Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Biol Chem ; 286(15): 13127-33, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21343304

RESUMO

A single mutation (C73R) in the enzyme uroporphyrinogen III synthase (UROIIIS) is responsible for more than one-third of all of the reported cases of the rare autosomal disease congenital erythropoietic porphyria (CEP). CEP patients carrying this hotspot mutation develop a severe phenotype of the disease, including reduced life expectancy. Here, we have investigated the molecular basis for the functional deficit in the mutant enzyme both in vitro and in cellular systems. We show that a Cys in position 73 is not essential for the catalytic activity of the enzyme but its mutation to Arg speeds up the process of irreversible unfolding and aggregation. In the mammalian cell milieu, the mutant protein levels decrease to below the detection limit, whereas wild type UROIIIS can be detected easily. The disparate response is not produced by differences at the level of transcription, and the results with cultured cells and in vitro are consistent with a model where the protein becomes very unstable upon mutation and triggers a degradation mechanism via the proteasome. Mutant protein levels can be restored upon cell treatment with the proteasome inhibitor MG132. The intracellularly recovered C73R-UROIIIS protein shows enzymatic activity, paving the way for a new line of therapeutic intervention in CEP patients.


Assuntos
Mutação de Sentido Incorreto , Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/metabolismo , Substituição de Aminoácidos , Catálise , Linhagem Celular , Cisteína/genética , Cisteína/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Humanos , Leupeptinas/farmacologia , Porfiria Eritropoética/tratamento farmacológico , Porfiria Eritropoética/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Uroporfirinogênio III Sintetase/genética
2.
J Gene Med ; 12(8): 637-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20586119

RESUMO

BACKGROUND: Congenital erythropoietic porphyria (CEP) is a severe autosomal recessive disorder characterized by a deficiency in uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme biosynthetic pathway. We recently demonstrated the definitive cure of a murine model of CEP by lentiviral vector-mediated hematopoietic stem cell (HSC) gene therapy. In the perspective of a gene therapy clinical trial, human cellular models are required to evaluate the therapeutic potential of lentiviral vectors in UROS-deficient cells. However, the rare incidence of the disease makes difficult the availability of HSCs derived from patients. METHODS: RNA interference (RNAi) has been used to develop a new human model of the disease from normal cord blood HSCs. Lentivectors were developed for this purpose. RESULTS: We were able to down-regulate the level of human UROS in human cell lines and primary hematopoietic cells. A 97% reduction of UROS activity led to spontaneous uroporphyrin accumulation in human erythroid bone marrow cells of transplanted immune-deficient mice, recapitulating the phenotype of cells derived from patients. A strong RNAi-induced UROS inhibition allowed us to test the efficiency of different lentiviral vectors with the aim of selecting a safer vector. Restoration of UROS activity in these small hairpin RNA-transduced CD34(+) cord blood cells by therapeutic lentivectors led to a partial correction of the phenotype in vivo. CONCLUSIONS: The RNAi strategy is an interesting new tool for preclinical gene therapy evaluation.


Assuntos
Terapia Genética/métodos , Porfiria Eritropoética/terapia , Interferência de RNA , Animais , Modelos Animais de Doenças , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células K562 , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , Uroporfirinogênio III Sintetase/metabolismo
3.
Mol Med ; 16(9-10): 381-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20485863

RESUMO

The first feline model of human congenital erythropoietic porphyria (CEP) due to deficient uroporphyrinogen III synthase (URO-synthase) activity was identified by its characteristic clinical phenotype, and confirmed by biochemical and molecular genetic studies. The proband, an adult domestic shorthair cat, had dark-red urine and brownish discolored teeth with red fluorescence under ultraviolet light. Biochemical studies demonstrated markedly increased uroporphyrinogen I in urine and plasma (2,650- and 10,700-fold greater than wild type, respectively), whereas urinary 5-aminolevulinic acid and porphobilinogen were lower than normal. Erythrocytic URO-synthase activity was <1% of mean wild-type activity, confirming the diagnosis and distinguishing it from feline phenocopies having acute intermittent porphyria. Sequencing of the affected cat's UROS gene revealed two missense mutations, c.140C>T (p.S47F) in exon 3 and c.331G>A (p.G111S) in exon 6, both of which were homozygous, presumably owing to parental consanguinity. Neither was present in 100 normal cat alleles. Prokaryotic expression and thermostability studies of the purified monomeric wild-type, p.S47F, p.G111S, and p.S47F/G111S enzymes showed that the p.S47F enzyme had 100% of wild-type specific activity but ~50% decreased thermostability, whereas the p.G111S and p.S47F/G111S enzymes had about 60% and 20% of wild-type specific activity, respectively, and both were markedly thermolabile. Molecular modeling results indicated that the less active/less stable p.G111S enzyme was further functionally impaired by a structural interaction induced by the presence of the S47F substitution. Thus, the synergistic interaction of two rare amino acid substitutions in the URO-synthase polypeptide caused the feline model of human CEP.


Assuntos
Doenças do Gato/enzimologia , Doenças do Gato/genética , Homozigoto , Mutação de Sentido Incorreto/genética , Porfiria Eritropoética/veterinária , Porfirinas/metabolismo , Uroporfirinogênio III Sintetase/genética , Animais , Doenças do Gato/sangue , Doenças do Gato/urina , Gatos , Eritrócitos/metabolismo , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Porfiria Eritropoética/sangue , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/urina , Porfirinas/sangue , Porfirinas/urina , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo
4.
Genomics ; 87(1): 84-92, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16314073

RESUMO

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


Assuntos
Substituição de Aminoácidos , Mutação de Sentido Incorreto , Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/genética , Animais , Transplante de Medula Óssea , Modelos Animais de Doenças , Terapia Genética , Camundongos , Camundongos Transgênicos , Porfiria Eritropoética/patologia , Porfiria Eritropoética/terapia , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinas/metabolismo
5.
An. bras. dermatol ; 73(2): 119-23, mar.-abr. 1998. ilus
Artigo em Português | LILACS | ID: lil-242355

RESUMO

A porfiria erotropoiética congênita é doença que resulta da deficiência da enzima uroporfirinogênio III sintease, do metabolismo do grupo heme da hemoglobina. Essa anormalidade desvia a produçäo de metabólitos dessa síntese para isômeros da série uroporfirina I e coproporfirina I), que se acumulam em praticamente todos os tecidos do organismo. Altos níveis de uroporfirina I säo encontrados na urina e nas fezes. Os depósitos cutâneos da uroporfirina induzem à reaçäo fototóxica com formaçäo de bolhas subepidérmicas. O presente artigo constitui-se no relato do caso clínico de um paciente de 13 anos de idade, do sexo masculino, cujo quadro clínico era caracterizado pela presença de vesículas e bolhas nas áreas da pele fotoexpostas, associada a outras manifestaçöes, como eritrodontia, hipertricose, atrofia e reabsorçäo das falanges, conjuntivite, cicatrizes cutâneas, mília, contraturas dos dedos, hiper e hipopigmentaçäo cutânea. A investigaçäo clínica e laboratorial dos órgäos internos demonstrou apenas discreta hepatoesplenomegalia. O estudo das porfirinas revelou aumentos muito significativos na excreçäo urinário da uroporfirina I e da coproporfirina I


Assuntos
Humanos , Masculino , Adolescente , Porfiria Eritropoética , Porfiria Eritropoética/complicações , Porfiria Eritropoética/diagnóstico , Porfiria Eritropoética/enzimologia , Porfirinas/urina , Manifestações Cutâneas , Pele/patologia , Uroporfirinogênio III Sintetase/deficiência
6.
Transfus Clin Biol ; 4(3): 263-6, 1997.
Artigo em Francês | MEDLINE | ID: mdl-9264783

RESUMO

CEP is a rare disease inherited as an autosomal recessive trait and characterized by an overproduction and accumulation of porphyrins in the bone-marrow. Because the predominant site of metabolic expression of the disease is the erythropoietic system, bone marrow transplantation represents a curative treatment for patients with severe phenotypes. This treatment can be considered in severe cases when the disease appears in the first few years of life. When bone marrow transplantation is not possible, gene therapy by transplantation of genetically modified hematopoietic cells is an attractive alternative for the future. In this report, we present the restoration of enzymatic activity and the metabolic correction of deficient cells in vitro after transduction with retroviral vectors. The future availability of a mouse model of the disease will permit ex vivo gene therapy experiments on the entire animal.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Porfiria Eritropoética/terapia , Animais , Medula Óssea/enzimologia , Células da Medula Óssea , Modelos Animais de Doenças , Vetores Genéticos , Camundongos , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/genética , Retroviridae , Uroporfirinogênio III Sintetase/metabolismo
8.
Arch Pediatr ; 2(8): 755-61, 1995 Aug.
Artigo em Francês | MEDLINE | ID: mdl-7550841

RESUMO

BACKGROUND: Congenital erythropoietic porphyria, an autosomal recessive disease, is characterized by deficiency of uroporphyrinogen III synthase. Clinical variability of the disease is related to the different mutations found in the patients. CASE REPORT: A newborn suffered one hour after birth from jaundice and polypnea with acute hemolysis. Severe cutaneous photosensitivity occurred after phototherapy. Congenital erythropoietic porphyria was suspected because of reddish-colored urine and confirmed by porphyrin analyses. The baby died one month later due to severe hemolytic anemia with hepatic failure. Uroporphyrinogen III synthase activity was decreased by 99% in bone marrow cells and established lymphoblastoid cells from the patient. Molecular biology studies demonstrated the presence of the Cys 73-->Arg substitution at the homozygous state in the patient. CONCLUSION: This mutation, the most frequently found in this disease, is responsible for a severe phenotype. Molecular characterization provides genotype/phenotype correlations in this porphyria and allows to clarify unusual cases of porphyrias.


Assuntos
Eritroblastose Fetal/complicações , Falência Hepática/complicações , Porfiria Eritropoética/metabolismo , Doença Aguda , Evolução Fatal , Hemólise , Humanos , Recém-Nascido , Masculino , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/genética , Porfirinas/metabolismo , Uroporfirinogênio III Sintetase/metabolismo
9.
Hum Gene Ther ; 6(1): 13-20, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7703283

RESUMO

Congenital erythropoietic porphyria (CEP) is a genetic disease characterized by an overproduction and accumulation of porphyrins in bone marrow. The enzyme defect concerns uroporphyrinogen III synthase (UROIIIS), the fourth enzyme of the heme biosynthetic pathway. It is the most severe porphyria and the treatment is largely symptomatic: gene therapy would represent a great therapeutic improvement. As a step toward the development of an effective gene therapy, we have constructed two retroviral vectors, LUSN and pMFG-US (with and without the selectable marker Neo), containing a full-length human cDNA for UROIIIS. Recombinant retroviruses were obtained by transfection of the LUSN or pMFG-US plasmid into the amphotropic packaging cell line psi CRIP. For each construct, three different producing clones were selected for their high titer (LUSN) or for their ability to express the message at a high level (pMFG-US). In vitro amplification of genomic DNA from target tissue demonstrated the presence of vector sequences. Murine fibroblasts infected in vitro expressed the human enzyme efficiently, as indicated by RNA and enzymatic studies. Retroviral-mediated gene transfer was then used to introduce the UROIIIS cDNA into human deficient cells. Enzyme activity was increased from 2% (deficient fibroblasts) to 121-274% of the normal value for the different clones. Transduced cells selected with G418 presented an 18-fold increase in enzyme activity compared to the normal cells. Furthermore, high gene transfer rate into peripheral blood progenitor cells (PBPB) was documented by in vitro amplification (PCR). These results demonstrate the potential usefulness of somatic gene therapy for the treatment of CEP.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Porfiria Eritropoética/terapia , Retroviridae/genética , Uroporfirinogênio III Sintetase/genética , Células 3T3 , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Humanos , Camundongos , Dados de Sequência Molecular , Porfiria Eritropoética/enzimologia , Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA