Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
IUBMB Life ; 61(6): 670-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19472184

RESUMO

Appropriate retinoic acid (RA) signaling is essential in the development of the central nervous system (CNS). Previous studies have shown that RA activates p38 mitogen-activated protein kinase (MAPK) and steroid receptor coactivator (SRC)-3 in tumor cells in vitro. It is unknown whether the activation of p38 MAPK and SRC-3 is involved in RA-mediated CNS development. The current study investigated a possible role for p38 MAPK in the regulation of (SRC)-3 phosphorylation/degradation and in retinoic acid receptor (RAR)alpha signaling in mouse fetal cortical neurons treated with all-trans retinoic acid (ATRA). Results showed that ATRA treatment rapidly activated p38 MAPK, which in turn resulted in phosphorylation with subsequent degradation of SRC-3. Inhibition of p38 MAPK by SB203580 blocked the phosphorylation and degradation of SRC-3. The binding of RARalpha to retinoic acid responsive element (RARE) was rapidly increased in neurons following ATRA treatment. Inhibition of p38 MAPK significantly enhanced the RARalpha-RARE binding activity, but had no effects on ATRA-induced decrease of RARalpha. Treatment of the fetal cortical neurons with ATRA significantly increased the expression of HOXd3, a retinoid-target gene. The increase of HOXd3 expression was augmented when p38 MAPK activity was inhibited by a specific inhibitor, SB203580. Results suggest that ATRA activates the p38 MAPK signal pathway with resultant degradation of SRC-3, and that p38 MAPK is involved in the regulation of RARalpha-mediated signaling in developing neurons.


Assuntos
Histona Acetiltransferases/metabolismo , Neurogênese/fisiologia , Receptores do Ácido Retinoico/fisiologia , Transativadores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/embriologia , Proteínas de Ligação a DNA/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/efeitos dos fármacos , Coativador 3 de Receptor Nuclear , Fosforilação , Receptor alfa de Ácido Retinoico , Transdução de Sinais , Tretinoína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
2.
J Appl Toxicol ; 29(4): 364-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19058293

RESUMO

Retinoic acid can cause malformations of the developing nervous system. Smad signaling is involved in embryonic development. The current study investigated all-trans-retinoic acid (ATRA)-induced alteration of Smad expression in the developing neural tubes of mice. Pregnant mice were treated with a single dose of 50 mg/kg ATRA by oral gavage on embryonic day E7. Western immunoblotting was used to examine Smads proteins, particularly phosphorylated (p-) Smad1, total Smad1 and Smad6 in the neural tissue of the embryos on E8-E11 following treatment. Results showed that ATRA treatment significantly increased expression of both p-Smad1 and total Smad1, while Smad6 was decreased in neural tissues of ATRA-exposed embryos in utero from E8 to E11, a critical period for neural tube formation. Data suggest that disruption of Smad signaling may be involved in ATRA-induced neural tube defects.


Assuntos
Neurônios/metabolismo , Proteínas Smad/biossíntese , Tretinoína/toxicidade , Actinas/biossíntese , Animais , Western Blotting , Feminino , Expressão Gênica/efeitos dos fármacos , Camundongos , Tubo Neural/efeitos dos fármacos , Tubo Neural/crescimento & desenvolvimento , Fosforilação , Gravidez , Proteínas Smad/genética , Proteína Smad1/biossíntese , Proteína Smad1/genética , Proteína Smad6/biossíntese , Proteína Smad6/genética
3.
Toxicol Pathol ; 36(1): 30-42, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18337219

RESUMO

beta-cyclodextrin (beta-CD) and other cyclodextrins (CDs) have utility for solubilizing and stabilizing drugs; however, some are nephrotoxic when administered parenterally. A number of workers have attempted to identify, prepare, and evaluate various CD derivatives with superior inclusion complexation and maximal in vivo safety for various biomedical uses. A systematic study led to SBE-beta-CD (Captisol), a polyanionic variably substituted sulfobutyl ether of beta-CD, as a non-nephrotoxic derivative and HP-beta-CD, a modified CD developed by Janssen. SBE-beta-CD and HP-beta-CD have undergone extensive safety studies and are currently used in six products approved by the Food and Drug Administration (four for Captisol and two for HP-beta-CD). They are also in use in numerous clinical and preclinical studies. This article will focus on the issues that led to the development of these two CDs, their safety, characterization, and applications, and especially their ability to improve drug delivery. SBE-beta-CD interacts very well with neutral drugs to facilitate solubility and chemical stability, and because of its polyanionic nature, it interacts particularly well with cationic drugs. Complexes between SBE-beta-CD and HP-beta-CD and various drugs have been shown to rapidly dissociate after parenteral drug administration, to have no tissue-irritating effects after intramuscular dosing, and to result in superior oral bioavailability of poorly water-soluble drugs. The pharmacokinetics, tissue distribution, and cellular effects of some representative CDs, including SBE-beta-CD and HP-beta-CD, are reviewed. The safety profiles of CDs are discussed, with emphasis on the biological effects of some CDs on the gastrointestinal tract, kidney, and reproduction and development and the carcinogenic potential of CDs. In addition, human experience with CD derivatives, specifically SBE-beta-CD and HP-beta-CD, indicates that these two CDs are well tolerated in humans and have no adverse effects on the kidneys or other organs following either oral or intravenous administration.


Assuntos
Química Farmacêutica/métodos , Ciclodextrinas , Portadores de Fármacos , Preparações Farmacêuticas , Animais , Ciclodextrinas/química , Ciclodextrinas/farmacocinética , Ciclodextrinas/toxicidade , Cães , Interações Medicamentosas , Estabilidade de Medicamentos , Humanos , Coelhos , Ratos , Solubilidade , Testes de Toxicidade
4.
Toxicol Sci ; 94(2): 388-97, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16960033

RESUMO

Sphingolipids are important components of cell structure and cell signaling. Both external and internal stimuli can alter levels of cellular sphingolipids by regulating enzyme activities associated with sphingolipid metabolism. Fumonisin B1, mycotoxin produced by Fusarium verticillioides, is a reportedly specific inhibitor of ceramide synthase. In order to test our hypothesis whether ceramide synthase inhibition by fumonisin B1 alters other sphingolipid-metabolizing enzymes, we investigated the changes in free sphingoid bases and sphingomyelin (SM) and activities of key enzymes for their metabolism, sphingomyelinase (SMase), serine palmitoyltransferase (SPT), and sphingosine kinase (SPHK) in mouse liver. The hepatic free sphingoid bases increased significantly following five daily treatments with fumonisin B1 in mice. The activity of acidic SMase was enhanced by fumonisin B1, accompanied with a decrease in liver SM content. The expression and activities of SPT and SPHK1 in liver increased significantly following fumonisin B1 treatment. Another hepatotoxicant acetaminophen caused liver regeneration similar to fumonisin B1 but did not produce similar effects on liver sphingolipid-metabolizing enzymes, suggesting that activation of sphingolipid metabolism was not a consequence of hepatocyte regeneration. Data suggest that ceramide synthase inhibition by fumonisin B1 treatment stimulates sphingolipid-metabolizing systems to maintain a balance of cellular sphingolipids.


Assuntos
Inibidores Enzimáticos/toxicidade , Fumonisinas/toxicidade , Fígado/efeitos dos fármacos , Micotoxinas/toxicidade , Oxirredutases/antagonistas & inibidores , Esfingolipídeos/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Relação Dose-Resposta a Droga , Feminino , Injeções Subcutâneas , Isoenzimas , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Serina C-Palmitoiltransferase/biossíntese , Serina C-Palmitoiltransferase/genética , Esfingomielina Fosfodiesterase/biossíntese , Esfingomielina Fosfodiesterase/genética , Esfingomielinas/metabolismo
5.
Toxicology ; 223(3): 191-201, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16690192

RESUMO

Fumonisin B1 is a mycotoxin produced by Fusarium verticillioides, frequently associated with corn. It produces species-specific and organ-specific toxicity, including equine leukoencephalomalacia, porcine pulmonary edema, and hepatic or renal damage in most animal species. Fumonisin B1 perturbs sphingolipid metabolism by inhibiting ceramide synthase. Our previous studies in male mice indicated that fumonisin B1-induced hepatotoxicity is modulated by the localized activation of cytokines in liver macrophages and other cell types. In the current study, male athymic nude mice and their wild type counterparts (WT), the latter with or without depletion of T cells, were treated subcutaneously with fumonisin B1 at 2.25 mg/kg/day for 5 days and sampled 24 h after the last injection. Depletion of T cells in WT was achieved by a single intravenous injection of 50 microg monoclonal antibody against Thy-1.2 surface antigen of mature peripheral T lymphocytes 24 h before the first fumonisin B1 treatment. The depletion of T cells nearly abolished fumonisin B1-mediated liver toxicity as indicated by the near normal concentrations of circulating liver enzymes and by enumeration of apoptotic hepatocytes. There was no difference in the fumonisin B1-induced elevation in circulating liver enzymes between WT and nude mice. Fumonisin B1-induced mRNA expression of tumor necrosis factor alpha and interleukin-1alpha was observed in nude and WT mice but not in T cell-depleted mice. Hepatotoxic response to fumonisin B1 was unaltered in mice lacking natural killer cells. This study suggested that T cells and corresponding proinflammatory cytokines have a vital role in mediating fumonisin B1-induced hepatic toxicity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Fumonisinas/toxicidade , Isoanticorpos/uso terapêutico , Linfócitos T/efeitos dos fármacos , Animais , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Citocinas/imunologia , Isoanticorpos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Nus , Linfócitos T/imunologia
6.
Toxicon ; 46(4): 404-13, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16054185

RESUMO

Fumonisin B1 (FB1), a mycotoxin from Fusarium verticillioides, disrupts sphingolipid metabolism by inhibiting ceramide synthase leading to modulation of cytokines including tumor necrosis factor (TNF) alpha. Current study investigated the effect of interrupting TNFalpha signaling, known to be involved in FB1 hepatotoxicity. Male C57BL/6N mice were injected intravenously once with anti-TNFalpha antibodies or treated with pentoxifylline at 150 mg/kg intraperitoneally twice a day for 5 days to inhibit TNFalpha production before and during subcutaneous injection of 2.25mg FB1/kg daily for 5 days; mice were sampled one day after the last treatment. Results showed that both anti-TNFalpha antibodies and pentoxifylline did not prevent FB1 hepatotoxicity; the latter was somewhat augmented, indicated by increases in circulating alanine aminotransferase and aspartate aminotransferase, and incidence of apoptotic hepatocytes. Anti-TNFalpha antibodies did not alter FB1-induced accumulation of free sphingoid bases or expression of TNFalpha in liver following the FB1 treatment. Pentoxifylline significantly reduced accumulation of free sphinganine and expression of TNFalpha. Neither anti-TNFalpha antibodies nor pentoxifylline altered FB1-induced expression of interleukin-12, interferongamma, lymphotoxinbeta, and c-myc. Expression of c-myc, an inducer of cell death, increased after interference with TNFalpha signaling. These findings suggest a dual role of TNFalpha signaling activation in FB1 hepatotoxicity.


Assuntos
Anticorpos/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Fumonisinas/toxicidade , Micotoxinas/toxicidade , Pentoxifilina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Alanina Transaminase/sangue , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Aspartato Aminotransferases/sangue , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Primers do DNA , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Esfingosina/análogos & derivados , Esfingosina/metabolismo
7.
Toxicology ; 207(1): 137-47, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15590129

RESUMO

Fumonisin B1 (FB1) is a toxic and carcinogenic mycotoxin produced by Fusarium verticillioides found on corn worldwide. The biological effects of FB1 are attributed to sphingolipid metabolism disruption as a result of ceramide synthase inhibition. Tumor necrosis factor alpha (TNFalpha) is an important modulator of FB1 hepatotoxicity. Kupffer cells are major source of cytokine production in liver. In the present study we investigated the effects of Kupffer cell depletion by gadolinium on FB1 hepatotoxicity in female BALB/c mice. Mice were given saline or 50 mg/kg of gadolinium chloride once via the tail vein; 16 h later they were treated with subcutaneous injections of vehicle or 2.25 mg/kg/day FB1 in saline for three successive days. Gadolinium significantly attenuated FB1-induced increases in the activities of circulating alanine aminotransferase and aspartate aminotransferase and reduced the FB1-induced hepatocyte apoptosis and free sphinganine accumulation in liver. Both gadolinium and FB1 treatments individually increased the expression of selected cell signal factors; e.g., TNFalpha, TNF receptor 1, TNF-related apoptosis-inducing ligand, lymphotoxin beta, interferon gamma, and transforming growth factor beta1; gadolinium chloride did not alter FB1-induced expression of the above genes. Results indicated that Kupffer cells play a role in FB1 hepatotoxicity. Decreased FB1-induced sphinganine accumulation and increased protective TNFalpha signaling by gadolinium chloride may in part account for its ameliorating effect on FB1 liver damage.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fumonisinas/toxicidade , Gadolínio/uso terapêutico , Hepatite Animal/etiologia , Células de Kupffer/efeitos dos fármacos , Alanina Transaminase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Aspartato Aminotransferases/metabolismo , Proliferação de Células/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Citocinas/biossíntese , Feminino , Expressão Gênica/efeitos dos fármacos , Hepatite Animal/metabolismo , Hepatite Animal/patologia , Hepatite Animal/prevenção & controle , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Injeções Intravenosas , Injeções Subcutâneas , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Citocinas/biossíntese
8.
Toxicology ; 216(1): 59-71, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16159691

RESUMO

Fumonisin B1 (FB1) is a mycotoxin produced by Fusarium verticillioides, commonly present in corn and other cereals. Exposure to FB1 causes organ-specific diseases in various species, e.g., equine leukoencephalomalacia and porcine pulmonary edema; in mice the response is hepatotoxicity. We earlier reported that ceramide synthase inhibition by FB1, the initial biochemical effect of this mycotoxin, results in modulation of cytokine network in response to accumulated free sphingoid bases. In the current study we used NZB/NZW-F1 (NZBW) mice that have modified cytokine expression and develop lupus beginning at 5 months of age. The NZBW and C57BL/6J (CBL) mice (appropriate control) were given five daily subcutaneous injections of either saline or 2.25 mg FB1/kg/day and euthanized 24 h after the last treatment. Peripheral leukocyte counts were higher after exposure to FB1 in CBL but not in NZBW. FB1 treatment caused increases of plasma alanine aminotransferase and aspartate aminotransferase activity in CBL mice indicating hepatotoxicity; no elevation of circulating liver enzymes was recorded in NZBW mice. Hepatotoxic responses were confirmed by microscopic evaluation of apoptotic cells. The FB1-induced proliferation of cells observed in CBL strain was abolished in NZBW animals. The sphinganine accumulation in liver after FB1 was equal in both strains of mice. The NZBW strain lacked the FB1-induced increases in the expression of liver tumor necrosis factor alpha, interferon gamma, receptor interacting protein (RIP), and tumor necrosis factor alpha-related apoptosis-inducing ligand (TRAIL), observed in CBL. Results confirmed our hypothesis that initial altered sphingolipid metabolism caused by FB1 leads to perturbation of liver cytokine network and ultimate cellular injury; the mice deficient in cytokine signaling are refractory to FB1 hepatotoxicity.


Assuntos
Citocinas/metabolismo , Fumonisinas/toxicidade , Hepatite/imunologia , Fígado/efeitos dos fármacos , Lúpus Vulgar/imunologia , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/complicações , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Citocinas/imunologia , Hepatite/complicações , Hepatite/metabolismo , Hepatite Animal/complicações , Hepatite Animal/imunologia , Hepatite Animal/metabolismo , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Fígado/imunologia , Fígado/metabolismo , Lúpus Vulgar/complicações , Lúpus Vulgar/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo
9.
Food Chem Toxicol ; 43(9): 1389-97, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15913876

RESUMO

Fumonisin B(1) (FB(1)), a mycotoxin produced by Fusarium verticillioides, causes equine leukoencephalomalacia and hepatotoxicity. We studied the modulation of FB(1) toxicity in brain and liver of female BALB/c mice after endotoxin administration to compromise the blood-brain barrier (BBB) integrity. Mice were injected intraperitoneally with saline or 3 mg/kg of lipopolysaccharide (LPS) followed 2 h later by either a single or three daily subcutaneous doses of 2.25 mg/kg of FB(1). After 4h of a single FB(1) injection the inhibition of sphingolipid biosynthesis occurred in liver. Circulating alanine aminotransferase increased by LPS alone at this time. In brain LPS triggered inflammation increasing the expression of tumor necrosis factor (TNF) alpha, interferon (IFN) gamma, interleukin (IL)-1beta, IL-6, and IL-12; no effect of FB(1) was observed. In liver LPS+FB(1) attenuated the expression TNFalpha and IFNgamma compared to LPS alone. One day after the 3-day FB(1) treatment the biosynthesis of sphingolipids was markedly reduced in brain and liver and it was further inhibited when LPS was given before FB(1). FB(1) induced hepatotoxicity, as measured by circulating liver enzymes, was reduced after the combined treatment with LPS+FB(1) compared to FB(1) alone. FB(1) decreased the LPS-induced brain expression of IFNgamma and IL-1beta, whereas the expression of IL-6 and IL-12 was augmented. In liver FB(1) also reduced the expression of IL-1beta and IFNgamma compared to LPS alone. Results indicated that endotoxemia concurrent with FB(1) intoxication facilitated the permeability of fumonisin in brain indicated by increased accumulation of sphinganine and endotoxin modified the effects of FB(1) in both brain and liver.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Endotoxinas/toxicidade , Fumonisinas/farmacologia , Fígado/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Citocinas/genética , Primers do DNA/farmacologia , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Fígado/enzimologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão/efeitos dos fármacos , RNA/biossíntese , RNA/genética , Esfingolipídeos/metabolismo
10.
Food Chem Toxicol ; 43(6): 969-79, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15811577

RESUMO

Fumonisin B(1) (FB(1)), a mycotoxin produced by Fusarium verticillioides present on corn and corn-based products, causes species- and organ-specific diseases. The hepatotoxic effects of FB(1) in mice have been closely correlated with the accumulation of free sphinganine, a marker for ceramide synthase inhibition, and reduced biosynthesis of more complex sphingolipids. It has been shown that FB(1) modulates expression of many cell signaling factors. In the current study we used myriocin, a specific inhibitor of serine palmitoyltransferase, to investigate the role of free sphinganine accumulation in FB(1)-induced hepatotoxicity and increased expression of selected signaling genes in BALB/c mice. The mice were pretreated daily with intraperitoneal injection of 1.0 mg/kg myriocin 30 min before subcutaneous injections of 2.25 mg/kg of FB(1) for 3 days. Results showed that myriocin alone was not hepatotoxic and the combination of myriocin plus FB(1) completely prevented the FB(1)-induced elevation of hepatic free sphinganine and prevented the FB(1)-induced induction of selected cell signaling genes, suggesting that accumulation of free sphinganine and/or its metabolites contribute to the FB(1)-modulation of the cell signaling factors. However, the combination of myriocin and FB(1) did not prevent FB(1)-increased concentration of plasma alanine aminotransferase and only slightly attenuated aspartate aminotransferase; it did not affect the FB(1)-induced hepatocyte apoptosis or increased cell proliferation. A longer combined treatment of myriocin and FB(1) was highly toxic. The hepatotoxic effects in mice seen in this study are most likely due to a combination of factors including accumulation of free sphinganine, depletion of more complex sphingolipids and sphingomyelin, or other unknown mechanisms.


Assuntos
Aciltransferases/antagonistas & inibidores , Antifúngicos/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Monoinsaturados/uso terapêutico , Fumonisinas/antagonistas & inibidores , Esfingolipídeos/biossíntese , Esfingosina/análogos & derivados , Aciltransferases/metabolismo , Aciltransferases/fisiologia , Animais , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Feminino , Fumonisinas/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Serina C-Palmitoiltransferase , Esfingosina/metabolismo
11.
J Interferon Cytokine Res ; 23(1): 13-23, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12639295

RESUMO

Fumonisin B(1) (FB(1)) produces species-specific and organ-specific toxicity, including equine leukoencephalomalacia, porcine pulmonary edema, and hepatic or renal damage in other animals. FB(1) causes inhibition of ceramide synthase, leading to accumulation of free sphingoid bases. We previously reported that such cytokines as tumor necrosis factor-alpha (TNF-alpha) modify FB(1)-induced hepatic apoptosis in male mice. FB(1) also caused induction of interferon-gamma (IFN-gamma) in mouse liver, and, therefore, it was worthwhile to determine the role IFN-gamma plays in FB(1) toxicity. In the current study, male IFN-gamma-knockout (GKO) mice and their wild-type (WT) counterparts, C57BL/6J, were treated subcutaneously (s.c.) with 2.25 mg/kg/day of FB(1) for 5 days and sampled 1 day after the last injection. The levels of circulating liver enzymes were increased in WT animals but considerably less in GKO mice. Reduced hepatotoxicity in GKO mice was evident by histologic evaluation and enumeration of apoptotic cells. The induction of TNF-alpha and interleukin-12 (IL-12) p40 by FB(1) in liver was less in GKO mice compared with WT animals. The GKO mice also had a reduced accumulation of liver sphinganine than did WT mice after FB(1) treatment. Results suggested the implication of IFN-gamma in FB(1)-induced hepatotoxicity, which can be explained by a lack of TNF-alpha and IL-12 amplification in the liver of the GKO mice. In addition, the GKO mice had altered expression of various apoptotic and antiapoptotic factors in liver. These changes were accompanied by a greater number of proliferating cells in the liver of GKO mice after FB(1) treatment, which may also contribute to the reduced hepatotoxicity of FB(1) in GKO mice. Whereas the GKO mice show reduced sensitivity to FB(1) and FB(1) treatment elevates IFN-gamma expression, decreased hepatotoxicity to FB(1) could result from alterations in sphingolipid metabolism in the GKO strain.


Assuntos
Citocinas/metabolismo , Fumonisinas/toxicidade , Mediadores da Inflamação/metabolismo , Interferon gama/deficiência , Fígado/efeitos dos fármacos , Fígado/imunologia , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Interferon gama/genética , Interferon gama/fisiologia , Interleucina-12/biossíntese , Fígado/patologia , Fígado/fisiopatologia , Regeneração Hepática/efeitos dos fármacos , Regeneração Hepática/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
12.
Toxicol Sci ; 80(2): 335-42, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15103051

RESUMO

Fumonisin B(1) (FB(1)) is a mycotoxin produced by Fusarium verticillioides found on corn and corn-based foods. It causes equine leukoencephalomalacia, porcine pulmonary edema, and liver and kidney damage in most animal species. Fumonisin B(1) perturbs sphingolipid metabolism by inhibiting ceramide synthase activity, leading to the production of cell signaling factors including tumor necrosis factor alpha (TNF-alpha). The signal pathways of TNF-alpha are important factors in the pathogenesis of FB(1) hepatotoxicity. In the present study, female BALB/c mice were treated daily with 750 mg/kg silymarin by gavage and 2.25 mg/kg FB(1) subcutaneously for 3 days. Then, 1 day after the last FB(1) injection, the mice were euthanized and blood and tissues were sampled for analyses. Silymarin significantly diminished FB(1)-induced elevation of plasma alanine aminotransferase and aspartate aminotransferase activities and the number of apoptotic hepatocytes, while it augmented hepatocyte proliferation indicated by an increase in proliferating cells. Silymarin dramatically potentiated FB(1)-induced accumulation of free sphinganine and sphingosine in both liver and kidney. Silymarin itself slightly increased expression of hepatic TNF-alpha; however, it prevented the FB(1)-induced increases in TNF-alpha, TNF receptor 1, TNF receptor-associated apoptosis-inducing ligand, lymphotoxin beta, and interferon gamma. The induction of transforming growth factor beta1 expression in liver following FB(1) treatment was not affected by silymarin. These findings suggest that silymarin protected against FB(1) liver damage by inhibiting biological functions of free sphingoid bases and increasing cellular regeneration.


Assuntos
Carcinógenos Ambientais/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Fumonisinas/toxicidade , Silimarina/farmacologia , Esfingosina/metabolismo , Aciltransferases/metabolismo , Alanina Transaminase/sangue , Animais , Apoptose/efeitos dos fármacos , Aspartato Aminotransferases/sangue , Proliferação de Células/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/biossíntese , Feminino , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Rim/enzimologia , Fígado/enzimologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ensaios de Proteção de Nucleases , Antígeno Nuclear de Célula em Proliferação , Serina C-Palmitoiltransferase
13.
Toxicology ; 203(1-3): 239-51, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15363598

RESUMO

Fumonisin B1, a common mycotoxin produced by Fusarium verticillioides found in corn, causes several fatal animal diseases. Liver and kidney are target organs of fumonisin B1 in laboratory animals, but primary rodent hepatocytes and liver slices were resistant to fumonisin B1-induced cytotoxic effects. We have shown that fumonisin B1 induces expression of tumor necrosis factor (TNF)alpha, interferon (IFN)gamma, and interleukine (IL) 12, in mouse liver. In various models of acute liver injury, a positive amplification loop involving TNFalpha, IFNgamma, and IL-12 has been implied that involves Kupffer cells (macrophages), hepatic lymphocytes and non-parenchymatous liver epithelial cells (NPECs). In the current study, cellular interactions in fumonisin B1-induced toxicity were investigated, using co-cultures of murine macrophages (J774A.1) and NPECs (NMuLi). Treatment of the co-cultures with fumonisin B1-produced cytotoxicity, whereas either J774A.1 or NMuLi cultures alone showed no response to the mycotoxin. Accumulation of sphinganine occurred to the similar extent in individual cultures as well as co-cultures. Expression of TNFalpha and IL-12 was increased in co-cultures but not in individual cultures. Transfer of conditioned medium from fumonisin B1-treated J774A.1 cells to NMuLi cultures produced an increase in IFNgamma expression in NMuLi cells. Results indicated that macrophages and liver epithelial cells interact in response to fumonisin B1 and potentiate the cytokines expression, which may have implications in making hepatocytes responsive to cytotoxicity of fumonisin B1.


Assuntos
Carcinógenos Ambientais/toxicidade , Citocinas/fisiologia , Células Epiteliais/fisiologia , Fumonisinas/toxicidade , Fígado/citologia , Macrófagos/fisiologia , Receptor Cross-Talk/fisiologia , Animais , Apoptose/efeitos dos fármacos , Morte Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultura , Células Epiteliais/efeitos dos fármacos , Imuno-Histoquímica , Inflamação/fisiopatologia , Interferon gama/antagonistas & inibidores , L-Lactato Desidrogenase/metabolismo , Fígado/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/farmacologia
14.
Toxicology ; 201(1-3): 67-75, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15297021

RESUMO

Myriocin is a naturally occurring fungal metabolite possessing potent immunosuppressive properties. The biochemical mechanism of action of this compound is inhibition of serine palmitoyltransferase (SPT), the key rate limiting enzyme in sphingolipid biosynthesis, intermediates of which are important mediators of immune signaling. Previous studies have shown that myriocin strongly suppressed immune function with T-lymphocyte functions being most sensitive. To further our understanding of the mechanisms of this effect, we investigated the impact of subacute treatment with myriocin on lymphocyte populations in the thymus and spleen of male BALB/c mice following intraperitoneal injection of myriocin at 0, 0.1, 0.3, and 1.0 mg/kg daily for 5 consecutive days. Cellular analysis of the thymus demonstrated that total cellularity was dose-dependently reduced and the reduction was significant in mice treated with 1.0 mg/kg myriocin. Phenotyping showed that CD4+ and CD4+/CD8+ double positive lymphocyte populations were sensitive to myriocin. No change in total cellularity of the spleen was noted but there was a significant reduction in the CD4+ lymphocyte population in mice treated with 1.0 mg/kg myriocin. There was a strong positive correlation between total CD4+ lymphocytes in the thymus and those in the spleen. Analysis of sphingolipid levels showed a dose-dependent reduction of sphinganine in the thymus, which were positively correlated with all reductions in lymphocyte populations. These results suggest that the immunosuppressive properties of myriocin may be due to diminished T-lymphocyte populations likely related to inhibition of SPT and disruption of sphingolipid homeostasis.


Assuntos
Ácidos Graxos Monoinsaturados/toxicidade , Homeostase/efeitos dos fármacos , Imunossupressores/toxicidade , Esfingolipídeos/biossíntese , Baço/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Timo/efeitos dos fármacos , Aciltransferases/metabolismo , Animais , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Serina C-Palmitoiltransferase , Esfingolipídeos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
Toxicology ; 180(3): 221-32, 2002 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-12393292

RESUMO

Tumor necrosis factor alpha (TNFalpha) is involved in fumonisin-induced hepatotoxic effects in mice. The hepatic response to fumonisin B(1) (FB(1)) was reduced in transgenic animals lacking either of the two TNFalpha receptors. In the present study, we hypothesized that the effect of a similar fumonisin treatment in animals lacking either TNFalpha or both TNFalpha receptors would be considerably less than their wild type (WT) counterparts. The FB(1)-induced increase in circulating liver enzymes was enhanced by deletion of TNFalpha or unchanged in mice lacking both TNFalpha receptors. These findings corresponded with the degree of toxicity as established by microscopic examination of liver. FB(1) induced the expression of TNFalpha in the liver of all strains, except the animals with a deleted TNFalpha gene. The FB(1)-mediated increases in liver sphingosine or sphinganine paralleled the hepatotoxic responses. It is apparent that the presence of TNFalpha is not necessary for FB(1)-induced hepatotoxicity in mice and a lack of the function of this cytokine may aggravate the hepatotoxic responses to fumonisins, perhaps by preventing repair mechanisms or by expression of other signaling molecules. These observations were in accordance with our previous finding where over-expression of TNFalpha also protected against FB(1)-mediated hepatotoxicity, and with the reported beneficial functions of low-level TNFalpha in tissue regeneration.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fumonisinas/toxicidade , Micotoxinas/toxicidade , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antígenos CD/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Primers do DNA , Contagem de Eritrócitos , Contagem de Leucócitos , Fígado/enzimologia , Camundongos , Camundongos Transgênicos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Transdução de Sinais/fisiologia , Esfingolipídeos/metabolismo
16.
Life Sci ; 71(17): 2015-23, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12175895

RESUMO

Fumonisin B(1) (FB(1)), produced by Fusarium verticillioides, is a common contaminant in foods and feeds. Increase in tissue free sphingoid bases resulting from the inhibition of ceramide synthase is a biomarker of fumonisin exposure. Tumor necrosis factor alpha (TNFalpha) is induced in liver in response to FB(1) treatment. This study determined whether fumonisin B(1) caused increases in free sphingoid bases and altered the expression of TNFalpha in heart and lung, organs that are not targets of FB(1) toxicity, of male and female mice treated with 5-daily subcutaneous injection of 2.25 mg/kg FB(1). A significant increase in free sphingoid bases was observed in both heart and lung of FB(1)-exposed mice. The magnitude of increases in free sphingoid bases in both organs of female mice was much higher than that in males. The expression of TNFalpha was increased by FB(1) treatment in the lung of male mice and in the heart of female mice, whereas the expression of interferon gamma was unaltered. Results suggest that both sphingolipid accumulation and TNFalpha induction are observed in the tissues of mice that are not associated with FB(1) toxicity.


Assuntos
Ácidos Carboxílicos/toxicidade , Carcinógenos Ambientais/toxicidade , Fumonisinas , Coração/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ácidos Carboxílicos/administração & dosagem , Carcinógenos Ambientais/administração & dosagem , Feminino , Injeções Subcutâneas , Interferon gama/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Fator de Necrose Tumoral alfa/genética
17.
Chem Biol Interact ; 146(2): 131-45, 2003 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-14597127

RESUMO

Fumonisin B1 (FB1), the most potent of the fumonisin mycotoxins, is a carcinogen and causes a wide range of species-specific toxicoses. FB1 modulates the activity of protein kinase C (PKC), a family of phospholipid-dependent serine/threonine kinases that play important role in modulating a variety of biologic responses ranging from regulation of cell growth to cell death. Although it has been demonstrated that FB1 induces apoptosis in many cell lines, the precise mechanism of apoptosis is not fully understood. In this study, we investigated the membrane localization of various PKC isoforms, PKC enzyme activity, and its downstream targets, namely nuclear factor-kappa B (NF-kappaB), tumor necrosis factor alpha (TNFalpha), and caspase 3, in porcine renal epithelial (LLC-PK1) cells. FB1 repressed cytosol to membrane translocation of PKC-alpha, -delta, -epsilon, and -zeta isoforms over 24-72 h. The FB1-induced membrane PKC repression was corroborated by a concentration-dependent decrease in total PKC activity. Exposure of cells to phorbol 12-myristate 13-acetate (PMA) for this duration also resulted in repressed PKC membrane localization and activity comparable to FB1. Exposure of cells to FB1 (10 microM) was associated with inhibition of cytosol to nuclear translocation of NF-kappaB and NF-kappaB-DNA binding at 72 h. The expression of TNFalpha was significantly inhibited at 24 and 48 h in response to 1 and 10 microM FB1. Increased caspase 3 activity was observed in LLC-PK1 cells exposed to > or =1 microM FB1 at 48 h. PMA also increased the caspase 3 activity at 24 and 48 h. Results suggest that FB1-induced apoptosis involves the activation of caspase 3, which is associated with the repression of PKC and possibly its down-stream effectors, NF-kappaB and TNFalpha.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Fumonisinas/toxicidade , NF-kappa B/metabolismo , Proteína Quinase C/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Animais , Carcinógenos Ambientais/toxicidade , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Isoenzimas , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Micotoxinas/toxicidade , NF-kappa B/genética , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , Suínos , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/genética
18.
Chem Biol Interact ; 151(1): 33-42, 2004 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-15607760

RESUMO

Fumonisin B1 induces cytotoxicity in sensitive cells by inhibiting ceramide synthase due to its structural similarity to the long-chain backbones of sphingolipids. The resulting accumulation of sphingoid bases has been established as a mechanism for fumonisin B1 cytotoxicity. We found that despite the accumulation of sphinganine, human embryonic kidney (HEK-293) cells are resistant to fumonisin B1 toxicity; 25 microM fumonisin B1 exposure for 48 h did not increase apoptosis in these cells, while it did so in sensitive porcine kidney epithelial (LLC-PK1) cells. In this study, DL-threo-dihydrosphingosine, the sphingosine kinase inhibitor (SKI), considerably increased the sensitivity of HEK-293 cells to fumonisin B1. Treatment of these cells with 25 microM fumonisin B1 and 2.5 microM SKI increased apoptosis. Sphingoid bases, sphinganine or sphingosine, added to cell cultures induced apoptosis by themselves and their effects were potentiated by SKI or fumonisin B1. Addition of physiological amounts of sphingosine-1-phosphate prevented the toxic effects induced by SKI inhibition and fumonisin B1. Results indicated that HEK-293 cells are resistant to fumonisin B1 due to rapid formation of sphingosine-1-phosphate that imparts survival properties. Taken together, these findings suggest that sphingoid base metabolism by sphingosine kinase may be a critical event in rendering the HEK-293 cells relatively resistant to fumonisin B1-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos Ambientais/toxicidade , Fumonisinas/farmacologia , Rim/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Humanos , Rim/citologia , Rim/embriologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Suínos
19.
Chem Biol Interact ; 145(3): 297-309, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12732456

RESUMO

Previous studies have shown that tumor necrosis factor alpha (TNFalpha) is involved in the pathogenic events following exposure to fumonisin B(1) (FB(1)), a potent inhibitor of ceramide synthase and sphingolipid biosynthesis. The intimate role of sphingolipid mediators in TNFalpha signaling and cellular death suggests that FB(1) may alter the sensitivity of cells to TNFalpha-induced apoptosis. We tested the hypothesis that FB(1) treatment will increase the sensitivity of porcine renal epithelial cells to TNFalpha. Porcine renal epithelial cells (LLC-PK(1)) were treated with FB(1) for 48 h prior to treatment with TNFalpha. A dose-dependent increase in TNFalpha-induced apoptosis was observed in cells pretreated with FB(1). Cells treated with FB(1) showed increased DNA fragmentation and terminal uridine nucleotide end labeling in response to TNFalpha treatment. FB(1) increased DNA synthesis and resulted in cell cycle arrest in the G(2)/M phase of the cell cycle. Flow cytometric analysis of the cell cycle indicated that TNFalpha predominantly killed cells in the G(2)/M phase. The activation of JNK, a mitogen-activated protein kinase (MAPK), was increased following 48 h exposure to FB(1). Phosphorylation of p38 and ERK remained unchanged following treatment with FB(1). FB(1) also increased free sphingoid base levels under identical treatment conditions. Results suggest that FB(1) increased free sphingoid base levels and the population of cells in the G(2)/M phase. This population was shown to be most susceptible to TNFalpha-induced apoptosis. Phosphorylation of pro-apoptotic JNK may play an important role in these effects.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fumonisinas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Células LLC-PK1/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Marcação In Situ das Extremidades Cortadas , Interfase/efeitos dos fármacos , Células LLC-PK1/patologia , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/biossíntese , Suínos
20.
Toxicol Lett ; 147(1): 87-94, 2004 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-14700532

RESUMO

Myriocin is a specific serine palmitoyltransferase (SPT) inhibitor whose effect on the brain is unknown. Brain amine metabolism and sphingolipid biosynthesis were studied in mice treated intraperitoneally with 0, 0.1, 0.3 or 1 mg/kg per day of myriocin for 5 days. Regional concentrations of dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 5-hydroxytryptamine (5-HT, serotonin), 5-hydroxyindoleacetic acid (5-HIAA) and norepinephrine (NE), were determined. Sphinganine (Sa) and sphingosine (So) concentrations and SPT activity in brain and liver were used to evaluate the impact of myriocin on sphingolipid biosynthesis. Myriocin treatment increased DA in striatum and hippocampus and reduced it in cortex. NE concentration decreased in cerebellum and 5-HT levels were reduced in cortex and in medulla oblongata. Changes in ratios for DOPAC/DA and HVA/DA were observed in hippocampus, cortex and midbrain. Brain Sa, So and SPT activity remained unchanged, whereas Sa and SPT activity decreased in liver. Results showed that myriocin may alter the levels and metabolism of brain amines and this effect is not related with inhibition of sphingolipid biosynthesis in the nervous system.


Assuntos
Aciltransferases/antagonistas & inibidores , Monoaminas Biogênicas/metabolismo , Encéfalo/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Ácidos Graxos Monoinsaturados/toxicidade , Esfingosina/análogos & derivados , Esfingosina/biossíntese , Aciltransferases/metabolismo , Animais , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Ácidos Graxos Monoinsaturados/administração & dosagem , Feminino , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos BALB C , Serina C-Palmitoiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA