Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
2.
Blood Adv ; 8(10): 2433-2441, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38330179

RÉSUMÉ

ABSTRACT: Pyruvate kinase (PK) deficiency is a rare, hereditary disease characterized by chronic hemolytic anemia. Iron overload is a common complication regardless of age, genotype, or transfusion history. Mitapivat, an oral, allosteric PK activator, improves anemia and hemolysis in adult patients with PK deficiency. Mitapivat's impact on iron overload and ineffective erythropoiesis was evaluated in adults with PK deficiency who were not regularly transfused in the phase 3 ACTIVATE trial and long-term extension (LTE) (#NCT03548220/#NCT03853798). Patients in the LTE received mitapivat throughout ACTIVATE/LTE (baseline to week 96; mitapivat-to-mitapivat [M/M] arm) or switched from placebo (baseline to week 24) to mitapivat (week 24 to week 96; placebo-to-mitapivat [P/M] arm). Changes from baseline in markers of iron overload and erythropoiesis were assessed to week 96. Improvements in hepcidin (mean, 4770.0 ng/L; 95% confidence interval [CI], -1532.3 to 11 072.3), erythroferrone (mean, -9834.9 ng/L; 95% CI, -14 328.4 to -5341.3), soluble transferrin receptor (mean, -56.0 nmol/L; 95% CI, -84.8 to -27.2), and erythropoietin (mean, -32.85 IU/L; 95% CI, -54.65 to -11.06) were observed in the M/M arm (n = 40) from baseline to week 24, sustained to week 96. No improvements were observed in the P/M arm (n = 40) to week 24; however, upon transitioning to mitapivat, improvements similar to those observed in the M/M arm were seen. Mean changes from baseline in liver iron concentration by magnetic resonance imaging at week 96 in the M/M arm and the P/M arm were -2.0 mg Fe/g dry weight (dw; 95% CI, -4.8 to -0.8) and -1.8 mg Fe/g dw (95% CI, -4.4 to 0.80), respectively. Mitapivat is the first disease-modifying pharmacotherapy shown to have beneficial effects on iron overload and ineffective erythropoiesis in patients with PK deficiency. This trial was registered at www.ClinicalTrials.gov as #NCT03548220 (ACTIVATE) and #NCT03853798 (LTE).


Sujet(s)
Anémie hémolytique congénitale non sphérocytaire , Érythropoïèse , Surcharge en fer , Pyruvate kinase , Erreurs innées du métabolisme du pyruvate , Humains , Surcharge en fer/étiologie , Surcharge en fer/traitement médicamenteux , Érythropoïèse/effets des médicaments et des substances chimiques , Adulte , Pyruvate kinase/déficit , Mâle , Femelle , Adulte d'âge moyen , Jeune adulte , Alanine/usage thérapeutique , Alanine/analogues et dérivés , Pipérazines , Quinoléines
3.
Int J Mol Sci ; 24(9)2023 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-37175706

RÉSUMÉ

The purpose of this study was to examine whether myeloid dendritic cells (mDCs) from patients with multiple sclerosis (MS) and healthy controls (HCs) become similarly tolerogenic when exposed to IL-27 as this may represent a potential mechanism of autoimmune dysregulation. Our study focused on natural mDCs that were isolated from HCs and MS patient peripheral blood mononuclear cells (PBMCs). After a 24-h treatment with IL-27 ± lipopolysaccharide (LPS), the mDCs were either harvested to identify IL-27-regulated gene expression or co-cultured with naive T-cells to measure how the treated DC affected T-cell proliferation and cytokine secretion. mDCs isolated from HCs but not untreated MS patients became functionally tolerogenic after IL-27 treatment. Although IL-27 induced both HC and untreated MS mDCs to produce similar amounts of IL-10, the tolerogenic HC mDCs expressed PD-L2, IDO1, and SOCS1, while the non-tolerogenic untreated MS mDCs expressed IDO1 and IL-6R. Cytokine and RNA analyses identified two signature blocks: the first identified genes associated with mDC tolerizing responses to IL-27, while the second was associated with the presence of MS. In contrast to mDCs from untreated MS patients, mDCs from HCs and IFNb-treated MS patients became tolerogenic in response to IL-27. The genes differentially expressed in the different donor IL-27-treated mDCs may contain targets that regulate mDC tolerogenic responses.


Sujet(s)
Interleukine-27 , Sclérose en plaques , Humains , Cellules cultivées , Cytokines/métabolisme , Cellules dendritiques , Interleukine-27/métabolisme , Agranulocytes/métabolisme , Sclérose en plaques/génétique , Sclérose en plaques/métabolisme , Lymphocytes T/métabolisme
4.
Lancet Haematol ; 9(10): e724-e732, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35988546

RÉSUMÉ

BACKGROUND: Mitapivat, an oral activator of pyruvate kinase (PK) in red blood cells (RBCs), has shown significant improvements in haemoglobin and haemolysis among patients with pyruvate kinase deficiency who were not receiving regular transfusions. We aimed to evaluate the efficacy and safety of mitapivat in adults with pyruvate kinase deficiency receiving regular transfusions. METHODS: ACTIVATE-T was an open-label, single-arm, phase 3 trial conducted in 20 centres across Europe, North America, and Asia. Eligible participants were adults (aged ≥18 years) with a clinical laboratory confirmation of pyruvate kinase deficiency receiving regular transfusions (at least six episodes in the previous year). Participants received oral mitapivat during a 16-week dose-optimisation period (5 mg, 20 mg, 50 mg twice daily) and 24-week fixed-dose period. The primary endpoint was a reduction in transfusion burden (≥33% reduction in number of RBC units transfused during the fixed-dose period, compared with the participant's individual historical transfusion burden, standardised to 24 weeks). Efficacy and safety were assessed in all participants who received at least one dose of mitapivat. This trial is registered with ClinicalTrials.gov, NCT03559699, and is complete. FINDINGS: Between June 26, 2018, and Feb 4, 2020, 27 participants (20 [74%] female and seven [26%] male; 20 [74%] White, three [11%] Asian, and four [15%] not reported) were enrolled and received at least one dose of mitapivat. Median duration of exposure to mitapivat was 40·3 weeks (IQR 40·0-41·3). A reduction in transfusion burden by at least 33% was found in ten (37%) participants (95% CI 19-58; p=0·0002). The most common treatment-emergent adverse events were increase in alanine aminotransferase (ten [37%] participants), headache (ten [37%]), increase in aspartate aminotransferase (five [19%]), fatigue (five [19%]), and nausea (five [19%]). Two grade 3 treatment-emergent adverse events were related to study treatment: joint swelling (one participant [4%]) and an increase in aspartate aminotransferase (one participant [4%]). Three participants had serious treatment-emergent adverse events, none related to the study treatment: increased blood triglycerides, ovarian cyst, and renal colic (each in one participant [4%]). No treatment-related deaths were observed. INTERPRETATION: Mitapivat represents a novel therapy that can reduce transfusion burden in some adults with pyruvate kinase deficiency receiving regular transfusions, and is the first disease-modifying agent approved in this disease. FUNDING: Agios Pharmaceuticals.


Sujet(s)
Hémoglobines , Pyruvate kinase , Adolescent , Adulte , Alanine transaminase , Anémie hémolytique congénitale non sphérocytaire , Aspartate aminotransferases , Femelle , Humains , Mâle , Préparations pharmaceutiques , Pipérazines , Pyruvate kinase/déficit , Erreurs innées du métabolisme du pyruvate , Quinoléines , Résultat thérapeutique , Triglycéride
5.
BMJ Neurol Open ; 4(1): e000240, 2022.
Article de Anglais | MEDLINE | ID: mdl-35720980

RÉSUMÉ

Objective: Slowly expanding lesions (SELs), a subgroup of chronic white matter lesions that gradually expand over time, have been shown to predict disability accumulation in primary progressive multiple sclerosis (MS) disease. However, the relationships between SELs, acute lesion activity (ALA), overall chronic lesion activity (CLA) and disability progression are not well understood. In this study, we examined the ASCEND phase III clinical trial, which compared natalizumab with placebo in secondary progressive MS (SPMS). Methods: Patients with complete imaging datasets between baseline and week 108 (N=600) were analysed for SEL prevalence (the number and volume of SELs), disability progression, ALA (assessed by gadolinium-enhancing lesions and new T2-hyperintense lesions) and CLA (assessed by T1-hypointense lesion volume increase within baseline T2-non-enhancing lesions identified as SELs and non-SELs). Results: CLA in both SELs and non-SELs was greater in patients with SPMS with confirmed disability progression than in those with no progression. In the complete absence of ALA at baseline and on study, SEL prevalence was significantly lower, while CLA within non-SELs remained associated with disability progression. Natalizumab decreased SEL prevalence and CLA in SELs and non-SELs compared with placebo. Conclusions: This study shows that CLA in patients with SPMS is decreased but persists in the absence of ALA and is associated with disability progression, highlighting the need for therapeutics targeting all mechanisms of CLA, including smouldering inflammation and neurodegeneration. Trial registration number: NCT01416181.

7.
N Engl J Med ; 386(15): 1432-1442, 2022 04 14.
Article de Anglais | MEDLINE | ID: mdl-35417638

RÉSUMÉ

BACKGROUND: Pyruvate kinase deficiency is a rare, hereditary, chronic condition that is associated with hemolytic anemia. In a phase 2 study, mitapivat, an oral, first-in-class activator of erythrocyte pyruvate kinase, increased the hemoglobin level in patients with pyruvate kinase deficiency. METHODS: In this global, phase 3, randomized, placebo-controlled trial, we evaluated the efficacy and safety of mitapivat in adults with pyruvate kinase deficiency who were not receiving regular red-cell transfusions. The patients were assigned to receive either mitapivat (5 mg twice daily, with potential escalation to 20 or 50 mg twice daily) or placebo for 24 weeks. The primary end point was a hemoglobin response (an increase from baseline of ≥1.5 g per deciliter in the hemoglobin level) that was sustained at two or more scheduled assessments at weeks 16, 20, and 24. Secondary efficacy end points were the average change from baseline in the hemoglobin level, markers of hemolysis and hematopoiesis, and the change from baseline at week 24 in two pyruvate kinase deficiency-specific patient-reported outcome measures. RESULTS: Sixteen of the 40 patients (40%) in the mitapivat group had a hemoglobin response, as compared with none of the 40 patients in the placebo group (adjusted difference, 39.3 percentage points; 95% confidence interval, 24.1 to 54.6; two-sided P<0.001). Patients who received mitapivat had a greater response than those who received placebo with respect to each secondary end point, including the average change from baseline in the hemoglobin level. The most common adverse events were nausea (in 7 patients [18%] in the mitapivat group and 9 patients [23%] in the placebo group) and headache (in 6 patients [15%] and 13 patients [33%], respectively). Adverse events of grade 3 or higher occurred in 10 patients (25%) who received mitapivat and 5 patients (13%) who received placebo. CONCLUSIONS: In patients with pyruvate kinase deficiency, mitapivat significantly increased the hemoglobin level, decreased hemolysis, and improved patient-reported outcomes. No new safety signals were identified in the patients who received mitapivat. (Funded by Agios Pharmaceuticals; ACTIVATE ClinicalTrials.gov number, NCT03548220.).


Sujet(s)
Pipérazines , Pyruvate kinase , Quinoléines , Adulte , Anémie hémolytique congénitale non sphérocytaire/traitement médicamenteux , Méthode en double aveugle , Hémoglobines/analyse , Hémoglobines/effets des médicaments et des substances chimiques , Hémolyse/effets des médicaments et des substances chimiques , Humains , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Pyruvate kinase/déficit , Erreurs innées du métabolisme du pyruvate/traitement médicamenteux , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique
8.
J Clin Invest ; 132(10)2022 05 16.
Article de Anglais | MEDLINE | ID: mdl-35298438

RÉSUMÉ

A disequilibrium between immunosuppressive Tregs and inflammatory IL-17-producing Th17 cells is a hallmark of autoimmune diseases, including multiple sclerosis (MS). However, the molecular mechanisms underlying the Treg and Th17 imbalance in CNS autoimmunity remain largely unclear. Identifying the factors that drive this imbalance is of high clinical interest. Here, we report a major disease-promoting role for microRNA-92a (miR-92a) in CNS autoimmunity. miR-92a was elevated in experimental autoimmune encephalomyelitis (EAE), and its loss attenuated EAE. Mechanistically, miR-92a mediated EAE susceptibility in a T cell-intrinsic manner by restricting Treg induction and suppressive capacity, while supporting Th17 responses, by directly repressing the transcription factor Foxo1. Although miR-92a did not directly alter Th1 differentiation, it appeared to indirectly promote Th1 cells by inhibiting Treg responses. Correspondingly, miR-92a inhibitor therapy ameliorated EAE by concomitantly boosting Treg responses and dampening inflammatory T cell responses. Analogous to our findings in mice, miR-92a was elevated in CD4+ T cells from patients with MS, and miR-92a silencing in patients' T cells promoted Treg development but limited Th17 differentiation. Together, our results demonstrate that miR-92a drives CNS autoimmunity by sustaining the Treg/Th17 imbalance and implicate miR-92a as a potential therapeutic target for MS.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , microARN , Sclérose en plaques , Lymphocytes T régulateurs , Animaux , Auto-immunité , Différenciation cellulaire , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Humains , Souris , Souris de lignée C57BL , microARN/génétique , Sclérose en plaques/génétique , Sclérose en plaques/immunologie , Lymphocytes auxiliaires Th1 , Cellules Th17
9.
Front Neurol ; 11: 606418, 2020.
Article de Anglais | MEDLINE | ID: mdl-33473248

RÉSUMÉ

Background: Pediatric multiple sclerosis (MS) is rare: only 1.5-5% of MS cases are diagnosed before 18 years of age, and data on disease-modifying therapies (DMTs) for pediatric MS are limited. The CONNECTED study assessed the long-term safety and efficacy of treatment with delayed-release dimethyl fumarate (DMF), an oral MS DMT, in pediatric patients with MS. Methods: CONNECTED is the 96-week extension to FOCUS, a 24-week phase 2 study of patients aged 13-17 years; participants received DMF 240 mg twice daily. Endpoints included (primary) incidence of adverse events (AEs), serious AEs, and DMF discontinuations due to an AE, and (secondary) T2 hyperintense lesion incidence by magnetic resonance imaging and annualized relapse rate (ARR). Results: Twenty participants [median (range) age, 17 (14-18) years; 65% female] who completed FOCUS enrolled into CONNECTED; 17 (85%) completed CONNECTED. Eighteen participants (90%) experienced AEs: the most frequent was flushing (25%). None experienced infections or fever related to low lymphocyte counts. Three participants experienced four serious AEs; none led to DMF discontinuation. Twelve of 17 participants (71%) had no new/newly enlarged T2 lesions from weeks 16-24, two (12%) had one, and one each (6%) had two, three, or five or more lesions [median (range), 0 (0-6)]. Over the full 120-week treatment period, ARR was 0.2, an 84.5% relative reduction (n = 20; 95% confidence interval: 66.8-92.8; p < 0.0001) vs. the year before DMF initiation. Conclusions: The long-term safety and efficacy observed in CONNECTED was consistent with adults, suggesting pediatric and adolescent patients with MS might benefit from DMF treatment.

10.
Cell Rep ; 28(13): 3353-3366.e5, 2019 09 24.
Article de Anglais | MEDLINE | ID: mdl-31553906

RÉSUMÉ

Smad7, a negative regulator of TGF-ß signaling, has been implicated in the pathogenesis and treatment of inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC). Here, we found that Smad7 mediates intestinal inflammation by limiting the PDL2/1-PD1 axis in dendritic cells (DCs) and CD4+T cells. Smad7 deficiency in DCs promotes TGF-ß responsiveness and the co-inhibitory molecules PDL2/1 on DCs, and it further imprints T cell-PD1 signaling to promote Treg differentiation. DC-specific Smad7 deletion mitigates DSS-induced colitis by inducing CD103+PDL2/1+DCs and Tregs. In addition, Smad7 deficiency in CD4+T cells promotes PD1 and PD1-induced Tregs in vitro. The transfer of Smad7-deficient CD4+T cells enhances Tregs in vivo and protects against T cell-mediated colitis. Furthermore, Smad7 antisense ameliorates DSS-induced UC, increasing TGF-ß and PDL2/1-PD1 signaling. Enhancing PD1 signaling directly via Fc-fused PDL2/1 is also beneficial. Our results identify how Smad7 mediates intestinal inflammation and leverages these pathways therapeutically, providing additional strategies for IBD intervention.


Sujet(s)
Auto-immunité/génétique , Inflammation/génétique , Intestins/anatomopathologie , Protéine Smad7/génétique , Humains , Transduction du signal
11.
Mult Scler ; 25(1): 63-71, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-29106333

RÉSUMÉ

BACKGROUND: Dimethyl fumarate (DMF) and its active metabolite monomethyl fumarate (MMF) effectively lead to reduction in disease relapses and active magnetic resonance imaging (MRI) lesions. DMF and MMF are known to be effective in modulating T- and B-cell responses; however, their effect on the phenotype and function of human myeloid dendritic cells (mDCs) is not fully understood. OBJECTIVE: To investigate the role of MMF on human mDCs maturation and function. METHODS: mDCs from healthy controls were isolated and cultured in vitro with MMF. The effect of MMF on mDC gene expression was determined by polymerase chain reaction (PCR) array after in vitro MMF treatment. The ability of mDCs to activate T cells was assessed by in vitro co-culture system. mDCs from DMF-treated multiple sclerosis (MS) patients were analyzed by flow cytometry and PCR. RESULTS: MMF treatment induced a less mature phenotype of mDCs with reduced expression of major histocompatibility complex class II (MHC-II), co-stimulatory molecules CD86, CD40, CD83, and expression of nuclear factor κB (NF-κB) subunits RELA and RELB. mDCs from DMF-treated MS patients also showed the same immature phenotype. T cells co-cultured with MMF-treated mDCs showed reduced proliferation with decreased production of interferon gamma (IFN-γ), interleukin-17 (IL-17), and granulocyte-macrophage colony-stimulating factor (GM-CSF) compared to untreated cells. CONCLUSION: We report that MMF can modulate immune response by affecting human mDC function.


Sujet(s)
Cellules dendritiques/effets des médicaments et des substances chimiques , Fumarate de diméthyle/pharmacologie , Fumarates/pharmacologie , Facteurs immunologiques/pharmacologie , Sclérose en plaques récurrente-rémittente/sang , Sclérose en plaques récurrente-rémittente/traitement médicamenteux , Cellules myéloïdes/effets des médicaments et des substances chimiques , Lymphocytes T/effets des médicaments et des substances chimiques , Humains
12.
Muscle Nerve ; 58(2): 261-269, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-29466830

RÉSUMÉ

INTRODUCTION: Amyotrophic lateral sclerosis (ALS) is a debilitating neurologic disorder with poor survival rates and no clear biomarkers for disease diagnosis and prognosis. METHODS: We compared serum microRNA (miRNA) expression from patients with ALS with healthy controls and patients with multiple sclerosis and Alzheimer disease. We also correlated miRNA expression in cross-sectional and longitudinal cohorts of ALS patients with clinical parameters. RESULTS: We identified 7 miRNAs (miR-192-5p, miR-192-3p, miR-1, miR-133a-3p, miR-133b, miR-144-5p, miR-19a-3p) that were upregulated and 6 miRNAs (miR-320c, miR-320a, let-7d-3p, miR-425-5p, miR-320b, miR-139-5p) that were downregulated in patients with ALS compared with healthy controls, patients with Alzheimer disease, and patients with multiple sclerosis. Changes in 4 miRNAs (miR-136-3p, miR-30b-5p, miR-331-3p, miR-496) correlated positively and change in 1 miRNA (miR-2110) correlated negatively with changes in clinical parameters in longitudinal analysis. DISCUSSION: Our findings identified serum miRNAs that can serve as biomarkers for ALS diagnosis and progression. Muscle Nerve 58: 261-269, 2018.


Sujet(s)
Sclérose latérale amyotrophique/sang , Sclérose latérale amyotrophique/physiopathologie , microARN/sang , Adulte , Maladie d'Alzheimer/sang , Maladie d'Alzheimer/physiopathologie , Marqueurs biologiques/sang , Études de cohortes , Études transversales , Évolution de la maladie , Femelle , Régulation de l'expression des gènes , Humains , Études longitudinales , Mâle , Adulte d'âge moyen , Sclérose en plaques/sang , Sclérose en plaques/physiopathologie
13.
Methods Mol Biol ; 1585: 127-140, 2017.
Article de Anglais | MEDLINE | ID: mdl-28477192

RÉSUMÉ

IL-9-producing Th9 cells are a novel subset of T helper cells that develop independently of other T helper subsets. Th9 cells have been implicated in the pathogenesis of allergic asthma and autoimmunity, while also serving as critical effector T cells in mediating antitumor immune responses. Concomitant presence of TGF-ß and IL-4 lead to the differentiation of naïve CD4+ T cells towards the Th9 phenotype. In addition, several cytokines, including IL-1ß, IL-2, IL-25, and IL-33, further amplify Th9 responses. Negative regulators of Th9 cells include other cytokines such as IFN-γ, IL-23, and IL-27. Here, we describe a detailed protocol for the analysis of STAT molecules involved in the differentiation of Th9 cells and Th9 inhibition by IL-27.


Sujet(s)
Cytométrie en flux/méthodes , Interleukine-9/métabolisme , Lymphocytes T CD4+/métabolisme , Interleukine-33/métabolisme , Interleukine-4/métabolisme , Interleukines/métabolisme , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-6/métabolisme , Lymphocytes T auxiliaires/métabolisme , Facteur de croissance transformant bêta/métabolisme
14.
Brain ; 139(Pt 7): 1939-57, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27246324

RÉSUMÉ

SEE WINGER AND ZAMVIL DOI101093/BRAIN/AWW121 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood-brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.


Sujet(s)
Astrocytes/immunologie , Antigènes CD3/immunologie , Encéphalomyélite auto-immune expérimentale/immunologie , Facteurs immunologiques/pharmacologie , Interleukine-10/immunologie , Muromonab-CD3/pharmacologie , Lymphocytes T régulateurs/immunologie , Administration par voie nasale , Animaux , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Femelle , Facteurs immunologiques/administration et posologie , Souris , Souris de lignée C57BL , Souris de lignée NOD , Muromonab-CD3/administration et posologie , Pneumonie à pneumocoques/immunologie
15.
J Immunol ; 189(11): 5277-83, 2012 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-23125412

RÉSUMÉ

IL-9-producing Th9 cells have been associated with autoimmune diseases, such as experimental autoimmune encephalitis. However, the factors that negatively regulate Th9 cells during autoimmune inflammation are unclear. In this article, we show that IFN-γ inhibits Th9 differentiation both in vitro and in vivo. This suppressive activity was dependent on the transcription factor STAT-1. In addition to its direct inhibitory effect on Th9 differentiation, IFN-γ suppressed Th9 cells through the induction of IL-27 from dendritic cells. In vitro, treatment of naive CD4(+) T cells with IL-27 suppressed the development of Th9 cells, which was partially dependent on the transcription factors STAT-1 and T-bet. Moreover, IL-27 treatment completely abrogated the encephalitogenicity of Th9 cells in the experimental autoimmune encephalomyelitis model. Thus, our results identify a previously unknown mechanism by which IFN-γ limits Th9-mediated autoimmune inflammation through dendritic cell modulation of IL-27.


Sujet(s)
Cellules dendritiques/effets des médicaments et des substances chimiques , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Inflammation/traitement médicamenteux , Interféron gamma/pharmacologie , Interleukine-9/immunologie , Interleukines/immunologie , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/immunologie , Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Encéphalomyélite auto-immune expérimentale/complications , Encéphalomyélite auto-immune expérimentale/immunologie , Expression des gènes/effets des médicaments et des substances chimiques , Inflammation/induit chimiquement , Inflammation/complications , Inflammation/immunologie , Interféron gamma/immunologie , Interleukine-17/biosynthèse , Interleukine-17/immunologie , Interleukine-9/biosynthèse , Interleukines/biosynthèse , Interleukines/pharmacologie , Souris , Souris de lignée C57BL , Souris knockout , Glycoprotéine MOG/pharmacologie , Fragments peptidiques/pharmacologie , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/immunologie , Lymphocytes T auxiliaires/cytologie , Lymphocytes T auxiliaires/immunologie
16.
PLoS One ; 7(6): e39576, 2012.
Article de Anglais | MEDLINE | ID: mdl-22768094

RÉSUMÉ

Inflammasomes are multi-protein complexes that control the production of pro-inflammatory cytokines such as IL-1ß. Inflammasomes play an important role in the control of immunity to tumors and infections, and also in autoimmune diseases, but the mechanisms controlling the activation of human inflammasomes are largely unknown. We found that human activated CD4+CD45RO+ memory T-cells specifically suppress P2X7R-mediated NLRP3 inflammasome activation, without affecting P2X7R-independent NLRP3 or NLRP1 inflammasome activation. The concomitant increase in pro-IL-1ß production induced by activated memory T-cells concealed this effect. Priming with IFNß decreased pro-IL-1ß production in addition to NLRP3 inflammasome inhibition and thus unmasked the inhibitory effect on NLRP3 inflammasome activation. IFNß suppresses NLRP3 inflammasome activation through an indirect mechanism involving decreased P2X7R signaling. The inhibition of pro-IL-1ß production and suppression of NLRP3 inflammasome activation by IFNß-primed human CD4+CD45RO+ memory T-cells is partly mediated by soluble FasL and is associated with down-regulated P2X7R mRNA expression and reduced response to ATP in monocytes. CD4+CD45RO+ memory T-cells from multiple sclerosis (MS) patients showed a reduced ability to suppress NLRP3 inflammasome activation, however their suppressive ability was recovered following in vivo treatment with IFNß. Thus, our data demonstrate that human P2X7R-mediated NLRP3 inflammasome activation is regulated by activated CD4+CD45RO+ memory T cells, and provide new information on the mechanisms mediating the therapeutic effects of IFNß in MS.


Sujet(s)
Lymphocytes T CD4+/immunologie , Protéines de transport/métabolisme , Régulation négative , Mémoire immunologique , Inflammasomes/métabolisme , Activation des lymphocytes/immunologie , Récepteurs purinergiques P2X7/génétique , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Caspase-1/métabolisme , Cellules cultivées , Cross-priming/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Ligand de Fas/métabolisme , Humains , Mémoire immunologique/effets des médicaments et des substances chimiques , Interféron bêta/pharmacologie , Interleukine-10/métabolisme , Interleukine-1 bêta/métabolisme , Antigènes CD45/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Masse moléculaire , Monocytes/métabolisme , Sclérose en plaques/immunologie , Sclérose en plaques/anatomopathologie , Protéine-3 de la famille des NLR contenant un domaine pyrine , ARN messager/génétique , ARN messager/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Solubilité
17.
Immunol Cell Biol ; 90(8): 831-5, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22430249

RÉSUMÉ

Interleukin (IL)-27 exerts an anti-inflammatory effect on human and mice CD4(+) T cells by inducing IL-10-producing T regulatory 1 cells through induction of IL-21. However, the role of IL-27 and how it regulates IL-21 from human CD8(+) T cells is unclear. Here, we show that the IL-27 receptor is expressed on human CD8(+) T cells and stimulation of human naïve CD8(+) T cells in the presence of IL-27 leads to an increase in IL-21 and interferon (IFN)-γ production. IL-21 induction in IL-27-stimulated human CD8(+) T cells correlates specifically with expression of the transcription factor T-bet. IL-27 stimulation of naïve CD8(+) T cells induces a double-positive T-bet(+) IL-21(+) expressing CD8(+) T-cell population. Furthermore, IL-27 stimulation of human naïve CD8(+) T cells greatly increases expression of granzyme B. Antibody-mediated neutralization of IL-21 abrogates IL-27-induced granzyme B expression. Moreover, direct addition of IL-21 greatly amplifies granzyme B expression in human naïve CD8(+) T cells. Our findings identify IL-27-induced IL-21 as a key autocrine regulator of granzyme B expression in human CD8(+) T cells.


Sujet(s)
Communication autocrine , Lymphocytes T CD8+/enzymologie , Lymphocytes T CD8+/immunologie , Granzymes/biosynthèse , Interleukines/métabolisme , Interleukines/pharmacologie , Animaux , Lymphocytes T CD8+/cytologie , Prolifération cellulaire , Induction enzymatique/immunologie , Humains , Interféron gamma/métabolisme , Activation des lymphocytes/immunologie , Souris
18.
PLoS One ; 6(10): e26173, 2011.
Article de Anglais | MEDLINE | ID: mdl-22039443

RÉSUMÉ

Interleukin-27 (IL-27) is a cytokine with multiple roles in regulating the immune response, but its effect on human CD56(bright) and CD56(dim) NK cell subsets is unknown. NK cell subsets interact with other components of the immune system, leading to cytotoxicity or immunoregulation depending on stimulating factors. We found that IL-27 treatment results in increased IL-10 and IFN-γ expression, increased viability and decreased proliferation in both CD56(bright) and CD56(dim) NK cell subsets. More importantly, IL-27 treatment imparts regulatory activity to CD56(bright) NK cells, which mediates its suppressive function on T cells in a contact-dependent manner. There is growing evidence that CD56(bright) NK cell-mediated immunoregulation plays an important role in the control of autoimmunity. Thus, understanding the role of IL-27 in NK cell function has important implications for treatment of autoimmune disorders.


Sujet(s)
Interleukine-17/physiologie , Cellules tueuses naturelles/immunologie , Prolifération cellulaire , Humains , Interféron gamma/métabolisme , Interleukine-10/métabolisme , Cellules tueuses naturelles/cytologie , Réaction de polymérisation en chaine en temps réel
19.
J Immunol ; 187(5): 2213-21, 2011 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-21788439

RÉSUMÉ

IFN-γ-producing Th1 and IL-17-producing Th17 cells are the key participants in various autoimmune diseases, including multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). Although both of these T cell subsets are known to be regulated by specific transcription factors and cytokines, the role of microRNAs that control these two inflammatory T cell subsets and whether targeting microRNAs can have therapeutic effects are not known. In this study, we show that microRNA-155 (Mir-155) expression is elevated in CD4(+) T cells during EAE, and Mir-155(-/-) mice had a delayed course and reduced severity of disease and less inflammation in the CNS. The attenuation of EAE in Mir-155(-/-) mice was associated with a decrease in Th1 and Th17 responses in the CNS and peripheral lymphoid organs. The T cell-intrinsic function of Mir-155(-/-) was demonstrated by the resistance of Mir-155(-/-) CD4(+) T cell-repleted Rag-1(-/-) mice to EAE. Finally, we found that anti-Mir-155 treatment reduced clinical severity of EAE when given before and after the appearance of clinical symptoms. These findings demonstrate that Mir-155 confers susceptibility to EAE by affecting inflammatory T cell responses and identify Mir-155 as a new target for therapeutic intervention in multiple sclerosis.


Sujet(s)
Lymphocytes T CD4+/immunologie , Encéphalomyélite auto-immune expérimentale/génétique , microARN/génétique , Sous-populations de lymphocytes T/immunologie , Animaux , Encéphalomyélite auto-immune expérimentale/immunologie , Extinction de l'expression des gènes , Inflammation/génétique , Inflammation/immunologie , Souris , Souris de lignée C57BL , Souris knockout , microARN/immunologie , RT-PCR
20.
Inflamm Bowel Dis ; 14(8): 1033-40, 2008 Aug.
Article de Anglais | MEDLINE | ID: mdl-18383179

RÉSUMÉ

BACKGROUND: The functional link between mutations in NOD2 and Crohn's disease (CD) has not been entirely elucidated. The 1007fs mutation results in loss of NF-kappaB activation in response to muramyl dipeptide (MDP) but has also been linked to an increased IL-1beta processing and IL-12 release. METHODS: We investigated the basal and MDP-triggered mRNA expression and protein release for TNF-alpha, IL-10, IL-1beta, and IL-12p40 in peripheral blood monocytes from 40 CD patients and 15 healthy individuals with different NOD2 genotypes. RESULTS: Monocytes from individuals with 2 mutated NOD2 alleles (homozygous and compound-heterozygous individuals) displayed an impaired release of TNF-alpha and IL-10 but also of IL-1beta and IL-12p40 in response to MDP. In contrast to other NOD2 variants, the presence of at least 1 1007fs allele in double-mutated individuals completely abrogated NOD2 receptor function. Interestingly, monocytes from CD patients with 2 mutated NOD2 alleles displayed significantly higher basal levels of IL-12p40 in cell culture supernatants compared to wildtype CD patients and control individuals (P = 0.002 and P = 0.008, respectively). This was regardless of the IL23R genotype and was not mirrored by increased IL-12p40 plasma levels in these individuals. CONCLUSIONS: The CD-associated NOD2 variants lead, in a dose- and mutation-dependent manner, to an impaired release of TNF-alpha, IL-10, IL-1beta, and IL-12p40 in response to MDP. The finding of increased basal levels for IL-12p40-related cytokines in monocytes with 2 mutated NOD2 alleles is likely to set a new link between NOD2 mutations and the inflammatory mechanisms underlying CD.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Maladie de Crohn/génétique , Sous-unité p40 de l'interleukine-12/métabolisme , Monocytes/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Acétylmuramyl alanyl isoglutamine/pharmacologie , Études cas-témoins , Relation dose-effet des médicaments , Femelle , Génotype , Humains , Interleukine-10/métabolisme , Interleukine-1 bêta/métabolisme , Mâle , Monocytes/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...