Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 296: 100774, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33992646

RESUMEN

Cathepsin H (CTSH) is a type 1 diabetes (T1D) risk gene; large-scale genetic and epidemiological studies found that T1D genetic risk correlates with high CTSH expression, rapid decline of beta-cell function, and early onset T1D. Counterintuitively, transcriptional downregulation of CTSH by proinflammatory cytokines has been shown to promote beta-cell apoptosis. Here, we potentially explain these observed contrasting effects, describing a new mechanism where proinflammatory cytokines and T1D genetic risk variants regulate CTSH transcription via differential DNA methylation. We show that, in human islets, CTSH downregulation by the proinflammatory cytokine cocktail interleukin 1ß + tumor necrosis factor α + interferon γ was coupled with DNA hypermethylation in an open chromatin region in CTSH intron 1. A luciferase assay in human embryonic kidney 293 cells revealed that methylation of three key cytosine-phosphate-guanine dinucleotide (CpG) residues in intron 1 was responsible for the reduction of promoter activity. We further found that cytokine-induced intron 1 hypermethylation is caused by lowered Tet1/3 activities, suggesting that attenuated active demethylation lowered CTSH transcription. Importantly, individuals who carry the T1D risk variant showed lower methylation variability at the intron 1 CpG residues, presumably making them less sensitive to cytokines, whereas individuals who carry the protective variant showed higher methylation variability, presumably making them more sensitive to cytokines and implying differential responses to environment between the two patient populations. These findings suggest that genetic and environmental influences on a T1D locus are mediated by differential variability and mean of DNA methylation.


Asunto(s)
Catepsina H/genética , Metilación de ADN , Diabetes Mellitus Tipo 1/genética , Epigénesis Genética , Islas de CpG , Interacción Gen-Ambiente , Humanos
2.
Telemed J E Health ; 27(8): 934-938, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33600728

RESUMEN

Telemedicine has been widely implemented during the coronavirus disease 2019 (COVID-19) pandemic; however, its impact on those providing care remains largely understudied. Provider documentation data collected by the electronic health record (EHR) represents an underutilized tool for assessing the provider experience. Through Epic Signal, we collected data regarding the actions logged in the EHR by health care providers of the Montefiore Health System (Bronx, NY) before and after the implementation of telemedicine during the pandemic. Focusing on five metrics (appointments per day, visits closed same day, time spent outside 7 AM-7 PM, time spent on unscheduled days, and pajama time), we performed a preliminary analysis of providers across the institution, by specialty, and according to demographic characteristics such as gender and years since graduation. We observed that after telemedicine implementation, a greater proportion of providers had fewer appointments per day, closed more notes same day, and spent less time in the EHR outside of normal working hours for each of the time-related metrics. We additionally found that providers who graduated longer ago as well as female providers spent more time documenting in the EHR after hours. This brief analysis highlights the potential of using EHR data to inform decisions based on provider well-being, specifically in the setting of telemedicine implementation.


Asunto(s)
COVID-19 , Telemedicina , Registros Electrónicos de Salud , Femenino , Humanos , Pandemias , SARS-CoV-2
3.
J Autoimmun ; 108: 102402, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980336

RESUMEN

We have previously identified a signature HLA-DR3 pocket variant, designated HLA-DRß1-Arg74 that confers a high risk for Graves' Disease (GD). In view of the key role of HLA-DRß1-Arg74 in triggering GD we hypothesized that thyroid-stimulating hormone receptor (TSHR) peptides that bind to the HLA-DRß1-Arg74 pocket with high affinity represent key pathogenic TSHR peptides triggering GD, and that blocking their presentation to CD4+ T-cells can be used as a novel therapeutic approach in GD. There were several previous attempts to identify the major pathogenic TSHR peptide utilizing different methodologies, however the results were inconsistent and inconclusive. Therefore, the aim of our study was to use TSHR peptide binding affinity to HLA-DRß1-Arg74 as a method to identify the key pathogenic TSHR peptides that trigger GD. Using virtual screening and ELISA and cellular binding assays we identified 2 TSHR peptides that bound with high affinity to HLA-DRß1-Arg74 - TSHR.132 and TSHR.197. Peptide immunization studies in humanized DR3 mice showed that only TSHR.132, but not TSHR.197, induced autoreactive T-cell proliferation and cytokine responses. Next, we induced experimental autoimmune Graves' disease (EAGD) in a novel BALB/c-DR3 humanized mouse model we created and confirmed TSHR.132 as a major DRß1-Arg74 binding peptide triggering GD in our mouse model. Furthermore, we demonstrated that Cepharanthine, a compound we have previously identified as DRß1-Arg74 blocker, could block the presentation and T-cell responses to TSHR.132 in the EAGD model.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Bencilisoquinolinas/farmacología , Antígeno HLA-DR3/inmunología , Péptidos/antagonistas & inhibidores , Péptidos/inmunología , Receptores de Tirotropina/inmunología , Secuencia de Aminoácidos , Animales , Bencilisoquinolinas/química , Mapeo Epitopo/métodos , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/tratamiento farmacológico , Enfermedad de Graves/inmunología , Antígeno HLA-DR3/genética , Humanos , Inmunohistoquímica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Péptidos/química , Unión Proteica , Receptores de Tirotropina/química , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
J Autoimmun ; 115: 102543, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32951964

RESUMEN

Over the past four decades, the number of people with Type 1 Diabetes (T1D) has increased by 4% per year, making it an important public health challenge. Currently, no curative therapy exists for T1D and the only available treatment is insulin replacement. HLA-DQ8 has been shown to present antigenic islet peptides driving the activation of CD4+ T-cells in T1D patients. Specifically, the insulin peptide InsB:9-23 activates self-reactive CD4+ T-cells, causing pancreatic beta cell destruction. The aim of the current study was to identify retro-inverso-d-amino acid based peptides (RI-D-peptides) that can suppress T-cell activation by blocking the presentation of InsB:9-23 peptide within HLA-DQ8 pocket. We identified a RI-D-peptide (RI-EXT) that inhibited InsB:9-23 binding to recombinant HLA-DQ8 molecule, as well as its binding to DQ8 expressed on human B-cells. RI-EXT prevented T-cell activation in a cellular antigen presentation assay containing human DQ8 cells loaded with InsB:9-23 peptide and murine T-cells expressing a human T-cell receptor specific for the InsB:9-23-DQ8 complex. Moreover, RI-EXT blocked T-cell activation by InsB:9-23 in a humanized DQ8 mice both ex vivo and in vivo, as shown by decreased production of IL-2 and IFN-γ and reduced lymphocyte proliferation. Interestingly, RI-EXT also blocked lymphocyte activation and proliferation by InsB:9-23 in PBMCs isolated from recent onset DQ8-T1D patients. In summary, we discovered a RI-D-peptide that blocks InsB:9-23 binding to HLA-DQ8 and its presentation to T-cells in T1D. These findings set the stage for using our approach as a novel therapy for patients with T1D and potentially other autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Antígenos HLA-DQ/metabolismo , Células Secretoras de Insulina/inmunología , Péptidos/antagonistas & inhibidores , Animales , Presentación de Antígeno/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Antígenos HLA-DQ/inmunología , Antígenos HLA-DQ/aislamiento & purificación , Humanos , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Simulación de Dinámica Molecular , Péptidos/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
5.
J Gen Intern Med ; 35(Suppl 2): 832-838, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32779140

RESUMEN

BACKGROUND: eConsult programs have been instituted to increase access to specialty expertise. Opt-in choice eConsult programs maintain primary care physician (PCP) autonomy to decide whether to utilize eConsults versus traditional specialty referrals, but little is known about how this intervention may impact PCP eConsult adoption and traditional referral demand. OBJECTIVE: We assessed the feasibility of implementing an opt-in choice eConsult program and examined whether this intervention reduces demand for in-person visits for primary care patients requiring specialty expertise. DESIGN: Stepped-wedge, cluster randomized trial conducted from July 2018 to June 2019. PARTICIPANTS: Sixteen primary care practices in a large, urban academic health care system. INTERVENTION: Our intervention was an opt-in choice eConsult available in addition to traditional specialty referral; our implementation strategy included in-person training, audit and feedback, and incentive payments. MAIN MEASURES: Our implementation outcome measure was the eConsult rate: weekly proportion of eConsults per PCP visit at each site. Our intervention outcome measure was traditional referral rate: weekly proportion of referrals per PCP visit at each site. We also assessed PCP experiences with questionnaires. KEY RESULTS: Of 305,915 in-person PCP visits, there were 31,510 traditional referrals to specialties participating in the eConsult program, and 679 eConsults. All but one primary care site utilized the opt-in choice eConsult program, with a weekly rate of 0.05 eConsults per 100 PCP visits by the end of the study period. The weekly rate of traditional referrals was 11 per 100 PCP visits at the end of the study period; this represents a significant increase in traditional referral rate after implementation of eConsults. PCPs were generally satisfied with the eConsult program and valued prompt provider-to-provider communication. CONCLUSIONS: Implementation of an opt-in choice eConsult program resulted in widespread PCP adoption; however, this did not decrease the demand for traditional referrals. Future studies should evaluate different strategies to incentivize and increase eConsult utilization while maintaining PCP choice.


Asunto(s)
Medicina , Médicos de Atención Primaria , Humanos , Atención Primaria de Salud , Derivación y Consulta , Encuestas y Cuestionarios
6.
J Autoimmun ; 94: 7-15, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30115527

RESUMEN

IFNα is a cytokine essential to a vast array of immunologic processes. Its induction early in the innate immune response provides a priming mechanism that orchestrates numerous subsequent pathways in innate and adaptive immunity. Despite its beneficial effects in viral infections IFNα has been reported to be associated with several autoimmune diseases including autoimmune thyroid disease, systemic lupus erythematosus, rheumatoid arthritis, primary biliary cholangitis, and recently emerged as a major cytokine that triggers Type 1 Diabetes. In this review, we dissect the role of IFNα in T1D, focusing on the potential pathophysiological mechanisms involved. Evidence from human and mouse studies indicates that IFNα plays a key role in enhancing islet expression of HLA-I in patients with T1D, thereby increasing autoantigen presentation and beta cell activation of autoreactive cytotoxic CD8 T-lymphocytes. The binding of IFNα to its receptor induces the secretion of chemokines, attracting monocytes, T lymphocytes, and NK cells to the infected tissue triggering autoimmunity in susceptible individuals. Furthermore, IFNα impairs insulin production through the induction of endoplasmic reticulum stress as well as by impairing mitochondrial function. Due to its central role in the early phases of beta cell death, targeting IFNα and its pathways in genetically predisposed individuals may represent a potential novel therapeutic strategy in the very early stages of T1D.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Diabetes Mellitus Tipo 1/inmunología , Regulación de la Expresión Génica/inmunología , Células Secretoras de Insulina/inmunología , Interferón-alfa/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Autoantígenos/inmunología , Autoinmunidad/efectos de los fármacos , Quimiocinas/genética , Quimiocinas/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/inmunología , Humanos , Insulina/agonistas , Insulina/biosíntesis , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/genética , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/inmunología , Terapia Molecular Dirigida/métodos , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/genética , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
7.
J Biol Chem ; 291(8): 4079-90, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703475

RESUMEN

We previously showed that an HLA-DR variant containing arginine at position 74 of the DRß1 chain (DRß1-Arg74) is the specific HLA class II variant conferring risk for autoimmune thyroid diseases (AITD). We also identified 5 thyroglobulin (Tg) peptides that bound to DRß1-Arg74. We hypothesized that blocking the binding of these peptides to DRß1-Arg74 could block the continuous T-cell activation in thyroiditis needed to maintain the autoimmune response to the thyroid. The aim of the current study was to identify small molecules that can block T-cell activation by Tg peptides presented within DRß1-Arg74 pockets. We screened a large and diverse library of compounds and identified one compound, cepharanthine that was able to block peptide binding to DRß1-Arg74. We then showed that Tg.2098 is the dominant peptide when inducing experimental autoimmune thyroiditis (EAT) in NOD mice expressing human DRß1-Arg74. Furthermore, cepharanthine blocked T-cell activation by thyroglobulin peptides, in particular Tg.2098 in mice that were induced with EAT. For the first time we identified a small molecule that can block Tg peptide binding and presentation to T-cells in autoimmune thyroiditis. If confirmed cepharanthine could potentially have a role in treating human AITD.


Asunto(s)
Alcaloides/farmacología , Presentación de Antígeno/efectos de los fármacos , Cadenas HLA-DRB1/inmunología , Tiroiditis Autoinmune/inmunología , Alcaloides/química , Animales , Cadenas HLA-DRB1/genética , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Transgénicos , Péptidos/genética , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Tiroglobulina/genética , Tiroglobulina/inmunología , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/patología
8.
J Autoimmun ; 80: 48-55, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28238527

RESUMEN

Despite substantial advances in the research exploring the pathogenesis of Type 1 Diabetes (T1D), the pathophysiological mechanisms involved remain unknown. We hypothesized in this study that interferon alpha (IFNα) participates in the early stages of T1D development by triggering endoplasmic reticulum (ER) stress. To verify our hypothesis, human islets and human EndoC-ßH1 cells were exposed to IFNα and tested for ER stress markers, glucose stimulated insulin secretion (GSIS) and insulin content. IFNα treatment induced upregulation of ER stress markers including Binding immunoglobulin Protein, phospho-eukaryotic translation initiation factor 2α, spliced- X-box binding protein-1, C/EBP homologous protein and activating transcription factor 4. Intriguingly, IFNα treatment did not impair GSIS but significantly decreased insulin production in both human islets and EndoC-ßH1 cells. Furthermore, IFNα decreased the expression of both proinsulin convertase 1 and proinsulin convertase 2, suggesting an altered functional state of the beta cells characterized by a slower proinsulin-insulin conversion. Pretreatment of both human islets and EndoC-ßH1 cells with chemical chaperones 4-phenylbutyric acid and tauroursodeoxycholic acid completely prevented IFNα effects, indicating an ER stress-mediated impairment of insulin production. We demonstrated for the first time that IFNα elicits ER stress in human beta cells providing a novel mechanistic role for this virus-induced cytokine in the development of T1D. Compounds targeting molecular processes altered in ER-stressed beta cells could represent a potential therapeutic strategy to prevent IFNα-induced beta cell dysfunction in the early onset of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Interferón-alfa/metabolismo , Apoptosis , Células Cultivadas , Citocinas/metabolismo , Humanos , Fenilbutiratos/farmacología , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 2/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Factor de Transcripción CHOP/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo
9.
J Autoimmun ; 76: 1-9, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27670087

RESUMEN

Autoimmune polyglandular syndrome 3 variant (APS3v) refers to the co-occurrence of autoimmune thyroiditis (AITD) and type 1 diabetes (T1D) within the same individual. HLA class II confers the strongest susceptibility to APS3v. We previously identified a unique amino acid signature of the HLA-DR pocket (designated APS3v HLA-DR pocket) that predisposes to APS3v. We hypothesized that both thyroid and islet peptides can be presented by the unique APS3v HLA-DR pocket, triggering AITD + T1D together. To test this hypothesis we screened islet and thyroid peptides for their ability to bind to the APS3v HLA-DR pocket. Virtual screen of all possible thyroglobulin (Tg), thyroid-stimulating hormone receptor (TSHR), thyroid peroxidase (TPO), insulin (Ins), and glutamic acid decarboxylase 65 (GAD65) peptides identified 36 peptides that bound to this unique pocket. In vitro binding assays using baculovirus-produced recombinant APS3v HLA-DR identified 11 thyroid/islet peptides (of the 36 predicted binders) that bound with high affinity. By immunizing humanized HLA-DR3 mice carrying the APS3v HLA-DR pocket we identified 4 peptides (Tg.1571, GAD.492, TPO.758, TPO.338) that were presented by antigen presenting cells and elicited T-cell response. We conclude that both thyroid and islet peptides can bind to this flexible APS3v HLA-DR pocket and induce thyroid and islet specific T-cell responses. These findings set the stage to developing specific inhibitors of the APS3v HLA-DR pocket as a precision medicine approach to treating or preventing APS3v in patients that carry this genetic HLA-DR pocket variant.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA-DR/inmunología , Péptidos/inmunología , Linfocitos T/inmunología , Tiroiditis Autoinmune/inmunología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Antígenos HLA-DR/química , Antígenos HLA-DR/metabolismo , Humanos , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo , Unión Proteica/inmunología , Conformación Proteica , Linfocitos T/metabolismo , Glándula Tiroides/inmunología , Tiroiditis Autoinmune/metabolismo
10.
Virol J ; 14(1): 237, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29258547

RESUMEN

BACKGROUND: An association between hepatitis C virus (HCV) and type 2 diabetes (T2D) is supported by numerous epidemiologic studies. We hypothesized that HCV could infect human pancreatic islet cells in vitro. METHODS: Measures of HCV RNA synthesis and protein production were used to evaluate HCV infection of pancreatic islets recovered from human donors. RESULTS: Significant co-staining of insulin and the HCV entry factor CD81 was observed in pancreatic islets. Positive- and negative-sense HCV RNA were detected in HCV-exposed islets at days 1, 3, 7, and 14 post-infection. The HCV core and NS3 proteins were expressed and increased with time providing further evidence of viral replication. Interferon and an HCV polymerase inhibitor reduced viral replication in islet cells. In HCV-infected islets, TNFα levels were elevated at days 1, 3, and 7 post-infection, while IL-6 levels were elevated at day 1 but not days 3 or 7. Overall, the expression of miR-122 was low in islets compared to the Huh7.5 hepatocyte-derived cell line, although the relative expression of miR-122 increased in islet cells after viral infection (1, 6.63, and 5.83 at days 1, 3, and 7, respectively). CONCLUSIONS: In this pilot study, viral infection was demonstrated in pancreatic islet cells from multiple donors using complementary measures of viral replication, thus providing evidence of in vitro infection. Altered cytokine expression may contribute to the development of insulin deficiency, and understanding the etiology of diabetes in individuals with HCV infection may facilitate the development of novel treatment modalities and prevention strategies. This in vitro system provides an important model for mechanistic studies of HCV-pancreas interactions and facilitates future studies of the potential impact of viral infection on islet cell function.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/virología , Islotes Pancreáticos/virología , Antivirales/farmacología , Células Cultivadas , Citocinas/metabolismo , Células HEK293 , Hepacivirus/efectos de los fármacos , Hepatitis C/sangre , Hepatocitos , Humanos , Interferón-alfa/farmacología , Islotes Pancreáticos/metabolismo , MicroARNs/metabolismo , ARN Viral , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sofosbuvir/farmacología , Proteínas no Estructurales Virales/metabolismo , Virión/fisiología , Internalización del Virus , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
11.
Proc Natl Acad Sci U S A ; 111(34): 12562-7, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25122677

RESUMEN

Graves disease (GD) is an autoimmune condition caused by interacting genetic and environmental factors. Genetic studies have mapped several single-nucleotide polymorphisms (SNPs) that are strongly associated with GD, but the mechanisms by which they trigger disease are unknown. We hypothesized that epigenetic modifications induced by microenvironmental influences of cytokines can reveal the functionality of GD-associated SNPs. We analyzed genome-wide histone H3 lysine 4 methylation and gene expression in thyroid cells induced by IFNα, a key cytokine secreted during viral infections, and overlapped them with known GD-associated SNPs. We mapped an open chromatin region overlapping two adjacent GD-associated SNPs (rs12101255 and rs12101261) in intron 1 of the thyroid stimulating hormone receptor (TSHR) gene. We then demonstrated that this region functions as a regulatory element through binding of the transcriptional repressor promyelocytic leukemia zinc finger protein (PLZF) at the rs12101261 site. Repression by PLZF depended on the rs12101261 disease susceptibility allele and was increased by IFNα. Intrathymic TSHR expression was decreased in individuals homozygous for the rs12101261 disease-associated genotype compared with carriers of the disease-protective allele. Our studies discovered a genetic-epigenetic interaction involving a noncoding SNP in the TSHR gene that regulates thymic TSHR gene expression and facilitates escape of TSHR-reactive T cells from central tolerance, triggering GD.


Asunto(s)
Autoinmunidad/genética , Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Receptores de Tirotropina/genética , Receptores de Tirotropina/inmunología , Sitios de Unión/genética , Línea Celular , Epigénesis Genética , Regulación de la Expresión Génica , Interacción Gen-Ambiente , Enfermedad de Graves/etiología , Histonas/química , Histonas/metabolismo , Humanos , Interferón-alfa/inmunología , Interferón-alfa/farmacología , Intrones , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Metilación , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Proteína de la Leucemia Promielocítica con Dedos de Zinc , ARN Mensajero/genética , ARN Mensajero/metabolismo , Timo/inmunología , Timo/metabolismo , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo
12.
J Autoimmun ; 64: 82-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26235382

RESUMEN

Both environmental and genetic triggers factor into the etiology of autoimmune thyroid disease (AITD), including Graves' disease (GD) and Hashimoto's thyroiditis (HT). Although the exact pathogenesis and causative interaction between environment and genes are unknown, GD and HT share similar immune-mediated mechanisms of disease. They both are characterized by the production of thyroid autoantibodies and by thyroidal lymphocytic infiltration, despite being clinically distinct entities with thyrotoxicosis in GD and hypothyroidism in HT. Family and population studies confirm the strong genetic influence and inheritability in the development of AITD. AITD susceptibility genes can be categorized as either thyroid specific (Tg, TSHR) or immune-modulating (FOXP3, CD25, CD40, CTLA-4, HLA), with HLA-DR3 carrying the highest risk. Of the AITD susceptibility genes, FOXP3 and CD25 play critical roles in the establishment of peripheral tolerance while CD40, CTLA-4, and the HLA genes are pivotal for T lymphocyte activation and antigen presentation. Polymorphisms in these immune-modulating genes, in particular, significantly contribute to the predisposition for GD, HT and, unsurprisingly, other autoimmune diseases. Emerging evidence suggests that single nucleotide polymorphisms (SNPs) in the immunoregulatory genes may functionally hinder the proper development of central and peripheral tolerance and alter T cell interactions with antigen presenting cells (APCs) in the immunological synapse. Thus, susceptibility genes for AITD contribute directly to the key mechanism underlying the development of organ-specific autoimmunity, namely the breakdown in self-tolerance. Here we review the major immune-modulating genes that are associated with AITD and their potential functional effects on thyroidal immune dysregulation.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Predisposición Genética a la Enfermedad , Inmunogenética , Enfermedades de la Tiroides/genética , Enfermedades de la Tiroides/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Sitios Genéticos , Humanos , Tolerancia Inmunológica/genética , Fenotipo , Enfermedades de la Tiroides/diagnóstico , Enfermedades de la Tiroides/metabolismo
13.
J Autoimmun ; 60: 32-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25936594

RESUMEN

Autoimmune thyroid diseases (AITD) and Type 1 diabetes (T1D) frequently occur in the same individual pointing to a strong shared genetic susceptibility. Indeed, the co-occurrence of T1D and AITD in the same individual is classified as a variant of the autoimmune polyglandular syndrome type 3 (designated APS3v). Our aim was to identify new genes and mechanisms causing the co-occurrence of T1D + AITD (APS3v) in the same individual using a genome-wide approach. For our discovery set we analyzed 346 Caucasian APS3v patients and 727 gender and ethnicity matched healthy controls. Genotyping was performed using the Illumina Human660W-Quad.v1. The replication set included 185 APS3v patients and 340 controls. Association analyses were performed using the PLINK program, and pathway analyses were performed using the MAGENTA software. We identified multiple signals within the HLA region and conditioning studies suggested that a few of them contributed independently to the strong association of the HLA locus with APS3v. Outside the HLA region, variants in GPR103, a gene not suggested by previous studies of APS3v, T1D, or AITD, showed genome-wide significance (p < 5 × 10(-8)). In addition, a locus on 1p13 containing the PTPN22 gene showed genome-wide significant associations. Pathway analysis demonstrated that cell cycle, B-cell development, CD40, and CTLA-4 signaling were the major pathways contributing to the pathogenesis of APS3v. These findings suggest that complex mechanisms involving T-cell and B-cell pathways are involved in the strong genetic association between AITD and T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Poliendocrinopatías Autoinmunes/genética , Tiroiditis Autoinmune/genética , Linfocitos B/inmunología , Antígenos CD40/genética , Antígeno CTLA-4/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Desequilibrio de Ligamiento/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Linfocitos T/inmunología
14.
J Autoimmun ; 50: 33-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24210274

RESUMEN

Type 1 diabetes (T1D) shows ∼40% concordance rate in monozygotic twins (MZ) suggesting a role for environmental factors and/or epigenetic modifications in the etiology of the disease. The aim of our study was to dissect the contribution of epigenetic factors, particularly, DNA methylation (DNAm), to the incomplete penetrance of T1D. We performed DNAm profiling in lymphocyte cell lines from 3 monozygotic (MZ) twin pairs discordant for T1D and 6 MZ twin pairs concordant for the disease using HumanMethylation27 BeadChip. This assay assesses the methylation state of 27,578 CpG sites, mostly located within proximal promoter regions. We identified 88 CpG sites displaying significant methylation changes in all T1D-discordant MZ twin pairs. Functional annotation of the genes with distinct CpG methylation profiles in T1D samples showed differential DNAm of immune response and defense response pathways between affected and unaffected twins. Integration of DNAm data with GWAS data mapped several known T1D associated genes, HLA, INS, IL-2RB, CD226, which showed significant differences in DNAm between affected and unaffected of twins. Our findings suggest that abnormalities of DNA methylation patterns, known to regulate gene transcription, may be involved in the pathogenesis of T1D.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 1/genética , Epigénesis Genética , Predisposición Genética a la Enfermedad , Adolescente , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Niño , Preescolar , Islas de CpG , Diabetes Mellitus Tipo 1/inmunología , Femenino , Perfilación de la Expresión Génica , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Lactante , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/inmunología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transcripción Genética , Gemelos Monocigóticos
15.
J Immunol ; 189(6): 3043-53, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22888137

RESUMEN

The CD40 gene, an important immune regulatory gene, is also expressed and functional on nonmyeloid-derived cells, many of which are targets for tissue-specific autoimmune diseases, including ß cells in type 1 diabetes, intestinal epithelial cells in Crohn's disease, and thyroid follicular cells in Graves' disease (GD). Whether target tissue CD40 expression plays a role in autoimmune disease etiology has yet to be determined. In this study, we show that target tissue overexpression of CD40 plays a key role in the etiology of autoimmunity. Using a murine model of GD, we demonstrated that thyroidal CD40 overexpression augmented the production of thyroid-specific Abs, resulting in more severe experimental autoimmune GD (EAGD), whereas deletion of thyroidal CD40 suppressed disease. Using transcriptome and immune-pathway analyses, we showed that in both EAGD mouse thyroids and human primary thyrocytes, CD40 mediates this effect by activating downstream cytokines and chemokines, most notably IL-6. To translate these findings into therapy, we blocked IL-6 during EAGD induction in the setting of thyroidal CD40 overexpression and showed decreased levels of thyroid stimulating hormone receptor-stimulating Abs and frequency of disease. We conclude that target tissue overexpression of CD40 plays a key role in the etiology of organ-specific autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Antígenos CD40/genética , Marcación de Gen/métodos , Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/prevención & control , Antígenos CD40/biosíntesis , Antígenos CD40/deficiencia , Células Cultivadas , Modelos Animales de Enfermedad , Enfermedad de Graves/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Cultivo Primario de Células , Quimera por Radiación/inmunología , Receptores de Tirotropina/inmunología , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/patología
16.
Curr Protoc ; 4(3): e938, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38436133

RESUMEN

The main challenge in the "post-GWAS" era is to determine the functional meaning of genetic variants and their contribution to disease pathogenesis. Development of suitable mouse models is critical because disease susceptibility is triggered by complex interactions between genetic, epigenetic, and environmental factors that cannot be modeled by in vitro models. Thyroglobulin (TG) is a key gene for autoimmune thyroid disease (AITD) and several single nucleotide polymorphisms (SNPs) in the TG coding region have been associated with AITD. The classical model of experimental autoimmune thyroiditis (EAT), based on immunization of genetically susceptible mouse strains with purified TG protein in adjuvant, does not allow testing the impact of TG sequence variants on the development of autoimmune thyroiditis. Here we describe a protocol for the induction of EAT by immunization of mice susceptible to thyroiditis with an adenovirus vector carrying full-length human TG cDNA (Ad-TG EAT). We also provide support protocols for evaluation of autoimmune thyroiditis including serological assessment of TG antibodies, in vitro splenocyte proliferation assay and cytokines secretion, thyroid histology, and evaluation of thyroid lymphocytic infiltration by immunostaining. This protocol for EAT induction allows manipulation of the TG cDNA to introduce variants associated with AITD, enabling the testing of the functional effects of susceptible variants and their haplotypes on the immunogenicity of TG. Furthermore, the Ad-TG EAT mouse model is a valuable model for studying the interactions of the TG variants with non-genetic factors influencing AITD development (e.g., cytokines, iodine exposure) or with variants of other susceptible genes (e.g., HLA-DRß1). © 2024 Wiley Periodicals LLC. Basic Protocol: Development of a mouse model of autoimmune thyroiditis induced by immunization with adenovirus containing full-length thyroglobulin cDNA Support Protocol 1: Splenocytes isolation Support Protocol 2: T cell stimulation and carboxyfluorescein diacetate succinimidyl ester (CFSE) based cell proliferation assay Support Protocol 3: Cytokine assays: measuring levels of interferon gamma (IFNγ) and interleukins IL-2, IL-4, and IL-10 in splenocyte supernatants Support Protocol 4: Evaluating thyroid histology and infiltration with immune cells: hematoxylin-eosin staining of mice thyroid glands Support Protocol 5: Immunohistochemistry of thyroid tissues: Immunofluorescence protocol of paraffin-embedded thyroid sections Support Protocol 6: Anti-thyroglobulin antibody measurement in mice sera by enzyme-linked immunosorbent assay (ELISA).


Asunto(s)
Infecciones por Adenoviridae , Enfermedad de Hashimoto , Tiroiditis Autoinmune , Humanos , Animales , Ratones , Tiroglobulina/genética , Adenoviridae/genética , ADN Complementario/genética , Inmunización , Tiroiditis Autoinmune/genética , Citocinas , Modelos Animales de Enfermedad
17.
J Autoimmun ; 44: 61-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23683877

RESUMEN

Autoimmune thyroid diseases (AITD) have become increasingly recognized as a complication of interferon-alpha (IFNα) therapy in patients with chronic Hepatitis C virus (HCV) infection. Interferon-induced thyroiditis (IIT) can manifest as clinical thyroiditis in approximately 15% of HCV patients receiving IFNα and subclinical thyroiditis in up to 40% of patients, possibly resulting in either dose reduction or discontinuation of IFNα treatment. However, the exact mechanisms that lead to the development of IIT are unknown and may include IFNα-mediated immune-recruitment as well as direct toxic effects on thyroid follicular cells. We hypothesized that IIT develops in genetically predisposed individuals whose threshold for developing thyroiditis is lowered by IFNα. Therefore, our aim was to identify the susceptibility genes for IIT. We used a genomic convergence approach combining genetic association data with transcriptome analysis of genes upregulated by IFNα. Integrating results of genetic association, transcriptome data, pathway, and haplotype analyses enabled the identification of 3 putative loci, SP100/110/140 (2q37.1), HLA (6p21.3), and TAP1 (6p21.3) that may be involved in the pathogenesis of IIT. Immune-regulation and apoptosis emerged as the predominant mechanisms underlying the etiology of IIT.


Asunto(s)
Apoptosis/genética , Interferón-alfa/inmunología , Complejo Mayor de Histocompatibilidad/genética , Tiroiditis/genética , Tiroiditis/inmunología , Apoptosis/inmunología , Femenino , Perfilación de la Expresión Génica/métodos , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad , Haplotipos/genética , Hepacivirus/inmunología , Hepatitis C Crónica/genética , Hepatitis C Crónica/inmunología , Humanos , Complejo Mayor de Histocompatibilidad/inmunología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Tiroiditis/patología
18.
J Immunol ; 186(8): 4693-706, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21402899

RESUMEN

IFN-α is known to play a key role in autoimmunity, but the mechanisms are uncertain. Although the induction of autoimmunity by IFN-α is consistent with primarily immunomodulatory effects, the high frequency of nonautoimmune inflammation suggests other mechanisms. We used thyroiditis as a model to dissect these possibilities. IFN-α treatment of cultured thyrocytes increased expression of thyroid differentiation markers, thyroglobulin, thyroid-stimulating hormone receptor, thyroid peroxidase, and sodium iodide transporter. RNAseq analysis demonstrated that pathways of Ag presentation, pattern recognition receptors, and cytokines/chemokines were also stimulated. These changes were associated with markedly increased nonapoptotic thyroid cell death, suggesting direct toxicity. To corroborate these in vitro findings, we created transgenic mice with thyroid-specific overexpression of IFN-α under control of the thyroglobulin promoter. Transgenic mice developed marked inflammatory thyroid destruction associated with immune cell infiltration of thyroid and surrounding tissues leading to profound hypothyroidism, findings consistent with our in vitro results. In addition, transgenic mice thyroids showed upregulation of pathways similar to those observed in cultured thyrocytes. In particular, expression of granzyme B, CXCL10, a subset of the tripartite motif-containing family, and other genes involved in recruitment of bystander cytotoxic immune responses were increased. Pathways associated with apoptosis and autophagy were not induced. Taken together, our data demonstrate that the induction of tissue inflammation and autoimmunity by IFN-α involves direct tissue toxic effects as well as provocation of destructive bystander immune responses.


Asunto(s)
Autoinmunidad/inmunología , Sistema Inmunológico/inmunología , Interferón-alfa/inmunología , Glándula Tiroides/inmunología , Animales , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Interferón-alfa/genética , Interferón-alfa/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Ratas , Receptor de Interferón alfa y beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Tiroiditis/genética , Tiroiditis/metabolismo , Factores de Tiempo
19.
Proc Natl Acad Sci U S A ; 107(39): 16899-903, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20837527

RESUMEN

There is strong genetic association between type 1A diabetes (T1D) and autoimmune thyroid disease (AITD). T1D and AITD frequently occur together in the same individual, a condition classified as a variant of the autoimmune polyglandular syndrome type 3 (APS3). Because T1D and AITD are individually strongly associated with different HLA class II sequences, we asked which HLA class II pocket sequence and structure confer joint susceptibility to both T1D and AITD in the same individual (APS3v). We sequenced the HLA-DR gene in 105 APS3v patients and 153 controls, and identified a pocket amino acid signature, DRß-Tyr-26, DRß-Leu-67, DRß-Lys-71, and DRß-Arg-74, that was strongly associated with APS3v (P = 5.4 × 10(-14), odds ratio = 8.38). Logistic regression analysis demonstrated that DRß-Leu-67 (P = 9.4 × 10(-13)) and DRß-Arg-74 (P = 1.21 × 10(-13)) gave strong independent effects on disease susceptibility. Structural modeling studies demonstrated that pocket 4 was critical for the development of T1D+AITD; all disease-associated amino acids were linked to areas of the pocket that interact directly with the peptide and, therefore, influence peptide binding. The disease-susceptible HLA-DR pocket was more positively charged (Lys-71, Arg-74) compared with the protective pocket (Ala-71, Gln-74). We conclude that a specific pocket amino acid signature confers joint susceptibility to T1D+AITD in the same individual by causing significant structural changes in the MHC II peptide binding pocket and influencing peptide binding and presentation. Moreover, Arg-74 is a major amino acid position for the development of several autoimmune diseases. These findings suggest that blocking the critical Arg-74 pocket might offer a method for treating certain autoimmune conditions.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Antígenos HLA-DR/genética , Tiroiditis Autoinmune/genética , Secuencias de Aminoácidos , Arginina/química , Arginina/genética , Sitios de Unión , Femenino , Antígenos HLA-DR/química , Humanos , Masculino , Unión Proteica , Conformación Proteica , Análisis de Secuencia de Proteína
20.
Best Pract Res Clin Endocrinol Metab ; 37(2): 101661, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35459628

RESUMEN

Hashimoto's thyroiditis (HT) and Graves' disease (GD) are prevalent autoimmune disorders, representing opposite ends of the clinical spectrum of autoimmune thyroid diseases (AITD). The pathogenesis involves a complex interplay between environment and genes. Specific susceptibility genes have been discovered that predispose to AITD, including thyroid-specific and immune-regulatory genes. Growing evidence has revealed that genetic and epigenetic variants can alter autoantigen presentation during the development of immune tolerance, can enhance self-peptide binding to MHC (major histocompatibility complex), and can amplify stimulation of T- and B-cells. These gene-driven mechanistic discoveries lay the groundwork for novel treatment targets. This review summarizes recent advances in our understanding of key AITD susceptibility genes (Tg1, TSHR, HLA-DR3, and CD40) and their translational therapeutic potential.


Asunto(s)
Enfermedades Autoinmunes , Enfermedad de Graves , Enfermedad de Hashimoto , Enfermedades de la Tiroides , Humanos , Predisposición Genética a la Enfermedad , Enfermedad de Hashimoto/genética , Enfermedad de Hashimoto/complicaciones , Enfermedades Autoinmunes/complicaciones , Epigénesis Genética , Enfermedades de la Tiroides/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA