rheumatoid arthritis who had not previously received treatment with biologic disease-modifying antirheumatic drugs (DMARDs). METHODS: In this phase 3 study involving 527 patients with an inadequate response to or unacceptable side effects associated with one or more tumor necrosis factor inhibitors, other biologic DMARDs, or both, we randomly assigned the patients in a 1:1:1 ratio to baricitinib at a dose of 2 or 4 mg daily or placebo for 24 weeks. End points, tested hierarchically at week 12 to control type 1 error, were the American College of Rheumatology 20% (ACR20) response (primary end point), the Health Assessment Questionnaire-Disability Index (HAQ-DI) score, the 28-joint Disease Activity Score based on C-reactive protein level (DAS28-CRP), and a Simplified Disease Activity Index (SDAI) score of 3.3 or less (on a scale of 0.1 to 86.0, with a score of 3.3 or less indicating remission). Comparisons with placebo were made first with the 4-mg dose of baricitinib and then with the 2-mg dose. RESULTS: Significantly more patients receiving baricitinib at the 4-mg dose than those receiving placebo had an ACR20 response at week 12 (55% vs. 27%, P<0.001). Differences between the higher-dose baricitinib group and the placebo group were also significant for the HAQ-DI score and the DAS28-CRP but not for an SDAI score of 3.3 or less. Adverse-event rates through 24 weeks were higher for patients receiving the 2-mg dose of baricitinib and those receiving the 4-mg dose than for patients receiving placebo (71% and 77%, respectively, vs. 64%), including infections (44% and 40%, vs. 31%). The rates of serious adverse events were 4%, 10%, and 7% in the three groups, respectively. Two nonmelanoma skin cancers and two major adverse cardiovascular events, including a fatal stroke, occurred in the higher-dose group. Baricitinib was associated with a small reduction in neutrophil levels and increases in serum creatinine and low-density lipoprotein cholesterol levels. CONCLUSIONS: In patients with rheumatoid arthritis and an inadequate response to biologic DMARDs, baricitinib at a daily dose of 4 mg was associated with clinical improvement at 12 weeks. (Funded by Eli Lilly and Incyte; ClinicalTrials.gov number, NCT01721044.).


Sujet(s)
Antirhumatismaux/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Azétidines/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Sulfonamides/usage thérapeutique , Sujet âgé , Antirhumatismaux/effets indésirables , Azétidines/effets indésirables , Femelle , Humains , Janus kinase 1/antagonistes et inhibiteurs , Kinase Janus-2/antagonistes et inhibiteurs , Modèles logistiques , Mâle , Adulte d'âge moyen , Inhibiteurs de protéines kinases/effets indésirables , Purines , Pyrazoles , Indice de gravité de la maladie , Sulfonamides/effets indésirables
10.
Nat Rev Rheumatol ; 12(1): 5-13, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26607389

RÉSUMÉ

Chemokines and chemokine receptors are involved in leukocyte recruitment and angiogenesis underlying the pathogenesis of rheumatoid arthritis (RA) and other inflammatory rheumatic diseases. Numerous chemokines, along with both conventional and atypical cell-surface chemokine receptors, are found in inflamed synovia. Preclinical studies carried out in animal models of arthritis involving agents targeting chemokines and chemokine receptors have yielded promising results. However, most human trials of treatment of RA with antibodies and synthetic compounds targeting chemokine signalling have failed to show clinical improvements. Chemokines can have overlapping actions, and their activities can be altered by chemical modification or proteolytic degradation. Effective targeting of chemokine pathways must take acount of these properties, and can also require high levels of receptor occupancy by therapeutic agents to prevent signalling. CCR1 is a promising target for chemokine-receptor blockade.


Sujet(s)
Polyarthrite rhumatoïde , Chimiokines/métabolisme , Facteurs immunologiques/usage thérapeutique , Animaux , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/thérapie , Chimiokines/effets des médicaments et des substances chimiques , Humains , Synovie/métabolisme
11.
Rheumatology (Oxford) ; 55(4): 745-54, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26705326

RÉSUMÉ

OBJECTIVES: Angiogenesis plays a critical role in SSc (scleroderma). The aim of this study was to examine the expression of growth-regulated protein-γ (Gro-γ/CXCL3), granulocyte chemotactic protein 2 (GCP-2/CXCL6) and their receptor CXCR2 in endothelial cells (ECs) isolated from SSc skin and determine whether these cells mount an angiogenic response towards pro-angiogenic chemokines. The downstream signalling pathways as well as the pro-angiogenic transcription factor inhibitor of DNA-binding protein 1 (Id-1) were also examined. METHODS: Skin biopsies were obtained from patients with dcSSc. ECs were isolated via magnetic positive selection. Angiogenesis was measured by EC chemotaxis assay. RESULTS: Gro-γ/CXCL3 and GCP-2/CXCL6 were minimally expressed in both skin types but elevated in SSc serum. Pro-angiogenic chemokine mRNA was greater in SSc ECs than in normal ECs. SSc ECs did not migrate to vascular endothelial growth factor (VEGF), Gro-γ/CXCL3, GCP-2/CXCL6 or CXCL16. The signalling pathways stimulated by these chemokines were also dysregulated. Id-1 mRNA in SSc ECs was lower compared with normal ECs, and overexpression of Id-1 in SSc ECs increased their ability to migrate towards VEGF and CXCL16. CONCLUSION: Our results show that SSc ECs are unable to respond to pro-angiogenic chemokines despite their increased expression in serum and ECs. This might be due to the differences in the signalling pathways activated by these chemokines in normal vs SSc ECs. In addition, the lower expression of Id-1 also decreases the angiogenic response. The inability of pro-angiogenic chemokines to promote EC migration provides an additional mechanism for the impaired angiogenesis that characterizes SSc.


Sujet(s)
Chimiokines/physiologie , Endothélium vasculaire/anatomopathologie , Néovascularisation pathologique/anatomopathologie , Sclérodermie systémique/anatomopathologie , Peau/vascularisation , Agents angiogéniques/pharmacologie , Études cas-témoins , Cellules cultivées , Chimiokines/biosynthèse , Chimiokines/pharmacologie , Chimiotaxie/effets des médicaments et des substances chimiques , Chimiotaxie/physiologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Endothélium vasculaire/métabolisme , Femelle , Humains , Protéine d'inhibition de la différenciation de type 1/physiologie , Mâle , Adulte d'âge moyen , Néovascularisation pathologique/métabolisme , Récepteurs à l'interleukine-8B/métabolisme , Sclérodermie systémique/métabolisme , Transduction du signal/physiologie , 12-Myristate-13-acétate de phorbol/analogues et dérivés , 12-Myristate-13-acétate de phorbol/pharmacologie
12.
J Invest Dermatol ; 135(12): 3153-3162, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26288351

RÉSUMÉ

The pathogenesis of scleroderma (SSc) includes components of autoimmunity, vascular dysfunction, and accumulation of extracellular matrix. 8-isoprostane, an oxidized lipid created by oxidative stress, activates the thromboxane A2 receptor (TXAR) and the Rho-associated kinase (ROCK) pathway. In this study, we determined whether the TXAR was activated by 8-isoprostane in SSc endothelial cells (ECs) and whether this pathway inhibited VEGF-induced angiogenesis. Elevated 8-isoprostane was observed in plasma and conditioned media from SSc patients. SSc-conditioned media inhibited EC tube formation, whereas addition of vitamin E, by reducing 8-isoprostane, increased tube formation. VEGF did not induce angiogenesis in SSc ECs, but vitamin E or TXAR inhibition restored its effect. The expression of TXAR, RhoA, and ROCK1/2 was elevated in SSc ECs. ROCK activity and 8-isoprostane-induced ROCK activation were significantly higher in SSc ECs, whereas VEGF had no effect. The hyper-activation of the TXAR leads to inhibition of VEGF-induced angiogenesis, as inhibition of the TXAR pathway results in a blockade of 8-isoprostane-induced ROCK activation and restoration of VEGF activity. These results suggest that the TXAR pathway has a crucial role in angiogenesis and that 8-isoprostane is not just a by-product of oxidative stress but also has a significant role in the impaired angiogenesis that characterizes SSc.


Sujet(s)
Dinoprost/analogues et dérivés , Néovascularisation physiologique , Récepteurs du thromboxane 2 et prostaglandine H2/physiologie , Sclérodermie systémique/physiopathologie , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Adulte , Collagène , Dinoprost/pharmacologie , Association médicamenteuse , Cellules endothéliales/physiologie , Femelle , Focal adhesion protein-tyrosine kinases/métabolisme , Humains , Laminine , Mâle , Adulte d'âge moyen , Stress oxydatif , Phosphorylation , Protéoglycanes , Récepteurs du thromboxane 2 et prostaglandine H2/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/pharmacologie , rho-Associated Kinases/physiologie
13.
J Biomed Nanotechnol ; 11(8): 1431-41, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26295143

RÉSUMÉ

Generation 5 (G5) poly(amidoamide) (PAMAM) dendrimers are synthetic polymers that have been broadly applied as drug delivery carriers. Methotrexate (MTX), an anti-folate metabolite, has been successfully used as an anti-inflammatory drug to treat rheumatoid arthritis (RA) in the clinic. In this study, we examine the therapeutic efficacy of G5 PAMAM dendrimer methotrexate conjugates (G5-MTX) that also have folic acid (FA) conjugated to the G5-MTX (G5-FA-MTX) to target inflammation-activated folate receptors overexpressing macrophages. These cells are thought to play an important role in the development of RA. With G5 serving as a control, the in vitro binding affinities of G5-FA-MTX and G5-MTX to activated macrophages were assessed in RAW264.7, NR8383 and primary rat peritoneal macrophages. The results indicated that the binding of either conjugate to macrophages was concentration- and temperature-dependent and could be blocked by the presence of 6.25 mM free FA (p < 0.005). The preventive effects of G5-MTX and G5-FA-MTX conjugates on the development of arthritis were explored on an adjuvant-induced inflammatory arthritis model and had similar preventive effects in inflammatory arthritis at a MTX equivalent dose of 4.95 µmol/kg. These studies indicated that when multiples of MTX are conjugated on dendritic polymers, they specifically bind to folate receptor overexpressing macrophages and have comparable anti-inflammatory effects to folate targeted MTX conjugated polymers.


Sujet(s)
Arthrite/traitement médicamenteux , Arthrite/métabolisme , Dendrimères/composition chimique , Acide folique/pharmacocinétique , Méthotrexate/administration et posologie , Nanocapsules/composition chimique , Animaux , Lignée cellulaire , Diffusion , Femelle , Récepteurs des folates à ancre GPI/métabolisme , Acide folique/composition chimique , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Méthotrexate/composition chimique , Nanocapsules/ultrastructure , Nanoconjugués/composition chimique , Nanoconjugués/ultrastructure , Rats , Rats de lignée LEW , Résultat thérapeutique
14.
Neuropsychopharmacology ; 40(3): 554-65, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25139063

RÉSUMÉ

Evidence in animal models suggests IL-1 family cytokines interact with central endogenous opioid neurotransmitter systems, inducing or perpetuating pathological states such as persistent pain syndromes, depression, substance use disorders, and their comorbidity. Understanding these interactions in humans is particularly relevant to understanding pathological states wherein this neurotransmitter system is implicated (ie, persistent pain, mood disorders, substance use disorders, etc). Here, we examined relationships between IL-1ß, IL-1ra, and functional measures of the endogenous opioid system in 34 healthy volunteers, in the absence and presence of a standardized sustained muscular pain challenge, a psychophysical challenge with emotionally and physically stressful components. Mu-opioid receptor availability in vivo was examined with [(11)C]carfentanil positron emission tomography (PET) scanning. Sex and neuroticism impacted IL-1 family cytokines; higher baseline IL-1ß and IL-1ra was identified in females with lower neuroticism. Higher baseline IL-1ß was also associated with reduced µ-opioid receptor availability (amygdala) and greater pain sensitivity. The pain challenge increased IL-1ß in females with high neuroticism. Strong associations between IL-1ra (an anti-nociceptive cytokine) and µ-opioid receptor activation (VP/NAcc) were identified during the pain challenge and the resulting analgesic effect of µ-opioid receptor activation was moderated by changes in IL-1ß whereby volunteers with greater pain induced increase in IL-1ß experienced less endogenous opioid analgesia. This study demonstrates the presence of relationships between inflammatory factors and a specific central neurotransmitter system and circuitry, of relevance to understanding interindividual variations in regulation of responses to pain and other physical and emotional stressors.


Sujet(s)
Antagoniste du récepteur à l'interleukine-1/sang , Interleukine-1 bêta/sang , Agents neuromédiateurs/métabolisme , Récepteur mu/métabolisme , Amygdale (système limbique)/métabolisme , Troubles anxieux/métabolisme , Radio-isotopes du carbone/métabolisme , Femelle , Fentanyl/analogues et dérivés , Fentanyl/métabolisme , Neuroimagerie fonctionnelle , Humains , Mâle , Neuroticisme , Douleur/métabolisme , Mesure de la douleur , Tomographie par émission de positons , Caractères sexuels
15.
Ann Rheum Dis ; 74(7): 1459-66, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-24665114

RÉSUMÉ

OBJECTIVES: Angiogenesis contributes to the pathogenesis of rheumatoid arthritis. Fucosyltransferases (Futs) are involved in angiogenesis and tumour growth. Here, we examined the role of Fut1 in angiogenesis and K/BxN serum transfer arthritis. METHODS: We examined Fut1 expression in human dermal microvascular endothelial cells (HMVECs) by quantitative PCR. We performed a number of angiogenesis assays to determine the role of Fut1 using HMVECs, Fut1 null (Fut1(-/-)), and wild type (wt) endothelial cells (ECs) and mice. K/BxN serum transfer arthritis was performed to determine the contribution of Fut1-mediated angiogenesis in Fut1(-/-) and wt mice. A static adhesion assay was implemented with RAW264.7 (mouse macrophage cell line) and mouse ECs. Quantitative PCR, immunofluorescence and flow cytometry were performed with Fut1(-/-) and wt ECs for adhesion molecule expression. RESULTS: Tumour necrosis factor-α induced Fut1 mRNA and protein expression in HMVECs. HMVECs transfected with Fut1 antisense oligodeoxynucleotide and Fut1(-/-) ECs formed significantly fewer tubes on Matrigel. Fut1(-/-) mice had reduced angiogenesis in Matrigel plug and sponge granuloma angiogenesis assays compared with wt mice. Fut1(-/-) mice were resistant to K/BxN serum transfer arthritis and had decreased angiogenesis and leucocyte ingress into inflamed joints. Adhesion of RAW264.7 cells to wt mouse ECs was significantly reduced when Fut1 was lacking. Fut1(-/-) ECs had decreased intercellular adhesion molecule-1 (ICAM-1) expression at mRNA and protein levels compared with wt ECs. ICAM-1 was also decreased in Fut1(-/-) arthritic ankle cryosections compared with wt ankles. CONCLUSIONS: Fut1 plays an important role in regulating angiogenesis and ICAM-1 expression in inflammatory arthritis.


Sujet(s)
Arthrite expérimentale/métabolisme , Arthrite expérimentale/physiopathologie , Fucosyltransferases/physiologie , Molécule-1 d'adhérence intercellulaire/métabolisme , Néovascularisation pathologique/physiopathologie , Animaux , Arthrite expérimentale/anatomopathologie , Adhérence cellulaire/physiologie , Lignée cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Endothélium vasculaire/anatomopathologie , Fucosyltransferases/déficit , Fucosyltransferases/génétique , Humains , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/anatomopathologie , Souris , Souris knockout , ARN messager/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie ,
17.
Arthritis Res Ther ; 16(5): 411, 2014.
Article de Anglais | MEDLINE | ID: mdl-25123250

RÉSUMÉ

INTRODUCTION: Systemic sclerosis (SSc) is a connective tissue disease characterized by fibrosis of the skin and organs. Increase in oxidative stress and platelet-derived growth factor receptor (PDGFR) activation promote collagen I (Col I) production, leading to fibrosis in SSc. Lipoic acid (LA) and its active metabolite dihydrolipoic acid (DHLA) are naturally occurring thiols that act as cofactors and antioxidants, and are produced by lipoic acid synthetase (LIAS). The goal of this study was to examine whether LA and LIAS was deficient in SSc patients and determine the effect of DHLA on the phenotype of SSc dermal fibroblasts. N-acetylcysteine (NAC), a commonly used thiol antioxidant, was included as a comparison. METHODS: Dermal fibroblasts were isolated from healthy subjects and patients with diffuse cutaneous SSc. Matrix metalloproteinase (MMPs), tissue inhibitors of MMPs (TIMP), plasminogen activator inhibitor-1 (PAI-1) and LIAS were measured by ELISA. The expression of Col I was measured by immunofluorescence, hydroxyproline assay, and quantitative PCR. PDGFR phosphorylation and α-smooth muscle actin (α-SMA) was measured by Western blotting. Student's t-tests were performed for statistical analysis and p-values of less than 0.05 with two-tailed analysis were considered statistically significant. RESULTS: The expression of LA and LIAS in SSc dermal fibroblasts was lower than normal fibroblasts, however LIAS was significantly higher in SSc plasma and appeared to be released from monocytes. DHLA lowered cellular oxidative stress, and decreased PDGFR phosphorylation, Col I, PAI-1, and α-SMA expression in SSc dermal fibroblasts. It also restored the activities of phosphatases that inactivated the PDGFR. SSc fibroblasts produced lower levels of MMP-1 and 3, and DHLA increased them. In contrast, TIMP-1 levels were higher in SSc but DHLA had minimal effect. Both DHLA and NAC increased MMP-1 activity when SSc cells were stimulated with PDGF. In general, DHLA showed better efficacy than NAC in most cases. CONCLUSIONS: DHLA not only acts as an antioxidant but also an antifibrotic since it has the ability to reverse the profibrotic phenotype of SSc dermal fibroblasts. Our study suggests that thiol antioxidants, including NAC and LA/DHLA, could be beneficial for patients with SSc.


Sujet(s)
Derme/métabolisme , Fibroblastes/métabolisme , Sclérodermie diffuse/métabolisme , Acide lipoïque/métabolisme , Adulte , Technique de Western , Cellules cultivées , Collagène de type I/génétique , Collagène de type I/métabolisme , Derme/anatomopathologie , Femelle , Expression des gènes , Humains , Mâle , Matrix metalloproteinases/métabolisme , Adulte d'âge moyen , Stress oxydatif , Phosphorylation , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Receptor-Like Protein Tyrosine Phosphatases, Class 3/génétique , Receptor-Like Protein Tyrosine Phosphatases, Class 3/métabolisme , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , RT-PCR , Sclérodermie diffuse/génétique , Sclérodermie diffuse/anatomopathologie , Sulfurtransferases/sang , Sulfurtransferases/métabolisme , Acide lipoïque/analogues et dérivés , Inhibiteur tissulaire des métalloprotéinases/métabolisme
19.
Arthritis Rheumatol ; 66(10): 2716-27, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24943990

RÉSUMÉ

OBJECTIVE: To examine whether the citrullinated chemokines epithelial neutrophil-activating peptide 78 (ENA-78)/CXCL5, macrophage inflammatory protein 1α/CCL3, and monocyte chemotactic protein 1/CCL2 are detected in the biologic fluid of patients with rheumatoid arthritis (RA), and if so, to determine the biologic activities of these chemokines. METHODS: Recombinant human chemokines were citrullinated by peptidylarginine deiminase. Enzyme-linked immunosorbent assays were performed to measure the concentrations of citrullinated chemokines in sera from patients with rheumatoid arthritis (RA) and normal individuals and in synovial fluid from patients with RA, patients with osteoarthritis (OA), and patients with other inflammatory rheumatic diseases. The correlation between the citrullinated chemokine levels and clinical data was analyzed. Monocyte and neutrophil chemotaxis assays were performed, and native (noncitrullinated) or citrullinated ENA-78/CXCL5 was injected into mouse knees to evaluate the biologic activities of these chemokines. RESULTS: The concentration of citrullinated ENA-78/CXCL5 was significantly higher in RA sera and SF than in normal sera and in SF from patients with other rheumatic diseases including OA. In RA SF, a strong correlation between the amount of citrullinated ENA-78/CXCL5 and the C-reactive protein level or the erythrocyte sedimentation rate was observed. Citrullinated ENA-78/CXCL5 induced monocyte chemotaxis via CXCR1 and CXCR2, while noncitrullinated ENA-78/CXCL5 did not. In a mouse model of inflammatory arthritis, citrullinated ENA-78/CXCL5 induced more severe inflammation and recruited more monocytes than did noncitrullinated ENA-78/CXCL5. CONCLUSION: Citrullinated ENA-78/CXCL5 is highly correlated with RA disease activity and, unlike noncitrullinated ENA-78/CXCL5, recruits monocytes. These results indicate that citrullinated ENA-78/CXCL5 may exert previously unrecognized inflammatory properties in RA by recruiting monocytes to inflamed joint tissue.


Sujet(s)
Polyarthrite rhumatoïde/métabolisme , Chimiokine CXCL5/métabolisme , Monocytes/métabolisme , Synovie/métabolisme , Animaux , Chimiokine CCL2/métabolisme , Chimiokine CCL3/métabolisme , Femelle , Humains , Articulation du genou/métabolisme , Mâle , Souris , Adulte d'âge moyen , Arthrose/métabolisme
20.
Arthritis Res Ther ; 16(2): R102, 2014 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-24762050

RÉSUMÉ

INTRODUCTION: Our objective was to examine the role of the janus-activated kinase (JAK) pathway in the modulation of tumor necrosis factor-α (TNF)-induced-IL-18 bioactivity by reduction of caspase-1 function. METHODS: Caspase-1 expression in rheumatoid arthritis (RA) synovial fibroblasts treated with TNF was assessed by qRT-PCR and Western blot. Interleukin (IL)-18 was assessed by enzyme-linked immunosorbent assay (ELISA) in cell lysates and conditioned media and detected by immunofluorescence (IF) staining in RA synovial fibroblasts. The critical pathways for TNF-induced caspase-1 expression were determined by using chemical inhibitors of signaling followed by TNF stimulation. IL-18 bioactivity was assessed using human myelomonocytic KG-1 cells. RESULTS: TNF induced RA synovial fibroblast caspase-1 expression at the protein level in a time-dependant manner (P < 0.05). Blocking the JAK pathway reduced TNF-induced-caspase-1 expression at the transcriptional and protein levels by approximately 60% and 40%, respectively (P < 0.05). Blocking the JAK pathway reduced TNF-induced-caspase-1 expression at the transcriptional, protein, and activity levels by approximately 60%, 40%, and 53%, respectively (P < 0.05). We then confirmed by IF that TNF-induced IL-18 and investigated roles of the ERK1/2 and JAK pathways. Blocking the JAK pathway, TNF induced intracytoplasmic granular IL-18 expression suggesting a defect of caspase-1. Finally, blocking the JAK pathway, we observed a reduction of IL-18 bioactivity by 52% in RA synovial fibroblasts (P < 0.05). CONCLUSIONS: These results provide a new way to regulate TNF-induced-IL-18 bioactivity by blocking capase-1. These data present a novel role for JAK inhibition in RA patients and emphasize JAK inhibition use as a new therapeutic option in RA management.


Sujet(s)
Polyarthrite rhumatoïde/immunologie , Caspase-1/immunologie , Interleukine-18/immunologie , Janus kinases/immunologie , Facteur de nécrose tumorale alpha/immunologie , Polyarthrite rhumatoïde/métabolisme , Technique de Western , Cellules cultivées , Test ELISA , Fibroblastes/immunologie , Fibroblastes/métabolisme , Technique d'immunofluorescence , Humains , Interleukine-18/métabolisme , Janus kinases/métabolisme , Réaction de polymérisation en chaine en temps réel , Transduction du signal/immunologie , Membrane synoviale/immunologie , Membrane synoviale/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...