Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Nat Immunol ; 23(7): 1098-1108, 2022 07.
Article in English | MEDLINE | ID: mdl-35761088

ABSTRACT

Patients with loss of function in the gene encoding the master regulator of central tolerance AIRE suffer from a devastating disorder called autoimmune polyendocrine syndrome type 1 (APS-1), characterized by a spectrum of autoimmune diseases and severe mucocutaneous candidiasis. Although the key mechanisms underlying the development of autoimmunity in patients with APS-1 are well established, the underlying cause of the increased susceptibility to Candida albicans infection remains less understood. Here, we show that Aire+MHCII+ type 3 innate lymphoid cells (ILC3s) could sense, internalize and present C. albicans and had a critical role in the induction of Candida-specific T helper 17 (TH17) cell clones. Extrathymic Rorc-Cre-mediated deletion of Aire resulted in impaired generation of Candida-specific TH17 cells and subsequent overgrowth of C. albicans in the mucosal tissues. Collectively, our observations identify a previously unrecognized regulatory mechanism for effective defense responses against fungal infections.


Subject(s)
Autoimmune Diseases , Candidiasis , Polyendocrinopathies, Autoimmune , Candida albicans , Candidiasis/genetics , Humans , Immunity, Innate , Polyendocrinopathies, Autoimmune/genetics , Th17 Cells
2.
Clin Exp Immunol ; 215(1): 47-57, 2024 01 09.
Article in English | MEDLINE | ID: mdl-37578839

ABSTRACT

Primary adrenal insufficiency (PAI) is most often caused by an autoimmune destruction of the adrenal cortex resulting in failure to produce cortisol and aldosterone. The aetiology is thought to be a combination of genetic and environmental risk factors, leading to breakdown of immunological tolerance. Regulatory T cells (Tregs) are deficient in many autoimmune disorders, but it is not known whether they contribute to development of PAI. We aimed to investigate the frequency and function of naive and expanded Tregs in patients with PAI and polyendocrine syndromes compared to age- and gender-matched healthy controls. Flow cytometry was used to assess the frequency and characterize functional markers of blood Tregs in PAI (N = 15). Expanded Treg suppressive abilities were assessed with a flow cytometry based suppression assay (N = 20), while bulk RNA-sequencing was used to examine transcriptomic differences (N = 16) and oxygen consumption rate was measured by a Seahorse cell metabolic assay (N = 11). Our results showed that Treg frequency and suppressive capacity were similar between patients and controls. An increased expression of killer-cell leptin-like receptors and mitochondrial genes was revealed in PAI patients, but their expanded Tregs did not display signs of mitochondrial dysfunction. Our findings do not support a clear role for Tregs in the contribution of PAI development.


Subject(s)
Addison Disease , T-Lymphocytes, Regulatory , Humans , Addison Disease/genetics , Immune Tolerance , Hydrocortisone/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism
3.
Immunity ; 42(6): 1185-96, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084028

ABSTRACT

The autoimmune regulator (AIRE) gene is crucial for establishing central immunological tolerance and preventing autoimmunity. Mutations in AIRE cause a rare autosomal-recessive disease, autoimmune polyendocrine syndrome type 1 (APS-1), distinguished by multi-organ autoimmunity. We have identified multiple cases and families with mono-allelic mutations in the first plant homeodomain (PHD1) zinc finger of AIRE that followed dominant inheritance, typically characterized by later onset, milder phenotypes, and reduced penetrance compared to classical APS-1. These missense PHD1 mutations suppressed gene expression driven by wild-type AIRE in a dominant-negative manner, unlike CARD or truncated AIRE mutants that lacked such dominant capacity. Exome array analysis revealed that the PHD1 dominant mutants were found with relatively high frequency (>0.0008) in mixed populations. Our results provide insight into the molecular action of AIRE and demonstrate that disease-causing mutations in the AIRE locus are more common than previously appreciated and cause more variable autoimmune phenotypes.


Subject(s)
DNA Mutational Analysis/methods , Genes, Dominant/genetics , Mutation/genetics , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , Adolescent , Adult , Amino Acid Sequence , Autoimmunity/genetics , Child , Child, Preschool , Female , Gene Frequency , Humans , Male , Microsatellite Repeats/genetics , Molecular Sequence Data , Norway , Organ Specificity/genetics , Pedigree , Penetrance , Phenotype , Russia , Young Adult , AIRE Protein
4.
Crit Rev Immunol ; 43(5): 25-52, 2023.
Article in English | MEDLINE | ID: mdl-37831521

ABSTRACT

T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.


Subject(s)
Autoimmune Diseases , Th17 Cells , Humans , Quality of Life , T-Lymphocytes, Regulatory , Inflammation , Cytokines/metabolism
5.
J Autoimmun ; 133: 102917, 2022 12.
Article in English | MEDLINE | ID: mdl-36191466

ABSTRACT

BACKGROUND: Autoantibodies against type I interferons (IFN) alpha (α) and omega (ω), and interleukins (IL) 17 and 22 are a hallmark of autoimmune polyendocrine syndrome type 1 (APS-1), caused by mutations in the autoimmune regulator (AIRE) gene. Such antibodies are also seen in a number of monogenic immunodeficiencies. OBJECTIVES: To determine whether screening for cytokine autoantibodies (anti-IFN-ω and anti-IL22) can be used to identify patients with monogenic immune disorders. METHODS: A novel ELISA assay was employed to measure IL22 autoantibodies in 675 patients with autoimmune primary adrenal insufficiency (PAI) and a radio immune assay (RIA) was used to measure autoantibodies against IFN-ω in 1778 patients with a variety of endocrine diseases, mostly of autoimmune aetiology. Positive cases were sequenced for all coding exons of the AIRE gene. If no AIRE mutations were found, we applied next generation sequencing (NGS) to search for mutations in immune related genes. RESULTS: We identified 29 patients with autoantibodies against IFN-ω and/or IL22. Of these, four new APS-1 cases with disease-causing variants in AIRE were found. In addition, we identified two patients with pathogenic heterozygous variants in CTLA4 and NFKB2, respectively. Nine rare variants in other immune genes were identified in six patients, although further studies are needed to determine their disease-causing potential. CONCLUSION: Screening of cytokine autoantibodies can efficiently identify patients with previously unknown monogenic and possible oligogenic causes of autoimmune and immune deficiency diseases. This information is crucial for providing personalised treatment and follow-up of patients and their relatives.


Subject(s)
Autoantibodies , Endocrine System Diseases , Humans , Cytokines
6.
Scand J Immunol ; 92(5): e12961, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32853446

ABSTRACT

The new era of immune and reconstitution therapy of autoimmune disorders is ongoing. However, endocrine autoimmune diseases comprise a group of elaborating pathologies where the development of new treatment strategies remains slow. Substitution of the missing hormones is still standard practice, taking care of the devastating symptoms but not the cause of disease. As our knowledge of the genetic contribution to the aetiology of endocrine disorders increases and early diagnostic tools are available, it is now possible to identify persons at risk before they acquire full-blown disease. This review summarizes current knowledge and treatment of endocrine autoimmune disorders, focusing on type 1 diabetes, Addison's disease, autoimmune thyroid diseases and primary ovarian insufficiency. We explore which new therapies might be used in the different stages of the disease, focus on legalized therapy and elaborate on the ongoing clinical studies for these diseases and the research front, before hypothesizing on the way ahead.


Subject(s)
Addison Disease/immunology , Autoimmune Diseases/immunology , Diabetes Mellitus, Type 1/immunology , Endocrine System Diseases/immunology , Primary Ovarian Insufficiency/immunology , Thyroid Diseases/immunology , Addison Disease/genetics , Addison Disease/therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Endocrine System Diseases/genetics , Endocrine System Diseases/therapy , Female , Humans , Immunotherapy/methods , Models, Immunological , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/therapy , Thyroid Diseases/genetics , Thyroid Diseases/therapy
7.
Scand J Immunol ; 92(5): e12964, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32869859

ABSTRACT

In the last decade, there has been a tremendous development of technologies focused on analysing various molecular attributes in single cells, with an ever-increasing number of parameters becoming available at the DNA, RNA and protein levels. Much of this progress has involved cells in suspension, but also in situ analysis of tissues has taken great leaps. Paralleling the development in the laboratory, and because of increasing complexity, the analysis of single-cell data is also constantly being updated with new algorithms and analysis platforms. Our immune system shares this complexity, and immunologists have therefore been in the forefront of this technological development. These technologies clearly open new avenues for immunology research, maybe particularly within autoimmunity where the interaction between the faulty immune system and the thymus or the target organ is important. However, the technologies currently available can seem overwhelming and daunting. The aim of this review is to remedy this by giving a balanced overview of the prospects of using single-cell analysis in basal and clinical autoimmunity research, with an emphasis on endocrine autoimmunity.


Subject(s)
Autoimmunity/immunology , Computational Biology/methods , Flow Cytometry/methods , Immune System/immunology , Single-Cell Analysis/methods , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/genetics , Gene Expression Profiling/methods , Humans , Immune System/cytology , Immune System/metabolism , Sequence Analysis, RNA/methods
8.
J Autoimmun ; 81: 24-33, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28318808

ABSTRACT

Next generation sequencing of T and B cell receptors is emerging as a valuable and effective method to diagnose and monitor hematopoietic malignancies. So far, this approach has not been fully explored in regard to autoimmune diseases. T cells develop in the thymus where they undergo positive and negative selection, and the autoimmune regulator (Aire) is central in the establishment of immunological tolerance. Loss of Aire leads to severe multiorgan autoimmune disease with infiltration of autoreactive T cells in affected organs. Here, we have utilized next generation sequencing technology to investigate the T cell receptor repertoire in autoimmunity induced by immunization of mice with a self-antigen, myeloperoxidase. By investigating the T cell receptor repertoire in peripheral blood, spleen and lumbar lymph nodes from naïve and immunized Aire -/- mice and wild type littermates, changes in the usage of V and J genes were evident. Our results identify TCR clonotypes which could be potential targets for immune therapy. Also, Aire -/- autoimmunity is driven by a variety of autoantigens where the autoimmune response is highly polyclonal, and access to the most adjacent immunologically active tissue is required to identify T cell receptor sequences that are potentially unique to the antigen in Aire-/- immunized mice.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/genetics , Cell Communication/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Autoimmune Diseases/metabolism , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Disease Models, Animal , Female , Genotype , Male , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Sequence Analysis, DNA
9.
J Immunol ; 193(8): 3880-90, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25230752

ABSTRACT

Patients with the autoimmune polyendocrine syndrome type I (APS-I), caused by mutations in the autoimmune regulator (AIRE) gene, and myasthenia gravis (MG) with thymoma, show intriguing but unexplained parallels. They include uncommon manifestations like autoimmune adrenal insufficiency (AI), hypoparathyroidism, and chronic mucocutaneous candidiasis plus autoantibodies neutralizing IL-17, IL-22, and type I IFNs. Thymopoiesis in the absence of AIRE is implicated in both syndromes. To test whether these parallels extend further, we screened 247 patients with MG, thymoma, or both for clinical features and organ-specific autoantibodies characteristic of APS-I patients, and we assayed 26 thymoma samples for transcripts for AIRE and 16 peripheral tissue-specific autoantigens (TSAgs) by quantitative PCR. We found APS-I-typical autoantibodies and clinical manifestations, including chronic mucocutaneous candidiasis, AI, and asplenia, respectively, in 49 of 121 (40%) and 10 of 121 (8%) thymoma patients, but clinical features seldom occurred together with the corresponding autoantibodies. Both were rare in other MG subgroups (n = 126). In 38 patients with APS-I, by contrast, we observed neither autoantibodies against muscle Ags nor any neuromuscular disorders. Whereas relative transcript levels for AIRE and 7 of 16 TSAgs showed the expected underexpression in thymomas, levels were increased for four of the five TSAgs most frequently targeted by these patients' autoantibodies. Therefore, the clinical and serologic parallels to APS-I in patients with thymomas are not explained purely by deficient TSAg transcription in these aberrant AIRE-deficient tumors. We therefore propose additional explanations for the unusual autoimmune biases they provoke. Thymoma patients should be monitored for potentially life-threatening APS-I manifestations such as AI and hypoparathyroidism.


Subject(s)
Autoantigens/immunology , Polyendocrinopathies, Autoimmune/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Transcription Factors/genetics , Adrenal Insufficiency/immunology , Adult , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/genetics , Candidiasis, Chronic Mucocutaneous , Female , Heterotaxy Syndrome/immunology , Humans , Hypoparathyroidism/immunology , Interferon Type I/immunology , Interleukin-17/immunology , Interleukins/immunology , Male , Middle Aged , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Polyendocrinopathies, Autoimmune/genetics , Thymoma/genetics , Thymus Neoplasms/genetics , AIRE Protein , Interleukin-22
10.
Clin Immunol ; 153(1): 220-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24792136

ABSTRACT

An important characteristic of autoimmune polyendocrine syndrome type 1 (APS 1) is the existence of neutralizing autoantibodies (nAbs) against the type I interferons (IFN) -α2 and -ω at frequencies close to 100%. Type 1 IFN autoantibodies are detected by antiviral neutralizing assays (AVA), binding assays with radiolabelled antigens (RLBA), enzyme-linked immunosorbent assay (ELISA), or by reporter-based cell assays. We here present a simple and reliable version of the latter utilizing a commercially available cell line (HEK-Blue IFN-α/ß). All 67 APS 1 patients were positive for IFN-ω nAbs, while 90% were positive for IFN-α2 nAbs, a 100% and 96% correlation with RLBA, respectively. All blood donors and non-APS 1 patients were negative. The dilution titer required to reduce the effect of IFN-ω nAbs correlated with the RLBA index. This cell-based autoantibody assay (CBAA) is easy to perform, suitable for high throughput, while providing high specificity and sensitivity.


Subject(s)
Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Immunoassay/methods , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/immunology , Antibodies, Neutralizing/blood , Autoantibodies/blood , Cell Line , Humans , Sensitivity and Specificity
11.
Front Immunol ; 15: 1327784, 2024.
Article in English | MEDLINE | ID: mdl-38455040

ABSTRACT

Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.


Subject(s)
Autoantibodies , Interferon Type I , Thymus Gland , Herpesvirus 3, Human
12.
iScience ; 27(4): 109610, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38632993

ABSTRACT

Immune tolerance fails in autoimmune polyendocrine syndrome type 1 (APS-1) because of AIRE mutations. We have used single cell transcriptomics to characterize regulatory T cells (Tregs) sorted directly from blood and from in vitro expanded Tregs in APS-1 patients compared to healthy controls. We revealed only CD52 and LTB (down) and TXNIP (up) as consistently differentially expressed genes in the datasets. There were furthermore no large differences of the TCR-repertoire of expanded Tregs between the cohorts, but unique patients showed a more restricted use of specific clonotypes. We also found that in vitro expanded Tregs from APS-1 patients had similar suppressive capacity as controls in co-culture assays, despite expanding faster and having more exhausted cells. Our results suggest that APS-1 patients do not have intrinsic defects in their Treg functionality, and that their Tregs can be expanded ex vivo for potential therapeutic applications.

13.
Front Endocrinol (Lausanne) ; 14: 1285901, 2023.
Article in English | MEDLINE | ID: mdl-38027140

ABSTRACT

The adrenal glands are small endocrine glands located on top of each kidney, producing hormones regulating important functions in our body like metabolism and stress. There are several underlying causes for adrenal insufficiency, where an autoimmune attack by the immune system is the most common cause. A number of genes are known to confer early onset adrenal disease in monogenic inheritance patterns, usually genetic encoding enzymes of adrenal steroidogenesis. Autoimmune primary adrenal insufficiency is usually a polygenic disease where our information recently has increased due to genome association studies. In this review, we go through the physiology of the adrenals before explaining the different reasons for adrenal insufficiency with a particular focus on autoimmune primary adrenal insufficiency. We will give a clinical overview including diagnosis and current treatment, before giving an overview of the genetic causes including monogenetic reasons for adrenal insufficiency and the polygenic background and inheritance pattern in autoimmune adrenal insufficiency. We will then look at the autoimmune mechanisms underlying autoimmune adrenal insufficiency and how autoantibodies are important for diagnosis. We end with a discussion on how to move the field forward emphasizing on the clinical workup, early identification, and potential targeted treatment of autoimmune PAI.


Subject(s)
Addison Disease , Adrenal Insufficiency , Humans , Addison Disease/diagnosis , Addison Disease/genetics , Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/genetics , Adrenal Insufficiency/therapy , Adrenal Glands , Autoantibodies , Kidney
14.
FEBS Lett ; 597(9): 1261-1274, 2023 05.
Article in English | MEDLINE | ID: mdl-37052889

ABSTRACT

Autoimmune polyendocrine syndrome type I (APS-1) is caused by mutations in the autoimmune regulator (AIRE) gene and characterised clinically by multiple autoimmune manifestations and serologically by autoantibodies against tissue proteins and cytokines. We here hypothesised that lack of AIRE expression in thymus affects blood immune cells and performed whole-blood microarray analysis (N = 16 APS-I patients vs 16 controls), qPCR verification, and bioinformatic deconvolution of cell subsets. We identified B cell responses as being downregulated in APS-1 patients, which was confirmed by qPCR; these results call for further studies on B cells in this disorder. The type I interferon (IFN-I) pathway was also downregulated in APS-1, and the presence of IFN antibodies is the likely reason for this mild overall downregulation of the IFN-I genes in most APS-1 patients.


Subject(s)
Interferon Type I , Polyendocrinopathies, Autoimmune , Humans , Polyendocrinopathies, Autoimmune/genetics , Interferon Type I/genetics , Autoantibodies/genetics , Cytokines/genetics , Mutation
15.
iScience ; 26(7): 107084, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37346050

ABSTRACT

A hallmark of patients with autoimmune polyendocrine syndrome type 1 (APS-1) is serological neutralizing autoantibodies against type 1 interferons (IFN-I). The presence of these antibodies has been associated with severe course of COVID-19. The aims of this study were to investigate SARS-CoV-2 vaccine tolerability and immune responses in a large cohort of patients with APS-1 (N = 33) and how these vaccinated patients coped with subsequent infections. We report that adult patients with APS-1 were able to mount adequate SARS-CoV-2 spike-specific antibody responses after vaccination and observed no signs of decreased tolerability. Compared with age- and gender-matched healthy controls, patients with APS-1 had considerably lower peak antibody responses resembling elderly persons, but antibody decline was more rapid in the elderly. We demonstrate that vaccination protected patients with APS-1 from severe illness when infected with SARS-CoV-2 virus, overriding the systemic danger of IFN-I autoantibodies observed in previous studies.

16.
Diabetes ; 72(1): 59-70, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35709010

ABSTRACT

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.


Subject(s)
Lipodystrophy, Congenital Generalized , Lipodystrophy , Humans , Animals , Mice , Perilipin-1/metabolism , Autoantibodies , Lipodystrophy, Congenital Generalized/metabolism , Lipodystrophy, Congenital Generalized/pathology , Lipodystrophy/metabolism , Adipose Tissue/metabolism
17.
iScience ; 26(6): 106818, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37235056

ABSTRACT

Autoimmune polyendocrine syndrome type 1 (APS-1) is an autosomal recessive disease characterized by severe and childhood onset organ-specific autoimmunity caused by mutations in the autoimmune regulator (AIRE) gene. More recently, dominant-negative mutations within the PHD1, PHD2, and SAND domains have been associated with an incompletely penetrant milder phenotype with later onset familial clustering, often masquerading as organ-specific autoimmunity. Patients with immunodeficiencies or autoimmunity where genetic analyses revealed heterozygous AIRE mutations were included in the study and the dominant-negative effects of the AIRE mutations were functionally assessed in vitro. We here report additional families with phenotypes ranging from immunodeficiency, enteropathy, and vitiligo to asymptomatic carrier status. APS-1-specific autoantibodies can hint to the presence of these pathogenic AIRE variants although their absence does not rule out their presence. Our findings suggest functional studies of heterozygous AIRE variants and close follow-up of identified individuals and their families.

18.
J Clin Immunol ; 32(2): 230-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22127461

ABSTRACT

Patients with autoimmune polyendocrine syndrome type I (APS I) or acquired thymoma-associated myasthenia gravis (MG) surprisingly share several common features, including defective expression of the transcription factor AIRE and autoantibodies against type I interferons. Here, we have adapted and validated the radioligand-binding assay we recently developed against (35)S-Met-interferon-ω, for rapid and specific screening for autoantibodies against interferons-α2 and -α8. We then investigated their potential for diagnosis and for predicting clinical manifestations in patients with APS I and different subgroups of MG. Autoantibodies against interferons-ω, -α2, and -α8 occurred more often in patients with APS I (100%) and MG with thymoma (73%) than in late-onset MG (39%) and early-onset MG (5%). These autoantibodies showed preferences for interferon-ω in APS I and for the interferon-αs in MG, hinting at thymic aberrations in both groups. The exact profile of type I interferon antibodies may indicate MG subtype and may hint at thymoma recurrence.


Subject(s)
Autoantibodies/immunology , Interferon Type I/immunology , Myasthenia Gravis/immunology , Polyendocrinopathies, Autoimmune/immunology , Radioligand Assay , Age Factors , Autoantibodies/blood , Female , Humans , Male , Myasthenia Gravis/epidemiology , Polyendocrinopathies, Autoimmune/epidemiology , Seroepidemiologic Studies , Thymoma/immunology , Thymus Neoplasms/immunology
19.
Biomedicines ; 9(9)2021 Sep 21.
Article in English | MEDLINE | ID: mdl-34572460

ABSTRACT

Autoimmune polyendocrine syndrome type 1 (APS-1) is a rare but severe monogenetic autoimmune endocrine disease caused by failure of the Autoimmune Regulator (AIRE). AIRE regulates the negative selection of T cells in the thymus, and the main pathogenic mechanisms are believed to be T cell-mediated, but little is known about the role of B cells. Here, we give an overview of the role of B cells in thymic and peripheral tolerance in APS-1 patients and different AIRE-deficient mouse models. We also look closely into which autoantibodies have been described for this disorder, and their implications. Based on what is known about B cell therapy in other autoimmune disorders, we outline the potential of B cell therapies in APS-1 and highlight the unresolved research questions to be answered.

20.
Commun Biol ; 4(1): 681, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083746

ABSTRACT

T cells rely for their development and function on the correct folding and turnover of proteins generated in response to a broad range of molecular cues. In the absence of the eukaryotic type II chaperonin complex, CCT, T cell activation induced changes in the proteome are compromised including the formation of nuclear actin filaments and the formation of a normal cell stress response. Consequently, thymocyte maturation and selection, and T cell homeostatic maintenance and receptor-mediated activation are severely impaired. In the absence of CCT-controlled protein folding, Th2 polarization diverges from normal differentiation with paradoxical continued IFN-γ expression. As a result, CCT-deficient T cells fail to generate an efficient immune protection against helminths as they are unable to sustain a coordinated recruitment of the innate and adaptive immune systems. These findings thus demonstrate that normal T cell biology is critically dependent on CCT-controlled proteostasis and that its absence is incompatible with protective immunity.


Subject(s)
Chaperonin Containing TCP-1/immunology , Proteostasis/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chaperonin Containing TCP-1/genetics , Chaperonin Containing TCP-1/metabolism , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Proteome/immunology , Proteome/metabolism , Proteostasis/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL