Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
1.
RSC Adv ; 14(31): 22244-22252, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-39010923

RÉSUMÉ

The prospect of being able to efficiently inject large plasmids in insulin-producing beta cells is very attractive for diabetes research. However, conventional transfection methods suffer from high cytotoxicity or low transfection efficiency, which negatively affect their outcome. In contrast, nanostraw electroporation is a gentle method that can provide a high transfection efficiency while maintaining high cell viability. While nanostraw electroporation has gone through some method optimization in the past, such as tuning the pulse frequency, amplitude, and duration, the effect of other parameters has not been thoroughly investigated. Here, we demonstrate efficient transfection of clonal beta cells and investigate the effect of voltage at a fixed inter-electrode distance, cell density, and cargo solution conductivity on transfection efficiency. We used GFP-encoding DNA plasmids stained with an intercalating dye to enable immediate analysis and assessment of the electrophoretic transport of cargo. Moreover, we ran simulations to assess how cargo buffer conductivity impacts the transfection efficiency by affecting the voltage drop on the nanostraws and cell membrane during electroporation. Both experiments and simulations show that MilliQ water as the cargo buffer yields the best transfection efficiency. We also show that the cell density should be adjusted to maximize the number of cells interfacing the nanostraws and avoid cell stacking. Finally, we compared the transfection efficiency when using nanostraws and nanopores. Whereas the amount of GFP plasmids injected using nanostraws is larger than for nanopores, the outcome in terms of GFP fluorescence 48 h after transfection was worse than for nanopores. Moreover, when using nanostraws, fewer cells were found on the substrate 48 h after transfection compared to when using nanopores. This suggests that injecting substantial amounts of plasmids in cells can affect their proliferation and/or viability, and that nanopore electroporation, as a simpler method, is an interesting alternative to nanostraws in achieving efficient and gentle clonal beta cell transfection.

2.
Nat Commun ; 14(1): 8040, 2023 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-38086799

RÉSUMÉ

Epigenetic dysregulation may influence disease progression. Here we explore whether epigenetic alterations in human pancreatic islets impact insulin secretion and type 2 diabetes (T2D). In islets, 5,584 DNA methylation sites exhibit alterations in T2D cases versus controls and are associated with HbA1c in individuals not diagnosed with T2D. T2D-associated methylation changes are found in enhancers and regions bound by ß-cell-specific transcription factors and associated with reduced expression of e.g. CABLES1, FOXP1, GABRA2, GLR1A, RHOT1, and TBC1D4. We find RHOT1 (MIRO1) to be a key regulator of insulin secretion in human islets. Rhot1-deficiency in ß-cells leads to reduced insulin secretion, ATP/ADP ratio, mitochondrial mass, Ca2+, and respiration. Regulators of mitochondrial dynamics and metabolites, including L-proline, glycine, GABA, and carnitines, are altered in Rhot1-deficient ß-cells. Islets from diabetic GK rats present Rhot1-deficiency. Finally, RHOT1methylation in blood is associated with future T2D. Together, individuals with T2D exhibit epigenetic alterations linked to mitochondrial dysfunction in pancreatic islets.


Sujet(s)
Diabète de type 2 , Cellules à insuline , Ilots pancréatiques , Humains , Rats , Animaux , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Sécrétion d'insuline , Insuline/métabolisme , Méthylation de l'ADN , Ilots pancréatiques/métabolisme , Cellules à insuline/métabolisme , Facteurs de transcription/métabolisme , Épigenèse génétique , Mitochondries/génétique , Mitochondries/métabolisme , Protéines de répression/métabolisme , Facteurs de transcription Forkhead/métabolisme
3.
J Clin Invest ; 133(4)2023 02 15.
Article de Anglais | MEDLINE | ID: mdl-36656641

RÉSUMÉ

Type 2 diabetes (T2D) is caused by insufficient insulin secretion from pancreatic ß cells. To identify candidate genes contributing to T2D pathophysiology, we studied human pancreatic islets from approximately 300 individuals. We found 395 differentially expressed genes (DEGs) in islets from individuals with T2D, including, to our knowledge, novel (OPRD1, PAX5, TET1) and previously identified (CHL1, GLRA1, IAPP) candidates. A third of the identified expression changes in islets may predispose to diabetes, as expression of these genes associated with HbA1c in individuals not previously diagnosed with T2D. Most DEGs were expressed in human ß cells, based on single-cell RNA-Seq data. Additionally, DEGs displayed alterations in open chromatin and associated with T2D SNPs. Mouse KO strains demonstrated that the identified T2D-associated candidate genes regulate glucose homeostasis and body composition in vivo. Functional validation showed that mimicking T2D-associated changes for OPRD1, PAX5, and SLC2A2 impaired insulin secretion. Impairments in Pax5-overexpressing ß cells were due to severe mitochondrial dysfunction. Finally, we discovered PAX5 as a potential transcriptional regulator of many T2D-associated DEGs in human islets. Overall, we have identified molecular alterations in human pancreatic islets that contribute to ß cell dysfunction in T2D pathophysiology.


Sujet(s)
Diabète de type 2 , Cellules à insuline , Ilots pancréatiques , Humains , Souris , Animaux , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Sécrétion d'insuline/génétique , Insuline/génétique , Insuline/métabolisme , Ilots pancréatiques/métabolisme , Cellules à insuline/métabolisme , Mixed function oxygenases/métabolisme , Protéines proto-oncogènes/métabolisme , Protéine activatrice spécifique des lymphocytes B/métabolisme
4.
Ann Rheum Dis ; 81(8): 1151-1161, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35470161

RÉSUMÉ

OBJECTIVE: Neonatal lupus erythematosus (NLE) may develop after transplacental transfer of maternal autoantibodies with cardiac manifestations (congenital heart block, CHB) including atrioventricular block, atrial and ventricular arrhythmias, and cardiomyopathies. The association with anti-Ro/SSA antibodies is well established, but a recurrence rate of only 12%-16% despite persisting maternal autoantibodies suggests that additional factors are required for CHB development. Here, we identify fetal genetic variants conferring risk of CHB and elucidate their effects on cardiac function. METHODS: A genome-wide association study was performed in families with at least one case of CHB. Gene expression was analysed by microarrays, RNA sequencing and PCR and protein expression by western blot, immunohistochemistry, immunofluorescence and flow cytometry. Calcium regulation and connectivity were analysed in primary cardiomyocytes and cells induced from pleuripotent stem cells. Fetal heart performance was analysed by Doppler/echocardiography. RESULTS: We identified DNAJC6 as a novel fetal susceptibility gene, with decreased cardiac expression of DNAJC6 associated with the disease risk genotype. We further demonstrate that fetal cardiomyocytes deficient in auxilin, the protein encoded by DNAJC6, have abnormal connectivity and Ca2+ homoeostasis in culture, as well as decreased cell surface expression of the Cav1.3 calcium channel. Doppler echocardiography of auxilin-deficient fetal mice revealed cardiac NLE abnormalities in utero, including abnormal heart rhythm with atrial and ventricular ectopias, as well as a prolonged atrioventricular time intervals. CONCLUSIONS: Our study identifies auxilin as the first genetic susceptibility factor in NLE modulating cardiac function, opening new avenues for the development of screening and therapeutic strategies in CHB.


Sujet(s)
Bloc atrioventriculaire , Auxilines , Animaux , Anticorps antinucléaires , Bloc atrioventriculaire/génétique , Autoanticorps , Coeur foetal , Étude d'association pangénomique , Bloc cardiaque/congénital , Lupus érythémateux disséminé/congénital , Souris
5.
Sci Adv ; 8(17): eabn1823, 2022 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-35476434

RÉSUMÉ

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS), in which pathological T cells, likely autoimmune, play a key role. Despite its central importance, the autoantigen repertoire remains largely uncharacterized. Using a novel in vitro antigen delivery method combined with the Human Protein Atlas library, we screened for T cell autoreactivity against 63 CNS-expressed proteins. We identified four previously unreported autoantigens in MS: fatty acid-binding protein 7, prokineticin-2, reticulon-3, and synaptosomal-associated protein 91, which were verified to induce interferon-γ responses in MS in two cohorts. Autoreactive profiles were heterogeneous, and reactivity to several autoantigens was MS-selective. Autoreactive T cells were predominantly CD4+ and human leukocyte antigen-DR restricted. Mouse immunization induced antigen-specific responses and CNS leukocyte infiltration. This represents one of the largest systematic efforts to date in the search for MS autoantigens, demonstrates the heterogeneity of autoreactive profiles, and highlights promising targets for future diagnostic tools and immunomodulatory therapies in MS.

6.
FASEB J ; 35(8): e21828, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34325494

RÉSUMÉ

Since prenatal glucocorticoids (GC) excess increases the risk of metabolic dysfunctions in the offspring and its effect on ß-cell recovery capacity remains unknown we investigated these aspects in offspring from mice treated with dexamethasone (DEX) in the late pregnancy. Half of the pups were treated with streptozotocin (STZ) on the sixth postnatal day (PN). Functional and molecular analyses were performed in male offspring on PN25 and PN225. Prenatal DEX treatment resulted in low birth weight. At PN25, both the STZ-treated offspring developed hyperglycemia and had lower ß-cell mass, in parallel with higher α-cell mass and glucose intolerance, with no impact of prenatal DEX on such parameters. At PN225, the ß-cell mass was partially recovered in the STZ-treated mice, but they remained glucose-intolerant, irrespective of being insulin sensitive. Prenatal exposition to DEX predisposed adult offspring to sustained hyperglycemia and perturbed islet function (lower insulin and higher glucagon response to glucose) in parallel with exacerbated glucose intolerance. ß-cell-specific knockdown of the Hnf4α in mice from the DS group resulted in exacerbated glucose intolerance. We conclude that high GC exposure during the prenatal period exacerbates the metabolic dysfunctions in adult life of mice exposed to STZ early in life, resulting in a lesser ability to recover the islets' function over time. This study alerts to the importance of proper management of exogenous GCs during pregnancy and a healthy postnatal lifestyle since the combination of adverse factors during the prenatal and postnatal period accentuates the predisposition to metabolic disorders in adult life.


Sujet(s)
Dexaméthasone/toxicité , Glucocorticoïdes/toxicité , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/physiologie , Animaux , Animal génétiquement modifié , Animaux nouveau-nés , Dexaméthasone/administration et posologie , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glucocorticoïdes/administration et posologie , Hyperglycémie provoquée , Insuline/pharmacologie , Souris , Tumeurs expérimentales , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque , ARN messager/génétique , ARN messager/métabolisme
7.
Sci Immunol ; 5(52)2020 10 16.
Article de Anglais | MEDLINE | ID: mdl-33067381

RÉSUMÉ

Multiple sclerosis (MS) is a leading cause of incurable progressive disability in young adults caused by inflammation and neurodegeneration in the central nervous system (CNS). The capacity of microglia to clear tissue debris is essential for maintaining and restoring CNS homeostasis. This capacity diminishes with age, and age strongly associates with MS disease progression, although the underlying mechanisms are still largely elusive. Here, we demonstrate that the recovery from CNS inflammation in a murine model of MS is dependent on the ability of microglia to clear tissue debris. Microglia-specific deletion of the autophagy regulator Atg7, but not the canonical macroautophagy protein Ulk1, led to increased intracellular accumulation of phagocytosed myelin and progressive MS-like disease. This impairment correlated with a microglial phenotype previously associated with neurodegenerative pathologies. Moreover, Atg7-deficient microglia showed notable transcriptional and functional similarities to microglia from aged wild-type mice that were also unable to clear myelin and recover from disease. In contrast, induction of autophagy in aged mice using the disaccharide trehalose found in plants and fungi led to functional myelin clearance and disease remission. Our results demonstrate that a noncanonical form of autophagy in microglia is responsible for myelin degradation and clearance leading to recovery from MS-like disease and that boosting this process has a therapeutic potential for age-related neuroinflammatory conditions.


Sujet(s)
Protéine-7 associée à l'autophagie/déficit , Encéphalomyélite auto-immune expérimentale/immunologie , Microglie/immunologie , Sclérose en plaques/immunologie , Phagocytose/immunologie , Animaux , Autophagie/immunologie , Protéine-7 associée à l'autophagie/génétique , Homologue de la protéine-1 associée à l'autophagie/déficit , Homologue de la protéine-1 associée à l'autophagie/génétique , Encéphale/cytologie , Encéphale/immunologie , Encéphale/anatomopathologie , Cellules cultivées , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Femelle , Humains , Mâle , Souris , Souris knockout , Microglie/métabolisme , Sclérose en plaques/anatomopathologie , Gaine de myéline/métabolisme , Culture de cellules primaires , Moelle spinale/cytologie , Moelle spinale/immunologie , Moelle spinale/anatomopathologie
8.
Sci Rep ; 9(1): 11996, 2019 08 19.
Article de Anglais | MEDLINE | ID: mdl-31427643

RÉSUMÉ

Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system with prominent neurodegenerative components. The triggering and progression of MS is associated with transcriptional and epigenetic alterations in several tissues, including peripheral blood. The combined influence of transcriptional and epigenetic changes associated with MS has not been assessed in the same individuals. Here we generated paired transcriptomic (RNA-seq) and DNA methylation (Illumina 450 K array) profiles of CD4+ and CD8+ T cells (CD4, CD8), using clinically accessible blood from healthy donors and MS patients in the initial relapsing-remitting and subsequent secondary-progressive stage. By integrating the output of a differential expression test with a permutation-based non-parametric combination methodology, we identified 149 differentially expressed (DE) genes in both CD4 and CD8 cells collected from MS patients. Moreover, by leveraging the methylation-dependent regulation of gene expression, we identified the gene SH3YL1, which displayed significant correlated expression and methylation changes in MS patients. Importantly, silencing of SH3YL1 in primary human CD4 cells demonstrated its influence on T cell activation. Collectively, our strategy based on paired sampling of several cell-types provides a novel approach to increase sensitivity for identifying shared mechanisms altered in CD4 and CD8 cells of relevance in MS in small sized clinical materials.


Sujet(s)
Immunomodulation , Sclérose en plaques/étiologie , Sclérose en plaques/métabolisme , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Adulte , Biologie informatique/méthodes , Méthylation de l'ADN , Prise en charge de la maladie , Évolution de la maladie , Prédisposition aux maladies , Femelle , Analyse de profil d'expression de gènes , Humains , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Mâle , Adulte d'âge moyen , Sclérose en plaques/diagnostic , Indice de gravité de la maladie , Transcriptome
9.
MethodsX ; 6: 1635-1641, 2019.
Article de Anglais | MEDLINE | ID: mdl-31367530

RÉSUMÉ

Reliable and sensitive detection of antigen specific cells is essential in several fields of research, whether it concerns monitoring responses to infectious agents or exploring the auto-antigen repertoire in autoimmune diseases. Identification of these cells is however difficult, especially when the cells often are rare and methods not sensitive, specific or practical enough. We propose a novel method of processing antigens before stimulation of cells which consists of covalently binding protein antigen to superparamagnetic micro-beads and using denaturing washes to remove contaminants. Peripheral blood mononuclear cells (PBMCs) from healthy donors were stimulated using both cytomegalovirus and tetanus-diphtheria antigen-beads as well as non-antigenic protein-beads as negative control in an IFNγ FluoroSpot assay in order to detect Th1 and CD8+ responses. The responses toward the antigen beads were both antigen specific and sensitive, with a detection threshold of 1 IFNγ producing T-cell per 18,000 PBMCs. •Covalently binding antigen to paramagnetic beads allows for harsh denaturing washes without loss of antigen.•Microbeads are phagocytosed by antigen presenting cells, resulting in efficient uptake, processing and presentation of the antigens.•The method allows the usage of relatively impure starting antigen material and whole PBMC samples without high background levels in follow up cellular assays.

10.
EBioMedicine ; 43: 411-423, 2019 May.
Article de Anglais | MEDLINE | ID: mdl-31053557

RÉSUMÉ

BACKGROUND: Multiple Sclerosis (MS) is a chronic inflammatory disease and a leading cause of progressive neurological disability among young adults. DNA methylation, which intersects genes and environment to control cellular functions on a molecular level, may provide insights into MS pathogenesis. METHODS: We measured DNA methylation in CD4+ T cells (n = 31), CD8+ T cells (n = 28), CD14+ monocytes (n = 35) and CD19+ B cells (n = 27) from relapsing-remitting (RRMS), secondary progressive (SPMS) patients and healthy controls (HC) using Infinium HumanMethylation450 arrays. Monocyte (n = 25) and whole blood (n = 275) cohorts were used for validations. FINDINGS: B cells from MS patients displayed most significant differentially methylated positions (DMPs), followed by monocytes, while only few DMPs were detected in T cells. We implemented a non-parametric combination framework (omicsNPC) to increase discovery power by combining evidence from all four cell types. Identified shared DMPs co-localized at MS risk loci and clustered into distinct groups. Functional exploration of changes discriminating RRMS and SPMS from HC implicated lymphocyte signaling, T cell activation and migration. SPMS-specific changes, on the other hand, implicated myeloid cell functions and metabolism. Interestingly, neuronal and neurodegenerative genes and pathways were also specifically enriched in the SPMS cluster. INTERPRETATION: We utilized a statistical framework (omicsNPC) that combines multiple layers of evidence to identify DNA methylation changes that provide new insights into MS pathogenesis in general, and disease progression, in particular. FUND: This work was supported by the Swedish Research Council, Stockholm County Council, AstraZeneca, European Research Council, Karolinska Institutet and Margaretha af Ugglas Foundation.


Sujet(s)
Méthylation de l'ADN , Immunité , Sclérose en plaques/étiologie , Sclérose en plaques/métabolisme , Transduction du signal , Adulte , Sujet âgé , Sous-populations de lymphocytes B/immunologie , Sous-populations de lymphocytes B/métabolisme , Marqueurs biologiques , Ilots CpG , Évolution de la maladie , Prédisposition aux maladies , Femelle , Humains , Immunophénotypage , Mâle , Adulte d'âge moyen , Sclérose en plaques/imagerie diagnostique , Sclérose en plaques/anatomopathologie , Sclérose en plaques chronique progressive/diagnostic , Sclérose en plaques chronique progressive/étiologie , Sclérose en plaques chronique progressive/métabolisme , Sclérose en plaques récurrente-rémittente/diagnostic , Sclérose en plaques récurrente-rémittente/étiologie , Sclérose en plaques récurrente-rémittente/métabolisme , Locus de caractère quantitatif , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme
11.
J Autoimmun ; 102: 38-49, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31054941

RÉSUMÉ

Autoreactive CD4+ T-cells are believed to be a main driver of multiple sclerosis (MS). Myelin oligodendrocyte glycoprotein (MOG) is considered an autoantigen, yet doubted in recent years. The reason is in part due to low frequency and titers of MOG autoantibodies and the challenge to detect MOG-specific T-cells. In this study we aimed to analyze T-cell reactivity and frequency utilizing a novel method for detection of antigen-specific T-cells with bead-bound MOG as stimulant. Peripheral blood mononuclear cells (PBMCs) from natalizumab treated persons with MS (n = 52) and healthy controls (HCs) (n = 24) were analyzed by IFNγ/IL-22/IL-17A FluoroSpot. A higher number of IFNγ (P = 0.001), IL-22 (P = 0.003), IL-17A (P < 0.0001) as well as double and triple cytokine producing MOG-specific T-cells were detected in persons with MS compared to HCs. Of the patients, 46.2-59.6% displayed MOG-reactivity. Depletion of CD4+ T-cells or monocytes or blocking HLA-DR completely eliminated the MOG specific response. Anti-MOG antibodies did not correlate with T-cell MOG-responses. In conclusion, we present a sensitive method to detect circulating autoreactive CD4+ T-cells producing IFNγ, IL-22 or IL-17A using MOG as a model antigen. Further, we demonstrate that MOG-specific T-cells are present in approximately half of persons with MS.


Sujet(s)
Lymphocytes T CD4+/immunologie , Interféron gamma/biosynthèse , Interleukine-17/biosynthèse , Interleukines/biosynthèse , Sclérose en plaques/immunologie , Glycoprotéine MOG/immunologie , Adolescent , Adulte , Autoanticorps/sang , Autoanticorps/immunologie , Autoantigènes/immunologie , Femelle , Antigènes HLA-DR/métabolisme , Humains , Interféron gamma/immunologie , Interleukine-17/immunologie , Interleukines/immunologie , Mâle , Adulte d'âge moyen , Sclérose en plaques/traitement médicamenteux , Glycoprotéine MOG/génétique , Natalizumab/usage thérapeutique , Jeune adulte ,
12.
Cell ; 175(1): 85-100.e23, 2018 09 20.
Article de Anglais | MEDLINE | ID: mdl-30173916

RÉSUMÉ

Multiple sclerosis is an autoimmune disease that is caused by the interplay of genetic, particularly the HLA-DR15 haplotype, and environmental risk factors. How these etiologic factors contribute to generating an autoreactive CD4+ T cell repertoire is not clear. Here, we demonstrate that self-reactivity, defined as "autoproliferation" of peripheral Th1 cells, is elevated in patients carrying the HLA-DR15 haplotype. Autoproliferation is mediated by memory B cells in a HLA-DR-dependent manner. Depletion of B cells in vitro and therapeutically in vivo by anti-CD20 effectively reduces T cell autoproliferation. T cell receptor deep sequencing showed that in vitro autoproliferating T cells are enriched for brain-homing T cells. Using an unbiased epitope discovery approach, we identified RASGRP2 as target autoantigen that is expressed in the brain and B cells. These findings will be instrumental to address important questions regarding pathogenic B-T cell interactions in multiple sclerosis and possibly also to develop novel therapies.


Sujet(s)
Lymphocytes B/anatomopathologie , Sous-types sérologiques HLA-DR/immunologie , Sclérose en plaques/immunologie , Autoantigènes/immunologie , Maladies auto-immunes/immunologie , Maladies auto-immunes/physiopathologie , Lymphocytes B/métabolisme , Encéphale/anatomopathologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/physiologie , Facteurs d'échange de nucléotides guanyliques/métabolisme , Sous-types sérologiques HLA-DR/génétique , Humains , Sclérose en plaques/génétique , Sclérose en plaques/physiopathologie , Récepteurs aux antigènes des cellules T , Lymphocytes auxiliaires Th1/physiologie
13.
Nat Commun ; 9(1): 2397, 2018 06 19.
Article de Anglais | MEDLINE | ID: mdl-29921915

RÉSUMÉ

The human leukocyte antigen (HLA) haplotype DRB1*15:01 is the major risk factor for multiple sclerosis (MS). Here, we find that DRB1*15:01 is hypomethylated and predominantly expressed in monocytes among carriers of DRB1*15:01. A differentially methylated region (DMR) encompassing HLA-DRB1 exon 2 is particularly affected and displays methylation-sensitive regulatory properties in vitro. Causal inference and Mendelian randomization provide evidence that HLA variants mediate risk for MS via changes in the HLA-DRB1 DMR that modify HLA-DRB1 expression. Meta-analysis of 14,259 cases and 171,347 controls confirms that these variants confer risk from DRB1*15:01 and also identifies a protective variant (rs9267649, p < 3.32 × 10-8, odds ratio = 0.86) after conditioning for all MS-associated variants in the region. rs9267649 is associated with increased DNA methylation at the HLA-DRB1 DMR and reduced expression of HLA-DRB1, suggesting a modulation of the DRB1*15:01 effect. Our integrative approach provides insights into the molecular mechanisms of MS susceptibility and suggests putative therapeutic strategies targeting a methylation-mediated regulation of the major risk gene.


Sujet(s)
Méthylation de l'ADN , Prédisposition génétique à une maladie/génétique , Chaines HLA-DRB1/génétique , Sclérose en plaques/génétique , Polymorphisme de nucléotide simple , Adulte , Sujet âgé , Cellules cultivées , Études de cohortes , Femelle , Régulation de l'expression des gènes , Humains , Mâle , Méta-analyse comme sujet , Adulte d'âge moyen , Sclérose en plaques/immunologie , Sclérose en plaques/anatomopathologie , Facteurs de risque , Jeune adulte
14.
Nat Immunol ; 19(5): 1-7, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29662171

RÉSUMÉ

The cytokine transforming growth factor-ß (TGF-ß) regulates the development and homeostasis of several tissue-resident macrophage populations, including microglia. TGF-ß is not critical for microglia survival but is required for the maintenance of the microglia-specific homeostatic gene signature1,2. Under defined host conditions, circulating monocytes can compete for the microglial niche and give rise to long-lived monocyte-derived macrophages residing in the central nervous system (CNS)3-5. Whether monocytes require TGF-ß for colonization of the microglial niche and maintenance of CNS integrity is unknown. We found that abrogation of TGF-ß signaling in CX3CR1+ monocyte-derived macrophages led to rapid onset of a progressive and fatal demyelinating motor disease characterized by myelin-laden giant macrophages throughout the spinal cord. Tgfbr2-deficient macrophages were characterized by high expression of genes encoding proteins involved in antigen presentation, inflammation and phagocytosis. TGF-ß is thus crucial for the functional integration of monocytes into the CNS microenvironment.


Sujet(s)
Encéphale/immunologie , Maladies démyélinisantes/immunologie , Macrophages/anatomopathologie , Moelle spinale/immunologie , Facteur de croissance transformant bêta/immunologie , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Maladies démyélinisantes/métabolisme , Maladies démyélinisantes/anatomopathologie , Macrophages/immunologie , Macrophages/métabolisme , Souris , Transduction du signal , Moelle spinale/métabolisme , Moelle spinale/anatomopathologie , Facteur de croissance transformant bêta/métabolisme
15.
Hum Mol Genet ; 27(5): 912-928, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29325110

RÉSUMÉ

Despite advancements in genetic studies, it is difficult to understand and characterize the functional relevance of disease-associated genetic variants, especially in the context of a complex multifactorial disease such as multiple sclerosis (MS). As a large proportion of expression quantitative trait loci (eQTLs) are context-specific, we performed RNA-Seq in peripheral blood mononuclear cells from MS patients (n = 145) to identify eQTLs in regions centered on 109 MS risk single nucleotide polymorphisms and 7 associated human leukocyte antigen variants. We identified 77 statistically significant eQTL associations, including pseudogenes and non-coding RNAs. Thirty-eight out of 40 testable eQTL effects were colocalized with the disease association signal. As many eQTLs are tissue specific, we aimed to detail their significance in different cell types. Approximately 70% of the eQTLs were replicated and characterized in at least one major peripheral blood mononuclear cell-derived cell type. Furthermore, 40% of eQTLs were found to be more pronounced in MS patients compared with non-inflammatory neurological diseases patients. In addition, we found two single nucleotide polymorphisms to be significantly associated with the proportions of three different cell types. Mapping to enhancer histone marks and predicted transcription factor binding sites added additional functional evidence for eight eQTL regions. As an example, we found that rs71624119, shared with three other autoimmune diseases and located in a primed enhancer (H3K4me1) with potential binding for STAT transcription factors, significantly associates with ANKRD55 expression. This study provides many novel and validated targets for future functional characterization of MS and other diseases.


Sujet(s)
Sclérose en plaques/génétique , Locus de caractère quantitatif , Études de cohortes , Régulation de l'expression des gènes , Prédisposition génétique à une maladie , Antigènes HLA/génétique , Humains , Interféron gamma/pharmacologie , Agranulocytes/physiologie , Déséquilibre de liaison , Lipopolysaccharides/pharmacologie , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Polymorphisme de nucléotide simple , Reproductibilité des résultats
16.
Mult Scler ; 24(10): 1288-1300, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-28766461

RÉSUMÉ

BACKGROUND: Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system caused by genetic and environmental factors. DNA methylation, an epigenetic mechanism that controls genome activity, may provide a link between genetic and environmental risk factors. OBJECTIVE: We sought to identify DNA methylation changes in CD4+ T cells in patients with relapsing-remitting (RR-MS) and secondary-progressive (SP-MS) disease and healthy controls (HC). METHODS: We performed DNA methylation analysis in CD4+ T cells from RR-MS, SP-MS, and HC and associated identified changes with the nearby risk allele, smoking, age, and gene expression. RESULTS: We observed significant methylation differences in the VMP1/MIR21 locus, with RR-MS displaying higher methylation compared to SP-MS and HC. VMP1/MIR21 methylation did not correlate with a known MS risk variant in VMP1 or smoking but displayed a significant negative correlation with age and the levels of mature miR-21 in CD4+ T cells. Accordingly, RR-MS displayed lower levels of miR-21 compared to SP-MS, which might reflect differences in age between the groups, and healthy individuals and a significant enrichment of up-regulated miR-21 target genes. CONCLUSION: Disease-related changes in epigenetic marking of MIR21 in RR-MS lead to differences in miR-21 expression with a consequence on miR-21 target genes.


Sujet(s)
Lymphocytes T CD4+/physiologie , Régulation de l'expression des gènes/physiologie , microARN/génétique , Sclérose en plaques chronique progressive/génétique , Sclérose en plaques récurrente-rémittente/génétique , Adulte , Méthylation de l'ADN , Femelle , Humains , Mâle , Adulte d'âge moyen , Sclérose en plaques chronique progressive/immunologie , Sclérose en plaques récurrente-rémittente/immunologie , Régulation positive
17.
Physiol Genomics ; 49(9): 447-461, 2017 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-28754822

RÉSUMÉ

Multiple sclerosis (MS) is a chronic inflammatory and demyelinating disease of the central nervous system. MS likely results from a complex interplay between predisposing causal gene variants (the strongest influence coming from HLA class II locus) and environmental risk factors such as smoking, infectious mononucleosis, and lack of sun exposure/vitamin D. However, little is known about the mechanisms underlying MS development and progression. Moreover, the clinical heterogeneity and variable response to treatment represent additional challenges to a comprehensive understanding and efficient treatment of disease. Epigenetic processes, such as DNA methylation and histone posttranslational modifications, integrate influences from the genes and the environment to regulate gene expression accordingly. Studying epigenetic modifications, which are stable and reversible, may provide an alternative approach to better understand and manage disease. We here aim to review findings from epigenetic studies in MS and further discuss the challenges and clinical opportunities arising from epigenetic research, many of which apply to other diseases with similar complex etiology. A growing body of evidence supports a role of epigenetic processes in the mechanisms underlying immune pathogenesis and nervous system dysfunction in MS. However, disparities between studies shed light on the need to consider possible confounders and methodological limitations for a better interpretation of the data. Nevertheless, translational use of epigenetics might offer new opportunities in epigenetic-based diagnostics and therapeutic tools for a personalized care of MS patients.


Sujet(s)
Recherche biomédicale , Épigenèse génétique , Sclérose en plaques/génétique , Animaux , Marqueurs biologiques/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Humains
18.
Proc Natl Acad Sci U S A ; 114(9): E1678-E1687, 2017 02 28.
Article de Anglais | MEDLINE | ID: mdl-28196884

RÉSUMÉ

Vitamin D exerts multiple immunomodulatory functions and has been implicated in the etiology and treatment of several autoimmune diseases, including multiple sclerosis (MS). We have previously reported that in juvenile/adolescent rats, vitamin D supplementation protects from experimental autoimmune encephalomyelitis (EAE), a model of MS. Here we demonstrate that this protective effect associates with decreased proliferation of CD4+ T cells and lower frequency of pathogenic T helper (Th) 17 cells. Using transcriptome, methylome, and pathway analyses in CD4+ T cells, we show that vitamin D affects multiple signaling and metabolic pathways critical for T-cell activation and differentiation into Th1 and Th17 subsets in vivo. Namely, Jak/Stat, Erk/Mapk, and Pi3K/Akt/mTor signaling pathway genes were down-regulated upon vitamin D supplementation. The protective effect associated with epigenetic mechanisms, such as (i) changed levels of enzymes involved in establishment and maintenance of epigenetic marks, i.e., DNA methylation and histone modifications; (ii) genome-wide reduction of DNA methylation, and (iii) up-regulation of noncoding RNAs, including microRNAs, with concomitant down-regulation of their protein-coding target RNAs involved in T-cell activation and differentiation. We further demonstrate that treatment of myelin-specific T cells with vitamin D reduces frequency of Th1 and Th17 cells, down-regulates genes in key signaling pathways and epigenetic machinery, and impairs their ability to transfer EAE. Finally, orthologs of nearly 50% of candidate MS risk genes and 40% of signature genes of myelin-reactive T cells in MS changed their expression in vivo in EAE upon supplementation, supporting the hypothesis that vitamin D may modulate risk for developing MS.


Sujet(s)
Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Vitamine D/pharmacologie , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Épigenèse génétique/effets des médicaments et des substances chimiques , Génomique/méthodes , Activation des lymphocytes/effets des médicaments et des substances chimiques , Sclérose en plaques/traitement médicamenteux , Rats , Transduction du signal/génétique , Transduction du signal/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Cellules Th17/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
19.
PLoS One ; 11(12): e0168276, 2016.
Article de Anglais | MEDLINE | ID: mdl-27959965

RÉSUMÉ

BACKGROUND: Genetic variation in the Laccase (multicopper oxidoreductase) domain-containing 1 (LACC1) gene has been shown to affect the risk of Crohn's disease, leprosy and, more recently, ulcerative colitis and juvenile idiopathic arthritis. LACC1 function appears to promote fatty-acid oxidation, with concomitant inflammasome activation, reactive oxygen species production, and anti-bacterial responses in macrophages. We sought to contribute to elucidating LACC1 biological function by extensive characterization of its expression in human tissues and cells, and through preliminary analyses of the regulatory mechanisms driving such expression. METHODS: We implemented Western blot, quantitative real-time PCR, immunofluorescence microscopy, and flow cytometry analyses to investigate fatty acid metabolism-immune nexus (FAMIN; the LACC1 encoded protein) expression in subcellular compartments, cell lines and relevant human tissues. Gene-set enrichment analyses were performed to initially investigate modulatory mechanisms of LACC1 expression. A small-interference RNA knockdown in vitro model system was used to study the effect of FAMIN depletion on peroxisome function. RESULTS: FAMIN expression was detected in macrophage-differentiated THP-1 cells and several human tissues, being highest in neutrophils, monocytes/macrophages, myeloid and plasmacytoid dendritic cells among peripheral blood cells. Subcellular co-localization was exclusively confined to peroxisomes, with some additional positivity for organelle endomembrane structures. LACC1 co-expression signatures were enriched for genes involved in peroxisome proliferator-activated receptors (PPAR) signaling pathways, and PPAR ligands downregulated FAMIN expression in in vitro model systems. CONCLUSION: FAMIN is a peroxisome-associated protein with primary role(s) in macrophages and other immune cells, where its metabolic functions may be modulated by PPAR signaling events. However, the precise molecular mechanisms through which FAMIN exerts its biological effects in immune cells remain to be elucidated.


Sujet(s)
Maladie de Crohn/génétique , Prédisposition génétique à une maladie , Protéines/génétique , Différenciation cellulaire , Lignée cellulaire tumorale , Acides gras/métabolisme , Analyse de profil d'expression de gènes , Cellules HeLa , Humains , Inflammasomes/métabolisme , Protéines et peptides de signalisation intracellulaire , Agranulocytes/cytologie , Ligands , Macrophages/cytologie , Macrophages/métabolisme , Oxygène/composition chimique , Petit ARN interférent/métabolisme , Espèces réactives de l'oxygène/métabolisme , Transduction du signal
20.
J Leukoc Biol ; 99(3): 437-46, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26516183

RÉSUMÉ

Dendritic cells are professional APCs that play a central role in the initiation of immune responses. The limited ex vivo availability of dendritic cells inspires the widespread use of bone marrow-derived dendritic cells as an alternative in research. However, the functional characteristics of bone marrow-derived dendritic cells are incompletely understood. Therefore, we compared functional and phenotypic characteristics of rat bone marrow-derived dendritic cells generated with GM-CSF/IL-4 or FLT3 ligand bone marrow-derived dendritic cells. A comparison of surface markers revealed that FLT3 ligand-bone marrow-derived dendritic cells expressed signal regulatory protein α, CD103, and CD4 and baseline levels of MHC class II, CD40, and CD86, which were highly up-regulated upon stimulation. Conversely, GM-CSF/IL-4-bone marrow-derived dendritic cells constitutively expressed signal regulatory protein α, CD11c, and CD11b but only mildly up-regulated MHC class II, CD40, or CD86 following stimulation. Expression of dendritic cell-associated core transcripts was restricted to FLT3 ligand-bone marrow-derived dendritic cells . GM-CSF/IL-4-bone marrow-derived dendritic cells were superior at phagocytosis but were outperformed by FLT3 ligand-bone marrow-derived dendritic cells at antigen presentation and T cell stimulation in vitro. Stimulated GM-CSF/IL-4-bone marrow-derived dendritic cells secreted more TNF, CCL5, CCL20, and NO, whereas FLT3 ligand-bone marrow-derived dendritic cells secreted more IL-6 and IL-12. Finally, whereas GM-CSF/IL-4-bone marrow-derived dendritic cell culture supernatants added to resting T cell cultures promoted forkhead box p3(+) regulatory T cell populations, FLT3 ligand-bone marrow-derived dendritic cell culture supernatants drove Th17 differentiation. We conclude that rat GM-CSF/IL-4-bone marrow-derived dendritic cells and FLT3 ligand-bone marrow-derived dendritic cells are functionally distinct. Our data support the current rationale that FLT3 ligand-bone marrow-derived dendritic cells mostly resemble classic dendritic cells but comprise additional minor subpopulations, whereas GM-CSF/IL-4-bone marrow-derived dendritic cells resemble monocyte-derived inflammatory dendritic cells (iNOS-positive monocyte-derived cells).


Sujet(s)
Cellules de la moelle osseuse/physiologie , Cellules dendritiques/physiologie , Facteur de stimulation des colonies de granulocytes et de macrophages/pharmacologie , Interleukine-4/pharmacologie , Protéines membranaires/pharmacologie , Animaux , Phénotype , Rats , Rats de lignée LEW
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE