Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters











Publication year range
1.
PLoS One ; 9(2): e89889, 2014.
Article in English | MEDLINE | ID: mdl-24587102

ABSTRACT

Uroporphyrinogen decarboxylase (UROD) catalyzes the conversion of uroporphyrinogen to coproporphyrinogen during heme biosynthesis. This enzyme was recently identified as a potential anticancer target; its inhibition leads to an increase in reactive oxygen species, likely mediated by the Fenton reaction, thereby decreasing cancer cell viability and working in cooperation with radiation and/or cisplatin. Because there is no known chemical UROD inhibitor suitable for use in translational studies, we aimed to design, synthesize, and characterize such a compound. Initial in silico-based design and docking analyses identified a potential porphyrin analogue that was subsequently synthesized. This species, a porphodimethene (named PI-16), was found to inhibit UROD in an enzymatic assay (IC50 = 9.9 µM), but did not affect porphobilinogen deaminase (at 62.5 µM), thereby exhibiting specificity. In cellular assays, PI-16 reduced the viability of FaDu and ME-180 cancer cells with half maximal effective concentrations of 22.7 µM and 26.9 µM, respectively, and only minimally affected normal oral epithelial (NOE) cells. PI-16 also combined effectively with radiation and cisplatin, with potent synergy being observed in the case of cisplatin in FaDu cells (Chou-Talalay combination index <1). This work presents the first known synthetic UROD inhibitor, and sets the foundation for the design, synthesis, and characterization of higher affinity and more effective UROD inhibitors.


Subject(s)
Models, Molecular , Porphyrins/chemical synthesis , Recombinant Proteins/metabolism , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Epithelial Cells/drug effects , Humans , Inhibitory Concentration 50 , Molecular Structure , Porphyrins/chemistry , Porphyrins/pharmacology , Recombinant Proteins/chemistry , Substrate Specificity , Uroporphyrinogen Decarboxylase/chemistry
2.
J Biomol Struct Dyn ; 31(12): 1358-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23140436

ABSTRACT

Overexpression of epidermal growth factor receptor (EGFR), Her2, and uroporphyrinogen decarboxylase (UROD) occurs in a variety of malignant tumor tissues. UROD has potential to modulate tumor response of radiotherapy for head and neck cancer, and EGFR and Her2 are common drug targets for the treatment of head and neck cancer. This study attempts to find a possible lead compound backbone from TCM Database@Taiwan ( http://tcm.cmu.edu.tw/ ) for EGFR, Her2, and UROD proteins against head and neck cancer using computational techniques. Possible traditional Chinese medicine (TCM) lead compounds had potential binding affinities with EGFR, Her2, and UROD proteins. The candidates formed stable interactions with residues Arg803, Thr854 in EGFR, residues Thr862, Asp863 in Her2 protein, and residues Arg37, Arg41 in UROD protein, which are key residues in the binding or catalytic domain of EGFR, Her2, and UROD proteins. Thus, the TCM candidates indicated a possible molecule backbone for evolving potential inhibitors for three drug target proteins against head and neck cancer.


Subject(s)
Enzyme Inhibitors/therapeutic use , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/prevention & control , Receptor, ErbB-2/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Computer Simulation , Databases, Factual , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Humans , Medicine, Chinese Traditional/methods , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Taiwan , Uroporphyrinogen Decarboxylase/chemistry , Uroporphyrinogen Decarboxylase/metabolism
3.
Ann Dermatol Venereol ; 137(1): 36-9, 2010 Jan.
Article in French | MEDLINE | ID: mdl-20110066

ABSTRACT

BACKGROUND: Voriconazole is a systemic antifungal drug that can induce phototoxic reactions suggestive of porphyria cutanea tarda (PCT); however, porphyrin levels in urine, blood and stool remain within the normal range. Superficial cheilitis is frequently associated with this clinical picture; it is believed to be related to drug-induced impairment of endogenous retinoid metabolism. We report a case of true PCT associated with cheilitis, both occurring soon after the introduction of voriconazole and partially disappearing after withdrawal of this drug. CASE REPORT: A 65-year-old man with a past history of excessive alcohol consumption presented with typical features of PCT associated with a mild superficial desquamating cheilitis. Both symptoms had appeared 12 days after initiation of oral voriconazole for a cavitary aspergillosis. Laboratory tests confirmed a sporadic case of PCT. Withdrawal of voriconazole (replaced by itraconazole) resulted in complete disappearance of the cheilitis but incomplete remission of the PCT. Ultimately, the patient was successfully treated by venous puncture. DISCUSSION: This patient had both voriconazole-induced superficial cheilitis and a true PCT which seemed related to the same drug. The mechanism by which voriconazole may have revealed PCT remains elusive and could possibly have involved decreased uroporphyrinogen decarboxylase activity in the liver or potentiation of the phototoxic effects of porphyrins by the cutaneous toxicity of voriconazole. CONCLUSION: On presentation of a clinical picture of PCT-like photosensitivity in a patient on voriconazole, laboratory investigations should be performed routinely to rule out true PCT, even in cases of associated cheilitis.


Subject(s)
Antifungal Agents/adverse effects , Photosensitivity Disorders/chemically induced , Porphyria Cutanea Tarda/diagnosis , Pyrimidines/adverse effects , Triazoles/adverse effects , Aged , Alcoholism/complications , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Cheilitis/chemically induced , Feces/chemistry , Humans , Liver/enzymology , Male , Porphyria Cutanea Tarda/complications , Porphyrins/analysis , Porphyrins/blood , Porphyrins/urine , Pulmonary Aspergillosis/complications , Pulmonary Aspergillosis/drug therapy , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Voriconazole
4.
Biomed Chromatogr ; 21(7): 661-3, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17516469

ABSTRACT

An uroporphomethene inhibitor of uroporphyrinogen decarboxylase, characterized by high-performance liquid chromatography/electrospray ionization tandem mass spectrometry, was reported recently (Phillips et al., Proceedings of the National Academy of Sciences of the United States of America 2007; 104: 5079-5084). Close examination of the tandem mass spectrometric fragmentation pattern of the compound showed that it is not a tetrapyrrole or an uroporphyrinogen or uroporphyrin related molecule. The product ion spectrum showed a fragmentation pattern typical of a poly(ethylene glycol) structure. Characteristic fragmentations of the side-chain acetic acid and propionic acid substituents of a uroporphyrin or uroporphyrinogen derivative were absent.


Subject(s)
Chromatography, High Pressure Liquid/methods , Enzyme Inhibitors/pharmacology , Porphyrinogens/pharmacology , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Uroporphyrinogen Decarboxylase/antagonists & inhibitors
5.
Proc Natl Acad Sci U S A ; 104(12): 5079-84, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17360334

ABSTRACT

Porphyria cutanea tarda (PCT), the most common form of porphyria in humans, is due to reduced activity of uroporphyrinogen decarboxylase (URO-D) in the liver. Previous studies have demonstrated that protein levels of URO-D do not change when catalytic activity is reduced, suggesting that an inhibitor of URO-D is generated in hepatocytes. Here, we describe the identification and characterization of an inhibitor of URO-D in liver cytosolic extracts from two murine models of PCT: wild-type mice treated with iron, delta-aminolevulinic acid, and polychlorinated biphenyls; and mice with one null allele of Uro-d and two null alleles of the hemochromatosis gene (Uro-d(+/-), Hfe(-/-)) that develop PCT with no treatments. In both models, we identified an inhibitor of recombinant human URO-D (rhURO-D). The inhibitor was characterized by solid-phase extraction, chromatography, UV-visible spectroscopy, and mass spectroscopy and proved to be uroporphomethene, a compound in which one bridge carbon in the uroporphyrinogen macrocycle is oxidized. We synthesized uroporphomethene by photooxidation of enzymatically generated uroporphyrinogen I or III. Both uroporphomethenes inhibited rhURO-D, but the III isomer porphomethene was a more potent inhibitor. Finally, we detected an inhibitor of rhURO-D in cytosolic extracts of liver biopsy samples of patients with PCT. These studies define the mechanism underlying clinical expression of the PCT phenotype, namely oxidation of uroporphyrinogen to uroporphomethene, a competitive inhibitor of URO-D. The oxidation reaction is iron-dependent.


Subject(s)
Porphyria Cutanea Tarda/etiology , Porphyrins/pharmacology , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Animals , Chromatography, High Pressure Liquid , Cytosol/drug effects , Cytosol/enzymology , Humans , Liver/drug effects , Liver/enzymology , Liver Extracts , Mass Spectrometry , Mice , Mice, Inbred C57BL , Porphyria Cutanea Tarda/chemically induced , Porphyrins/analysis , Porphyrins/chemistry , Recombinant Proteins/metabolism , Uroporphyrinogens/chemistry
6.
Arch Dermatol Res ; 295(3): 109-11, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12811577

ABSTRACT

In an investigation of autoimmune antibodies using indirect immunofluorescence and Western blot analysis in a group of porphyria cutanea tarda patients we did not find any cytosolic antibodies potentially able to inhibit uroporphyrinogen decarboxylase. Furthermore, no known etiological factors were present in any of our patients. We therefore consider the development of the recently reported autoantibody with a molecular weight of 40 kDa a reaction to infection with the hepatitis C virus. The origin of mostly antinuclear antibodies against liver antigens (50, 45 and 56 kDa), detected in seven patients, was not identified and their etiopathogenetic implications remain unknown.


Subject(s)
Autoantibodies/analysis , Porphyria Cutanea Tarda/immunology , Antibodies, Antinuclear/analysis , Antibodies, Antinuclear/immunology , Antigens/immunology , Autoantibodies/physiology , Coproporphyrins/urine , Cytosol/immunology , Female , Fluorescent Antibody Technique, Indirect , Hepatitis C Antibodies/analysis , Humans , Immunoblotting , Liver/immunology , Male , Osmolar Concentration , Porphyria Cutanea Tarda/urine , Porphyrins/urine , Uroporphyrinogen Decarboxylase/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 98(1): 259-64, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11134514

ABSTRACT

Approximately one-third of patients with porphyria cutanea tarda (PCT), the most common porphyria in humans, inherit a single mutant allele of the uroporphyrinogen decarboxylase (URO-D) gene. PCT associated with URO-D mutations is designated familial PCT. The phenotype is characterized by a photosensitive dermatosis with hepatic accumulation and urinary excretion of uroporphyrin and hepta-carboxylic porphyrins. Most heterozygotes for URO-D mutations do not express a porphyric phenotype unless hepatic siderosis is present. Hemochromatosis gene (HFE) mutations are frequently found when the phenotype is expressed. We used homologous recombination to disrupt one allele of murine URO-D. URO-D(+/-) mice had half-wild type (wt) URO-D protein and enzymatic activity in all tissues but did not accumulate hepatic porphyrins, indicating that half-normal URO-D activity is not rate limiting. When URO-D(+/-) mice were injected with iron-dextran and given drinking water containing delta-aminolevulinic acid for 21 days, hepatic porphyrins accumulated, and hepatic URO-D activity was reduced to 20% of wt. We bred mice homozygous for an HFE gene disruption (HFE(-/-)) to URO-D(+/-) mice, generating mice with the URO-D(+/-)/HFE(-/-) genotype. These animals developed a porphyric phenotype by 14 weeks of age without ALA supplementation, and URO-D activity was reduced to 14% of wt. These data indicate that iron overload alone is sufficient to reduce URO-D activity to rate-limiting levels in URO-D(+/-) mice. The URO-D(+/-) mouse serves as an excellent model of familial PCT and affords the opportunity to define the mechanism by which iron influences URO-D activity.


Subject(s)
Disease Models, Animal , Hemochromatosis/genetics , Porphyria Cutanea Tarda/genetics , Uroporphyrinogen Decarboxylase/genetics , Aminolevulinic Acid/pharmacology , Animals , Cloning, Molecular , Coproporphyrinogens/chemistry , Coproporphyrinogens/metabolism , Enzyme Inhibitors/pharmacology , Gene Deletion , Gene Targeting , Genotype , Humans , Iron/analysis , Iron-Dextran Complex/administration & dosage , Iron-Dextran Complex/pharmacology , Liver/chemistry , Liver/metabolism , Mice , Mice, Knockout , Phenotype , Porphyria Cutanea Tarda/chemically induced , Porphyria Cutanea Tarda/enzymology , Porphyria Cutanea Tarda/metabolism , Porphyrins/analysis , Porphyrins/urine , Stem Cells/metabolism , Uroporphyrinogen Decarboxylase/analysis , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/metabolism , Uroporphyrinogens/chemistry , Uroporphyrinogens/metabolism
8.
Comp Biochem Physiol C Toxicol Pharmacol ; 127(2): 165-75, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11083027

ABSTRACT

The aim of the present study was to determine whether short-term administration of hexachlorobenzene (HCB) (1 g/kg body wt., suspended in water, 5 days/week), could cause and maintain marked porphyria in the absence of the exogenous drug, and whether porphyria parameters can be useful as biomarkers of HCB persistence in rats. Hepatic uroporphyrinogen decarboxylase activity, its inhibitor formation, porphyrin content and composition were studied in Wistar rats treated with the fungicide for 1, 2, 3, or 4 weeks and then withdrawn for a 20-week period. The time course of urinary porphyrin excretion was studied for 7 weeks either by continuous treatment for the entire period, or a 1-week HCB administration. The degree of porphyria achieved by rats after 20 weeks of suspended HCB administration was severe, independent of the length of the treatment, and even higher than that observed in animals analysed immediately at the end of each treatment. Rats treated with HCB for 1 week showed a modest decrease in uroporphyrinogen decarboxylase and low inhibitor formation, and exhibited a greater enzyme inhibition, inhibitor formation, hepatic porphyrin accumulation, and an altered pattern of porphyrin composition in the absence of the exogenous drug. Independent of the treatment, urinary porphyrins rose after a delay of 5 weeks. Substantial amounts of HCB were still found in fat of rats treated with HCB for 1 week, after a withdrawal period of 20 weeks. These results suggest that the high persistence of HCB in tissues acts as a continuous source of the xenobiotic, and stimulus for heme biosynthesis derangement. The alterations induced by HCB within 1 week of treatment could be regarded as an initial trigger for irreversible damage on heme metabolism. Thus, abnormalities in heme biosynthesis can be considered effective markers of HCB persistence in rats or of irreversible HCB-induced damage. Taking into account the delayed and enhanced metabolic effects of HCB, it is advisable that porphyria parameters should be evaluated not only immediately after exposure, but also some time afterwards, especially in susceptible and occupationally-exposed populations.


Subject(s)
Fungicides, Industrial/toxicity , Heme/metabolism , Hexachlorobenzene/toxicity , Porphyrias/chemically induced , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Animals, Outbred Strains , Biomarkers/analysis , Drug Administration Schedule , Enzyme Inhibitors/toxicity , Female , Fungicides, Industrial/administration & dosage , Fungicides, Industrial/pharmacokinetics , Hexachlorobenzene/administration & dosage , Hexachlorobenzene/pharmacokinetics , Liver/drug effects , Liver/enzymology , Porphyrias/metabolism , Rats , Rats, Wistar , Time Factors , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/metabolism
9.
Kidney Blood Press Res ; 23(1): 20-6, 2000.
Article in English | MEDLINE | ID: mdl-10567850

ABSTRACT

In the present study, the effects of hexachlorobenzene (HCB) on lipid peroxidation and heme metabolism in the different constitutive suborgans of the kidney were determined. For this purpose, conjugated diene and malondialdehyde levels, as lipid peroxidation parameters, and porphyrin accumulation, uroporphyrinogen decarboxylase activity, and its inhibitor formation, as measures of heme metabolism, were determined in renal cortex, medulla, and papilla. Adult Wistar rats were treated with HCB during 1, 2, 3, or 4 weeks. A significant increase in cortical conjugated dienes was observed from the 1st week of treatment. The malondialdehyde levels rose by 47, 34, and 28% after 2, 3, and 4 weeks of intoxication, respectively. The porphyrin content showed a tenfold increase after 4 weeks of treatment, and the uroporphyrinogen decarboxylase activity was reduced by 26 and 58% with respect to control values after 3 and 4 weeks of treatment, respectively. The results demonstrate a direct correlation between the oxidative environment and the effect elicited by the drug on heme metabolism in the renal cortex. In contrast, in papilla and medulla, where the antioxidant systems were higher, HCB showed no porphyrinogenic effect.


Subject(s)
Heme/metabolism , Hexachlorobenzene/toxicity , Kidney/cytology , Lipid Peroxidation/drug effects , Porphyrias/metabolism , Animals , Biomarkers , Enzyme Inhibitors/pharmacology , Female , Kidney/drug effects , Kidney Cortex/metabolism , Liver/metabolism , Malondialdehyde/metabolism , Porphyrias/chemically induced , Porphyrins/metabolism , Rats , Rats, Wistar , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/metabolism
10.
Toxicol Appl Pharmacol ; 145(1): 23-33, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9221820

ABSTRACT

Hexachlorobenzene (HCB)-induced porphyria occurs in female, but not male, rats after a delay of 35 days following HCB treatment. Uroporphyrinogen decarboxylase (UROD) inhibition has been proposed as a primary causative event. To determine whether there also exists a delay phase and a sexual dimorphism for UROD inhibition, groups of male and female rats were given HCB (100 mg/kg/day) from Days 1 to 5. Hepatic uroporphyrin III was markedly increased only after Day 33. Liver cytosol UROD activity in HCB-treated female rats with porphyria at Days 33, 40, 47, 54, and 100 was decreased by over 70% compared to concurrent control, whereas treated male rats as well as nonporphyric female rats had UROD activity comparable to control levels at Days 6, 12, 19, 26, 33, 40, 47, and 54. Level of immunoreactive UROD in cytosol of porphyric rats was not modified by HCB. No gender-related differences in liver cytosol radiolabel level ([14C]HCB given as the fifth dose) were found at Days 6 and 30. Chromatography of liver cytosol showed nonspecific binding of radiolabel to proteins for males, porphyric and nonporphyric females, and loss of UROD activity did not correlate with the amount of radiolabel in the UROD-containing fractions. Thus, the gender-specific decrease in UROD activity observed when porphyria develops in female rats (delay of about 4 weeks), as well as the persistence of low activity and porphyria for months, suggests that UROD inhibition was causally related to porphyria.


Subject(s)
Fungicides, Industrial/toxicity , Hexachlorobenzene/toxicity , Liver/drug effects , Porphyrias, Hepatic/enzymology , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Analysis of Variance , Animals , Carbon Radioisotopes , Chromatography, Gel , Chromatography, High Pressure Liquid , Cytosol/drug effects , Cytosol/enzymology , Dialysis , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Female , Fungicides, Industrial/administration & dosage , Hexachlorobenzene/administration & dosage , Isotope Labeling , Liver/enzymology , Liver/metabolism , Male , Porphyrias, Hepatic/chemically induced , Rats , Rats, Sprague-Dawley , Sex Characteristics , Uroporphyrinogen Decarboxylase/blood , Uroporphyrins/metabolism , Uroporphyrins/urine
12.
Hum Exp Toxicol ; 14(6): 475-83, 1995 Jun.
Article in English | MEDLINE | ID: mdl-8519522

ABSTRACT

1. A time-course study was carried out in mice subchronically exposed to As III (as sodium arsenite) or As V (as sodium arsenate), via drinking water, relating the pattern of urinary porphyrin excretion to the renal and hepatic enzyme activities of porphobilinogen deaminase (PBGD), uroporphyrinogen III synthetase (URO III-S), uroporphyrinogen decarboxylase (URO-D) and coproporphyrinogen oxidase (COPRO-O), as well as to the hepatic porphyrin accumulation in the treated animals. 2. A time-dependent, wave-like porphyric response was found in mice exposed to As V, and the increases seen in total urinary porphyrins (at 3 weeks of exposure) corresponded to an increased activity of PBGD and Uro III-S in liver. 3. Significant decreases in renal URO-D and hepatic and renal COPRO-O activities were found in treated mice; these inhibitions were more pronounced in animals exposed to As III. 4. The combination of these enzymic effects may explain the time-dependent porphyric response of mice subchronically exposed to As. Finally, the relative magnitudes of URO-D and COPRO-O inhibitions may determine the pattern of porphyrin concentration observed in urine and tissues. 5. The decrease in renal URO-D activity may help to explain the inversion in the coproporphyrin/uroporphyrin ratio previously reported in humans chronically exposed to As; however, there were differences between the urinary porphyrin profiles found in both species. The possible reasons for the similarities and differences are briefly discussed.


Subject(s)
Arsenates/toxicity , Arsenites/toxicity , Porphyrins/urine , Sodium Compounds/toxicity , Sulfhydryl Reagents/toxicity , Administration, Oral , Animals , Arsenates/administration & dosage , Arsenites/administration & dosage , Coproporphyrinogen Oxidase/antagonists & inhibitors , Dose-Response Relationship, Drug , Hydroxymethylbilane Synthase/antagonists & inhibitors , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Porphyrins/metabolism , Sodium Compounds/administration & dosage , Sulfhydryl Reagents/administration & dosage , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogen III Synthetase/antagonists & inhibitors
13.
Int J Biochem ; 26(2): 259-62, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8174760

ABSTRACT

1. The effect of URO I on the activity of ALA-D, PBGase, deaminase and URO-D, both in aerobiosis and anaerobiosis, was studied. 2. Photoinactivation of the enzymes was much lower in an anaerobic than in an aerobic atmosphere. 3. Dark inactivation in the absence of oxygen was lower than its presence. 4. Preincubation in the presence of ALA or PBG protected the enzymic activity of ALA-D, PBGase and deaminase against URO I-inactivation both under u.v. light and in the dark. 5. Photoinactivating action of URO I would be mediated by reactive oxygen species generated by the excited porphyrin after its absorption of light. Dark inactivation, in aerobiosis, can also be partly mediated by amino acid oxidation, although to a lesser extent than that observed under u.v. light.


Subject(s)
Darkness , Enzyme Inhibitors/pharmacology , Light , Uroporphyrins/pharmacology , Aerobiosis , Ammonia-Lyases/antagonists & inhibitors , Anaerobiosis , Humans , Hydroxymethylbilane Synthase/antagonists & inhibitors , Porphobilinogen Synthase/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/antagonists & inhibitors
14.
Biochem J ; 293 ( Pt 3): 703-12, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-8352737

ABSTRACT

Uroporphyrinogen decarboxylase (EC 4.1.1.37) was purified 600-fold from Rhodobacter sphaeroides grown anaerobically in the light. The enzyme, under both denaturing and non-denaturing conditions, is a monomer of M(r) 41,000. The Km values are 1.8 microM and 6.0 microM for the conversion of uroporphyrinogen I and III to coproporphyrinogen I and III respectively. The enzyme is susceptible to inhibition by both uroporphyrinogen and uroporphyrin. The pH optimum is 6.8 and the isoelectric point is 4.4. The importance of cysteine and arginine residues is implicated from studies with inhibitors. The sequence of the first 29 amino acids of the N-terminus shows a high degree of similarity to the primary structures of other uroporphyrinogen decarboxylases. Studies on the order of decarboxylation of the four acetic acid side chains of uroporphyrinogen III suggest that at high substrate levels a random route is preferred.


Subject(s)
Rhodobacter sphaeroides/enzymology , Uroporphyrinogen Decarboxylase/isolation & purification , Amino Acid Sequence , Carboxylic Acids/chemistry , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Electrophoresis, Polyacrylamide Gel , Hot Temperature , Indicators and Reagents , Isoelectric Point , Kinetics , Molecular Sequence Data , Molecular Weight , Porphyrins/pharmacology , Protein Denaturation , Sequence Homology, Amino Acid , Sulfhydryl Compounds/analysis , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogen Decarboxylase/chemistry , Uroporphyrinogen Decarboxylase/metabolism
15.
Biochem J ; 291 ( Pt 1): 29-35, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-8471047

ABSTRACT

Iron overload causes inhibition of hepatic uroporphyrinogen decarboxylase (UROD) and uroporphyria in C57BL/10ScSn but not DBA/2 mice [Smith, Cabral, Carthew, Francis and Manson (1989) Int. J. Cancer 43, 492-496]. We have investigated the induction of uroporphyria in 12 inbred strains of mice 25 weeks after iron treatment (600 mg/kg) to determine if there was any correlation with the Ah locus. Under these conditions, inhibition of UROD occurred to varying degrees in Ahd mice (SWR and AKR) as well as nominally Ahb-1 (C57BL/6J, C57BL/10ScSn and C57BL/10-cc) and Ahb-2 strains (BALB/c and C3H/HeJ). Five other Ahb or Ahd strains (C57BL/Ks, A/J, CBA/J, LP and DBA/2) were unaffected. Thus there appeared to be no correlation with the Ah phenotype and this illustrated that some other variable inherited factors are involved. Comparisons between another susceptible strain, A2G, and the congenic A2G-hr/+strain (carrying the recessive hr gene) showed a modulating influence associated with the hr locus. In contrast with individual mice of inbred strains, which showed consistent responses to iron, those of the outbred MF1 strain showed a spectrum of sensitivities as might be expected for a heterogeneic stock. The rate of porphyria development was accelerated by administration of 5-aminolaevulinic acid (5-ALA) in the drinking water, but this did not overcome strain differences. Among four strains the order of susceptibility was SWR > C57BL/10ScSn > C57B1/6J > DBA/2 (the last strain was completely resistant). With degrees of iron loading greater than 600 mg of Fe/kg (1200-1800 mg of Fe/kg) C57BL/10ScSn mice (after 20 weeks) and SWR mice (after 5 weeks which included 4 weeks of 5-ALA treatment) had less inhibition of UROD and a lower uroporphyric response, showing that there was an optimum level of liver iron concentration. Studies on selected microsomal enzyme activities associated with cytochrome P-450 showed no correlation with the propensities of strains to develop porphyria. These activities included the NADPH-dependent oxidation of uroporphyrinogen I to uroporphyrin I.


Subject(s)
Iron/toxicity , Porphyrias/genetics , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogens/metabolism , Aminolevulinic Acid/pharmacology , Animals , Drug Tolerance/genetics , Hydrocarbons/pharmacology , Iron/administration & dosage , Liver/metabolism , Mice , Mice, Hairless , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Porphyrias/chemically induced
16.
Can J Physiol Pharmacol ; 70(7): 939-42, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1451030

ABSTRACT

A variety of xenobiotics, viz., 3,3',4,4'-tetrachlorobiphenyl (TCBP), sodium phenobarbital (PB), 3,5-diethoxycarbonyl-2, 4,6-trimethylpyridine (OX-DDC), and nifedipine, cause a decrease in uroporphyrinogen decarboxylase (UROG-D) activity, accompanied by uroporphyrin accumulation, in chick embryo hepatocytes in culture. In this study the activity of 17-day-old chick embryo hepatic UROG-D was determined by measuring the conversion of pentacarboxylporphyrinogen I to coproporphyrinogen I, and it was shown that a UROG-D inhibitor, previously reported to accumulate in TCBP-treated and PB-treated chick embryo hepatocytes in culture, also accumulates in OX-DDC-treated and nifedipine-treated chick embryo hepatocytes in culture. It was concluded that the accumulation of a UROG-D inhibitor provides an explanation for the UROG-D inhibition observed in this culture system with xenobiotics that cause uroporphyrin accumulation. Studies of the UROG-D inhibitory fraction isolated from the 10,000 x g, 40,000 x g, and 100,000 x g supernatant fractions of cultured chick embryo hepatocyte homogenate led to the conclusion that the UROG-D inhibitor is derived from a soluble component of the homogenate.


Subject(s)
Liver/enzymology , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Xenobiotics/pharmacology , Animals , Cells, Cultured , Chick Embryo , Chromatography, High Pressure Liquid , Coproporphyrinogens/metabolism
17.
J Enzyme Inhib ; 5(3): 225-33, 1991.
Article in English | MEDLINE | ID: mdl-1669450

ABSTRACT

The action of uroporphyrin I (URO I) on the activity of red cell uroporphyrinogen decarboxylase (URO-D) in the dark and under UV light was studied. Light-dependent-and light-independent inactivation was observed. Both effects increased at increasing concentrations of URO I, the former reached its maximum at 150 microM of sensitizer. At 100 microM of URO I, both light and dark inactivation were temperature dependent amounting to about 50% at 30-37 degrees C. The velocity of dark inactivation increased with increasing temperature in the range of 0 to 45 degrees C. Photoinactivation can be ascribed to primary oxidation of essential amino acids, very likely histidyl residues, followed by secondary inter or intrapeptide cross-linking. Dark inactivation could be the result of both oxidation and cross-linking (although to a less degree than that produced by light) and also direct inhibition of the enzyme by induced conformational changes at its active site through binding of the porphyrin to the protein. When the action of URO I was tested on partially purified URO-D, the enzyme appeared to be more susceptible to the dark than to the light effect.


Subject(s)
Erythrocytes/enzymology , Photosensitizing Agents/pharmacology , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrins/pharmacology , Darkness , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Molecular Weight , Ultraviolet Rays , Uroporphyrinogen Decarboxylase/blood , Uroporphyrinogen Decarboxylase/radiation effects
19.
Biochem Pharmacol ; 40(9): 2059-68, 1990 Nov 01.
Article in English | MEDLINE | ID: mdl-2173599

ABSTRACT

A marked sex difference in the development of uroporphyria occurred after administration of polychlorinated and polybrominated biphenyls (PCBs and PBBs), as well as hexachlorobenzene (HCB), to F344 rats for 15 weeks. Thus the propensity of female rats to develop uroporphyria appears to be a general response to this class of halogenated chemicals. A heat-stable inhibitor(s) of liver uroporphyrinogen decarboxylase was extractable from uroporphyric livers. Although oxidation of uroporphyrinogen I to uroporphyrin I by hepatic microsomes from rats pretreated with porphyrogenic regimes of HCB and PCBs was induced, there was no correlation with the in vivo sex difference in porphyria development. Levels of total cytochrome P450 and pentoxyresorufin and benzyloxyresorufin dealkylase activities (associated with cytochrome P450IIB1) were greater in microsomes from control, HCB, PCB and PBB treated male rats than females. In contrast, ethoxyresorufin deethylase activity (associated with cytochrome P450IA1) was always significantly greater in females. These findings were confirmed by immunoblotting with polyclonal antibodies to cytochromes P450IA1, IA2 and IIB1. Immunocytochemical studies showed that, even after 30 weeks of HCB exposure, cytochromes P450IA1 and P450IA2 were still more highly induced in female liver, especially in the centrilobular region. The results are consistent with the association of cytochrome P450IA isoenzymes with uroporphyria development, although the sex difference in P450IA levels alone may not be marked enough to provide the complete explanation for the pronounced susceptibility of females to HCB.


Subject(s)
Chemical and Drug Induced Liver Injury , Cytochrome P-450 Enzyme System/biosynthesis , Hexachlorobenzene/toxicity , Polybrominated Biphenyls/toxicity , Porphyrias/chemically induced , Uroporphyrins/urine , Animals , Aroclors/toxicity , Enzyme Induction/drug effects , Female , Immunohistochemistry , Liver Diseases/enzymology , Liver Diseases/metabolism , Male , Microsomes, Liver/metabolism , Oxidation-Reduction , Porphyrias/enzymology , Porphyrias/metabolism , Rats , Rats, Inbred F344 , Sex Factors , Uroporphyrinogen Decarboxylase/antagonists & inhibitors
20.
Biochem J ; 269(2): 437-41, 1990 Jul 15.
Article in English | MEDLINE | ID: mdl-2117439

ABSTRACT

It was previously shown that uroporphyrinogen oxidation is catalysed by a form of cytochrome P-450 induced by 3-methylcholanthrene [Sinclair, Lambrecht & Sinclair (1987) Biochem. Biophys. Res. Commun. 146, 1324-1329]. We have now measured uroporphyrinogen oxidation and uroporphyrinogen decarboxylation simultaneously in 10,000 g supernatants from the livers of methylcholanthrene-treated mice and chick embryos incubated with an NADPH-generating system. We found that uroporphyrinogen oxidation is associated with inhibition of uroporphyrinogen decarboxylase activity. The decreased uroporphyrinogen decarboxylase activity was not due to depletion of substrate, since decarboxylase activity was not increased by a 2.6-fold increase in uroporphyrinogen. Uroporphyrinogen oxidation and the associated inhibition of decarboxylase activity were also observed with liver supernatant from methylcholanthrene-treated chick embryo; both actions required the addition of 3,3',4,4'-tetrachlorobiphenyl. Uroporphyrinogen oxidation catalysed by microsomes from a methylcholanthrene-treated mouse inhibited the uroporphyrinogen decarboxylase activity in the 100,000 g supernatant. Ketoconazole, an inhibitor of cytochrome P-450, prevented both uroporphyrinogen oxidation and the inhibition of uroporphyrinogen decarboxylation. The addition of ketoconazole to mouse supernatant actively oxidizing uroporphyrinogen inhibited the oxidation and restored decarboxylation. The latter finding suggested that a labile inhibitor was formed during the oxidation. These results suggest uroporphyrinogen oxidation may be important in the mechanism of chemically induced uroporphyria.


Subject(s)
Carboxy-Lyases/antagonists & inhibitors , Cytochrome P-450 Enzyme System/metabolism , Liver/enzymology , Porphyrinogens/metabolism , Uroporphyrinogen Decarboxylase/antagonists & inhibitors , Uroporphyrinogens/metabolism , Animals , Chick Embryo , Cytochrome P-450 Enzyme Inhibitors , Ketoconazole/pharmacology , Kinetics , Liver/embryology , Male , Methylcholanthrene/pharmacology , Mice , Mice, Inbred C57BL , Microsomes, Liver/enzymology , NADP/metabolism , Oxidation-Reduction , Polychlorinated Biphenyls/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL