Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 194(9): 4154-61, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25825442

RESUMO

Abs that stimulate the thyrotropin receptor (TSHR), the cause of Graves' hyperthyroidism, only develop in humans. TSHR Abs can be induced in mice by immunization, but studying pathogenesis and therapeutic intervention requires a model without immunization. Spontaneous, iodine-accelerated, thyroid autoimmunity develops in NOD.H2(h4) mice associated with thyroglobulin and thyroid-peroxidase, but not TSHR, Abs. We hypothesized that transferring the human TSHR A-subunit to NOD.H2(h4) mice would result in loss of tolerance to this protein. BALB/c human TSHR A-subunit mice were bred to NOD.H2(h4) mice, and transgenic offspring were repeatedly backcrossed to NOD.H2(h4) mice. All offspring developed Abs to thyroglobulin and thyroid-peroxidase. However, only TSHR-transgenic NOD.H2(h4) mice (TSHR/NOD.H2(h4)) developed pathogenic TSHR Abs as detected using clinical Graves' disease assays. As in humans, TSHR/NOD.H2(h4) female mice were more prone than male mice to developing pathogenic TSHR Abs. Fortunately, in view of the confounding effect of excess thyroid hormone on immune responses, spontaneously arising pathogenic human TSHR Abs cross-react poorly with the mouse TSHR and do not cause thyrotoxicosis. In summary, the TSHR/NOD.H2(h4) mouse strain develops spontaneous, iodine-accelerated, pathogenic TSHR Abs in female mice, providing a unique model to investigate disease pathogenesis and test novel TSHR Ag-specific immunotherapies aimed at curing Graves' disease in humans.


Assuntos
Autoanticorpos/imunologia , Modelos Animais de Doenças , Doença de Graves/imunologia , Iodo , Receptores da Tireotropina/imunologia , Animais , Feminino , Doença de Graves/induzido quimicamente , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Transgênicos
2.
J Basic Microbiol ; 56(3): 214-28, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26344826

RESUMO

A total of 1502 samples, including feces of sheep (793) and cattle (348), pasture soil (118), dung compost (147) and barn soil (96), were examined between October 2012 and August 2014 to discover potential strains of nematophagous fungi for the biological control of livestock-parasitic nematodes. These samples were collected from 87 sites located in 48 counties of 20 provinces (autonomous regions/municipalities) of China. Fungi were identified down to a species level. Four hundred and seventy-seven isolates, which were distributed in 8 genera and 28 taxa, were identified as nematophagous fungi. Nematode-trapping fungi included 17 species and one unidentified species of Arthrobotrys, two of Dactylella, Drechslerella dactyloides, and Duddingtonia flagrans. Five identified species and two unidentified species of endoparasitic fungi were isolated. The predominant species from all regions were Arthrobotrys oligospora, followed by Arthrobotrys musiformis, Arthrobotrys (Monacrosporium) thaumasiun, and Arthrobotrys (Monacrosporium) microscaphoides. Species with adhesive networks were the most frequently isolated. Among the endoparasitic fungi, Podocrella harposporifera (Harposporium anguillulae) was the most common species, followed by Harposporium lilliputanum and Harposporium arcuatum. Based on Shannon diversity index, the diversity levels of nematophagous fungi were relatively higher in samples associated with cattle, barn soil, and subtropical monsoon climate zone. Three species isolated from this study, namely, Duddingtonia flagrans, Arthrobotrys salina (Monacrosporium salinum), and Arthrobotrys oligospora var. sarmatica, are newly recorded in China, and 20 species (including one unidentified species) are newly recorded in sheep and cattle barn soils worldwide.


Assuntos
Doenças dos Bovinos/prevenção & controle , Doenças dos Bovinos/parasitologia , Fungos/isolamento & purificação , Nematoides/microbiologia , Infecções por Nematoides/veterinária , Doenças dos Ovinos/prevenção & controle , Doenças dos Ovinos/parasitologia , Animais , Biodiversidade , Bovinos , China , Sistema Digestório/microbiologia , Sistema Digestório/parasitologia , Fezes/microbiologia , Fezes/parasitologia , Fungos/classificação , Infecções por Nematoides/microbiologia , Infecções por Nematoides/prevenção & controle , Controle Biológico de Vetores/métodos , Ovinos
3.
J Basic Microbiol ; 55(8): 992-1001, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25847583

RESUMO

Nematophagous fungi are considered to have the best potential as biological agents for the control of gastrointestinal nematodes in domestic animals. However, relatively few studies have been conducted with the genus Monacrosporium, especially with strains native to China. In the present study, we isolated and identified nematophagous fungi from fresh sheep feces. A pure fungal strain was molecularly characterized, and its nematophagous activity was evaluated. The morphological plasticity of the isolated strain, as well as its interaction with the nematode targets, was observed by scanning electron microscopy of the infected Trichostrongylus colubriformis L3 and the free-living nematode Caenorhabditis elegans. Three isolated fungal strains from the 30 fresh fecal samples of sheep from Inner Mongolia, China exhibited predatory activity; however, only a single strain was successfully purified (SF 0459). The SF 0459 strain was characterized by morphological analysis of its conidia and sequencing of its ITS1-5.8S rDNA-ITS2 region. This strain was identified to be Monacrosporium salinum (GenBank ID: KP036623). Nematophagous fungus helper bacteria were found at the interaction points between fungi and nematodes. The percentage of live T. colubriformis L3 was reduced by 83.79-88.69% based on the in vitro assay.


Assuntos
Ascomicetos/isolamento & purificação , Ascomicetos/fisiologia , Caenorhabditis elegans/microbiologia , Fezes/microbiologia , Trichostrongylus/microbiologia , Animais , Ascomicetos/classificação , Ascomicetos/citologia , China , Microscopia Eletrônica de Varredura , Dados de Sequência Molecular , Controle Biológico de Vetores , Filogenia , Análise de Sequência de DNA , Ovinos , Esporos Fúngicos/isolamento & purificação , Esporos Fúngicos/ultraestrutura
4.
J Biol Chem ; 286(8): 6219-24, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21190937

RESUMO

Thyroid-stimulating hormone (TSH)-induced reduction in ligand binding affinity (negative cooperativity) requires TSH receptor (TSHR) homodimerization, the latter involving primarily the transmembrane domain (TMD) but with the extracellular domain (ECD) also contributing to this association. To test the role of the TMD in negative cooperativity, we studied the TSHR ECD tethered to the cell surface by a glycosylphosphatidylinositol (GPI) anchor that multimerizes despite the absence of the TMD. Using the infinite ligand dilution approach, we confirmed that TSH increased the rate of dissociation (k(off)) of prebound (125)I-TSH from CHO cells expressing the TSH holoreceptor. Such negative cooperativity did not occur with TSHR ECD-GPI-expressing cells. However, even in the absence of added TSH, (125)I-TSH dissociated much more rapidly from the TSHR ECD-GPI than from the TSH holoreceptor. This phenomenon, suggesting a lower TSH affinity for the former, was surprising because both the TSHR ECD and TSH holoreceptor contain the entire TSH-binding site, and the TSH binding affinities for both receptor forms should, theoretically, be identical. In ligand competition studies, we observed that the TSH binding affinity for the TSHR ECD-GPI was significantly lower than that for the TSH holoreceptor. Further evidence for a difference in ligand binding kinetics for the TSH holoreceptor and TSHR ECD-GPI was obtained upon comparison of the TSH K(d) values for these two receptor forms at 4 °C versus room temperature. Our data provide the first evidence that the wild-type TSHR TMD influences ligand binding affinity for the ECD, possibly by altering the conformation of the closely associated hinge region that contributes to the TSH-binding site.


Assuntos
Receptores da Tireotropina/metabolismo , Tireotropina/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Humanos , Cinética , Estrutura Terciária de Proteína , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Tireotropina/química , Tireotropina/genética
5.
Colloids Surf B Biointerfaces ; 213: 112410, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35176603

RESUMO

Conducting polymer has been directly polymerized around living neural cells or in the cortex with the aim of creating an intimate contact between implantable electrical devices and electrogenetic cells. The long term cellular effect after conductive polymer coating, a critical issue for practical applications, has not been reported. In this study, poly(3,4-ethylenedioxythiophene) PEDOT was directly polymerized around the living primary neural and PC12 cells under varying current densities, potentials and charge-balanced current pulses. The cell morphology, nuclei evolution, and cell viability post PEDOT polymerization were studied at different time points. The aim of this study was to investigate the immediate and long-term cellular response towards in-situ polymerization of conductive polymers and to provide experimental information on the feasibility of this technique in practical applications.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes , Polímeros , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Condutividade Elétrica , Neurônios/fisiologia , Polimerização , Ratos
6.
Sci Rep ; 11(1): 12850, 2021 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145333

RESUMO

Acoustic-gravity waves are generally considered to be one of the major factors that drive changes of the total electron content in the ionosphere. However, causal mechanisms of couplings between sources in the lithosphere and responses in the atmosphere and the ionosphere are not fully understood, yet. A barometer in the cave of the SBCB station records an unusual phenomenon of larger amplitudes in air pressure changes inside than those at the Xinwu station (outside). Accordingly, the comparison between the recorded data at the SBCB and Xinwu station can drive investigations of potential sources of the unusual phenomenon. Analytical results of phase angle differences reveal that the air pressure outside the cave at the Xinwu station often leads air pressure changes inside at the SBCB station at relatively low frequency bands. In contrast, the larger pressure changes at frequencies > ~ 5 × 10-4 Hz inside the cave at the SBCB station lead smaller changes outside at the Xinwu station. To expose causal mechanisms of the unusual phenomenon, continuous seismic waveforms are further conducted for examination. When the horizontal and vertical ground velocities of ground vibrations yield a difference in the phase angle close to 90°, coherence values between the air pressure changes and ground vibrations become large. This suggests that the pressure-shear vertical ground vibrations can drive air pressure changes. Meanwhile, the results shed light on investigating the existence of acoustic waves near the Earth's surface using a partially confined space underground due to that the assumptions of the waves can propagate upward into the atmosphere driving changes in the ionosphere.

7.
J Nanosci Nanotechnol ; 10(7): 4500-4, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21128447

RESUMO

The growth of uniform nanostructures requires simple and reproducible ways to control the size. It is found that Co atoms can form two-dimensional structural islands on √3 x √3-Ag/Ge(111) surfaces. Temperature and Co coverage are two factors to modulate the island size. By using scanning tunneling microscopy, the surface structures and morphology for different annealing temperatures and variable Co coverage have been investigated. For 100 degrees C annealing temperature, Co atoms are difficult to condense into structural islands at 0.35 ML whereas several structural Co islands are found at 1.4 ML. This difference is due to the quantity of Co atoms per unit area for forming structural islands. As the temperature increases, Co atoms get more energy to diffuse. Therefore, the average island size increases with rising temperatures until the coverage of 3.5 ML. Yet, the island size stops growing above the coverage of 3.5 ML because of the limitation for the Co covered area. Therefore the Co islands increase their height rather than their size. In addition, the shape of Co islands can also be controlled. It transforms from random shapes to the hexagonal shape with increasing temperature.

8.
Mol Endocrinol ; 22(5): 1171-82, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18218728

RESUMO

The glycoprotein hormone receptor hinge region connects the leucine-rich and transmembrane domains. The prevalent concept is that the hinge does not play a significant role in ligand binding and signal transduction. Portions of the hinge are redundant and can be deleted by mutagenesis or are absent in certain species. A minimal hinge will be more amenable to future investigation of its structure and function. We, therefore, combined and progressively extended previous deletions (Delta) in the TSH receptor (TSHR) hinge region (residues 277-418). TSHRDelta287-366, Delta287-371, Delta287-376, and Delta287-384 progressively lost their response to TSH stimulation of cAMP generation in intact cells, consistent with a progressive loss of TSH binding. The longest deletion (TSHRDelta287-384), reducing the hinge region from 141 to 43 amino acids, totally lost both functions. Surprisingly, however, with deletions extending from residues 371-384, constitutive (ligand-independent) activity increased severalfold, reversing the suppressive (inverse agonist) effect of the TSHR extracellular domain. TSHR-activating point mutations I486F and I568T in the first and second extracellular loops (especially the former) had reduced activity on a background of TSHRDelta287-371. In summary, our data support the concept that the TSHR hinge contributes significantly to ligand binding affinity and signal transduction. Residues within the hinge, particularly between positions 371-384, appear involved in ectodomain inverse agonist activity. In addition, the hinge is necessary for functionality of activating mutations in the first and second extracellular loops. Rather than being an inert linker between the leucine-rich and transmembrane domains, the TSHR hinge is a signaling-specificity domain.


Assuntos
Leucina/metabolismo , Receptores da Tireotropina/metabolismo , Transdução de Sinais/fisiologia , Animais , Sítios de Ligação/genética , Células COS , Chlorocebus aethiops , Citometria de Fluxo , Leucina/genética , Ligantes , Modelos Biológicos , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Transdução de Sinais/genética
9.
Endocrinology ; 149(7): 3427-34, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18388191

RESUMO

CS-17 is a murine monoclonal antibody to the human TSH receptor (TSHR) with both inverse agonist and antagonist properties. Thus, in the absence of ligand, CS-17 reduces constitutive TSHR cAMP generation and also competes for TSH binding to the receptor. The present data indicate that for both of these functions, the monovalent CS-17 Fab (50 kDa) behaves identically to the intact, divalent IgG molecule (150 kDa). The surprising observation that CS-17 competes for TSH binding to the human but not porcine TSHR enabled identification of a number of amino acids in its epitope. Replacement of only three human TSHR residues (Y195, Q235, and S243) with the homologous porcine TSHR residues totally abolishes CS-17 binding as detected by flow cytometry. TSH binding is unaffected. Of these residues, Y195 is most important, with Q235 and S243 contributing to CS-17 binding to a much lesser degree. The functional effects of CS-17 IgG and Fab on constitutive cAMP generation by porcinized human TSHR confirm the CS-17 binding data. The location of TSHR amino acid residues Y195, Q235, and S243 deduced from the crystal structure of the FSH receptor leucine-rich domain provides valuable insight into the CS-17 and TSH binding sites. Whereas hormone ligands bind primarily to the concave surface of the leucine-rich domains, a major portion of the CS-17 epitope lies on the opposite convex surface with a minor component in close proximity to known TSH binding residues.


Assuntos
Aminoácidos/imunologia , Anticorpos Monoclonais/imunologia , Epitopos/imunologia , Receptores da Tireotropina/imunologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Aminoácidos/química , Animais , Anticorpos Monoclonais/farmacologia , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Epitopos/química , Citometria de Fluxo , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/antagonistas & inibidores , Suínos
10.
Endocrinology ; 149(4): 2001-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162518

RESUMO

Susceptibility genes for TSH receptor (TSHR) antibodies and hyperthyroidism can be probed in recombinant inbred (RI) mice immunized with adenovirus expressing the TSHR A-subunit. The RI set of CXB strains, derived from susceptible BALB/c and resistant C57BL/6 (B6) mice, were studied previously. High-resolution genetic maps are also available for RI BXH strains, derived from B6 and C3H/He parents. We found that C3H/He mice develop TSHR antibodies, and some animals become hyperthyroid after A-subunit immunization. In contrast, the responses of the F1 progeny of C3H/He x B6 mice, as well as most BXH RI strains, are dominated by the B6 resistance to hyperthyroidism. As in the CXB set, linkage analysis of BXH strains implicates different chromosomes (Chr) or loci in the susceptibility to induced TSHR antibodies vs. hyperthyroidism. Importantly, BXH and CXB mice share genetic loci controlling the generation of TSHR antibodies (Chr 17, major histocompatibility complex region, and Chr X) and development of hyperthyroidism (Chr 1 and 3). Moreover, some chromosomal linkages are unique to either BXH or CXB strains. An interesting candidate gene linked to thyroid-stimulating antibody generation in BXH mice is the Ig heavy chain locus, suggesting a role for particular germline region genes as precursors for these antibodies. In conclusion, our findings reinforce the importance of major histocompatibility complex region genes in controlling the generation of TSHR antibodies measured by TSH binding inhibition. Moreover, these data emphasize the value of RI strains to dissect the genetic basis for induced TSHR antibodies vs. their effects on thyroid function in Graves' disease.


Assuntos
Autoanticorpos/genética , Predisposição Genética para Doença , Doença de Graves/genética , Animais , Ligação Genética , Hipertireoidismo/genética , Imunoglobulinas Estimuladoras da Glândula Tireoide , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Locos de Características Quantitativas , Recombinação Genética , Cromossomo X
11.
Thyroid ; 28(7): 933-940, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29845889

RESUMO

BACKGROUND: The high constitutive, or ligand-independent, activity of the thyrotropin receptor (TSHR) is of clinical importance in some thyroid conditions, particularly well-differentiated thyroid carcinoma remnants following incomplete ablative therapy (surgery and radioiodine). Under these conditions, even total suppression of TSH by thyroid hormone administration does not fully reduce TSHR activity, a driver of thyrocyte growth. METHODS: CS-17 is a murine monoclonal antibody that has inverse agonist activity in that it suppresses TSHR constitutive activity. This study crystallized the CS-17 Fab and determined its atomic structure at a resolution of 3.4 Å. RESULTS: In silico docking of this structure to that of the TSHR extracellular domain was accomplished by targeting to TSHR residue tyrosine 195 (Y195) known to contribute to the CS-17 epitope. High affinity interaction between these two molecules, primarily by the CS-17 immunoglobulin heavy chain, was validated by energetic analysis (KD of 8.7 × 10-11 M), as well as by previously obtained data on a number of individual TSHR amino acids in three regions whose mutagenesis reduced CS-17 binding as detected by flow cytometry. CONCLUSIONS: Structural insight at atomic resolution of a TSHR antibody with inverse agonist activity opens the way for the development of a molecule with therapeutic potential, particularly in thyroid carcinoma. For this purpose, CS-17 will require "humanization" by substitution of its constant region (Fc component). In addition, with its epitope defined, the CS-17 affinity can be increased further by mutagenesis of selected amino acids in its heavy- and light-chain complementarity determining regions.


Assuntos
Anticorpos Monoclonais/química , Epitopos , Receptores da Tireotropina/imunologia , Animais , Células CHO , Cricetulus , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide
12.
Endocrinology ; 148(5): 2375-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17272389

RESUMO

TSH binding to the TSH receptor (TSHR) induces thyrocyte growth and proliferation primarily by activating the adenylyl cyclase signaling pathway. Relative to the other glycoprotein hormone receptors, the TSHR has considerable ligand-independent (constitutive) activity. We describe a TSHR monoclonal antibody (CS-17) with the previously unrecognized property of being an inverse agonist for TSHR constitutive activity. This property is retained, even when constitutive activity is extremely high consequent to diverse TSHR extracellular region mutations. A similar effect on an activating mutation at the base of the sixth transmembrane helix (not accessible to direct CS-17 contact) indicates that CS-17 is acting allosterically. Administered to mice in vivo, CS-17 reduces serum T(4) levels. The CS-17 epitope is conformational and a significant portion lies in the C-terminal region of the TSHR leucine-rich domain (residues 260-289). By interacting with the large TSHR extracellular domain, CS-17 is, to our knowledge, the first antibody reported to be an inverse agonist for a member of the G protein receptor superfamily. After humanization of its murine constant region, CS-17 has the potential to be an adjunctive therapeutic agent in athyreotic patients with residual well-differentiated thyroid carcinoma as well as pending definitive treatment in some selected hyperthyroidism states.


Assuntos
Anticorpos Monoclonais/imunologia , Hipertireoidismo/imunologia , Hipertireoidismo/terapia , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/imunologia , Regulação Alostérica/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Células COS , Chlorocebus aethiops , Cricetinae , Feminino , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação , Receptores da Tireotropina/genética , Tiroxina/sangue , Transfecção
13.
Endocrinology ; 148(12): 5724-33, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823263

RESUMO

Hyperthyroidism in Graves' disease is caused by thyroid-stimulating autoantibodies to the TSH receptor (TSHR), whereas hypothyroidism in Hashimoto's thyroiditis is associated with thyroid peroxidase and thyroglobulin autoantibodies. In some Graves' patients, thyroiditis becomes sufficiently extensive to cure the hyperthyroidism with resultant hypothyroidism. Factors determining the balance between these two diseases, the commonest organ-specific autoimmune diseases affecting humans, are unknown. Serendipitous findings in transgenic BALB/c mice, with the human TSHR A-subunit targeted to the thyroid, shed light on this relationship. Of three transgenic lines, two expressed high levels and one expressed low intrathyroidal A-subunit levels (Hi- and Lo-transgenics, respectively). Transgenics and wild-type littermates were depleted of T regulatory cells (Treg) using antibodies to CD25 (CD4(+) T cells) or CD122 (CD8(+) T cells) before TSHR-adenovirus immunization. Regardless of Treg depletion, high-expressor transgenics remained tolerant to A-subunit-adenovirus immunization (no TSHR antibodies and no hyperthyroidism). Tolerance was broken in low-transgenics, although TSHR antibody levels were lower than in wild-type littermates and no mice became hyperthyroid. Treg depletion before immunization did not significantly alter the TSHR antibody response. However, Treg depletion (particularly CD25) induced thyroid lymphocytic infiltrates in Lo-transgenics with transient or permanent hypothyroidism (low T(4), elevated TSH). Neither thyroid lymphocytic infiltration nor hypothyroidism developed in similarly treated wild-type littermates. Remarkably, lymphocytic infiltration was associated with intermolecular spreading of the TSHR antibody response to other self thyroid antigens, murine thyroid peroxidase and thyroglobulin. These data suggest a role for Treg in the natural progression of hyperthyroid Graves' disease to Hashimoto's thyroiditis and hypothyroidism in humans.


Assuntos
Doença de Graves/imunologia , Doença de Hashimoto/imunologia , Linfócitos T Reguladores/imunologia , Adenoviridae/genética , Animais , Autoanticorpos/imunologia , Doença de Graves/patologia , Doença de Hashimoto/patologia , Humanos , Hipertireoidismo/imunologia , Hipertireoidismo/patologia , Imunização , Imunoglobulinas Estimuladoras da Glândula Tireoide/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia , Linfócitos T Reguladores/metabolismo , Glândula Tireoide/imunologia , Glândula Tireoide/metabolismo
14.
J Clin Invest ; 111(12): 1897-904, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12813025

RESUMO

Graves disease, a common organ-specific autoimmune disease affecting humans, differs from all other autoimmune diseases in being associated with target organ hyperfunction rather than organ damage. Clinical thyrotoxicosis is directly caused by autoantibodies that activate the thyrotropin receptor (TSHR). The etiology of Graves disease is multifactorial, with nongenetic factors playing an important role. Of the latter, there is the intriguing possibility that the molecular structure of the target antigen contributes to the development of thyroid-stimulatory autoantibodies (TSAb's). Among the glycoprotein hormone receptors, only the TSHR undergoes intramolecular cleavage into disulfide-linked subunits with consequent shedding of some of the extracellular, autoantibody-binding A subunits. Functional autoantibodies do not arise to the noncleaving glycoprotein hormone receptors. Recently, TSAb's were found to preferentially recognize shed, rather than attached, A subunits. Here we use a new adenovirus-mediated animal model of Graves disease to show that goiter and hyperthyroidism occur to a much greater extent when the adenovirus expresses the free A subunit as opposed to a genetically modified TSHR that cleaves minimally into subunits. These data show that shed A subunits induce or amplify the immune response leading to hyperthyroidism and provide new insight into the etiology of Graves disease.


Assuntos
Autoantígenos/química , Doença de Graves/imunologia , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Adenoviridae/genética , Animais , Anticorpos Bloqueadores/metabolismo , Autoanticorpos/metabolismo , Autoantígenos/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Doença de Graves/etiologia , Doença de Graves/genética , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Mutagênese , Subunidades Proteicas , Receptores da Tireotropina/genética , Glândula Tireoide/patologia
15.
J Clin Invest ; 110(2): 209-17, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12122113

RESUMO

Graves disease is directly caused by thyroid-stimulating autoantibodies (TSAb's) that activate the thyrotropin receptor (TSHR). We observed upon flow cytometry using intact cells that a mouse mAb (3BD10) recognized the TSHR ectodomain with a glycosidylphosphatidylinositol (ECD-GPI) anchor approximately tenfold better than the same ectodomain on the wild-type TSHR, despite the far higher level of expression of the latter. The 3BD10 epitope contains the N-terminal cysteine cluster critical for TSAb action. Consequently, we hypothesized and confirmed that TSAb (but not thyrotropin-blocking autoantibodies [TBAb's]) also poorly recognize the wild-type TSHR relative to the ECD-GPI. Despite poor recognition by TSAb of the holoreceptor, soluble TSHR A subunits (known to be shed from surface TSHR) fully neutralized autoantibody-binding activity. These data indicate that the epitope(s) for TSAb's, but not for TBAb's, are partially sterically hindered on the holoreceptor by the plasma membrane, the serpentine region of the TSHR, or by TSHR dimerization. However, the TSAb epitope on the soluble A subunit is freely accessible. This observation, as well as other evidence, supports the concept that A subunit shedding either initiates or amplifies the autoimmune response to the TSHR, thereby causing Graves disease in genetically susceptible individuals.


Assuntos
Autoanticorpos/metabolismo , Doença de Graves/imunologia , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/metabolismo , Animais , Anticorpos Monoclonais , Especificidade de Anticorpos , Células CHO , Cricetinae , Reagentes de Ligações Cruzadas , Doença de Graves/metabolismo , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide , Camundongos , Testes de Neutralização , Subunidades Proteicas , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Tireotropina/metabolismo
16.
Endocrinology ; 147(6): 2789-800, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16543368

RESUMO

Immunization with adenovirus encoding the TSH receptor (TSHR) or its A-subunit induces Graves' hyperthyroidism in BALB/c and BALB/c x C57BL/6 offspring but not C57BL/6 mice. High-resolution genetic maps are available for 13 recombinant inbred CXB strains generated from BALB/c x C57BL/6 progeny by repeated brother x sister matings to establish fully inbred lines. CXB strains were studied before and after A-subunit adenovirus immunization for TSHR antibodies (TBI, inhibition of TSH binding), serum T4, and thyroid histology. All strains developed TBI activity (at variable levels), six strains became hyperthyroid, and one was overtly thyrotoxic. No low TBI responders became hyperthyroid, but high TBI did not predict hyperthyroidism. Preimmunization T4 levels varied in different CXB strains and was unrelated to subsequent T4 elevation. Linkage analysis indicated that different chromosomes were involved in generating TSHR antibodies and serum T4 before and after immunization. TBI activity was linked in part with major histocompatibility (MHC) genes on chromosome 17 (Chr 17) but induced Graves' disease involved non-MHC genes (Chr 19 and 10). The Chr 10 locus is close to the Trhde gene that encodes TSH-releasing hormone degrading enzyme. Expression of Trhde is controlled by thyroid hormones and linkage with a thyroid function-related gene is intriguing. Our data, the first genome scan in murine Graves' disease, provides insight into the role of MHC and non-MHC genes in human and murine Graves' disease. Finally, our study demonstrates the potential of recombinant inbred mice for discriminating between immune-response genes and thyroid function susceptibility genes in Graves' disease.


Assuntos
Autoanticorpos/biossíntese , Hipertireoidismo/genética , Receptores da Tireotropina/imunologia , Adenoviridae/genética , Animais , Formação de Anticorpos/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Ligação Genética , Hipertireoidismo/etiologia , Imunoglobulinas Estimuladoras da Glândula Tireoide , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Subunidades Proteicas , Receptores da Tireotropina/biossíntese , Receptores da Tireotropina/genética , Recombinação Genética , Tiroxina/sangue
17.
Endocrinology ; 147(6): 3107-13, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16497802

RESUMO

The prevailing concept is that, in human thyroid tissue in vivo, all cell-surface TSH receptors (TSHR) cleave into disulfide linked A and B subunits. Because this viewpoint is based on studies using homogenized thyroid tissue and because of TSHR fragility, we studied TSHR subunit structure in intact thyroid cells, primary human thyrocyte cultures, FRTL-5 rat thyroid cells, and WRO (follicular) and NPA (papillary) thyroid cancer cell lines. To overcome the handicap of very low TSHR expression in thyroid cells, we generated a TSHR-expressing adenovirus (TSHR-Ad-RGD) with an integrin-binding RGD motif enabling efficient entry into cells lacking the coxsackie-adenovirus receptor. Two days after TSHR-Ad-RGD infection, [125I]TSH cross-linking to intact cells revealed uncleaved, single-chain TSHR as well as cleaved TSHR A subunits on the surface of all four thyroid cell types. The extent of TSHR cleavage, which is independent of the level of TSHR expression, was consistently lower in the human thyroid cancer cell lines than in the other cell lines. In flow cytometry studies after TSHR-Ad-RGD infection, strong signals were detected in all four thyroid cell types using a monoclonal antibody that primarily recognizes the uncleaved TSHR. Finally, using the same monoclonal antibody, confocal microscopy confirmed the presence of single-chain TSHR on TSHR-Ad-RGD-infected thyroid cells. In summary, TSH covalent cross-linking, flow cytometry, and confocal microscopy demonstrate the presence of uncleaved TSHR on the human thyrocyte surface. These data provide stronger evidence for this alternative than the contrary view based on the finding of only cleaved TSHR in homogenized thyroid cells.


Assuntos
Receptores da Tireotropina/análise , Glândula Tireoide/química , Animais , Células CHO , Membrana Celular/química , Cricetinae , Citometria de Fluxo , Camundongos , Microscopia Confocal , Ensaio Radioligante , Receptores da Tireotropina/química , Glândula Tireoide/citologia , Tireotropina/metabolismo
18.
Thyroid ; 16(5): 427-34, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16756463

RESUMO

OBJECTIVE: Immune responses require costimulatory interactions between molecules on antigen-presenting cells and T cells: CD40 binding to CD40 ligand and B7 binding to CD28. Graves' hyperthyroidism is induced in BALB/c mice by immunization with thyrotropin receptor (TSHR) A-subunit adenovirus (Ad-A-subunit). We attempted to modulate Ad-A-subunit-induced Graves' disease using adenoviruses expressing costimulation "decoys": CD40-IgG-Fc (CD40-Ig) to block CD40:CD40-ligand interactions and CTLA4-Fc (CTLA4-Ig) to prevent B7:CD28 binding. OUTCOME: Unexpectedly, coimmunizing mice with Ad-A-subunit and excess control adenovirus (1:10 Ad-A-subunit:Ad-control) reduced TSHR antibody levels (thyrotropin binding inhibition [TBI]). Furthermore, only 15% of mice developed hyperthyroidism versus 75% using the same Ad-A-subunit dose (10(8) particles) without Ad-control. This effect was related to the dose of control adenovirus but not to the adenovirus insert, the timing or immunization site. Increasing the Ad-subunit dose (10(9) particles) and decreasing the control adenovirus dose (10:1 Ad-A-subunit:Ad-control) induced high TBI levels and 80% of mice were hyperthyroid. Coimmunization with Ad-CD40-Ig (but not Ad-CTLA4-Ig) reduced the incidence of hyperthyroidism to 40%. CONCLUSIONS: Using appropriate controls and adenovirus ratios, our data suggest the importance of CD40:CD40-ligand interactions for inducing Graves' hyperthyroidism by Ad-A-subunit. Furthermore, our observations emphasize the potential pitfalls of non-specific inhibition by coimmunization with two adenovirus species.


Assuntos
Adenoviridae/genética , Células Apresentadoras de Antígenos/citologia , Doença de Graves/terapia , Receptores da Tireotropina/genética , Linfócitos T/citologia , Animais , Antígenos CD28/biossíntese , Antígenos CD40/biossíntese , Ligante de CD40/biossíntese , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Hipertireoidismo/patologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Ligação Proteica
19.
Endocrinology ; 146(1): 294-300, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15459116

RESUMO

NOD.H-2h4 mice are genetically predisposed to thyroid autoimmunity and spontaneously develop thyroglobulin autoantibodies (TgAb) and thyroiditis. Iodide administration enhances TgAb levels and the incidence and severity of thyroiditis. Using these mice, we investigated the interactions between TSH receptor (TSHR) antibodies induced by vaccination and spontaneous or iodide-enhanced thyroid autoimmunity (thyroiditis and TgAb). Mice were immunized with adenovirus expressing the TSHR A-subunit (or control adenovirus). Thyroid antibodies, histology, and serum thyroxine levels were compared in animals on a regular diet or on a high-iodide diet (0.05% NaI-supplemented water). Thyroiditis severity and TgAb levels were enhanced by iodide administration and were independent of the type of adenovirus used for immunization. In contrast, TSHR antibodies, measured by TSH-binding inhibition, thyroid-stimulating activity, and TSH-blocking activity, were induced in the majority of animals immunized with TSHR (but not control) adenovirus and were unaffected by dietary iodide. The NOD.2h4 strain of mice was less susceptible than BALB/c or BALB/k mice to TSHR adenovirus-induced hyperthyroidism. Nevertheless, hyperthyroidism developed in approximately one third of TSHR adenovirus-injected NOD.2h4 mice. This hyperthyroidism was suppressed by a high-iodide diet, probably by a nonimmune mechanism. The fact that inducing an immune response to the TSHR had no effect on thyroiditis raises the possibility that the TSHR may not be the target involved in the variable thyroiditis component in some humans with Graves' disease.


Assuntos
Autoanticorpos/imunologia , Hipertireoidismo/imunologia , Iodetos , Camundongos Endogâmicos NOD/genética , Receptores da Tireotropina/imunologia , Tireoidite/induzido quimicamente , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Formação de Anticorpos , Autoanticorpos/biossíntese , Hipertireoidismo/patologia , Hipertireoidismo/fisiopatologia , Imunização , Imunoglobulinas Estimuladoras da Glândula Tireoide , Camundongos , Receptores da Tireotropina/genética , Especificidade da Espécie , Tireoglobulina/imunologia , Glândula Tireoide/patologia , Glândula Tireoide/fisiopatologia , Tireoidite/sangue , Tireoidite/patologia , Tireoidite/fisiopatologia , Tiroxina/sangue
20.
Endocrinology ; 146(11): 4961-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16081633

RESUMO

We investigated the relationship between thyroid peroxidase (TPO) antibody and T lymphocyte epitopes in TPO-adenovirus (TPO-Ad) immunized BALB/c mice and mice transgenic for the human class II molecule DR3 associated with human thyroid autoimmunity. TPO autoantibodies are largely restricted to an immunodominant region (IDR). BALB/c mice immunized with fewer (10(7) vs. 10(9)) TPO-Ad particles developed TPO antibodies with lower titers that displayed greater restriction to the IDR. However, as with higher-dose TPO-Ad immunization, T cell epitopes (assessed by splenocyte interferon-gamma response to TPO in vitro) were highly diverse and variable in different animals. In contrast, DR3 mice immunized the higher TPO-Ad dose (10(9) particles) had high TPO antibody levels that showed relative focus on the IDR. Moreover, T cell epitopes recognized by splenocytes from DR3 mice showed greater restriction than BALB/c mice. Antibody affinities for TPO were higher in DR3 than in BALB/c mice. The present study indicates that weak TPO-Ad immunization of BALB/c mice (with consequent low TPO antibody titers) is required for enhanced IDR focus yet is not associated with T cell epitopic restriction. Humanized DR3 transgenic mice, despite stronger TPO-Ad immunization, develop higher titer TPO antibodies that do focus on the autoantibody IDR with T cells that recognize a more limited range of TPO peptides. These data suggest a relationship between major histocompatibility complex class II molecules and the development of antibodies to the IDR, a feature of human thyroid autoimmunity.


Assuntos
Autoanticorpos/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-DR3/imunologia , Epitopos Imunodominantes/imunologia , Iodeto Peroxidase/imunologia , Adenoviridae/genética , Animais , Células Cultivadas , Feminino , Vetores Genéticos , Antígeno HLA-DR3/genética , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Baço/citologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA