Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
1.
Mymensingh Med J ; 32(2): 584-586, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37002774

RESUMO

Porphyria cutanea tarda is a rare disorder of heme metabolism due to deficiency of the enzyme uroporphyrinogen decarboxylase which is manifested as some typical dermatological features and hepatic dysfunction. The Hepatitis-C virus co-infection is common and it can be aggravated by other environmental factors. We report a case of porphyria cutanea tarda in a 37-year-old woman, who presented with recurrent skin blisters and has concomitant Hepatitis-C virus infection. She was taking oestrogen containing oral contraceptive pill for a long duration. The diagnosis of porphyria cutanea tarda was considered on the basis of clinical features and high level of urine porphyrin level. She was put on hydroxychloroquine and combination drugs for Hepatitis-C virus with significant improvement after 3 months of therapy.


Assuntos
Coinfecção , Hepatite C , Porfiria Cutânea Tardia , Dermatopatias , Feminino , Humanos , Adulto , Porfiria Cutânea Tardia/complicações , Porfiria Cutânea Tardia/diagnóstico , Porfiria Cutânea Tardia/terapia , Coinfecção/diagnóstico , Coinfecção/complicações , Uroporfirinogênio Descarboxilase/metabolismo , Hepacivirus/metabolismo
2.
F1000Res ; 11: 945, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37360939

RESUMO

Background: Porphyria cutanea tarda (PCT) is a complex metabolic disease resulting from altered activity of the enzyme uroporphyrinogen decarboxylase (UROD) in the liver resulting in accumulation of uroporphyrin. PCT presents as a blistering photodermatitis with skin fragility, vesicles, scarring and milia. Case: We report a case of PCT in a 67-year-old man with hemochromatosis (HFE) gene mutation who, following a major syncopal episode in response to venesection was commenced on low dose hydroxychloroquine. Conclusions: Low dose hydroxychloroquine provided a safe and effective alternative to venesection in this patient who was needle phobic.


Assuntos
Hemocromatose , Porfiria Cutânea Tardia , Masculino , Humanos , Idoso , Porfiria Cutânea Tardia/tratamento farmacológico , Porfiria Cutânea Tardia/genética , Hidroxicloroquina/uso terapêutico , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/metabolismo , Hemocromatose/genética
3.
J Biol Chem ; 295(6): 1539-1550, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914409

RESUMO

Apicomplexan parasites such as Toxoplasma gondii possess an unusual heme biosynthesis pathway whose enzymes localize to the mitochondrion, cytosol, or apicoplast, a nonphotosynthetic plastid present in most apicomplexans. To characterize the involvement of the apicoplast in the T. gondii heme biosynthesis pathway, we investigated the role of the apicoplast-localized enzyme uroporphyrinogen III decarboxylase (TgUroD). We found that TgUroD knockdown impaired parasite proliferation, decreased free heme levels in the parasite, and decreased the abundance of heme-containing c-type cytochrome proteins in the parasite mitochondrion. We validated the effects of heme loss on mitochondrial cytochromes by knocking down cytochrome c/c1 heme lyase 1 (TgCCHL1), a mitochondrial enzyme that catalyzes the covalent attachment of heme to c-type cytochromes. TgCCHL1 depletion reduced parasite proliferation and decreased the abundance of c-type cytochromes. We further sought to characterize the overall importance of TgUroD and TgCCHL1 for both mitochondrial and general parasite metabolism. TgUroD depletion decreased cellular ATP levels, mitochondrial oxygen consumption, and extracellular acidification rates. By contrast, depletion of TgCCHL1 neither diminished ATP levels in the parasite nor impaired extracellular acidification rate, but resulted in specific defects in mitochondrial oxygen consumption. Together, our results indicate that the apicoplast has a key role in heme biology in T. gondii and is important for both mitochondrial and general parasite metabolism. Our study highlights the importance of heme and its synthesis in these parasites.


Assuntos
Apicoplastos/metabolismo , Heme/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Uroporfirinogênio Descarboxilase/metabolismo , Vias Biossintéticas , Heme/análise , Humanos , Mitocôndrias/metabolismo , Proteínas de Protozoários/análise , Toxoplasma/enzimologia , Toxoplasmose/parasitologia , Uroporfirinogênio Descarboxilase/análise
4.
Protein Sci ; 29(3): 789-802, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31930600

RESUMO

Acinetobacter baumannii is well known for causing hospital-associated infections due in part to its intrinsic antibiotic resistance as well as its ability to remain viable on surfaces and resist cleaning agents. In a previous publication, A. baumannii strain AB5075 was studied by transposon mutagenesis and 438 essential gene candidates for growth on rich-medium were identified. The Seattle Structural Genomics Center for Infectious Disease entered 342 of these candidate essential genes into our pipeline for structure determination, in which 306 were successfully cloned into expression vectors, 192 were detectably expressed, 165 screened as soluble, 121 were purified, 52 crystalized, 30 provided diffraction data, and 29 structures were deposited in the Protein Data Bank. Here, we report these structures, compare them with human orthologs where applicable, and discuss their potential as drug targets for antibiotic development against A. baumannii.


Assuntos
Acinetobacter baumannii/química , Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Genoma Bacteriano/efeitos dos fármacos , Genoma Bacteriano/genética , Acinetobacter baumannii/genética , Proteínas de Bactérias/genética , Coproporfirinogênio Oxidase/química , Coproporfirinogênio Oxidase/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Metionina tRNA Ligase/química , Metionina tRNA Ligase/metabolismo , Modelos Moleculares , Conformação Proteica , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo
5.
Mol Genet Metab ; 128(3): 164-177, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31326287

RESUMO

Porphyrias, is a general term for a group of metabolic diseases that are genetic in nature. In each specific porphyria the activity of specific enzymes in the heme biosynthetic pathway is defective and leads to accumulation of pathway intermediates. Phenotypically, each disease leads to either neurologic and/or photocutaneous symptoms based on the metabolic intermediate that accumulates. In each porphyria the distinct patterns of these substances in plasma, erythrocytes, urine and feces are the basis for diagnostically defining the metabolic defect underlying the clinical observations. Porphyrias may also be classified as either erythropoietic or hepatic, depending on the principal site of accumulation of pathway intermediates. The erythropoietic porphyrias are congenital erythropoietic porphyria (CEP), and erythropoietic protoporphyria (EPP). The acute hepatic porphyrias include ALA dehydratase deficiency porphyria, acute intermittent porphyria (AIP), hereditary coproporphyria (HCP) and variegate porphyria (VP). Porphyria cutanea tarda (PCT) is the only porphyria that has both genetic and/or environmental factors that lead to reduced activity of uroporphyrinogen decarboxylase in the liver. Each of the 8 enzymes in the heme biosynthetic pathway have been associated with a specific porphyria (Table 1). Mutations affecting the erythroid form of ALA synthase (ALAS2) are most commonly associated with X-linked sideroblastic anemia, however, gain-of-function mutations of ALAS2 have also been associated with a variant form of EPP. This overview does not describe the full clinical spectrum of the porphyrias, but is meant to be an overview of the biochemical steps that are required to make heme in both erythroid and non-erythroid cells.


Assuntos
Heme/biossíntese , Porfirias/genética , Animais , Vias Biossintéticas , Meio Ambiente , Humanos , Fígado/fisiopatologia , Camundongos , Mutação , Sintase do Porfobilinogênio/deficiência , Porfiria Aguda Intermitente , Porfirias/classificação , Porfirias/congênito , Porfirias Hepáticas , Uroporfirinogênio Descarboxilase/metabolismo
6.
J Med Case Rep ; 13(1): 17, 2019 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-30661508

RESUMO

BACKGROUND: The porphyrias are a rare group of metabolic disorders that can either be inherited or acquired. Along the heme biosynthetic pathway, porphyrias can manifest with neurovisceral and/or cutaneous symptoms, depending on the defective enzyme. Porphyria cutanea tarda, the most common type of porphyria worldwide, is caused by a deficiency of uroporphyrinogen decarboxylase, a crucial enzyme in heme biosynthesis, which results in an accumulation of photosensitive byproducts, such as uroporphyrinogen, which leads to the fragility and blistering of sun-exposed skin. Porphyria cutanea tarda is a condition that affects the liver and skin by reduction and inhibition of uroporphyrinogen decarboxylase enzyme in erythrocytes. Areas of skin that are exposed to the sun can generate blisters, hyperpigmentation, and, sometimes, lesions that heal leaving a scar or keratosis. Liver damage might present in a wide range of ways from liver function test abnormalities to hepatocellular carcinoma. The toxic effect of iron plays a role in liver damage pathogenesis. CASE PRESENTATION: A 59-year-old Turkish man presented with hyperpigmented skin lesions, fatigue, and elevated ferritin level and liver function tests. He was diagnosed as having porphyria cutanea tarda after a clinical investigation and treated with phlebotomy. CONCLUSION: Porphyria cutanea tarda is a rare condition of the liver but it must be remembered in a differential diagnosis of liver disease with typical skin involvement to decrease morbidity and health costs with early treatment.


Assuntos
Hiperpigmentação/patologia , Flebotomia/métodos , Porfiria Cutânea Tardia/diagnóstico , Uroporfirinogênio Descarboxilase/metabolismo , Fadiga/etiologia , Humanos , Hiperpigmentação/etiologia , Masculino , Pessoa de Meia-Idade , Porfiria Cutânea Tardia/terapia , Resultado do Tratamento
7.
Mol Genet Metab ; 128(3): 271-281, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30683557

RESUMO

Porphyria cutanea tarda (PCT) is the most common human porphyria, due to hepatic deficiency of uroporphyrinogen decarboxylase (UROD), which is acquired in the presence of iron overload and various susceptibility factors, such as alcohol abuse, smoking, hepatitis C virus (HCV) infection, HIV infection, iron overload with HFE gene mutations, use of estrogens, and UROD mutation. Patients with familial or type II PCT due to autosomal dominant UROD mutation also require other susceptibility factors, as the disease phenotype requires hepatic UROD deficiency to below 20% of normal. PCT clinically manifests with increased skin fragility and blistering skin lesions on sun exposed areas. The common age of presentation is 5th to 6th decade and occurs slightly more commonly in males. Although mild liver biochemical profile are common, advanced fibrosis and cirrhosis with hepatocellular carcinoma (HCC) can occasionally develop. Screening for HCC using ultrasound examination is recommended in PCT patients, especially with cirrhosis and advanced fibrosis. PCT is effectively and readily treatable with the use of either repeated phlebotomy or use of 100 mg hydroxychloroquine orally twice a week, and both the treatments are equally effective and safe. With the advent of new or direct antiviral agents for HCV infection, treatment of concomitant HCV has become safer and effective. Data are emerging on the benefit of these drugs as monotherapy for both PCT and HCV. After the achievement of remission of PCT, there remains a potential for relapse, especially when the susceptibility factors are not adequately controlled. Scanty data from retrospective and observational studies shows the relapse rate to be somewhat higher after remission with low-dose hydroxychloroquine as compared to phlebotomy induced remission. Future studies are needed on exploring mechanism of action of 4-aminoquinolines, understanding interaction of HCV and PCT, and relapse of PCT on long-term follow-up.


Assuntos
Mutação , Porfiria Cutânea Tardia/complicações , Porfiria Cutânea Tardia/genética , Aminoquinolinas/uso terapêutico , Animais , Carcinoma Hepatocelular/etiologia , Predisposição Genética para Doença , Humanos , Fígado/patologia , Neoplasias Hepáticas/etiologia , Camundongos , Recidiva Local de Neoplasia/etiologia , Estudos Observacionais como Assunto , Flebotomia , Porfiria Cutânea Tardia/tratamento farmacológico , Recidiva , Fatores de Risco , Uroporfirinogênio Descarboxilase/metabolismo
8.
Microbiol Mol Biol Rev ; 81(1)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28123057

RESUMO

The advent of heme during evolution allowed organisms possessing this compound to safely and efficiently carry out a variety of chemical reactions that otherwise were difficult or impossible. While it was long assumed that a single heme biosynthetic pathway existed in nature, over the past decade, it has become clear that there are three distinct pathways among prokaryotes, although all three pathways utilize a common initial core of three enzymes to produce the intermediate uroporphyrinogen III. The most ancient pathway and the only one found in the Archaea converts siroheme to protoheme via an oxygen-independent four-enzyme-step process. Bacteria utilize the initial core pathway but then add one additional common step to produce coproporphyrinogen III. Following this step, Gram-positive organisms oxidize coproporphyrinogen III to coproporphyrin III, insert iron to make coproheme, and finally decarboxylate coproheme to protoheme, whereas Gram-negative bacteria first decarboxylate coproporphyrinogen III to protoporphyrinogen IX and then oxidize this to protoporphyrin IX prior to metal insertion to make protoheme. In order to adapt to oxygen-deficient conditions, two steps in the bacterial pathways have multiple forms to accommodate oxidative reactions in an anaerobic environment. The regulation of these pathways reflects the diversity of bacterial metabolism. This diversity, along with the late recognition that three pathways exist, has significantly slowed advances in this field such that no single organism's heme synthesis pathway regulation is currently completely characterized.


Assuntos
Archaea/metabolismo , Bactérias/metabolismo , Heme/análogos & derivados , Ferro/química , Tetrapirróis/biossíntese , Ácido Aminolevulínico/metabolismo , Coproporfirinogênio Oxidase/metabolismo , Coproporfirinas/metabolismo , Heme/biossíntese , Protoporfirinas/biossíntese , Protoporfirinas/metabolismo , Uroporfirinogênio Descarboxilase/metabolismo
9.
Acta Biochim Pol ; 62(3): 509-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317124

RESUMO

Heme biosynthesis pathway is conserved in yeast and humans and hem12 yeast mutants mimic porphyria cutanea tarda (PCT), a hereditary human disease caused by mutations in the UROD gene. Even though mutations in other genes also affect UROD activity and predispose to sporadic PCT, the regulation of UROD is unknown. Here, we used yeast as a model to study regulation of Hem12 by ubiquitination and involvement of Rsp5 ubiquitin ligase in this process. We found that Hem12 is monoubiquitinated in vivo by Rsp5. Hem12 contains three conserved lysine residues located on the protein surface that can potentially be ubiquitinated and lysine K8 is close to the 36-LPEY-39 (PY) motif which binds WW domains of the Rsp5 ligase. The hem12-K8A mutation results in a defect in cell growth on a glycerol medium at 38°C but it does not affect the level of Hem12. The hem12-L36A,P37A mutations which destroy the PY motif result in a more profound growth defect on both, glycerol and glucose-containing media. However, after several passages on the glucose medium, the hem12-L36A,P37A cells adapt to the growth medium owing to higher expression of hem12-L36A,P37A gene and higher stability of the mutant Hem12-L36A,P37A protein. The Hem12 protein is downregulated upon heat stress in a Rsp5-independent way. Thus, Rsp5-dependent Hem12 monoubiquitination is important for its functioning, but not required for its degradation. Since Rsp5 has homologs among the Nedd4 family of ubiquitin ligases in humans, a similar regulation by ubiquitination might be also important for functioning of the human UROD.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Regulação Fúngica da Expressão Gênica , Mutação , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Complexos Ubiquitina-Proteína Ligase/metabolismo , Uroporfirinogênio Descarboxilase/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Regulação para Baixo , Glucose/química , Glicerol/química , Heme/metabolismo , Humanos , Lisina/química , Conformação Molecular , Dados de Sequência Molecular , Porfiria Cutânea Tardia/genética , Proteínas de Saccharomyces cerevisiae/fisiologia , Homologia de Sequência de Aminoácidos , Ubiquitina/química , Ubiquitinação , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/fisiologia
10.
Environ Sci Pollut Res Int ; 22(8): 5877-86, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25352395

RESUMO

Bisphenol A (BPA), as an emerging environmental pollutant, is potentially harmful to plant growth. Chlorophyll (Chl) is critical in photosynthesis that provides matter and energy for plant growth. How BPA affects the chlorophyll content remains largely unknown. Here, the effects of BPA on Chl synthesis in soybean seedlings were investigated. Exposure to 1.5 mg/L BPA decreased the 5-aminolevulinic acid (ALA) content and increased protoporphyrin IX (Proto IX), magnesium protoporphyrin, and protochlorophyll contents and 5-aminolaevulinic acid dehydratase, porphobilinogen deaminase, uroporphyrinogen III synthase, uroporphyrinogen III decarboxylase, and protoporphyrinogen oxidase activities. Exposure to 17.2 and 50.0 mg/L BPA exerted the opposite effects on these four intermediates and five enzymes. Following the withdrawal of BPA exposure, the aforementioned parameters gradually recovered, except magnesium protoporphyrin content in exposure to 50.0 mg/L BPA. Our findings revealed that exposure to low-concentration BPA increased the Chl content in soybean seedlings through improving Chl synthesis, especially the conversion from ALA to Proto IX, whereas exposure to high-concentration BPA decreased the Chl content through inhibiting Chl synthesis, especially the conversion from ALA to Proto IX. The dual effects of BPA were largely reversed following the withdrawal of BPA exposure.


Assuntos
Compostos Benzidrílicos/toxicidade , Vias Biossintéticas/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Glycine max/efeitos dos fármacos , Fenóis/toxicidade , Fotossíntese/efeitos dos fármacos , Plântula/efeitos dos fármacos , Ácido Aminolevulínico/metabolismo , Análise de Variância , Clorofila/análogos & derivados , Clorofila/metabolismo , Hidroximetilbilano Sintase/metabolismo , Folhas de Planta/química , Sintase do Porfobilinogênio/metabolismo , Protoporfirinogênio Oxidase/metabolismo , Protoporfirinas/metabolismo , Plântula/metabolismo , Glycine max/metabolismo , Espectrometria de Fluorescência , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinogênio III Sintetase/metabolismo
11.
Oxid Med Cell Longev ; 2013: 932472, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23970950

RESUMO

Uroporphyrinogen decarboxylase (Hem12p) and transketolase (Tkl1p) are key mediators of two critical processes within the cell, heme biosynthesis, and the nonoxidative part of the pentose phosphate pathway (PPP). The redox properties of both Hem12p and Tkl1p from Saccharomyces cerevisiae were investigated using proteomic techniques (SRM and label-free quantification) and biochemical assays in cell extracts and in vitro with recombinant proteins. The in vivo analysis revealed an increase in oxidized Cys-peptides in the absence of Grx2p, and also after treatment with H2O2 in the case of Tkl1p, without corresponding changes in total protein, demonstrating a true redox response. Out of three detectable Cys residues in Hem12p, only the conserved residue Cys52 could be modified by glutathione and efficiently deglutathionylated by Grx2p, suggesting a possible redox control mechanism for heme biosynthesis. On the other hand, Tkl1p activity was sensitive to thiol redox modification and although Cys622 could be glutathionylated to a limited extent, it was not a natural substrate of Grx2p. The human orthologues of both enzymes have been involved in certain cancers and possess Cys residues equivalent to those identified as redox sensitive in yeast. The possible implication for redox regulation in the context of tumour progression is put forward.


Assuntos
Heme/biossíntese , Via de Pentose Fosfato/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Compostos de Sulfidrila/metabolismo , Transcetolase/metabolismo , Uroporfirinogênio Descarboxilase/metabolismo , Peróxido de Hidrogênio/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos
12.
J Biomol Struct Dyn ; 31(12): 1358-69, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23140436

RESUMO

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


Assuntos
Inibidores Enzimáticos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/prevenção & controle , Receptor ErbB-2/antagonistas & inibidores , Uroporfirinogênio Descarboxilase/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Simulação por Computador , Bases de Dados Factuais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Receptores ErbB/química , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Medicina Tradicional Chinesa/métodos , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Taiwan , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo
13.
PLoS One ; 7(11): e50087, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209648

RESUMO

Uroporphyrinogen decarboxylase (UROD) has been suggested as a protectant against radiation for head and neck cancer (HNC). In this study, we employed traditional Chinese medicine (TCM) compounds from TCM Database@Taiwan (http://tcm.cmu.edu.tw/) to screen for drug-like candidates with potential UROD inhibition characteristics using virtual screening techniques. Isopraeroside IV, scopolin, and nodakenin exhibited the highest Dock Scores, and were predicted to have good Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) properties. Two common moieties, 2H-chromen-2-one and glucoside, were observed among the top TCM candidates. Cross comparison of the docking poses indicated that candidates formed stable interactions with key binding and catalytic residues of UROD through these two moieties. The 2H-chromen-2-one moiety enabled pi-cation interactions with Arg37 and H-bonds with Tyr164. The glucoside moiety was involved in forming H-bonds with Arg37 and Asp86. From our computational results, we propose isopraeroside IV, scopolin, and nodakenin as ligands that might exhibit drug-like inhibitory effects on UROD. The glucoside and 2H-chromen-2-one moieties may potentially be used for designing inhibitors of UROD.


Assuntos
Medicina Tradicional Chinesa/métodos , Uroporfirinogênio Descarboxilase/metabolismo , Algoritmos , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X/métodos , Desenho de Fármacos , Glucosídeos/metabolismo , Humanos , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Ligantes , Modelos Químicos , Simulação de Dinâmica Molecular , Ligação Proteica , Software , Tirosina/genética , Uroporfirinogênio Descarboxilase/química
14.
Blood Cells Mol Dis ; 47(4): 249-54, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21880518

RESUMO

Cytochrome P4501A2 (Cyp1a2) is important in the development of uroporphyria in mice, a model of porphyria cutanea tarda in humans. Heretofore, mice homozygous for the Cyp1a2-/- mutation do not develop uroporphyria with treatment regimens that result in uroporphyria in wild-type mice. Here we report uroporphyria development in Cyp1a2-/- mice additionally null for both alleles of the hemochromatosis (Hfe) gene and heterozygous for deletion of the uroporphyrinogen decarboxylase (Urod) gene (genotype: Cyp1a2-/-;Hfe-/-;Urod+/-), demonstrating that upon adding porphyria-predisposing genetic manipulations, Cyp1a2 is not essential. Cyp1a2-/-;Hfe-/-;Urod+/- mice were treated with various combinations of an iron-enriched diet, parenteral iron-dextran, drinking water containing δ-aminolevulinic acid and intraperitoneal Aroclor 1254 (a polychlorinated biphenyl mixture) and analyzed for uroporphyrin accumulation. Animals fed an iron-enriched diet alone did not develop uroporphyria but uroporphyria developed with all treatments that included iron supplementation and δ-aminolevulinic acid, even with a regimen without Aroclor 1254. Hepatic porphyrin levels correlated with low UROD activity and high levels of an inhibitor of UROD but marked variability in the magnitude of the porphyric response was present in all treatment groups. Gene expression profiling revealed no major differences between genetically identical triple cross mice exhibiting high and low magnitude porphyric responses from iron-enriched diet and iron-dextran supplementation, and δ-aminolevulinic acid. Even though the variation in porphyric response did not parallel the hepatic iron concentration, the results are compatible with the presence of a Cyp1a2-independent, iron-dependent pathway for the generation of uroporphomethene, the UROD inhibitor required for the expression of uroporphyria in mice and PCT in humans.


Assuntos
Citocromo P-450 CYP1A2/genética , Porfiria Cutânea Tardia/genética , Animais , Citocromo P-450 CYP1A2/metabolismo , Modelos Animais de Doenças , Genótipo , Ferro/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Porfiria Cutânea Tardia/dietoterapia , Porfiria Cutânea Tardia/metabolismo , Porfirinas/metabolismo , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/metabolismo
15.
Br J Dermatol ; 165(3): 499-505, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21668429

RESUMO

BACKGROUND: Hepatoerythropoietic porphyria (HEP) is a rare form of porphyria that results from a deficiency of uroporphyrinogen decarboxylase (UROD). The disease is caused by homoallelism or heteroallelism for mutations in the UROD gene. OBJECTIVE: To study a 19-year-old woman from Equatorial Guinea, one of the few cases of HEP of African descent and to characterize a new mutation causing HEP. METHODS: Excretion of porphyrins and residual UROD activity in erythrocytes were measured and compared with those of other patients with HEP. The UROD gene of the proband was sequenced and a new mutation identified. The recombinant UROD protein was purified and assayed for enzymatic activity. The change of amino acid mapped to the UROD protein and the functional consequences were predicted. RESULTS: The patient presented a novel homozygous G170D missense mutation. Porphyrin excretion showed an atypical pattern in stool with a high pentaporphyrin III to isocoproporphyrin ratio. Erythrocyte UROD activity was 42% of normal and higher than the activity found in patients with HEP with a G281E mutation. The recombinant UROD protein showed a relative activity of 17% and 60% of wild-type to uroporphyrinogen I and III respectively. Molecular modelling showed that glycine 170 is located on the dimer interface of UROD, in a loop containing residues 167-172 that are critical for optimal enzymatic activity and that the carboxyl side chain from aspartic acid is predicted to cause negative interactions between the protein and the substrate. CONCLUSIONS: The results emphasize the complex relationship between the genetic defects and the biochemical phenotype in homozygous porphyria.


Assuntos
Mutação de Sentido Incorreto/genética , Porfiria Hepatoeritropoética/genética , Uroporfirinogênio Descarboxilase/genética , Cromatografia Líquida de Alta Pressão , Eritrócitos/enzimologia , Feminino , Técnicas de Genotipagem , Homozigoto , Humanos , Proteínas Recombinantes , Uroporfirinogênio Descarboxilase/metabolismo , Adulto Jovem
17.
Sci Transl Med ; 3(67): 67ra7, 2011 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-21270338

RESUMO

Head and neck cancer (HNC) is the eighth most common malignancy worldwide, comprising a diverse group of cancers affecting the head and neck region. Despite advances in therapeutic options over the last few decades, treatment toxicities and overall clinical outcomes have remained disappointing, thereby underscoring a need to develop novel therapeutic approaches in HNC treatment. Uroporphyrinogen decarboxylase (UROD), a key regulator of heme biosynthesis, was identified from an RNA interference-based high-throughput screen as a tumor-selective radiosensitizing target for HNC. UROD knockdown plus radiation induced caspase-mediated apoptosis and cell cycle arrest in HNC cells in vitro and suppressed the in vivo tumor-forming capacity of HNC cells, as well as delayed the growth of established tumor xenografts in mice. This radiosensitization appeared to be mediated by alterations in iron homeostasis and increased production of reactive oxygen species, resulting in enhanced tumor oxidative stress. Moreover, UROD was significantly overexpressed in HNC patient biopsies. Lower preradiation UROD mRNA expression correlated with improved disease-free survival, suggesting that UROD could potentially be used to predict radiation response. UROD down-regulation also radiosensitized several different models of human cancer, as well as sensitized tumors to chemotherapeutic agents, including 5-fluorouracil, cisplatin, and paclitaxel. Thus, our study has revealed UROD as a potent tumor-selective sensitizer for both radiation and chemotherapy, with potential relevance to many human malignancies.


Assuntos
Técnicas de Silenciamento de Genes , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/radioterapia , Radiossensibilizantes/metabolismo , Radiossensibilizantes/uso terapêutico , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos da radiação , Modelos Animais de Doenças , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Homeostase , Humanos , Ferro/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Estresse Oxidativo , Interferência de RNA
18.
J Comput Chem ; 32(5): 822-34, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20941734

RESUMO

In humans, uroporphyrinogen decarboxylase is intimately involved in the synthesis of heme, where the decarboxylation of the uroporphyrinogen-III occurs in a single catalytic site. Several variants of the mechanistic proposal exist; however, the exact mechanism is still debated. Thus, using an ONIOM quantum mechanical/molecular mechanical approach, the mechanism by which uroporphyrinogen decarboxylase decarboxylates ring D of uroporphyrinogen-III has been investigated. From the study performed, it was found that both Arg37 and Arg50 are essential in the decarboxylation of ring D, where experimentally both have been shown to be critical to the catalytic behavior of the enzyme. Overall, the reaction was found to have a barrier of 10.3 kcal mol(-1) at 298.15 K. The rate-limiting step was found to be the initial proton transfer from Arg37 to the substrate before the decarboxylation. In addition, it has been found that several key interactions exist between the substrate carboxylate groups and backbone amides of various active site residues as well as several other functional groups.


Assuntos
Simulação de Dinâmica Molecular , Porfirinas/biossíntese , Teoria Quântica , Uroporfirinogênio Descarboxilase/química , Sítios de Ligação , Biocatálise , Biologia Computacional , Descarboxilação , Humanos , Modelos Moleculares , Conformação Molecular , Uroporfirinogênio Descarboxilase/metabolismo
19.
J Phys Chem B ; 114(27): 8994-9001, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20553007

RESUMO

Uroporphyrinogen III decarboxylase catalyzes the fifth step in heme biosynthesis, the elimination of carboxyl groups from the four acetate side chains of uroporphyrinogen III to yield coproporphyrinogen III. We have previously found that the rate-limiting step of uroporphyrinogen III decarboxylase is substrate protonation rather than the decarboxylation reaction. This protonation can be effected by an arginine residue (Arg37) in close proximity to the substrate. In this report, we present evidence for the function of this arginine residue as a general acid catalyst. Although substrate protonation by H(3)O(+) is both exergonic and very fast, our density functional calculations show that in the presence of a protonated Arg37 substrate, decarboxylation becomes rate-limiting, and the substrate spontaneously breaks upon protonation. These results suggest that the active site must be shielded from solvent protons. Consequently, H(3)O(+) can be excluded from a role in both protonations proposed for the enzyme mechanism. In agreement with these conclusions, a second arginine residue (Arg41) is uniquely positioned to act as donor of the second proton, with an activation barrier below 2 kcal mol(-1). Generated mutant uroporphyrinogen III decarboxylase variants carrying amino acid exchanges in the position of both arginine residues (R41A, R41K, R37A, and R37K) failed to produce coproporphyrinogen III. The proposed unusual use of two basic residues as general acids in two different proton donation steps by uroporphyrinogen III decarboxylase provides an elegant solution to the problem of simultaneously binding the very negative uroporphyrinogen (which requires a positively charged active site), and selectively protonating it while preventing excessive carboxylate stabilization by positive charges.


Assuntos
Arginina , Oniocompostos , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Domínio Catalítico , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Prótons , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênios/metabolismo
20.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 40-5, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19656450

RESUMO

Porphyria cutanea tarda (PCT) is caused by inhibition of uroporphyrinogen decarboxylase (URO-D) activity in hepatocytes. Subnormal URO-D activity results in accumulation and urinary excretion of uroporphyrin and heptacarboxyl porphyrin. Heterozygosity for mutations in the URO-D gene is found in the familial form of PCT (F-PCT). Over 70 mutations of URO-D have been described but very few have been characterized structurally. Here we characterize 3 mutations in the URO-D gene found in patients with F-PCT, G318R, K297N, and D306Y. Expression of the D306Y mutation results in an insoluble recombinant protein. G318R and K297N have little effect on the structure or activity of recombinant URO-D, but the proteins display reduced stability in vitro.


Assuntos
Uroporfirinogênio Descarboxilase/metabolismo , Adulto , Idoso , Domínio Catalítico , Cristalografia por Raios X , Feminino , Genótipo , Heterozigoto , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Porfiria Cutânea Tardia/etiologia , Porfiria Cutânea Tardia/genética , Estabilidade Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA