Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Liver Int ; 42(1): 161-172, 2022 01.
Article in English | MEDLINE | ID: mdl-34717041

ABSTRACT

BACKGROUND & AIMS: Upregulation of hepatic delta-aminolevulinic acid synthase 1 with accumulation of potentially toxic heme precursors delta-aminolevulinic acid and porphobilinogen is fundamental to the pathogenesis of acute hepatic porphyria. AIMS: evaluate long-term efficacy and safety of givosiran in acute hepatic porphyria. METHODS: Interim analysis of ongoing ENVISION study (NCT03338816), after all active patients completed their Month 24 visit. Patients with acute hepatic porphyria (≥12 years) with recurrent attacks received givosiran (2.5 mg/kg monthly) (n = 48) or placebo (n = 46) for 6 months (double-blind period); 93 received givosiran (2.5 mg or 1.25 mg/kg monthly) in the open-label extension (continuous givosiran, n = 47/48; placebo crossover, n = 46/46). Endpoints included annualized attack rate, urinary delta-aminolevulinic acid and porphobilinogen levels, hemin use, daily worst pain, quality of life, and adverse events. RESULTS: Patients receiving continuous givosiran had sustained annualized attack rate reduction (median 1.0 in double-blind period, 0.0 in open-label extension); in placebo crossover patients, median annualized attack rate decreased from 10.7 to 1.4. Median annualized days of hemin use were 0.0 (double-blind period) and 0.0 (open-label extension) for continuous givosiran patients and reduced from 14.98 to 0.71 for placebo crossover patients. Long-term givosiran led to sustained lowering of delta-aminolevulinic acid and porphobilinogen and improvements in daily worst pain and quality of life. Safety findings were consistent with the double-blind period. CONCLUSIONS: Long-term givosiran has an acceptable safety profile and significantly benefits acute hepatic porphyria patients with recurrent attacks by reducing attack frequency, hemin use, and severity of daily worst pain while improving quality of life.


Subject(s)
Porphyria, Acute Intermittent , Porphyrias, Hepatic , Acetylgalactosamine/analogs & derivatives , Humans , Porphyria, Acute Intermittent/chemically induced , Porphyria, Acute Intermittent/drug therapy , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/drug therapy , Pyrrolidines , Quality of Life
2.
Article in English | MEDLINE | ID: mdl-32784669

ABSTRACT

BACKGROUND: Lead inhibits the enzymes in heme biosynthesis, mainly reducing δ-aminolevulinic acid dehydratase (ALAD) activity, which could be an available biomarker. The aim of this study was to detect the threshold of δ-aminolevulinic acid dehydratase activity reduced by lead exposure. METHODS: We collected data on 121 lead workers and 117 non-exposed workers when annual health examinations were performed. ALAD activity was determined by the standardized method of the European Community. ALAD G177C (rs1800435) genotyping was conducted using the polymerase chain reaction and restricted fragment length polymorphism (PCR-RFLP) method. In order to find a threshold effect, we used generalized additive models (GAMs) and scatter plots with smoothing curves, in addition to multiple regression methods. RESULTS: There were 229 ALAD1-1 homozygotes and 9 ALAD1-2 heterozygotes identified, and no ALAD2-2 homozygotes. Lead workers had significantly lower ALAD activity than non-exposed workers (41.6 ± 22.1 vs. 63.3 ± 14.0 U/L, p < 0.001). The results of multiple regressions showed that the blood lead level (BLL) was an important factor inversely associated with ALAD activity. The possible threshold of BLL affecting ALAD activity was around 5 µg/dL. CONCLUSIONS: ALAD activity was inhibited by blood lead at a possible threshold of 5 µg/dL, which suggests that ALAD activity could be used as an indicator for lead exposure regulation.


Subject(s)
Lead/blood , Occupational Exposure/adverse effects , Porphobilinogen Synthase/blood , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/genetics , Adult , Biomarkers/analysis , Genotype , Humans , Lead/toxicity , Male , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Porphobilinogen Synthase/genetics , Porphyrias, Hepatic/chemically induced , Risk Factors
3.
Med Hypotheses ; 131: 109314, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31443750

ABSTRACT

Metabolic targeting of liver 5-aminolevulinate synthase (5-ALAS) by inhibition of heme utilisation by tryptophan (Trp) 2,3-dioxygenase (TDO) or the use of tryptophan is proposed as a therapy of acute hepatic porphyrias. 5-ALAS, the rate-limiting enzyme of heme biosynthesis, is under negative feedback control by a small regulatory heme pool in the hepatic cytosol. Acute porphyric attacks, precipitated by fasting, certain hormones and some drugs, involve induction of 5-ALAS secondarily to depletion of the above pool, and the resultant elevation of 5-ALA levels initiates the abdominal and neurological symptoms of attacks. By utilising the regulatory heme, cytosolic TDO undermines the feedback control, thus allowing 5-ALAS induction to occur, e.g. upon glucocorticoid induction of TDO during fasting (starvation) and exogenous glucocorticoid administration. Currently, glucose therapy is the preferred strategy for reversing moderate attacks induced by fasting (calorie restriction), with more severe attacks being treated by intravenous heme preparations. Reversal of fasting-induced attacks by glucose is explained by the previously demonstrated reversal of increased heme utilisation by TDO. Inhibitors of this utilisation are therefore potential therapeutic targets in acute attacks and also for maintenance of a symptomless state. Existing TDO inhibitors other than glucose include allopurinol, nicotinamide and recently developed potent inhibitors such as LM10 used in cancer therapy. Based on studies in rats, the hypothesis predicts that the safety or otherwise of drugs in the hepatic porphyrias is determined by their ability to inhibit TDO utilisation of heme under basal conditions or after glucocorticoid induction or heme activation of TDO, in parallel with reciprocal changes in 5-ALAS induction. Tryptophan is also proposed as a potential therapy of acute attacks either alone or as an adjunct to the recently proposed 5-ALAS1 gene silencing. Trp increases heme biosynthesis by enhancing 5-ALA dehydratase activity and, based on a Trp-5-ALA model presented herein, Trp offers several advantages over heme therapy, namely rapid conversion of 5-ALA into heme, a greatly enhanced heme availability, a near complete inhibition of 5-ALAS induction, assumed rapid clearance of 5-ALA and hence accelerated resolution of symptoms of attacks, and finally provision of the neuroprotective metabolite kynurenic acid to neutralise the neurological symptoms. The hypothesis also addresses heme regulation in species lacking the TDO free apoenzyme and its glucocorticoid induction mechanism and proposes detailed assessment of heme biosynthesis in these species. Detailed proposals for testing the hypothesis are presented.


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Heme/metabolism , Molecular Targeted Therapy , Porphyrias, Hepatic/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/therapeutic use , 5-Aminolevulinate Synthetase/genetics , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Enzyme Induction/drug effects , Fasting/metabolism , Feedback, Physiological , Gene Silencing , Glucose/metabolism , Glucose/therapeutic use , Guinea Pigs , Heme/therapeutic use , Humans , Kynurenine/metabolism , Liver/drug effects , Liver/enzymology , Models, Biological , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/metabolism , Rodentia , Species Specificity , Tryptophan/adverse effects , Tryptophan/pharmacology
4.
Postgrad Med ; 126(7): 108-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25387219

ABSTRACT

The porphyrias are a group of metabolic diseases caused by inherited or acquired enzymatic deficiency in the metabolic pathway of heme biosynthesis. Simplistically, they can be considered as storage diseases, because the partial enzymatic defect gives rise to a metabolic "bottleneck" in the biosynthetic pathway and hence to an accumulation of different metabolic intermediates, potentially toxic and responsible for the various (cutaneous or neurovisceral) clinical manifestations observed in these diseases. In the acute porphyrias (acute intermittent porphyria, hereditary coproporphyria, variegate porphyria, and the very rare delta-aminolevulinic acid dehydratase ALAD-d porphyria), the characteristic severe neurovisceral involvement is mainly ascribed to a tissue accumulation of delta-aminolevulinic acid, a neurotoxic nonporphyrin precursor. Many different factors, both endogenous and exogenous, may favor the accumulation of this precursor in patients who are carriers of an enzymatic defect consistent with an acute porphyria, thus contributing to trigger the serious (and potentially fatal) clinical manifestations of the disease (acute porphyric attacks). To date, many different drugs are known to be able to precipitate an acute porphyric attack, so that the acute porphyrias are also considered as pharmacogenetic or toxygenetic diseases. This article reviews the different biochemical mechanisms underlying the capacity of many drugs to precipitate a porphyric acute attack (drug porphyrogenicity) in carriers of genetic mutations responsible for acute porphyrias, and addresses the issue of prescribing drugs for patients affected by these rare, but extremely complex, diseases.


Subject(s)
Porphyrias, Hepatic/chemically induced , Porphyrias/chemically induced , Carrier State , Cytochromes/metabolism , Heme/metabolism , Humans , Metabolism , Porphyrias/genetics , Porphyrias/physiopathology , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/physiopathology
5.
Toxicol Sci ; 141(2): 353-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24973095

ABSTRACT

A new antiepileptic synaptic vesicle 2a (SV2a) ligand drug candidate was tested in 4-week oral toxicity studies in rat and dog. Brown pigment inclusions were found in the liver of high-dose dogs. The morphology of the deposits and the accompanying liver changes (increased plasma liver enzymes, increased total hepatic porphyrin level, decreased liver ferrochelatase activity, combined induction, and inactivation of cytochrome P-450 CYP2B11) suggested disruption of the heme biosynthetic cascade. None of these changes was seen in rat although this species was exposed to higher parent drug levels. Toxicokinetic analysis and in vitro metabolism assays in hepatocytes showed that dog is more prone to oxidize the drug candidate than rat. Mass spectrometry analysis of liver samples from treated dogs revealed an N-alkylprotoporphyrin adduct. The elucidation of its chemical structure suggested that the drug transforms into a reactive metabolite which is structurally related to a known reference porphyrogenic agent allylisopropylacetamide. That particular metabolite, primarily produced in dog but neither in rat nor in human, has the potential to alkylate the prosthetic heme of CYP. Overall, the data suggested that the drug candidate should not be porphyrogenic in human. This case study further exemplifies the species variability in the susceptibility to drug-induced porphyria.


Subject(s)
Anticonvulsants/pharmacokinetics , Anticonvulsants/toxicity , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Porphyrias, Hepatic/chemically induced , Administration, Oral , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/blood , Aryl Hydrocarbon Hydroxylases/metabolism , Biotransformation , Cells, Cultured , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/diagnosis , Cytochrome P450 Family 2 , Dogs , Female , Ferrochelatase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Isoenzymes , Liver/enzymology , Liver/pathology , Male , Molecular Structure , Oxidation-Reduction , Porphyrias, Hepatic/blood , Porphyrias, Hepatic/diagnosis , Porphyrins/metabolism , Rats , Rats, Wistar , Risk Assessment , Species Specificity , Steroid Hydroxylases/metabolism
6.
In. Rodríguez Rivera, Luis. Para no cometer errores en la atención al paciente epiléptico. La Habana, Ecimed, 2013. .
Monography in Spanish | CUMED | ID: cum-55906
8.
Rev Esp Anestesiol Reanim ; 57(6): 395-6, 2010.
Article in Spanish | MEDLINE | ID: mdl-20645499
9.
Chem Res Toxicol ; 23(4): 712-23, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20099833

ABSTRACT

Many toxicological disorders, in common with numerous human diseases, are probably the consequence of multigene interactions with a variety of chemical and physiological factors. The importance of genetic factors may not be obvious initially from association studies because of their complexity and variable penetrance. The human disease, porphyria cutanea tarda (PCT), is a skin disease caused by the photosensitizing action of porphyrins arising secondary to the decreased activity of an enzyme of heme biosynthesis, uroporphyrinogen decarboxylase (UROD), in the liver. It is triggered by idiosyncratic hepatic interaction between genetic factors and chemicals such as alcohol, estrogenic drugs, and polyhalogenated aromatics. PCT and its animal models are known collectively as the hepatic uroporphyrias. There is strong evidence for the participation of iron in the pathogenesis of these conditions. Mouse models have been used to explore the relative importance of a variety of agents such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), alcohol, and iron in the development of uroporphyria and to elucidate the mechanism of the depression of hepatic UROD activity. Mutations of the UROD and hemochromatosis (HFE) genes are genetic factors in some PCT patients which can be mimicked in mice heterozygous for the Hfe and Urod null genes. Association studies of uroporphyria induced by TCDD or hexachlorobenzene with DNA markers in mouse intercrosses have shown the participation of other, unknown, genetic factors in addition to the strong influence of the Ahr gene. The pathogenesis of hepatic uroporphyrias exemplifies the complexity of the interactions between chemical and genetic factors that can contribute to the hepatotoxicity of chemicals.


Subject(s)
Porphyrias, Hepatic/genetics , Animals , Disease Models, Animal , Hemochromatosis/genetics , Humans , Iron/metabolism , Liver/enzymology , Mice , Photosensitizing Agents/toxicity , Polychlorinated Dibenzodioxins/toxicity , Porphyrias, Hepatic/chemically induced , Porphyrins/metabolism , Uroporphyrinogen Decarboxylase/genetics
10.
São Paulo; s.n; 2009. 219 p. ilus, tab, graf.
Thesis in Portuguese | LILACS | ID: lil-593589

ABSTRACT

Para otimizar urn modelo experimental para o estudo do desbalanço redox em porfirias relacionadas ao acúmulo de ácido 5-aminolevulinico-(ALA), via inibição da ALA desidratase-(ALA-D), ratos foram tratados com o éster metílico de succinilacetona-(SAME), um catabólito da tirosina que inibe fortemente a ALA-D, mimetizando o estado metabólico observado nos portadores de porfirias e tirosinemias. Estabeleceram-se modelos de tratamento agudo por 36 e 18 h. No primeiro, os animais receberam 3 injeções de SAME (10, 40 ou 80 mg/kg, grupos All-IV). No segundo, os animais receberam 3 injeções de 40 mg/kg de SAME, ALA ou éster metílico de ALA (grupos BII-IV), ALA:SAME (30:10 mg/kg, grupo BV), ou 10 mg/kg SAME (grupo BVI). Paralelamente, avaliou-se se os sintomas neurológicos característicos das porfirias decorriam de danos oxidativos mitocondriais. Para isso, aplicou-se uma tecnologia óptica para medidas da difusão da depressão cortical que determinou a oxigenação e o estado redox do cit c em mitocôndrias do córtex cerebral de ratos submetidos ao tratamento crônico com ALA (40 mg/kg), SAME (10 e 40 mg/kg) e ALA:SAME (30:10 mg/kg), a cada 48 h, durante 30 dias. Tratamento agudo/36 h: Os níveis de ALA no plasma, fígado, cérebro e urina e o clearance renal do ALA aumentaram nos grupos tratados. A atividade de ALA-D e a coproporfirina urinaria reduziram. A marcação para proteínas carboniladas, ferro e ferritina aumentou no fígado e cérebro dos grupos tratados, especialmente no All. Os níveis de malondialdeído hepática aumentaram no grupo AIV. A razão GSH/GSH+GSSG e a atividade de GPx cerebrais aumentaram nos grupos AIV e AIII, respectivamente. Consistentemente com estes dados indicando um desbalanço oxidativo induzido pelo SAME, alterações mitocondriais e citosólicas ultraestruturais foram reveladas, especialmente no fígado./Tratamento agudo/18 h: Os níveis de ALA plasmáticos aumentaram nos grupos tratados, exceto em BIV. 0 grupo Bll mostrou aumento dos níveis hepáticos...


To optimize an experimental model for studying redox imbalance in porphyrias related to 5-aminolevulinic acid (ALA) accumulation through the inhibition of ALA dehydratase (ALA-D), rats were treated with methyl ester of succinylacetone (SAME), a tyrosine catabolite that strongly inhibits ALA-D, what mimics the metabolic state observed in patients suffering from porphyrias and tyrosinemias. Models of acute treatment were established during 36 and 18 h. In the first model, animals received 3 injections of SAME (10, 40 or 80 mg/kg, groups All-IV). In the second model, animals received 3 injections of 40 mg/kg SAME, ALA or methyl ester of ALA (groups BII-IV), ALA:SAME (30:10 mg/kg, group BV), or 10 mg/kg SAME (group BVI). Concomitantly, we evaluated if the neurologic symptoms characteristics of porphyrias were a consequence of the oxidative mitochondria! impairment. For this, an optical technology for the measurement of cortical spreading depression was applied. This techonology determined the cerebral oxygenation and the redox state of cit c in mitochondria of the cerebral cortex of rats submitted to a chronic treatment with ALA (40 mg/kg), SAME (10 and 40 mg/kg) and ALA:SAME (30:10 mg/kg), alternate days, during 30 days. Acute treatment/36 h: ALA levels in plasma, liver and urine and clearance of renal ALA increased in treated groups. ALA-D activities and urinary coproporphyrin were found to be decreased. Liver and brain proteins carbonyl, iron and ferritin were higher in the liver of treated groups, especially in All. Liver j malondialdehyde levels were higher in group AIV. Cerebral GSH/GSH+GSSG ratio and GPx activities increased in groups AIV and AIII, respectively. Consistently with these data indicating SAME-induced oxidative imbalance, mitochondrial and cytosolic ultrastructural changes were revealed, especially in the liver. Acute treatment/18 h: Plasma ALA levels increased in all treated groups but BIV. Group BII showed increased hepatic ALA levels…


Subject(s)
Animals , Male , Young Adult , Rats , Aminolevulinic Acid/antagonists & inhibitors , Disease Models, Animal , Clinical Trial , Hydro-Lyases , Oxidative Stress , Porphyrias, Hepatic/chemically induced , Mitochondria , Porphyria, Acute Intermittent , Tyrosinemias
11.
J Toxicol Sci ; 33(5): 599-608, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19043281

ABSTRACT

Most patients with hepatic porphyria exhibit neuropsychiatric symptoms, including abdominal pain, peripheral neuropathy, confusion, insomnia and mental disturbances such as anxiety and depression. Although heme deficiency and accumulation of heme precursors are thought to be responsible for neuropsychiatric manifestations in patients with acute porphyria, the pathogenetic mechanisms remain poorly understood. In the present study, we observed psychiatric behaviors in mice with hepatic porphyria induced by the ingestion of a griseofulvin (GF)-containing diet over a period of 12 weeks. GF ingestion by the mice caused an accumulation of porphyrins in the feces and a decrease in heme in the liver; these effects were observed throughout the entire duration of the experiment, with maximum levels observed after circa 1 week of ingestion of this diet. In addition, the mice developed enlargement of the liver, hepatocyte injury, and cholestasis. Mice with hepatic porphyria manifested an anxiety-like behavior by the long-term treatment (over 5 weeks) in a GF-dose and duration dependent manner. The hepatic porphyria mice also manifested depression-like behaviors by the short-term treatment (3 weeks) of GF2.0, which was reversed by administration of anti-depressant, imipramine. In conclusion, this study for the first time demonstrated psychiatric manifestations in GF-induced hepatic porphyria mice. The present results suggest that model animals could be useful for elucidating the mechanisms underlying psychiatric manifestations in syndromes such as hepatic porphyria and hepatic encephalopathy that are associated with the impairment of hepatic function.


Subject(s)
Anxiety/psychology , Behavior, Animal , Depression/psychology , Griseofulvin/toxicity , Heme/biosynthesis , Porphyrias, Hepatic/psychology , Animals , Anxiety/etiology , Anxiety/metabolism , Behavior, Animal/drug effects , Cytochrome P-450 Enzyme System/metabolism , Depression/etiology , Depression/metabolism , Disease Models, Animal , Enzymes/blood , Feces/chemistry , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred ICR , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/complications , Porphyrias, Hepatic/metabolism , Swimming
12.
Toxicol Pathol ; 33(6): 720-5, 2005.
Article in English | MEDLINE | ID: mdl-16263697

ABSTRACT

As part of regulatory safety testing program, a 13-week oral toxicity study with a new antipsychotic drug candidate was performed in beagle dogs. During this study, dark red/brown feces were recorded in treated dogs and increases in liver parameters (alanine aminotransferase, alkaline phosphatase, bilirubin) were measured biochemically. At the end of the study, livers of high-dose (50 mg/kg) animals were (mottled) dark brown, sometimes with pale foci. Histopathological examination of these livers showed dark globular pigment deposits in the hepatocellular cytoplasm and within the bile canaliculi. Varying numbers of inflammatory cell infiltrates were additionally present in association with the deposits. These pigment deposits showed birefringency with characteristic "Maltese Cross"-like structures under polarized light. Electronmicroscopy revealed the typical, so-called "sunburst" pattern with radiating double-lined crystalline structures. These morphologic characteristics strongly indicated at the presence of porphyrins, which was definitely confirmed biochemically. Published reports of drug-induced hepatic porphyria in dogs are rare. The possible underlying mechanism in the dog and man is discussed.


Subject(s)
Antipsychotic Agents/toxicity , Liver/drug effects , Porphyrias, Hepatic/chemically induced , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Antipsychotic Agents/administration & dosage , Bilirubin/blood , Blood Coagulation/drug effects , Dogs , Dose-Response Relationship, Drug , Feces/chemistry , Female , Hyperplasia , Liver/enzymology , Liver/pathology , Male , Organ Size/drug effects , Partial Thromboplastin Time , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , Protoporphyrins/analysis , Protoporphyrins/metabolism , Thymus Gland/drug effects , Thymus Gland/pathology , Toxicity Tests
13.
Toxicol Appl Pharmacol ; 204(2): 187-95, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15808524

ABSTRACT

Hexaclorobenzene (HCB), one of the most persistent environmental pollutants, can cause a wide range of toxic effects including cancer in animals, and hepatotoxicity and porphyria both in humans and animals. In the present study, liver microsomal cytochrome P450 (CYP)-dependent arachidonic acid (AA) metabolism, hepatic PGE production, and cytosolic phospholipase A2 (cPLA2) activity were investigated in an experimental model of porphyria cutanea tarda induced by HCB. Female Wistar rats were treated with a single daily dose of HCB (100 mg kg(-1) body weight) for 5 days and were sacrificed 3, 10, 17, and 52 days after the last dose. HCB treatment induced the accumulation of hepatic porphyrins from day 17 and increased the activities of liver ethoxyresorufin O-deethylase (EROD), methoxyresorufin O-demethylase (MROD), and aminopyrine N-demethylase (APND) from day 3 after the last dose. Liver microsomes from control and HCB-treated rats generated, in the presence of NADPH, hydroxyeicosatetraenoic acids (HETEs), epoxyeicosatrienoic acids (EETs), 11,12-Di HETE, and omega-OH/omega-1-OH AA. HCB treatment caused an increase in total NADPH CYP-dependent AA metabolism, with a higher response at 3 days after the last HCB dose than at the other time points studied. In addition, HCB treatment markedly enhanced PGE production and release in liver slices. This HCB effect was time dependent and reached its highest level after 10 days. At this time cPLA2 activity was shown to be increased. Unexpectedly, HCB produced a significant decrease in cPLA2 activity on the 17th and 52nd day. Our results demonstrated for the first time that HCB induces both the cyclooxygenase and CYP-dependent AA metabolism. The effects of HCB on AA metabolism were previous to the onset of a marked porphyria and might contribute to different aspects of HCB-induced liver toxicity such as alterations of membrane fluidity and membrane-bound protein function. Observations also suggested that a possible role of cPLA2 in the early increase of AA metabolism cannot be excluded. However, the existence of other pathway(s) for metabolizable AA generation different from cPLA2 activation is also proposed.


Subject(s)
Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/metabolism , Hexachlorobenzene/adverse effects , Liver/chemistry , Liver/metabolism , Animals , Arachidonic Acid/chemistry , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Disease Models, Animal , Female , Hexachlorobenzene/administration & dosage , Hydroxyeicosatetraenoic Acids/biosynthesis , Intubation, Gastrointestinal , Methods , Microsomes, Liver/chemistry , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NADP/metabolism , Oxidoreductases/biosynthesis , Phospholipases A/metabolism , Phospholipases A2 , Porphyrias, Hepatic/chemically induced , Prostaglandins E/biosynthesis , Rats , Rats, Wistar , Time Factors
14.
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
15.
Environ Toxicol Chem ; 24(2): 450-6, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15720007

ABSTRACT

Tralkoxydim is the active ingredient in a postemergent herbicide used in cereal crops. During preregistration trials, tralkoxydim was observed to cause hepatic porphyria and cholestasis in laboratory mice. Porphyria was not seen in similarly exposed rats or hamsters, but data were not collected regarding the susceptibility of any wild small mammal species to the tralkoxydim-induced porphyria. To address this data gap, we exposed small mammals to tralkoxydim, to 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC; a known porphyrinogenic chemical), or to sunflower oil alone. We studied small mammal species that might be exposed following agricultural applications of the herbicide, including the white-footed mouse (Peromyscus leucopus), the deer mouse (P. maniculatus), and the meadow vole (Microtus pennsylvanicus). Because of their known susceptibility to both tralkoxydim- and DDC-induced porphyria, commercially supplied Mus musculus (CD-1 Swiss mice) were exposed as positive-control animals. We also exposed offspring of wild-caught M. musculus to compare their responses to those of the commercially supplied animals. Potential hepatotoxicity was determined by assessing the accumulation of liver protoporphyrin. Of the species tested, only M. musculus was susceptible to the porphyrinogenic action of tralkoxydim, and no significant accumulation of protoporphyrin was observed in any of the other species exposed to the herbicide.


Subject(s)
Cyclohexanones/toxicity , Herbicides/toxicity , Imines/toxicity , Peromyscus/physiology , Porphyrias, Hepatic/chemically induced , Animals , Dicarbethoxydihydrocollidine/toxicity , Liver/chemistry , Liver/metabolism , Mice , Plant Oils/toxicity , Protoporphyrins/analysis , Protoporphyrins/metabolism , Rats , Species Specificity , Sunflower Oil
16.
Epilepsia ; 45(5): 559-60, 2004 May.
Article in English | MEDLINE | ID: mdl-15101839

ABSTRACT

We report a patient with focal epilepsy and latent hereditary coproporphyria who had exacerbation of clinical symptoms of porphyria under treatment with valproate and primidone and was then treated with levetiracetam without exacerbation of clinically latent porphyria.


Subject(s)
Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Epilepsies, Partial/drug therapy , Piracetam/therapeutic use , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/drug therapy , Acute Disease , Adult , Comorbidity , Epilepsies, Partial/epidemiology , Female , Humans , Levetiracetam , Piracetam/analogs & derivatives , Porphyrias, Hepatic/epidemiology , Primidone/adverse effects , Primidone/therapeutic use , Valproic Acid/adverse effects , Valproic Acid/therapeutic use
17.
Toxicol Appl Pharmacol ; 189(1): 28-38, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12758057

ABSTRACT

Hepatic uroporphyria can be readily induced by a variety of treatments in mice of the C57BL strains, whereas DBA/2 mice are almost completely resistant. However, feeding of the protoporphyrinogen oxidase-inhibiting herbicide fomesafen (0.25% in the diet for 18 weeks) induced hepatic uroporphyria in male DBA/2N mice (liver porphyrin content up to 150 nmol/g, control animals 1 nmol/g), whereas fomesafen-treated male C57BL/6N mice displayed only a slight elevation of liver porphyrins (approximately 5 nmol/g). The profile of accumulated hepatic porphyrins in fomesafen-treated DBA/2N mice resembled the well-characterised uroporphyria induced by polyhalogenated aromatic hydrocarbons, while histological examination confirmed the presence of uroporphyria-specific cytoplasmic inclusions in the hepatocytes. Uroporphyrinogen decarboxylase activity decreased to about 30% of control values in fomesafen-treated DBA/2N mice; microsomal methoxyresorufin O-dealkylase activity was slightly reduced. The amount of CYP1A1 and CYP1A2 mRNA, as determined by real-time PCR, was not significantly changed; mRNA encoding the housekeeping 5-aminolevulinic acid synthase was elevated 10-fold. Total liver iron was slightly increased. A similar uroporphyria was induced by the herbicide formulation Blazer, containing a structurally related herbicide acifluorfen, when fed to DBA/2N mice at a dose corresponding to 0.25% of acifluorfen in the diet. Since DBA/2 mice are almost completely resistant to all well-characterised porphyrogenic chemicals, the results suggest the possible existence of a yet unknown mechanism of uroporphyria induction, to which the DBA/2 mouse strain is more sensitive than the C57BL strain.


Subject(s)
Benzamides/toxicity , Herbicides/toxicity , Phenyl Ethers/toxicity , Porphyrias, Hepatic/chemically induced , Uroporphyrins/biosynthesis , Animals , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Polymerase Chain Reaction , Porphyrias, Hepatic/enzymology , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , RNA, Messenger/biosynthesis , Species Specificity , Uroporphyrins/analysis
18.
Bol. Hosp. San Juan de Dios ; 50(2): 84-98, mar.-abr. 2003. tab
Article in Spanish | LILACS | ID: lil-348158

ABSTRACT

Las variedades agudas de porfirias suelen presentar episodios conocidos como crisis agudas, que pueden ser muy graves y aún fatales. La causa desencadenante más frecuente de estas crisis es el consumo de ciertos medicamentos. Así como algunos fármacos están identificados como productores de estas crisis, otros no tienen ese riesgo. Se puede así distinguir medicamentos sin peligro o "seguros", otros que conllevan el riesgo de desencadenar crisis o "no seguros" y otros en que la información es contradictoria. Esta publicación tiene por objeto facilitar, tanto a pacientes como a médicos tratantes, el listado de principios activos, incluyendo los fármacos que los contienen, ordenados según sean "seguros", "no seguros" y con "información contradictoria" y clasificados según acción farmacológica. A fin de facilitar aún más la selección de medicamentos a usar, se incorpora una segunda lista de productos farmacéuticos que se expenden en el mercado chileno, ordenados también según su riesgo de producir crisis agudas. El mercado de medicamentos es muy dinámico, constantemente están incorporándose nuevos y retirando otros que se dejan de usar, por lo que nuestra Unidad mantiene actualizadas estas listas en nuestra página Web (http://porfiria.med.uchile.cl)


Subject(s)
Humans , Drug-Related Side Effects and Adverse Reactions , Porphyrias , Porphyria Cutanea Tarda/chemically induced , Porphyria, Hepatoerythropoietic/chemically induced , Porphyrias, Hepatic/chemically induced , Porphyria, Acute Intermittent
19.
Z Gastroenterol ; 39(6): 453-5, 458, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11475000

ABSTRACT

Patients with porphyria cutanea tarda (PCT) develop hepatocellular carcinoma as a late consequence. Pre-loading of C57BL/10ScSn mice with iron greatly sensitizes them to the induction of hepatic porphyria caused by hexachlorobenzene (HCB). HCB will also cause liver tumors in experimental animals. Elevated liver iron stores are implicated in the development of some human liver cancers in connection with its known catalytic role in generation of highly reactive activated oxygen species. The aim of this study was to determine the lipid and DNA oxidative damage in iron and HCB-induced porphyric mice. C57BL/10ScSn mice received i.p. injections of dextran sulfate (control), iron (Imferon) or combined iron and HCB. 6 weeks after treatment plasma ALT levels and hepatic free iron, porphyrin, lipid peroxides and 8-hydroxyguanosine (8-OHdG) levels were analyzed. Hepatic porphyrin level was significantly (p < 0.001) increased following combined iron/HCB treatment as compared to control mice. The level of lipid peroxides increased 9-fold (p = 0.001) and 35-fold (p < 0.001) after iron and iron/HCB treatment respectively, whereas the level of 8-OHdG was increased 2.5-fold (p = 0.002) and 7.5-fold (p < 0.001) after iron and iron/HCB treatment respectively as compared to control mice. The authors conclude that iron overload in conjugation with HCB induce lipid and DNA oxidative damage in C57BL/10ScSn mice. DNA oxidative damage may be important in the early events of hepatic carcinogenesis in experimental porphyria.


Subject(s)
DNA Damage/physiology , Hemochromatosis/pathology , Lipid Peroxidation/physiology , Liver Neoplasms, Experimental/pathology , Porphyria Cutanea Tarda/pathology , Porphyrias, Hepatic/pathology , Animals , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Hemochromatosis/chemically induced , Hexachlorobenzene/toxicity , Iron-Dextran Complex/toxicity , Liver/drug effects , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred C57BL , Porphyria Cutanea Tarda/chemically induced , Porphyrias, Hepatic/chemically induced
20.
Toxicol Appl Pharmacol ; 173(2): 89-98, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11384210

ABSTRACT

The effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the liver of C57BL/6J mice is a model for clinical sporadic porphyria cutanea tarda (PCT). There is massive uroporphyria, inhibition of uroporphyrinogen decarboxylase (UROD) activity, and hepatocellular damage. A variety of evidence implicates the CYP1A2 enzyme as necessary for mouse uroporphyria. Here we report that, 5 weeks after a single oral dose of TCDD (75 microg/kg), Cyp1a2(+/+) wild-type mice showed severe uroporphyria and greater than 90% decreases in UROD activity; in contrast, despite exposure to this potent agent Cyp1a2(-/-) knockout mice displayed absolutely no increases in hepatic porphyrin levels, even after prior iron overload, and no detectable inhibition of UROD activity. Plasma levels of alanine-aminotransferase (ALT) and aspartate aminotransferase (AST)-although elevated in both genotypes after TCDD exposure-were significantly less in Cyp1a2(-/-) than in Cyp1a2(+/+) mice, suggesting that the absence of CYP1A2 also affords partial protection against TCDD-induced liver toxicity. Histological examination confirmed a decrease in hepatocellular damage in TCDD-treated Cyp1a2(-/-) mice; in particular, there was no bile duct damage or proliferation that in the Cyp1a2(+/+) mice might be caused by uroporphyrin. We conclude that CYP1A2 is both necessary and essential for the potent uroporphyrinogenic effects of TCDD in mice, and that CYP1A2 also plays a role in contributing to TCDD-induced hepatocellular injury. This study has implications for both the toxicity assessment of TCDD and the hepatic injury seen in PCT patients.


Subject(s)
Cytochrome P-450 CYP1A2/deficiency , Polychlorinated Dibenzodioxins/toxicity , Porphyrias, Hepatic/enzymology , Porphyrias, Hepatic/prevention & control , Uroporphyrins/urine , Animals , Atrophy/chemically induced , Crosses, Genetic , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Environmental Pollutants/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Porphyrias, Hepatic/chemically induced , Thymus Gland/drug effects , Thymus Gland/pathology , Uroporphyrinogen Decarboxylase/metabolism , Uroporphyrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...