Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(18)2021 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-34576284

RESUMO

5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX) fluorescence is widely used for the intraoperative detection of malignant tumors. However, the fluorescence emission profiles of the accompanying necrotic regions of these tumors have yet to be determined. To address this, we performed fluorescence and high-performance liquid chromatography (HPLC) analyses of necrotic tissues of squamous cancer after 5-ALA administration. In resected human lymph nodes of metastatic squamous cell carcinoma, we found a fluorescence peak at approximately 620 nm in necrotic lesions, which was distinct from the PpIX fluorescence peak at 635 nm for viable cancer lesions. Necrotic lesions obtained from a subcutaneous xenograft model of human B88 oral squamous cancer also emitted the characteristic fluorescence peak at 620 nm after light irradiation: the fluorescence intensity ratio (620 nm/635 nm) increased with the energy of the irradiation light. HPLC analysis revealed a high content ratio of uroporphyrin I (UPI)/total porphyrins in the necrotic cores of murine tumors, indicating that UPI is responsible for the 620 nm peak. UPI accumulation in necrotic tissues after 5-ALA administration was possibly due to the failure of the heme biosynthetic pathway. Taken together, fluorescence imaging of UPI after 5-ALA administration may be applicable for the evaluation of tumor necrosis.


Assuntos
Ácido Aminolevulínico/administração & dosagem , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Uroporfirinas/metabolismo , Idoso , Ácido Aminolevulínico/uso terapêutico , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Biológicos , Necrose , Espectrometria de Fluorescência
2.
Environ Microbiol ; 19(1): 106-118, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27486032

RESUMO

The sulfate-reducing bacteria of the Desulfovibrio genus make three distinct modified tetrapyrroles, haem, sirohaem and adenosylcobamide, where sirohydrochlorin acts as the last common biosynthetic intermediate along the branched tetrapyrrole pathway. Intriguingly, D. vulgaris encodes two sirohydrochlorin chelatases, CbiKP and CbiKC , that insert cobalt/iron into the tetrapyrrole macrocycle but are thought to be distinctly located in the periplasm and cytoplasm respectively. Fusing GFP onto the C-terminus of CbiKP confirmed that the protein is transported to the periplasm. The structure-function relationship of CbiKP was studied by constructing eleven site-directed mutants and determining their chelatase activities, oligomeric status and haem binding abilities. Residues His154 and His216 were identified as essential for metal-chelation of sirohydrochlorin. The tetrameric form of the protein is stabilized by Arg54 and Glu76, which form hydrogen bonds between two subunits. His96 is responsible for the binding of two haem groups within the main central cavity of the tetramer. Unexpectedly, CbiKP is shown to bind two additional haem groups through interaction with His103. Thus, although still retaining cobaltochelatase activity, the presence of His96 and His103 in CbiKP , which are absent from all other known bacterial cobaltochelatases, has evolved CbiKP a new function as a haem binding protein permitting it to act as a potential haem chaperone or transporter.


Assuntos
Proteínas de Bactérias/genética , Desulfovibrio vulgaris/enzimologia , Desulfovibrio vulgaris/genética , Heme/análogos & derivados , Liases/genética , Tetrapirróis/metabolismo , Uroporfirinas/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/genética , Desulfovibrio vulgaris/metabolismo , Ferroquelatase/genética , Ferroquelatase/metabolismo , Heme/metabolismo , Proteínas Ligantes de Grupo Heme , Hemeproteínas/genética , Histidina/metabolismo
3.
BMC Struct Biol ; 13: 10, 2013 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-23688113

RESUMO

BACKGROUND: In the anaerobic pathway of cobalamin (vitamin B12) synthesis, the CbiT enzyme plays two roles, as a cobalt-precorrin-7 C15-methyltransferase and a C12-decarboxylase, to produce the intermediate, cobalt-precorrin 8. RESULTS: The primary structure of the hypothetical protein MJ0391, from Methanocaldococcus jannaschii, suggested that MJ0391 is a putative CbiT. Here, we report the crystal structure of MJ0391, solved by the MAD procedure and refined to final R-factor and R-free values of 19.8 & 27.3%, respectively, at 2.3 Å resolution. The asymmetric unit contains two NCS molecules, and the intact tetramer generated by crystallographic symmetry may be functionally important. The overall tertiary structure and the tetrameric arrangements are highly homologous to those found in MT0146/CbiT from Methanobacterium thermoautotrophicum. CONCLUSIONS: The conservation of functional residues in the binding site for the co-factor, AdoMet, and in the putative precorrin-7 binding pocket suggested that MJ0391 may also possess CbiT activity. The putative function of MJ0391 is discussed, based on structural homology.


Assuntos
Proteínas Arqueais/química , Methanocaldococcus/enzimologia , Metiltransferases/química , Vitamina B 12/biossíntese , Sequência de Aminoácidos , Proteínas Arqueais/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Metiltransferases/metabolismo , Dados de Sequência Molecular , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Alinhamento de Sequência , Uroporfirinas/química , Uroporfirinas/metabolismo
4.
FEBS J ; 276(20): 5973-82, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19754882

RESUMO

Biosynthesis of heme d(1), the essential prosthetic group of the dissimilatory nitrite reductase cytochrome cd(1), requires the methylation of the tetrapyrrole precursor uroporphyrinogen III at positions C-2 and C-7. We produced Pseudomonas aeruginosa NirE, a putative S-adenosyl-L-methionine (SAM)-dependent uroporphyrinogen III methyltransferase, as a recombinant protein in Escherichia coli and purified it to apparent homogeneity by metal chelate and gel filtration chromatography. Analytical gel filtration of purified NirE indicated that the recombinant protein is a homodimer. NirE was shown to be a SAM-dependent uroporphyrinogen III methyltransferase that catalyzes the conversion of uroporphyrinogen III into precorrin-2 in vivo and in vitro. A specific activity of 316.8 nmol of precorrin-2 h(-1) x mg(-1) of NirE was found for the conversion of uroporphyrinogen III to precorrin-2. At high enzyme concentrations NirE catalyzed an overmethylation of uroporphyrinogen III, resulting in the formation of trimethylpyrrocorphin. Substrate inhibition was observed at uroporphyrinogen III concentrations above 17 microM. The protein did bind SAM, although not with the same avidity as reported for other SAM-dependent uroporphyrinogen III methyltransferases involved in siroheme and cobalamin biosynthesis. A P. aeruginosa nirE transposon mutant was not complemented by native cobA encoding the SAM-dependent uroporphyrinogen III methyltransferase involved in cobalamin formation. However, bacterial growth of the nirE mutant was observed when cobA was constitutively expressed by a complementing plasmid, underscoring the special requirement of NirE for heme d(1) biosynthesis.


Assuntos
Proteínas de Bactérias/metabolismo , Heme/análogos & derivados , Metiltransferases/química , Metiltransferases/metabolismo , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Teste de Complementação Genética , Heme/biossíntese , Metiltransferases/genética , Dados de Sequência Molecular , Ligação Proteica , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , S-Adenosilmetionina/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Uroporfirinogênios/metabolismo , Uroporfirinas/metabolismo
5.
Biometals ; 22(2): 345-51, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18956144

RESUMO

Accumulating evidence, including experiments using cytochrome P450 1a2 (Cyp1a2) gene knock-out mice (Cyp1a2(-/-)), indicates that the development of chemically induced porphyria requires the expression of CYP1A2. It has also been demonstrated that iron enhances and expedites the development of experimental uroporphyria, but that iron alone without CYP1A2 expression, as in Cyp1a2(-/-) mice, does not cause uroporphyria. The role of iron in the development of porphyria has not been elucidated. We examined the in vivo effect of iron deficiency on hepatic URO accumulation in experimental porphyria. Mice were fed diets containing low (iron-deficient diet (IDD), 8.5 mg iron/kg) or normal (normal diet (ND), 213.7 mg iron/kg) levels of iron. They were treated with 3-methylcholanthrene (MC), an archetypal inducer of CYP1A, and 5-aminolevulinate (ALA), precursors of porphyrin and heme. We found that uroporphyrin (URO) levels and uroporphyrinogen oxidation (UROX) activity were markedly increased in ND mice treated with MC and ALA, while the levels were not raised in IDD mice with the same treatments. CYP1A2 levels and methoxyresorufin O-demethylase (MROD) activities, the CYP1A2-mediated reaction, were markedly induced in the livers of both ND and IDD mice treated with MC and ALA. UROX activity, supposedly a CYP1A2-dependent activity, was not enhanced in iron-deficient mice in spite of the fact of induction of CYP1A2. We showed that a sufficient level of iron is essential for the development of porphyria and UROX activity.


Assuntos
Ácido Aminolevulínico/farmacologia , Ferro/metabolismo , Metilcolantreno/farmacologia , Porfirias/induzido quimicamente , Uroporfirinas/metabolismo , Animais , Citocromo P-450 CYP1A2/genética , Ferro/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/química , Porfirias/metabolismo , Fatores de Tempo , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo , Uroporfirinas/química
6.
Hepatology ; 45(1): 187-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17187429

RESUMO

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


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Ferro/farmacologia , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/metabolismo , Ácido Aminolevulínico , Animais , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ferro/metabolismo , Complexo Ferro-Dextran/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Bifenilos Policlorados , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/tratamento farmacológico
7.
J Bacteriol ; 188(24): 8666-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17028275

RESUMO

The methanogenic archaeon Methanosarcina barkeri synthesizes protoheme via precorrin-2, which is formed from uroporphyrinogen III in two consecutive methylation reactions utilizing S-adenosyl-L-methionine. The existence of this pathway, previously exclusively found in the sulfate-reducing delta-proteobacterium Desulfovibrio vulgaris, was demonstrated for M. barkeri via the incorporation of two methyl groups from methionine into protoheme.


Assuntos
Heme/biossíntese , Methanosarcina barkeri/metabolismo , Metionina/análogos & derivados , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Meios de Cultura , Methanosarcina barkeri/genética , Methanosarcina barkeri/crescimento & desenvolvimento , Metionina/metabolismo , Metilação , Uroporfirinogênios/metabolismo , Uroporfirinas/metabolismo
8.
Genomics ; 87(1): 84-92, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16314073

RESUMO

Congenital erythropoietic porphyria (CEP) is a recessive autosomal disorder characterized by a deficiency in uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme biosynthetic pathway. The severity of the disease, the lack of specific treatment except for allogeneic bone marrow transplantation, and the knowledge of the molecular lesions are strong arguments for gene therapy. An animal model of CEP has been designed to evaluate the feasibility of retroviral gene transfer in hematopoietic stem cells. We have previously demonstrated that the knockout of the Uros gene is lethal in mice (Uros(del) model). This work describes the achievement of a knock-in model, which reproduces a mutation of the UROS gene responsible for a severe UROS deficiency in humans (P248Q missense mutant). Homozygous mice display erythrodontia, moderate photosensitivity, hepatosplenomegaly, and hemolytic anemia. Uroporphyrin (99% type I isomer) accumulates in urine. Total porphyrins are increased in erythrocytes and feces, while Uros enzymatic activity is below 1% of the normal level in the different tissues analyzed. These pathological findings closely mimic the CEP disease in humans and demonstrate that the Uros(mut248) mouse represents a suitable model of the human disease for pathophysiological, pharmaceutical, and therapeutic purposes.


Assuntos
Substituição de Aminoácidos , Mutação de Sentido Incorreto , Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/genética , Animais , Transplante de Medula Óssea , Modelos Animais de Doenças , Terapia Genética , Camundongos , Camundongos Transgênicos , Porfiria Eritropoética/patologia , Porfiria Eritropoética/terapia , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinas/metabolismo
9.
Biochem Biophys Res Commun ; 331(1): 147-52, 2005 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15845371

RESUMO

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.


Assuntos
Citocromo P-450 CYP1A2/metabolismo , Poluentes Ambientais/toxicidade , Ferro/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Bifenilos Policlorados/toxicidade , Porfirias Hepáticas/induzido quimicamente , Animais , Citocromo P-450 CYP1A2/genética , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas Experimentais/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Porfirias Hepáticas/enzimologia , Porfirias Hepáticas/patologia , Ratos , Uroporfirinas/metabolismo
10.
Biochem Pharmacol ; 65(4): 545-50, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12566081

RESUMO

In experimental animals, CYP1A2 is absolutely required for the development of uroporphyria induced by treatment with polyhalogenated aromatic compounds or other compounds. Although the role of this CYP in clinical uroporphyria, porphyria cutanea tarda (PCT), is not clear, Cyp1a2(-/-) mice are resistant to the development of uroporphyria. Here, we compared the abilities of human and mouse CYP1A2 expressed in mouse hepatoma Hepa-1 cells to: (i) catalyze CYP1A2-dependent methoxyresorufin demethylase (MROD), and (ii) support uroporphyrin (URO) accumulation. Both CYP1A2 orthologs were expressed at similar levels as indicated by immunodetectable CYP1A2 proteins and MROD activities. URO accumulation was increased in cultures expressing either ortholog when supplemented with 5-aminolevulinic acid, the porphyrin precursor. Cells expressing mouse CYP1A2 produced more URO than cells expressing human CYP1A2. The results indicate that human CYP1A2 can support URO accumulation in hepatoma cells and thus may play a role in human PCT.


Assuntos
Carcinoma Hepatocelular/metabolismo , Citocromo P-450 CYP1A2/fisiologia , Porfiria Cutânea Tardia/enzimologia , Uroporfirinas/metabolismo , Ácido Aminolevulínico/farmacologia , Animais , Citocromo P-450 CYP1A2/biossíntese , Citocromo P-450 CYP1A2/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Immunoblotting , Camundongos , Oxirredutases/metabolismo , Porfiria Cutânea Tardia/metabolismo , Transfecção , Células Tumorais Cultivadas
11.
Hepatology ; 35(4): 912-21, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11915039

RESUMO

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


Assuntos
Citocromo P-450 CYP1A2/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Porfiria Cutânea Tardia/metabolismo , Ácido Aminolevulínico/farmacologia , Animais , Citocromo P-450 CYP1A2/genética , Limiar Diferencial , Modelos Animais de Doenças , Hepatócitos/metabolismo , Ferro/farmacologia , Células de Kupffer/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Bifenilos Policlorados , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/genética , Porfiria Cutânea Tardia/patologia , Distribuição Tecidual , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinas/metabolismo
12.
Electrophoresis ; 22(11): 2210-6, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11504054

RESUMO

The present work demonstrates that affinity capillary electrophoresis (ACE) can be employed as a valuable and powerful tool for studying the interactions between porphyrins and proteins in biological and biomedical research, such as the development of porphyrins and related compounds as efficient and selective photosensitizers in the photodynamic therapy of cancers. Binding constants of human serum albumin (HSA) to four biological porphyrins (uroporphyrin I, heptacarboxylporphyrin, coproporphyrin I, protoporphyrin IX), which possess a wide range of hydrophobicity, were estimated by ACE. Based on 1:1 molecular association between these individual porphyrins and HSA, the change of the electrophoretic mobility of HSA as a function of porphyrin concentration in the run buffer was measured and the binding constants were calculated from the slope of the Scatchard plots. The binding constant values were found to be 8.80 +/- 0.51 x 10(4) M(-1), 2.39 +/- 0.16 x 10(5) M(-1), 1.61 +/- 0.11 x 10(6) M(-1), and 9.34 +/- 0.30 x 10(6) M(-1) for uroporphyrin I, heptacarboxylporphyrin, coproporphyrin I, and protoporphyrin IX, respectively, and most of these results are in good agreement with those reported in the literature using conventional methods for binding measurements. Additionally, experimental binding constant data obtained using ACE was found to exhibit very good correlation with theoretical hydrophobicity values calculated using the Rekker's hydrophobic fragmental constant method, thus further supporting the hypothesis that the hydrophobicity of the porphyrin side chains play an important role in governing the hydrophobic interaction of porphyrins with serum proteins such as HSA.


Assuntos
Eletroforese Capilar/métodos , Porfirinas/metabolismo , Albumina Sérica/metabolismo , Marcadores de Afinidade , Coproporfirinas/metabolismo , Humanos , Técnicas In Vitro , Cinética , Ligação Proteica , Protoporfirinas/metabolismo , Uroporfirinas/metabolismo
13.
Prikl Biokhim Mikrobiol ; 37(6): 660-8, 2001.
Artigo em Russo | MEDLINE | ID: mdl-11771318

RESUMO

Methods of synthesis of coproporphyrin and uroporphyrin by using bacteria of the genus Arthrobacter are proposed. Metal complexes of coproporphyrin and uroporphyrin with Pt, Pd, and Zn were synthesized. Their structures were identified by spectrophotometry, IR spectrometry, 1H-NMR, mass spectrometry, and HPLC. Data showing the possibility to use coproporphyrin III-metal complexes as luminophores for fluorescence detection of tumors. The current and prospective uses of metal complexes of water-soluble natural porphyrins in advanced immunofluorescence assays are discussed.


Assuntos
Coproporfirinas , Corantes Fluorescentes , Metaloporfirinas , Neoplasias/química , Uroporfirinas , Animais , Arthrobacter/metabolismo , Cromatografia Líquida de Alta Pressão , Coproporfirinas/química , Coproporfirinas/metabolismo , Meios de Cultura , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Imunoensaio , Medições Luminescentes , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Metaloporfirinas/síntese química , Metaloporfirinas/química , Neoplasias/diagnóstico , Especificidade de Órgãos , Paládio , Platina , Propionibacterium/metabolismo , Uroporfirinas/química , Uroporfirinas/metabolismo , Zinco
14.
Biochem Biophys Res Commun ; 272(3): 946-50, 2000 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-10860855

RESUMO

In Saccharomyces cerevisiae, as in all eukaryotic organisms, delta-aminolevulinic acid (ALA) is a precursor of porphyrin biosynthesis, a very finely regulated pathway. ALA enters yeast cells through the gamma-aminobutyric acid (GABA) permease Uga4. The incorporation of a metabolite into the cells may be a limiting step for its intracellular metabolization. To determine the relationship between ALA transport and ALA metabolization, ALA incorporation was measured in yeast mutant strains deficient in the delta-aminolevulinic acid-synthase, uroporphyrinogen III decarboxylase, and ferrochelatase, three enzymes involved in porphyrin biosynthesis. Results presented here showed that neither intracellular ALA nor uroporphyrin or protoporphyrin regulates ALA incorporation, indicating that ALA uptake and its subsequent metabolization are not related to each other. Thus a key metabolite as it is, ALA does not have a transport system regulated according to its role.


Assuntos
Ácido Aminolevulínico/metabolismo , Transportadores de Ânions Orgânicos , Porfirinas/biossíntese , Porfirinas/metabolismo , Saccharomyces cerevisiae/metabolismo , 5-Aminolevulinato Sintetase/deficiência , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Ácido Aminolevulínico/farmacologia , Transporte Biológico , Ferroquelatase/genética , Ferroquelatase/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA , Genes Fúngicos/genética , Cinética , Proteínas de Membrana Transportadoras/metabolismo , Mutação/genética , Porfobilinogênio/metabolismo , Sintase do Porfobilinogênio/metabolismo , Protoporfiria Eritropoética , Protoporfirinas/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae , Uroporfirinogênio Descarboxilase/deficiência , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinas/metabolismo
15.
Toxicol Appl Pharmacol ; 162(1): 60-7, 2000 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10631128

RESUMO

Using Cyp1a2(-/-) mice we previously showed that CYP1A2 is absolutely required for hepatic uroporphyrin accumulation caused by iron and 5-aminolevulinate (ALA) treatment, both in the presence and absence of an inducer of CYP1A2. In this study we have used these mice to investigate whether CYP1A2 has an obligatory role in hepatic uroporphyria caused by hexachlorobenzene (HCBZ), an inducer of CYP2B and CYP3A, as well as CYP1A2. Here we treated mice with HCBZ and iron, with and without the porphyrin precursor, ALA, in the drinking water. In iron-loaded wild-type mice given a single dose of HCBZ and ALA, hepatic uroporphyrin (URO) accumulated to 300 nmol/g liver after 37 days, whereas in Cyp1a2(-/-) mice, there was no hepatic URO, even after an additional dose of HCBZ, and a further 29 days of ALA treatment. A similar requirement for CYP1A2 was found in uroporphyria produced in HCBZ and iron-treated mice in the absence of ALA. As detected by Western immunoblotting, HCBZ induced small increases in CYP2B and CYP3A in the livers of all animals. In the wild-type animals, HCBZ also induced CYP1A2 and associated enzyme activities, including uroporphyrinogen oxidation, by about 2-3-fold. In the Cyp1a2(-/-) mice, HCBZ did not increase hepatic microsomal uroporphyrinogen oxidation. These results indicate that, in mice, CYP1A2 is essential in the process leading to HCBZ-induced uroporphyria. Contributions by other CYP forms induced by HCBZ appear to be minimal.


Assuntos
Citocromo P-450 CYP1A2/fisiologia , Indução Enzimática/efeitos dos fármacos , Imidazóis/toxicidade , Ferro/toxicidade , Microssomos Hepáticos/metabolismo , Uroporfirinas/metabolismo , Ácido Aminolevulínico/farmacologia , Animais , Anticorpos/imunologia , Western Blotting , Doença Hepática Induzida por Substâncias e Drogas , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Fígado/química , Masculino , Metilcolantreno/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Oxazinas/metabolismo , Oxirredução , Fenobarbital/toxicidade , Fatores de Tempo , Uroporfirinogênios/metabolismo
16.
J Clin Gastroenterol ; 29(4): 327-31, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10599635

RESUMO

A strong association between hepatitis C virus (HCV) infection and porphyria cutanea tarda (PCT) has been observed, but the implications of the viral infection in the metabolism of porphyrins in patients without clinical manifestations of PCT are not known. The levels of porphyrin in plasma and uroporphyrin (URO) and coproporphyrin (COPRO) in 24-hour urine were measured in 156 patients with chronic HCV infection showing no clinical evidence of PCT. Levels of URO higher than the upper limit were observed in 35 of 156 patients (22.4%). The range and the mean values +/- standard deviation were 26-1,196 microg/24 hours and 82 +/- 204 microg/24 hours. Increased levels of COPRO and plasma porphyrin were observed in 12 of 156 patients (7.7%) and 2 of 156 patients (1.3%) respectively. There were no differences between patients with increased URO levels and patients with normal URO levels in terms of gender, age, risk factors for HCV infection, alcohol abuse, or hepatitis B viral infection. Transferrin saturation (p = 0.040), gamma glutamyl transpeptidase (p < 0.0001), aspartate aminotransferase (p = 0.006), and alanine aminotransferase (p = 0.040) were significantly higher in patients with abnormal URO than in patients with normal URO. The frequency of cirrhosis was higher, but not significantly different, in patients with increased URO (16.7%) compared with patients with normal URO (3.8%). The authors demonstrated that even without a clinical manifestation of PCT it is possible to detect abnormalities in the metabolism of porphyrins in patients with chronic HCV infection. The implications of these findings deserve additional investigation.


Assuntos
Hepatite C Crônica/urina , Porfiria Cutânea Tardia/urina , Porfirinas/metabolismo , Uroporfirinas/urina , Adolescente , Adulto , Idoso , Biópsia , Feminino , Hepatite C Crônica/complicações , Hepatite C Crônica/metabolismo , Hepatite C Crônica/patologia , Humanos , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Porfiria Cutânea Tardia/complicações , Porfiria Cutânea Tardia/metabolismo , Pele/química , Pele/patologia , Uroporfirinas/metabolismo
17.
Biochem Pharmacol ; 58(2): 375-82, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10423181

RESUMO

Previous work has implicated CYP1A2 in experimental uroporphyria caused by polyhalogenated aromatic compounds, and in uroporphyria caused by iron and 5-aminolevulinate (ALA) in the absence of inducers of CYP1A2. Here we examined whether the different susceptibilities of SWR and C57BL/6 strains of mice to uroporphyria in the absence of inducers of CYP1A2 are related to different levels of CYP1A2. Enzymological assays (ethoxy- and methoxyresorufin dealkylases, and uroporphyrinogen oxidation) and immunoblots indicated that there was about twice the amount of hepatic CYP1A2 in SWR mice compared with C57BL/6 mice. Immunohistochemistry revealed that CYP1A2 was located centrilobularly in the liver, and the staining was more intense in SWR mice than in C57BL/6 mice. Hepatic non-heme iron was about double in SWR compared with C57BL/6 mice. In SWR mice given iron dextran, hepatic iron was 1.7-fold that of C57BL/6 mice given iron dextran. SWR mice administered ALA in the drinking water accumulated much less hepatic protoporphyrin than did C57BL/6 mice. To confirm the importance of small increases in CYP1A2, C57BL/6 mice were given a low dose of 3-methylcholanthrene (MC) (15 mg/kg), as well as iron and ALA. There was about a 5- to 6-fold increase in hepatic uroporphyrin accumulation after 32 days on ALA compared with animals not given MC. In these animals, CYP1A2 was increased by 10-fold at 2 days, but returned to basal levels by 14 days. We conclude that small and transient differences in CYP1A2 may be important in the development of uroporphyria.


Assuntos
Ácido Aminolevulínico/farmacologia , Citocromo P-450 CYP1A2/metabolismo , Ferro/farmacologia , Fígado/enzimologia , Uroporfirinas/urina , Animais , Citocromo P-450 CYP1A2/análise , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade da Espécie , Uroporfirinas/metabolismo
18.
Pharmacogenetics ; 8(6): 485-94, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9918132

RESUMO

Genetic susceptibility to toxic chemicals is of major importance but most studies concentrate on candidate genes and searches for unknown susceptibility genes are uncommon. Human sporadic porphyria cutanea tarda is usually precipitated by alcohol, oestrogens, hepatitis viruses, HIV or haemodialysis. The mechanism is not known but there is a role for iron metabolism and an underlying genetic predisposition is suspected. A similar porphyria in humans has also been caused by hexachlorobenzene. These human porphyrias can be modelled in iron-loaded mice exposed to hexachlorobenzene, in which C57BL/10ScSn is a prototype susceptible strain whereas DBA/2 mice are extremely resistant. A search for susceptibility genes was undertaken using complex trait analysis with DNA microsatellite markers of 'high' and 'low' responders from an F2 intercross. Correlation of markers with susceptibility, defined as accumulation of uroporphyrin in the liver, was assessed by chi-squared test for the proportion of C57BL/10ScSn and DBA/2 alleles present. Susceptibility loci on chromosomes 12, 14 and 17 were identified. Further analysis of markers on chromosomes 14 and 17 by MAPMAKER/EXP and MAPMAKER/QTL gave LOD scores of 7.3 and 3.6, respectively. Typing of chromosome 12 for the Ahr gene, using a restriction fragment length polymorphism distinguishing between the b-1 and d alleles, gave significant but not perfect linkage. However, no strong association between alleles or expression of Cyp1a1/2 genes, regulated by Ahr, and susceptibility for porphyria was detected. The results demonstrate that the porphyria induced by hexachlorobenzene in C57BL/10ScSn mice is a complex trait determined by at least three genes, which may be of relevance to susceptibility in the development of sporadic porphyria cutanea tarda and unknown aspects of liver damage.


Assuntos
Mapeamento Cromossômico , Ligação Genética , Hexaclorobenzeno/toxicidade , Ferro/toxicidade , Porfiria Cutânea Tardia/genética , Animais , Sequência de Bases , Primers do DNA , Sinergismo Farmacológico , Marcadores Genéticos , Predisposição Genética para Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/metabolismo , Especificidade da Espécie , Uroporfirinas/biossíntese , Uroporfirinas/metabolismo
19.
Chem Biol ; 4(6): 445-51, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9224567

RESUMO

BACKGROUND: In order to study the biosynthesis of vitamin B12, it is necessary to produce various intermediates along the biosynthetic pathway by enzymic methods. Recently, information on the organisation of the biosynthetic pathway has permitted the selection of the set of enzymes needed to biosynthesise any specific identified intermediate. The aim of the present work was to use recombinant enzymes in reconstituted multi-enzyme systems to biosynthesise particular intermediates. RESULTS: The products of the cobG and cobJ genes from Pseudomonas denitrificans were expressed heterologously in Escherichia coli to afford good levels of activity of the corresponding enzymes, CobG and CobJ. Aerobic incubation of precorrin-3A with the CobG enzyme alone yielded precorrin-3B. When CobJ and S-adenosyl-L-methionine were included in the incubation, the product was precorrin-4. Both precorrin 3B and precorrin-4 are known precursors of vitamin B12 and their availability has allowed new mechanistic studies of enzymic transformations. CONCLUSIONS: Our results show that the expression of the CobG and CobJ enzymes has been successful, thus facilitating the biosynthesis of two precursors of vitamin B12. This lays the foundation for the structure determination of CobG and CobJ as well as future enzymic experiments focusing on later steps of vitamin B12 biosynthesis.


Assuntos
Proteínas de Bactérias , Metiltransferases/metabolismo , Complexos Multienzimáticos/metabolismo , Oxigenases/metabolismo , Pseudomonas/enzimologia , Uroporfirinas/biossíntese , Vitamina B 12/biossíntese , Clonagem Molecular , Escherichia coli/genética , Expressão Gênica , Genes Bacterianos , Metiltransferases/genética , Estrutura Molecular , Complexos Multienzimáticos/genética , Mutagênese Sítio-Dirigida/genética , Oxigenases/genética , Plasmídeos , Proteínas Recombinantes/metabolismo , Uroporfirinas/genética , Uroporfirinas/metabolismo
20.
Toxicol Appl Pharmacol ; 136(1): 79-86, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8560483

RESUMO

Exposure of iron-loaded C57BL/10ScSn mice to the polychlorinated biphenyls (PCBs) mixture Aroclor 1254 in the diet (0.01%) for 5 weeks caused massive hepatic porphyria far greater than occurred with PCBs alone. This regime eventually causes hepatocellular carcinoma. Hepatic microsomal ethoxy-, pentoxy-, and benzyloxyresorufin dealkylase activities (respectively EROD, PROD, and BROD) catalyzed primarily by cytochrome P4501A1 and 2B isoenzymes were markedly induced after 2 weeks of diet (when no porphyria had developed) but showed little effect of iron. EROD activity in the nuclear membrane was also induced by the PCBs as was CYP1A1 protein when shown by immunoblotting. Nuclear dealkylase activities of PCBs-treated mice were considerably less than microsomal activities but were stimulated by iron pretreatment. The mechanism of the iron-enhanced toxicity may be due to oxidative damage associated with chronic induction of CYP1A1 isoforms. Lucigenin-enhanced chemiluminescence (CL) by microsomes and nuclear membranes was used as a method to estimate their potential to form reactive oxygen species. Despite CL being induced by PCBs it was less with microsomes from iron-treated mice. In a comparison of a variety of inducers of microsomal cytochrome P450 there was no correlation between inducer, uroporphyrogenic agent, and intensity of CL. On the other hand, cytosolic glutathione S-transferase (GST) activities with 1-chloro-2,4-dinitrobenzene and 1,2-dichloro-4-nitrobenzene (DCNB) as substrates, were also induced by the PCBs mixture, the induction with DCNB being synergistically potentiated by iron pretreatment. Complementary results were observed by immunocytochemistry using anti alpha-GST antibody. In contrast, total glutathione peroxidase activity and selenium-dependent glutathione peroxidase activity were depressed by PCBs but particularly in mice also administered iron. The results illustrate that PCBs not only induce CYP1A1 in microsomes but also in the nuclear membrane, which may be of significance in the mechanism of the iron-enhanced carcinogenicity of these chemicals. The iron-enhanced induction of GST with accompanying depletion of glutathione peroxidase provides evidence for oxidative processes induced in vivo by the PCBs.


Assuntos
Arocloros/toxicidade , Carcinógenos/toxicidade , Poluentes Ambientais/toxicidade , Ferro/farmacologia , Fígado/efeitos dos fármacos , Animais , Arocloros/administração & dosagem , Arocloros/metabolismo , Carcinógenos/administração & dosagem , Carcinógenos/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Citocromo P-450 CYP1A1 , Sistema Enzimático do Citocromo P-450/metabolismo , Citosol/efeitos dos fármacos , Citosol/enzimologia , Citosol/metabolismo , Sinergismo Farmacológico , Eletroforese em Gel de Poliacrilamida , Poluentes Ambientais/administração & dosagem , Indução Enzimática/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Glutationa Transferase/metabolismo , Immunoblotting , Ferro/metabolismo , Isoenzimas , Fígado/enzimologia , Fígado/ultraestrutura , Medições Luminescentes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Nitrobenzenos/administração & dosagem , Nitrobenzenos/toxicidade , Oxirredutases/metabolismo , Selênio/farmacologia , Uroporfirinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA