Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Exp Immunol ; 196(1): 52-58, 2019 04.
Article in English | MEDLINE | ID: mdl-30566234

ABSTRACT

NOD.H2h4 mice are the most commonly used model for human autoimmune thyroiditis. Because thyroid autoimmunity develops slowly (over months), NOD.H2h4 mice are usually exposed to excess dietary iodide to accelerate and amplify the process. However, unlike the female bias in human thyroid autoimmunity, autoantibodies to thyroglobulin (TgAb) are reported to be similar in male and female NOD.H2h4 . We sought evidence for sexual dimorphism in other parameters in this strain maintained on regular or iodized water. Without iodide, TgAb levels are higher in males than in females, the reverse of human disease. In humans, autoantibodies to thyroid peroxidase (TPOAb) are a better marker of disease than TgAb. In NOD.H2h4 mice TPOAb develop more slowly than TgAb, being detectable at 6 months of age versus 4 months for the latter. Remarkably, unlike TgAb, TPOAb levels are higher in female than male NOD.H2h4 mice on both regular and iodized water. As previously observed, serum T4 levels are similar in both sexes. However, thyroid-stimulating hormone (TSH) levels are significantly higher in males than females with or without iodide exposure. TSH levels correlate with TgAb levels in male NOD.H2h4 mice, suggesting a possible role for TSH in TgAb development. However, there is no correlation between TSH and TPOAb levels, the latter more important than TgAb in human disease. In conclusion, if the goal of an animal model is to closely reflect human disease, TPOAb rather than TgAb should be measured in older female NOD.H2h4 mice, an approach requiring patience and the use of mouse TPO protein.


Subject(s)
Aging/immunology , Iodide Peroxidase/immunology , Sex Factors , Thyroiditis, Autoimmune/immunology , Animals , Antibody Formation , Autoantibodies/metabolism , Diet Therapy , Disease Models, Animal , Female , Humans , Iodides/administration & dosage , Male , Mice , Mice, Inbred NOD , Sex Characteristics , Thyroglobulin/immunology , Thyroiditis, Autoimmune/diagnosis , Thyrotropin/blood
2.
Clin Exp Immunol ; 188(2): 243-253, 2017 05.
Article in English | MEDLINE | ID: mdl-28099999

ABSTRACT

The thyrotrophin receptor (TSHR) A-subunit is the autoantigen targeted by pathogenic autoantibodies that cause Graves' hyperthyroidism, a common autoimmune disease in humans. Previously, we reported that pathogenic TSHR antibodies develop spontaneously in thyroiditis-susceptible non-obese diabetic (NOD).H2h4 mice bearing a human TSHR A-subunit transgene, which is expressed at low levels in both the thyroid and thymus (Lo-expressor transgene). The present study tested recent evidence that high intrathymic TSHR expression protects against the development of pathogenic TSHR antibodies in humans. By successive back-crossing, we transferred to the NOD.H2h4 background a human TSHR A-subunit transgene expressed at high levels in the thyroid and thymus (Hi-expressor transgene). In the sixth back-cross generation (> 98% NOD.H2h4 genome), only transgenic offspring produced spontaneously immunoglobulin (Ig)G class non-pathogenic human TSHR A-subunit antibodies. In contrast, both transgenic and non-transgenic offspring developed antibodies to thyroglobulin and thyroid peroxidase. However, non-pathogenic human TSHR antibody levels in Hi-expressor offspring were lower than in Lo-expressor transgenic mice. Moreover, pathogenic TSHR antibodies, detected by inhibition of TSH binding to the TSHR, only developed in back-cross offspring bearing the Lo-expressor, but not the Hi-expressor, transgene. High versus low expression human TSHR A-subunit in the NOD.H2h4 thymus was not explained by the transgene locations, namely chromosome 2 (127-147 Mb; Hi-expressor) and chromosome 1 (22.9-39.3 Mb; low expressor). Nevertheless, using thyroiditis-prone NOD.H2h4 mice and two transgenic lines, our data support the association from human studies that low intrathymic TSHR expression is associated with susceptibility to developing pathogenic TSHR antibodies, while high intrathymic TSHR expression is protective.


Subject(s)
Immunoglobulins, Thyroid-Stimulating/biosynthesis , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Thymus Gland/metabolism , Thyroid Gland/immunology , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/physiopathology , Animals , Autoantibodies/blood , Graves Disease/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Iodide Peroxidase/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Thyroglobulin/immunology , Thyroid Gland/cytology , Thyroid Gland/pathology , Thyroiditis, Autoimmune/pathology
3.
Genes Immun ; 16(4): 268-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25811933

ABSTRACT

NOD.H2(k) and NOD.H2(h4) mice carry the major histocompatibility complex (MHC) class II molecule I-A(k) associated with susceptibility to experimentally induced thyroiditis. Dietary iodine-enhanced spontaneous thyroid autoimmunity, well known in NOD.H2(h4) mice, has not been investigated in NOD.H2(k) mice. We compared NOD.H2(h4) and NOD.H2(k) strains for thyroiditis and autoantibodies to thyroglobulin (TgAb) and thyroid peroxidase (TPOAb) without or with dietary sodium iodide (NaI) for up to 32 weeks. TgAb levels were significantly higher in NOD.H2(h4) compared with NOD.H2(k) mice on NaI, and TPOAb developed in NOD.H2(h4) mice but not in NOD.H2(k) mice. DNA exome analysis revealed, in addition to the differences in the chromosome (Chr) 17 MHC regions, that NOD.H2(k) mice, and particularly NOD.H2(h4) mice, have substantial non-MHC parental DNA. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis highlighted thyroid autoimmunity and immune-response genes on Chr 17 but not on Chr 7, and 15 parental B10.A4R DNA. Studies of parental strains provided no evidence for non-MHC gene contributions. The exon 10 Tg haplotype, associated with experimentally induced thyroiditis, is absent in NOD.H2(h4) and NOD.H2(k) mice and is not a marker for spontaneous murine thyroid autoimmunity. In conclusion, the absence of I-E is a likely explanation for the difference between NOD.H2(h4) and NOD.H2(k) mice in TgAb levels and, as in humans, autoantibody spreading to TPO.


Subject(s)
Autoantibodies/immunology , Histocompatibility Antigens Class II/immunology , Thyroglobulin/metabolism , Thyroid Gland/immunology , Animals , Autoantibodies/metabolism , Autoimmunity/immunology , Exome , Haplotypes , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Iodide Peroxidase/immunology , Male , Mice, Inbred NOD/genetics , Mice, Inbred NOD/immunology , Sodium Iodide/adverse effects , Thyroglobulin/genetics , Thyroglobulin/immunology , Thyroiditis/genetics , Thyroiditis/immunology , Thyroiditis, Autoimmune/chemically induced , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/immunology
4.
Clin Exp Immunol ; 154(3): 305-15, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18811696

ABSTRACT

Transgenic BALB/c mice that express intrathyroidal human thyroid stimulating hormone receptor (TSHR) A-subunit, unlike wild-type (WT) littermates, develop thyroid lymphocytic infiltration and spreading to other thyroid autoantigens after T regulatory cell (T(reg)) depletion and immunization with human thyrotropin receptor (hTSHR) adenovirus. To determine if this process involves intramolecular epitope spreading, we studied antibody and T cell recognition of TSHR ectodomain peptides (A-Z). In transgenic and WT mice, regardless of T(reg) depletion, TSHR antibodies bound predominantly to N-terminal peptide A and much less to a few downstream peptides. After T(reg) depletion, splenocytes from WT mice responded to peptides C, D and J (all in the A-subunit), but transgenic splenocytes recognized only peptide D. Because CD4(+) T cells are critical for thyroid lymphocytic infiltration, amino acid sequences of these peptides were examined for in silico binding to BALB/c major histocompatibility complex class II (IA-d). High affinity subsequences (inhibitory concentration of 50% < 50 nm) are present in peptides C and D (not J) of the hTSHR and mouse TSHR equivalents. These data probably explain why transgenic splenocytes do not recognize peptide J. Mouse TSHR mRNA levels are comparable in transgenic and WT thyroids, but only transgenics have human A-subunit mRNA. Transgenic mice can present mouse TSHR and human A-subunit-derived peptides. However, WT mice can present only mouse TSHR, and two to four amino acid species differences may preclude recognition by CD4+ T cells activated by hTSHR-adenovirus. Overall, thyroid lymphocytic infiltration in the transgenic mice is unrelated to epitopic spreading but involves human A-subunit peptides for recognition by T cells activated using the hTSHR.


Subject(s)
Receptors, Thyrotropin/immunology , T-Lymphocytes, Regulatory/immunology , Thyroiditis, Autoimmune/immunology , Amino Acid Sequence , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Chemotaxis, Leukocyte/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Immunization , Lymphocyte Depletion/methods , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , RNA, Messenger/genetics , Receptors, Thyrotropin/genetics , Spleen/immunology , Thyroid Gland/immunology , Thyroid Gland/physiopathology , Thyroiditis, Autoimmune/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...