Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Endocrinol Diabetes Obes ; 22(5): 381-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26181432

RESUMO

PURPOSE OF REVIEW: The purpose of this article is to summarize the recent advances on experimental Graves' hyperthyroidism and orbitopathy as studied in two widely used mouse models, which involve repetitive genetic vaccinations using either adenovirus or in-vivo electroporation of the eukaryotic expression plasmid expressing the thyrotropin receptor (TSHR) as a vector. RECENT FINDINGS: The models have been improved by using different types of antigens, including the holo receptor, the receptor A-subunit, an alternatively spliced form of variant receptor lacking a single leucine-rich repeat in the codomain, the receptors of human or mouse origin; different mice such as wild-type, TSHR knockout, TSHR transgenic and different inbred mice; and different immunization protocols. They are now useful for elucidating the pathogenic mechanisms of not only Graves' hyperthyroidism but also Graves' orbitopathy. SUMMARY: This review summarizes the literature of mouse models of Graves' hyperthyroidism and orbitopathy published over the last 3 years.


Assuntos
Doença de Graves/patologia , Oftalmopatia de Graves/patologia , Animais , Modelos Animais de Doenças , Doença de Graves/genética , Oftalmopatia de Graves/genética , Humanos , Camundongos , Receptores da Tireotropina/genética
2.
Endocr J ; 61(3): 297-302, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24335009

RESUMO

Transforming growth factor ß (TGF-ß) members, pleiotropic cytokines, play a critical role for carcinogenesis generally as a tumor suppressor in the early cancer development, but as a tumor promoter in the late stage of cancer progression. The present study was designed to clarify the role for TGF-ß signaling in early thyroid carcinogenesis using the conditional Tgfbr2(floxE2/floxE2) knock-in mice, having 2 loxP sites at introns 1 and 2 of Tgfb2r gene. When these mice were crossed with thyroid peroxidase (TPO)-Cre or fibroblast-specific protein-1 (FSP1)-Cre, the resultant mice, Tgfbr2(tpoKO) and Tgfbr2(fspKO), lost TGF-ß II receptor expression (thereby TGF-ß signaling) specifically in the thyroid follicular epithelial cells or fibroblasts, respectively. The thyroid morphology was monitored up to 52 weeks in these mice, showing no tumor development, except one Tgfbr2(tpoKO) mouse developing follicular adenoma like-lesion. Our data suggest that TGF-ß signaling in mesenchymal or follicular epithelial cells of the thyroid does not appear to function as a tumor suppressive barrier at the early stage of thyroid carcinogenesis.


Assuntos
Transdução de Sinais , Glândula Tireoide/citologia , Neoplasias da Glândula Tireoide/etiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Carcinogênese , Células Epiteliais/fisiologia , Feminino , Fibroblastos/metabolismo , Masculino , Camundongos
3.
Endocrinology ; 154(11): 4423-30, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23970782

RESUMO

The mutant BRAF (BRAF(V600E)) is the most common genetic alteration in papillary thyroid carcinomas (PTCs). The oncogenicity of this mutation has been shown by some genetically engineered mouse models. However, in these mice, BRAF(V600E) is expressed in all the thyroid cells from the fetal periods, and suppresses thyroid function, thereby leading to TSH elevation, which by itself promotes thyroid tumorigenesis. To overcome these problems, we exploited 2 different approaches, both of which allowed temporally and spatially restricted expression of BRAF(V600E) in the thyroid glands. First, we generated conditional transgenic mice harboring the loxP-neo(R)-loxP-BRAF(V600E)-internal ribosome entry site-green fluorescent protein sequence [Tg(LNL-BRAF(V600E))]. The double transgenic mice (LNL-BRAF(V600E);TPO-Cre) were derived from a high expressor line of Tg(LNL-BRAF(V600E)) mice and TPO-Cre mice; the latter expresses Cre DNA recombinase under the control of thyroid-specific thyroid peroxidase (TPO) promoter and developed PTC-like lesions in early life under normal serum TSH levels due to mosaic recombination. In contrast, injection of adenovirus expressing Cre under the control of another thyroid-specific thyroglobulin (Tg) promoter (Ad-TgP-Cre) into the thyroids of LNL-BRAF(V600E) mice did not induce tumor formation despite detection of BRAF(V600E) and pERK in a small fraction of thyroid cells. Second, postnatal expression of BRAF(V600E) in a small number of thyroid cells was also achieved by injecting the lentivirus expressing loxP-green fluorescent protein-loxP-BRAF(V600E) into the thyroids of TPO-Cre mice; however, no tumor development was again observed. These results suggest that BRAF(V600E) does not appear to induce PTC-like lesions when expressed in a fraction of thyroid cells postnatally under normal TSH concentrations.


Assuntos
Carcinoma Papilar/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Genes Transgênicos Suicidas , Iodeto Peroxidase , Camundongos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Tiroxina/metabolismo
4.
Endocrinology ; 153(4): 2034-42, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22334716

RESUMO

We have recently shown that wild type mice are highly tolerant, whereas thyrotropin receptor (TSHR) knockout (KO) mice are susceptible to immunization with the mouse TSHR, the autoantigen in Graves' disease. However, because TSHR KO mice lack the endogenous TSHR, Graves-like hyperthyroidism cannot be expected to occur in these mice. We therefore performed adoptive transfer of splenocytes from TSHR KO mice into nude mice expressing the endogenous TSHR. Anti-TSHR autoantibodies were detected in approximately 50 % recipient mice 4 wk after adoptive transfer of splenocytes (5 × 107/mouse) from TSHR KO mice immunized with adenovirus expressing mTSHR A subunit and persisted for 24 wk. Depletion of regulatory T cells by anti-CD25 antibody in the donor mice increased successful transfer rates without increasing antibody levels. Some recipient mice showed transient increases in thyroid-stimulating antibodies and T4 levels 4-8 wk after transfer, but many became thyroid-blocking antibody positive and hypothyroid 24 wk later. Adoptive transfer of splenocytes from naïve TSHR KO mice transiently induced very low antibody titers when the recipient mice were treated with anticytotoxic lymphocyte antigen 4 and antiprogrammed cell death 1 ligand 1 antibodies for 8 wk after transfer. Histologically, macrophages infiltrated the retrobulbar adipose tissues and extraocular muscles in a small fraction of the recipients. Our findings demonstrate successful adoptive transfer of anti-TSHR immune response from TSHR KO mice to nude mice. Although the recipient mice developed only transient and infrequent hyperthyroidism, followed by eventual hypothyroidism, induction of orbital inflammation suggests the possible role of anti-TSHR immune response for Graves' orbitopathy.


Assuntos
Transferência Adotiva/métodos , Autoimunidade/imunologia , Camundongos Knockout/imunologia , Camundongos Nus/imunologia , Receptores da Tireotropina/deficiência , Receptores da Tireotropina/imunologia , Baço/imunologia , Animais , Anticorpos Anti-Idiotípicos/metabolismo , Modelos Animais de Doenças , Feminino , Doença de Graves/complicações , Doença de Graves/imunologia , Doença de Graves/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Doenças Orbitárias/etiologia , Doenças Orbitárias/imunologia , Doenças Orbitárias/fisiopatologia , Receptores da Tireotropina/genética , Baço/citologia , Glândula Tireoide/fisiopatologia , Transplantes
5.
Autoimmunity ; 44(6): 504-10, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21306188

RESUMO

The nonobese diabetic (NOD) mouse spontaneously develops several autoimmune diseases, including type 1 diabetes and to a lesser extent thyroiditis and sialitis. Imbalance between effector T cells (Teffs) and regulatory T cells (Tregs) has recently been proposed as a mechanism for the disease pathogenesis in NOD mice, but previous studies have shown the various outcomes by different timing and methods of Treg-depletion. This study was, therefore, designed to compare the consequences of Treg-depletion by the same method (anti-CD25 antibody) on the spectrum of organ-specific autoimmune diseases in NOD mice of different ages. Treg-depletion by anti-CD25 antibody at 10 days of age accelerated development of all three diseases we examined (insulitis/diabetes, thyroiditis, and sialitis); Treg-depletion at 4 weeks of age accelerated only diabetes but not thyroiditis or sialitis; and Treg-depletion at 12 weeks of age hastened only development of thyroiditis and exhibited little influence on diabetes or sialitis. Increased levels of insulin autoantibodies (IAA) were, however, observed in mice depleted of Tregs at 10 days of age, not in those at 4 weeks. Thus, the consequences of Treg-depletion on the spectrum of organ-specific autoimmune diseases depend on the timing of anti-CD25 antibody injection in NOD mice. Aging gradually tips balance between Teffs and Tregs toward Teff-dominance for diabetes, but this balance for thyroiditis and sialitis likely alters more intricately. Our data also suggest that the levels of IAA are not necessarily correlated with diabetes development.


Assuntos
Doenças Autoimunes/imunologia , Depleção Linfocítica/efeitos adversos , Linfócitos T Reguladores/imunologia , Envelhecimento , Animais , Anticorpos Monoclonais , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Feminino , Subunidade alfa de Receptor de Interleucina-2/sangue , Subunidade alfa de Receptor de Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Sialadenite/imunologia , Tireoidite/imunologia
6.
Endocrinology ; 151(8): 4047-54, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20555026

RESUMO

Graves-like hyperthyroidism is induced in BALB/c mice by immunization with adenovirus expressing the human TSH receptor (TSHR) A-subunit (amino acids 1-289). However, because of nonidentity between the human and mouse TSHR ( approximately 87% amino acid homology), we compared the responses of mice immunized with adenoviruses expressing either the mouse or the human TSHR A-subunit. Wild-type (wt) BALB/c mice immunized with the mouse A-subunit developed neither TSHR antibodies (measured by flow cytometry) nor thyroid lymphocytic infiltration. However, wt C57BL/6 mice developed sparse intrathyroidal lymphocyte infiltration without antibody production. Depletion of naturally occurring regulatory CD4(+)CD25(+) T cells had little effect. These results indicate the inability to break tolerance to the mouse TSHR in wt mice. In contrast, TSHR knockout (KO) BALB/c mice generated mouse TSHR antibodies in response to mouse A-subunit immunization and augmented human TSHR antibody response to human A-subunit immunization. Thyroid-stimulating antibody titers measured in a functional bioassay were comparable in human A-subunit immunized wt mice and in TSHR KO mice immunized with either the mouse or human A-subunit. In conclusion, immune response to the mouse TSHR is readily induced in TSHR KO but not in wt mice. Only in the former does immunization with adenovirus expressing the mouse A-subunit generate antibodies capable of activating the mouse TSHR. TSHR KO mice are, therefore, of value for future studies dissecting the autoimmune response to the mouse TSHR.


Assuntos
Imunização , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia , Animais , Autoimunidade/genética , Células CHO , Cricetinae , Cricetulus , Feminino , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regulação para Cima/imunologia
7.
Endocrinology ; 150(3): 1545-51, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18988676

RESUMO

One approach to prevent tissue destruction by autoimmune attack in organ-specific autoimmune diseases is to protect the target tissue from autoimmune reaction, regardless of its persistent activity. To provide proof-of-principle for the feasibility of this approach, the immunoregulatory molecules, TNF-related apoptosis-inducing ligand (TRAIL) and indoleamine 2, 3-dioxygenase, were expressed in the thyroid glands using adenovirus vector in nonobese diabetic-H2(h4) mice that spontaneously develop thyroiditis. Mice were anesthetized, and the thyroid glands were exposed by neck dissection, followed by in situ infection with adenovirus vector (5 x 10(10) particles per mouse) twice or thrice, starting 1 d or 4 wk before mice were supplied with sodium iodine (NaI) water. After 8 wk NaI provision, the extent of thyroiditis, serum titers of antithyroglobulin antibodies, and cytokine expression in the spleen were examined. In situ infection of adenovirus expressing TRAIL or indoleamine 2, 3-dioxygenase, but not green fluorescent protein, significantly suppressed thyroiditis scores. However, antithyroglobulin antibody titers and expression levels of cytokines (interferon-gamma and IL-4) in the spleen remained unaltered. Importantly, adenovirus infection 4 wk after NaI provision was also effective at suppressing thyroiditis. The suppressive effect of TRAIL appears to be mediated at least partly by accumulation of CD4(+)Foxp3(+) regulatory T cells into the thyroid glands. Thus, localized expression of immunoregulatory molecules efficiently protected the thyroid glands from autoimmune attack without changing the systemic autoimmunity in nonobese diabetic-H2(h4) mice. This kind of immunological intervention, although it does not suppress autoimmune reactivity, may have a potential for treating organ-specific autoimmune diseases.


Assuntos
Fatores Imunológicos/genética , Fatores Imunológicos/fisiologia , Glândula Tireoide/metabolismo , Tireoidite Autoimune/genética , Tireoidite Autoimune/prevenção & controle , Adenoviridae/genética , Animais , Células COS , Chlorocebus aethiops , Citocinas/sangue , Citocinas/metabolismo , Citoproteção/genética , Citoproteção/imunologia , Feminino , Terapia Genética/métodos , Fatores Imunológicos/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Especificidade de Órgãos/genética , Linfócitos T Reguladores/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/imunologia , Tireoidite Autoimune/imunologia , Tireoidite Autoimune/metabolismo
8.
Biochem Biophys Res Commun ; 367(4): 719-24, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18194666

RESUMO

Insulin peptide B:9-23 is a major autoantigen in type 1 diabetes. Combined treatment with B:9-23 peptide and polyinosinic-polycytidylic acid (poly I:C), but neither alone, induce insulitis in normal BALB/c mice. In contrast, the combined treatment accelerated insulitis, but prevented diabetes in NOD mice. Our immunofluorescence study with anti-CD4/anti-Foxp3 revealed that the proportion of Foxp3 positive CD4(+)CD25(+) regulatory T cells (Tregs) was elevated in the islets of NOD mice treated with B:9-23 peptide and poly I:C, as compared to non-treated mice. Depletion of Tregs by anti-CD25 antibody hastened spontaneous development of diabetes in non-treated NOD mice, and abolished the protective effect of the combined treatment and conversely accelerated the onset of diabetes in the treated mice. These results indicate that poly I:C combined with B:9-23 peptide promotes infiltration of both pathogenic T cells and predominantly Tregs into the islets, thereby inhibiting progression from insulitis to overt diabetes in NOD mice.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Fatores de Transcrição Forkhead/metabolismo , Insulina/administração & dosagem , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Fragmentos de Peptídeos/administração & dosagem , Poli I-C/administração & dosagem , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/patologia , Feminino , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/patologia
9.
Endocrinology ; 148(12): 6040-6, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823258

RESUMO

Graves' disease is a thyroid-specific autoimmune disease mediated by stimulatory autoantibodies against the TSH receptor (TSHR). We have previously shown in our mouse model with adenovirus expressing the TSHR that antibody mediated depletion of CD4(+)CD25(+) regulatory T cells (Tregs) enhances incidence and severity of hyperthyroidism in resistant and susceptible mouse strains, respectively. These data indicate that balance between effector T cells and Tregs is critical for disease development. This study was designed to evaluate the role played by another recently identified type of Treg, CD8(+)CD122(+) T cells, in our mouse model to delineate the significance of different types of Tregs in Graves' disease. Flow cytometry analysis showed that CD4(+)CD25(+) and CD8(+)CD122(+) T cells are distinct cell types, and anti-CD122 antibody effectively and selectively depleted CD8(+)CD122(+) T cells. As for CD4(+)CD25(+) Treg, CD8(+)CD122(+) T cell depletion increased the incidence of hyperthyroidism both in resistant and susceptible mice. Of interest, intrathyroidal lymphocytic infiltration was observed in some CD8(+)CD122(+) T cell-depleted, hyperthyroid resistant mice. These results indicate that in addition to CD4(+)CD25(+) T cells, CD8(+)CD122(+) T cells also play a crucial role in disease susceptibility in mouse Graves' disease. Thus, different types of Tregs appear to be involved in tolerance to a self-antigen, the TSHR.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença de Graves/imunologia , Hipertireoidismo/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Doença de Graves/sangue , Hipertireoidismo/sangue , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores da Tireotropina/imunologia , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Hormônios Tireóideos/sangue , Hormônios Tireóideos/metabolismo
10.
J Autoimmun ; 29(2-3): 195-202, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17826032

RESUMO

NOD-H2(h4) mice, which express I-A(k) on the NOD background, spontaneously develop autoimmune thyroiditis, a model of Hashimoto thyroiditis in humans, by adding iodide in the drinking water. Parental NOD mice have previously been shown to have intrinsic numerical abnormalities in peripheral lymphocytes and lymphocyte subpopulations such as CD4(+)CD25(+) naturally occurring regulatory T cells (Treg). Therefore we first investigated whether the similar abnormalities exist in NOD-H2(h4) mice. We observed that, compared with other non-autoimmune disease prone BALB/c and C57BL/6 mice, NOD-H2(h4) mice have lower numbers of splenocytes, CD3(+)T, CD4(+)T and CD8(+)T cells but the ratios of Treg to CD4(+)T cells were comparable. Increasing the numbers of peripheral lymphocytes by Complete Freund's Adjuvant immunization or splenocyte transfer did not affect development of thyroiditis, indicating that lymphopenia does not play a critical role in the pathogenesis of thyroiditis. We next examined the significance of Treg by depleting this lymphocyte subset with anti-CD25 antibody. Treg depletion, performed 4days before the administration of NaI water for 8 weeks, significantly exacerbated thyroiditis (p<0.01). Anti-thyroglobulin antibody titers also increased by Treg depletion (p<0.01) without changing the IgG2b to IgG1 ratios. In addition, expression levels of mRNA for IFN-gamma and IL-4 were enhanced in parallel. However, T(4) levels were similar between antibody-treated and untreated groups. Additional anti-CD25 administration at 3 weekly intervals did not influence these results. These data, together with previous studies on other mouse models of inducible thyroiditis and Graves' disease, indicate the role played by Treg in keeping anti-thyroid autoimmune reaction in check in experimental autoimmune thyroid diseases.


Assuntos
Linfopenia/imunologia , Linfócitos T Reguladores/imunologia , Tireoidite Autoimune/imunologia , Animais , Autoanticorpos/análise , Autoanticorpos/imunologia , Subunidade alfa de Receptor de Interleucina-2/análise , Subunidade alfa de Receptor de Interleucina-2/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos NOD , Iodeto de Sódio/administração & dosagem , Linfócitos T Reguladores/metabolismo , Tireoglobulina/imunologia , Glândula Tireoide/citologia , Glândula Tireoide/imunologia , Glândula Tireoide/metabolismo , Tireoidite Autoimune/induzido quimicamente , Tireoidite Autoimune/metabolismo , Tiroxina/sangue , Tiroxina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...