Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Neurogastroenterol Motil ; 27(11): 1657-66, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26376608

ABSTRACT

BACKGROUND: Gastrointestinal (GI) manifestations are found in Ehlers Danlos syndrome (EDS) hypermobility subtype (HM). We aimed to assess associations between EDS HM and other EDS subtypes with GI manifestations. METHODS: We reviewed medical records of EDS patients evaluated at Mayo Clinic's Medical Genetics Clinic 1994-2013. We extracted information regarding EDS subtypes, GI manifestations, and treatments. KEY RESULTS: We identified 687 patients; 378 (56%) had associated GI manifestations (female 86.8%, diagnosis mean age 29.6 years). Of the patients identified, 58.9% (43/73) had EDS classic, 57.5% (271/471) EDS HM, 47.3% (27/57) EDS vascular subtypes. In addition, 86 patients had EDS that could not be classified in any of those three subtypes. Commonest GI symptoms were: abdominal pain (56.1%), nausea (42.3%), constipation (38.6%), heartburn (37.6%), and irritable bowel syndrome-like symptoms (27.5%). Many GI symptoms were commoner in EDS HM than the other subtypes together. Among 37.8% of the 378 patients who underwent esophagogastroduodenoscopy, the commonest abnormalities were gastritis, hiatal hernia and reflux esophagitis. Abnormal gastric emptying was observed in 22.3% (17/76): 11.8% delayed and 10.5% accelerated. Colonic transit was abnormal in 28.3% (13/46): 19.6% delayed and 8.7% accelerated. Rectal evacuation disorder was confirmed in 18/30 patients who underwent anorectal manometry. Angiography showed aneurysms in abdominal vessels in EDS vascular type. Proton pump inhibitors (38%) and drugs for constipation (23%) were the most commonly used medications. A minority underwent colectomy (2.9%) or small bowel surgery (4%). CONCLUSIONS & INFERENCES: EDS HM and other subtypes should be considered in patients with chronic functional GI symptoms and abdominal vascular lesions.


Subject(s)
Ehlers-Danlos Syndrome/complications , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/etiology , Adult , Ehlers-Danlos Syndrome/classification , Female , Humans , Male , Prevalence , Retrospective Studies
3.
J Genet Couns ; 23(3): 289-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24306141

ABSTRACT

Intellectual disability is a heterogeneous disorder with a wide phenotypic spectrum. Over 1,700 OMIM genes have been associated with this condition, many of which reside on the X-chromosome. The IQSEC2 gene is located on chromosome Xp11.22 and is known to play a significant role in the maintenance and homeostasis of the brain. Mutations in IQSEC2 have been historically associated with nonsyndromic X-linked intellectual disability. Case reports of affected probands show phenotypic overlap with conditions associated with pathogenic MECP2, FOXG1, CDKL5, and MEF2C gene mutations. Affected individuals, however, have also been identified as presenting with additional clinical features including seizures, autistic-behavior, psychiatric problems, and delayed language skills. To our knowledge, only 5 deleterious mutations and 2 intragenic duplications have been previously reported in IQSEC2. Here we report two novel IQSEC2 de novo truncating mutations identified through diagnostic exome sequencing in two severely affected unrelated male probands manifesting developmental delay, seizures, hypotonia, plagiocephaly, and abnormal MRI findings. Overall, diagnostic exome sequencing established a molecular diagnosis for two patients in whom traditional testing methods were uninformative while expanding on the mutational and phenotypic spectrum. In addition, our data suggests that IQSEC2 may be more common than previously appreciated, accounting for approximately 9 % (2/22) of positive findings among patients with seizures referred for diagnostic exome sequencing. Further, these data supports recently published data suggesting that IQSEC2 plays a more significant role in the development of X-linked intellectual disability with seizures than previously anticipated.


Subject(s)
Exome , Genetic Counseling , Genetic Diseases, X-Linked/genetics , Guanine Nucleotide Exchange Factors/genetics , Intellectual Disability/genetics , Mutation , Seizures/complications , Amino Acid Sequence , Animals , Child, Preschool , Female , Genetic Diseases, X-Linked/complications , Guanine Nucleotide Exchange Factors/chemistry , Humans , Intellectual Disability/complications , Male , Molecular Sequence Data , Pedigree , Sequence Homology, Amino Acid
4.
Transpl Immunol ; 26(2-3): 107-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22101306

ABSTRACT

Using a novel cytokine flow cytometry test (allo-CFC), we have previously shown that incubation of allogeneic cells with peripheral blood from highly-HLA sensitized (HS) patients results in reproducible gamma-interferon (IFNγ production in CD3(-) cells, and high (+) allo-CFC levels correlated with risk for antibody-mediated rejection (AMR). Here we report on identification of the cells and mechanisms responsible. The allo-CFC with/without modification was performed using blood from HS or normal individuals. IFNγ producing cells were CD3(-)/CD19(-), but CD3(-)/CD56(+). In vitro and in vivo B cell-depletion did not affect IFNγ production, demonstrating NK cells as the cells responsible for IFNγ production. NK cells from allo-CFC(+) or (-) individuals released significant amounts of IFNγ against target cells treated with serum from allo-CFC(+) individuals, but not allo-CFC(-) individuals. IFNγ release was abrogated by protein A/G treatment of the pretreated target cells, suggesting mediation by antibodies via FcγRIIIa (CD16). In conclusion, NK cell IFNγ release after allo-antigen exposure is mediated primarily through antibody-dependent cellular cytotoxicity (ADCC)-like mechanisms, suggesting that NK cells may be partially responsible for graft injury during AMR including C4d(-) AMR via ADCC, and could be a potential target for modification of this process.


Subject(s)
Antigens, CD/immunology , HLA Antigens/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Female , HLA Antigens/metabolism , Humans , Interferon-gamma/biosynthesis , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Male
5.
Clin Exp Immunol ; 139(2): 216-24, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15654820

ABSTRACT

Thyroid autoantigens require internalization and processing by antigen-presenting cells to induce immune responses. Besides pinocytosis, antigen uptake can be receptor-mediated. The mannose receptor (ManR) has a cysteine rich domain (CR) and eight carbohydrate recognition domains (CRD) that bind glycosylated proteins. The TSH receptor (TSHR), thyroid peroxidase (TPO) and thyroglobulin (Tg) are glycoproteins. To investigate a role for the ManR in thyroid autoimmunity, we tested the interaction between these autoantigens and chimeric ManRs. Plasmids encoding the CR-domain linked to IgG-Fc (CR-Fc) and CDR domains 4-7 linked to IgG-Fc (CDR4-7-Fc) were expressed and purified with Protein A. Enzyme-linked immunosorbent assay (ELISA) plates were coated with human thyroid autoantigens and CR-Fc or CRD4-7-Fc binding detected with peroxidase-conjugated anti-IgG-Fc. CRD4-7-Fc binding was highest for the TSHR, followed by Tg and was minimal for TPO. CR-Fc bound to Tg but not to TSHR or TPO. The interaction between the TSHR and CRD-Fc was calcium-dependent; it was inhibited by mannose (not galactose), and required a glycosylated TSHR A-subunit. Moreover, precomplexing the TSHR A-subunit with CRD-Fc (but not CR-Fc), or adding mannose (but not galactose), decreased in vitro responses of splenocytes from TSHR-immunized mice. Our data indicate that the ManR may participate in autoimmune responses to Tg and the TSHR but not to TPO. Most important, ManR binding of heavily glycosylated TSHR A-subunits suggests a mechanism by which the minute amounts of A-subunit protein shed from the thyroid may be captured by antigen-presenting cells located in the gland or in draining lymph nodes.


Subject(s)
Autoantigens/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes/immunology , Thyroid Hormones/immunology , Thyroiditis, Autoimmune/immunology , Animals , Antigen Presentation , Carbohydrates/analysis , Cell Line , Female , Humans , Iodide Peroxidase/chemistry , Iodide Peroxidase/immunology , Mannose Receptor , Mice , Mice, Inbred BALB C , Protein Binding , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Thyroglobulin/chemistry , Thyroglobulin/immunology
6.
Endocrinology ; 145(11): 4927-33, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15284197

ABSTRACT

We investigated why TSH receptor (TSHR) adenovirus immunization induces hyperthyroidism more commonly in BALB/c than in C57BL/6 mice. Recent modifications of the adenovirus model suggested that using adenovirus expressing the TSHR A subunit (A-subunit-Ad), rather than the full-length TSHR, and injecting fewer viral particles would increase the frequency of hyperthyroidism in C57BL/6 mice. This hypothesis was not fulfilled; 65% of BALB/c but only 5% of C57BL/6 mice developed hyperthyroidism. TSH binding inhibitory antibody titers were similar in each strain. Functional TSHR antibody measurements provided a better indication for this strain difference. Whereas thyroid-stimulating antibody activity was higher in C57BL/6 than BALB/c mice, TSH blocking antibody activity was more potent in hyperthyroid-resistant C57BL/6 mice. F(1) hybrids (BALB/c x C57BL/6) responded to A-subunit-Ad immunization with hyperthyroidism and TSHR antibody profiles similar to those of the hyperthyroid-susceptible parental BALB/c strain. In contrast, ELISA of TSHR antibodies revealed that the IgG subclass distribution in the F(1) mice resembled the disease-resistant C57BL/6 parental strain. Because the IgG subclass distribution is dependent on the T helper 1/T helper 2 cytokine balance, this paradigm can likely be excluded as an explanation for susceptibility to hyperthyroidism. In summary, our data for BALB/c, C57BL/6, and F(1) strains suggest that BALB/c mice carry a dominant gene(s) for susceptibility to induction of a thyroid-stimulating antibody/TSH blocking antibody balance that results in hyperthyroidism. Study of this genetic influence will provide useful information on potential candidate genes in human Graves' disease.


Subject(s)
Graves Disease/immunology , Graves Disease/physiopathology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Adenoviridae/genetics , Animals , Antibodies/blood , Disease Models, Animal , Disease Susceptibility , Female , Gene Transfer Techniques , Graves Disease/epidemiology , Incidence , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Thyrotropin/metabolism
7.
Clin Exp Immunol ; 135(3): 391-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15008970

ABSTRACT

Immunization with thyrotropin receptor (TSHR)-adenovirus is an effective approach for inducing thyroid stimulating antibodies and Graves' hyperthyroidism in BALB/c mice. In contrast, mice of the same strain vaccinated with TSHR-DNA have low or absent TSHR antibodies and their T cells recognize restricted epitopes on the TSHR. In the present study, we tested the hypothesis that immunization with TSHR-adenovirus induces a wider, or different, spectrum of TSHR T cell epitopes in BALB/c mice. Because TSHR antibody levels rose progressively from one to three TSHR-adenovirus injections, we compared T cell responses from mice immunized once or three times. Mice in the latter group were subdivided into animals that developed hyperthyroidism and those that remained euthyroid. Unexpectedly, splenocytes from mice immunized once, as well as splenocytes from hyperthyroid and euthyroid mice (three injections), all produced interferon-gamma in response to the same three synthetic peptides (amino acid residues 52-71, 67-86 and 157-176). These peptides were also the major epitopes recognized by TSHR-DNA plasmid vaccinated mice. We observed lesser responses to a wide range of additional peptides in mice injected three times with TSHR-adenovirus, but the pattern was more consistent with increased background 'noise' than with spreading from primary epitopes to dominant secondary epitopes. In conclusion, these data suggest that factors other than particular TSHR T cell epitopes (such as adenovirus-induced expression of conformationally intact TSHR protein), contribute to the generation of thyroid stimulating antibodies with consequent hyperthyroidism in TSHR-adenovirus immunized mice.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Graves Disease/immunology , Receptors, Thyrotropin/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Amino Acid Sequence , Animals , Autoantibodies/biosynthesis , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Female , Immunization/methods , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/immunology , Receptors, Thyrotropin/genetics , Spleen/immunology
8.
Clin Exp Immunol ; 134(3): 396-402, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14632743

ABSTRACT

The role of B cells as antigen-presenting cells is being recognized increasingly in immune responses to infections and autoimmunity. We compared T cell responses in wild-type and B cell-deficient mice immunized with the thyrotrophin receptor (TSHR), the major autoantigen in Graves' disease. Three B cell-deficient mouse strains were studied: JHD (no B cells), mIgM (membrane-bound monoclonal IgM+ B cells) and (m + s)IgM (membrane-bound and secreted monoclonal IgM). Wild-type and B cell-deficient mice (BALB/c background) were studied 8 weeks after three injections of TSHR or control adenovirus. Only wild-type mice developed IgG class TSHR antibodies and hyperthyroidism. After challenge with TSHR antigen, splenocyte cultures were tested for cytokine production. Splenocytes from TSHR adenovirus injected wild-type and mIgM-mice, but not from JHD- or (m + s)IgM- mice, produced interferon (IFN)-gamma in response to TSHR protein. Concanavalin A and pokeweed mitogen induced comparable IFN-gamma secretion in all groups of mice except in the JHD strain in which responses were reduced. The absence in (m + s)IgM mice and presence in mIgM mice of an anamnestic response to TSHR antigen was unrelated to lymphoid cell types. Surprisingly, although TSHR-specific antibodies were undetectable, low levels of serum IgG were present in mIgM- but not (m + s)IgM mice. Moreover, IFN-gamma production by antigen-stimulated splenocytes correlated with IgG levels. In conclusion, T cell responses to TSHR antigen developed only in mice with IgG-secreting B cells. Consequently, in the TSHR-adenovirus model of Graves' disease, some normal B cells appear to be required for the development of memory T cells.


Subject(s)
B-Lymphocytes/immunology , Graves Disease/immunology , Immunologic Memory , Receptors, Thyrotropin/genetics , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Autoantibodies/blood , Female , Genetic Vectors/administration & dosage , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Models, Animal , Receptors, Thyrotropin/immunology , Thyroxine/blood , Transduction, Genetic/methods
9.
Clin Exp Immunol ; 132(3): 408-15, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12780686

ABSTRACT

Plasmid and adenoviral vectors have been used to generate antibodies in mice that resemble human autoantibodies to the thyrotrophin receptor. No such studies, however, have been performed for thyroid peroxidase (TPO), the major autoantigen in human thyroiditis. We constructed plasmid and adenovirus vectors for in vivo expression of TPO. BALB/c mice were immunized directly by intramuscular injection of TPO-plasmid or TPO-adenovirus, as well as by subcutaneous injection of dendritic cells (DC) infected previously with TPO-adenovirus. Intramuscular TPO-adenovirus induced the highest, and TPO-plasmid the lowest, TPO antibody titres. Mice injected with TPO-transfected DC developed intermediate levels. Antibodies generated by all three approaches had similar affinities (Kd approximately 10(-9)M) and recognized TPO expressed on the cell-surface. Their epitopes were analysed in competition assays using monoclonal human autoantibodies that define the TPO immunodominant region (IDR) recognized by patients with thyroid autoimmune disease. Surprisingly, high titre antibodies generated using adenovirus interacted with diverse TPO epitopes largely outside the IDR, whereas low titre antibodies induced by DNA-plasmid recognized restricted epitopes in the IDR. This inverse relationship between antibody titre and restriction to the IDR is likely to be due to epitope spreading following strong antigenic stimulation provided by the adenovirus vector. However, TPO antibody epitope spreading does not occur in Hashimoto's thyroiditis, despite high autoantibody levels. Consequently, these data support the concept that in human thyroid autoimmunity, factors besides titre must play a role in shaping an autoantibody epitopic profile.


Subject(s)
Adenoviridae/genetics , Autoantibodies/biosynthesis , Genetic Vectors/immunology , Iodide Peroxidase/immunology , Plasmids/genetics , Animals , Antibody Affinity , Antibody Specificity/immunology , Autoantigens/genetics , Autoantigens/immunology , Cell Transplantation , Dendritic Cells/transplantation , Female , Immunization/methods , Immunodominant Epitopes/immunology , Immunoglobulin G/biosynthesis , Injections, Intramuscular , Iodide Peroxidase/genetics , Mice , Mice, Inbred BALB C , Thyroid Gland/immunology
10.
Clin Exp Immunol ; 131(2): 234-40, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12562382

ABSTRACT

Dendritic cells (DCs) are the most potent antigen-presenting cells and a prerequisite for the initiation of primary immune response. This study was performed to investigate the contribution of DCs to the initiation of Graves' hyperthyroidism, an organ-specific autoimmune disease in which the thyrotrophin receptor (TSHR) is the major autoantigen. DCs were prepared from bone marrow precursor cells of BALB/c mice by culturing with granulocyte macrophage-colony stimulating factor and interleukin-4. Subcutaneous injections of DCs infected with recombinant adenovirus expressing the TSHR (but not beta-galactosidase) in syngeneic female mice induced Graves'-like hyperthyroidism (8 and 35% of mice after two and three injections, respectively) characterized by stimulating TSHR antibodies, elevated serum thyroxine levels and diffuse hyperplasitc goiter. TSHR antibodies determined by ELISA were of both IgG1 (Th2-type) and IgG2a (Th1-type) subclasses, and splenocytes from immunized mice secreted interferon-gamma (a Th1 cytokine), not interleukin-4 (a Th2 cytokine), in response to TSHR antigen. Surprisingly, IFN-gamma secretion, and induction of antibodies and disease were almost completely suppressed by co-administration of alum/pertussis toxin, a Th2-dominant adjuvant, whereas polyriboinosinic polyribocytidylic acid, a Th1-inducer, enhanced splenocyte secretion of IFN-gamma without changing disease incidence. These observations demonstrate that DCs efficiently present the TSHR to naive T cells to induce TSHR antibodies and Graves'-like hyperthyroidism in mice. In addition, our results challenge the previous concept of Th2 dominance in Graves' hyperthyroidism and provide support for the role of Th1 immune response in disease pathogenesis.


Subject(s)
Autoantigens/immunology , Dendritic Cells/immunology , Graves Disease/immunology , Receptors, Thyrotropin/immunology , Adenoviridae/metabolism , Animals , Autoantibodies/biosynthesis , Female , Immunization/methods , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Spleen/immunology , Th1 Cells/immunology , Th2 Cells/immunology
11.
Endocrinology ; 142(8): 3530-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459799

ABSTRACT

Two approaches have been developed to induce TSH receptor antibodies in mice with properties resembling those in Graves' disease, the Shimojo model of injecting live fibroblasts coexpressing the TSH receptor and major histocompatibility complex antigen Class II, and TSH receptor-DNA vaccination. Thyroid-stimulating antibodies appear to occur less commonly after DNA vaccination, but there has been no direct comparison of these models. We performed a three-way comparison of 1) AKR/N and 2) BALB/c mice vaccinated with TSH receptor-DNA and 3) AKR/N mice injected with fibroblasts expressing the TSH receptor and the major histocompatibility complex antigen class II of AKR/N mice. TSH receptor-DNA vaccinated mice had low or undetectable levels of TSH receptor antibodies determined by ELISA or flow cytometry. Nonspecific binding precluded comparisons with sera from Shimojo mice by these assays. TSH binding inhibition and thyroid-stimulating antibody were undetectable in TSH receptor-DNA vaccinated mice. In Shimojo mice, TSH binding inhibition was positive in approximately 60%, and thyroid-stimulating antibodies were positive in hyperthyroid animals. Unlike the negative antibody data, splenocytes from TSH receptor-vaccinated (but not Shimojo) mice proliferated and produced the Th1 cytokine interferon-gamma in response to TSH receptor antigen. In conclusion, DNA vaccination is less effective at inducing TSH receptor antibodies than the Shimojo approach, but it permits the future characterization of TSH receptor-specific T cells generated without adjuvant.


Subject(s)
Interferon-gamma/biosynthesis , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccination , Vaccines, DNA/immunology , Animals , Antibodies/analysis , Antibody Formation/physiology , Antigens/pharmacology , Cell Division/drug effects , Humans , Interleukin-4/biosynthesis , Lymphocytes/cytology , Male , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/metabolism , Thyroid Gland/cytology
12.
Thyroid ; 11(4): 301-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11349828

ABSTRACT

Human autoantibodies to thyroid peroxidase (TPO) interact with a restricted or immunodominant region (IDR) on intact TPO. However, a smaller proportion of polyclonal serum TPO autoantibodies bind outside this region. To isolate monoclonal nonimmunodominant region (non-IDR) TPO autoantibodies, we screened a thyroid-derived immunoglobulin gene phage display library while "epitope masking" the TPO IDR with four human TPO monoclonal autoantibodies that define the IDR. Among 31 non-IDR autoantibodies obtained (expressed as Fab), 8 representatives were analyzed further based on their restriction digestion profiles. All are encoded by almost identical H chains (VH3 family), with extremely long D regions, paired with three different types of light chains. In contrast, IDR TPO Fab from the same patient utilize seven different heavy chains (VH1 and VH5 families) paired nonpromiscuously with different light chains. Use of VH5 genes has not been reported previously for TPO autoantibodies. Both non-IDR and IDR Fab bind specifically to TPO and not to other proteins. The non-IDR Fab affinities for TPO are moderately high (Kd 1-2 x 10(-9) M), somewhat lower than those for most IDR Fab (Kd 1-4 x 10(-10) M). The epitopes of the three types of non-IDR Fab overlap with each other, indicating a major role for their heavy chain in TPO binding. Most importantly, the epitopes of non-IDR Fab are recognized by patients' serum autoantibodies. In summary, we provide the first insight into the immunoglobulin genes, affinities and epitopes of human monoclonal autoantibodies that bind outside the TPO-immunodominant region.


Subject(s)
Antibodies, Monoclonal/immunology , Autoantibodies/immunology , Epitopes, B-Lymphocyte , Iodide Peroxidase/immunology , Amino Acid Sequence , Antibody Affinity , Antibody Specificity , Genes, Immunoglobulin , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Variable Region/genetics , Molecular Sequence Data
13.
J Clin Endocrinol Metab ; 86(3): 1287-93, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238522

ABSTRACT

A secreted recombinant TSH receptor (TSHR) ectodomain variant (TSHR-289) neutralizes TSHR autoantibodies in Graves' disease, but is heterogeneous in containing both immunologically active and inactive molecules and is also unstable. We have now purified each form of TSHR-289 using sequential affinity chromatography with a mouse mAb (3BD10) specific for the inactive form, and a mAb to C-terminal His residues that recognizes both forms. The immunological difference between active and inactive TSHR-289 was unrelated to primary amino acid sequence or carbohydrate content and was, therefore, attributable to its folded state. The epitopes for Graves' autoantibodies and 3BD10 overlap, and both are destroyed by denaturation. Therefore, reciprocal binding by autoantibodies and 3BD10 to conformational determinants involving the same TSHR segment suggests a prion-like shift between two folded states of the molecule. Despite purification, immunologically active TSHR-289 remained labile, as determined by loss of autoantibody, and gain of 3BD10, recognition. However, using chemical chaperones we have, for the first time, been able to stabilize purified TSHR antigen in immunologically intact form. In summary, purification of immunologically active and stable antigen in milligram quantities provides a powerful tool for future diagnostic and therapeutic studies in Graves' disease.


Subject(s)
Autoantibodies/immunology , Graves Disease/immunology , Protein Conformation , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Amino Acid Sequence , Animals , Antibody Specificity , CHO Cells , Carbohydrates/analysis , Chromatography, Affinity , Cricetinae , Drug Stability , Epitopes/chemistry , Epitopes/immunology , Humans , Immunoglobulins, Thyroid-Stimulating , Methylamines/pharmacology , Mice , Proline/pharmacology , Receptors, Thyrotropin/genetics , Recombinant Proteins/chemistry , Structure-Activity Relationship , Thyrotropin/metabolism
14.
Clin Exp Immunol ; 122(2): 170-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11091271

ABSTRACT

AKR/N mice injected with fibroblasts expressing MHC class II (RT4.15HP cells) and the TSH receptor (TSHR) develop antibodies similar to those in Graves' disease. We were unable to analyse the subclass of these antibodies because of unexpectedly high non-specific binding by ELISA or flow cytometry. The non-specific binding reflected generalized immune activation which occurred even when the fibroblasts did not express the TSHR. However, the IgG subclasses were determined for thyroid peroxidase (TPO) antibodies induced using TPO-expressing RT4.14HP cells and found to be IgG2a > IgG1. This Thl pattern is consistent with spontaneous secretion of interferon-gamma (but not IL-4 or IL-10) by splenocytes from injected mice. The Th1 bias was related to fibroblast injection because conventional immunization of the same mouse strain with purified TPO and adjuvant induced a Th2 response (IgG1 >> IgG2a). Further, untransfected fibroblasts themselves induced powerful, non-specific proliferative responses when used as antigen-presenting cells (APC) in vitro. Flow cytometry revealed that the RT4.15HP fibroblasts (and TSHR- and TPO-transfected derivatives) expressed B7-1. Unexpected constitutive expression of this key molecule may bypass the requirement for up-regulation of other costimulatory molecules involved in T cell stimulation. Our data support the concept that RT4.15HP fibroblasts present the TSHR (or TPO), at least for initiating the immune response. However, the accompanying generalized immune stimulation creates difficulties for analysis of TSHR-specific T and B lymphocytes. On the other hand, extension of the model to TPO, an easier antigen to study, will facilitate analysis of murine T cell responses likely to resemble those in human thyroid autoimmunity.


Subject(s)
Cytokines/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/classification , Thyroiditis, Autoimmune/immunology , Animals , Antibody Specificity , Antigen Presentation , Autoantigens/administration & dosage , Cell Line , Disease Models, Animal , Female , Fibroblasts/immunology , Histocompatibility Antigens Class II/administration & dosage , Humans , In Vitro Techniques , Iodide Peroxidase/administration & dosage , Iodide Peroxidase/immunology , Lymphocyte Activation , Mice , Mice, Inbred AKR , Receptors, Thyrotropin/administration & dosage , Receptors, Thyrotropin/immunology , Thyroid Gland/immunology , Thyroiditis, Autoimmune/etiology
15.
Thyroid ; 9(11): 1057-61, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10595452

ABSTRACT

There is, at present, no assay in clinical use for the direct assay of autoantibody binding to the thyrotropin receptor (TSHR). We now describe a direct thyrotropin receptor autoantibody binding assay (DTAb) using a secreted form of the TSHR ectodomain (TSHR-289) without the need for antigen purification. The assay compensates for the low TSHR autoantibody concentration in serum by capturing a relatively large amount of patient immunoglobulin G (IgG) on high-capacity beads, a reversal of standard methods that typically first immobilize antigen. TSHR-289 captured by Graves' IgG was detected in a colorimetric reaction using a biotinylated murine monoclonal antibody to the poly-histidine tail engineered into the antigen. By this approach, sera from 11 normal individuals provided a mean optical density (OD) value of 0.20 +/- 0.08 SD (range 0.06-0.33). Of 38 sera from unselected patients with a history of Graves' disease (untreated and treated), 29 (76%) generated OD values > 0.37 (2 SD above the mean for the normal sera), the highest being OD 1.38. Surprisingly, 3 of 13 (23%) sera from TPO autoantibody-positive patients with Hashimoto's thyroiditis also provided values > 2 SD above the normal sera. The extent of direct autoantibody binding to the TSHR correlated closely with the thyrotropin binding inhibition (TBI) values (r = 0.881; p < 0.001). One serum was clearly positive in only the direct binding assay and another in only the TBI assay. The data obtained with the direct binding assay correlated less well with the thyroid-stimulating antibody (TSAb) assay (r = 0.582; p < 0.001). In summary, we describe a new direct DTAb assay that correlates more closely with the TBI than with the TSI assays. Future studies in a large series of clinically defined patients will be needed to evaluate the clinical utility of the DTAb assay.


Subject(s)
Autoantibodies/blood , Receptors, Thyrotropin/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL