Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Photochem Photobiol B ; 199: 111585, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31450131

RESUMO

Thiosemicarbazone derivatives are known for their broad biological activity including their antitumor potency. The aim of the current study was to examine the effect of a novel series of non-toxic iron chelators on the accumulation of protoporphyrin IX after external 5-aminolevulonic acid administration. From this series we selected one the most promising derivative which causes a pronounced increase in the concentration of protoporphyrin IX. The increase of the photosensitizer concentration is necessary for the trigger the efficient therapeutic effect of the photodynamic reaction. For selected compound 2 we performed an examination of a panel of the genes that are involved in the heme biosynthesis and degradation. Results indicated the crucial roles of ferrochelatase and heme oxygenase in the described processes. Surprisingly, there was a strict dependence on the type of the tested cell line. A decrease in the expression of the two aforementioned enzymes after incubation with compound 2 and 5-aminolevulonic acid is a commonly known fact and we detected this trend for the MCF-7 and HCT 116 cell lines. However, we noticed the upregulation of the tested targets for the Hs683 cells. These unconventional results prompted us to do a more in-depth analysis of the described processes. In conclusion, we found that compound 2 is a novel, highly effective booster of photodynamic therapy that has prospective applications.


Assuntos
Antineoplásicos/síntese química , Ferro/química , Fármacos Fotossensibilizantes/síntese química , Protoporfirinas/química , Tiossemicarbazonas/metabolismo , Células A549 , Ácido Aminolevulínico/química , Ácido Aminolevulínico/metabolismo , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Ferroquelatase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Células MCF-7 , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/farmacologia , Tiossemicarbazonas/síntese química
2.
J Biol Chem ; 291(33): 17417-26, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27317660

RESUMO

The cellular transport of the cofactor heme and its biosynthetic intermediates such as protoporphyrin IX is a complex and highly coordinated process. To investigate the molecular details of this trafficking pathway, we created a synthetic lesion in the heme biosynthetic pathway by deleting the gene HEM15 encoding the enzyme ferrochelatase in S. cerevisiae and performed a genetic suppressor screen. Cells lacking Hem15 are respiratory-defective because of an inefficient heme delivery to the mitochondria. Thus, the biogenesis of mitochondrial cytochromes is negatively affected. The suppressor screen resulted in the isolation of respiratory-competent colonies containing two distinct missense mutations in Nce102, a protein that localizes to plasma membrane invaginations designated as eisosomes. The presence of the Nce102 mutant alleles enabled formation of the mitochondrial respiratory complexes and respiratory growth in hem15Δ cells cultured in supplemental hemin. Respiratory function in hem15Δ cells can also be restored by the presence of a heterologous plasma membrane heme permease (HRG-4), but the mode of suppression mediated by the Nce102 mutant is more efficient. Attenuation of the endocytic pathway through deletion of the gene END3 impaired the Nce102-mediated rescue, suggesting that the Nce102 mutants lead to suppression through the yeast endocytic pathway.


Assuntos
Endossomos/metabolismo , Heme/metabolismo , Mitocôndrias/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transporte Biológico Ativo/fisiologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Endossomos/genética , Ferroquelatase/genética , Ferroquelatase/metabolismo , Heme/genética , Mitocôndrias/genética , Mutação de Sentido Incorreto , Consumo de Oxigênio/fisiologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
3.
Toxicol Sci ; 141(2): 353-64, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24973095

RESUMO

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.


Assuntos
Anticonvulsivantes/farmacocinética , Anticonvulsivantes/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fígado/efeitos dos fármacos , Porfirias Hepáticas/induzido quimicamente , Administração Oral , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/sangue , Hidrocarboneto de Aril Hidroxilases/metabolismo , Biotransformação , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Família 2 do Citocromo P450 , Cães , Feminino , Ferroquelatase/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Isoenzimas , Fígado/enzimologia , Fígado/patologia , Masculino , Estrutura Molecular , Oxirredução , Porfirias Hepáticas/sangue , Porfirias Hepáticas/diagnóstico , Porfirinas/metabolismo , Ratos , Ratos Wistar , Medição de Risco , Especificidade da Espécie , Esteroide Hidroxilases/metabolismo
4.
Appl Microbiol Biotechnol ; 97(22): 9773-85, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24113826

RESUMO

Heme is a suggested limiting factor in peroxidase production by Aspergillus spp., which are well-known suitable hosts for heterologous protein production. In this study, the role of genes coding for coproporphyrinogen III oxidase (hemF) and ferrochelatase (hemH) was analyzed by means of deletion and overexpression to obtain more insight in fungal heme biosynthesis and regulation. These enzymes represent steps in the heme biosynthetic pathway downstream of the siroheme branch and are suggested to play a role in regulation of the pathway. Based on genome mining, both enzymes deviate in cellular localization and protein domain structure from their Saccharomyces cerevisiae counterparts. The lethal phenotype of deletion of hemF or hemH could be remediated by heme supplementation confirming that Aspergillus niger is capable of hemin uptake. Nevertheless, both gene deletion mutants showed an extremely impaired growth even with hemin supplementation which could be slightly improved by media modifications and the use of hemoglobin as heme source. The hyphae of the mutant strains displayed pinkish coloration and red autofluorescence under UV indicative of cellular porphyrin accumulation. HPLC analysis confirmed accumulation of specific porphyrins, thereby confirming the function of the two proteins in heme biosynthesis. Overexpression of hemH, but not hemF or the aminolevulinic acid synthase encoding hemA, modestly increased the cellular heme content, which was apparently insufficient to increase activity of endogenous peroxidase and cytochrome P450 enzyme activities. Overexpression of all three genes increased the cellular accumulation of porphyrin intermediates suggesting regulatory mechanisms operating in the final steps of the fungal heme biosynthesis pathway.


Assuntos
Aspergillus niger/enzimologia , Aspergillus niger/metabolismo , Vias Biossintéticas/genética , Coproporfirinogênio Oxidase/metabolismo , Ferroquelatase/metabolismo , Heme/biossíntese , Aspergillus niger/genética , Aspergillus niger/crescimento & desenvolvimento , Coproporfirinogênio Oxidase/genética , Ferroquelatase/genética , Deleção de Genes , Expressão Gênica , Regulação Fúngica da Expressão Gênica , Genômica , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética
5.
PLoS Pathog ; 9(8): e1003522, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935500

RESUMO

Heme metabolism is central to malaria parasite biology. The parasite acquires heme from host hemoglobin in the intraerythrocytic stages and stores it as hemozoin to prevent free heme toxicity. The parasite can also synthesize heme de novo, and all the enzymes in the pathway are characterized. To study the role of the dual heme sources in malaria parasite growth and development, we knocked out the first enzyme, δ-aminolevulinate synthase (ALAS), and the last enzyme, ferrochelatase (FC), in the heme-biosynthetic pathway of Plasmodium berghei (Pb). The wild-type and knockout (KO) parasites had similar intraerythrocytic growth patterns in mice. We carried out in vitro radiolabeling of heme in Pb-infected mouse reticulocytes and Plasmodium falciparum-infected human RBCs using [4-(14)C] aminolevulinic acid (ALA). We found that the parasites incorporated both host hemoglobin-heme and parasite-synthesized heme into hemozoin and mitochondrial cytochromes. The similar fates of the two heme sources suggest that they may serve as backup mechanisms to provide heme in the intraerythrocytic stages. Nevertheless, the de novo pathway is absolutely essential for parasite development in the mosquito and liver stages. PbKO parasites formed drastically reduced oocysts and did not form sporozoites in the salivary glands. Oocyst production in PbALASKO parasites recovered when mosquitoes received an ALA supplement. PbALASKO sporozoites could infect mice only when the mice received an ALA supplement. Our results indicate the potential for new therapeutic interventions targeting the heme-biosynthetic pathway in the parasite during the mosquito and liver stages.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Anopheles/parasitologia , Ferroquelatase/metabolismo , Heme/biossíntese , Fígado/parasitologia , Malária Falciparum/enzimologia , Plasmodium berghei/enzimologia , Plasmodium falciparum/enzimologia , 5-Aminolevulinato Sintetase/genética , Animais , Ferroquelatase/genética , Heme/genética , Hemeproteínas/biossíntese , Hemeproteínas/genética , Humanos , Fígado/patologia , Malária Falciparum/genética , Camundongos , Oocistos/enzimologia , Plasmodium berghei/genética , Plasmodium falciparum/genética , Esporozoítos/enzimologia
6.
Exp Hematol ; 39(7): 784-94, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21627978

RESUMO

OBJECTIVE: Most patients with erythropoietic protoporphyria have deficient ferrochelatase (FECH) activity due to changes in FECH DNA. We evaluated seven patients with erythropoietic protoporphyria phenotype in whom abnormalities of FECH DNA were not found by conventional analysis. The major focus was mitoferrin-1 (MFRN1), the mitochondrial transporter of Fe used for heme formation by FECH and for 2Fe2S cluster synthesis, which is critical to FECH activity/stability. MATERIALS AND METHODS: Four patients had a deletion in ALAS2 that causes enzyme gain-of-function, resulting in increased formation of protoporphyrin; one had a heterozygous major deletion in FECH DNA. All had an abnormal transcript of MFRN1 in messenger RNA extracted from blood leukocytes and/or liver tissue. The abnormal transcript contained an insert of intron 2 that had a stop codon. The consequences of abnormal MFRN1 expression were examined using zebrafish and yeast MFRN-deficient strains and cultured lymphoblasts from the patients. RESULTS: Abnormal human MFRN1 complementary DNA showed loss-of-function in zebrafish and yeast mutants, whereas normal human MFRN1 complementary DNA rescued both. Using cultured lymphoblasts, quantitative reverse transcription polymerase chain reaction showed increased formation of abnormal transcript that was accompanied by decreased formation of normal transcript and reduced FECH activity in patients compared to normal lines. A positive correlation coefficient (0.75) was found between FECH activity and normal MFRN1 messenger RNA in lymphoblasts. However, no obvious cause for increased formation of abnormal transcript was identified in MFRN1 exons and splice junctions. CONCLUSIONS: Abnormal MFRN1 expression can contribute to erythropoietic protoporphyria phenotype in some patients, probably by causing a reduction in FECH activity.


Assuntos
Proteínas de Transporte de Cátions/genética , Ferroquelatase/genética , Expressão Gênica , Proteínas Mitocondriais/genética , Protoporfiria Eritropoética/genética , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Adolescente , Adulto , Idoso , Animais , Sequência de Bases , Células COS , Proteínas de Transporte de Cátions/metabolismo , Criança , Chlorocebus aethiops , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Feminino , Ferroquelatase/metabolismo , Teste de Complementação Genética , Humanos , Células K562 , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Mutação , Protoporfiria Eritropoética/metabolismo , Protoporfiria Eritropoética/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Xenopus/embriologia , Xenopus/genética , Leveduras/genética , Leveduras/crescimento & desenvolvimento , Adulto Jovem
7.
Biosci Biotechnol Biochem ; 74(7): 1415-20, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20622448

RESUMO

At the terminal step of heme biosynthesis, ferrochelatase (FECH) catalyzes the insertion of Fe2+ into protoporphyrin to form heme. It is located on the inner membrane of the mitochondria of animals. The enzyme inserts divalent metal ions, including Fe2+, Co2+, and Zn2+, into porphyrins in vitro. We have reported that it can remove Fe2+ from heme. To characterize the iron-removal reverse activity of FECH, we examined its properties in porcine liver and muscle mitochondria, and isolated porcine FECH cDNA. The amino acid sequence of porcine FECH showed high homology with bovine (91%), human (85%), mouse (87%), and rat (76%) equivalents. It was expressed in Escherichia coli, and purified, and the kinetic properties of the zinc-chelating and iron-removal activities were examined. Both activities peaked at 45 degrees C, but different optimal pH values, of 7.5-8.0 for zinc-ion insertion and 5.5-6.0 for the reverse reaction were found. The K(m) values for mesoporphyrin IX and Zn2+ were 6.6 and 1.1 microM, respectively, and the K(m) for heme was 5.7 microM. The k(cat) value of the forward reaction was about 11-fold higher than that of the reverse reaction, indicating that the enzyme preferably catalyzes the forward reaction rather than the iron-removal reaction. Reverse activity was stimulated by fatty acids and phospholipids, similarly to the case of the forward reaction, indicating that lipids play a role in regulating both enzyme activities.


Assuntos
Ferroquelatase/metabolismo , Heme/metabolismo , Ferro/metabolismo , Protoporfirinas/metabolismo , Suínos , Sequência de Aminoácidos , Animais , Biocatálise , Bovinos , Clonagem Molecular , DNA Complementar/genética , Ferroquelatase/química , Ferroquelatase/genética , Ferroquelatase/isolamento & purificação , Humanos , Cinética , Fígado/enzimologia , Camundongos , Mitocôndrias/enzimologia , Dados de Sequência Molecular , Músculos/citologia , Ratos
8.
Artigo em Chinês | MEDLINE | ID: mdl-21158025

RESUMO

AIM: To investigate the possible role of rate-limiting enzyme of heme metabolism and globin in the development of the low hemoglobin (Hb), red blood (cell) count (RBC) and hematocrit (Hct) after long-term exercise, and effect of nutrition supplement on sports anemia. METHODS: Male Wistar rats were randomly assigned to three groups (n = 10): control (C), exercise (P) and exercise + nutrition (G). Animals in the P and G groups started treadmill running at 30 m/min, 0% grade, 1 min/time. Running time was gradually increased with 2 min/time during initial 5 weeks and final 4 weeks. In addition, running frequency was 2 times/day except initial 2 weeks. At the end of eleventh week, gene expression of 5-aminolevulinate synthase (ALAS), ferrochelatase, alpha-globin and beta-globin in bone marrow were measured with RT-PCR. Mean-while heme oxygenase 1 (HO-1) activity in liver was measured with immunohistochemical method. RESULTS: Eleven weeks of exercise induced a significant increase in HO-1 and a significant increase in gene expression of beta-globin (P < 0.01, P < 0.05, respectively). Treatment with anti-sports anemia compound dosage led to no significant differences in rate-limiting enzyme of heme metabolism and globin in the exercised rats. The G group had a significantly higher HO-1 level in liver than the C group (P < 0.01). These finds showed that exercise was associated with no significant difference in heme synthetase and alpha-globin gene expression, and significant difference in heme catabolic enzyme and beta-globin gene expression. CONCLUSION: The increase of HO-1 activity in liver might be one of the causes of the lower Hb, RBC and Hct status in exercised rats.


Assuntos
Anemia/etiologia , Suplementos Nutricionais , Regulação Enzimológica da Expressão Gênica/fisiologia , Heme Oxigenase (Desciclizante)/metabolismo , Condicionamento Físico Animal/efeitos adversos , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Anemia/metabolismo , Anemia/fisiopatologia , Animais , Ferroquelatase/genética , Ferroquelatase/metabolismo , Globinas/metabolismo , Heme Oxigenase (Desciclizante)/genética , Hidroximetilbilano Sintase/genética , Hidroximetilbilano Sintase/metabolismo , Masculino , Atividade Motora , Distribuição Aleatória , Ratos , Ratos Wistar
9.
Plant Physiol ; 139(4): 1946-58, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16306143

RESUMO

Protoporphyrinogen IX oxidase (PPO) catalyzes the last common step in chlorophyll and heme synthesis, and ferrochelatase (FeC) catalyzes the last step of the heme synthesis pathway. In plants, each of these two enzymes is encoded by two or more genes, and the enzymes have been reported to be located in the chloroplasts or in the mitochondria. We report that in the green alga Chlamydomonas reinhardtii, PPO and FeC are each encoded by a single gene. Phylogenetic analysis indicates that C. reinhardtii PPO and FeC are most closely related to plant counterparts that are located only in chloroplasts. Immunoblotting results suggest that C. reinhardtii PPO and FeC are targeted exclusively to the chloroplast, where they are associated with membranes. These results indicate that cellular needs for heme in this photosynthetic eukaryote can be met by heme that is synthesized in the chloroplast. It is proposed that the multiplicity of genes for PPO and FeC in higher plants could be related to differential expression in differently developing tissues rather than to targeting of different gene products to different organelles. The FeC content is higher in C. reinhardtii cells growing in continuous light than in cells growing in the dark, whereas the content of PPO does not significantly differ in light- and dark-grown cells. In cells synchronized to a light/dark cycle, the level of neither enzyme varied significantly with the phase of the cycle. These results indicate that heme synthesis is not directly regulated by the levels of PPO and FeC in C. reinhardtii.


Assuntos
Chlamydomonas reinhardtii/enzimologia , Ferroquelatase/metabolismo , Protoporfirinogênio Oxidase/metabolismo , Sequência de Aminoácidos , Animais , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/efeitos da radiação , DNA de Algas/genética , DNA Complementar/genética , DNA de Protozoário/genética , Escherichia coli/genética , Ferroquelatase/genética , Dosagem de Genes , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Genes de Protozoários , Luz , Dados de Sequência Molecular , Filogenia , Protoporfirinogênio Oxidase/genética , RNA de Algas/genética , RNA de Algas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Protozoário/genética , RNA de Protozoário/metabolismo , Homologia de Sequência de Aminoácidos , Frações Subcelulares/enzimologia
10.
J Bacteriol ; 187(15): 5084-9, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16030200

RESUMO

The heme prosthetic group of heme proteins contains iron, which can be a limiting nutrient. Here, we show that cytochrome c1 protein from Bradyrhizobium japonicum was strongly affected by the iron status, with low expression in cells grown under iron limitation. This control was not affected in mutants encoding the iron regulator Irr or Fur. Furthermore, cytochrome c1 mRNA was not influenced by the iron status, suggesting control at a posttranscriptional step. Cytochrome c1 protein levels were very low in mutants defective in the genes encoding delta-aminolevulinic acid (ALA) synthase and ferrochelatase, enzymes that catalyze the first and final steps of the heme biosynthetic pathway, respectively. Iron-dependent cytochrome c1 expression was restored in the ALA synthase mutant by supplementation of the medium with the heme precursor ALA. Supplementation with heme resulted in high levels of cytochrome c1 protein in the wild type and in both mutants, but expression was no longer iron dependent. Cytochrome c1 is synthesized as a protein precursor fused with cytochrome b. A plasmid-borne construct encoding only cytochrome c1 was expressed in an iron- and heme-dependent manner similar to that of the wild-type gene, indicating that control by those effectors is not linked to posttranslational processing of the fusion protein. Mutation of the cytochrome c1 cysteines involved in covalent binding to heme nearly abolished immunodetectable protein. Thus, defects in heme synthesis or heme binding abrogate cytochrome c1 accumulation, apparently due to protein degradation. We suggest that iron-dependent cytochrome c1 expression is mediated by heme availability for heme protein formation.


Assuntos
Bradyrhizobium/metabolismo , Citocromos c1/metabolismo , Heme/metabolismo , Ferro/metabolismo , 5-Aminolevulinato Sintetase/metabolismo , Bradyrhizobium/crescimento & desenvolvimento , Citocromos c1/biossíntese , Ferroquelatase/metabolismo , Heme/biossíntese , Ferro/farmacologia
11.
Biochem J ; 384(Pt 2): 429-36, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15307818

RESUMO

Our previous studies have demonstrated de novo haem biosynthesis in the malarial parasite (Plasmodium falciparum and P. berghei). It has also been shown that the first enzyme of the pathway is the parasite genome-coded ALA (delta-aminolaevulinate) synthase localized in the parasite mitochondrion, whereas the second enzyme, ALAD (ALA dehydratase), is accounted for by two species: one species imported from the host red blood cell into the parasite cytosol and another parasite genome-coded species in the apicoplast. In the present study, specific antibodies have been raised to PfFC (parasite genome-coded ferrochelatase), the terminal enzyme of the haem-biosynthetic pathway, using recombinant truncated protein. With the use of these antibodies as well as those against the hFC (host red cell ferrochelatase) and other marker proteins, immunofluorescence studies were performed. The results reveal that P. falciparum in culture manifests a broad distribution of hFC and a localized distribution of PfFC in the parasite. However, PfFC is not localized to the parasite mitochondrion. Immunoelectron-microscopy studies reveal that PfFC is indeed localized to the apicoplast, whereas hFC is distributed in the parasite cytoplasm. These results on the localization of PfFC are unexpected and are at variance with theoretical predictions based on leader sequence analysis. Biochemical studies using the parasite cytosolic and organellar fractions reveal that the cytosol containing hFC accounts for 80% of FC enzymic activity, whereas the organellar fraction containing PfFC accounts for the remaining 20%. Interestingly, both the isolated cytosolic and organellar fractions are capable of independent haem synthesis in vitro from [4-14C]ALA, with the cytosol being three times more efficient compared with the organellar fraction. With [2-14C]glycine, most of the haem is synthesized in the organellar fraction. Thus haem is synthesized in two independent compartments: in the cytosol, using the imported host enzymes, and in the organellar fractions, using the parasite genome-coded enzymes.


Assuntos
Ferroquelatase/metabolismo , Plasmodium falciparum/química , Animais , Anticorpos Antiprotozoários/metabolismo , Especificidade de Anticorpos , Clonagem Molecular , DNA Complementar/genética , DNA de Protozoário/genética , Eritrócitos/parasitologia , Ferroquelatase/genética , Ferroquelatase/imunologia , Heme/biossíntese , Humanos , Organelas/metabolismo , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Frações Subcelulares/metabolismo
12.
J Gene Med ; 5(9): 737-47, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12950064

RESUMO

BACKGROUND: Erythropoietic protoporphyria (EPP) is an inherited disease characterised by a ferrochelatase (FECH) deficiency, the latest enzyme of the heme biosynthetic pathway, leading to the accumulation of toxic protoporphyrin in the liver, bone marrow and spleen. We have previously shown that a successful gene therapy of a murine model of the disease was possible with lentiviral vectors even in the absence of preselection of corrected cells, but lethal irradiation of the recipient was necessary to obtain an efficient bone marrow engraftment. To overcome a preconditioning regimen, a selective growth advantage has to be conferred to the corrected cells. METHODS: We have developed a novel bicistronic lentiviral vector that contains the human alkylating drug resistance mutant O(6)-methylguanine DNA methyltransferase (MGMT G156A) and FECH cDNAs. We tested their capacity to protect hematopoietic cell lines efficiently from alkylating drug toxicity and correct enzymatic deficiency. RESULTS: EPP lymphoblastoid (LB) cell lines, K562 and cord-blood-derived CD34(+) cells were transduced at a low multiplicity of infection (MOI) with the bicistronic constructs. Resistance to O(6)-benzylguanine (BG)/N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU) was clearly shown in transduced cells, leading to the survival and expansion of provirus-containing cells. Corrected EPP LB cells were selectively amplified, leading to complete restoration of enzymatic activity and the absence of protoporphyrin accumulation. CONCLUSIONS: This study demonstrates that a lentiviral vector including therapeutic and G156A MGMT genes followed by BG/BCNU exposure can lead to a full metabolic correction of deficient cells. This vector might form the basis of new EPP mouse gene therapy protocols without a preconditioning regimen followed by in vivo selection of corrected hematopoietic stem cells.


Assuntos
Terapia Genética , Vetores Genéticos , Lentivirus/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Porfiria Hepatoeritropoética/terapia , Animais , Antígenos CD34/imunologia , Antineoplásicos/farmacologia , Carmustina/farmacologia , Linhagem Celular , DNA Complementar/genética , DNA Complementar/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ferroquelatase/genética , Ferroquelatase/metabolismo , Regulação da Expressão Gênica , Regulação Viral da Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Mutação Puntual , Porfiria Hepatoeritropoética/genética , Porfiria Hepatoeritropoética/metabolismo , Regiões Promotoras Genéticas , Linfócitos T/imunologia , Fatores de Tempo , Transgenes
13.
Arch Biochem Biophys ; 398(2): 170-8, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11831847

RESUMO

Ferrochelatase, the last enzyme of the heme biosynthetic pathway, has for years been considered to be active as a monomer. The crystal structure of Bacillus subtilis ferrochelatase confirmed its monomeric structure. However, animal ferrochelatase was found to form a functional dimer. Data presented here indicate that ferrochelatase from the yeast Saccharomyces cerevisiae is also dimeric. Following two-hybrid studies that had shown an interaction of two ferrochelatase molecules, we employed several different, complementary approaches, such as chemical crosslinking, affinity chromatography, and complementation analysis, to prove that in the yeast cells ferrochelatase forms an active dimer. We have isolated a double mutant, hem15D246V/Y248F, which is probably dimerization-defective. We propose a structural model of yeast ferrochelatase, based on the known structure of the human enzyme, which helps us to understand the differences in dimerization between the wild-type and mutant proteins.


Assuntos
Ferroquelatase/metabolismo , Saccharomyces cerevisiae/enzimologia , Reagentes de Ligações Cruzadas , Dimerização , Ferroquelatase/química , Ferroquelatase/genética , Humanos , Modelos Moleculares , Mutação , Relação Estrutura-Atividade
14.
Photochem Photobiol ; 72(2): 226-33, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10946577

RESUMO

5-Aminolevulinic acid (ALA)-supported fluorescence endoscopy of the urinary bladder results in a detection rate of bladder cancer superior to that of white light endoscopy. The different accumulation of the metabolite protoporphyrin IX (PPIX) in tumor cells after ALA instillation is poorly understood; however, it is crucial to optimize diagnosis and potential phototherapy. For systematic analysis of cell-type specific PPIX accumulation and metabolism two human bladder carcinoma cell lines (RT4 and J82), a normal urothelial cell line (UROtsa), and a fibroblast cell line (N1) were chosen, and grown in two different growth states to model important tissue components of the urinary bladder, i.e. tumor, normal epithelium and stroma. To quantitate PPIX content, fluorescence intensities measured by flow cytometry were matched with cellular PPIX extraction values, and related to relative ferrochelatase activity, cellular iron content, number of transferrin receptors per cell and porphobilinogen deaminase (PBGD) activity. For in vitro experiments, the initial correlation of relative flow cytometric and spectrometric measurements of PPIX provides a calibration curve for consequent flow cytometric PPIX quantification. Lower fluorescence of normal cells could be explained by significant differences of ferrochelatase activity and iron content in comparison to tumor cells. However, the content of iron was not related to transferrin receptor content. PBGD activity seemed to play a minor role for the differential accumulation of PPIX in urothelial cells. In conclusion, the in vitro culture of urothelial cells and fibroblasts indicates that the most important metabolic step for PPIX accumulation in the urinary bladder is the transition from PPIX to heme. Further investigation of PPIX metabolism does support the validation of photodynamic diagnosis, and might also lead the way to a highly specific tumor related molecule.


Assuntos
Fármacos Fotossensibilizantes/metabolismo , Protoporfirinas/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Divisão Celular , Ferroquelatase/metabolismo , Humanos , Hidroximetilbilano Sintase/metabolismo , Técnicas In Vitro , Ferro/metabolismo , Fotoquimioterapia , Receptores da Transferrina , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia
15.
Alcohol Alcohol ; 35(2): 109-25, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10787385

RESUMO

Alcohol is a porphyrinogenic agent which may cause disturbances in porphyrin metabolism in healthy persons as well as biochemical and clinical manifestations of acute and chronic hepatic porphyrias. After excessive consumption of alcohol, a temporary, clinically asymptomatic secondary hepatic coproporphyrinuria is observable, which can become persistent in cases of alcohol-induced liver damage. Nowadays, the alcohol-liver-porphyrinuria syndrome is the first to be mentioned in secondary hepatic disturbances of porphyrin metabolism. Acute hepatic porphyrias (acute intermittent porphyria, variegate porphyria and hereditary coproporphyria) are considered to be molecular regulatory diseases, in contrast to non-acute, chronic hepatic porphyria, clinically appearing as porphyria cutanea tarda (PCT). Porphyrins do not accumulate in the liver in acute porphyrias, whereas in chronic hepatic porphyrias they do. Thus, chronic hepatic porphyria is a porphyrin-accumulation disease, whereas acute hepatic porphyrias are haem-pathway-dysregulation diseases, characterized in general by induction of delta-aminolevulinic acid synthase in the liver and excessive stimulation of the pathway without storage of porphyrins in the liver. The clinical expression of acute hepatic porphyrias can be triggered by alcohol, because alcohol augments the inducibility of delta-aminolevulinic acid synthase. In chronic hepatic porphyrias, however, which are already associated with liver damage, alcohol potentiates the disturbance of the decarboxylation of uro- and heptacarboxyporphyrinogen, which is followed by a hepatic accumulation of uro- and heptacarboxyporphyrin and their sometimes extreme urinary excretion. Especially in persons with a genetic deficiency of uroporphyrinogen decarboxylase, but also in patients with the so-called sporadic variety of PCT, alcohol is able to transform an asymptomatic coproporphyrinuria into PCT. Alcohol has many biochemical and clinical effects on porphyrin and haem synthesis both in humans and laboratory animals. Ethanol suppresses the activity of porphobilinogen synthase (synonym: delta-aminolevulinic acid dehydratase), uroporphyrinogen decarboxylase, coproporphyrinogen oxidase and ferrochelatase, whereas it induces the first and rate-limiting enzyme in the pathway, delta-aminolevulinic acid synthase and also porphobilinogen deaminase. Therefore, teetotalism is a therapeutically and prophylactically important measure in all types of hepatic porphyrias.


Assuntos
Etanol/efeitos adversos , Porfirinas/metabolismo , Doença Aguda , Animais , Doença Crônica , Coproporfirinogênio Oxidase/metabolismo , DNA Complementar/genética , Progressão da Doença , Ferroquelatase/metabolismo , Humanos , Hidroximetilbilano Sintase/metabolismo , Sintase do Porfobilinogênio/metabolismo , Porfirias/classificação , Porfirias/diagnóstico , Porfirias/genética
16.
Plant J ; 15(4): 531-41, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9753778

RESUMO

Ferrochelatase is the last enzyme of haem biosynthesis. We have isolated 27 independent ferrochelatase cDNAs from Arabidopsis thaliana by functional complementation of a yeast mutant. Twenty-two of these cDNAs were similar to a previously isolated clone, AF3, and although they varied in length at the 5' and 3' ends, their nucleotide sequences were identical, indicating that they were derived from the same gene (ferrochelatase-I). The remaining five cDNAs all encoded a separate ferrochelatase isoform (ferrochelatase-II), which was 69% identical at the amino acid level to ferrochelatase-I. Using RFLP analysis in recombinant inbred lines, the ferrochelatase-I gene was mapped to chromosome V and that for ferrochelatase-II to chromosome II. Northern analysis showed that both ferrochelatase genes are expressed in leaves, stems and flowers, and expression in the leaves is higher in the light than in the dark. However, in roots only ferrochelatase-I transcripts were detected. High levels of sucrose stimulated expression of ferrochelatase-I, but had no effect, or repressed slightly, the expression of the ferrochelatase-II isoform. Import experiments into isolated chloroplasts and mitochondria showed that the ferrochelatase-II gene encodes a precursor which is imported solely into the chloroplast, in contrast to ferrochelatase-I which is targeted to both organelles. The significance of these results for haem biosynthesis and the production of haemoproteins, both within the plant cell and in different plant tissues, is discussed.


Assuntos
Arabidopsis/genética , Precursores Enzimáticos/metabolismo , Ferroquelatase/genética , Ferroquelatase/metabolismo , Genes de Plantas/genética , Sequência de Aminoácidos , Arabidopsis/enzimologia , Cloroplastos/enzimologia , Clonagem Molecular , DNA Complementar/genética , DNA de Plantas/genética , Regulação da Expressão Gênica de Plantas/fisiologia , Mitocôndrias/enzimologia , Dados de Sequência Molecular , Polimorfismo de Fragmento de Restrição , RNA Mensageiro/análise , RNA de Plantas/análise , Homologia de Sequência de Aminoácidos
17.
J Biol Chem ; 273(35): 22311-6, 1998 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9712849

RESUMO

In a previous study, site-directed mutagenesis experiments identified three of the four ligands to the [2Fe-2S] cluster in animal ferrochelatase as conserved cysteines in the COOH-terminal extension, Cys-403, Cys-406, and Cys-411 in human ferrochelatase (Crouse, B. R., Sellers, V. M., Finnegan, M. G., Dailey, H. A. & Johnson, M. K. (1996) Biochemistry 35, 16222-16229). The nature of the fourth ligand was left unresolved, and spectroscopic studies raised the possibility of one noncysteinyl, oxygenic ligand. In this work, we report two lines of evidence that strongly suggest the fourth ligand is a cysteine residue. Cysteine at position 196 in human recombinant ferrochelatase when changed to a serine results in an inactive enzyme that is lacking the [2Fe-2S] cluster. Furthermore, whole cell EPR studies demonstrate that in the C196S mutant the cluster fails to assemble. Additionally, the cloning and expression of Drosophila melanogaster ferrochelatase has allowed the identification, by EPR and UV-visible spectroscopy, of a [2Fe-2S]2+ cluster with properties analogous to those of animal ferrochelatases. The observation that Drosophila ferrochelatase contains only four conserved cysteines at positions 196, 403, 406, and 411, is in accord with the proposal that these residues function as cluster ligands. In the case of the ferrochelatase iron-sulfur cluster ligands, NH2-Cys-X206-Cys-X2-Cys-X4-Cys-COOH, the position distant from other ligands may lead to a spatial positioning of the cluster near the enzyme active site or at the interface of two domains, thereby explaining the loss of enzyme activity that accompanies cluster degradation and reinforcing the idea that the cluster functions as a regulatory switch.


Assuntos
Cisteína/metabolismo , Drosophila melanogaster/enzimologia , Ferroquelatase/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Cisteína/química , DNA Complementar , Espectroscopia de Ressonância de Spin Eletrônica , Ferroquelatase/química , Ferroquelatase/genética , Humanos , Ligantes , Espectrometria de Massas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Homologia de Sequência de Aminoácidos
18.
J Biol Chem ; 272(44): 27565-71, 1997 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-9346891

RESUMO

Ferrochelatase is the last enzyme of heme biosynthesis and in higher plants is found in both chloroplasts and mitochondria. We have isolated cDNAs for two isoforms of ferrochelatase from Arabidopsis thaliana, both of which are imported into isolated chloroplasts. In this paper we show that ferrochelatase-I is also imported into isolated pea mitochondria with approximately the same efficiency as into chloroplasts. Processing of the precursor was observed with both chloroplast stroma and mitochondrial matrix extracts. This was inhibited by EDTA, indicating it was due to the specific processing proteases. The specificity of import was verified by the fact that the mitochondrial preparation did not import the precursor of the light-harvesting chlorophyll a/b protein precursor or the precursor of porphobilinogen deaminase, an earlier enzyme of tetrapyrrole biosynthesis, both of which are exclusively chloroplast-located. Furthermore, import of ferrochelatase-I precursor into mitochondria was inhibited by valinomycin, but this had no effect on its import into chloroplasts. Thus a single precursor molecule is recognized by the import machinery of the two organelles. The implications for the targeting of ferrochelatase in a possible protective role against photooxidative stress are discussed.


Assuntos
Arabidopsis/metabolismo , Cloroplastos/metabolismo , Ferroquelatase/metabolismo , Mitocôndrias/metabolismo , Precursores de Proteínas/metabolismo , Transporte Biológico , DNA Complementar , Ferroquelatase/genética , Pisum sativum/metabolismo , Processamento de Proteína Pós-Traducional
19.
Hum Gene Ther ; 6(10): 1285-90, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8590732

RESUMO

Protoporphyria is generally an autosomal dominant disease characterized genetically by mutations in the ferrochelatase gene. The interaction between the wild-type and mutant ferrochelatase protein is unknown. The aim of this study was to evaluate the ability to correct the enzymatic and biochemical defects in cells from patients with protoporphyria, using a replication-defective human adenovirus for gene transfer. Overexpression of ferrochelatase was accomplished by construction of a vector in which expression of the wild-type ferrochelatase cDNA was driven by the constitutive cytomegalovirus (CMV) promoter, introduction and packaging of the cDNA into human adenovirus dl309, and transduction of normal and protoporphyric fibroblasts. Fibroblasts from controls and patients were infected with the ferrochelatase adenovirus or a control adenovirus and assayed for ferrochelatase activity and the accumulation of protoporphyrin upon challenge with the precursor delta-aminolevulinic acid (ALA). At a multiplicity of infection (moi) of 10, greater than 85% of both the wild-type and protoporphyric fibroblasts were infected. The recombinant adenovirus increased the ferrochelatase protein content and activity in the wild-type and protoporphyric fibroblasts with equal efficiency. Therefore, the presence of the mutant ferrochelatase protein did not inhibit the ferrochelatase activity expressed by the transgene.


Assuntos
Adenoviridae/genética , Ferroquelatase/genética , Ferroquelatase/metabolismo , Porfiria Hepatoeritropoética/terapia , Adenoviridae/química , Adenoviridae/patogenicidade , Ácido Aminolevulínico/metabolismo , Células Cultivadas , Citomegalovirus/genética , DNA Complementar , Deuteroporfirinas/metabolismo , Ferroquelatase/farmacologia , Fibroblastos/virologia , Células HeLa/metabolismo , Células HeLa/virologia , Humanos , Immunoblotting , Porfiria Hepatoeritropoética/genética , Porfiria Hepatoeritropoética/patologia , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transfecção , beta-Galactosidase/genética
20.
Br J Cancer ; 71(4): 733-7, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7710937

RESUMO

The use of photodynamic therapy (PDT) as an adjunct to curative tumour resection was investigated in a tumour recurrence model, using rat mammary adenocarcinoma BN472. Tumours were inoculated subcutaneously in 60 animals and resected after 21 days of growth. Immediately after removal, the operation site was exposed to 320-450 nm light of 0.1 W cm-2 and 60 J cm-2 after photosensitisation with either Photofrin (5 mg kg-1 i.v. 48 h before illumination) or 5-aminolaevulinic acid (ALA) (2 mg ml-1 in drinking water for 9 days). Porphyrin concentrations were measured in tissue samples. After 28 days, animals treated with adjunctive PDT had a significantly longer tumour-free interval than controls (P < 0.01); median 25 days (Photofrin), 18 days (ALA), 14 days (controls). Moreover, in the PDT groups significantly fewer rats had lymph node metastasis. A prophyrin concentration ratio between tumour and mammary tissue of 2:1 was found after Photofrin and 4:1 after ALA. The results indicate that adjuvant intraoperative PDT may be a safe and effective method of destroying residual tumour, thereby preventing locoregional tumour recurrence.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/cirurgia , Ácido Aminolevulínico/uso terapêutico , Derivado da Hematoporfirina/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/cirurgia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Ácido Aminolevulínico/administração & dosagem , Análise de Variância , Animais , Quimioterapia Adjuvante , Feminino , Ferroquelatase/metabolismo , Derivado da Hematoporfirina/administração & dosagem , Hidroximetilbilano Sintase/metabolismo , Luz , Metástase Linfática/prevenção & controle , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Recidiva Local de Neoplasia/prevenção & controle , Porfirinas/metabolismo , Ratos , Ratos Endogâmicos BN
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA