Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Res Toxicol ; 21(2): 330-40, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18163543

ABSTRACT

The dysfunction of hepatic heme synthesis by 2,3,7,8-tetrachlordibenzo- p-dioxin (TCDD) in mice, enhanced by iron, leads to accumulation of uroporphyrins I and III (uroporphyria) and resembles the human disorder porphyria cutanea tarda (PCT) precipitated by alcohol and estrogenic drugs. Although consequences of TCDD are considered entirely dependent on the aryl hydrocarbon receptor (AHR), this is not proven for uroporphyria. Administration of TCDD (75 microg/kg) caused uroporphyria in susceptible C57BL/6J mice with high-affinity AHR after 5 weeks (>600-fold increase in hepatic uroporphyrins). Transcriptomics showed significant modified gene expressions for intermediary, heme, and iron metabolism as well as for oxidative stress and cell injury. Resistant low-affinity AHR DBA/2 mice (no increase in porphyrins) showed far fewer changes. At this dose of TCDD, persistent up-regulation of some traditional AH battery genes occurred in both strains. Essentiality of AHR was demonstrated with C57BL/6 Ahr knockout mice. Elevation of hepatic uroporphyrins was 964-fold in Ahr (+/+) mice, lower in Ahr (+/-) (60-fold), but undetectable with Ahr (-/-) . Consistent with an oxidative mechanism, iron overload enhanced porphyria as well as general liver injury in Ahr (+/+) and Ahr (+/-) mice but had no interactive effect in Ahr (-/-) . In contrast, when iron-treated mice received, instead of TCDD, the heme precursor 5-aminolevulinic acid (ALA), causing uroporphyia in Ahr (+/+) mice (242-fold rise in uroporphyrins), elevation of uroporphyrins I and III (42-fold) also occurred in Ahr (-/-) mice and was seemingly associated with AHR-independent expression of Cyp1a2. The findings prove that AHR is a key factor in porphyria induced in mice by TCDD. However, in other models of human PCT, participation of AHR may not be an essential requirement.


Subject(s)
Environmental Pollutants/metabolism , Heme/metabolism , Polychlorinated Dibenzodioxins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Aminolevulinic Acid/pharmacology , Animals , Cytochrome P-450 CYP1A2/metabolism , Disease Models, Animal , Environmental Pollutants/toxicity , Female , Gene Expression/drug effects , Gene Expression Profiling , Gene Silencing , Heme/genetics , Iron Overload/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Oxidative Stress/drug effects , Polychlorinated Dibenzodioxins/toxicity , Porphyria Cutanea Tarda/chemically induced , Porphyria Cutanea Tarda/genetics , Porphyria Cutanea Tarda/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Up-Regulation , Uroporphyrins/analysis
2.
Chem Res Toxicol ; 19(12): 1660-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17173380

ABSTRACT

Previous work has shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes porphyria, enhanced by iron, in C57BL/6J mice with marked accumulation in the liver of uroporphyrin I and III isomers and heptacarboxylic acid III and is one model of human porphyria cutanea tarda. Preliminary examination by HPLC also indicated the presence of some oxygenated side chain uroporphyrin derivatives. Here, the porphyrin constituents of TCDD-induced porphyric liver have been examined by HPLC/electrospray ionization quadrupole time-of-flight mass spectrometry (HPLC/ESI-Q-TOFMS) to characterize the major and minor porphyrins present in hepatic tissue. As well as the major constituents uroporphyrins I and III, we identified the isomers of heptacarboxylic, hexacarboxylic, and pentacarboxylic acid porphyrins arising from intermediates in the stepwise decarboxylation of uroporphyrinogen I and III to coproporphyrinogens. In addition, monohydroxy analogues of uroporphyrin isomers were detected hydroxylated in the acetic acid and beta-positions of propionic acid side chains and in the meso ring position. Of particular note, for the first time for human and experimental porphyrias, we found chlorins (dihydroxy-, hydroxyspirolactone- ,and dihydroxyspirolactone-urochlorins) consistent with those derived from an epoxyurochlorin structure, formed by oxidation of the double bond of a pyrrole ring of uroporphyrinogen I and III isomers. The findings demonstrate that oxygen insertion into the pyrrole rings of uroporphyrinogens occurs under pathological circumstances in vivo and support the evidence for an oxidative cellular environment present in TCDD-treated porphyric tissue.


Subject(s)
Liver/drug effects , Polychlorinated Dibenzodioxins/toxicity , Spironolactone/metabolism , Uroporphyrinogens/metabolism , Uroporphyrins/metabolism , Animals , Chromatography, High Pressure Liquid , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
3.
Hepatology ; 44(1): 174-85, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16799992

ABSTRACT

Polymorphisms of genes linked to iron metabolism may account for individual variability in hemochromatosis and iron status connected with liver and cardiovascular diseases, cancers, toxicity, and infection. Mouse strains exhibit marked differences in levels of non-heme iron, with C57BL/6J and SWR showing low and high levels, respectively. The genetic basis for this variability was examined using quantitative trait loci (QTL) analysis together with expression profiling and chromosomal positions of known iron-related genes. Non-heme iron levels in liver and spleen of C57BL/6J x SWR F2 mice were poorly correlated, indicating independent regulation. Highly significant (P < .01) polymorphic loci were found on chromosomes 2 and 16 for liver and on chromosomes 8 and 9 for spleen. With sex as a covariate, additional significant or suggestive (P < 0.1) QTL were detected on chromosomes 7, 8, 11, and 19 for liver and on chromosome 2 for spleen. A gene array showed no clear association between most loci and differential iron-related gene expression. The gene for transferrin and a transferrin-like gene map close to the QTL on chromosome 9. Transferrin saturation was significantly lower in C57BL/6J mice than in SWR mice, but there was no significant difference in the serum level of transferrin, hepatic expression, or functional change in cDNA sequence. beta2-Microglobulin, which, unlike other loci, was associated with C57BL/6J alleles, is a candidate for the chromosome 2 QTL for higher iron. In conclusion, the findings show the location of polymorphic genes that determine basal iron status in wild-type mice. Human equivalents may be pertinent in predisposition to hepatic and other disorders.


Subject(s)
Hemochromatosis/genetics , Iron/metabolism , Liver/metabolism , Polymorphism, Genetic , Quantitative Trait Loci , RNA, Messenger/genetics , Spleen/metabolism , Animals , Chromosomes, Mammalian/genetics , Genetic Predisposition to Disease , Genotype , Hemochromatosis/metabolism , Hemochromatosis/pathology , Mice , Mice, Inbred C57BL , Transferrin/metabolism
4.
Biochem Biophys Res Commun ; 331(1): 147-52, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15845371

ABSTRACT

Aryl hydrocarbon receptor ligands, such as polychlorinated biphenyls (PCBs), cause inhibition of the heme biosynthesis enzyme, uroporphyrinogen decarboxylase; this leads to uroporphyria and hepatic tumors, which are markedly enhanced by iron overload in C57BL/10 and C57BL/6 strains of mice. Cyp1a2(-/-) knockout mice were used to compare the effects of CYP1A2 expression on uroporphyria and liver carcinogenesis. PCBs in the diet (100ppm) of Cyp1a2(+/+) wild-type mice caused hepatic uroporphyria, which was strongly increased by iron-dextran (800mg Fe/kg). In contrast, uroporphyria was not detected in Cyp1a2(-/-) knockout mice, although expression of CYP1A1 and CYP2B10 was greatly induced. After 57 weeks on this diet, hepatic preneoplastic foci and tumors were seen in the Cyp1a2(+/+) mice; numbers and severity were enhanced by iron. No foci or tumors were detected in Cyp1a2(-/-) mice, although evidence for other forms of liver injury was observed. Our findings suggest a link not only between CYP1A2, iron metabolism, and the induction of uroporphyria by PCBs, but also with subsequent hepatocarcinogenesis.


Subject(s)
Cytochrome P-450 CYP1A2/metabolism , Environmental Pollutants/toxicity , Iron/toxicity , Liver Neoplasms, Experimental/chemically induced , Polychlorinated Biphenyls/toxicity , Porphyrias, Hepatic/chemically induced , Animals , Cytochrome P-450 CYP1A2/genetics , Drug Synergism , Humans , Liver Neoplasms, Experimental/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Porphyrias, Hepatic/enzymology , Porphyrias, Hepatic/pathology , Rats , Uroporphyrins/metabolism
5.
Am J Pathol ; 166(4): 1041-53, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15793285

ABSTRACT

BALB/c Fech(m1Pas) mice have a mutated ferrochelatase gene resulting in protoporphyria that models the hepatic injury occurring sporadically in human erythropoietic protoporphyria. We used this mouse model to study the development of the injury and to compare the dysfunction of heme synthesis with hepatic gene expression of liver metabolism, oxidative stress, and cellular injury/inflammation. From an early age expression of total cytochrome P450 and many of its isoforms was significantly lower than in wild-type mice. However, despite massive accumulation of protoporphyrin in the liver, expression of the main genes controlling heme synthesis and catabolism (Alas1 and Hmox1, respectively) were only modestly affected even in the presence of the cytochrome P450-inducing CAR agonist 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene. In contrast, in BALB/c mice exhibiting griseofulvin-induced hepatic protoporphyria with induction and destruction of cytochrome P450, both Alas1 and Hmox1 genes were markedly up-regulated. Other expression profiles in BALB/c Fech(m1Pas) mice identified roles for oxidative mechanisms in liver injury while modulated gene expression of hepatocyte transport proteins and cholesterol and bile acid synthesis illustrated the development of cholestasis. Subsequent inflammation and cirrhosis were also shown by the up-regulation of cytokine, cell cycling, and procollagen genes. Thus, gene expression profiles studied in Fech(m1Pas) mice may provide candidates for human polymorphisms that explain the sporadic hepatic consequences of erythropoietic protoporphyria.


Subject(s)
Aging , Heme/metabolism , Liver/pathology , Protoporphyria, Erythropoietic/genetics , Animals , Antifungal Agents/toxicity , Cholestasis/chemically induced , Cholestasis/genetics , Cholestasis/pathology , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Griseofulvin/toxicity , Heme/genetics , Hemeproteins/genetics , Hemeproteins/metabolism , Immunoblotting , Liver/physiology , Male , Mice , Protoporphyria, Erythropoietic/chemically induced , Protoporphyria, Erythropoietic/pathology , Protoporphyrins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
EHP Toxicogenomics ; 111(1T): 37-43, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12735108

ABSTRACT

Erythropoietic protoporphyria patients can develop cholestasis, severe hepatic damage, fibrosis, and cirrhosis. We modeled this hepatic pathology in C57BL/6J and BALB/c mice using griseofulvin and analyzed 3,127 genes for alteration of expression in the liver before and during the onset of protoporphyria, cholestasis, inflammation, and hepatic fibrosis. The two mouse strains developed different levels of pathologic damage in response to the griseofulvin. Characteristic gene expression profiles could be associated with griseofulvin-induced gene expression, disruption of lipid metabolism, and the pathologic states of inflammation, early fibrosis, and cholestasis. Additionally, some genes individually indicated an alteration of homeostasis. or pathologic state; for example, fibroblast proliferation was potentially indicated by increased calcyclin (SA100a6) expression. Changes in cytochrome P450 (Cyp) gene expression were particularly pronounced, with increased expression of the Cyp2a, Cyp2b, and Cyp3a families. Decreased Cyp4a10 and Cyp4a14 expression was observed that could be associated with early pathologic change. A potential decrease in bile acid and steroid biosynthesis was indicated by the decreased expression of Cyp7b1 and Hsd3b4, respectively. DNA damage was indicated by induction of GADD45. This study illustrates how transcriptional programs can be associated with different stimuli in the same experiment. The time course of change in the gene expression profile compared with changes in pathology and clinical chemistry shows the potential of this approach for modeling causative, predictive, and adaptive changes in gene expression during pathologic change.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/metabolism , Gene Expression Profiling , Griseofulvin/toxicity , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Animals , Antigens, Ly/genetics , Cholestasis/chemically induced , Collagen/genetics , Cytochrome P-450 Enzyme System/genetics , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Mol Pharmacol ; 61(3): 674-81, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11854449

ABSTRACT

Among the actions of 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) in mice is the induction of hepatic porphyria. This is similar to the most common disease of this type in humans, sporadic porphyria cutanea tarda (PCT). Evidence is consistent with the actions of dioxin being mediated through binding to the aryl hydrocarbon receptor (AHR) with different Ahr alleles in mouse strains apparently accounting for differential downstream gene expression and susceptibility. However, studies of dioxin-induced porphyria and liver injury indicate that the mechanisms must involve interactions with other genes, perhaps associated with iron metabolism. We performed a quantitative trait locus (QTL) analysis of an F(2) cross between susceptible C57BL/6J (Ahr(b1) allele) and the highly resistant DBA/2 (Ahr(d) allele) strains after treatment with dioxin and iron. For porphyria we found QTLs on chromosomes 11 and 14 in addition to the Ahr gene (chromosome 12). Studies with C57BL/6.D2 Ahr(d) mice confirmed that the Ahr(d) allele alone did not completely negate the response. SWR mice are syngenic for the Ahr(d) allele with the DBA/2 strain but are susceptible to porphyria after elevation of hepatic iron. Analysis of SWRxD2 F(2) mice treated with iron and dioxin showed a QTL on chromosome 11, as well as finding other loci on chromosomes 1 (and possibly 9), for both porphyria and liver injury. These findings show for the first time the location of genes, other than Ahr, that modulate the mechanism of hepatic porphyria and injury caused by dioxin in mice. Orthologous loci may contribute to the pathogenesis of human sporadic PCT.


Subject(s)
Porphyrias/genetics , Receptors, Aryl Hydrocarbon/genetics , Respiratory Distress Syndrome/genetics , Animals , Chromosome Mapping , Chromosomes , Cytochrome P-450 CYP1A2/biosynthesis , Dioxins , Disease Models, Animal , Drug Interactions , Genotype , Iron , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Porphyrias/chemically induced , Quantitative Trait, Heritable , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Distress Syndrome/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...