Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
iScience ; 25(7): 104684, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35856023

RESUMEN

SMCHD1 (structural maintenance of chromosomes hinge domain containing 1) is a noncanonical SMC protein that mediates long-range repressive chromatin structures. SMCHD1 is required for X chromosome inactivation in female cells and repression of imprinted and clustered autosomal genes, with SMCHD1 mutations linked to human diseases facioscapulohumeral muscular dystrophy (FSHD) and bosma arhinia and micropthalmia syndrome (BAMS). We used a conditional mouse model to investigate SMCHD1 in hematopoiesis. Smchd1-deleted mice maintained steady-state hematopoiesis despite showing an impaired reconstitution capacity in competitive bone marrow transplantations and age-related hematopoietic stem cell (HSC) loss. This phenotype was more pronounced in Smchd1-deleted females, which showed a loss of quiescent HSCs and fewer B cells. Gene expression profiling of Smchd1-deficient HSCs and B cells revealed known and cell-type-specific SMCHD1-sensitive genes and significant disruption to X-linked gene expression in female cells. These data show SMCHD1 is a regulator of HSCs whose effects are more profound in females.

2.
Epigenetics Chromatin ; 15(1): 26, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35843975

RESUMEN

Embryonic development is dependent on the maternal supply of proteins through the oocyte, including factors setting up the adequate epigenetic patterning of the zygotic genome. We previously reported that one such factor is the epigenetic repressor SMCHD1, whose maternal supply controls autosomal imprinted expression in mouse preimplantation embryos and mid-gestation placenta. In mouse preimplantation embryos, X chromosome inactivation is also an imprinted process. Combining genomics and imaging, we show that maternal SMCHD1 is required not only for the imprinted expression of Xist in preimplantation embryos, but also for the efficient silencing of the inactive X in both the preimplantation embryo and mid-gestation placenta. These results expand the role of SMCHD1 in enforcing the silencing of Polycomb targets. The inability of zygotic SMCHD1 to fully restore imprinted X inactivation further points to maternal SMCHD1's role in setting up the appropriate chromatin environment during preimplantation development, a critical window of epigenetic remodelling.


Asunto(s)
Proteínas Cromosómicas no Histona , ARN Largo no Codificante , Inactivación del Cromosoma X , Animales , Blastocisto/fisiología , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Desarrollo Embrionario , Impresión Genómica , Ratones , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromosoma X
3.
Nat Commun ; 13(1): 1658, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351876

RESUMEN

The process of epigenetic silencing, while fundamentally important, is not yet completely understood. Here we report a replenishable female mouse embryonic stem cell (mESC) system, Xmas, that allows rapid assessment of X chromosome inactivation (XCI), the epigenetic silencing mechanism of one of the two X chromosomes that enables dosage compensation in female mammals. Through a targeted genetic screen in differentiating Xmas mESCs, we reveal that the BAF complex is required to create nucleosome-depleted regions at promoters on the inactive X chromosome during the earliest stages of establishment of XCI. Without this action gene silencing fails. Xmas mESCs provide a tractable model for screen-based approaches that enable the discovery of unknown facets of the female-specific process of XCI and epigenetic silencing more broadly.


Asunto(s)
ARN Largo no Codificante , Inactivación del Cromosoma X , Animales , Cromatina/genética , Compensación de Dosificación (Genética) , Epigénesis Genética , Femenino , Ratones , ARN Largo no Codificante/genética , Cromosoma X/genética , Inactivación del Cromosoma X/genética
4.
Elife ; 92020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33186096

RESUMEN

Genomic imprinting establishes parental allele-biased expression of a suite of mammalian genes based on parent-of-origin specific epigenetic marks. These marks are under the control of maternal effect proteins supplied in the oocyte. Here we report epigenetic repressor Smchd1 as a novel maternal effect gene that regulates the imprinted expression of ten genes in mice. We also found zygotic SMCHD1 had a dose-dependent effect on the imprinted expression of seven genes. Together, zygotic and maternal SMCHD1 regulate three classic imprinted clusters and eight other genes, including non-canonical imprinted genes. Interestingly, the loss of maternal SMCHD1 does not alter germline DNA methylation imprints pre-implantation or later in gestation. Instead, what appears to unite most imprinted genes sensitive to SMCHD1 is their reliance on polycomb-mediated methylation as germline or secondary imprints, therefore we propose that SMCHD1 acts downstream of polycomb imprints to mediate its function.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Impresión Genómica/genética , Animales , Blastocisto , Proteínas Cromosómicas no Histona/genética , Metilación de ADN , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Genotipo , Proteínas Fluorescentes Verdes , Masculino , Ratones , Células-Madre Neurales
5.
Artículo en Inglés | MEDLINE | ID: mdl-27195021

RESUMEN

BACKGROUND: The presence of histone 3 lysine 9 (H3K9) methylation on the mouse inactive X chromosome has been controversial over the last 15 years, and the functional role of H3K9 methylation in X chromosome inactivation in any species has remained largely unexplored. RESULTS: Here we report the first genomic analysis of H3K9 di- and tri-methylation on the inactive X: we find they are enriched at the intergenic, gene poor regions of the inactive X, interspersed between H3K27 tri-methylation domains found in the gene dense regions. Although H3K9 methylation is predominantly non-genic, we find that depletion of H3K9 methylation via depletion of H3K9 methyltransferase Set domain bifurcated 1 (Setdb1) during the establishment of X inactivation, results in failure of silencing for around 150 genes on the inactive X. By contrast, we find a very minor role for Setdb1-mediated H3K9 methylation once X inactivation is fully established. In addition to failed gene silencing, we observed a specific failure to silence X-linked long-terminal repeat class repetitive elements. CONCLUSIONS: Here we have shown that H3K9 methylation clearly marks the murine inactive X chromosome. The role of this mark is most apparent during the establishment phase of gene silencing, with a more muted effect on maintenance of the silent state. Based on our data, we hypothesise that Setdb1-mediated H3K9 methylation plays a role in epigenetic silencing of the inactive X via silencing of the repeats, which itself facilitates gene silencing through alterations to the conformation of the whole inactive X chromosome.

6.
Blood ; 126(2): 167-75, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26036803

RESUMEN

Polycomb repressive complex 2 (PRC2) is a chromatin modifier that regulates stem cells in embryonic and adult tissues. Loss-of-function studies of PRC2 components have been complicated by early embryonic dependence on PRC2 activity and the partial functional redundancy of enhancer of zeste homolog 1 (Ezh1) and enhancer of zeste homolog 2 (Ezh2), which encode the enzymatic component of PRC2. Here, we investigated the role of PRC2 in hematopoiesis by conditional deletion of suppressor of zeste 12 protein homolog (Suz12), a core component of PRC2. Complete loss of Suz12 resulted in failure of hematopoiesis, both in the embryo and the adult, with a loss of maintenance of hematopoietic stem cells (HSCs). In contrast, partial loss of PRC2 enhanced HSC self-renewal. Although Suz12 was required for lymphoid development, deletion in individual blood cell lineages revealed that it was dispensable for the development of granulocytic, monocytic, and megakaryocytic cells. Collectively, these data reveal the multifaceted role of PRC2 in hematopoiesis, with divergent dose-dependent effects in HSC and distinct roles in maturing blood cells. Because PRC2 is a potential target for cancer therapy, the significant consequences of modest changes in PRC2 activity, as well as the cell and developmental stage-specific effects, will need to be carefully considered in any therapeutic context.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Linfopoyesis/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Proliferación Celular/genética , Células Cultivadas , Feto/inmunología , Feto/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/genética
7.
Blood ; 125(12): 1890-900, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25645357

RESUMEN

Polycomb repressive complex 2 (PRC2) plays a key role in hematopoietic stem and progenitor cell (HSPC) function. Analyses of mouse mutants harboring deletions of core components have implicated PRC2 in fine-tuning multiple pathways that instruct HSPC behavior, yet how PRC2 is targeted to specific genomic loci within HSPCs remains unknown. Here we use short hairpin RNA-mediated knockdown to survey the function of PRC2 accessory factors that were defined in embryonic stem cells (ESCs) by testing the competitive reconstitution capacity of transduced murine HSPCs. We find that, similar to the phenotype observed upon depletion of core subunit Suz12, depleting Jarid2 enhances the competitive transplantation capacity of both fetal and adult mouse HSPCs. Furthermore, we demonstrate that depletion of JARID2 enhances the in vitro expansion and in vivo reconstitution capacity of human HSPCs. Gene expression profiling revealed common Suz12 and Jarid2 target genes that are enriched for the H3K27me3 mark established by PRC2. These data implicate Jarid2 as an important component of PRC2 that has a central role in coordinating HSPC function.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Complejo Represivo Polycomb 2/metabolismo , Animales , Antígenos CD34/metabolismo , Linaje de la Célula , Perfilación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/citología , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fenotipo , ARN Interferente Pequeño/metabolismo , Células Madre/citología
8.
Front Genet ; 5: 329, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25309581

RESUMEN

The advent of high-throughput sequencing has allowed genome wide profiling of histone modifications by Chromatin ImmunoPrecipitation (ChIP) followed by sequencing (ChIP-seq). In this assay the histone mark of interest is enriched through a chromatin pull-down assay using an antibody for the mark. Due to imperfect antibodies and other factors, many of the sequenced fragments do not originate from the histone mark of interest, and are referred to as background reads. Background reads are not uniformly distributed and therefore control samples are usually used to estimate the background distribution at any given genomic position. The Encyclopedia of DNA Elements (ENCODE) Consortium guidelines suggest sequencing a whole cell extract (WCE, or "input") sample, or a mock ChIP reaction such as an IgG control, as a background sample. However, for a histone modification ChIP-seq investigation it is also possible to use a Histone H3 (H3) pull-down to map the underlying distribution of histones. In this paper we generated data from a hematopoietic stem and progenitor cell population isolated from mouse fetal liver to compare WCE and H3 ChIP-seq as control samples. The quality of the control samples is estimated by a comparison to pull-downs of histone modifications and to expression data. We find minor differences between WCE and H3 ChIP-seq, such as coverage in mitochondria and behavior close to transcription start sites. Where the two controls differ, the H3 pull-down is generally more similar to the ChIP-seq of histone modifications. However, the differences between H3 and WCE have a negligible impact on the quality of a standard analysis.

9.
J Biol Chem ; 287(24): 20652-63, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22535952

RESUMEN

Eukaryotic cells generate energy in the form of ATP, through a network of mitochondrial complexes and electron carriers known as the oxidative phosphorylation system. In mammals, mitochondrial complex I (CI) is the largest component of this system, comprising 45 different subunits encoded by mitochondrial and nuclear DNA. Humans diagnosed with mutations in the gene NDUFS4, encoding a nuclear DNA-encoded subunit of CI (NADH dehydrogenase ubiquinone Fe-S protein 4), typically suffer from Leigh syndrome, a neurodegenerative disease with onset in infancy or early childhood. Mitochondria from NDUFS4 patients usually lack detectable NDUFS4 protein and show a CI stability/assembly defect. Here, we describe a recessive mouse phenotype caused by the insertion of a transposable element into Ndufs4, identified by a novel combined linkage and expression analysis. Designated Ndufs4(fky), the mutation leads to aberrant transcript splicing and absence of NDUFS4 protein in all tissues tested of homozygous mice. Physical and behavioral symptoms displayed by Ndufs4(fky/fky) mice include temporary fur loss, growth retardation, unsteady gait, and abnormal body posture when suspended by the tail. Analysis of CI in Ndufs4(fky/fky) mice using blue native PAGE revealed the presence of a faster migrating crippled complex. This crippled CI was shown to lack subunits of the "N assembly module", which contains the NADH binding site, but contained two assembly factors not present in intact CI. Metabolomic analysis of the blood by tandem mass spectrometry showed increased hydroxyacylcarnitine species, implying that the CI defect leads to an imbalanced NADH/NAD(+) ratio that inhibits mitochondrial fatty acid ß-oxidation.


Asunto(s)
Elementos Transponibles de ADN , Complejo I de Transporte de Electrón/metabolismo , Enfermedad de Leigh/enzimología , Mitocondrias/enzimología , Mutación , NAD/metabolismo , Animales , Sitios de Unión , Complejo I de Transporte de Electrón/genética , Humanos , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Enfermedad de Leigh/fisiopatología , Metabolómica/métodos , Ratones , Ratones Mutantes , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/patología , NAD/genética , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , Proteómica/métodos , Empalme del ARN/genética
10.
EMBO J ; 31(3): 692-706, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22124325

RESUMEN

The role of specific members of the NF-κB family of transcription factors in CD8 T-cell selection and development is largely unknown. Here, we show that mice lacking NF-κB1 develop a unique population of conventional CD8 single-positive (SP) thymocytes with memory T cell-like properties that populate peripheral immune organs. Development of this memory-like population is not due to PLZF(+) thymocytes and instead coincides with changes in CD8 T-cell selection. These include a reduction in the efficiency of negative selection and a dependence on MHC class Ia or Ib expressed by haematopoietic cells. These findings indicate that NF-κB1 regulates multiple events in the thymus that collectively inhibit the excess development of CD8(+) thymocytes with memory cell characteristics.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/fisiología , FN-kappa B/fisiología , Timo/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inmunofenotipificación , Interleucina-4/biosíntesis , FN-kappa B/genética , Transducción de Señal
11.
Mol Immunol ; 49(3): 518-26, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22036612

RESUMEN

Mutations in the AIRE gene cause autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED), which is associated with autoimmunity towards several peripheral organs. The AIRE protein is almost exclusively expressed in medullary thymic epithelial cells (mTEC) and CpG methylation in the promoter of the AIRE gene has been suggested to control its tissue-specific expression pattern. We found that in human AIRE-positive medullary and AIRE-negative cortical epithelium, the AIRE promoter is hypomethylated, whereas in thymocytes, the promoter had high level of CpG methylation. Likewise, in mouse mTECs the AIRE promoter was uniformly hypomethylated. In the same vein, the AIRE promoter was hypomethylated in AIRE-negative thymic epithelial tumors (thymomas) and in several peripheral tissues. Our data are compatible with the notion that promoter hypomethylation is necessary but not sufficient for tissue-specific regulation of the AIRE gene. In contrast, a positive correlation between AIRE expression and histone H3 lysine 4 trimethylation, an active chromatin mark, was found in the AIRE promoter in human and mouse TECs.


Asunto(s)
Metilación de ADN , Células Epiteliales/metabolismo , Regiones Promotoras Genéticas , Timoma/metabolismo , Timo/metabolismo , Neoplasias del Timo/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteína AIRE
12.
Blood ; 118(9): 2462-72, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21505196

RESUMEN

To investigate the role of Aire in thymic selection, we examined the cellular requirements for generation of ovalbumin (OVA)-specific CD4 and CD8 T cells in mice expressing OVA under the control of the rat insulin promoter. Aire deficiency reduced the number of mature single-positive OVA-specific CD4(+) or CD8(+) T cells in the thymus, independent of OVA expression. Importantly, it also contributed in 2 ways to OVA-dependent negative selection depending on the T-cell type. Aire-dependent negative selection of OVA-specific CD8 T cells correlated with Aire-regulated expression of OVA. By contrast, for OVA-specific CD4 T cells, Aire affected tolerance induction by a mechanism that operated independent of the level of OVA expression, controlling access of antigen presenting cells to medullary thymic epithelial cell (mTEC)-expressed OVA. This study supports the view that one mechanism by which Aire controls thymic negative selection is by regulating the indirect presentation of mTEC-derived antigens by thymic dendritic cells. It also indicates that mTECs can mediate tolerance by direct presentation of Aire-regulated antigens to both CD4 and CD8 T cells.


Asunto(s)
Presentación de Antígeno , Antígenos/metabolismo , Supresión Clonal/inmunología , Células Dendríticas/inmunología , Células Epiteliales/inmunología , Tolerancia Inmunológica/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Antígenos/inmunología , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Cruzamientos Genéticos , Citocinas/metabolismo , Regulación de la Expresión Génica/inmunología , Insulina/genética , Ratones , Ratones Transgénicos , Ovalbúmina/genética , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Regiones Promotoras Genéticas , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Timo/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína AIRE
13.
PLoS One ; 6(3): e18276, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21483837

RESUMEN

Previous studies on the epigenetic regulator DNA methyltransferase 3-Like (DNMT3L), have demonstrated it is an essential regulator of paternal imprinting and early male meiosis. Dnmt3L is also a paternal effect gene, i.e., wild type offspring of heterozygous mutant sires display abnormal phenotypes suggesting the inheritance of aberrant epigenetic marks on the paternal chromosomes. In order to reveal the mechanisms underlying these paternal effects, we have assessed X chromosome meiotic compaction, XY chromosome aneuploidy rates and global transcription in meiotic and haploid germ cells from male mice heterozygous for Dnmt3L. XY bodies from Dnmt3L heterozygous males were significantly longer than those from wild types, and were associated with a three-fold increase in XY bearing sperm. Loss of a Dnmt3L allele resulted in deregulated expression of a large number of both X-linked and autosomal genes within meiotic cells, but more prominently in haploid germ cells. Data demonstrate that similar to embryonic stem cells, DNMT3L is involved in an auto-regulatory loop in germ cells wherein the loss of a Dnmt3L allele resulted in increased transcription from the remaining wild type allele. In contrast, however, within round spermatids, this auto-regulatory loop incorporated the alternative non-coding alternative transcripts. Consistent with the mRNA data, we have localized DNMT3L within spermatids and sperm and shown that the loss of a Dnmt3L allele results in a decreased DNMT3L content within sperm. These data demonstrate previously unrecognised roles for DNMT3L in late meiosis and in the transcriptional regulation of meiotic and post-meiotic germ cells. These data provide a potential mechanism for some cases of human Klinefelter's and Turner's syndromes.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Transcripción Genética/genética , Cromosoma X/metabolismo , Alelos , Animales , Western Blotting , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , Femenino , Células Germinativas , Heterocigoto , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Meiosis/genética , Meiosis/fisiología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Espermátides/metabolismo , Espermatozoides/metabolismo , Cromosoma X/genética , Cromosoma Y/genética , Cromosoma Y/metabolismo
14.
Eur J Immunol ; 40(12): 3499-509, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108470

RESUMEN

The autoimmune regulator (AIRE) promotes "promiscuous" expression of tissue-restricted antigens (TRA) in thymic medullary epithelial cells to facilitate thymic deletion of autoreactive T-cells. Here, we show that AIRE-deficient mice showed an earlier development of myelin oligonucleotide glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). To determine the outcome of ectopic Aire expression, we used a retroviral transduction system to over-express Aire in vitro, in cell lines and in bone marrow (BM). In the cell lines that included those of thymic medullary and dendritic cell origin, ectopically expressed Aire variably promoted expression of TRA including Mog and Ins2 (proII) autoantigens associated, respectively, with the autoimmune diseases multiple sclerosis and type 1 diabetes. BM chimeras generated from BM transduced with a retrovirus encoding Aire displayed elevated levels of Mog and Ins2 expression in thymus and spleen. Following induction of EAE with MOG(35-55), transplanted mice displayed significant delay in the onset of EAE compared with control mice. To our knowledge, this is the first example showing that in vivo ectopic expression of AIRE can modulate TRA expression and alter autoimmune disease development.


Asunto(s)
Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Encefalomielitis Autoinmune Experimental/inmunología , Células Epiteliales/metabolismo , Factores de Transcripción/metabolismo , Animales , Presentación de Antígeno/genética , Autoantígenos/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Línea Celular , Clonación Molecular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/terapia , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Glicoproteínas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/inmunología , Timo/patología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Transgenes/genética , Proteína AIRE
15.
Nucleic Acids Res ; 38(7): 2168-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20056656

RESUMEN

A fundamental question in microarray analysis is the estimation of the number of expressed probes in different RNA samples. Negative control probes available in the latest microarray platforms, such as Illumina whole genome expression BeadChips, provide a unique opportunity to estimate the number of expressed probes without setting a threshold. A novel algorithm was proposed in this study to estimate the number of expressed probes in an RNA sample by utilizing these negative controls to measure background noise. The performance of the algorithm was demonstrated by comparing different generations of Illumina BeadChips, comparing the set of probes targeting well-characterized RefSeq NM transcripts with other probes on the array and comparing pure samples with heterogenous samples. Furthermore, hematopoietic stem cells were found to have a larger transcriptome than progenitor cells. Aire knockout medullary thymic epithelial cells were shown to have significantly less expressed probes than matched wild-type cells.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Sondas de Oligonucleótidos/análisis , Animales , Células Madre Hematopoyéticas/metabolismo , Ratones , ARN Mensajero/análisis , Células Madre/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Proteína AIRE
16.
Arthritis Rheum ; 60(6): 1683-93, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19479827

RESUMEN

OBJECTIVE: Autoimmune regulator (Aire) promotes the ectopic expression of tissue-restricted antigens in medullary thymic epithelial cells (mTECs), leading to negative selection of autoreactive T cells. This study was undertaken to determine whether loss of central tolerance renders Aire-deficient (Aire-/-) mice more susceptible to the induction of autoimmune arthritis. METHODS: Medullary TECs were isolated from Aire-/- and wild-type C57BL/6 mice for gene expression analysis. Collagen-induced arthritis (CIA) was elicited by injection of chick type II collagen (CII) in adjuvant. Cellular and humoral immune responses to CII were evaluated. Chimeric mice were created by reconstituting lymphocyte-deficient mice with either Aire-/- or wild-type CD4 T cells and wild-type B cells. RESULTS: Wild-type, but not Aire-/-, mTECs expressed the CII gene Col2a1. Aire-/- mice developed more rapid and severe CIA, showing elevated serum anti-CII IgG levels, with earlier switching to arthritogenic IgG subclasses. No evidence was found of enhanced T cell responsiveness to CII in Aire-/- mice; however, Aire-/- CD4 T cells were more efficient at stimulating wild-type B cells to produce anti-CII IgG following immunization of chimeric mice with CII. CONCLUSION: Our findings indicate that Aire-dependent expression of CII occurs in mTECs, implying that there is central tolerance to self antigens found in articular cartilage. Reduced central tolerance to CII in Aire-/- mice manifests as increased CD4 T cell help to B cells for cross-reactive autoantibody production and enhanced CIA. Aire and central tolerance help prevent cross-reactive autoimmune responses to CII initiated by environmental stimuli and limit spontaneous autoimmunity.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/patología , Autoanticuerpos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Factores de Transcripción/metabolismo , Animales , Artritis Experimental/epidemiología , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Autoinmunidad/fisiología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Riesgo , Índice de Severidad de la Enfermedad , Timo/metabolismo , Timo/patología , Factores de Transcripción/genética , Proteína AIRE
17.
J Immunol ; 182(6): 3902-18, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265170

RESUMEN

Autoimmune regulator (AIRE) is an important transcription regulator that mediates a role in central tolerance via promoting the "promiscuous" expression of tissue-specific Ags in the thymus. Although several mouse models of Aire deficiency have been described, none has analyzed the phenotype induced by a mutation that emulates the common 13-bp deletion in human APECED (autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy) by disrupting the first plant homeodomain in exon 8. Aire-deficient mice with a corresponding mutation showed some disturbance of the medullary epithelial compartment, but at the phenotypic level their T cell compartment appeared relatively normal in the thymus and periphery. An increase in the number of activated T cells was evident, and autoantibodies against several organs were detected. At the histological level, lymphocytic infiltration of several organs indicated the development of autoimmunity, although symptoms were mild and the quality of life for Aire-deficient mice appeared equivalent to wild-type littermates, with the exception of male infertility. Vbeta and CDR3 length analysis suggested that each Aire-deficient mouse developed its own polyclonal autoimmune repertoire. Finally, given the prevalence of candidiasis in APECED patients, we examined the control of infection with Candida albicans in Aire-deficient mice. No increase in disease susceptibility was found for either oral or systemic infection. These observations support the view that additional genetic and/or environmental factors contribute substantially to the overt nature of autoimmunity associated with Aire mutations, even for mutations identical to those found in humans with APECED.


Asunto(s)
Imitación Molecular/genética , Imitación Molecular/inmunología , Mutagénesis Sitio-Dirigida , Fenotipo , Poliendocrinopatías Autoinmunes/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Emparejamiento Base/genética , Secuencia de Bases , Línea Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Datos de Secuencia Molecular , Poliendocrinopatías Autoinmunes/inmunología , Poliendocrinopatías Autoinmunes/metabolismo , Homología de Secuencia de Aminoácido , Timo/inmunología , Timo/metabolismo , Timo/patología , Factores de Transcripción/biosíntesis , Proteína AIRE
18.
J Immunol ; 180(6): 3824-32, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322189

RESUMEN

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy is an autoimmune disorder caused by mutations in the autoimmune regulator gene AIRE. We examined the expression of Aire in different organs (thymus, spleen, and lymph nodes) in C57BL/6 mice, using a novel rat mAb, specific for murine Aire. Using flow cytometry, directly fluorochrome-labeled mAb revealed Aire expression in a rare thymic cellular subset that was CD45(-), expressed low levels of Ly51, and was high for MHC-II and EpCam. This subset also expressed a specific pattern of costimulatory molecules, including CD40, CD80, and PD-L1. Immunohistochemical analysis revealed that Aire(+) cells were specifically localized to the thymus or, more precisely, to the cortico-medulla junction and medulla, correlating with the site of negative selection. Although in agreement with previous studies, low levels of Aire mRNA was detected in all dendritic cell subtypes however lacZ staining, immunohistochemistry and flow cytometry failed to detect Aire protein. At a cellular level, Aire was expressed in perinuclear speckles within the nucleus. This report provides the first detailed analysis of Aire protein expression, highlighting the precise location at both the tissue and cellular level.


Asunto(s)
Anticuerpos Monoclonales/análisis , Especificidad de Anticuerpos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Timo/citología , Timo/inmunología , Factores de Transcripción/biosíntesis , Factores de Transcripción/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Reacciones Antígeno-Anticuerpo , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Puntos Cuánticos , Ratas , Timo/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína AIRE
19.
N Engl J Med ; 358(10): 1018-28, 2008 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-18322283

RESUMEN

BACKGROUND: Autoimmune polyendocrine syndrome type 1 (APS-1) is a multiorgan autoimmune disorder caused by mutations in AIRE, the autoimmune regulator gene. Though recent studies concerning AIRE deficiency have begun to elucidate the molecular pathogenesis of organ-specific autoimmunity in patients with APS-1, the autoantigen responsible for hypoparathyroidism, a hallmark of APS-1 and its most common autoimmune endocrinopathy, has not yet been identified. METHODS: We performed immunoscreening of a human parathyroid complementary DNA library, using serum samples from patients with APS-1 and hypoparathyroidism, to identify patients with reactivity to the NACHT leucine-rich-repeat protein 5 (NALP5). Subsequently, serum samples from 87 patients with APS-1 and 293 controls, including patients with other autoimmune disorders, were used to determine the frequency and specificity of autoantibodies against NALP5. In addition, the expression of NALP5 was investigated in various tissues. RESULTS: NALP5-specific autoantibodies were detected in 49% of the patients with APS-1 and hypoparathyroidism but were absent in all patients with APS-1 but without hypoparathyroidism, in all patients with other autoimmune endocrine disorders, and in all healthy controls. NALP5 was predominantly expressed in the cytoplasm of parathyroid chief cells. CONCLUSIONS: NALP5 appears to be a tissue-specific autoantigen involved in hypoparathyroidism in patients with APS-1. Autoantibodies against NALP5 appear to be highly specific and may be diagnostic for this prominent component of APS-1.


Asunto(s)
Autoanticuerpos/sangre , Autoantígenos/inmunología , Hipoparatiroidismo/diagnóstico , Glándulas Paratiroides/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Autoanticuerpos/análisis , Autoantígenos/genética , Biomarcadores/análisis , Biomarcadores/sangre , ADN Complementario/análisis , Biblioteca de Genes , Humanos , Hipoparatiroidismo/etiología , Hipoparatiroidismo/inmunología , Proteínas Mitocondriales , Proteínas Nucleares , Glándulas Paratiroides/química , Poliendocrinopatías Autoinmunes/complicaciones , ARN Mensajero/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...