Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Cell Metab ; 35(2): 228-230, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36754015

RESUMEN

Although metabolic rewiring of Treg cells constitutes a hallmark in autoimmune diseases, extrinsic and intrinsic mechanisms that imprint on this re-programming remain poorly understood. In this issue of Cell Metabolism, Côrte-Real and colleagues demonstrate that high salt exposure disturbs the mitochondrial respiration in Treg cells, promoting a pro-inflammatory phenotype, loss of function, and associated breakdown of self-tolerance.


Asunto(s)
Enfermedades Autoinmunes , Linfocitos T Reguladores , Humanos , Autotolerancia/genética , Tolerancia Inmunológica
2.
FEBS Lett ; 595(14): 1962-1974, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34080184

RESUMEN

Regulatory T cells (Tregs) are indispensable for the maintenance of immunological self-tolerance and homeostasis. Heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) is required for optimal Treg induction. Here, we reveal that human-induced Tregs (iTregs) lacking hnRNPA1 show reduced expression of the transcription factor FOXP3, increased ubiquitination level of FOXP3, and impaired suppressive abilities. Human naïve CD4 T cells with hnRNPA1 knockdown show a decreased Treg differentiation ratio. hnRNPA1 could interact with FOXP3 as well as with the E3 ligase Stub1. The phosphorylation at hnRNPA1 S199 could increase both interactions. The overexpression of FOXP3 in Tregs containing shhnRNPA1 could not recover the phenotype caused by hnRNPA1 knockdown. Therefore, there might be multiple essential pathways regulated by hnRNPA1 in Tregs. In conclusion, we present a new role of hnRNPA1 in promoting Treg function, indicating it as a promising target for tumor therapies.


Asunto(s)
Factores de Transcripción Forkhead/genética , Ribonucleoproteína Nuclear Heterogénea A1/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Ubiquitina-Proteína Ligasas/genética , Diferenciación Celular , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Células HEK293 , Ribonucleoproteína Nuclear Heterogénea A1/antagonistas & inhibidores , Ribonucleoproteína Nuclear Heterogénea A1/inmunología , Homeostasis/inmunología , Humanos , Fosforilación , Cultivo Primario de Células , Unión Proteica , Estabilidad Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Autotolerancia/genética , Transducción de Señal , Linfocitos T Citotóxicos/citología , Linfocitos T Reguladores/citología , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
3.
Front Immunol ; 12: 635521, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34017326

RESUMEN

Highly polymorphic loci evolved many times over the history of species. These polymorphic loci are involved in three types of functions: kind recognition, self-incompatibility, and the jawed vertebrate adaptive immune system (AIS). In the first part of this perspective, we reanalyzed and described some cases of polymorphic loci reported in the literature. There is a convergent evolution within each functional category and between functional categories, suggesting that the emergence of these self/non-self recognition loci has occurred multiple times throughout the evolutionary history. Most of the highly polymorphic loci are coding for proteins that have a homophilic interaction or heterophilic interaction between linked loci, leading to self or non-self-recognition. The highly polymorphic MHCs, which are involved in the AIS have a different functional mechanism, as they interact through presented self or non-self-peptides with T cell receptors, whose diversity is generated by somatic recombination. Here we propose a mechanism called "the capacity of recognition competition mechanism" that might contribute to the evolution of MHC polymorphism. We propose that the published cases corresponding to these three biological categories represent a small part of what can be found throughout the tree of life, and that similar mechanisms will be found many times, including the one where polymorphic loci interact with somatically generated loci.


Asunto(s)
Inmunidad Adaptativa/genética , Evolución Molecular , Genes Codificadores de los Receptores de Linfocitos T/genética , Tolerancia Inmunológica/genética , Complejo Mayor de Histocompatibilidad/genética , Animales , Sitios Genéticos , Humanos , Plantas/genética , Plantas/inmunología , Polimorfismo Genético , Autotolerancia/genética , Especificidad de la Especie
4.
Front Immunol ; 12: 669856, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33986757

RESUMEN

Effective tolerogenic intervention in Rheumatoid Arthritis (RA) will rely upon understanding the evolution of articular antigen specific CD4 T cell responses. TCR clonality of endogenous CD4 T cell infiltrates in early inflammatory arthritis was assessed to monitor evolution of the TCR repertoire in the inflamed joint and associated lymph node (LN). Mouse models of antigen-induced breach of self-tolerance and chronic polyarthritis were used to recapitulate early and late phases of RA. The infiltrating endogenous, antigen experienced CD4 T cells in inflamed joints and LNs were analysed using flow cytometry and TCRß sequencing. TCR repertoires from inflamed late phase LNs displayed increased clonality and diversity compared to early phase LNs, while inflamed joints remained similar with time. Repertoires from late phase LNs accumulated clones with a diverse range of TRBV genes, while inflamed joints at both phases contained clones expressing similar TRBV genes. Repertoires from LNs and joints at the late phase displayed reduced CDR3ß sequence overlap compared to the early disease phase, however the most abundant clones in LNs accumulate in the joint at the later phase. The results indicate CD4 T cell repertoire clonality and diversity broadens with progression of inflammatory arthritis and is first reflected in LNs before mirroring in the joint. These observations imply that antigen specific tolerogenic therapies could be more effective if targeted at earlier phases of disease when CD4 T cell clonality is least diverse.


Asunto(s)
Artritis Experimental/inmunología , Linfocitos T CD4-Positivos/inmunología , Evolución Clonal , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Articulaciones/inmunología , Ganglios Linfáticos/inmunología , Autotolerancia , Animales , Artritis Experimental/genética , Artritis Experimental/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Progresión de la Enfermedad , Femenino , Articulaciones/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Ratones Endogámicos C57BL , Fenotipo , Autotolerancia/genética , Factores de Tiempo
5.
Int J Immunogenet ; 48(2): 193-200, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33112034

RESUMEN

The implementation of the immune checkpoint blockade as a therapeutic option in contemporary oncology is one of the significant immunological achievements in the last century. Constantly accumulating evidence suggests that the response to immune checkpoint inhibitors (ICIs) is not universal. Therefore, it is critical to identify determinants for response, resistance and adverse effects of immune checkpoint therapy that could be developed as prognostic and predictive markers. Recent large scale analyses of cancer genome data revealed the key role of HLA class I and class II molecules in cancer immunoediting, and it appears that HLA diversity can predict response to ICIs. In the present review, we summarize the emerging data on the role of HLA germline variations as a marker for response to ICIs.


Asunto(s)
Antígenos de Neoplasias/inmunología , Resistencia a Antineoplásicos/inmunología , Antígenos HLA/fisiología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia , Neoplasias/inmunología , Presentación de Antígeno , Antígenos de Neoplasias/genética , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/inmunología , Resistencia a Antineoplásicos/genética , Genes MHC Clase I , Genes MHC Clase II , Variación Genética , Genotipo , Mutación de Línea Germinal , Antígenos HLA/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Pronóstico , Receptor de Muerte Celular Programada 1/inmunología , Autotolerancia/genética , Autotolerancia/inmunología , Linfocitos T Citotóxicos/inmunología , Escape del Tumor
6.
Cancer Immunol Immunother ; 69(5): 683-687, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32152702

RESUMEN

More than 2000 immuno-oncology agents are being tested or are in use as a result of the cancer immunotherapy revolution. Manipulation of co-inhibitory receptors has achieved tumor eradication in a minority of patients, but widespread immune-related adverse events (irAEs) compromised tolerance to healthy self-tissues in the majority. We have proposed that a major mechanism of irAEs is similar to a graft-versus-malignancy effect of graft-versus-host disease. To verify our hypothesis, we retrieved post-marketing data of adverse events from the U.S. Food and Drug Administration Adverse Event Reporting System. A significant positive correlation was revealed in 7677 patients between the reporting odds ratio of irAEs during immune checkpoint inhibitor therapy and the corresponding tumor mutational burden across 19 cancer types. These results can be interpreted to mean that the ICI drugs unleashed T cells against "altered-self," self, and tumors resulting in better overall survival.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Neoplasias/tratamiento farmacológico , Autotolerancia/genética , Linfocitos T/efectos de los fármacos , Sistemas de Registro de Reacción Adversa a Medicamentos/estadística & datos numéricos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Humanos , Mutación , Neoplasias/genética , Neoplasias/inmunología , Autotolerancia/efectos de los fármacos , Linfocitos T/inmunología
7.
Cell Immunol ; 347: 103995, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31708111

RESUMEN

Graves' disease (GD) is the commonest cause of hyperthyroidism in populations with adequate iodine intake. It results from an abnormality in the immune system, which produces unique antibodies causing over production of thyroid hormones and glandular hyperplasia in individuals with genetic susceptibility. The Cytotoxic Lymphocyte Associated Antigen-4 (CTLA4) gene product serves the important function of immunomodulation, thereby helping in maintenance of peripheral self-tolerance. Studies on the association of the CTLA4 SNPs with GD have shown variations in the results from different populations. Since no such study has been carried out in ethnic Kashmiri population, we aimed to study a possible association of the CTLA4 SNPs (+49 A/G, -318C/T, CT 60 A/G and -1661 A/G) with GD. A total of 285 individuals (135 patients with GD and 150 healthy individuals) were genotyped using PCR-RFLP method and the results showed statistically significant differences in genotypic and allelic frequencies of cases and controls for + 49 A/G SNP (p=<0.001; OR = 5.14; CI = 2.17-12.19) and CT 60 A/G SNP (p = < 0.001; OR = 6.9; CI = 2.8-16.6), while -318C/T and -1661 A/G SNPs showed no significant association. We also studied the mRNA expression of the CTLA4 in patients with GD and healthy individuals by Real-Time PCR and found a decreased expression of the CTLA4 mRNA in PBMCs of patients with GD as compared to healthy controls with a -3.71-fold change. We conclude that the CTLA4 + 49 A/G and CT 60 A/G SNPs have a significant association with the risk of GD development in Kashmiri population and CTLA4 mRNA expression is significantly decreased in GD.


Asunto(s)
Antígeno CTLA-4/genética , Predisposición Genética a la Enfermedad/genética , Enfermedad de Graves/genética , Autotolerancia/genética , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes/genética , Genotipo , Enfermedad de Graves/inmunología , Humanos , India/etnología , Masculino , Persona de Mediana Edad , Polimorfismo de Longitud del Fragmento de Restricción/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Autotolerancia/inmunología , Hormonas Tiroideas/biosíntesis , Adulto Joven
8.
Front Immunol ; 10: 1695, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379878

RESUMEN

Graves' disease (GD) involves the presence of agonistic auto-antibodies against the thyrotropin receptor (TSHR), which are responsible for the clinical symptoms. While failure of TSHR tolerance is central to GD pathogenesis, the process leading to this failure remains poorly understood. Two mechanisms intimately linked to tolerance have been proposed to explain the association of SNPs located in TSHR intron 1 to GD: (1) differential alternative splicing in the thyroid; and (2) modulation of expression in the thymus. To elucidate the relative contribution to these two mechanisms to GD pathogenesis, we analyzed the level of full-length and ST4 and ST5 isoform expression in the thyroid (n = 49) and thymus (n = 39) glands, and the influence of intron 1-associated SNPs on such expression. The results show that: (1) the level of flTSHR and ST4 expression in the thymus was unexpectedly high (20% that of the thyroid); (2) while flTSHR is the predominant isoform, the levels are similar to ST4 (ratio flTSHR/ST4 = 1.34 in the thyroid and ratio flTSHR/ST4 in the thymus = 1.93); (3) next-generation sequencing confirmed the effect of the TSHR intron 1 polymorphism on TSHR expression in the thymus with a bias of 1.5 ± 0.2 overexpression of the protective allele in the thymus compared to the thyroid; (4) GD-associated intron 1 SNPs did not influence TSHR alternative splicing of ST4 and ST5 in the thyroid and thymus; and (5) three-color confocal imaging showed that TSHR is associated with both thymocytes, macrophages, and dendritic cells in the thymus. Our findings confirm the effect of intron 1 polymorphisms on thymic TSHR expression and we present evidence against an effect on the relative expression of isoforms. The high level of ST4 expression in the thymus and its distribution within the tissue suggest that this would most likely be the isoform that induces central tolerance to TSHR thus omitting most of the hinge and transmembrane portion. The lack of central tolerance to a large portion of TSHR may explain the relatively high frequency of autoimmunity related to TSHR and its clinical consequence, GD.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Enfermedad de Graves , Receptores de Tirotropina/biosíntesis , Autotolerancia , Timo , Glándula Tiroides , Empalme Alternativo , Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Humanos , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas , Receptores de Tirotropina/genética , Autotolerancia/genética , Autotolerancia/inmunología
9.
Nat Immunol ; 20(7): 879-889, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31182807

RESUMEN

CD8+ T cells and natural killer (NK) cells are central cellular components of immune responses against pathogens and cancer, which rely on interleukin (IL)-15 for homeostasis. Here we show that IL-15 also mediates homeostatic priming of CD8+ T cells for antigen-stimulated activation, which is controlled by a deubiquitinase, Otub1. IL-15 mediates membrane recruitment of Otub1, which inhibits ubiquitin-dependent activation of AKT, a kinase that is pivotal for T cell activation and metabolism. Otub1 deficiency in mice causes aberrant responses of CD8+ T cells to IL-15, rendering naive CD8+ T cells hypersensitive to antigen stimulation characterized by enhanced metabolic reprograming and effector functions. Otub1 also controls the maturation and activation of NK cells. Deletion of Otub1 profoundly enhances anticancer immunity by unleashing the activity of CD8+ T cells and NK cells. These findings suggest that Otub1 controls the activation of CD8+ T cells and NK cells by functioning as a checkpoint of IL-15-mediated priming.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Cisteína Endopeptidasas/metabolismo , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Cisteína Endopeptidasas/deficiencia , Enzimas Desubicuitinizantes/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-15/genética , Melanoma Experimental , Ratones , Ratones Transgénicos , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Interleucina-15/metabolismo , Autotolerancia/genética , Autotolerancia/inmunología , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T , Ubiquitinación
10.
Cancer Immunol Immunother ; 68(7): 1143-1155, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31177328

RESUMEN

Enhancement of endogenous immunity to tumor-associated self-antigens and neoantigens is the goal of preventive vaccination. Toward this goal, we compared the efficacy of the following HER2 DNA vaccine constructs: vaccines encoding wild-type HER2, hybrid HER2 vaccines consisting of human HER2 and rat Neu, HER2 vaccines with single residue substitutions and a novel human HER2 DNA vaccine, ph(es)E2TM. ph(es)E2TM was designed to contain five evolution-selected substitutions: M198V, Q398R, F425L, H473R and A622T that occur frequently in 12 primate HER2 sequences. These ph(es)E2TM substitutions score 0 to 1 in blocks substitutions matrix (BLOSUM), indicating minimal biochemical alterations. h(es)E2TM recombinant protein is recognized by a panel of anti-HER2 mAbs, demonstrating the preservation of HER2 protein structure. Compared to native human HER2, electrovaccination of HER2 transgenic mice with ph(es)E2TM induced a threefold increase in HER2-binding antibody (Ab) and elevated levels of IFNγ-producing T cells. ph(es)E2TM, but not pE2TM immune serum, recognized HER2 peptide p95 355LPESFDGDPASNTAP369, suggesting a broadening of epitope recognition induced by the minimally modified HER2 vaccine. ph(es)E2TM vaccination reduced tumor growth more effectively than wild-type HER2 or HER2 vaccines with more extensive modifications. The elevation of tumor immunity by ph(es)E2TM vaccination would create a favorable tumor microenvironment for neoantigen priming, further enhancing the protective immunity. The fundamental principle of exploiting evolution-selected amino acid substitutions is novel, effective and applicable to vaccine development in general.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Neoplasias Mamarias Experimentales/terapia , Receptor ErbB-2/inmunología , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Animales , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral/trasplante , Células Dendríticas/inmunología , Evolución Molecular , Femenino , Inmunogenicidad Vacunal/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptor ErbB-2/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Autotolerancia/genética , Microambiente Tumoral/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico
11.
Nature ; 565(7740): 495-499, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30626970

RESUMEN

Regulatory T cells (Treg cells), a distinct subset of CD4+ T cells, are necessary for the maintenance of immune self-tolerance and homeostasis1,2. Recent studies have demonstrated that Treg cells exhibit a unique metabolic profile, characterized by an increase in mitochondrial metabolism relative to other CD4+ effector subsets3,4. Furthermore, the Treg cell lineage-defining transcription factor, Foxp3, has been shown to promote respiration5,6; however, it remains unknown whether the mitochondrial respiratory chain is required for the T cell-suppression capacity, stability and survival of Treg cells. Here we report that Treg cell-specific ablation of mitochondrial respiratory chain complex III in mice results in the development of fatal inflammatory disease early in life, without affecting Treg cell number. Mice that lack mitochondrial complex III specifically in Treg cells displayed a loss of T cell-suppression capacity without altering Treg cell proliferation and survival. Treg cells deficient in complex III showed decreased expression of genes associated with Treg function, whereas Foxp3 expression remained stable. Loss of complex III in Treg cells increased DNA methylation as well as the metabolites 2-hydroxyglutarate (2-HG) and succinate that inhibit the ten-eleven translocation (TET) family of DNA demethylases7. Thus, Treg cells require mitochondrial complex III to maintain immune regulatory gene expression and suppressive function.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Mitocondrias/enzimología , Autotolerancia/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Desmetilación del ADN , Metilación de ADN , Transporte de Electrón , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Glutaratos/metabolismo , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Autotolerancia/genética , Ácido Succínico/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/enzimología
12.
Front Immunol ; 9: 1989, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319599

RESUMEN

Mammalian genomes encode a plethora of long non-coding RNA (lncRNA). These transcripts are thought to regulate gene expression, influencing biological processes from development to pathology. Results from the few lncRNA that have been studied in the context of the immune system have highlighted potentially critical functions as network regulators. Here we explored the nature of the lncRNA transcriptome in regulatory T cells (Tregs), a subset of CD4+ T cells required to establish and maintain immunological self-tolerance. The identified Treg lncRNA transcriptome showed distinct differences from that of non-regulatory CD4+ T cells, with evidence of direct shaping of the lncRNA transcriptome by Foxp3, the master transcription factor driving the distinct mRNA profile of Tregs. Treg lncRNA changes were disproportionally reversed in the absence of Foxp3, with an enrichment for colocalisation with Foxp3 DNA binding sites, indicating a direct coordination of transcription by Foxp3 independent of the mRNA coordination function. We further identified a novel lncRNA Flatr, as a member of the core Treg lncRNA transcriptome. Flatr expression anticipates Foxp3 expression during in vitro Treg conversion, and Flatr-deficient mice show a mild delay in in vitro and peripheral Treg induction. These results implicate Flatr as part of the upstream cascade leading to Treg conversion, and may provide clues as to the nature of this process.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , ARN Largo no Codificante/metabolismo , Autotolerancia/genética , Linfocitos T Reguladores/inmunología , Animales , Sistemas CRISPR-Cas/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Largo no Codificante/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo
13.
Front Immunol ; 9: 2249, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30349529

RESUMEN

Discussion of the antibody repertoire usually emphasizes diversity, but a conspicuous feature of the light chain repertoire is its lack of diversity. The diversity of reported allelic variants of germline light chain genes is also limited, even in well-studied species. In this review, the implications of this lack of diversity are considered. We explore germline and rearranged light chain genes in a variety of species, with a particular focus on human and mouse genes. The importance of the number, organization and orientation of the genes for the control of repertoire development is discussed, and we consider how primary rearrangements and receptor editing together shape the expressed light chain repertoire. The resulting repertoire is dominated by just a handful of IGKV and IGLV genes. It has been hypothesized that an important function of the light chain is to guard against self-reactivity, and the role of secondary rearrangements in this process could explain the genomic organization of the light chain genes. It could also explain why the light chain repertoire is so limited. Heavy and light chain genes may have co-evolved to ensure that suitable light chain partners are usually available for each heavy chain that forms early in B cell development. We suggest that the co-evolved loci of the house mouse often became separated during the inbreeding of laboratory mice, resulting in new pairings of loci that are derived from different sub-species of the house mouse. A resulting vulnerability to self-reactivity could explain at least some mouse models of autoimmune disease.


Asunto(s)
Anticuerpos/inmunología , Reordenamiento Génico/inmunología , Genes de las Cadenas Ligeras de las Inmunoglobulinas/inmunología , Cadenas Ligeras de Inmunoglobulina/inmunología , Ratones Endogámicos/inmunología , Receptores Inmunológicos/inmunología , Autotolerancia/inmunología , Animales , Anticuerpos/genética , Reordenamiento Génico/genética , Genes de las Cadenas Ligeras de las Inmunoglobulinas/genética , Variación Genética/genética , Variación Genética/inmunología , Cadenas Ligeras de Inmunoglobulina/genética , Ratones Endogámicos/clasificación , Ratones Endogámicos/genética , Receptores Inmunológicos/genética , Autotolerancia/genética , Especificidad de la Especie
14.
Rheumatol Int ; 38(10): 1763-1775, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29766256

RESUMEN

Systemic lupus erythematosus (SLE) is a complex disease with different genetic, immunologic, and environmental factors contributing to the pathogenesis. Monogenic SLE could help us understand the main phases of immune dysregulation in SLE. The aim of this review is to summarize the current knowledge on monogenic SLE with the implications of the respective genes on disease pathogenesis. A comprehensive literature search on monogenic SLE was conducted utilizing the Cochrane Library and MEDLINE/PubMed databases. The main affected pathways in disease pathogenesis are identified as follows: complement system, apoptosis, nucleic acid degradation, nucleic acid sensing, self-tolerance, and type I interferon production. Further studies on monogenic SLE can make precision medicine possible for SLE by increasing our understanding of disease pathogenesis.


Asunto(s)
Apoptosis , Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Ácidos Nucleicos/genética , Autotolerancia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Activación de Complemento/efectos de los fármacos , Activación de Complemento/genética , Proteínas del Sistema Complemento/genética , Predisposición Genética a la Enfermedad , Humanos , Factores Inmunológicos/uso terapéutico , Interferón Tipo I/genética , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ácidos Nucleicos/metabolismo , Fenotipo , Autotolerancia/efectos de los fármacos , Autotolerancia/genética
15.
Curr Stem Cell Res Ther ; 13(5): 336-344, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-26832137

RESUMEN

MicroRNAs are small non-coding RNAs that can modulate gene expression at posttranscriptional level, and they participate in almost all important biological processes. Immune system is elaborately regulated to maintain the equilibrium between immunity and tolerance. Recent studies have revealed significant functions of microRNAs in the maintenance of immune homeostasis using both cell and transgenic mouse models. In collaboration with various transcriptional factors and cytokines, microRNAs constitute an effective and flexible regulatory network governing the development and activation of immune cells; as well as maintenance of immune tolerance. In this review, microRNAs involved in T cell development, proliferation, and lineage differentiation will be summarized. Based on current knowledge, the function of microRNAs in establishing and maintaining immune tolerance will also be discussed in relation to determining the outcome of allograft transplantation.


Asunto(s)
Aloinjertos/inmunología , Inmunidad Innata/genética , MicroARNs/fisiología , Procesamiento Postranscripcional del ARN , Autotolerancia/genética , Animales , Homeostasis , Humanos , Inmunidad Innata/inmunología , MicroARNs/antagonistas & inhibidores , MicroARNs/clasificación , Modelos Animales , Autotolerancia/inmunología , Linfocitos T/inmunología , Linfocitos T/fisiología , Tolerancia al Trasplante/genética , Tolerancia al Trasplante/inmunología , Trasplante Homólogo
16.
J Exp Med ; 214(7): 1925-1935, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28611158

RESUMEN

The chemokine receptor CCR7 directs T cell relocation into and within lymphoid organs, including the migration of developing thymocytes into the thymic medulla. However, how three functional CCR7 ligands in mouse, CCL19, CCL21Ser, and CCL21Leu, divide their roles in immune organs is unclear. By producing mice specifically deficient in CCL21Ser, we show that CCL21Ser is essential for the accumulation of positively selected thymocytes in the thymic medulla. CCL21Ser-deficient mice were impaired in the medullary deletion of self-reactive thymocytes and developed autoimmune dacryoadenitis. T cell accumulation in the lymph nodes was also defective. These results indicate a nonredundant role of CCL21Ser in the establishment of self-tolerance in T cells in the thymic medulla, and reveal a functional inequality among CCR7 ligands in vivo.


Asunto(s)
Tolerancia Central/inmunología , Quimiocina CCL21/inmunología , Autotolerancia/inmunología , Linfocitos T/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Tolerancia Central/genética , Quimiocina CCL21/genética , Quimiocina CCL21/metabolismo , Dacriocistitis/genética , Dacriocistitis/inmunología , Dacriocistitis/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Microscopía Confocal , Receptores CCR7/inmunología , Receptores CCR7/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Autotolerancia/genética , Linfocitos T/metabolismo , Timocitos/inmunología , Timocitos/metabolismo , Timo/inmunología , Timo/metabolismo
17.
Curr Opin Immunol ; 46: 53-57, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28477557

RESUMEN

The processing and presentation of major histocompatibility complex (MHC)-associated antigens depend on the intracellular digestion of self- and nonself-proteins, the loading of digested peptides onto MHC molecules, and the traffic of peptide-MHC complexes to plasma membrane surface for display to interacting T cells. Recent studies have revealed unique machineries for antigen processing and presentation in thymic antigen-presenting cells that display self-antigens to developing thymocytes for the formation of functionally competent yet self-tolerant T cell repertoire. Here, we briefly summarize those machineries, focusing on the biology of cortical and medullary thymic epithelial cells.


Asunto(s)
Presentación de Antígeno/inmunología , Selección Clonal Mediada por Antígenos/inmunología , Subgrupos de Linfocitos T/inmunología , Timo/inmunología , Animales , Presentación de Antígeno/genética , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Selección Clonal Mediada por Antígenos/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Transporte de Proteínas , Proteolisis , Autotolerancia/genética , Autotolerancia/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo
18.
Rheumatology (Oxford) ; 56(suppl_1): i55-i66, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28375453

RESUMEN

SLE is a chronic autoimmune disease caused by perturbations of the immune system. The clinical presentation is heterogeneous, largely because of the multiple genetic and environmental factors that contribute to disease initiation and progression. Over the last 60 years, there have been a number of significant leaps in our understanding of the immunological mechanisms driving disease processes. We now know that multiple leucocyte subsets, together with inflammatory cytokines, chemokines and regulatory mediators that are normally involved in host protection from invading pathogens, contribute to the inflammatory events leading to tissue destruction and organ failure. In this broad overview, we discuss the main pathways involved in SLE and highlight new findings. We describe the immunological changes that characterize this form of autoimmunity. The major leucocytes that are essential for disease progression are discussed, together with key mediators that propagate the immune response and drive the inflammatory response in SLE.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Citocinas/inmunología , Ambiente , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología , Autoinmunidad/genética , Predisposición Genética a la Enfermedad , Humanos , Inflamación/genética , Lupus Eritematoso Sistémico/genética , Autotolerancia/genética , Autotolerancia/inmunología
19.
Nat Commun ; 7: 13381, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27830696

RESUMEN

Self-tolerance by clonal anergy of B cells is marked by an increase in IgD and decrease in IgM antigen receptor surface expression, yet the function of IgD on anergic cells is obscure. Here we define the RNA landscape of the in vivo anergy response, comprising 220 induced sequences including a core set of 97. Failure to co-express IgD with IgM decreases overall expression of receptors for self-antigen, but paradoxically increases the core anergy response, exemplified by increased Sdc1 encoding the cell surface marker syndecan-1. IgD expressed on its own is nevertheless competent to induce calcium signalling and the core anergy mRNA response. Syndecan-1 induction correlates with reduction of surface IgM and is exaggerated without surface IgD in many transitional and mature B cells. These results show that IgD attenuates the response to self-antigen in anergic cells and promotes their accumulation. In this way, IgD minimizes tolerance-induced holes in the pre-immune antibody repertoire.


Asunto(s)
Linfocitos B/inmunología , Anergia Clonal/inmunología , Inmunoglobulina D/inmunología , Inmunoglobulina M/inmunología , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Señalización del Calcio/genética , Señalización del Calcio/inmunología , Anergia Clonal/genética , Perfilación de la Expresión Génica/métodos , Inmunoglobulina D/genética , Inmunoglobulina M/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones Endogámicos C57BL , Mutación , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Autotolerancia/genética , Autotolerancia/inmunología , Sindecano-1/genética , Sindecano-1/inmunología , Sindecano-1/metabolismo
20.
Cell ; 166(3): 530-531, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471960

RESUMEN

Meyer et al. find that subjects lacking the AIRE gene, critical for self-tolerance in T lymphocytes, show a broad range of autoantibody specificities, which can have extremely high affinities. The data also suggest that some of these autoantibodies can, surprisingly, prevent some types of autoimmunity, particularly type I diabetes.


Asunto(s)
Autoinmunidad/inmunología , Autotolerancia/genética , Autoanticuerpos/inmunología , Humanos , Tolerancia Inmunológica , Linfocitos T/inmunología , Timo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA