Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 30: 393-427, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22224765

RESUMEN

Monogenic autoimmune syndromes provide a rare yet powerful glimpse into the fundamental mechanisms of immunologic tolerance. Such syndromes reveal not only the contribution of an individual breakpoint in tolerance but also patterns in the pathogenesis of autoimmunity. Disturbances in innate immunity, a system built for ubiquitous sensing of danger signals, tend to generate systemic autoimmunity. For example, defects in the clearance of self-antigens and chronic stimulation of type 1 interferons lead to the systemic autoimmunity seen in C1q deficiency, SPENCDI, and AGS. In contrast, disturbances of adaptive immunity, which is built for antigen specificity, tend to produce organ-specific autoimmunity. Thus, the loss of lymphocyte homeostasis, whether through defects in apoptosis, suppression, or negative selection, leads to organ-specific autoimmunity in ALPS, IPEX, and APS1. We discuss the unique mechanisms of disease in these prominent syndromes as well as how they contribute to the spectrum of organ-specific or systemic autoimmunity. The continued study of rare variants in autoimmune disease will inform future investigations and treatments directed at rare and common autoimmune diseases alike.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Autoinmunidad/genética , Animales , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología
2.
Nat Immunol ; 19(7): 659-664, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29925986

RESUMEN

Although the thymus has long been recognized as a key organ for T cell selection, the intricate details linking these selection events to human autoimmunity have been challenging to decipher. Over the last two decades, there has been rapid progress in understanding the role of thymic tolerance mechanisms in autoimmunity through genetics. Here we review some of the recent progress in understanding key thymic tolerance processes that are critical for preventing autoimmune disease.


Asunto(s)
Autoinmunidad , Tolerancia Inmunológica , Timo/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T Reguladores/inmunología , Factores de Transcripción/fisiología , Proteína AIRE
3.
Clin Transl Sci ; 17(2): e13732, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38593352

RESUMEN

Molnupiravir is an oral prodrug of the broadly active, antiviral ribonucleoside analog N-hydroxycytidine (NHC). The primary circulating metabolite NHC is taken up into cells and phosphorylated to NHC-triphosphate (NHC-TP). NHC-TP serves as a competitive substrate for viral RNA-dependent RNA polymerase (RdRp), which results in an accumulation of errors in the viral genome, rendering virus replication incompetent. Molnupiravir has demonstrated activity against SARS-CoV-2 both clinically and preclinically and has a high barrier to development of viral resistance. Little to no molnupiravir is observed in plasma due to rapid hydrolysis to NHC. Maximum concentrations of NHC are reached at 1.5 h following administration in a fasted state. The effective half-life of NHC is 3.3 h, reflecting minimal accumulation in the plasma following twice-daily (Q12H) dosing. The terminal half-life of NHC is 20.6 h. NHC-TP exhibits a flatter profile with a lower peak-to-trough ratio compared with NHC, which supports Q12H dosing. Renal and hepatic pathways are not major routes of elimination, as NHC is primarily cleared by metabolism to uridine and cytidine, which then mix with the endogenous nucleotide pools. In a phase III study of nonhospitalized patients with COVID-19 (MOVe-OUT), 5 days of treatment with 800 mg molnupiravir Q12H significantly reduced the incidence of hospitalization or death compared with placebo. Patients treated with molnupiravir also had a greater reduction in SARS-CoV-2 viral load and improved clinical outcomes, compared with those receiving placebo. The clinical effectiveness of molnupiravir has been further demonstrated in several real-world evidence studies. Molnupiravir is currently authorized or approved in more than 25 countries.


Asunto(s)
Citidina/análogos & derivados , Ribonucleósidos , Ciencia Traslacional Biomédica , Humanos , Citidina/farmacología , Hidroxilaminas , SARS-CoV-2
4.
Diabetes ; 72(1): 59-70, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35709010

RESUMEN

Acquired lipodystrophy is often characterized as an idiopathic subtype of lipodystrophy. Despite suspicion of an immune-mediated pathology, biomarkers such as autoantibodies are generally lacking. Here, we used an unbiased proteome-wide screening approach to identify autoantibodies to the adipocyte-specific lipid droplet protein perilipin 1 (PLIN1) in a murine model of autoimmune polyendocrine syndrome type 1 (APS1). We then tested for PLIN1 autoantibodies in human subjects with acquired lipodystrophy with two independent severe breaks in immune tolerance (including APS1) along with control subjects using a specific radioligand binding assay and indirect immunofluorescence on fat tissue. We identified autoantibodies to PLIN1 in these two cases, including the first reported case of APS1 with acquired lipodystrophy and a second patient who acquired lipodystrophy as an immune-related adverse event following cancer immunotherapy. Lastly, we also found PLIN1 autoantibodies to be specifically enriched in a subset of patients with acquired generalized lipodystrophy (17 of 46 [37%]), particularly those with panniculitis and other features of autoimmunity. These data lend additional support to new literature that suggests that PLIN1 autoantibodies represent a marker of acquired autoimmune lipodystrophies and further link them to a break in immune tolerance.


Asunto(s)
Lipodistrofia Generalizada Congénita , Lipodistrofia , Humanos , Animales , Ratones , Perilipina-1/metabolismo , Autoanticuerpos , Lipodistrofia Generalizada Congénita/metabolismo , Lipodistrofia Generalizada Congénita/patología , Lipodistrofia/metabolismo , Tejido Adiposo/metabolismo
5.
Nat Med ; 28(10): 2092-2099, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36192552

RESUMEN

Hyperglucagonemia contributes to hyperglycemia in patients with type 1 diabetes (T1D); however, novel therapeutics that block glucagon action could improve glycemic control. This phase 2 study evaluated the safety and efficacy of volagidemab, an antagonistic monoclonal glucagon receptor (GCGR) antibody, as an adjunct to insulin therapy in adults with T1D. The primary endpoint was change in daily insulin use at week 12. Secondary endpoints included changes in hemoglobin A1c (HbA1c) at week 13, in average daily blood glucose concentration and time within target range as assessed by continuous blood glucose monitoring (CGM) and seven-point glucose profile at week 12, incidence of hypoglycemic events, the proportion of subjects who achieve HbA1c reduction of ≥0.4%, volagidemab drug concentrations and incidence of anti-drug antibodies. Eligible participants (n = 79) were randomized to receive weekly subcutaneous injections of placebo, 35 mg volagidemab or 70 mg volagidemab. Volagidemab produced a reduction in total daily insulin use at week 12 (35 mg volagidemab: -7.59 units (U) (95% confidence interval (CI) -11.79, -3.39; P = 0.040 versus placebo); 70 mg volagidemab: -6.64 U (95% CI -10.99, -2.29; P = 0.084 versus placebo); placebo: -1.27 U (95% CI -5.4, 2.9)) without meeting the prespecified significance level (P < 0.025). At week 13, the placebo-corrected reduction in HbA1c percentage was -0.53 (95% CI -0.89 to -0.17, nominal P = 0.004) in the 35 mg volagidemab group and -0.49 (95% CI -0.85 to -0.12, nominal P = 0.010) in the 70 mg volagidemab group. No increase in hypoglycemia was observed with volagidemab therapy; however, increases in serum transaminases, low-density lipoprotein (LDL)-cholesterol and blood pressure were observed. Although the primary endpoint did not meet the prespecified significance level, we believe that the observed reduction in HbA1c and tolerable safety profile provide a rationale for further randomized studies to define the long-term efficacy and safety of volagidemab in patients with T1D.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Diabetes Mellitus Tipo 1 , Receptores de Glucagón , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Glucemia , Automonitorización de la Glucosa Sanguínea , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Método Doble Ciego , Glucagón , Hemoglobina Glucada/análisis , Hemoglobina Glucada/uso terapéutico , Humanos , Insulina/uso terapéutico , Lipoproteínas LDL/uso terapéutico , Receptores de Glucagón/antagonistas & inhibidores , Transaminasas/uso terapéutico , Resultado del Tratamiento
6.
Elife ; 92020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32410729

RESUMEN

The identification of autoantigens remains a critical challenge for understanding and treating autoimmune diseases. Autoimmune polyendocrine syndrome type 1 (APS1), a rare monogenic form of autoimmunity, presents as widespread autoimmunity with T and B cell responses to multiple organs. Importantly, autoantibody discovery in APS1 can illuminate fundamental disease pathogenesis, and many of the antigens found in APS1 extend to more common autoimmune diseases. Here, we performed proteome-wide programmable phage-display (PhIP-Seq) on sera from a cohort of people with APS1 and discovered multiple common antibody targets. These novel APS1 autoantigens exhibit tissue-restricted expression, including expression in enteroendocrine cells, pineal gland, and dental enamel. Using detailed clinical phenotyping, we find novel associations between autoantibodies and organ-restricted autoimmunity, including a link between anti-KHDC3L autoantibodies and premature ovarian insufficiency, and between anti-RFX6 autoantibodies and diarrheal-type intestinal dysfunction. Our study highlights the utility of PhIP-Seq for extensively interrogating antigenic repertoires in human autoimmunity and the importance of antigen discovery for improved understanding of disease mechanisms.


The immune system uses antibodies to fight microbes that cause disease. White blood cells pump antibodies into the bloodstream, and these antibodies latch onto bacteria and viruses, targeting them for destruction. But sometimes, the immune system gets it wrong. In autoimmune diseases, white blood cells mistakenly make antibodies that target the body's own tissues. Detecting these 'autoantibodies' in the blood can help doctors to diagnose autoimmune diseases. But the identities and targets of many autoantibodies remain unknown. In one rare disease, called autoimmune polyendocrine syndrome type 1 (APS-1), a faulty gene makes the immune system much more likely to make autoantibodies. People with this disease can develop an autoimmune response against many different healthy organs. Although APS-1 is rare, some of the autoantibodies made by individuals with the disease are the same as the ones in more common autoimmune diseases, like type 1 diabetes. Therefore, investigating the other autoantibodies produced by individuals with APS-1 could reveal the autoantibodies driving other autoimmune diseases. Autoantibodies bind to specific regions of healthy proteins, and one way to identify them is to use hundreds of thousands of tiny viruses in a technique called proteome-wide programmable phage-display, or PhIP-Seq. Each phage carries one type of protein segment. When mixed with blood serum from a patient, the autoantibodies stick to the phages that carry the target proteins for that autoantibody. These complexes can be isolated using biochemical techniques. Sequencing the genes of these phages then reveals the identity of the autoantibodies' targets. Using this technique, Vazquez et al successfully pulled 23 known autoantibodies from the serum of patients with APS-1. Then, experiments to search for new targets began. This revealed many new autoantibodies, targeting proteins found only in specific tissues. They included one that targets a protein found on cells in the gut, and another that targets a protein found on egg cells in the ovaries. Matching the PhIP-Seq data to patient symptoms confirmed that these new antibodies correlate with the features of specific autoimmune diseases. For example, patients with antibodies that targeted the gut protein were more likely to have gut symptoms, while patients with antibodies that targeted the egg cell protein were more likely to have problems with their ovaries. Further investigations using PhIP-Seq could reveal the identities of even more autoantibodies. This might pave the way for new antibody tests to diagnose autoimmune diseases and identify tissues at risk of damage. This could be useful not only for people with APS-1, but also for more common autoimmune diseases that target the same organs.


Asunto(s)
Autoanticuerpos/sangre , Autoantígenos/sangre , Autoinmunidad , Técnicas de Visualización de Superficie Celular , Poliendocrinopatías Autoinmunes/sangre , Proteoma , Proteómica , Fosfatasa Ácida/sangre , Fosfatasa Ácida/inmunología , Autoantígenos/inmunología , Biomarcadores/sangre , Femenino , Células HEK293 , Humanos , Masculino , Biblioteca de Péptidos , Poliendocrinopatías Autoinmunes/diagnóstico , Poliendocrinopatías Autoinmunes/inmunología , Proteínas/inmunología , Factores de Transcripción del Factor Regulador X/sangre , Factores de Transcripción del Factor Regulador X/inmunología
7.
J Clin Invest ; 129(11): 4676-4681, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31369399

RESUMEN

While improvements in genetic analysis have greatly enhanced our understanding of the mechanisms behind pancreatitis, it continues to afflict many families for whom the hereditary factors remain unknown. Recent evaluation of a patient with a strong family history of pancreatitis sparked us to reexamine a large kindred originally reported over 50 years ago with an autosomal dominant inheritance pattern of chronic pancreatitis, diabetes and pancreatic adenocarcinoma. Whole exome sequencing analysis identified a rare missense mutation in the gene encoding pancreas-specific protease Elastase 3B (CELA3B) that cosegregates with disease. Studies of the mutant protein in vitro, in cell lines and in CRISPR-Cas9 engineered mice indicate that this mutation causes translational upregulation of CELA3B, which upon secretion and activation by trypsin leads to uncontrolled proteolysis and recurrent pancreatitis. Although lesions in several other pancreatitic proteases have been previously linked to hereditary pancreatitis, this is the first known instance of a mutation in CELA3B and a defect in translational control contributing to this disease.


Asunto(s)
Adenocarcinoma/genética , Enfermedades Genéticas Congénitas/genética , Predisposición Genética a la Enfermedad , Mutación , Proteínas de Neoplasias/genética , Elastasa Pancreática/genética , Neoplasias Pancreáticas/genética , Pancreatitis/genética , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/patología , Humanos , Ratones , Proteínas de Neoplasias/metabolismo , Elastasa Pancreática/biosíntesis , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Pancreatitis/enzimología , Pancreatitis/patología , Regulación hacia Arriba , Secuenciación del Exoma , Neoplasias Pancreáticas
10.
J Clin Invest ; 125(6): 2250-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25985270

RESUMEN

Immune tolerance is critical to the avoidance of unwarranted immune responses against self antigens. Multiple, non-redundant checkpoints are in place to prevent such potentially deleterious autoimmune responses while preserving immunity integral to the fight against foreign pathogens. Nevertheless, a large and growing segment of the population is developing autoimmune diseases. Deciphering cellular and molecular pathways of immune tolerance is an important goal, with the expectation that understanding these pathways will lead to new clinical advances in the treatment of these devastating diseases. The vast majority of autoimmune diseases develop as a consequence of complex mechanisms that depend on genetic, epigenetic, molecular, cellular, and environmental elements and result in alterations in many different checkpoints of tolerance and ultimately in the breakdown of immune tolerance. The manifestations of this breakdown are harmful inflammatory responses in peripheral tissues driven by innate immunity and self antigen-specific pathogenic T and B cells. T cells play a central role in the regulation and initiation of these responses. In this Review we summarize our current understanding of the mechanisms involved in these fundamental checkpoints, the pathways that are defective in autoimmune diseases, and the therapeutic strategies being developed with the goal of restoring immune tolerance.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Tolerancia Inmunológica , Inmunidad Innata , Linfocitos T/inmunología , Animales , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Linfocitos B/inmunología , Linfocitos B/patología , Humanos , Especificidad de Órganos/inmunología , Linfocitos T/patología
11.
Nat Genet ; 47(6): 654-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25894502

RESUMEN

Unbiased genetic studies have uncovered surprising molecular mechanisms in human cellular immunity and autoimmunity. We performed whole-exome sequencing and targeted sequencing in five families with an apparent mendelian syndrome of autoimmunity characterized by high-titer autoantibodies, inflammatory arthritis and interstitial lung disease. We identified four unique deleterious variants in the COPA gene (encoding coatomer subunit α) affecting the same functional domain. Hypothesizing that mutant COPA leads to defective intracellular transport via coat protein complex I (COPI), we show that COPA variants impair binding to proteins targeted for retrograde Golgi-to-ER transport. Additionally, expression of mutant COPA results in ER stress and the upregulation of cytokines priming for a T helper type 17 (TH17) response. Patient-derived CD4(+) T cells also demonstrate significant skewing toward a TH17 phenotype that is implicated in autoimmunity. Our findings uncover an unexpected molecular link between a vesicular transport protein and a syndrome of autoimmunity manifested by lung and joint disease.


Asunto(s)
Artritis/genética , Enfermedades Autoinmunes/genética , Proteína Coatómero/genética , Aparato de Golgi/metabolismo , Enfermedades Pulmonares Intersticiales/genética , Secuencia de Aminoácidos , Preescolar , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Lactante , Escala de Lod , Masculino , Datos de Secuencia Molecular , Linaje , Transporte de Proteínas
12.
Curr Opin Endocrinol Diabetes Obes ; 20(4): 271-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23770732

RESUMEN

PURPOSE OF REVIEW: Advances in human genetics and investigations in animal models of autoimmune disease have allowed insight into the basic mechanisms of immunologic tolerance. These advances allow us to understand the pathogenesis of type 1 diabetes and other autoimmune diseases as never before. Here, we discuss the tolerance mechanisms of the autoimmune polyendocrine syndromes and their relevance to type 1 diabetes. RECENT FINDINGS: Defects in central tolerance with alteration of self-antigen expression levels in the thymus are a potent cause of autoimmunity. Peripheral tolerance defects that alter T-cell activation and signaling also play an important role in the pathogenesis of diabetes and other associated autoimmune disorders, with multiple modest defects working in concert to produce disease. Regulation of the immune response through the action of regulatory T cells is a potent mode of tolerance induction in autoimmunity that is important in type 1 diabetes. SUMMARY: Rare syndromes of autoimmunity provide a valuable window into the breakdown of tolerance and identify multiple checkpoints that are critical for generation of autoimmunity. Understanding the application of these in type 1 diabetes will allow the development of future immunomodulatory therapies in the treatment and prevention of disease.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Animales , Humanos , Tolerancia Inmunológica , Poliendocrinopatías Autoinmunes/metabolismo , Linfocitos T Reguladores/inmunología
13.
J Clin Invest ; 123(12): 5135-45, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24270423

RESUMEN

Autoantibodies against cytokines, chemokines, and growth factors inhibit normal immunity and are implicated in inflammatory autoimmune disease and diseases of immune deficiency. In an effort to evaluate serum from autoimmune and immunodeficient patients for Abs against cytokines, chemokines, and growth factors in a high-throughput and unbiased manner, we constructed a multiplex protein microarray for detection of serum factor-binding Abs and used the microarray to detect autoantibody targets in SLE. We designed a nitrocellulose-surface microarray containing human cytokines, chemokines, and other circulating proteins and demonstrated that the array permitted specific detection of serum factor-binding probes. We used the arrays to detect previously described autoantibodies against cytokines in samples from individuals with autoimmune polyendocrine syndrome type 1 and chronic mycobacterial infection. Serum profiling from individuals with SLE revealed that among several targets, elevated IgG autoantibody reactivity to B cell-activating factor (BAFF) was associated with SLE compared with control samples. BAFF reactivity correlated with the severity of disease-associated features, including IFN-α-driven SLE pathology. Our results showed that serum factor protein microarrays facilitate detection of autoantibody reactivity to serum factors in human samples and that BAFF-reactive autoantibodies may be associated with an elevated inflammatory disease state within the spectrum of SLE.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Factor Activador de Células B/inmunología , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/inmunología , Análisis por Matrices de Proteínas , Animales , Especificidad de Anticuerpos , Autoanticuerpos/sangre , Citocinas/inmunología , Humanos , Inmunoglobulina G/sangre , Inflamación , Interferón-alfa/inmunología , Ratones , Infecciones por Mycobacterium/sangre , Infecciones por Mycobacterium/inmunología , Poliendocrinopatías Autoinmunes/sangre , Poliendocrinopatías Autoinmunes/inmunología , Proteínas Recombinantes/inmunología
14.
Sci Transl Med ; 5(206): 206ra139, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24107778

RESUMEN

Interstitial lung disease (ILD) is a complex and heterogeneous disorder that is often associated with autoimmune syndromes. Despite the connection between ILD and autoimmunity, it remains unclear whether ILD can develop from an autoimmune response that specifically targets the lung parenchyma. We examined a severe form of autoimmune disease, autoimmune polyglandular syndrome type 1 (APS1), and established a strong link between an autoimmune response to the lung-specific protein BPIFB1 (bactericidal/permeability-increasing fold-containing B1) and clinical ILD. Screening of a large cohort of APS1 patients revealed autoantibodies to BPIFB1 in 9.6% of APS1 subjects overall and in 100% of APS1 subjects with ILD. Further investigation of ILD outside the APS1 disorder revealed BPIFB1 autoantibodies present in 14.6% of patients with connective tissue disease-associated ILD and in 12.0% of patients with idiopathic ILD. The animal model for APS1, Aire⁻/⁻ mice, harbors autoantibodies to a similar lung antigen (BPIFB9); these autoantibodies are a marker for ILD. We found that a defect in thymic tolerance was responsible for the production of BPIFB9 autoantibodies and the development of ILD. We also found that immunoreactivity targeting BPIFB1 independent of a defect in Aire also led to ILD, consistent with our discovery of BPIFB1 autoantibodies in non-APS1 patients. Overall, our results demonstrate that autoimmunity targeting the lung-specific antigen BPIFB1 may contribute to the pathogenesis of ILD in patients with APS1 and in subsets of patients with non-APS1 ILD, demonstrating the role of lung-specific autoimmunity in the genesis of ILD.


Asunto(s)
Autoantígenos/inmunología , Proteínas Portadoras/metabolismo , Glicoproteínas/metabolismo , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/patología , Pulmón/inmunología , Pulmón/patología , Proteínas/metabolismo , Traslado Adoptivo , Animales , Autoanticuerpos/inmunología , Autoantígenos/metabolismo , Autoinmunidad/inmunología , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión a Ácidos Grasos , Genotipo , Humanos , Tolerancia Inmunológica/inmunología , Ratones , Especificidad de Órganos , Poliendocrinopatías Autoinmunes/inmunología , Ensayo de Unión Radioligante , Reproducibilidad de los Resultados , Timo/inmunología , Timo/trasplante , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína AIRE
15.
Semin Reprod Med ; 29(4): 308-16, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21969265

RESUMEN

Ovarian autoimmunity is increasingly implicated in the etiology of primary ovarian insufficiency (POI), previously termed PREMATURE OVARIAN FAILURE or PREMATURE MENOPAUSE. Links to autoimmunity in human POI have long been noted due to the close association of POI with several autoimmune diseases and syndromes such as Addison's disease and Autoimmune polyglandular syndrome 1. However, diagnosis of autoimmune-mediated POI (aPOI) remains challenging because of the lack of sensitive or specific markers of disease. Autoimmunity can arise from the breakdown of immunological tolerance in several ways. How then may we discern what constitutes a relevant target and what represents a downstream phenomenon? The answer lies in the study of pathogenic mechanisms in translational models of disease. From examples in humans and mice, we see that ovarian autoimmunity likely arises from a limited number of antigens targeted in the ovary that are organ specific. These antigens may be conserved but not limited to those seen in animal models of autoimmune ovarian disease. Recent advances in these areas have begun to define the relevant antigens and mechanisms of immune tolerance breakdown in the ovary. Work in translational models continues to provide insight into mechanisms of disease pathogenesis that will allow more accurate diagnosis and, ultimately, improved interventions for women with aPOI.


Asunto(s)
Ovario/inmunología , Insuficiencia Ovárica Primaria/inmunología , Animales , Autoantígenos/inmunología , Autoinmunidad/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Endocrinology ; 152(6): 2465-73, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21447630

RESUMEN

Primary ovarian insufficiency (POI) resulting from ovarian autoimmunity is a poorly understood clinical condition lacking in effective treatments. Understanding the targets of the autoimmune response and induction of ovarian-specific tolerance would allow development of focused therapies to preserve fertility in an at-risk population. MATER (maternal antigen that embryos require) is a known ovarian autoantigen targeted in autoimmune syndromes of POI. We attempt to induce ovarian-specific tolerance via transgenic expression of the MATER antigen on potentially tolerogenic antigen-presenting cells (APC), which typically present antigen via the major histocompatibility complex (MHC) class II molecule. We hypothesize that expression of MATER in a MHC class II-dependent manner on APC can mediate induction of ovarian tolerance. We utilized a well-characterized murine model of ovarian autoimmunity, whereby oophoritis develops after d 3 neonatal thymectomy (NTx). Wild-type and transgenic mice, carrying an MHC Class II-driven Mater gene (IE-Mater), were subjected to NTx and assessed for evidence of autoimmune oophoritis. After disease induction by NTx, female mice carrying the IE-Mater transgene had significant reductions in histological oophoritis (56%) and circulating ovarian autoantibodies (28%) compared with wild-type females (94% and 82%, respectively). Incidence of other autoimmunity was unaffected as assessed by antinuclear autoantibodies. Transgenic expression of MATER in APC can induce antigen-specific tolerance with a significant reduction in ovarian autoimmunity. Lack of complete disease protection suggests that other antigens may also play a role in autoimmune oophoritis. As a known autoantigen in the human APS1 (autoimmune polyglandular syndrome type 1), which is associated with POI, MATER may represent a relevant target for future diagnostic and therapeutic clinical interventions.


Asunto(s)
Antígenos/genética , Antígenos/inmunología , Proteínas del Huevo/genética , Proteínas del Huevo/inmunología , Ooforitis/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Animales , Antígenos/farmacología , Modelos Animales de Enfermedad , Proteínas del Huevo/farmacología , Femenino , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Tolerancia Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ooforitis/tratamiento farmacológico , Ooforitis/genética , Ovario/inmunología , Poliendocrinopatías Autoinmunes/tratamiento farmacológico , Poliendocrinopatías Autoinmunes/genética
17.
Trends Immunol ; 28(7): 321-7, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17556019

RESUMEN

Unraveling the mechanisms underlying autoimmune disease remains a difficult challenge. Recent lessons learned from the study of AIRE (autoimmune regulator), the gene responsible for the rare monogenic human syndrome APS-1, highlight the power of genetics to reveal disease pathogenesis. With the discovery of AIRE, central tolerance has re-emerged as a crucial check against autoimmunity. Aire-mediated regulation of diverse self-antigens in the thymus serves as a paradigm for the importance of promiscuous gene expression in the prevention of autoimmune disease. Recent characterization of Aire-targeted antigens continues to bear this out. Here, we review the current progress surrounding the role of Aire in central tolerance from a molecular, genetic and developmental basis.


Asunto(s)
Autoinmunidad/inmunología , Factores de Transcripción/inmunología , Animales , Antígenos/inmunología , Diferenciación Celular/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Timo/citología , Timo/inmunología , Proteína AIRE
18.
Clin Immunol ; 125(2): 131-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17825626

RESUMEN

UNLABELLED: High titer antibodies to type 1 interferons have been recently reported as being highly specific for patients with autoimmune polyglandular syndrome type 1 (APS1) in Finnish and Norwegian patients with mutations in the AIRE gene. Those studies employed a complex neutralization assay to define the type 1 interferon autoantibodies. Here we have established a competitive europium time resolved fluorescence assay for IFN-alpha autoantibodies and measured sera from subjects with APS1, first degree relatives of APS1 patients, patients with Addison's disease or Type 1 diabetes. The europium-based immunoassay utilizes plate bound human IFN-alpha incubated with sera with or without competition with fluid phase IFN-alpha, followed by anti-IgG biotinylated antibody and detection with streptavidin-europium. The index of IFN-alpha Ab was calculated as (CPS (Counts per second) without competition-CPS with competition)/(CPS positive standard sera without competition-CPS positive standard sera with competition). RESULTS are reported for raw CPS and indices and are compared across the different subjects. RESULTS: For normal controls (n=100) CPS without competition were 31,237+/-17,328 CPS while after subtracting the competition value, the results were -6563+/-10,303 CPS. The initial APS1 patient (used to create the index as 1.0) gave 394,063 CPS without competition and a delta of 363,662+/-31,587 CPS with competition. Scatchard plot analysis of this patient sample revealed a high avidity for IFN-alpha (K(d) of 0.5 nM). The CPS, delta, and index for 6/7 APS1 patients were strongly positive and 3 standard deviations or more above that of the normal controls. Using a cut-off of 2 standard deviations above normal controls, relatives of APS1 patients were negative for type I interferon autoantibodies as were 71 patients with Addison's disease (non-APS1) and 141 Type 1 diabetes patients. This simple high throughput competitive europium time resolved fluorescence assay had a sensitivity of > or =86% or greater and a specificity of >99.5%.


Asunto(s)
Autoanticuerpos/sangre , Fluoroinmunoensayo/métodos , Interferón-alfa/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Adolescente , Adulto , Especificidad de Anticuerpos , Niño , Preescolar , Europio/farmacología , Fluorometría , Humanos , Sustancias Luminiscentes/farmacología , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA