Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Clin Med ; 13(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38673639

ABSTRACT

Hypocalcemia is a common occurrence in pediatric patients, attributed to various causes and presenting with diverse clinical manifestations. A prompt evaluation is necessary to determine its underlying cause, whether it presents acutely or chronically, and to tailor treatment based on its severity. Among the potential causes of chronic hypocalcemia, primary hypoparathyroidism stands out. The case of a seven-year-old male patient with hypocalcemia reported in this article serves as an illustration, wherein targeted next-generation sequencing revealed a homozygous p.R257X mutation in the AIRE gene, indicative of autoimmune polyendocrine syndrome type 1 (APS-1). It poses challenges due to its multisystemic nature and involvement of specific autoantibodies, often leading to underdiagnosis, owing to its rarity, varied manifestations, and incomplete penetrance. A comprehensive review of the APS-1 literature was conducted to provide insights into the clinical manifestations, genetic spectrum, potential immunological mechanisms, and current medical strategies. Additionally, the recognition of AIRE gene mutations is crucial for facilitating genetic diagnosis, prognosis, and potential treatment strategies for APS-1. The management of such cases involves individualized approaches to treatment, regular monitoring, medication adjustments, and the early identification of associated conditions.

2.
In Vivo ; 38(2): 774-784, 2024.
Article in English | MEDLINE | ID: mdl-38418155

ABSTRACT

BACKGROUND/AIM: Autoimmune regulator (AIRE) is a transcription factor that plays pivotal role in controlling autoimmunity. In the thymus, it supports the presentation of peripheral tissue antigens to developing T cells, where recognition of these self-antigens negatively selects the autoimmune naïve T-cells by central tolerance. Studies demonstrated that single-nucleotide polymorphisms (SNPs) in AIRE alter transcription and propagate clonal survival of autoimmune T cells, therefore increase susceptibility to autoimmune diseases. This study intended to identify SNPs in exon and intron sequences that determine AIRE transcription, where their genotypes are associated with rheumatoid arthritis (RA) risk and clinical parameters. PATIENTS AND METHODS: After a thorough in silico research, we enrolled 100 patients with RA and 100 healthy controls to analyze the association of SNP rs870881(C>T) and rs1003854(T>C) in AIRE coding sequence with RA risk by using five different genetic models and selected clinical parameters. Multiplex quantitative polymerase chain reaction was used to determine allelic discrimination of SNPs. RA risk was assessed by odds ratios (ORs) and confidence intervals (CIs). RESULTS: In a recessive model of rs878081, minor allele TT homozygotes were associated with RA (p=0.032, OR=5.44, 95%CI=1.16-25.52); in a recessive model of rs1003854, minor allele CC homozygotes were associated with RA (p=0.047, OR=4.84, 95%CI=1.02-23.02). Higher C-reactive protein (CRP) levels in patients with RA were significantly associated with minor allele homozygotes in recessive and codominant genetic models (p=0.029 and p=0.043, respectively) of rs1003854. CONCLUSION: Genotypes for minor alleles of rs878081 and rs1003854 might be involved in RA pathogenesis and risk prediction.


Subject(s)
Arthritis, Rheumatoid , Genetic Predisposition to Disease , Humans , Alleles , Arthritis, Rheumatoid/genetics , Case-Control Studies , Genotype , Hungary , Polymorphism, Single Nucleotide
3.
Cell Mol Immunol ; 20(12): 1472-1486, 2023 12.
Article in English | MEDLINE | ID: mdl-37990032

ABSTRACT

The expression of self-antigens in medullary thymic epithelial cells (mTECs) is essential for the establishment of immune tolerance, but the regulatory network that controls the generation and maintenance of the multitude of cell populations expressing self-antigens is poorly understood. Here, we show that Insm1, a zinc finger protein with known functions in neuroendocrine and neuronal cells, is broadly coexpressed with an autoimmune regulator (Aire) in mTECs. Insm1 expression is undetectable in most mimetic cell populations derived from mTECs but persists in neuroendocrine mimetic cells. Mutation of Insm1 in mice downregulated Aire expression, dysregulated the gene expression program of mTECs, and altered mTEC subpopulations and the expression of tissue-restricted antigens. Consistent with these findings, loss of Insm1 resulted in autoimmune responses in multiple peripheral tissues. We found that Insm1 regulates gene expression in mTECs by binding to chromatin. Interestingly, the majority of the Insm1 binding sites are co-occupied by Aire and enriched in superenhancer regions. Together, our data demonstrate the important role of Insm1 in the regulation of the repertoire of self-antigens needed to establish immune tolerance.


Subject(s)
Immune Tolerance , Thymus Gland , Mice , Animals , Mice, Inbred C57BL , Epithelial Cells/metabolism , Autoantigens/metabolism , Cell Differentiation , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Calcif Tissue Int ; 112(6): 675-682, 2023 06.
Article in English | MEDLINE | ID: mdl-36944707

ABSTRACT

Autoimmune polyendocrine syndrome type-1 (APS1) is characterized by autoimmune manifestations affecting different organs from early childhood on. Immunological abnormalities, the resulting endocrinopathies, and their treatments may compromise bone health. For the first time in APS1, we analyzed transiliac bone biopsy samples by bone histomorphometry and quantitative backscattered electron imaging in three adult patients (female P1, 38 years; male P2, 47 years; male P3, 25 years). All had biallelic mutations in the autoimmune regulator gene and in addition to endocrinopathies, also significant bone fragility. Histomorphometry showed bone volume in the lower normal range for P1 (BV/TV, - 0.98 SD) and P3 (- 1.34 SD), mainly due to reduced trabecular thickness (TbTh, - 3.63 and - 2.87 SD). In P1, osteoid surface was low (OS/BS, - 0.96 SD); active osteoblasts and double labeling were seen only on cortical bone. P3 showed a largely increased bone turnover rate (BFR/BV, + 4.53 SD) and increased mineralization lag time (Mlt, + 3.40 SD). Increased osteoid surface (OS/BS, + 2.03 and + 4.71 SD for P2 and P3) together with a large proportion of lowly mineralized bone area (Trab CaLow, + 2.22 and + 9.81 SD for P2 and P3) and focal mineralization defects were consistent with abnormal mineralization. In all patients, the density and area of osteocyte lacunae in cortical and trabecular bone were similar to healthy adults. The bone tissue characteristics were variable and included decreased trabecular thickness, increased amount of osteoid, and abnormal mineralization which are likely to contribute to bone fragility in patients with APS1.


Subject(s)
Bone Density , Polyendocrinopathies, Autoimmune , Adult , Humans , Male , Child, Preschool , Female , Polyendocrinopathies, Autoimmune/genetics , Bone and Bones , Cortical Bone , Bone Matrix
5.
Int J Mol Sci ; 24(5)2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36902438

ABSTRACT

This study aimed to assess the possible association between two single nucleotide polymorphisms (SNPs) of the autoimmune regulator (AIRE) gene (rs2075876 G/A and rs760426 A/G) with the risk of primary immune thrombocytopenia (ITP), as well as AIRE serum levels, in the Egyptian population. In this case-control study, 96 cases with primary ITP and 100 healthy subjects were included. Two SNPs of the AIRE gene (rs2075876 G/A and rs760426 A/G) were genotyped via Taqman allele discrimination real-time polymerase chain reaction (PCR). Additionally, serum AIRE levels were measured using the enzyme-linked immunosorbent assay (ELISA) technique. After adjusting for age, gender, and family history of ITP, the AIRE rs2075876 AA genotype and A allele were associated with increased ITP risk (adjusted odds ratio (aOR): 4.299, p = 0.008; aOR: 1.847, p = 0.004, respectively). Furthermore, there was no significant association between AIRE rs760426 A/G different genetic models and ITP risk. A linkage disequilibrium revealed that A-A haplotypes were associated with an increased ITP risk (aOR: 1.821, p = 0.020). Serum AIRE levels were found to be significantly lower in the ITP group, positively correlated with platelet counts, and were even lower in the AIRE rs2075876 AA genotype and A allele, as well as A-G and A-A haplotype carriers (all p < 0.001). The AIRE rs2075876 genetic variants (AA genotype and A allele) and A-A haplotype are associated with an increased ITP risk in the Egyptian population and lower serum AIRE levels, whereas the SNP rs760426 A/G is not.


Subject(s)
Purpura, Thrombocytopenic, Idiopathic , Transcription Factors , Humans , Case-Control Studies , Genetic Predisposition to Disease , Genotype , Polymorphism, Single Nucleotide , Purpura, Thrombocytopenic, Idiopathic/genetics , Transcription Factors/genetics , AIRE Protein
6.
J Pathol ; 260(2): 222-234, 2023 06.
Article in English | MEDLINE | ID: mdl-36853094

ABSTRACT

Autoimmune regulator (Aire) and TGF-ß signaling play important roles in central tolerance and peripheral tolerance, respectively, by eliminating or suppressing the activity of autoreactive T cells. We previously demonstrated that dnTGFßRII mice develop a defect in peripheral tolerance and a primary biliary cholangitis (PBC)-like disease. We hypothesized that by introducing the Aire gene to this model, we would observe a more severe PBC phenotype. Interestingly, however, we demonstrated that, while dnTGFßRII Aire-/- mice do manifest key histological and serological features of autoimmune cholangitis, they also develop mild to moderate interface hepatitis and show high levels of alanine transaminase (ALT) and antinuclear antibodies (ANA), characteristics of autoimmune hepatitis (AIH). To further understand this unique phenotype, we performed RNA sequencing (RNA-seq) and flow cytometry to explore the functional pathways and immune cell pathways in the liver of dnTGFßRII Aire-/- mice. Our data revealed enrichments of programmed cell death pathways and predominant CD8+ T cell infiltrates. Depleting CD8+ T cells using an anti-CD8α antibody significantly alleviated hepatic inflammation and prolonged the life span of these mice. Finally, RNA-seq data indicated the clonal expansion of hepatic CD8+ T cells. In conclusion, these mice developed an autoreactive CD8+ T-cell-mediated autoimmune cholangitis with concurrent hepatitis that exhibited key histological and serological features of the AIH-PBC overlap syndrome, representing a novel model for the study of tolerance and autoimmune liver disease. © 2023 The Pathological Society of Great Britain and Ireland.


Subject(s)
Cholangitis , Hepatitis, Autoimmune , Liver Cirrhosis, Biliary , Mice , Animals , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/metabolism , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , CD8-Positive T-Lymphocytes , Cholangitis/genetics , Cholangitis/metabolism
7.
Am J Reprod Immunol ; 89(2): e13637, 2023 02.
Article in English | MEDLINE | ID: mdl-36305192

ABSTRACT

PROBLEM: Autoimmune polyendocrinopathy-candidiasis- ectodermal dystrophy (APECED) pathology due to autoimmune regulator (AIRE) gene mutations leads to loss of central tolerance triggering immune attack, a factor causing infertility. One of the targets of autoimmune attack is ovary and its repercussion results in polycystic ovarian syndrome (PCOS). Although reduced Tregs have been reported in PCOS, a lacunae exists on the status of AIRE gene expression and its role in treg insufficiency via HIF1A-FOXP3 axis in PCOS. METHOD OF STUDY: This is a case-control cohort study recruiting 40 normal and 40 PCOS volunteers for peripheral blood sample collection and PCOS diagnoses were based on Rotterdam Consensus criteria. AIRE and HIF1A expression status was analysed by qRT PCR and western blot. FACS analyses was conducted on AIRE silenced peripheral blood mononuclear cells (PBMCs) after Treg induction. RESULTS: Our results indicate a reduced AIRE (fold change log2 (RQ) = -2.6, P < .01) and increased HIF1A (fold change log2 (RQ) = 3.6, P < .02) in PBMCs of PCOS subjects compared to age-matched controls. Western blot of AIRE and HIF1A corroborates with qRT PCR data. Our CHIP data demonstrate AIRE mediated HIF1A promoter regulation. Silencing of AIRE in PBMCs contributes to the upregulation of HIF1A transcripts by two-fold (P < .0015) and downregulation in FOXP3 expression by three-fold (P < .0017). FACS analyses revealed that silencing of AIRE reduces Tcell to Treg conversion. CONCLUSIONS: Our consolidated results derive a new connection among AIRE-HIF1A-FOXP3 with AIRE reduction enabling increased HIF1A resulting in reduced FOXP3 in PBMCs of PCOS patients leading to Treg insufficiency.


Subject(s)
Forkhead Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Polycystic Ovary Syndrome , Transcription Factors , Female , Humans , Case-Control Studies , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukocytes, Mononuclear/metabolism , Polycystic Ovary Syndrome/genetics , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/metabolism , AIRE Protein
8.
Front Cell Dev Biol ; 10: 948350, 2022.
Article in English | MEDLINE | ID: mdl-36072346

ABSTRACT

Autoimmune polyendocrine syndrome type-1 (APS-1) is a rare inherited monogenic autoimmune disease characterized by the presence of at least two of three following major clinical features: chronic mucocutaneous candidiasis, hypoparathyroidism, and adrenal insufficiency. Mutations in autoimmune regulator (AIRE) gene have been found to contribute to APS-1. In the present study, we reported a 36-years-old male APS-1 patient who presented with hypoparathyroidism and Addison's disease. The proband underwent complete clinical examinations and mutation screening was performed by Sanger sequencing on AIRE gene. A novel homozygous mutation in exon 9 of the AIRE gene (c.1024C>T) was identified. Based on sequencing findings, HEK293T cell-based assays were conducted to analyze the subcellular localization and mutant transcript processing. Our results revealed that p.Q342X mutant localized in nuclear speckles and exerted a dominant-negative effect on wildtype AIRE function. We reported the c.1024C>T mutation of AIRE gene for the first time, which enriched the AIRE mutation database and contributed to further understanding of APS-1.

9.
Autoimmun Rev ; 21(9): 103141, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35840039

ABSTRACT

Auto-immune regulator (AIRE) is a transcription factor that is mainly known for its crucial role in the thymus. Here, AIRE ensures central tolerance by promoting the expression of peripheral tissue antigens in thymic epithelial cells, which is essential for the negative selection of autoreactive T cells. Intriguingly, AIRE expressing cells have recently been identified in other tissues outside the thymus as well. However, the exact function of these extrathymic AIRE expressing cells (eTACs) remains largely enigmatic. Human eTACs are mainly found in secondary lymphoid tissues under homeostatic conditions, but are also found in pathologies such as the inflamed tissues of patients with autoimmune diseases and in various cancer tissues. eTACs have been demonstrated to express dendritic cell (DC)-like markers, such as MHCII, CD40 and CD127, but also CCR7, IDO and PD-L1. Interestingly, eTACs lack high expression of co-stimulatory molecules, such as CD80 or CD86. In mice, different types of peripheral AIRE expressing cells have been described, including cells with an innate lymphoid cell-like phenotype and antigen presenting cell (APC) function. These findings suggest that eTACs are APCs with the possibility to modulate or inhibit immune responses, which is confirmed by functional murine studies demonstrating the ability of eTACs to induce tolerance in autoreactive T cells. The potential immunomodulatory function of eTACs makes them promising targets to restore tolerance in autoimmunity or improve immunotherapy in cancer settings. Yet, this requires a better understanding of these cells and the molecular mechanisms involved. In this review we aim to summarize the current knowledge and understanding of eTACs, including their putative roles in health and disease.


Subject(s)
Autoimmune Diseases , Neoplasms , Animals , Autoimmunity , Humans , Immune Tolerance , Immunity, Innate , Lymphocytes , Mice , Neoplasms/therapy , Thymus Gland
10.
Cell Mol Life Sci ; 79(7): 355, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35678896

ABSTRACT

Thymically-derived Foxp3+ regulatory T cells (Treg) critically control immunological tolerance. These cells are generated in the medulla through high affinity interactions with medullary thymic epithelial cells (mTEC) expressing the Autoimmune regulator (Aire). Recent advances have revealed that thymic Treg contain not only developing but also recirculating cells from the periphery. Although Aire is implicated in the generation of Foxp3+ Treg, its role in the biology of recirculating Treg remains elusive. Here, we show that Aire regulates the suppressive signature of recirculating Treg independently of the remodeling of the medullary 3D organization throughout life where Treg reside. Accordingly, the adoptive transfer of peripheral Foxp3+ Treg in AireKO recipients led to an impaired suppressive signature upon their entry into the thymus. Furthermore, recirculating Treg from AireKO mice failed to attenuate the severity of multiorgan autoimmunity, demonstrating that their suppressive function is altered. Using bone marrow chimeras, we reveal that mTEC-specific expression of Aire controls the suppressive signature of recirculating Treg. Finally, mature mTEC lacking Aire were inefficient in stimulating peripheral Treg both in polyclonal and antigen-specific co-culture assays. Overall, this study demonstrates that Aire confers to mTEC the ability to restimulate recirculating Treg, unravelling a novel function for this master regulator in Treg biology.


Subject(s)
Immune Tolerance , T-Lymphocytes, Regulatory , Animals , Autoimmunity , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Thymus Gland
11.
Autoimmun Rev ; 21(5): 103058, 2022 May.
Article in English | MEDLINE | ID: mdl-35108619

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that causes a deficit of pancreatic islet ß cells. Millions of individuals worldwide have T1D, and its incidence increases annually. Recent clinical trials have highlighted the limits of conventional immunotherapy in T1D and underscore the need for novel treatments that not only overcome multiple immune dysfunctions, but also help restore islet ß-cell function. To address these two key issues, we have developed a unique and novel procedure designated the Stem Cell Educator therapy, based on the immune education by cord-blood-derived multipotent stem cells (CB-SC). Over the last 10 years, this technology has been evaluated through international multi-center clinical studies, which have demonstrated its clinical safety and efficacy in T1D and other autoimmune diseases. Mechanistic studies revealed that Educator therapy could fundamentally correct the autoimmunity and induce immune tolerance through multiple molecular and cellular mechanisms such as the expression of a master transcription factor autoimmune regulator (AIRE) in CB-SC for T-cell modulation, an expression of Galectin-9 on CB-SC to suppress activated B cells, and secretion of CB-SC-derived exosomes to polarize human blood monocytes/macrophages into type 2 macrophages. Educator therapy is the leading immunotherapy to date to safely and efficiently correct autoimmunity and restore ß cell function in T1D patients.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Autoimmunity , Diabetes Mellitus, Type 1/therapy , Fetal Blood/metabolism , Humans , Insulin-Secreting Cells/metabolism , Stem Cells
12.
Acta Anatomica Sinica ; (6): 453-460, 2022.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1015303

ABSTRACT

[Abstract] Objective To investigate the relationship between the expression of Wnt signaling pathway, autoimmune regulator (AIRE) and type 1 diabetes (T1D) tissue specific antigen (TSAs) insulin 2(Ins2) and glutamic acid decarboxybase(GAD67) in thymus and the occurrence of T1D in NOD/ Ltj mice with spontaneous type 1 diabetes (T1D). Methods Sixty female NOD/ Ltj mice were divided into three groups: 3 weeks group, 16 weeks non-onset group and 16 weeks onset group. Two consecutive non-fasting blood glucose levels ≥ 11. 1 mmol/ L were considered as the occurrence of T1D. Pancreatic HE staining was used to observe the occurrence of islet inflammation. Anti-Ins and CD45 immunohistochemical staining showed islet cells or infiltrating inflammatory cells. The protein levels and mRNA expressions of Wnt7a, -catenin, AIRE, Ins and GAD67 in thymus were detected by Western blotting and Real-time PCR. The proportion of T cells in thymus was analyzed by flow cytometry. Results 1. With the occurrence of T1D, the islet structure was destroyed, a large number of lymphocytes infiltrated, and the remaining islet cells were reduced. A large number of CD45

13.
BMC Endocr Disord ; 21(1): 154, 2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34344344

ABSTRACT

BACKGROUND: Autoimmune polyendocrine syndrome type 1 (APS-1) is a rare monogenic inherited disease caused by mutations of the autoimmune regulator gene (AIRE). The three major components of this syndrome are chronic mucocutaneous candidiasis, hypoparathyroidism and adrenocortical insufficiency. CASE PRESENTATION: We report a 20-year-old male who was clinically diagnosed with APS-1 at the age of 15. He was admitted to our department this time for suffering from polyuria and polydipsia for 6 months and was finally diagnosed with diabetes insipidus. Whole-exome sequencing (WES) revealed a novel compound heterozygous mutation of the AIRE gene -the c.239 T > G (p.Val80Gly) variant on one allele and the copy number variant (CNV) of 21q22.3(chr21:45,670,150-45,706,528)*1 on the other. CONCLUSIONS: This case suggests that diabetes insipidus is a rare component of APS-1 and expands the variety of mutations on AIRE gene.


Subject(s)
Diabetes Insipidus/pathology , Genetic Predisposition to Disease , Mutation , Polyendocrinopathies, Autoimmune/pathology , Transcription Factors/genetics , Adult , Diabetes Insipidus/complications , Diabetes Insipidus/genetics , Humans , Male , Phenotype , Polyendocrinopathies, Autoimmune/complications , Polyendocrinopathies, Autoimmune/genetics , Prognosis , Young Adult , AIRE Protein
14.
Front Endocrinol (Lausanne) ; 12: 645279, 2021.
Article in English | MEDLINE | ID: mdl-34194389

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a rare monogenic disorder, associated with endocrine deficiencies and non-endocrine involvement. Gastrointestinal (GI) manifestations appear in approximately 25% of patients and are the presenting symptom in about 10% of them. Limited awareness among pediatricians of autoimmune enteropathy (AIE) caused by destruction of the gut endocrine cells in APECED patients delays diagnosis and appropriate therapy. We describe an 18-year-old female presenting at the age of 6.10 years with hypoparathyroidism, oral candidiasis and vitiligo. The clinical diagnosis of APECED was confirmed by sequencing the autoimmune regulator-encoding (AIRE) gene. Several characteristics of the disease-Hashimoto's thyroiditis, Addison's disease, diabetes mellitus type 1 and primary ovarian insufficiency-developed over the years. She had recurrent episodes of severe intractable hypocalcemia. Extensive GI investigations for possible malabsorption, including laboratory analyses, imaging and endoscopy with biopsies were unremarkable. Revision of the biopsies and chromogranin A (CgA) immunostaining demonstrated complete loss of enteroendocrine cells in the duodenum and small intestine, confirming the diagnosis of AIE. Management of hypocalcemia was challenging. Only intravenous calcitriol maintained calcium in the normal range. Between hypocalcemic episodes, the proband maintained normal calcium levels, suggesting a fluctuating disease course. Repeated intestinal biopsy revealed positive intestinal CgA immunostaining. The attribution of severe hypocalcemic episodes to AIE emphasizes the need for increased awareness of this unique presentation of APECED. The fluctuating disease course and repeated intestinal biopsy showing positive CgA immunostaining support a reversible effect of GI involvement. CgA immunostaining is indicated in patients with APECED for whom all other investigations have failed to reveal an explanation for the malabsorption.


Subject(s)
Hypocalcemia/immunology , Hypocalcemia/physiopathology , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/physiopathology , Adolescent , Biopsy , Calcitriol/metabolism , Candidiasis/complications , Chromogranin A/pharmacology , Endocrine Cells , Female , Humans , Hypocalcemia/complications , Hypoparathyroidism/complications , Intestines/metabolism , Polyendocrinopathies, Autoimmune/complications , Rheumatology , Sequence Analysis, DNA , Transcription Factors/genetics , Vitamin D/metabolism , Vitiligo/complications , AIRE Protein
15.
Acta Pharm Sin B ; 11(2): 309-321, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643814

ABSTRACT

Cullin-RING ligases (CRLs) recognize and interact with substrates for ubiquitination and degradation, and can be targeted for disease treatment when the abnormal expression of substrates involves pathologic processes. Phosphorylation, either of substrates or receptors of CRLs, can alter their interaction. Phosphorylation-dependent ubiquitination and proteasome degradation influence various cellular processes and can contribute to the occurrence of various diseases, most often tumorigenesis. These processes have the potential to be used for tumor intervention through the regulation of the activities of related kinases, along with the regulation of the stability of specific oncoproteins and tumor suppressors. This review describes the mechanisms and biological functions of crosstalk between phosphorylation and ubiquitination, and most importantly its influence on tumorigenesis, to provide new directions and strategies for tumor therapy.

16.
Semin Immunopathol ; 43(1): 65-83, 2021 02.
Article in English | MEDLINE | ID: mdl-33532929

ABSTRACT

During their intrathymic development, nascent T cells are empowered to protect against pathogens and to be operative for a life-long acceptance of self. While autoreactive effector T (Teff) cell progenitors are eliminated by clonal deletion, the intrathymic mechanisms by which thymic regulatory T cell (tTreg) progenitors maintain specificity for self-antigens but escape deletion to exert their regulatory functions are less well understood. Both tTreg and Teff development and selection result from finely coordinated interactions between their clonotypic T cell receptors (TCR) and peptide/MHC complexes expressed by antigen-presenting cells, such as thymic epithelial cells and thymic dendritic cells. tTreg function is dependent on expression of the FOXP3 transcription factor, and induction of FOXP3 gene expression by tTreg occurs during their thymic development, particularly within the thymic medulla. While initial expression of FOXP3 is downstream of TCR activation, constitutive expression is fixed by interactions with various transcription factors that are regulated by other extracellular signals like TCR and cytokines, leading to epigenetic modification of the FOXP3 gene. Most of the understanding of the molecular events underlying tTreg generation is based on studies of murine models, whereas gaining similar insight in the human system has been very challenging. In this review, we will elucidate how inborn errors of immunity illuminate the critical non-redundant roles of certain molecules during tTreg development, shedding light on how their abnormal development and function cause well-defined diseases that manifest with autoimmunity alone or are associated with states of immune deficiency and autoinflammation.


Subject(s)
Autoimmunity , Forkhead Transcription Factors , Animals , Epithelial Cells , Forkhead Transcription Factors/genetics , Humans , Mice , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory
17.
Eur J Immunol ; 51(2): 311-318, 2021 02.
Article in English | MEDLINE | ID: mdl-32845012

ABSTRACT

Autoimmune regulator+ (Aire) medullary thymic epithelial cells (mTECs) play a critical role in tolerance induction. Several studies demonstrated that Aire+ mTECs differentiate further into Post-Aire cells. Yet, the identification of terminal stages of mTEC maturation depends on unique fate-mapping mouse models. Herein, we resolve this limitation by segmenting the mTEChi (MHCIIhi CD80hi ) compartment into mTECA/hi (CD24- Sca1- ), mTECB/hi (CD24+ Sca1- ), and mTECC/hi (CD24+ Sca1+ ). While mTECA/hi included mostly Aire-expressing cells, mTECB/hi contained Aire+ and Aire- cells and mTECC/hi were mainly composed of cells lacking Aire. The differential expression pattern of Aire led us to investigate the precursor-product relationship between these subsets. Strikingly, transcriptomic analysis of mTECA/hi , mTECB/hi , and mTECC/hi sequentially mirrored the specific genetic program of Early-, Late- and Post-Aire mTECs. Corroborating their Post-Aire nature, mTECC/hi downregulated the expression of tissue-restricted antigens, acquired traits of differentiated keratinocytes, and were absent in Aire-deficient mice. Collectively, our findings reveal a new and simple blueprint to survey late stages of mTEC differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/immunology , Epithelial Cells/immunology , Keratinocytes/immunology , Thymus Gland/immunology , Transcription Factors/genetics , Animals , Down-Regulation/genetics , Down-Regulation/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Transcription Factors/immunology , AIRE Protein
18.
Front Cardiovasc Med ; 8: 812769, 2021.
Article in English | MEDLINE | ID: mdl-35097028

ABSTRACT

Atherosclerosis is a chronic, lipid-driven disease of medium sized arteries which causes myocardial infarction and stroke. Recently, an adaptive immune response against the plaque-associated autoantigen Apolipoprotein B100 (ApoB), the structural protein component of low-density lipoprotein, has been implicated in atherogenesis. In healthy individuals, CD4+ T cells responding to ApoB mainly comprised regulatory T cells, which confer immune tolerance and atheroprotection. Mice and patients with atherosclerosis harbor increased numbers of proatherogenic ApoB-reactive T-helper cell subsets. Given the lack of therapies targeting proatherogenic immunity, clarification of the underlying mechanisms is of high clinical relevance. T cells develop in the thymus, where strong autoreactive T cells are eliminated in the process of negative selection. Herein, we investigated whether the transcription factor autoimmune regulator (AIRE), which controls expression of numerous tissue-restricted self-antigens in the thymus, is involved in mediating tolerance to ApoB and whether Aire deficiency might contribute to atherogenesis. Mice deficient for Aire were crossbred to apolipoprotein E-deficient mice to obtain atherosclerosis-prone Aire -/- Apoe -/- mice, which were fed a regular chow diet (CD) or western-type diet (WD). CD4+ T cells responding to the ApoB peptide p6 were analyzed by flow cytometry. We demonstrate that Aire deficiency influences neither generation nor activation of ApoB-reactive T cells and has only minor and overall inconsistent impacts on their phenotype. Furthermore, we show that atherosclerotic plaque size is not affected in Aire -/- Apoe -/- compared to Aire +/+ Apoe -/-, irrespective of diet and gender. In conclusion, our data suggests that AIRE is not involved in regulating thymic expression of ApoB or atherosclerosis. Alternative mechanisms how ApoB-reactive CD4 T cells are selected in the thymus will have to be investigated.

19.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 411-420, 2021 02.
Article in English | MEDLINE | ID: mdl-32686020

ABSTRACT

Senescence-related decline of thymus affects immune function in the elderly population and contributes to the prevalence of many relevant diseases like cancer, autoimmune diseases, and other chronic diseases. In this study, we investigated the therapeutic effects of curcumin, an agent that could counter aging, and explored its optimal intake and the alteration of autoimmune regulator (Aire) after curcumin treatment in the D-galactose (D-gal)-induced accelerated aging mice. ICR mice were intraperitoneally injected with D-gal for 8 weeks to establish the accelerated aging model and given curcumin with 50, 100, and 200 mg/kg body weight per day by gavage, respectively, for 6 weeks. It indicated that the D-gal-treated mice developed structural changes in the thymi compared with the control group without D-gal and curcumin treatment. As the supplements of curcumin, it resulted in a restoration of the normal thymic anatomy with an increase of proliferating cells and a reduction of apoptotic cells in the thymi of the D-gal-induced aging model mice. Curcumin administration could also expand the expression level of Aire from mRNA level and protein level. The current study demonstrated that curcumin could ameliorate senescence-related thymus involution via upregulating Aire expression, suggesting that curcumin can rejuvenate senescence-associated alterations of thymus induced by D-gal accumulation.


Subject(s)
Cellular Senescence/drug effects , Curcumin/pharmacology , Protective Agents/pharmacology , Thymus Gland/drug effects , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Galactose , Mice, Inbred ICR , Thymus Gland/metabolism , Transcription Factors/genetics , AIRE Protein
20.
Acta Pharmaceutica Sinica B ; (6): 309-321, 2021.
Article in English | WPRIM (Western Pacific) | ID: wpr-881138

ABSTRACT

Cullin-RING ligases (CRLs) recognize and interact with substrates for ubiquitination and degradation, and can be targeted for disease treatment when the abnormal expression of substrates involves pathologic processes. Phosphorylation, either of substrates or receptors of CRLs, can alter their interaction. Phosphorylation-dependent ubiquitination and proteasome degradation influence various cellular processes and can contribute to the occurrence of various diseases, most often tumorigenesis. These processes have the potential to be used for tumor intervention through the regulation of the activities of related kinases, along with the regulation of the stability of specific oncoproteins and tumor suppressors. This review describes the mechanisms and biological functions of crosstalk between phosphorylation and ubiquitination, and most importantly its influence on tumorigenesis, to provide new directions and strategies for tumor therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...