Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Leuk Res ; 39(8): 835-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26038120

RESUMO

We investigated protracted low-dose oral Clofarabine for the treatment of myelodysplastic syndromes (MDS). Adults with an International Prognostic Scoring System (IPSS) score of INT-1 or higher who had failed first line therapy were eligible. INT-1 patients had to be transfusion-dependent. We started with oral Clofarabine at 5mg (fixed dose) daily for 10 consecutive days on a 28-day cycle. Toxicity prompted a modification to 1mg PO daily for 10 days and then 1mg PO daily for 7 days. Patients received treatment indefinitely until loss of response or unacceptable toxicity. Nine patients (5 women) were enrolled and evaluable (median age 65 years; range 55-81). A 10-day regimen of oral Clofarabine at 5mg/day induced Grade IV pancytopenia. A dose of 1 mg/day for 7/28 days was very well tolerated without significant toxicity. Three patients had responses (2 with responses lasting up to 21 and 51 cycles) defined as stable disease in spite of no significant change on bone marrow evaluation. Low-dose oral Clofarabine (1mg daily for 7/28 days) proved both effective and safe for patients with MDS who had failed prior therapy. This patient population is particularly sensitive to more protracted Clofarabine treatment schedules.


Assuntos
Nucleotídeos de Adenina/administração & dosagem , Antineoplásicos/administração & dosagem , Arabinonucleosídeos/administração & dosagem , Síndromes Mielodisplásicas/tratamento farmacológico , Nucleotídeos de Adenina/efeitos adversos , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Arabinonucleosídeos/efeitos adversos , Clofarabina , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Falha de Tratamento
2.
Clin Biochem ; 48(12): 788-95, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25959086

RESUMO

OBJECTIVE: In mammalian cells the rate-limiting step in heme biosynthesis is the formation of δ-aminolevulinic acid (ALA). The reaction intermediates, porphyrins and iron and the final product, heme can be highly cytotoxic if allowed to accumulate. The importance of maintaining the levels of metabolic intermediates and heme within a narrow range is apparent based on the complex homeostatic system(s) that have developed. Ultimately, determining the enzymatic activity of ALA synthase (ALAS) present in the mitochondria is highly beneficial to confirm the effects of the transcriptional, translational and post-translational events. The aim of this study was to develop a highly sensitive assay for ALAS that could be used on whole tissue or cellular homogenates. DESIGN AND METHODS: A systematic approach was used to optimize steps in formation of ALA by ALAS. Reducing the signal to noise ratio for the assay was achieved by derivatizing the ALA formed into a fluorescent product that could be efficiently separated by ultra performance liquid chromatography (UPLC) from other derivatized primary amines. The stability of ALAS activity in whole tissue homogenate and cellular homogenate was determined after extended storage at -80 °C. CONCLUSIONS: A method for assaying ALAS has been developed that can be used with tissue homogenates or cellular lysates. There is no need to purify mitochondria and radiolabeled substrates are not needed for this assay. General laboratory reagents can be used to prepare the samples. Standard UPLC chromatography will resolve the derivatized ALA peak. Samples of tissue homogenate can be stored for approximately one year without significant loss of enzymatic activity.


Assuntos
5-Aminolevulinato Sintetase/análise , 5-Aminolevulinato Sintetase/metabolismo , Animais , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Corantes Fluorescentes/química , Humanos , Leucemia Eritroblástica Aguda/enzimologia , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL
3.
Obesity (Silver Spring) ; 22(10): 2120-2, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044717

RESUMO

OBJECTIVE: Levels of tissue iron contribute to determining diabetes risk, but little is known about the effects of higher iron levels on weight, and on the interaction of weight and iron overload on diabetes risk. Therefore, the effect of iron on body mass index and diabetes in individuals with iron overload from hereditary hemochromatosis (HH), compared to non-HH siblings and historical controls was examined. METHODS: Chart reviews were performed on a cohort of adults (age ≥40, N = 101) with the common C282Y/C282Y HFE genotype, compared to wild type siblings (N = 32) and comparable NHANES cohorts, with respect to body mass index and diabetes status. RESULTS: Males with HH have lower body mass index (BMI) than control siblings. Females had a trend toward decreased BMI that was not significant, possibly related to decreased degrees of iron overload. In both males and females, increased rates of diabetes were seen, especially in the overweight or obese. CONCLUSIONS: High tissue iron levels may be both pro- and anti-diabetic. The prevalence of obesity and diabetes in HH is likely dependent upon the degree of iron overload, caloric intake, and other genetic and environmental factors, contributing to the observed heterogeneity in the frequency of disease-related morbidities in HH.


Assuntos
Diabetes Mellitus/etiologia , Hemocromatose/complicações , Obesidade/prevenção & controle , Adulto , Índice de Massa Corporal , Feminino , Genótipo , Hemocromatose/genética , Hemocromatose/metabolismo , Antígenos de Histocompatibilidade Classe I , Humanos , Ferro/sangue , Masculino , Pessoa de Meia-Idade , Prevalência , Fatores de Risco
4.
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
6.
J Clin Invest ; 120(7): 2395-405, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20530874

RESUMO

Hepcidin is a peptide hormone that regulates iron homeostasis and acts as an antimicrobial peptide. It is expressed and secreted by a variety of cell types in response to iron loading and inflammation. Hepcidin mediates iron homeostasis by binding to the iron exporter ferroportin, inducing its internalization and degradation via activation of the protein kinase Jak2 and the subsequent phosphorylation of ferroportin. Here we have shown that hepcidin-activated Jak2 also phosphorylates the transcription factor Stat3, resulting in a transcriptional response. Hepcidin treatment of ferroportin-expressing mouse macrophages showed changes in mRNA expression levels of a wide variety of genes. The changes in transcript levels for half of these genes were a direct effect of hepcidin, as shown by cycloheximide insensitivity, and dependent on the presence of Stat3. Hepcidin-mediated transcriptional changes modulated LPS-induced transcription in both cultured macrophages and in vivo mouse models, as demonstrated by suppression of IL-6 and TNF-alpha transcript and secreted protein. Hepcidin-mediated transcription in mice also suppressed toxicity and morbidity due to single doses of LPS, poly(I:C), and turpentine, which is used to model chronic inflammatory disease. Most notably, we demonstrated that hepcidin pretreatment protected mice from a lethal dose of LPS and that hepcidin-knockout mice could be rescued from LPS toxicity by injection of hepcidin. The results of our study suggest a new function for hepcidin in modulating acute inflammatory responses.


Assuntos
Inflamação/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos , Transporte Biológico , Proteínas de Transporte de Cátions , Citocinas/metabolismo , Citocinas/farmacologia , Hepcidinas , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Ferro/metabolismo , Ferro/farmacologia , Ferro da Dieta/metabolismo , Ferro da Dieta/farmacologia , Janus Quinase 2/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fosforilação , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
7.
Blood ; 115(17): 3425-6, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20430964
8.
Chem Biol Interact ; 182(2-3): 220-6, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-19772856

RESUMO

The response of hepatic mono-oxygenase activities to Aroclor 1254 or 3-methylcholanthrene was investigated in wild-type and Cyp1a2(-/-) mice. Cytochrome P450 concentrations were similar in naïve Cyp1a2(-/-) and wild-type mice. There was no difference between naïve wild-type and Cyp1a2(-/-) animals in 7-ethoxyresorufin and 7-ethoxy-4-trifluoromethylcoumarin dealkylase activities, nor was the induction response after 3-methylcholanthrene any different between the two genotypes. However, both activities were induced to a higher extent in Cyp1a2(-/-) mice after Aroclor 1254. In contrast, 7-pentoxyresorufin dealkylation activity was lower in Cyp1a2(-/-) mice and this differential was maintained during induction by both agents. 7-Methoxy- and 7-benzoxyresorufin dealkylation activities were also lower than wild-type in naïve Cyp1a2(-/-) animals and during 3-methylcholanthrene induction, but showed accelerated induction in Cyp1a2(-/-) mice with Aroclor 1254. Bufuralol 1'- and testosterone 6beta-hydroxylation activities, and P450 characteristics were evaluated 48h after inducer administration. Bufuralol 1'-hydroxylation, a sexual dimorphic activity (female>male) showed no genotype differences in naïve animals. Activity changes varied across gender and genotype, with 3-methylcholanthrene and Aroclor 1254 inducing in male Cyp1a2(-/-), and Aroclor 1254 inducing in female wild-type. Testosterone 6beta-hydroxylation activity was 16% higher in Cyp1a2(-/-) mice and neither 3-methylcholanthrene nor Aroclor 1254 elicited induction. After Aroclor 1254, a 24% increase in P450 concentration with a hypsochromic shift in the ferrous-CO maximum characteristic of CYP1A enzymes occurred in wild-type, compared to no change in either parameter in Cyp1a2(-/-) mice. Induction changes with 3-methylcholanthrene were greater in wild-type mice, a 60% increase in concentration and approximately 2 nm hypsochromic shift versus a 10% increase and approximately 1nm hypsochromic shift in Cyp1a2(-/-) mice. The study demonstrates that deletion of a single P450 can profoundly affect the induction response, as monitored with activities of other P450s, in a manner unrelated to the contribution of the deleted P450 to the activity.


Assuntos
/farmacologia , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Metilcolantreno/farmacologia , Animais , Citocromo P-450 CYP1A2/genética , Feminino , Deleção de Genes , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bifenilos Policlorados/análise
9.
Blood ; 114(9): 1913-8, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19587376

RESUMO

Hepcidin is the major regulator of systemic iron homeostasis in mammals. Hepcidin is produced mainly by the liver and is increased by inflammation, leading to hypoferremia. We measured serum levels of bioactive hepcidin and its effects on serum iron levels in mice infected with Borrelia burgdorferi. Bioactive hepcidin was elevated in the serum of mice resulting in hypoferremia. Infected mice produced hepcidin in both liver and spleen. Both intact and sonicated B burgdorferi induced hepcidin expression in cultured mouse bone marrrow macrophages. Hepcidin production by cultured macrophages represents a primary transcriptional response stimulated by B burgdorferi and not a secondary consequence of cytokine elaboration. Hepcidin expression induced by B burgdorferi was mediated primarily by activation of Toll-like receptor 2.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Borrelia burgdorferi/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptores Toll-Like/metabolismo , Animais , Células da Medula Óssea/citologia , Citocinas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Hepcidinas , Humanos , Inflamação , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos
10.
J Mol Biol ; 389(2): 306-14, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19362562

RESUMO

Uroporphyrinogen decarboxylase (URO-D; EC 4.1.1.37), the fifth enzyme of the heme biosynthetic pathway, is required for the production of heme, vitamin B12, siroheme, and chlorophyll precursors. URO-D catalyzes the sequential decarboxylation of four acetate side chains in the pyrrole groups of uroporphyrinogen to produce coproporphyrinogen. URO-D is a stable homodimer, with the active-site clefts of the two subunits adjacent to each other. It has been hypothesized that the two catalytic centers interact functionally, perhaps by shuttling of reaction intermediates between subunits. We tested this hypothesis by construction of a single-chain protein (single-chain URO-D) in which the two subunits were connected by a flexible linker. The crystal structure of this protein was shown to be superimposable with wild-type activity and to have comparable catalytic activity. Mutations that impaired one or the other of the two active sites of single-chain URO-D resulted in approximately half of wild-type activity. The distributions of reaction intermediates were the same for mutant and wild-type sequences and were unaltered in a competition experiment using I and III isomer substrates. These observations indicate that communication between active sites is not required for enzyme function and suggest that the dimeric structure of URO-D is required to achieve conformational stability and to create a large active-site cleft.


Assuntos
Coproporfirinogênios/biossíntese , Uroporfirinogênio Descarboxilase/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênios/metabolismo
11.
Anal Biochem ; 384(1): 74-8, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18845122

RESUMO

Porphyrinogens serve as substrates for three heme biosynthetic enzymes. Porphyrinogens are highly unstable and must be generated as an integral part of enzyme assays. Methods commonly employed to generate porphyrinogens include chemical reduction using sodium amalgam or sodium borohydride and enzymatic generation from porphobilinogen. Chemical reduction yields porphyrinogens in highly alkaline solutions with high ionic strength, whereas enzymatic generation requires purified enzymes, deproteination, and complete buffer replacement. This article describes an improved method for reducing porphyrins to porphyrinogens using palladium on carbon as a catalyst under hydrogen at ambient temperature and pressure in the dark. The palladium catalyst is removed by filtration, the filtrate is blown dry with an inert gas, and the dried porphyrinogen can be dissolved in a buffer compatible with biological studies.


Assuntos
Carbono/química , Paládio/química , Porfirinogênios/química , Porfirinas/química , Catálise , Oxirredução
12.
Blood ; 112(12): 4723-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18809758

RESUMO

Hepatic siderosis is common in patients with porphyria cutanea tarda (PCT). Mutations in the hereditary hemochromatosis (hh) gene (HFE) explain the siderosis in approximately 20% patients, suggesting that the remaining occurrences result from additional genetic and environmental factors. Two genes known to modify iron loading in hh are hepcidin (HAMP) and hemojuvelin (HJV). To determine if mutations in or expression of these genes influenced iron overload in PCT, we compared sequences of HAMP and HJV in 96 patients with PCT and 88 HFE C282Y homozygotes with marked hepatic iron overload. We also compared hepatic expression of these and other iron-related genes in a group of patients with PCT and hh. Two intronic polymorphisms in HJV were associated with elevated serum ferritin in HFE C282Y homozygotes. No exonic polymorphisms were identified. Sequencing of HAMP revealed exonic polymorphisms in 2 patients with PCT: heterozygosity for a G-->A transition (G71D substitution) in one and heterozygosity for an A-->G transition (K83R substitution) in the other. Hepatic HAMP expression in patients with PCT was significantly reduced, regardless of HFE genotype, when compared with patients with hh but without PCT with comparable iron overload. These data indicate that the hepatic siderosis associated with PCT likely results from dysregulated HAMP.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Porfiria Cutânea Tardia/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Análise Mutacional de DNA , Regulação para Baixo/fisiologia , Feminino , Perfilação da Expressão Gênica , Proteína da Hemocromatose , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Lactente , Hepatopatias/etiologia , Hepatopatias/genética , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Porfiria Cutânea Tardia/complicações , Siderose/etiologia , Siderose/genética , Adulto Jovem
13.
Cell Metab ; 8(2): 146-56, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18680715

RESUMO

Mammalian iron homeostasis is regulated by the interaction of the liver-produced peptide hepcidin and its receptor, the iron transporter ferroportin. Hepcidin binds to ferroportin resulting in degradation of ferroportin and decreased cellular iron export. We identify the hepcidin-binding domain (HBD) on ferroportin and show that a synthetic 19 amino acid peptide corresponding to the HBD recapitulates the characteristics and specificity of hepcidin binding to cell-surface ferroportin. The binding of mammalian hepcidin to ferroportin or the HBD shows an unusual temperature dependency with an increased rate of dissociation at temperatures below 15 degrees C. The increased rate of dissociation is due to temperature- dependent changes in hepcidin structure. In contrast, hepcidin from poikilothermic vertebrates, such as fish or frogs, binds the HBD in a temperature-independent fashion. The affinity of hepcidin for the HBD permits a rapid, sensitive assay of hepcidin from all species and yields insights into the evolution of hepcidin.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Anuros/metabolismo , Sítios de Ligação/fisiologia , Temperatura Corporal/fisiologia , Linhagem Celular , Sequência Conservada/fisiologia , Evolução Molecular , Peixes/metabolismo , Hepcidinas , Humanos , Peptídeos/química , Peptídeos/metabolismo , Estrutura Terciária de Proteína/fisiologia , Especificidade da Espécie
14.
J Biol Chem ; 282(52): 37501-7, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17971451

RESUMO

Hereditary hemochromatosis is an inherited disorder of increased iron absorption that can result in cirrhosis, diabetes, and other morbidities. We have investigated the mechanisms underlying supranormal glucose tolerance despite decreased insulin secretion in a mouse model of hemochromatosis with deletion of the hemochromatosis gene (Hfe(-/-)). Hfe(-/-) mice on 129Sv or C57BL/6J backgrounds have decreased glucose excursions after challenge compared with controls. In the C57BL/6J/ Hfe(-/-), for example, incremental area under the glucose curve is reduced 52% (p < 0.001) despite decreased serum insulin, and homeostasis model assessment insulin resistance is decreased 50% (p < 0.05). When studied by the euglycemic clamp technique 129Sv/Hfe(-/-) mice exhibit a 20% increase in glucose disposal (p < 0.05) at submaximal insulin but no increase at maximal insulin compared with wild types. [1,2-(13)C]D-glucose clearance from plasma is significantly increased in Hfe(-/-) mice (19%, p < 0.05), and lactate derived from glycolysis is elevated 5.1-fold in Hfe(-/-) mice (p < 0.0001). Basal but not insulin-stimulated glucose uptake is elevated in isolated soleus muscle from Hfe(-/-) mice (p < 0.03). Compared with controls Hfe(-/-) mice exhibit no differences in serum lipid, insulin, glucagon, or thyroid hormone levels; adiponectin levels are elevated 41% (p < 0.05), and the adiponectin message in adipocytes is increased 83% (p = 0.04). Insulin action measured by phosphorylation of Akt is not enhanced in muscle, but phosphorylation of AMP-dependent kinase is increased. We conclude that supranormal glucose tolerance in iron overload is characterized by increased glucose disposal that does not result from increased insulin action. Instead, the Hfe(-/-) mice demonstrate increased adiponectin levels and activation of AMP-dependent kinase.


Assuntos
Regulação da Expressão Gênica , Hemocromatose/genética , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Adipócitos/metabolismo , Animais , Modelos Animais de Doenças , Glucose/metabolismo , Ferro/metabolismo , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Transdução de Sinais
15.
Blood ; 110(10): 3780-3, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17726163

RESUMO

The term hemochromatosis represents a group of inherited disorders leading to iron overload. Mutations in HFE, HJV, and TfR2 cause autosomal-recessive forms of hemochromatosis. Mutations in ferroportin, however, result in dominantly inherited iron overload. Some mutations (H32R and N174I) in ferroportin lead to macrophage iron loading, while others (NI44H) lead to hepatocyte iron loading. Expression of H32R or N174I ferroportin cDNA in zebrafish leads to severe iron-limited erythropoiesis. Expression of wild-type ferroportin or hepcidin-resistant ferroportin (N144H) does not affect erythropoiesis. Zebrafish provides a facile way of identifying which ferroportin mutants may lead to macrophage iron loading.


Assuntos
Proteínas de Transporte de Cátions/genética , Doenças Genéticas Inatas/etiologia , Modelos Animais , Mutação/fisiologia , Peixe-Zebra/genética , Animais , Proteínas de Transporte de Cátions/fisiologia , Embrião não Mamífero , Doenças Genéticas Inatas/genética , Ferro/metabolismo , Macrófagos/metabolismo
16.
Blood Cells Mol Dis ; 39(2): 199-205, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17493847

RESUMO

The highly variable clinical phenotype observed in patients homozygous for the C282Y mutation of the hereditary hemochromatosis gene (HFE) is likely due to the influence of non-HFE modifier genes. The primary functional abnormality causing iron overload in hemochromatosis is hyper-absorption of dietary iron. We found that iron absorption in inbred mice varies in a strain-specific manner, as does the pattern of iron distribution to the liver and spleen. A/J mice absorbed approximately twice the amount of 59Fe delivered by gavage compared to the C57BL/6 strain. Genetic comparisons between A/J and C57BL/6 were facilitated by the availability of consomic chromosome substitution strains (CSS). Each CSS has an individual chromosome pair from A/J on an otherwise C57BL/6J background. We found that iron absorption and iron content in liver and in spleen were continuous variables suggesting that each trait is under multigenic control. No trait co-segregated among the CSS. Chromosome 5 from A/J, however, imparted the highest iron absorption phenotype and multiple CSS had absorption levels equivalent to A/J. Chromosomes 9 and X were associated with high spleen iron content. These data suggest that multiple genes contribute to the regulation of iron absorption and that individual organ iron phenotypes are independently regulated.


Assuntos
Mapeamento Cromossômico , Cromossomos de Mamíferos , Padrões de Herança , Ferro/metabolismo , Animais , Ferro/análise , Fígado/química , Camundongos , Camundongos Mutantes , Fenótipo , Baço/química
17.
Proc Natl Acad Sci U S A ; 104(12): 5079-84, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17360334

RESUMO

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


Assuntos
Porfiria Cutânea Tardia/etiologia , Porfirinas/farmacologia , Uroporfirinogênio Descarboxilase/antagonistas & inibidores , Animais , Cromatografia Líquida de Alta Pressão , Citosol/efeitos dos fármacos , Citosol/enzimologia , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Extratos Hepáticos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Porfiria Cutânea Tardia/induzido quimicamente , Porfirinas/análise , Porfirinas/química , Proteínas Recombinantes/metabolismo , Uroporfirinogênios/química
18.
Transl Res ; 149(2): 85-91, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17240319

RESUMO

Hepatoerythropoietic porphyria (HEP) is a rare form of porphyria in humans. The disorder is caused by homozygosity or compound heterozygosity for mutations of the uroporphyrinogen decarboxylase (URO-D) gene. Subnormal URO-D activity results in accumulation of uroporphyrin in the liver, which ultimately mediates the photosensitivity that clinically characterizes HEP. Two previously undescribed URO-D mutations found in a 2-year-old Caucasian boy with HEP, a maternal nonsense mutation (Gln71Stop), and a paternal missense mutation (Gly168Arg) are reported here. Recombinant Gly168Arg URO-D retained 65% of wild-type URO-D activity and studies in Epstein-Barr Virus (EBV)-transformed lymphoblasts indicated that protein levels are reduced, suggesting that the mutant protein might be subjected to accelerated turnover. The crystal structure of Gly168Arg was determined both as the apo-enzyme and with the reaction product bound. These studies revealed little distortion of the active site, but a loop containing residues 167-172 was displaced, possibly indicating small changes in the catalytic geometry or in substrate binding or increased accessibility to a cellular proteolytic pathway. A second pregnancy occurred in this family, and in utero genotyping revealed a fetus heterozygous for the maternal nonsense mutation (URO-D genotype WT/Gln71Stop). A healthy infant was born with no clinical evidence of porphyria.


Assuntos
Códon sem Sentido , Mutação de Sentido Incorreto , Porfiria Hepatoeritropoética/genética , Uroporfirinogênio Descarboxilase/genética , Adulto , Linhagem Celular Transformada , Pré-Escolar , Coproporfirinogênios/biossíntese , Coproporfirinogênios/química , Cristalografia , Ativação Enzimática/genética , Feminino , Genótipo , Heterozigoto , Homozigoto , Humanos , Masculino , Fenótipo , Porfiria Hepatoeritropoética/metabolismo , Gravidez , Diagnóstico Pré-Natal , Estrutura Terciária de Proteína , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo
19.
Blood ; 109(6): 2618-21, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17148589

RESUMO

Congenital erythropoietic porphyria (CEP), an autosomal recessive disorder, is due to mutations of uroporphyrinogen III synthase (UROS). Deficiency of UROS results in excess uroporphyrin I, which causes photosensitization. We evaluated a 3-year-old boy with CEP. A hypochromic, microcytic anemia was present from birth, and platelet counts averaged 70 x 10(9)/L (70,000/microL). Erythrocyte UROS activity was 21% of controls. Red cell morphology and globin chain labeling studies were compatible with beta-thalassemia. Hb electrophoresis revealed 36.3% A, 2.4% A(2), 59.5% F, and 1.8% of an unidentified peak. No UROS or alpha- and beta-globin mutations were found in the child or the parents. The molecular basis of the phenotype proved to be a mutation of GATA1, an X-linked transcription factor common to globin genes and heme biosynthetic enzymes in erythrocytes. A mutation at codon 216 in the child and on one allele of his mother changed arginine to tryptophan (R216W). This is the first report of a human porphyria due to a mutation in a trans-acting factor and the first association of CEP with thalassemia and thrombocytopenia. The Hb F level of 59.5% suggests a role for GATA-1 in globin switching. A bone marrow allograft corrected both the porphyria and the thalassemia.


Assuntos
Fator de Transcrição GATA1/genética , Mutação/genética , Porfiria Eritropoética/genética , Ativação Transcricional/genética , Sequência de Aminoácidos , Pré-Escolar , Feminino , Fator de Transcrição GATA1/química , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Porfiria Eritropoética/etiologia , Porfiria Eritropoética/patologia , Ligação Proteica , Dedos de Zinco
20.
Biochim Biophys Acta ; 1763(7): 723-36, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16839620

RESUMO

Most iron in mammalian systems is routed to mitochondria to serve as a substrate for ferrochelatase. Ferrochelatase inserts iron into protoporphyrin IX to form heme which is incorporated into hemoglobin and cytochromes, the dominant hemoproteins in mammals. Tissue-specific regulatory features characterize the heme biosynthetic pathway. In erythroid cells, regulation is mediated by erythroid-specific transcription factors and the availability of iron as Fe/S clusters. In non-erythroid cells the pathway is regulated by heme-mediated feedback inhibition. All of the enzymes in the heme biosynthetic pathway have been crystallized and the crystal structures have permitted detailed analyses of enzyme mechanisms. All of the genes encoding the heme biosynthetic enzymes have been cloned and mutations of these genes are responsible for a group of human disorders designated the porphyrias and for X-linked sideroblastic anemia. The biochemistry, structural biology and the mechanisms of tissue-specific regulation are presented in this review along with the key features of the porphyric disorders.


Assuntos
Heme/biossíntese , Mamíferos/metabolismo , Animais , Cristalografia por Raios X , Ferroquelatase/química , Ferroquelatase/metabolismo , Modelos Moleculares , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA