Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mol Genet Metab ; 128(3): 363-366, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30514647

RESUMEN

Porphyria Cutanea Tarda (PCT) is a cutaneous porphyria that results from the hepatic inhibition of the heme biosynthetic enzyme uroporphyrinogen decarboxylase (UROD), and can occur either in the absence or presence of an inherited heterozygous UROD mutation (PCT subtypes 1 and 2, respectively). A heterozygous UROD mutation causes half-normal levels of UROD activity systemically, which is a susceptibility factor but is not sufficient alone to cause type 2 PCT. In both Types 1 and 2 PCT, the cutaneous manifestations are precipitated by additional factors that lead to generation of an inhibitor that more profoundly reduces hepatic UROD activity. PCT is an iron-related disorder, and many of its known susceptibility factors, which include infections (e.g. hepatitis C virus, HIV), high alcohol consumption, smoking, estrogens, and genetic traits (e.g. hemochromatosis mutations) can increase hepatic iron accumulation. Hepatoerythropoietic Porphyria (HEP) is a rare autosomal recessive disease that results from homozygosity or compound heterozygosity for UROD mutations and often causes infantile or childhood onset of both erythropoietic and cutaneous manifestations. During the 11-year period from 01/01/2007 through 12/31/2017, the Mount Sinai Porphyrias Diagnostic Laboratory provided molecular diagnostic testing for 387 unrelated patients with PCT and four unrelated patients with HEP. Of the 387 unrelated individuals tested for Type 2 PCT, 79 (20%) were heterozygous for UROD mutations. Among 26 family members of mutation-positive PCT patients, eight (31%) had the respective family mutation. Additionally, of the four unrelated HEP patients referred for UROD mutation analyses, all had homozygosity or compound heterozygosity for UROD mutations, and all eight asymptomatic family members were heterozygotes for UROD mutations. Of the UROD mutations identified, 19 were novel, including nine missense, two nonsense, one consensus splice-site, and seven insertions and deletions. These results expand the molecular heterogeneity of PCT and HEP by adding a total of 19 novel UROD mutations. Moreover, the results document the usefulness of molecular testing to confirm a genetic susceptibility trait in Type 2 PCT, confirm a diagnosis in HEP, and identify heterozygous family members.


Asunto(s)
Heterogeneidad Genética , Mutación , Porfiria Cutánea Tardía/genética , Porfiria Hepatoeritropoyética/genética , Uroporfirinógeno Descarboxilasa/genética , Niño , Familia , Tamización de Portadores Genéticos , Predisposición Genética a la Enfermedad , Hemo/biosíntesis , Humanos , Masculino , Técnicas de Diagnóstico Molecular
2.
Folia Biol (Praha) ; 61(6): 219-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26789143

RESUMEN

Porphyrias are metabolic disorders resulting from mutations in haem biosynthetic pathway genes. Hepatoerythropoietic porphyria (HEP) is a rare type of porphyria caused by the deficiency of the fifth enzyme (uroporphyrinogen decarboxylase, UROD) in this pathway. The defect in the enzymatic activity is due to biallelic mutations in the UROD gene. Currently, 109 UROD mutations are known. The human disease has an early onset, manifesting in infancy or early childhood with red urine, skin photosensitivity in sun-exposed areas, and hypertrichosis. Similar defects and links to photosensitivity and hepatopathy exist in several animal models, including zebrafish and mice. In the present study, we report a new mutation in the UROD gene in Egyptian patients with HEP. We show that the homozygous c.T163A missense mutation leads to a substitution of a conserved phenylalanine (amino acid 55) for isoleucine in the enzyme active site, causing a dramatic decrease in the enzyme activity (19 % of activity of wild-type enzyme). Inspection of the UROD crystal structure shows that Phe-55 contacts the substrate and is located in the loop that connects helices 2 and 3. Phe-55 is strictly conserved in both prokaryotic and eukaryotic UROD. The F55I substitution likely interferes with the enzyme-substrate interaction.


Asunto(s)
Alelos , Predisposición Genética a la Enfermedad , Mutación/genética , Porfiria Hepatoeritropoyética/enzimología , Porfiria Hepatoeritropoyética/genética , Uroporfirinógeno Descarboxilasa/genética , Adolescente , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Cicatriz/complicaciones , Análisis Mutacional de ADN , Egipto , Familia , Femenino , Humanos , Hipertricosis/complicaciones , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Tasa de Mutación , Linaje , Porfiria Hepatoeritropoyética/complicaciones , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Uroporfirinógeno Descarboxilasa/química
4.
Nat Genet ; 20(3): 239-43, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9806541

RESUMEN

Defects in the enzymes involved in the haem biosynthetic pathway can lead to a group of human diseases known as the porphyrias. yquem (yqe(tp61)) is a zebrafish mutant with a photosensitive porphyria syndrome. Here we show that the porphyric phenotype is due to an inherited homozygous mutation in the gene encoding uroporphyrinogen decarboxylase (UROD); a homozygous deficiency of this enzyme causes hepatoerythropoietic porphyria (HEP) in humans. The zebrafish mutant represents the first genetically 'accurate' animal model of HEP, and should be useful for studying the pathogenesis of UROD deficiency and evaluating gene therapy vectors. We rescued the mutant phenotype by transient and germline expression of the wild-type allele.


Asunto(s)
Porfiria Hepatoeritropoyética/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN/genética , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Terapia Genética , Homocigoto , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Porfiria Hepatoeritropoyética/enzimología , Porfiria Hepatoeritropoyética/terapia , Homología de Secuencia de Aminoácido , Transfección , Uroporfirinógeno Descarboxilasa/deficiencia , Uroporfirinógeno Descarboxilasa/genética , Pez Cebra/metabolismo
5.
Nat Genet ; 26(2): 216-20, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017081

RESUMEN

The sequencing of the zebrafish genome should be completed by the end of 2002. Direct assignment of function on the basis of this information would be facilitated by the development of a rapid, targeted 'knockdown' technology in this model vertebrate. We show here that antisense, morpholino-modified oligonucleotides (morpholinos) are effective and specific translational inhibitors in zebrafish. We generated phenocopies of mutations of the genes no tail (ref. 2), chordin (ref. 3), one-eyed-pinhead (ref. 4), nacre (ref. 5) and sparse (ref. 6), removing gene function from maternal through post-segmentation and organogenesis developmental stages. We blocked expression from a ubiquitous green fluorescent protein (GFP) transgene, showing that, unlike tissue-restricted limitations found with RNA-based interference in the nematode, all zebrafish cells readily respond to this technique. We also developed also morpholino-based zebrafish models of human disease. Morpholinos targeted to the uroporphyrinogen decarboxylase gene result in embryos with hepatoerythropoietic porphyria. We also used morpholinos for the determination of new gene functions. We showed that embryos with reduced sonic hedgehog (ref. 9) signalling and reduced tiggy-winkle hedgehog (ref. 10) function exhibit partial cyclopia and other specific midline abnormalities, providing a zebrafish genetic model for the common human disorder holoprosencephaly. Conserved vertebrate processes and diseases are now amenable to a systematic, in vivo, reverse-genetic paradigm using zebrafish embryos.


Asunto(s)
Genoma , Oligodesoxirribonucleótidos Antisentido/farmacología , Transactivadores , Uroporfirinógeno Descarboxilasa/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes , Proteínas Hedgehog , Humanos , Proteínas Luminiscentes/genética , Morfogénesis , Morfolinas , Porfiria Hepatoeritropoyética/genética , Proteínas/genética , Transducción de Señal , Uroporfirinógeno Descarboxilasa/deficiencia , Pez Cebra/embriología , Proteínas de Pez Cebra , Cigoto
6.
Mol Genet Metab ; 105(4): 629-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22382040

RESUMEN

Porphyria cutanea tarda (PCT) is caused by decreased activity of uroporphyrinogen decarboxylase (UROD) in the liver. The disease usually occurs in adulthood and is characterized by cutaneous photosensitivity, hyperpigmentation, skin fragility and hypertrichosis, due to the accumulation of porphyrins produced by oxidation of uroporphyrinogen and other highly carboxylated porphyrinogens overproduced as a result of the enzyme deficiency. PCT is generally sporadic, but about 20-30% of patients have familial-PCT (F-PCT) which is associated with heterozygosity of mutations in the UROD gene. In the present study we have found the molecular defect in seventeen unrelated Argentinean patients with F-PCT, identifying a total of eleven UROD gene mutations: four novel and seven previously described. The novel mutations were: a guanine insertion at the 5' splice junction of intron 2, a three nucleotide deletion causing the lost of valine 90, a deletion of 22 bp in exon 6 and a deletion of part of the polyadenylation signal. Prokaryotic expression studies showed that the novel amino acid deletion resulted in an inactive protein. Mutations c.10insA and p.M165R, previously found in Argentinean patients, were recurrent in this study; they are the most frequent in Argentina accounting for 40% of the mutant alleles characterized to date.


Asunto(s)
Predisposición Genética a la Enfermedad , Mutación/genética , Porfiria Cutánea Tardía/enzimología , Porfiria Cutánea Tardía/genética , Uroporfirinógeno Descarboxilasa/genética , Adolescente , Adulto , Argentina , Niño , Preescolar , ADN/genética , Exones/genética , Femenino , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia , Adulto Joven
7.
F1000Res ; 11: 945, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37360939

RESUMEN

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.


Asunto(s)
Hemocromatosis , Porfiria Cutánea Tardía , Masculino , Humanos , Anciano , Porfiria Cutánea Tardía/tratamiento farmacológico , Porfiria Cutánea Tardía/genética , Hidroxicloroquina/uso terapéutico , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo , Hemocromatosis/genética
8.
Blood Cells Mol Dis ; 47(4): 249-54, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21880518

RESUMEN

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.


Asunto(s)
Citocromo P-450 CYP1A2/genética , Porfiria Cutánea Tardía/genética , Animales , Citocromo P-450 CYP1A2/metabolismo , Modelos Animales de Enfermedad , Genotipo , Hierro/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Porfiria Cutánea Tardía/dietoterapia , Porfiria Cutánea Tardía/metabolismo , Porfirinas/metabolismo , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo
9.
Br J Dermatol ; 165(3): 499-505, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21668429

RESUMEN

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.


Asunto(s)
Mutación Missense/genética , Porfiria Hepatoeritropoyética/genética , Uroporfirinógeno Descarboxilasa/genética , Cromatografía Líquida de Alta Presión , Eritrocitos/enzimología , Femenino , Técnicas de Genotipaje , Homocigoto , Humanos , Proteínas Recombinantes , Uroporfirinógeno Descarboxilasa/metabolismo , Adulto Joven
10.
Chem Res Toxicol ; 23(4): 712-23, 2010 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-20099833

RESUMEN

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


Asunto(s)
Porfirias Hepáticas/genética , Animales , Modelos Animales de Enfermedad , Hemocromatosis/genética , Humanos , Hierro/metabolismo , Hígado/enzimología , Ratones , Fármacos Fotosensibilizantes/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Porfirias Hepáticas/inducido químicamente , Porfirinas/metabolismo , Uroporfirinógeno Descarboxilasa/genética
11.
Clin Chem ; 55(4): 795-803, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19233912

RESUMEN

BACKGROUND: Porphyria cutanea tarda (PCT) occurs in sporadic (sPCT) and familial (fPCT) forms, which are generally clinically indistinguishable and have traditionally been differentiated by erythrocyte uroporphyrinogen decarboxylase (UROD, EC 4.1.1.37) activity. We used UROD gene sequencing as the reference standard in assessing the diagnostic accuracy of UROD activity, evaluating the mutation spectrum of the UROD gene, determining the frequency and disease attributes of PCT and its subtypes in Norway, and developing diagnostic models that use clinical and laboratory characteristics for differentiating fPCT and sPCT. METHODS: All consecutive patients with PCT diagnosed within a 6-year period were used for incidence calculations. UROD activity analysis, UROD gene sequencing, analysis of hemochromatosis mutations, and registration of clinical and laboratory data were carried out for 253 patients. RESULTS: Fifty-three percent of the patients had disease-relevant mutations, 74% of which were c.578G>C or c.636+1G>C. The UROD activity at the optimal cutoff had a likelihood ratio (LR) of 9.2 for fPCT, whereas a positive family history had an LR of 19. A logistic regression model indicated that low UROD activity, a high uroporphyrin-heptaporphyrin ratio, a young age at diagnosis, male sex, and low alcohol consumption were predictors of fPCT. The incidence of PCT was 1 in 100 000. CONCLUSIONS: Two commonly occurring mutations are responsible for the high frequency of fPCT in Norway. UROD activity has a high diagnostic accuracy for differentiating the 2 PCT types, and a model that takes into account both clinical information and laboratory test results can be used to predict fPCT.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Porfiria Cutánea Tardía/diagnóstico , Porfiria Cutánea Tardía/genética , Secuencia de Bases , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Genotipo , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Mutación/genética , Porfiria Cutánea Tardía/epidemiología , Porfiria Cutánea Tardía/metabolismo , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo
12.
Clin Genet ; 75(4): 346-53, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19419417

RESUMEN

Porphyria cutanea tarda (PCT) arises from decreased hepatic activity of uroporphyrinogen decarboxylase (UROD). Both genetic and environmental factors interplay in the precipitation of clinically overt PCT, but these factors may vary between different geographic areas. Decreased activity of UROD in erythrocytes was used to identify patients with UROD mutations among a group of 130 Spanish PCT patients. Nineteen patients (14.6%) were found to harbor a mutation in the UROD gene. Eight mutations were novel: M1I, 5del10, A22V, D79N, F84I, Q116X, T141I and Y182C. Five others were previously described: F46L, V134Q, R142Q, P150L and E218G. The new missense mutations and P150L were expressed in Escherichia coli. D79N and P150L resulted in proteins that were localized to inclusion bodies. The other mutations produced recombinant proteins that were purified and showed reduced activity (range: 2.3-73.2% of wild type). These single amino acid changes were predicted to produce complex structural alterations and/or reduced stability of the enzyme. Screening of relatives of the probands showed that 37.5% of mutation carriers demonstrated increased urinary porphyrins. This study emphasizes the role of UROD mutations as a strong risk factor for PCT even in areas where environmental factors (hepatitis C virus) have been shown to be highly associated with the disease.


Asunto(s)
Mutación , Porfiria Cutánea Tardía/enzimología , Uroporfirinógeno Descarboxilasa/genética , Adulto , Anciano , Anciano de 80 o más Años , Clonación Molecular , Análisis Mutacional de ADN , Eritrocitos/enzimología , Escherichia coli/genética , Salud de la Familia , Humanos , Persona de Mediana Edad , Mutación Missense , Porfiria Cutánea Tardía/etiología , Porfiria Cutánea Tardía/genética , Porfirinas/orina , Adulto Joven
13.
Science ; 234(4777): 732-4, 1986 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-3775362

RESUMEN

Uroporphyrinogen decarboxylase deficiency in man is responsible for familial porphyria cutanea tarda and hepatoerythropoietic porphyria. A recent study of a family with hepatoerythropoietic porphyria showed that the enzyme defect resulted from rapid degradation of the protein in vivo. Cloning and sequencing of a complementary DNA for the mutated gene revealed that the mutation was due to the replacement of a glycine residue by a glutamic acid residue at position 281. This base change leads to a protein that is very rapidly degraded in the presence of cell lysate. Characterization of the mutation will allow comparison of this defect in a homozygous patient with defects in other patients with familial porphyria cutanea tarda.


Asunto(s)
Carboxiliasas/genética , Porfirias/genética , Uroporfirinógeno Descarboxilasa/genética , Secuencia de Aminoácidos , Clonación Molecular , ADN/genética , Humanos , Hepatopatías/genética , Mutación , Enfermedades de la Piel/genética , Relación Estructura-Actividad , Uroporfirinógeno Descarboxilasa/deficiencia , Uroporfirinógeno Descarboxilasa/metabolismo
14.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 61-5, 2009 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-19268003

RESUMEN

Hepatoerythropoietic Porphyria (HEP) is the rare homozygous form of Porphyria Cutanea Tarda (PCT). It is characterized clinically by the early onset of severe skin manifestations which can be confused with Congenital Erythropoietic Porphyria (CEP) or with PCT when the symptoms are mild. We describe the case of a 14 year-old child with skin manifestations similar to those observed in PCT. The biochemical assays ruled out a CEP as well as they suggested the development of a HEP. Although his symptoms were not severe enough to be HEP, the enzymatic activity was dramatically reduced to a 5% of normal values and the molecular analysis revealed the presence of two already known different mutations on the patient's URO-D gene, c.703 C>T and IVS9-1. Each parent carry one of the mutations, but they were absent in the brother. This is the first Argentinean HEP case ever described which appeared in a compound heterozygous form and less residual URO-D activity but associated to a mild phenotype.


Asunto(s)
Porfiria Hepatoeritropoyética/diagnóstico , Porfiria Hepatoeritropoyética/genética , Adolescente , Argentina , Análisis Mutacional de ADN , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Porfiria Hepatoeritropoyética/patología , Porfiria Hepatoeritropoyética/orina , Uroporfirinógeno Descarboxilasa/genética
15.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 40-5, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19656450

RESUMEN

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.


Asunto(s)
Uroporfirinógeno Descarboxilasa/metabolismo , Adulto , Anciano , Dominio Catalítico , Cristalografía por Rayos X , Femenino , Genotipo , Heterocigoto , Humanos , Cinética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Porfiria Cutánea Tardía/etiología , Porfiria Cutánea Tardía/genética , Estabilidad Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Uroporfirinógeno Descarboxilasa/química , Uroporfirinógeno Descarboxilasa/genética
16.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 46-54, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19656451

RESUMEN

Most rodent models of porphyria cutanea tarda (PCT) share in common the administration of iron and agents that induce transcription of cytochrome P450s. Dissection of changes related to porphyrin accumulation required generation of a genetic model free from exogenous precipitants. Mice heterozygous for a null Urod mutation and homozygous for null Hfe alleles spontaneously develop major increases in hepatic and urinary porphyrins several months after weaning but the high % uroporphyrin signature of PCT is established earlier, before total hepatic and urinary porphyrins rise. Total porphyrin levels eventually plateau at higher levels in females than in males. Porphyrinogens were the dominant tetrapyrroles accumulating in hepatocytes. Hepatic Urod activity is markedly reduced but total hepatic heme content does not diminish. Microsomal heme, however, is reduced and in vitro metabolism of prototype substrates showed that some but not all cytochrome P450 activities are reduced. High hepatic levels of uroporphyrinogen are also associated with increased glutathione S-transferase activity and elevated mRNA of 2 transporters, Abcc1 and Abcc4. This murine model of familial PCT affords the opportunity to study changes in porphyrinogen and porphyrin accumulation and transport in the absence of exogenous factors that alter P450 activity and transmembrane transporters.


Asunto(s)
Porfiria Cutánea Tardía/genética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Glutatión Transferasa/metabolismo , Hemo/análisis , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Hígado/metabolismo , Estudios Longitudinales , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Porfiria Cutánea Tardía/patología , Porfirinas/sangre , Porfirinas/orina , Uroporfirinógeno Descarboxilasa/deficiencia , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo , Uroporfirinógenos/metabolismo
17.
Clin Exp Dermatol ; 33(5): 602-5, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18462440

RESUMEN

Three children (two boys and one girl) from the same family presented with photosensitivity, hyperpigmentation, hypertrichosis, mild skin fragility, blistering and scarring in childhood. On examination, the cutaneous lesions were found to have improved since their previous examinations. Laboratory tests showed raised plasma and urine carboxyporphyrins and decreased uroporphyrinogen decarboxylase enzyme activity in red blood cells. Triggering factors for porphyria were not detected except for a hepatitis C virus infection in the younger boy. The girl's clinical symptoms recurred in late adolescence, after iron and oestrogen treatments. Mutation analysis of the UROD gene detected two missense mutations, 19 A-->G M1V (novel) and 703C-->T P235S (previously reported), in an uncommon compound heterozygous manner in the three siblings.


Asunto(s)
Mutación Missense/genética , Porfiria Cutánea Tardía/enzimología , Uroporfirinógeno Descarboxilasa/genética , Adolescente , Adulto , Niño , Análisis Mutacional de ADN , Femenino , Heterocigoto , Humanos , Masculino , Linaje , Uroporfirinógeno Descarboxilasa/metabolismo
18.
J Clin Invest ; 86(5): 1416-22, 1990 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2243121

RESUMEN

Uroporphyrinogen decarboxylase (URO-D) is a cytosolic heme-biosynthetic enzyme that converts uroporphyrinogen to coproporphyrinogen. Defects at the uroporphyrinogen decarboxylase locus cause the human genetic disease familial porphyria cutanea tarda. A splice site mutation has been found in a pedigree with familial porphyria cutanea tarda that causes exon 6 to be deleted from the mRNA. The intron/exon junctions on either side of exon 6 fall between codons, so the resulting protein is shorter than the normal protein, missing only the amino acids coded by exon 6. The shortened protein lacks catalytic activity, is rapidly degraded when exposed to human lymphocyte lysates, and is not detectable by Western blot analysis in lymphocyte lysates derived from affected individuals. The mutation was detected in five of 22 unrelated familial porphyria cutanea tarda pedigrees tested, so it appears to be common. This is the first splice site mutation to be found at the URO-D locus, and the first mutation that causes familial porphyria cutanea tarda to be found in more than one pedigree.


Asunto(s)
Exones , Mutación , Porfirias/genética , Enfermedades de la Piel/genética , Uroporfirinógeno Descarboxilasa/genética , Secuencia de Aminoácidos , Secuencia de Bases , Células Cultivadas , Genes , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Porfirias/enzimología , Biosíntesis de Proteínas , Enfermedades de la Piel/enzimología , Uroporfirinógeno Descarboxilasa/metabolismo
19.
J Clin Invest ; 77(2): 431-5, 1986 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3753711

RESUMEN

In order to determine the molecular basis of uroporphyrinogen (URO) decarboxylase deficiency responsible for hepatoerythropoietic porphyria (HEP) and familial porphyria cutanea tarda, we used a human URO decarboxylase cDNA to analyze the organization and expression of the URO decarboxylase gene in lymphoblastoid cells from normal individuals and from two patients with HEP. We could detect neither deletions nor rearrangements in the URO decarboxylase gene. Synthesis, processing, and cell-free translation of the specific transcripts appeared to be normal. The half-life of the abnormal protein was 12 times shorter than that of the normal enzyme. The results indicate that the enzyme defect is due to a rapid degradation of the protein in vivo. This study is the first to provide information regarding the molecular mechanism responsible for the URO decarboxylase deficiency in HEP.


Asunto(s)
Carboxiliasas/deficiencia , Eritropoyesis , Hepatopatías , Porfirias/genética , Uroporfirinógeno Descarboxilasa/deficiencia , Línea Celular , ADN/genética , Semivida , Humanos , Linfocitos/metabolismo , Hibridación de Ácido Nucleico , Porfirias/enzimología , Biosíntesis de Proteínas , ARN Mensajero/genética , Uroporfirinógeno Descarboxilasa/genética
20.
Orv Hetil ; 148(23): 1095-100, 2007 Jun 10.
Artículo en Húngaro | MEDLINE | ID: mdl-17545118

RESUMEN

In the last decades, the role of oxidative stress and trace elements was proven to play an important role in the pathogenesis of more and more diseases. This is why a great importance is attributed lately to the antioxidant therapy, and lots of studies are dealing with this issue. In porphyria cutanea tarda (PCT) the biosynthesis of hem is damaged, because of the reduced activity of uroporphyrinogen-decarboxylase enzyme. The hem precursors are accumulating in blood, liver and skin. The hem precursors and porphyrin are eliminated with urine and stool. The enzyme defect is autosomal dominant. The skin symptoms are intensified by sun exposure. This is because the accumulation of uroporphyrins and heptacarboxylporphyrin in the skin causes photosensitivity, and the accumulated iron has a lipid-peroxidation effect. Besides the genetical origin, the alcohol consumption, the hepatotoxic drugs, estrogen and viral infections can also determine the development of the disease. The applied treatment is phlebotomy. In the case of PCT that appears in the field of liver damage, the accumulation of iron is responsible for the development of oxidative stress. The patient's redox homeostasis is changed, and the level of antioxidants is decreased. The redox state of liver and the effects of additional antioxidant treatment in phlebotomized PCT patients were determined by biochemical and trace element analytical methods. According to the clinical data, phlebotomy proved to be an effective treatment in PCT patients. Phlebotomy improved the phototoxic skin symptoms, but it did not improve the ratios of trace elements to each other in the blood of the patients.


Asunto(s)
Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Estrés Oxidativo , Porfiria Cutánea Tardía/metabolismo , Oligoelementos/metabolismo , Adulto , Anciano , Humanos , Hepatopatías Alcohólicas/metabolismo , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Flebotomía , Porfiria Cutánea Tardía/genética , Porfiria Cutánea Tardía/terapia , Porfirinas/metabolismo , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA