Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
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 ; 171(3): 157-64, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11243915

RESUMO

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates many of the toxic effects induced by exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a high-affinity AhR ligand and a potent immunotoxicant. AhR-deficient mice have been constructed, and there are reports that the animals display altered splenic architecture and cellularity with an apparent increased incidence of infection. These observations have led to speculation that the immune system of these animals might be compromised, however, their functional immune response has not been directly tested. In the studies presented here, we examined the immune response of two strains of 8- to 10-week-old AhR-deficient mice. Mice were challenged with model antigens, allogeneic P815 tumor cells, or sheep red blood cells, and their ability to generate cell-mediated and humoral immune responses was examined. In addition, to address the obligatory role of the AhR in TCDD-induced immune suppression, we examined the immune response of the AhR-null animals following exposure to an immunosuppressive dose of TCDD. Results from these studies showed that AhR-deficient mice were able to mount normal productive immune responses to both model antigens and that neither the cellular nor the humoral response was suppressed by exposure to TCDD. Interestingly, however, we found that the immune response of heterozygous AhR(+/-) mice was less sensitive to TCDD than homozygous AhR(+/+) mice. The results of these studies suggest that the absence of the AhR does not impact the function of the immune system, but confirm the findings of previous studies that have indicated the AhR plays an obligatory role in TCDD-induced immune suppression.


Assuntos
Formação de Anticorpos/genética , Imunossupressores/farmacologia , Dibenzodioxinas Policloradas/farmacologia , Receptores de Hidrocarboneto Arílico/deficiência , Animais , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/imunologia , Resistência a Medicamentos , Eritrócitos/imunologia , Citometria de Fluxo , Receptores de Hidrocarboneto Arílico/genética , Ovinos , Baço/citologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
3.
Carcinogenesis ; 22(1): 171-7, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11159756

RESUMO

This laboratory has previously reported data suggesting that aryl hydrocarbon receptor (AhR) signaling may have a net potentiating effect on the DNA damaging potential of cigarette smoke. The experiments described in this report extend these studies by testing whether the potent AhR antagonist 3'-methoxy-4'-nitroflavone (3'M4'NF) can modify the in vivo genetic toxicity of benzo[a]pyrene (B[a]P) and the complex mixture of chemicals in cigarette smoke condensate (CSC). Initial experiments were designed to determine 3'M4'NF doses which can antagonize AhR in vivo but which have little effect on constitutive cytochrome P4501A (CYP1A) activity. These experiments took three forms: (i) zoxazolamine paralysis tests, a functional assay of cytochrome P450 CYP1A activity in 3'M4'NF-treated C57Bl/6J mice; (ii) co-treatment of AHR: null allele mice with 150 mg/kg B[a]P plus a range of 3'M4'NF concentrations in order to evaluate the potential of the flavone to interact with non-AhR targets which may affect B[a]P toxicity; (iii) an evaluation of the in vivo AhR antagonist activity of 3'M4'NF using transgenic mice which carry a dioxin-responsive element-regulated lacZ reporter. Once an appropriate dose range was determined, C57Bl/6J mice were challenged with B[a]P or CSC with and without 3'M4'NF co-treatment. Chromosome damage was measured by scoring the frequency of micronuclei in peripheral blood reticulocytes. Data presented herein suggest that 3'M4'NF can protect mice from B[a]P-induced bone marrow cytotoxicity and genotoxicity. Furthermore, CSC-associated genotoxicity was abolished by the flavonoid. These data add support to our hypothesis that AhR signaling has a net potentiating effect on the genetic toxicity and, presumably, carcinogenicity of cigarette smoke.


Assuntos
Benzo(a)pireno/toxicidade , Nicotiana , Plantas Tóxicas , Receptores de Hidrocarboneto Arílico/fisiologia , Transdução de Sinais/fisiologia , Fumaça/efeitos adversos , Animais , Benzo(a)pireno/antagonistas & inibidores , Cromossomos/efeitos dos fármacos , Citocromo P-450 CYP1A1/antagonistas & inibidores , Inibidores do Citocromo P-450 CYP1A2 , Dano ao DNA , Flavonoides/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/genética , Transdução de Sinais/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/patologia
5.
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
6.
Genesis ; 27(2): 64-75, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10890980

RESUMO

The cardiac neural crest (CNC) plays a central role in development of the thymus gland and cardiovascular system. Through morphological and histological characterization of embryos homozygous for the Del(7)Tyr(c-112K) and Del(7)Tyr(c-3H) albino deletions, we identified abnormalities that are consistent with aberrant development of tissues requiring CNC contributions. The defects include incompletely penetrant heart and great vessel patterning defects and hypoplastic thymus glands. The CNC phenotype is complemented by the partially overlapping deletion Del(7)Tyr(c-23DVT). Combined, these results suggest that a functional region necessary for development of CNC derived tissues is located between the Del(7)Tyr(c-23DVT) and Del(7)Tyr(c-112K) distal deletion breakpoints. This interval encompasses a functional region previously identified as important for juvenile survival (juvenile development and fertility, jdf). Using deletion mapping, we localized the Frizzled4 (Fzd4) gene to the jdf/thymus and cardiac development intervals.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Coração/fisiologia , Proteínas/genética , Timo/embriologia , Timo/fisiologia , Animais , Mapeamento Cromossômico , Desenvolvimento Embrionário e Fetal/genética , Receptores Frizzled , Camundongos , Mutação , Biossíntese de Proteínas , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G
7.
Endocrinology ; 141(7): 2309-18, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10875230

RESUMO

Although estrogens and estrogen receptors (ERs) are known to function in the male brain and reproductive tract, few studies have evaluated their involvement in the male hematopoietic and immune systems. This study was undertaken to determine the role of ERalpha in hematopoietic progenitor and B lymphocyte maturation. ERalpha knockout (ER-/-), wild-type (ER+/+), and radiation chimeric (ERalpha positive or negative in either nonhematopoietic or hematopoietic elements, or both) male mice were used to determine target tissues. ER-/- and ER+/+ animals showed similar hematopoietic progenitor profiles, but the ER-/- animals had fewer cells in all bone marrow B lymphocyte subpopulations. Animals receiving a pharmacological dose (5 mg/kg BW) of 17beta-estradiol (E2) with both elements, ER+/+, had decreased early hematopoietic progenitors and a shift toward a mature B cell subpopulation, whereas animals with both elements, ER-/-, showed changes only in early hematopoietic progenitors. Hematopoietic element ER+/+ animals exhibited greater E2-induced hematopoietic progenitor and B lymphocyte alterations than those having only nonhematopoietic ERalpha. These data indicate that 1) ERalpha is not necessary for regulating male mouse normal hematopoietic progenitor cell proportions, but is involved in B cell regulation; and 2) ERalpha in hematopoietic elements is predominantly responsible for mediating E2-induced hematopoietic and B cell changes.


Assuntos
Linfócitos B/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Receptores de Estrogênio/fisiologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Quimera , Estradiol/farmacologia , Receptor alfa de Estrogênio , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Receptores de Estrogênio/genética
8.
Toxicol Appl Pharmacol ; 165(3): 227-36, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10860871

RESUMO

The ligand-activated aryl hydrocarbon receptor (AHR) is a cytosolic DNA binding protein. Although no biologic role for AHR has been elucidated, it mediates the immunotoxicity of xenobiotics such as 2, 3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and its targeted inactivation produces abnormal immune system development. While investigators have demonstrated AHR's involvement in TCDD-induced B lymphocyte functional alterations, little is known about the receptor's possible role in early B cell maturation and whether exogenous ligands change this process. The purpose of this study was to determine, (1) whether bone marrow B lymphocyte maturation is affected by AHR presence, (2) if so, its relative importance in hematopoietic and/or nonhematopoietic elements and, (3) whether TCDD alters this process. Radiation chimeras were produced that were AHR positive (Ahr+/+) or negative (Ahr-/-) in either their nonhematopoietic or hematopoietic elements, or both. Marrow cells were analyzed for alterations in B lymphocyte maturation stage cell numbers in both vehicle- and TCDD-treated animals. Our results showed that (1) Ahr-/- animals had significantly higher numbers of pro/pre-B cells than Ahr+/+ animals, (2) TCDD treatment of Ahr+/+ animals produced a decrease in pro/pre-B cell numbers, whereas no effect was observed on Ahr-/- animals, and (3) AHR is required in both hematopoietic and stromal elements for maintenance of B cell subset maturation profiles.


Assuntos
Linfócitos B/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Linfócitos B/citologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
Biochem Pharmacol ; 60(2): 189-96, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10825463

RESUMO

This laboratory has studied a number of flavone derivatives for aryl hydrocarbon receptor (AhR) agonist and antagonist potential using cell-free and cell culture systems. The current report extends these investigations by testing the potent AhR antagonist 3'-methoxy-4'-nitroflavone (3'M4'NF) for in vivo activity. Wild-type C57Bl/6 male mice were treated with solvent, benzo[a]pyrene (B[a]P; 150 mg/kg), or concurrently with B[a]P and 3'M4'NF (60 mg/kg; delivered as a split dose). Since B[a]P is bioactivated to genotoxic metabolites by AhR-regulated enzymes, we measured B[a]P-induced chromosomal damage in peripheral blood (i.e. micronuclei) to characterize the antagonistic potential of 3'M4'NF in vivo. The influence of AhR signal transduction was investigated further by challenging wild-type and Ahr null allele mice with B[a]P with and without a 3'M4'NF co-treatment. The micronucleus data obtained from these experiments indicated that 3'M4'NF can attenuate the genotoxicity of B[a]P significantly. Since 3'M4'NF also protected Ahr null allele mice from B[a]P-induced genetic damage, it was apparent that AhR-independent mechanisms contribute to the effects observed. However, as opposed to the protective effects observed with the micronucleus endpoint, histological observations and lethality data indicated that some B[a]P effects are enhanced by 3'M4'NF. Potentiated B[a]P toxicity may be explained by inhibition of basal and induced CYP1A1/2 activities. Both in vitro and in vivo data presented herein support this hypothesis.


Assuntos
Benzo(a)pireno/antagonistas & inibidores , Flavonoides/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Animais , Antimutagênicos/farmacologia , Benzo(a)pireno/toxicidade , Citocromo P-450 CYP1A1/antagonistas & inibidores , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Inibidores do Citocromo P-450 CYP1A2 , Interações Medicamentosas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Mutagenicidade , Mutagênicos/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo
10.
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
11.
Toxicol Appl Pharmacol ; 158(1): 33-40, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10387930

RESUMO

Pathologic changes associated with 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) exposure have been reported in the livers of a wide range of species. While these changes have been extensively described, the mechanisms of toxic interaction(s) that produce these lesions remain unclear. Using an aryl hydrocarbon receptor (Ahr) knockout male mouse chimeric model, we investigated whether the presence of this receptor in hematopoietic and/or parenchymal cells affects TCDD-induced hepatotoxicity. Bone marrow chimeras were produced by hematopoietic reconstitution of irradiated mice. Specifically, chimeras were generated with aryl hydrocarbon receptor (AHR) positive hematopoietic and parenchymal cells (Ahr+/+ animal bone marrow cells into irradiated Ahr+/+ animals), AHR positive hematopoietic and negative parenchymal cells (Ahr+/+ into Ahr-/-), AHR negative hematopoietic and positive parenchymal cells (Ahr-/- into Ahr+/+), and AHR negative hematopoietic and parenchymal cells (Ahr-/- into Ahr-/-). Male wild-type (Ahr+/+) and knockout (Ahr-/-) animals were used as nonchimeric controls. Following TCDD treatment (30 microg/kg body wt), liver sections from mice in each control and chimeric group were histologically evaluated for necrotic and inflammatory changes. TCDD treatment produced moderate inflammation in Ahr+/+ controls and Ahr+/+ into Ahr+/+ chimeras. This response was mild in TCDD-treated Ahr-/-, Ahr-/- into Ahr-/-, Ahr+/+ into Ahr-/-, and Ahr-/- into Ahr+/+ animals and was not different from the corresponding vehicle-treated groups. Moderate necrosis was observed in all TCDD-treated controls or chimeras with AHR-positive parenchyma. No or mild necrosis was observed in TCDD- and vehicle-treated animals containing AHR-negative parenchyma. These data indicate that the presence of AHR in hepatic parenchyma alone is sufficient for TCDD induction of hepatic necrosis, and its presence in hematopoietic cells is necessary for the inflammatory response to TCDD-induced hepatic lesions.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Inflamação/induzido quimicamente , Fígado/efeitos dos fármacos , Fígado/patologia , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Masculino , Camundongos , Camundongos Knockout , Necrose , Quimera por Radiação , Distribuição Aleatória , Receptores de Hidrocarboneto Arílico/genética
12.
Carcinogenesis ; 19(11): 2037-42, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9855021

RESUMO

The role of aromatic hydrocarbon receptor (AhR)-mediated events on the genotoxicity of mainstream cigarette smoke condensate was investigated. In vitro studies with mouse hepatoma cells stably transfected with a DRE-dependent luciferase reporter indicate that cigarette smoke condensate is able to transform AhR to an active form which is capable of initiating gene transcription. Micronucleus formation in two hepatoma cell lines was used as an index of genotoxicity. Cigarette smoke condensate was observed to induce a higher frequency of micronuclei in Hepa1c1c7 cells relative to TAOc1BP(r)c1 cells, which express approximately 10-fold less AhR. Furthermore, the frequency of micronuclei was potentiated when Hepa1c1c7 cells were pretreated with 2,3,7,8-tetrachlorodibenzo-p-dioxin, a high affinity ligand of AhR. These in vitro studies were followed by an in vivo experiment with Ahr+/+ and Ahr-/- mice. Animals were dosed for three consecutive days with cigarette smoke condensate (0.5-10 microg/kg/day, i.p. injection). The frequency of micronuclei in reticulocytes and total erythrocytes was determined in peripheral blood samples collected 24 h after the last administration. While condensate was found to increase the incidence of micronucleated reticulocytes in Ahr+/+ mice, no increase was observed in the null allele animals. Furthermore, the frequency of micronucleated erythrocytes, a measure of basal chromosome-damaging activity, was slightly but significantly higher in Ahr+/+ relative to Ahr-/- mice. Together, these data suggest that cigarette smoke contains chemicals which transform the AhR to an active transcription factor and AhR-regulated enzyme induction plays an important role in mediating the genotoxicity of this complex environmental pollutant.


Assuntos
Mutagênicos/toxicidade , Nicotiana , Plantas Tóxicas , Receptores de Hidrocarboneto Arílico/fisiologia , Fumaça/efeitos adversos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade
13.
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
14.
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
15.
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
16.
Biochem Pharmacol ; 47(11): 2039-48, 1994 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8010988

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has both agonist and antagonist effects on estrogen-mediated activities and estrogen receptor (ER) levels in epithelial tissues following exposure. We previously demonstrated that TCDD alters bone marrow lymphocyte stem cells, including prothymocytes, as measured by functional assays and alterations in the lymphocyte stem cell-specific markers terminal deoxynucleotidyl transferase (TdT) and recombinase activating gene-1 (RAG-1). We have also shown that 17 beta-estradiol valerate (E2V) affects lymphocyte stem cells by reducing TdT and RAG-1 mRNA. It has been suggested that the effect of TCDD on these lymphocyte stem cells may be mediated directly or indirectly through estrogenic action and/or the ER. Studies were designed to evaluate whether endogenous estrogens or the ER mediate TCDD-elicited bone marrow alterations and thymic atrophy. Ovariectomy did not alter the sensitivity of mice to TCDD-induced thymic atrophy or to a reduction in TdT biosynthesis in bone marrow cells compared with either intact or sham-operated mice. The pure estrogen antagonist ICI 164,384 blocked E2V-induced uterine hypertrophy, thymic atrophy and reductions in lymphocyte stem cell markers. However, the antiestrogen failed to protect against TCDD-elicited thymic atrophy or bone marrow alterations in intact animals. The results are consistent with the hypothesis that the effects of TCDD on the thymus and/or bone marrow are mediated by mechanisms independent of estrogens or the ER.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Timo/efeitos dos fármacos , Animais , Peso Corporal , DNA Nucleotidilexotransferase/biossíntese , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Ovariectomia , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/efeitos dos fármacos , Timo/patologia , Útero/efeitos dos fármacos , Útero/patologia
17.
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
18.
Toxicol Appl Pharmacol ; 124(2): 242-7, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8122269

RESUMO

Our previous studies have shown that bone marrow lymphocyte stem cells are affected following perinatal or adult exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). These alterations may, in part, be responsible for thymic atrophy that is also observed following TCDD exposure. However, other investigators have suggested that the thymus or thymic-derived lymphocytes can affect bone marrow stem cell development. The purpose of these studies was to determine whether the TCDD-elicited effects that we have observed on lymphocyte stem cells in bone marrow were secondary to the actions of this chemical on the thymus. A single intraperitoneal dose of TCDD (30 micrograms/kg) to sham-operated or neonatally thymectomized female BALB/c mice reduced the levels of mRNA in the bone marrow for the lymphocyte stem cell-specific enzymes terminal deoxynucleotidyl transferase (TdT) and recombinase activating gene (RAG-1). TdT biosynthesis was also reduced by TCDD treatment. Thus, neonatal thymectomy had no effect on the TCDD-elicited reduction of TdT or RAG-1 mRNAs or TdT biosynthesis. Genetically athymic (nu/nu) mice were used to further determine if the actions of TCDD on the thymus or long-lived T-cells altered lymphocyte stem cell development. As observed in BALB/c mice, TCDD treatment decreased the expression of TdT and RAG-1 mRNAs in bone marrow from athymic nu/nu and intact nu/+ littermates. We conclude that TCDD-elicited alterations in bone marrow lymphocyte stem cells are not secondary to any actions, direct or indirect, that TCDD has on the thymus or thymic-derived T-cells.


Assuntos
Medula Óssea/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Células-Tronco/efeitos dos fármacos , Timo/fisiologia , Animais , Medula Óssea/ultraestrutura , Células Cultivadas , DNA Nucleotidiltransferases/biossíntese , Feminino , Genes RAG-1/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfócitos/metabolismo , Linfócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tamanho do Órgão/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Células-Tronco/ultraestrutura , Timectomia , Timo/efeitos dos fármacos , Timo/metabolismo
19.
Exp Clin Immunogenet ; 11(2-3): 94-101, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7826670

RESUMO

We here summarize evidence that thymic atrophy induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can be mediated, at least in part, by damage to extrathymic T-cell precursors in bone marrow and fetal liver. This atrophy induction does not involve apoptotic mechanisms in thymocytes affected by the bcl-2 proto-oncogene. TCDD mediates atrophy induction through its specific receptor (the AhR) and not through effects on the estrogen receptor. Both TCDD and estradiol induce extrathymic T-cell differentiation in the liver. These extrathymic T-cell populations include cells expressing elevated levels of V beta T-cell receptors that are normally deleted in thymic development.


Assuntos
Linfócitos/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Timo/efeitos dos fármacos , Animais , Atrofia/induzido quimicamente , Diferenciação Celular/efeitos dos fármacos , Dexametasona/toxicidade , Estradiol/toxicidade , Humanos , Fígado/efeitos dos fármacos , Proto-Oncogene Mas , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Receptores de Estradiol/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Timo/citologia , Timo/patologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...