Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Thyroid ; 33(4): 492-500, 2023 04.
Article in English | MEDLINE | ID: mdl-36762945

ABSTRACT

Background: Autoimmune thyroid diseases (AITD) represent the most common autoimmune diseases. However, current therapies focus on relieving the symptoms instead of curing AITD, and new therapies to reverse the autoimmune attack on the thyroid are needed. HLA-DRß1-Arg74 is the key HLA class II allele that triggers AITD by presenting pathogenic thyroglobulin (Tg) peptides that activate thyroid self-reactive T cells. We hypothesized that blocking the presentation of Tg peptides to T cells within the HLA-DRß1-Arg74 peptide binding cleft could reverse the autoimmune response to the thyroid in AITD. Methods: The approach we used to block Tg peptide presentation within HLA-DRß1-Arg74 is to design retro-inverso D-amino acid (RID) peptides that have high affinity to the HLA-DRß1-Arg74 peptide binding pocket. Results: By using computational approaches and molecular dynamics simulations, we designed two RID peptides, RT-15 and VT-15, that blocked peptide binding to recombinant HLA-DRß1-Arg74 molecule, as well as T cell activation in vitro. Furthermore, RT-15 and VT-15 blocked in vivo T cell activation by thyroglobulin in humanized NOD-DR3 mice induced with experimental autoimmune thyroiditis. Conclusions: In summary, we discovered two RID peptides that block thyroglobulin peptide binding to HLA-DRß1-Arg74 and their presentation to T cells in AITD. These findings set the stage for a personalized medicine therapeutic approach for AITD patients who carry the DRß1-Arg74 allele. This antigen-specific therapeutic strategy can potentially be extended to other autoimmune diseases.


Subject(s)
Autoimmune Diseases , Hashimoto Disease , Thyroiditis, Autoimmune , Mice , Animals , Thyroiditis, Autoimmune/drug therapy , Thyroglobulin , Autoimmunity , Antigen Presentation , Mice, Inbred NOD , Peptides/chemistry
2.
Best Pract Res Clin Endocrinol Metab ; 37(2): 101661, 2023 03.
Article in English | MEDLINE | ID: mdl-35459628

ABSTRACT

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


Subject(s)
Autoimmune Diseases , Graves Disease , Hashimoto Disease , Thyroid Diseases , Humans , Genetic Predisposition to Disease , Hashimoto Disease/genetics , Hashimoto Disease/complications , Autoimmune Diseases/complications , Epigenesis, Genetic , Thyroid Diseases/complications
3.
J Clin Endocrinol Metab ; 106(11): e4680-e4687, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34143178

ABSTRACT

CONTEXT: Genetic risk factors play a major role in the pathoetiology of autoimmune thyroid diseases (AITD). So far, only common risk variants have been identified in AITD susceptibility genes. Recently, rare genetic variants have emerged as important contributors to complex diseases, and we hypothesized that rare variants play a key role in the genetic susceptibility to AITD. OBJECTIVE: We aimed to identify new rare variants that are associated with familial AITD. METHODS: We performed deep sequencing of 3 previously mapped AITD-linked loci (10q, 12q, and 14q) in a dataset of 34 families in which AITD clustered (familial AITD). RESULTS: We identified 13 rare variants, located in the inositol polyphosphate multikinase (IPMK) gene, that were associated with AITD (ie, both Graves' disease [GD] and Hashimoto's thyroiditis [HT]); 2 rare variants, within the dihydrolipoamide S-succinyltransferase (DLST) and zinc-finger FYVE domain-containing protein (ZFYVE1) genes, that were associated with GD only; and 3 rare variants, within the phosphoglycerate mutase 1 pseudogene 5 (PGAM1P5), LOC105369879, and methionine aminopeptidase 2 (METAP2) genes, that were associated with HT only. CONCLUSION: Our study demonstrates that, in addition to common variants, rare variants also contribute to the genetic susceptibility to AITD. We identified new rare variants in 6 AITD susceptibility genes that predispose to familial AITD. Of these, 3 genes, IPMK, ZFYVE1, and METAP2, are mechanistically involved in immune pathways and have been previously shown to be associated with autoimmunity. These genes predispose to thyroid autoimmunity and may serve as potential therapeutic targets in the future.


Subject(s)
Autoimmune Diseases/pathology , Biomarkers/metabolism , Genetic Load , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing/methods , Polymorphism, Single Nucleotide , Thyroid Diseases/pathology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Female , Genotype , Humans , Male , Prognosis , Thyroid Diseases/genetics , Thyroid Diseases/metabolism
4.
Front Immunol ; 12: 796552, 2021.
Article in English | MEDLINE | ID: mdl-34987519

ABSTRACT

Autoimmune polyglandular syndrome type 3 variant (APS3v) refers to an autoimmune condition in which both type 1 diabetes (T1D) and autoimmune thyroiditis (AITD) develop in the same individual. HLA-DR3 confers the strongest susceptibility to APS3v. Previously we reported a unique amino acid signature pocket that predisposes to APS3v. We found that this pocket is flexible and can trigger APS3v by presenting both thyroid (Tg.1571, TPO.758) and islet (GAD.492) peptides to induce autoimmune response. We hypothesized that blocking the specific APS3v-HLA-DR3 pocket from presenting thyroid/islet antigens can block the autoimmune response in APS3v. To test this hypothesis we performed a virtual screen of small molecules blocking APS3v-HLA-DR3, and identified 11 small molecules hits that were predicted to block APS3v-HLA-DR3. Using the baculovirus-produced recombinant APS3v-HLA-DR3 protein we tested the 11 small molecules in an in vitro binding assay. We validated 4 small molecule hits, S9, S5, S53 and S15, that could block the APS3v-HLA-DR3 pocket in vitro. We then developed a novel humanized APS3v mouse model induced by co-immunizing a peptide mix of Tg.1571, TPO.758 and GAD.492. The immunized mice developed strong T-cell and antibody responses to the thyroid/islet peptides, as well as mouse thyroglobulin. In addition, the mice showed significantly lower free T4 levels compared to controls. Using the APS3v mouse model, we showed that one of the 4 small molecules, Cepharanthine (S53), blocked T-cell activation by thyroid/islet peptides ex vivo and in vivo. These findings suggested Cepharanthine may have a therapeutic potential in APS3v patients carrying the specific APS3v-HLA-DR3 pocket.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzylisoquinolines/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , HLA-DR3 Antigen/metabolism , Islets of Langerhans/immunology , Polyendocrinopathies, Autoimmune/drug therapy , T-Lymphocytes/immunology , Thyroiditis, Autoimmune/drug therapy , Animals , Antigen Presentation , Autoantigens/immunology , Binding Sites/genetics , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Genetic Predisposition to Disease , Glutamate Decarboxylase/immunology , HLA-DR3 Antigen/genetics , Humans , Immunity, Humoral , Immunization , Iodide Peroxidase/immunology , Iron-Binding Proteins/immunology , Lymphocyte Activation , Mice , Mice, SCID , Peptide Fragments/genetics , Peptide Fragments/immunology , Polyendocrinopathies, Autoimmune/immunology , Thyroglobulin/genetics , Thyroglobulin/immunology , Thyroiditis, Autoimmune/immunology
5.
J Autoimmun ; 108: 102402, 2020 03.
Article in English | MEDLINE | ID: mdl-31980336

ABSTRACT

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


Subject(s)
Antigen Presentation/drug effects , Antigen Presentation/immunology , Benzylisoquinolines/pharmacology , HLA-DR3 Antigen/immunology , Peptides/antagonists & inhibitors , Peptides/immunology , Receptors, Thyrotropin/immunology , Amino Acid Sequence , Animals , Benzylisoquinolines/chemistry , Epitope Mapping/methods , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Graves Disease/diagnosis , Graves Disease/drug therapy , Graves Disease/immunology , HLA-DR3 Antigen/genetics , Humans , Immunohistochemistry , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Models, Molecular , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Peptides/chemistry , Protein Binding , Receptors, Thyrotropin/chemistry , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Endocrinology ; 158(2): 410-418, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27929668

ABSTRACT

CD40, a tumor necrosis factor receptor, is a major immune-modulating susceptibility gene for Graves disease (GD) as well as for a variety of other autoimmune diseases. Its broad association with autoimmunity underscores its paramount role in the development of a normal adaptive immune response, primarily in coordinating effective antigen presentation. The molecular pathways by which CD40 activation in the thyroid induces GD are unknown. In this study, we investigated whether NF-κB, a ubiquitious family of transcription factors, mediates the downstream effects of thyroid-specific CD40 activation. Cultured primary human thyrocytes, from patients with and without GD, underwent CD40 stimulation. Once stimulated, cytokines and transcription factors specific for either the canonical nuclear factor κB (NF-κB)1 pathway [interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α], which primarily recruits cells for innate immunity, or the noncanonical NF-κB2 pathway [B cell-activating factor of the TNF family, CC chemokine ligand (CCL)21], which directs B cell viability, were analyzed. Significant upregulation in the messenger RNA and protein levels of both canonical and noncanonical pathway cytokines was observed. Western blot analyses of the specific transcription factors for the NF-κB1 and NF-κB2 pathways (p65 and p100/p52, respectively) demonstrated that p65 is constitutively expressed. In contrast, CD40 stimulation robustly increased the expression of the NF-κB2 p52 transcription factor, and the upregulation was significantly more profound in the GD tissue than in the normal thyroid tissue. Our data show that CD40 activity in thyrocytes is prominently mediated via NF-κB and furthermore suggest that the NF-κB1 and NF-κB2 pathways both contribute to the triggering and the progression of GD.


Subject(s)
CD40 Antigens/metabolism , Graves Disease/etiology , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Thyroid Epithelial Cells/metabolism , Cells, Cultured , Graves Disease/metabolism , Humans
7.
J Autoimmun ; 76: 1-9, 2017 01.
Article in English | MEDLINE | ID: mdl-27670087

ABSTRACT

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


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DR Antigens/immunology , Peptides/immunology , T-Lymphocytes/immunology , Thyroiditis, Autoimmune/immunology , Amino Acid Sequence , Animals , Binding Sites , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , HLA-DR Antigens/chemistry , HLA-DR Antigens/metabolism , Humans , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Binding/immunology , Protein Conformation , T-Lymphocytes/metabolism , Thyroid Gland/immunology , Thyroiditis, Autoimmune/metabolism
8.
J Biol Chem ; 291(8): 4079-90, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26703475

ABSTRACT

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


Subject(s)
Alkaloids/pharmacology , Antigen Presentation/drug effects , HLA-DRB1 Chains/immunology , Thyroiditis, Autoimmune/immunology , Alkaloids/chemistry , Animals , HLA-DRB1 Chains/genetics , Humans , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Peptides/genetics , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thyroglobulin/genetics , Thyroglobulin/immunology , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/pathology
9.
J Autoimmun ; 64: 82-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26235382

ABSTRACT

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


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Genetic Predisposition to Disease , Immunogenetics , Thyroid Diseases/genetics , Thyroid Diseases/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Genetic Loci , Humans , Immune Tolerance/genetics , Phenotype , Thyroid Diseases/diagnosis , Thyroid Diseases/metabolism
10.
Gene ; 556(2): 142-8, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25481456

ABSTRACT

We have previously shown that a (TC)n microsatellite in intron 5 of the Forkhead Box Protein 3 (FOXP3) gene was associated with a variant of the autoimmune polyglandular syndrome type 3 (APS3v), that is defined as the co-occurrence of type 1 diabetes (T1D) and autoimmune thyroiditis (AITD). Allele 10, containing 25 repeats of the microsatellite (long repeats), is preferentially transmitted to offspring with APS3v, while allele 2, containing 14 repeats of the microsatellite (short repeats), is protective. We hypothesized that the long repeats of the intron 5 microsatellite decrease FOXP3 splicing and function, thereby reducing regulatory T cell activity and promoting the development of APS3v. We cloned genomic DNA from two males hemizygous for the long and short repeats of the microsatellite on their X-chromosomes and transfected them into human embryonic kidney 293 (HEK 293) cells to perform direct splicing analysis. We identified a novel splice variant of FOXP3 lacking exon 6, and showed that it is expressed in human thymus and lymph node. However, the length of the repeats in the microsatellite did not significantly influence the expression of this FOXP3 splice variant in vitro. Interestingly, this splice variant was expressed in human regulatory T cells, suggesting that it may play a role in their function. In conclusion, we identified a novel splice variant FOXP3Δ6. The role of its expression in regulatory T cells in the development of autoimmunity remains to be determined.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Thyroiditis, Autoimmune/genetics , Alternative Splicing , Chromosomes, Human, X , Exons , Genetic Predisposition to Disease , HEK293 Cells , Humans , Lymph Nodes/metabolism , Male , Microsatellite Repeats , Protein Isoforms/genetics , Protein Isoforms/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/metabolism
11.
Proc Natl Acad Sci U S A ; 111(34): 12562-7, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25122677

ABSTRACT

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


Subject(s)
Autoimmunity/genetics , Graves Disease/genetics , Graves Disease/immunology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Binding Sites/genetics , Cell Line , Epigenesis, Genetic , Gene Expression Regulation , Gene-Environment Interaction , Graves Disease/etiology , Histones/chemistry , Histones/metabolism , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Introns , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Methylation , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Thyroid Gland/immunology , Thyroid Gland/metabolism
12.
Biol Reprod ; 89(3): 55, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843234

ABSTRACT

In the zebrafish, the dynamic expression of the activin-inhibin-follistatin system during folliculogenesis and its exclusive localization (except follistatin) in follicle cells suggests that the system plays important roles in follicle development and that its expression is subject to tight controls, probably by external factors including those derived from the oocyte. We have previously identified zebrafish bone morphogenetic proteins (BMPs) as oocyte factors that may act on follicle cells; however, the targets of BMPs in the follicle cells remain unknown. Considering their spatiotemporal expression in the follicle, we hypothesized that members of the activin-inhibin-follistatin system in follicle cells could be potential target genes of BMPs. In the present study, we developed a novel coculture system to co-incubate zebrafish bone morphogenetic protein 2b or 4 (zfBMP2b/4)-producing Chinese hamster ovary (CHO) cells with zebrafish follicle cells. During incubation, the zfBMPs secreted from the CHO cells would act directly on the follicle cells in a paracrine manner. Our results showed that all activin beta subunits (inhbaa, inhbab, and inhbb) were down-regulated by both zfBMP2b and zfBMP4, while follistatin (fst, an activin-binding protein) and inhibin alpha (inha, an activin antagonist) were significantly up-regulated. The specificity of bone morphogenetic protein (BMP) actions was confirmed by short interfering RNA knockdown of zfBMP4 expression in the CHO cells. The robust response of inha to zfBMPs, together with our previous observation that inha expression surged at the full-grown stage prior to oocyte maturation, led us to hypothesize that the full-grown oocyte may signal upper levels of the hypothalamic-pituitary-gonadal axis its readiness to mature by releasing BMPs, which in turn stimulate inhibin production. As an ovarian hormone and activin antagonist, inhibin may suppress the action of activin in the pituitary to reduce follicle-stimulating hormone but increase luteinizing hormone (LH) biosynthesis. Meanwhile, by increasing the local follistatin level and reducing the activin production, BMPs could help prevent precocious maturation before preovulatory LH surge.


Subject(s)
Activins/genetics , Bone Morphogenetic Proteins/physiology , Follistatin/genetics , Gene Expression Regulation , Inhibins/genetics , Ovarian Follicle/growth & development , Zebrafish/physiology , Activins/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Follistatin/metabolism , Inhibins/metabolism , Ovarian Follicle/metabolism , Signal Transduction/physiology , Zebrafish/genetics
13.
J Immunol ; 189(6): 3043-53, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22888137

ABSTRACT

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


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , CD40 Antigens/genetics , Gene Targeting/methods , Graves Disease/genetics , Graves Disease/immunology , Animals , Autoantibodies/biosynthesis , Autoimmune Diseases/prevention & control , CD40 Antigens/biosynthesis , CD40 Antigens/deficiency , Cells, Cultured , Disease Models, Animal , Graves Disease/prevention & control , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Primary Cell Culture , Radiation Chimera/immunology , Receptors, Thyrotropin/immunology , Thyroid Gland/immunology , Thyroid Gland/metabolism , Thyroid Gland/pathology
14.
Gen Comp Endocrinol ; 176(3): 420-5, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22240277

ABSTRACT

Follicle-stimulating hormone receptor (fshr) and luteinizing hormone/choriogonadotropin receptor (lhcgr) exhibit differential temporal expression patterns during zebrafish folliculogenesis with fshr being dominant during vitellogenic growth and lhcgr increasing its expression dramatically before maturation. The dynamic and distinct expression patterns of fshr and lhcgr suggest that they are under tight regulatory control. However, the underlying mechanisms for the differential expression of the two receptors remain unknown. We have recently demonstrated that members of bone morphogenetic protein (BMP) family are largely expressed in the oocyte, while their receptors are exclusively localized on the follicle cells, suggesting a potential paracrine signaling from the oocyte to the follicle cells by BMPs. In this study, we investigated the effects of zebrafish BMP2b (zfBmp2b) and BMP4 (zfBmp4) on the expression of fshr and lhcgr using a novel co-culture approach. The recombinant zfBmp2b or zfBmp4-producing CHO cells were co-cultured with the zebrafish follicle cells followed by real-time qPCR analysis of fshr and lhcgr expression. Our results showed that zfBmp2b and zfBmp4 both down-regulated fshr, while up-regulated lhcgr expression at 24 h of co-culturing. This finding, together with the high expression level of BMP receptors in the follicle cells prior to oocyte maturation, strongly suggests a potential role for BMPs in the differential expression of fshr and lhcgr, especially in the full-grown follicles before maturation. As BMPs are largely expressed in the oocyte, this also implies an important role for the oocyte in orchestrating the differentiation and function of the follicle cells.


Subject(s)
Bone Morphogenetic Proteins/physiology , Gene Expression Regulation/physiology , Ovarian Follicle/physiology , Receptors, FSH/physiology , Receptors, LH/physiology , Zebrafish/physiology , Animals , CHO Cells , Coculture Techniques , Cricetinae , Female , Oocytes/physiology , Ovarian Follicle/cytology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Receptors, FSH/genetics , Receptors, LH/genetics
15.
Biol Reprod ; 85(5): 977-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21734261

ABSTRACT

The bone morphogenetic proteins (BMPs), originally identified by their abilities to induce bone and/or cartilage formation, have been reported to be involved in various growth and differentiation processes, including reproduction. Although mammalian models are more frequently used to study the BMP system in reproduction, we have extended the study to the zebrafish, an excellent model for studying female reproduction in teleosts. Reverse transcription-PCR analysis revealed the expression of the Bmp ligands (bmp2a, bmp2b, bmp4, bmp6, and bmp7a) and the type II Bmp receptors (bmpr2a and bmpr2b) in various tissues, including the ovary. Spatiotemporal distribution of these Bmp ligands and receptors in the ovary was then investigated in this study. Reverse transcription-PCR on isolated follicle layers and denuded oocytes demonstrated that all Bmp ligands examined were exclusively or abundantly expressed in the oocyte, whereas the two receptors were expressed exclusively in the follicle layers, strongly suggesting a potential paracrine signaling from the oocyte towards the follicle layer by various Bmp ligands. This supports the current view that instead of being passively controlled and nurtured by the follicle layer for its growth and development, the oocyte may play an active role by releasing various growth differentiation factors to regulate follicle layer function. Quantitative analysis of temporal expression profiles during folliculogenesis revealed an increased expression of bmp2a, bmp2b, bmp4, and bmp6 from primary growth (stage I) to previtellogenic (stage II) stages, followed by steady declines toward the end of folliculogenesis when the follicles became fully grown. On the contrast, the BMP receptors (bmpr2a and bmpr2b) consistently showed an increase in expression during folliculogenesis, with the peak levels reached at the full-grown stage prior to final oocyte maturation. The spatiotemporal expression patterns of the Bmp family in the zebrafish follicles provide important insights into potential roles for Bmps during follicle development as oocyte-derived factors. Further experiments using recombinant zebrafish Bmp4 showed that Bmp4 had an inhibitory effect on spontaneous oocyte maturation in vitro, but not 17alpha,20beta-dihydroxy-4-pregnen-3-one (DHP)-induced oocyte maturation in vitro.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Cell Communication/physiology , Oocytes/cytology , Ovarian Follicle/cytology , Ovary/metabolism , Paracrine Communication/physiology , Zebrafish/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Female , Ligands , Models, Animal , Oocytes/physiology , Ovarian Follicle/physiology , Phosphorylation , Phylogeny , Smad Proteins/metabolism , Tissue Distribution , Zebrafish Proteins/metabolism
16.
J Biol Chem ; 286(36): 31168-79, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21757724

ABSTRACT

Autoimmune thyroid diseases (AITD) arise from complex interactions between genetic, epigenetic, and environmental factors. Whole genome linkage scans and association studies have established thyroglobulin (TG) as a major AITD susceptibility gene. However, the causative TG variants and the pathogenic mechanisms are unknown. Here, we describe a genetic/epigenetic mechanism by which a newly identified TG promoter single-nucleotide polymorphism (SNP) variant predisposes to AITD. Sequencing analyses followed by case control and family-based association studies identified an SNP (-1623A→G) that was associated with AITD in the Caucasian population (p = 0.006). We show that the nucleotide substitution introduced by SNP (-1623A/G) modified a binding site for interferon regulatory factor-1 (IRF-1), a major interferon-induced transcription factor. Using chromatin immunoprecipitation, we demonstrated that IRF-1 binds to the 5' TG promoter motif, and the transcription factor binding correlates with active chromatin structure and is marked by enrichment of mono-methylated Lys-4 residue of histone H3, a signature of active transcriptional enhancers. Using reporter mutations and siRNA approaches, we demonstrate that the disease-associated allele (G) conferred increased TG promoter activity through IRF-1 binding. Finally, treatment of thyroid cells with interferon α, a known trigger of AITD, increased TG promoter activity only when it interacted with the disease-associated variant through IRF-1 binding. These results reveal a new mechanism of interaction between environmental (IFNα) and genetic (TG) factors to trigger AITD.


Subject(s)
Autoimmunity/genetics , Epigenesis, Genetic , Interferon-alpha/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Thyroglobulin/genetics , Thyroid Diseases/genetics , Binding Sites , Case-Control Studies , Cell Line , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Thyroid Diseases/immunology
17.
J Biol Chem ; 284(49): 34231-43, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19776016

ABSTRACT

Previously, we have shown that statistical synergism between amino acid variants in thyroglobulin (Tg) and specific HLA-DR3 pocket sequence signatures conferred a high risk for autoimmune thyroid disease (AITD). Therefore, we hypothesized that this statistical synergism mirrors a biochemical interaction between Tg peptides and HLA-DR3, which is key to the pathoetiology of AITD. To test this hypothesis, we designed a recombinant HLA-DR3 expression system that was used to express HLA-DR molecules harboring either AITD susceptibility or resistance DR pocket sequences. Next, we biochemically generated the potential Tg peptidic repertoire available to HLA-DR3 by separately treating 20 purified human thyroglobulin samples with cathepsins B, D, or L, lysosomal proteases that are involved in antigen processing and thyroid biology. Sequences of the cathepsin-generated peptides were then determined by matrix-assisted laser desorption ionization time-of-flight-mass spectroscopy, and algorithmic means were employed to identify putative AITD-susceptible HLA-DR3 binders. From four predicted peptides, we identified two novel peptides that bound strongly and specifically to both recombinant AITD-susceptible HLA-DR3 protein and HLA-DR3 molecules expressed on stably transfected cells. Intriguingly, the HLA-DR3-binding peptides we identified had a marked preference for the AITD-susceptibility DR signatures and not to those signatures that were AITD-protective. Structural analyses demonstrated the profound influence that the pocket signatures have on the interaction of HLA-DR molecules with Tg peptides. Our study suggests that interactions between Tg and discrete HLA-DR pocket signatures contribute to the initiation of AITD.


Subject(s)
Gene Expression Regulation , HLA-DR3 Antigen/metabolism , Recombinant Proteins/chemistry , Algorithms , Animals , Autoimmune Diseases , Cathepsins/chemistry , Cell Line , HeLa Cells , Histocompatibility Antigens Class II , Humans , Peptides/chemistry , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thyroglobulin/chemistry , Thyroid Diseases/immunology , Thyroid Gland/metabolism
18.
Proc Natl Acad Sci U S A ; 105(37): 14034-9, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18779568

ABSTRACT

Hashimoto's thyroiditis (HT) is associated with HLA, but the associated allele is still controversial. We hypothesized that specific HLA-DR pocket-sequence variants are associated with HT and that similar variants in the murine I-E locus (homologous to HLA-DR) predispose to experimental autoimmune thyroiditis (EAT), a classical mouse model of HT. Therefore, we sequenced the polymorphic exon 2 of the HLA-DR gene in 94 HT patients and 149 controls. In addition, we sequenced exon 2 of the I-E gene in 22 strains of mice, 12 susceptible to EAT and 10 resistant. Using logistic regression analysis, we identified a pocket amino acid signature, Tyr-26, Tyr-30, Gln-70, Lys-71, strongly associated with HT (P = 6.18 x 10(-5), OR = 3.73). Lys-71 showed the strongest association (P = 1.7 x 10(-8), OR = 2.98). This association was seen across HLA-DR types. The 5-aa haplotype Tyr-26, Tyr-30, Gln-70, Lys-71, Arg-74 also was associated with HT (P = 3.66 x 10(-4)). In mice, the I-E pocket amino acids Val-28, Phe-86, and Asn-88 were strongly associated with EAT. Structural modeling studies demonstrated that pocket P4 was critical for the development of HT, and pockets P1 and P4 influenced susceptibility to EAT. Surprisingly, the structures of the HT- and EAT-susceptible pockets were different. We conclude that specific MHC II pocket amino acid signatures determine susceptibility to HT and EAT by causing structural changes in peptide-binding pockets that may influence peptide binding, selectivity, and presentation. Because the HT- and EAT-associated pockets are structurally different, it is likely that distinct antigenic peptides are associated with HT and EAT.


Subject(s)
HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Peptides/immunology , Peptides/metabolism , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/metabolism , Amino Acids/metabolism , Animals , Binding Sites , Disease Models, Animal , HLA-DR Antigens/chemistry , HLA-DR Antigens/genetics , Humans , Mice , Peptides/chemistry , Sequence Analysis , Thyroiditis, Autoimmune/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...