Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Clin Lab Sci ; 49(4): 423-424, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31471330
2.
Hepatology ; 46(6): 1927-834, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17854053

RESUMO

UNLABELLED: Porphyria cutanea tarda is a liver disease characterized by elevated hepatic iron and excessive production of uroporphyrin (URO). Phlebotomy is an effective treatment that probably acts by reducing hepatic iron. Here we used Hfe(-/-) mice to compare the effects on hepatic URO accumulation of two different methods of hepatic iron depletion: iron chelation using deferiprone (L1) versus iron-deficient diets. Hfe(-/-) mice in a 129S6/SvEvTac background were fed 5-aminolevulinic acid (ALA), which results in hepatic URO accumulation, and increasing doses of L1 in the drinking water. Hepatic URO accumulation was completely prevented at low L1 doses, which partially depleted hepatic nonheme iron. By histological assessment, the decrease in hepatic URO accumulation was associated with greater depletion of iron from hepatocytes than from Kupffer cells. The L1 treatment had no effect on levels of hepatic cytochrome P4501A2 (CYP1A2). L1 also effectively decreased hepatic URO accumulation in C57BL/6 Hfe(-/-) mice treated with ALA and a CYP1A2 inducer. ALA-treated mice maintained on defined iron-deficient diets, rather than chow diets, did not develop uroporphyria, even when the animals were iron-supplemented either directly in the diet or by iron dextran injection. CONCLUSION: The results suggest that dietary factors other than iron are involved in the development of uroporphyria and that a modest depletion of hepatocyte iron by L1 is sufficient to prevent URO accumulation.


Assuntos
Quelantes de Ferro/uso terapêutico , Deficiências de Ferro , Porfiria Cutânea Tardia/dietoterapia , Porfiria Cutânea Tardia/tratamento farmacológico , Piridonas/uso terapêutico , Animais , Deferiprona , Modelos Animais de Doenças , Fígado/química , Masculino , Camundongos , Uroporfirinas/análise
3.
Drug Metab Dispos ; 35(7): 1223-31, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17392391

RESUMO

CYP2E1 is widely accepted as the sole form of cytochrome P450 responsible for alcohol-mediated increases in acetaminophen (APAP) hepatotoxicity. However, we previously found that alcohol [ethanol and isopentanol (EIP)] causes increases in APAP hepatotoxicity in Cyp2e1(-/-) mice, indicating that CYP2E1 is not essential. Here, using wild-type and Cyp2e1(-/-) mice, we investigated the relative roles of CYP2E1 and CYP3A in EIP-mediated increases in APAP hepatotoxicity. We found that EIP-mediated increases in APAP hepatotoxicity occurred at lower APAP doses in wild-type mice (300 mg/kg) than in Cyp2e1(-/-) mice (600 mg/kg). Although this result suggests that CYP2E1 has a role in the different susceptibilities of these mouse lines, our findings that EIP-mediated increases in CYP3A activities were greater in wild-type mice compared with Cyp2e1(-/-) mice raises the possibility that differential increases in CYP3A may also contribute to the greater APAP sensitivity in EIP-pretreated wild-type mice. At the time of APAP administration, which followed an 11 h withdrawal from the alcohols, alcohol-induced levels of CYP3A were sustained in both mouse lines, whereas CYP2E1 was decreased to constitutive levels in wild-type mice. The CYP3A inhibitor triacetyloleandomycin (TAO) decreased APAP hepatotoxicity in EIP-pretreated wild-type and Cyp2e1(-/-) mice. TAO treatment in vivo resulted in inhibition of microsomal CYP3A-catalyzed activity, measured in vitro, with no inhibition of CYP1A2 and CYP2E1 activities. In conclusion, these findings suggest that both CYP3A and CYP2E1 contribute to APAP hepatotoxicity in alcohol-treated mice.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Citocromo P-450 CYP2E1/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Etanol/toxicidade , Fígado/efeitos dos fármacos , Fígado/enzimologia , Pentanóis/toxicidade , Acetaminofen , Alanina Transaminase/sangue , Animais , Benzoquinonas/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2E1/deficiência , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/biossíntese , Modelos Animais de Doenças , Sinergismo Farmacológico , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glucuronídeos/metabolismo , Glutationa/metabolismo , Hidroxilação , Iminas/metabolismo , Fígado/patologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Masculino , Camundongos , Camundongos Knockout , Índice de Gravidade de Doença , Testosterona/metabolismo , Troleandomicina/farmacologia
4.
Hepatology ; 45(1): 187-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17187429

RESUMO

UNLABELLED: Excess hepatic iron is known to enhance both porphyria cutanea tarda (PCT) and experimental uroporphyria. Since previous studies have suggested a role for ascorbate (AA) in suppressing uroporphyria in AA-requiring rats (in the absence of excess iron), the present study investigated whether AA could suppress uroporphyria produced by excess hepatic iron. Hepatic URO accumulation was produced in AA-requiring Gulo(-/-) mice by treatment with 3,3',4,4',5-pentachlorbiphenyl, an inducer of CYP1A2, and 5-aminolevulinic acid. Mice were administered either sufficient AA (1000 ppm) in the drinking water to maintain near normal hepatic AA levels or a lower intake (75 ppm) that resulted in 70 % lower hepatic AA levels. The higher AA intake suppressed hepatic URO accumulation in the absence of administered iron, but not when iron dextran (300-500 mg Fe/kg) was administered. This effect of iron was not due to hepatic AA depletion since hepatic AA content was not decreased. The effect of iron to prevent AA suppression of hepatic URO accumulation was not observed until a high hepatic iron threshold was exceeded. At both low and high AA intakes, hepatic malondialdehyde (MDA), an indicator of oxidative stress, was increased three-fold by high doses of iron dextran. MDA was considerably increased even at low iron dextran doses, but without any increase in URO accumulation. The level of hepatic CYP1A2 was unaffected by either AA intake. CONCLUSION: In this mouse model of PCT, AA suppresses hepatic URO accumulation at low, but not high hepatic iron levels. These results may have implications for the management of PCT.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Ferro/farmacologia , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/metabolismo , Ácido Aminolevulínico , Animais , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ferro/metabolismo , Complexo Ferro-Dextran/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Bifenilos Policlorados , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/tratamento farmacológico
5.
Fetal Pediatr Pathol ; 25(4): 211-31, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17162528

RESUMO

Even as a rare multiple congenital anomalies/mental retardation syndrome, the C-syndrome (CS, or Opitz C-trigonoecephaly syndrome) is, at long last, beginning to attract attention because of its developmental and causal complexity. Also, the possibility that the apparently balanced translocation recently described in an affected Japanese boy may soon provide a molecular/causal insight into this disorder. The manifestations recorded in the previously published patients, those autopsied within recent years, and the unpublished instances in our files suggest that the CS is a heterogeneous genetic disorder, predominantly sporadic but with sufficient familial cases (at times with consanguinity) to allow postulation of an entity due to autosomal dominant mutations with a high rate of germinal mosaicism, or due to both autosomal dominant mutations and an autosomal recessive genocopy. In any event, elucidation of cause and pathogenesis of CS will, in due time, shed light on its developmental pleiotropy, rarity in liveborn infants, prevalence in stillborn fetuses, recurrence risk in humans, and occurrence in other animals (e.g., mice) to further understanding of pathogenesis.


Assuntos
Anormalidades Múltiplas/mortalidade , Anormalidades Múltiplas/patologia , Osso Frontal/anormalidades , Deficiência Intelectual/mortalidade , Deficiência Intelectual/patologia , Anormalidades Múltiplas/genética , Animais , Transtornos Cromossômicos/genética , Modelos Animais de Doenças , Evolução Fatal , Feminino , Genes Dominantes , Humanos , Recém-Nascido , Deficiência Intelectual/genética , Masculino , Camundongos , Mutação , Síndrome
6.
Am J Physiol Gastrointest Liver Physiol ; 290(6): G1269-79, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16439473

RESUMO

The objective of this study was to determine whether Toll-like receptor 4 (TLR4) has a role in alcohol-mediated acetaminophen (APAP) hepatotoxicity. TLR4 is involved in the inflammatory response to endotoxin. Others have found that ethanol-mediated liver disease is decreased in C3H/HeJ mice, which have a mutated TLR4 resulting in a decreased response to endotoxin compared with endotoxin-responsive mice. In the present study, short-term (1 wk) pretreatment with ethanol plus isopentanol, the predominant alcohols in alcoholic beverages, caused no histologically observed liver damage in either C3H/HeJ mice or endotoxin-responsive C3H/HeN mice, despite an increase in nitrotyrosine levels in the livers of C3H/HeN mice. In C3H/HeN mice pretreated with the alcohols, subsequent exposure to APAP caused a transient decrease in liver nitrotyrosine formation, possibly due to competitive interaction of peroxynitrite with APAP producing 3-nitroacetaminophen. Treatment with APAP alone resulted in steatosis in addition to congestion and necrosis in both C3H/HeN and C3H/HeJ mice, but the effects were more severe in endotoxin-responsive C3H/HeN mice. In alcohol-pretreated endotoxin-responsive C3H/HeN mice, subsequent exposure to APAP resulted in further increases in liver damage, including severe steatosis, associated with elevated plasma levels of TNF-alpha. In contrast, alcohol pretreatment of C3H/HeJ mice caused little to no increase in APAP hepatotoxicity and no increase in plasma TNF-alpha. Portal blood endotoxin levels were very low and were not detectably elevated by any of the treatments. In conclusion, this study implicates a role of TLR4 in APAP-mediated hepatotoxicity.


Assuntos
Acetaminofen/efeitos adversos , Etanol/efeitos adversos , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Receptor 4 Toll-Like/metabolismo , Analgésicos não Narcóticos/efeitos adversos , Animais , Sinergismo Farmacológico , Fígado Gorduroso/patologia , Feminino , Fígado/patologia , Camundongos
7.
Drug Metab Dispos ; 33(12): 1827-36, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16141365

RESUMO

The pregnane X receptor (PXR) is a transcriptional regulator of xenobiotic metabolizing enzymes, including cytochrome P450 3A (CYP3A), and transporters. Pretreatment of mice and rats with inducers of CYP3A increases acetaminophen (APAP) hepatotoxicity. In untreated mice, the amount of hepatic CYP3A11 mRNA is 4-fold greater in PXR(-/-) mice compared to wild-type mice (Guo et al., 2003), a finding anticipated to increase APAP hepatotoxicity in PXR(-/-) mice. We investigated APAP hepatotoxicity in wild-type and PXR(-/-) mice in a C57BL/6 background, with APAP administered by gavage. Despite a 2.5-fold higher level of total hepatic CYP3A protein and a 3.6-fold higher level of CYP3A activity compared to wild-type mice, PXR(-/-) mice were less sensitive to APAP hepatotoxicity. Hepatic levels of CYP2E1 were identical in the two mouse lines, but hepatic CYP1A2 levels were 3-fold greater in wild-type mice compared to PXR(-/-) mice. Caffeine, an inhibitor of CYP1A2 activity and an enhancer of CYP3A activity, decreased APAP hepatotoxicity in wild-type mice. APAP uptake was 1.5-fold greater in wild-type mice compared to PXR(-/-) mice. No significant differences in the formation of APAP glucuronide and sulfate-conjugated metabolites were observed between wild-type and PXR(-/-) mice. Glutathione levels were similar in the two mouse lines and were transiently decreased to similar amounts after APAP administration. Our finding that APAP hepatotoxicity was decreased in PXR(-/-) mice indicates that PXR is an important modulator of APAP hepatotoxicity, through positive modulation of constitutive CYP1A2 expression and possibly through increased APAP absorption.


Assuntos
Acetaminofen/toxicidade , Fígado/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Esteroides/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Acetaminofen/metabolismo , Animais , Benzoquinonas/metabolismo , Transporte Biológico , Cafeína/farmacologia , Citocromo P-450 CYP1A2/análise , Citocromo P-450 CYP2E1/análise , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/análise , Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Iminas/metabolismo , Absorção Intestinal , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Pregnano X , Transcrição Gênica/efeitos dos fármacos
9.
J Biochem Mol Toxicol ; 19(6): 396-405, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16421895

RESUMO

Prodrugs of L-selenocysteine have potential utility in cancer chemoprevention. This study reports the efficacy of three selenazolidine-4(R)-carboxylic acids, (2-unsubstituted, 2-oxo, and 2-methyl derivatives; SCA, OSCA, and MSCA, respectively) against tobacco-related lung tumorigenesis in a mouse model. Seven days after initiation of an AIN-76A diet supplemented with sodium selenite (5 ppm Se), L-selenomethionine (3.75 ppm Se), Se-methyl-L-selenocysteine (3 ppm Se), L-selenocystine (15 ppm Se), SCA (15 ppm Se), OSCA (15 ppm Se), or MSCA (15 ppm Se), mice received 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK; 10 micromol, i.p.). After an additional 16 weeks on the diets, two compounds, OSCA and selenocystine, significantly reduced lung adenoma multiplicity from 7.2 tumors per mouse in the NNK group to 4.5 and 4.6 tumors per mouse, respectively. Neither selenium concentration nor glutathione peroxidase activity in either RBCs or liver served as surrogate indicators of tumor reduction. Hepatic selenium levels were significantly elevated by all selenium-containing compounds except Se-methyl-L-selenocysteine and SCA; RBC selenium levels by all except sodium selenite and MSCA. With the exception of L-selenomethionine, RBC glutathione peroxidase activity was increased along with the elevated selenium levels. Hepatic glutathione peroxidase activity was elevated by all Se-compounds except SCA. The two compounds showing significant tumor reduction (OSCA and selenocystine) were the only two compounds that showed ubiquity of changes, elevating both selenium levels and GPx activity in both liver and RBC.


Assuntos
Carcinógenos/toxicidade , Neoplasias Pulmonares/prevenção & controle , Nicotiana/química , Nitrosaminas/toxicidade , Pró-Fármacos/administração & dosagem , Selenocisteína/administração & dosagem , Animais , Peso Corporal , Feminino , Glutationa Peroxidase/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Neoplasias Pulmonares/induzido quimicamente , Camundongos , Tamanho do Órgão , Selênio/sangue , Selênio/metabolismo
10.
Hepatology ; 40(4): 942-50, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15382179

RESUMO

Two major risk factors for porphyria cutanea tarda (PCT) are alcohol consumption and homozygosity for the C282Y mutation in the hereditary hemochromatosis gene (HFE). We recently described an animal model for alcohol-induced uroporphyria, using Hfe(-/-) mice. In the present study we show that this effect is dependent on genetic background and ethanol dose. In the 129S6/SvEvTac (129) strain, treatment with 15% ethanol in the drinking water for 6.5 months produced an accumulation of hepatic uroporphyrin (URO) 4-fold higher than that observed with 10% ethanol, a 90% decrease in uroporphyrinogen decarboxylase activity (UROD), and further increased the activities of hepatic 5-aminolevulinate synthase (ALAS) and CYP1A2. Hepatic nonheme iron (NHFe) and hepatocyte iron staining were not further increased by 15% compared to 10% ethanol. Treatment of C57BL/6 Hfe(-/-) mice with 15% ethanol for 6.5 months did not increase hepatic URO. Although NHFe was increased by ethanol, the resulting level was only half that of ethanol-treated 129 Hfe(-/-) mice. ALAS induction was similar in both Hfe(-/-) strains. In wild-type 129 mice treated with ethanol for 6 to 7 months, administration of iron dextran increased hepatic URO accumulation and decreased UROD activity. In conclusion, this study demonstrates a strong effect of genetic background on ethanol-induced uroporphyria, which is probably due to a greater effect of ethanol on iron metabolism in the susceptible strain.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Porfiria Cutânea Tardia/genética , 5-Aminolevulinato Sintetase/metabolismo , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Citocromo P-450 CYP1A2/metabolismo , Proteína da Hemocromatose , Ferro/metabolismo , Complexo Ferro-Dextran/farmacologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Porfiria Cutânea Tardia/etiologia , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/metabolismo
11.
Laryngoscope ; 114(8): 1406-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15280717

RESUMO

OBJECTIVES/HYPOTHESIS: Chondroitin sulfate (CS) is a glycosaminoglycan in the extracellular matrix of all vertebrates. A biocompatible, nonimmunogenic, pliable hydrogel preparation of CS has recently been produced and has shown benefit in wound healing in murine and porcine epidermis. The objective of the current experiment is to compare the wound healing properties of CS hydrogel versus no treatment in wounds of the maxillary sinus mucosa. STUDY DESIGN: Prospective investigation in an animal model. METHODS: A 6 mm wound was created in bilateral maxillary sinuses of 17 New Zealand white rabbits. CS hydrogel (case) and no dressing (control) were randomly assigned to one side each as wound treatment. Wounds were examined ex vivo at 2, 4, 6, 10, and 14 day postinjury intervals. Wound diameter was measured microscopically by a blinded investigator. Representative specimens were examined histologically. RESULTS: The CS disc was partially integrated into the wounds at the 4-day interval and completely integrated by the 6-day interval. The average wound diameters for the case versus control side were similar at 2 days (3.75 mm vs. 4.42 mm) but differed significantly at 4 days (2.86 mm. vs. 3.80 mm., P =.035). At 6 days, the wounds could not be discerned on either the case or control sides. However, histologic analysis revealed accelerated healing with the CS treatment. The treated wounds displayed respiratory epithelium as opposed to the squamous epithelium exhibited on the untreated sides. CONCLUSIONS: Despite some limitations, the New Zealand white rabbit is an effective model for the study of sinonasal wound healing. CS hydrogel accelerates wound healing in sinonasal mucosa at a 4-day endpoint. We propose that the CS hydrogel acts as a surrogate extracellular matrix, serving as a repository for cytokines and growth factors produced by the regenerating mucosa. It may also provide a structural framework for fibroblasts and epithelial regeneration. Further study is necessary to establish this relationship.


Assuntos
Bandagens , Materiais Biocompatíveis/administração & dosagem , Sulfatos de Condroitina/administração & dosagem , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Seio Maxilar , Mucosa Nasal/patologia , Cicatrização , Animais , Sulfatos de Condroitina/fisiologia , Coelhos , Cicatrização/fisiologia
12.
J Biomed Mater Res B Appl Biomater ; 70(1): 130-8, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15199593

RESUMO

An adverse tissue response was reported in patients implanted with porous coated implants containing a mineral oil contaminant. A comparative staining technique was developed in a rabbit and sheep model to identify the presence of a mineral oil contaminant on porous coated implants by examining the surrounding tissues and end organs. The hypothesis for the study was that mineral oil, a saturated lipid, could be distinguished from unsaturated lipids such as animal fats. Frozen rabbit and sheep tissues were stained with hematoxylin and eosin, oil-red-O, and bromide-silver nitrate. Twenty-eight positive control rabbits were injected with mineral oil and 12 negative control rabbits were injected with saline into the paravertebral muscle and synovial cavity of the knee joint. Comparative analysis of tissue was conducted after 1, 3, 5, and 12 weeks. Thermally cleaned porous coated implants were implanted for 6, 12, and 24 weeks into cancellous bone of the distal femur of sheep to allow for a weight-bearing model. The comparative histological analysis of the positive control rabbit tissue allowed for detection of mineral oil in all tissues examined. Histological analysis of sheep tissue and saline-injected rabbit tissue showed no evidence of mineral oil or adverse tissue response.


Assuntos
Óleo Mineral/análise , Próteses e Implantes , Coloração e Rotulagem/métodos , Animais , Materiais Revestidos Biocompatíveis/efeitos adversos , Materiais Revestidos Biocompatíveis/química , Endotoxinas/metabolismo , Feminino , Histocitoquímica/métodos , Humanos , Masculino , Óleo Mineral/toxicidade , Coelhos , Ovinos , Fatores de Tempo
13.
Life Sci ; 75(4): 447-59, 2004 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-15147831

RESUMO

We have previously reported the synthesis and characterization of two new classes of selenazolidine-4(R)-carboxylic acids (2-oxo and 2-methyl-SCAs) (OSCA and MSCA, respectively), as well as the "parent" compound, selenazolidine-4(R)-carboxylic acid (SCA, selenaproline). These compounds were designed as prodrugs of L-selenocysteine with potential application in cancer chemoprevention or other clinical uses. We will be exploring the chemopreventive activity of the new compounds in the well-established A/J mouse model of tobacco-induced lung carcinogenesis. The objectives of the present study were to investigate several fundamental biochemical endpoints after selenazolidine administration compared with other selenium-containing agents. Groups of mice were fed either AIN-76A diet alone or the diet supplemented with the following selenium compounds (ppm Se): sodium selenite (5), L-selenomethionine (3.75), L-selenocystine (15), Se-methyl-L-selenocysteine (3), MSCA (5, 10, or 15), OSCA (5, 10, or 15), or SCA (5, 10, or 15). After 28 days of supplementation, toxicity of the selenazolidines was not evident, as measured by outward appearance and behavior, body and organ weight changes, and histological evaluation of liver and lung tissue. Select treatment groups showed significant increases in selenium levels in blood and tissues. Increased activity of selenium-dependent glutathione peroxidase (GPx) in blood and liver illustrated that the selenazolidines provided a source of biologically-available selenium.


Assuntos
Glutationa Peroxidase/biossíntese , Compostos Organosselênicos/toxicidade , Pró-Fármacos/toxicidade , Prolina/análogos & derivados , Prolina/toxicidade , Selênio/farmacocinética , Selenocisteína/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Indução Enzimática , Feminino , Glutationa Peroxidase/sangue , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Compostos Organosselênicos/farmacocinética , Compostos Organosselênicos/farmacologia , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Prolina/farmacocinética , Prolina/farmacologia , Selênio/sangue , Selenocisteína/farmacocinética , Selenocisteína/farmacologia , Distribuição Tecidual , Testes de Toxicidade
14.
Hepatology ; 37(2): 351-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12540785

RESUMO

Two major risk factors for the development of porphyria cutanea tarda (PCT) are alcohol consumption and homozygosity for the C282Y mutation in the hereditary hemochromatosis gene (HFE). To develop an animal model, Hfe knockout mice were treated continuously with 10% ethanol in drinking water. By 4 months, uroporphyrin (URO) was detected in the urine. At 6 to 7 months, hepatic URO was increased and hepatic uroporphyrinogen decarboxylase (UROD) activity was decreased. Untreated Hfe(-/-) mice or wild-type mice treated with or without ethanol did not show any of these biochemical changes. Treatment with ethanol increased hepatic nonheme iron and hepatic 5-aminolevulinate synthase activity in Hfe(-/-) but not wild-type mice. The increases in nonheme iron in Hfe(-/-) mice were associated with diffuse increases in iron staining of parenchymal cells but without evidence of significant liver injury. In conclusion, the results of this study suggest that the uroporphyrinogenic effect of ethanol is mediated by its effects on hepatic iron metabolism. Ethanol-treated Hfe(-/-) mice seem to be an excellent model for studies of alcohol-mediated PCT.


Assuntos
Etanol/farmacologia , Proteínas de Membrana/deficiência , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/urina , 5-Aminolevulinato Sintetase/metabolismo , Animais , Citocromo P-450 CYP1A2/metabolismo , Modelos Animais de Doenças , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Ferro/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout/genética , Distribuição Tecidual
15.
Semin Ultrasound CT MR ; 23(4): 275-87, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12465685

RESUMO

The pathologic diagnosis of diffuse lung disease is based on the recognition of specific patterns closely correlated with the clinical history and the radiologic findings. The current pathologic classification of the idiopathic interstitial pneumonias is in flux, with the new creation of cellular and fibrotic subtypes of nonspecific interstitial pneumonia and further refinements in the criteria for usual interstitial pneumonia, desquamative interstitial pneumonia, and respiratory bronchiolitis-associated lung disease. The radiologist plays a pivotal role in the diagnosis of these conditions by using high-resolution computed tomography. The radiologist also provides an important service by guiding the clinician or surgeon, based on the site and extent of disease, in obtaining a sample that will provide the pathologist with appropriate diagnostic material. This article reviews the current pathologic criteria for the classification of idiopathic interstitial pneumonias, smoking-related disorders, hypersensitivity pneumonitis, as well as the lymphoproliferative disorder lymphocytic interstitial pneumonitis, with a focus on differential diagnoses.


Assuntos
Doenças Pulmonares Intersticiais/patologia , Fumar/efeitos adversos , Diagnóstico Diferencial , Humanos , Pulmão/diagnóstico por imagem , Doenças Pulmonares Intersticiais/classificação , Doenças Pulmonares Intersticiais/etiologia , Doenças Pulmonares Intersticiais/mortalidade , Radiografia , Taxa de Sobrevida
16.
Hepatology ; 35(4): 912-21, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11915039

RESUMO

In mice treated with 5-aminolevulinic acid (ALA) and polyhalogenated aromatic compounds, the levels of both hepatic cytochrome P450 (CYP)1A2 and iron-which can be quite different among inbred strains-are critical in causing experimental uroporphyria. Here we investigate the development of uroporphyria as a function of CYP1A2 and iron levels in the liver of mice having a common C57BL/6 genetic background. We compared Cyp1a2(-/-) knockout mice, Cyp1a2(+/-) heterozygotes, Cyp1a2(+/+) wild type, and Cyp1a2(+/+) mice pretreated with a low dose of 3,3',4,4',5-pentachlorobiphenyl (PCB126) (4 microg/kg). Cyp1a2(+/-) mice contain about 60% of the hepatic CYP1A2 content of Cyp1a2(+/+) mice, and the PCB126-pretreated Cyp1a2(+/+) mice have about twice the wild-type levels of CYP1A2. ALA- and iron-treated Cyp1a2(+/+) mice are known to accumulate hepatic uroporphyrin; this accumulation was increased 7-fold by pretreatment with the low dose of PCB126. ALA- and iron-treated Cyp1a2(+/-) heterozygote mice accumulated no uroporphyrin in 4 weeks, but by 8 weeks accumulated significant amounts of uroporphyrin. As previously reported, the ALA- and iron-treated Cyp1a2(-/-) knockout mouse has no CYP1A2 and exhibits no detectable uroporphyrin accumulation. Iron dose-response curves in ALA- and PCB126-treated Cyp1a2(+/+) mice showed that hepatic iron levels greater than 850 microg/g liver were required to produce significant uroporphyrin accumulation in the liver. Other measures of hepatic effects of iron (iron-response element-binding protein [IRP]-iron response element [IRE] binding activity and accumulation of protoporphyrin from ALA) decreased when the level of iron was considerably lower than 850 microg/g liver. At low iron doses, accumulation of iron was principally in Kupffer cells, whereas at the higher doses (required to stimulate uroporphyrin accumulation), more iron was found in parenchymal cells. We conclude that small changes in hepatic CYP1A2 levels can dramatically affect uroporphyria in C57BL/6 mice, providing the animals have been sufficiently loaded with iron; these data might be clinically relevant to acquired (sporadic) porphyria cutanea tarda, because humans show greater than 60-fold genetic differences in hepatic basal CYP1A2.


Assuntos
Citocromo P-450 CYP1A2/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Porfiria Cutânea Tardia/metabolismo , Ácido Aminolevulínico/farmacologia , Animais , Citocromo P-450 CYP1A2/genética , Limiar Diferencial , Modelos Animais de Doenças , Hepatócitos/metabolismo , Ferro/farmacologia , Células de Kupffer/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Bifenilos Policlorados , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/genética , Porfiria Cutânea Tardia/patologia , Distribuição Tecidual , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...