Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lupus ; 21(3): 288-301, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22065096

RESUMO

While it has been shown that estradiol treatment accelerates the onset of lupus nephritis with autoantibody production and kidney damage in both male and female lupus-prone mice, the specific mechanism(s) involved are unknown. Our previous work has shown that alterations in Id(LN)F(1)-reactive T cells and Id(LN)F(1)+ antibodies correlated closely with the onset of autoimmune nephritis in female F(1) progeny of SWR and NZB (SNF(1)) mice, supporting a critical role for the Id(LN)F(1) idiotype in the development of disease. Since male SNF(1) mice normally do not develop nephritis, we tested whether administration of 17ß-estradiol (E-2) to male SNF(1) mice would increase Id(LN)F(1) IgG levels and autoreactive T cells, and further, induce nephritis. We found that E-2-treated male SNF(1) mice developed nephritis with the same time course and mean survival as normal female SNF(1) mice. Moreover, it appeared that the mechanism involved increased serum Id(LN)F(1)(+)IgG and its deposition in kidney glomeruli, preceded by a striking twofold increase in T-lymphocytes expressing the memory phenotype (CD44(+)CD45RB(lo)) predominantly in the Id(LN)F(1)-reactive T-cell population. In addition, we noted that cells with this phenotype were increased in the nephritic kidneys of treated mice, suggesting a direct involvement of those cells in the renal pathology. E-2 treatment also induced increased numbers of pathogenic Id(LN)F(1)+ antibody-producing B cells and elevated presentation of pathogenic Id(LN)F(1)+ peptide. Taken together, these results suggest a mechanism of E-2-induced acceleration of autoimmune disease in lupus-prone mice may involve expansion of autoreactive idiotypic T and B-cell populations.


Assuntos
Estradiol/toxicidade , Glomerulonefrite/fisiopatologia , Nefrite Lúpica/fisiopatologia , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Feminino , Glomerulonefrite/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Idiótipos de Imunoglobulinas/imunologia , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Nefrite Lúpica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NZB , Fatores Sexuais , Sobrevida , Fatores de Tempo
2.
Toxicol Appl Pharmacol ; 168(1): 15-24, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11000096

RESUMO

Both the estrogenic drug diethylstilbestrol (DES) and the pervasive environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibit thymocyte development. The mechanisms by which either agent induces thymic atrophy are still undetermined. We previously found that TCDD and DES inhibited C57BL/6 murine fetal thymocyte organ cultures (FTOC) at different stages of development. Now, using bcl-2 transgenic (TG) mice, we have further investigated their effects on FTOC proliferation, differentiation, maturation, and apoptosis. As with C57BL/6 mice, thymocyte development in C3H/bcl-2 FTOCs was inhibited by either TCDD (10 nM) or DES (20 microM) in both bcl-2 TG- and TG+ littermates. However, the percentage reduction of cell number induced by DES in bcl-2 TG+ FTOCs was significantly less than the level of inhibition in TG- FTOCs. There was no difference in the level of reduction from TCDD-exposed TG+ or TG- FTOC. Whereas TCDD increased production of mature CD8 cells in either strain, DES mainly yielded cells in the CD4(-)CD8(-)(DN) stage in TG- mice. The anti-apoptotic bcl-2 transgene overcame some DES blocking of DN thymocyte development, allowing more cells to differentiate into CD4 single-positive cells. Analysis of cell cycle showed that TCDD inhibited entry into S phase, whereas DES blocked cell cycling in the G2/M phase. TCDD did not induce detectable apoptosis in FTOC. However, unlike the effects of 17 beta-estradiol (E2) in vivo, DES induced apoptosis in the TG- FTOC, and these apoptotic cells were mainly in the DN subpopulation. This apoptosis could be prevented by the overexpression of bcl-2 in the TG+ mice. Our results demonstrate that, in addition to inhibition of fetal thymocytes at different stages of development by TCDD and DES, DES also induces thymic atrophy by both bcl-2-inhibitable apoptosis and by inducing cell cycle arrest in G2/M in the latest stage in the stem cell compartment. TCDD, on the other hand, does not induce apoptosis, but inhibits entry into cell cycle in the earliest stage in the stem cell compartment.


Assuntos
Apoptose/efeitos dos fármacos , Dietilestilbestrol/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Linfócitos T/efeitos dos fármacos , Timo/efeitos dos fármacos , Animais , Atrofia , Ciclo Celular/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Gravidez , Linfócitos T/fisiologia , Timo/patologia
3.
J Immunol ; 163(8): 4168-74, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10510352

RESUMO

Estrogens affect the development, maturation, and function of multiple organ systems, including the immune system. One of the main targets of estrogens in the immune system is the thymus, which undergoes atrophy and phenotypic alterations when exposed to elevated levels of estrogen. To determine how estrogens influence the thymus and affect T cell development, estrogen receptor alpha (ERalpha) knockout (ERKO) mice were examined. ERKO mice have significantly smaller thymi than their wild-type (WT) littermates. Construction of ER radiation bone marrow chimeras indicated that the smaller thymi were due to a lack of ERalpha in radiation-resistant tissues rather than hemopoietic elements. ERKO mice were also susceptible to estradiol-induced thymic atrophy, but the extent of their atrophy was less than what was seen in WT mice. The estradiol-treated ERKO mice failed, however, to manifest alterations in their thymic CD4/CD8 phenotypes compared with WT mice. Therefore, ERalpha is essential in nonhemopoietic cells to obtain a full-sized thymus, and ERalpha also mediates some of the response of the thymus to elevated estrogen levels. Finally, these results suggest that in addition to ERalpha, another receptor pathway is involved in estradiol-induced thymic atrophy.


Assuntos
Adjuvantes Imunológicos/fisiologia , Estradiol/farmacologia , Receptores de Estrogênio/fisiologia , Timo/crescimento & desenvolvimento , Timo/patologia , Adjuvantes Imunológicos/genética , Animais , Atrofia , Tamanho Celular/imunologia , Receptor alfa de Estrogênio , Feminino , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Imunofenotipagem , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quimera por Radiação/imunologia , Receptores de Estrogênio/genética , Células Estromais/metabolismo , Células Estromais/fisiologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/efeitos da radiação , Timo/imunologia , Timo/metabolismo
4.
Toxicol Appl Pharmacol ; 151(1): 200-10, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9705904

RESUMO

Dexamethasone (Dex), estradiol (E2), and 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) all affect the immune system, causing immunosuppression and thymic atrophy. It is still uncertain how and where these compounds act to induce thymic atrophy. However, it has been suggested that these compounds may have similar actions and targets, i.e., apoptosis of immature thymocytes for Dex and TCDD and preferential targeting of double-positive cells by Dex and E2. The lckpr-bcl-2 transgenic mouse has been shown to be protected against Dex-induced thymic atrophy. We used this murine model to determine if bcl-2 expression would also protect against E2- and TCDD-induced thymic atrophy. Our results indicate that, although the bcl-2 transgenic (TG+) mice were fully protected from atrophy induced by a single dose of Dex, atrophy was still induced in these mice following treatment with E2 or TCDD. Phenotypic analysis of thymocytes from TG- and TG+ mice also showed distinct consequences of atrophy induced by Dex, E2, and TCDD. Finally, since there are alternative pathways for apoptosis that are bcl-2 independent, both TG- and TG+ thymocytes were examined directly for indications of apoptosis using the TUNEL assay. After TCDD and E2 treatment there were no detectable signs of apoptosis in either TG- or TG+ mice even at early time points and at elevated dose levels. These results indicate that there are distinct mechanisms for the actions of Dex, E2, and TCDD in the thymus and that apoptosis is not a key mechanism of E2- and TCDD-induced thymic atrophy.


Assuntos
Estradiol/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Timo/efeitos dos fármacos , Animais , Apoptose/genética , Atrofia/induzido quimicamente , Atrofia/prevenção & controle , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Contagem de Células , Dexametasona/toxicidade , Proteínas de Ligação ao GTP/biossíntese , Proteínas de Ligação ao GTP/genética , Camundongos , Camundongos Transgênicos , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Timo/metabolismo , Timo/patologia
5.
Toxicol Appl Pharmacol ; 149(2): 167-77, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9571985

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and estrogen induce thymic atrophy and alter thymocyte development. In the present study we investigate whether TCDD and the synthetic estrogen diethylstilbestrol (DES) alter intrathymic development by the same or different mechanisms. We compared the effects of TCDD and DES on thymocyte development in fetal thymus organ culture (FTOC) and found that both compounds caused a reduction in cell yield. TCDD- and DES-treated FTOCs yielded fewer CD4 + CD8+ double-positive cells. However TCDD treatment also led to a greater percentage of cells in the CD8+ single-positive compartment. At lower dioxin concentrations, our results demonstrated an actual increase in CD8+ cells, whereas DES-treated fetal thymocytes were mainly enriched in CD4-CD8- double-negative cells. More alpha beta-TCR+ positive cells were seen in TCDD- but not in DES-exposed cultures. Furthermore, in this study we found that TCDD and DES also alter intrathymic development at different stages in the CD4-CD8- double-negative compartment. TCDD induced a relative increase in c-kit + CD44 + CD25-HSA-thymocytes, while DES induced an relative increase in c-kit-CD44-CD25 + HSA+ cells. RT-PCR revealed that TCDD reduced RAG-1, RAG-2, and TdT gene expression in the CD4-CD8- double-negative thymocytes. Co-treatment by TCDD and DES in FTOC yielded a mixture of effects induced by each agent. Taken together, our results demonstrate that TCDD and DES affect thymocytes at different stages of development, suggesting distinct mechanisms for induction of thymic atrophy.


Assuntos
Adjuvantes Imunológicos/toxicidade , Dietilestilbestrol/toxicidade , Estrogênios não Esteroides/toxicidade , Imunossupressores/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Timo/efeitos dos fármacos , Animais , Relação CD4-CD8/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Contagem de Células/efeitos dos fármacos , Dietilestilbestrol/administração & dosagem , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Dibenzodioxinas Policloradas/administração & dosagem , Receptores de Antígenos de Linfócitos T/biossíntese , Timo/embriologia , Timo/patologia
6.
J Immunol ; 160(8): 3844-54, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9558089

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related congeners affect the immune system, causing immunosuppression and thymic atrophy in a variety of animal species. TCDD is believed to exert its effects primarily through the ligand-activated transcription factor, the aryl hydrocarbon receptor (AhR). Although the AhR is found at high levels in both thymocytes and thymic stroma, it is uncertain in which cells TCDD is activating the AhR to cause alterations in the thymus. Some investigators have suggested that stromal elements, primarily epithelial cells, within the thymus are the primary targets for TCDD. Others have suggested that atrophy is due to a direct effect on thymocytes, either by apoptosis or by altering the development of progenitor cells. By producing chimeric mice with TCDD-responsive (AhR[+/+]) stromal components and TCDD-unresponsive (AhR[-/-]) hemopoietic components, or the reverse, we have clarified the role of stromal vs hemopoietic elements in TCDD-induced thymic alterations. Our results show that the targets for TCDD-induced thymic atrophy and phenotypic alterations are strictly in the hemopoietic compartment and that TCDD activation of epithelial cells in the stroma is not required for thymic alterations. Furthermore, changes observed in the putative stem cell populations of these chimeric mice are also dependent on TCDD activation of the AhR in hemopoietic elements.


Assuntos
Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Timo/efeitos dos fármacos , Timo/metabolismo , Animais , Atrofia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A1/genética , Hematopoese/efeitos dos fármacos , Receptores de Hialuronatos/metabolismo , Imunossupressores/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Quimera por Radiação , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/imunologia , Receptores de Interleucina-2/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/imunologia
7.
Toxicol Appl Pharmacol ; 126(2): 248-59, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7516097

RESUMO

The effects of single doses of dexamethasone (DEX), beta-estradiol-17-valerate (E2), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the kinetics of thymic atrophy and related bone marrow and thymocyte phenotype alterations were examined. The results imply differences in the mechanisms by which these compounds act. Of the three compounds, DEX induced maximal atrophy by 3 days with complete recovery by Day 12. At the point of maximal atrophy, the RAG-1+TdT+CD4+8+3int thymocyte population was proportionately the most depleted. In contrast, TCDD and E2 caused maximal thymic atrophy by Day 12. E2 treatment, like DEX, resulted in a preferential decrease in the RAG-1+TdT+CD4+8+3int population, but unlike DEX, this decrease persisted. TCDD-induced thymic atrophy resulted from a proportional loss of all classes of thymocytes. There was no significant relative reduction of TdT+RAG-1+ cells by TCDD in the thymus. A slow and persistent reduction of TdT and RAG-1 in bone marrow by both TCDD and E2 contrasted with the rapid reduction and quick recovery of these markers in marrow from DEX-treated animals. Additional studies showed that only DEX-induced atrophy was accompanied by the induction of thymocyte apoptosis, as detected by multiple nucleosomal length DNA fragments within the first 24 hr. The different kinetics and proportions of subsets in the atrophied thymuses, as well as the distinct patterns of alterations of RAG and TdT expression, and the presence or the absence of apoptosis provide evidence for different mechanisms of thymic atrophy by these agents. The slow induction and longer persistence of thymic atrophy induced by E2 and TCDD, as well as their effects on bone marrow stem cell markers, suggest that bone marrow thymocyte precursors are major targets for these agents.


Assuntos
Medula Óssea/efeitos dos fármacos , Dexametasona/toxicidade , Estradiol/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Timo/efeitos dos fármacos , Animais , Antígenos de Superfície/análise , Apoptose/efeitos dos fármacos , Medula Óssea/imunologia , Células da Medula Óssea , DNA/análise , Feminino , Imunofluorescência , Linfoma de Células T/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão/efeitos dos fármacos , RNA/análise , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Timo/citologia , Timo/imunologia , Células Tumorais Cultivadas
8.
J Pharmacol Exp Ther ; 255(1): 128-32, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2213547

RESUMO

The mechanism by which exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) produces thymic atrophy and cell-mediated immune suppression in experimental animals is poorly understood. A previous study from our laboratory found that terminal deoxynucleotidyl transferase-synthesizing lymphocyte stem cell populations in fetal liver and neonatal bone marrow, but not thymus, were profoundly altered after perinatal TCDD exposure, implying that a defect in the prothymocyte population in liver and marrow may play a role in the etiology of thymic atrophy in TCDD-exposed animals. In this report, we present results of experiments designed to directly assess the prothymocyte compartment in mice exposed to TCDD perinatally by examining the ability of these stem cells to reconstitute an irradiated thymus. Maternal TCDD exposure (15 micrograms/kg) caused a significant impairment of both fetal liver and neonatal bone marrow prothymocyte activity. These alterations occurred at tissue concentrations less than 200 fg of TCDD per mg. TCDD treatment also resulted in a mild reduction in colony-forming unit-spleen in these organs and a decrease in colony-forming unit-granulocyte-macrophage in fetal and neonatal liver, but not bone marrow. Overall, these data provide evidence that alterations to early stages of T-lymphopoiesis, at the level of the prothymocyte, may be involved in the development of TCDD-induced thymic atrophy and cell-mediated immunosuppression.


Assuntos
Feto/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Linfócitos T/efeitos dos fármacos , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Gravidez , Timo/efeitos dos fármacos , Timo/patologia
9.
J Exp Med ; 138(4): 925-38, 1973 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-4744012

RESUMO

AKR mice with spontaneous leukemia were infused with normal serum from a variety of species. Leukemia cell destruction was produced by serum from strains of mice possessing the full spectrum of complement components, but not by serum from strains with a genetically determined deficiency of C5. Serum from guinea pigs, horses, and humans also causes destruction of leukemia cells. The antileukemic factor in normal serum was heat labile (56 degrees C for 35 min) and could be inactivated by cobra venom factor (CVF). Tests of individual complement factors from guinea pig serum and from human serum suggest that C5 is the antileukemic complement component in normal serum. Evidence was obtained that complement also plays a role in the antileukemic effect of interferon and endotoxin.


Assuntos
Proteínas do Sistema Complemento , Soros Imunes , Leucemia Experimental/imunologia , Linfonodos/imunologia , Camundongos Endogâmicos AKR , Baço/imunologia , Animais , Complexo Antígeno-Anticorpo , Modelos Animais de Doenças , Endotoxinas , Cobaias , Cavalos , Humanos , Interferons , Leucemia Experimental/patologia , Camundongos , Especificidade da Espécie , Peçonhas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...