Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Genet Metab ; 128(3): 304-308, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31076252

RESUMO

Deficiency in ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway, leads to an accumulation of protoporphyrin IX (PPIX) that causes a severely painful phototoxic reaction of the skin in patients with erythropoietic protoporphyria (EPP). Besides phototoxicity of the skin, EPP patients often present with symptoms of iron deficiency in form of a microcytic and hypochromic anemia with low serum iron and ferritin. In addition, elevated aminolevulinic acid synthase 2 (ALAS2) both at the mRNA and protein levels have been observed among EPP patients. ALAS is the first enzyme in the pathway and exists in two isoforms, whereby the isoform 2 (ALAS2) is expressed exclusively in erythropoiesis. The mRNA of ALAS2 contains an iron response element (IRE) at its 5'UTR. When iron is limited, iron response element binding protein 2 (IRP2) binds to the IRE of ALAS2 mRNA and suppresses its translation. In this study, we demonstrated that iron deprivation increased the amount of ALAS2 mRNA as well as the ratio of ALAS2 to FECH mRNAs in cultured erythroleukemic K562 cells. At the protein level, however, iron deprivation in the cell line caused reductions in both enzymes as shown by the Western blot analysis. A comparable increase in the ratio of ALAS2 to FECH mRNAs was also found in EPP patients indicating an imbalance in heme biosynthesis. As iron cannot be completely missing from an organism, we assume that in EPP patients, a certain amount of ALAS2 mRNA is translated despite a partial deficiency of FECH. The increase in ALAS2 enzyme contributes to the accumulation in PPIX in the patients. Targeted inhibition of ALAS2 could therefore be a treatment option for EPP.


Assuntos
5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Ferro/metabolismo , Protoporfiria Eritropoética/enzimologia , Vias Biossintéticas , Ferroquelatase/genética , Humanos , Ferro/sangue , Proteína 2 Reguladora do Ferro/metabolismo , Proteínas Reguladoras de Ferro/metabolismo , Células K562 , Protoporfiria Eritropoética/terapia , Protoporfirinas/metabolismo
2.
Clin Genet ; 92(5): 495-502, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28075030

RESUMO

Erythropoietic protoporphyria (EPP) is a rare cutaneous and systemic disease caused by mutations in the ferrochelatase gene (FECH). The molecular underpinnings of EPP in Middle Eastern populations and relative to other ethnic groups secondary to increased consanguinity are unknown. To understand the molecular pathogenesis of Middle Eastern EPP, we surveyed clinicopathological and molecular features in 6 large consanguineous families from Lebanon and Syria presenting with cutaneous and systemic features consistent with EPP. We observed 30% increased liver disease and 20% elevated end-stage liver complications in our EPP cohort compared to EPP patients previously reported elsewhere. In addition, Middle Eastern EPP patients in our cohort exhibited uniquely an increased incidence of colon cancer. Sequence analysis revealed 2 novel non-synonymous FECH mutations in the studied families designated p.M294T and p.I230M. In addition, FECH activity was significantly decreased (6%) in fibroblasts obtained from sun-exposed sites in a patient with p.M294T mutation, whereas in sharp contrast, protected sites from the same patient exhibited 54% activity for the gene. We also found that sun-exposed fibroblasts, relative to sun-protected and control fibroblasts, exhibited suppressed growth and atypical morphology in vitro, and that these effects were alleviated when the cells were co-cultured with sun-protected fibroblasts. Our findings on the increased incidence of colon cancer in EPP patients prompted us to survey FECH expression patterns in cancer. Using publicly available microarray datasets we found that FECH mRNA was largely significantly decreased in colon adenocarcinomas relative to normal colon tissues. Our findings suggest that families with autosomal recessive EPP should be screened more extensively for systemic involvement including liver diseases and colon cancer, and point to a previously unknown yet plausible tumor suppressor role for FECH in colon malignancy.


Assuntos
Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Ferroquelatase/genética , Genes Supressores de Tumor , Protoporfiria Eritropoética/enzimologia , Protoporfiria Eritropoética/genética , Adolescente , Sequência de Bases , Criança , Técnicas de Cocultura , Consanguinidade , Família , Feminino , Ferroquelatase/metabolismo , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Líbano , Hepatopatias/complicações , Masculino , Mutação de Sentido Incorreto/genética , Linhagem , Fenótipo , Protoporfiria Eritropoética/mortalidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
3.
Blood Cells Mol Dis ; 54(1): 71-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25179834

RESUMO

The activity of the erythroid-specific isoenzyme of 5-aminolevulinic acid synthase (ALAS2), the first and rate-limiting enzyme in heme biosynthesis, is down-regulated during iron-deficiency. Ferrochelatase (FECH), the last enzyme of this pathway, inserts iron into protoporphyrin IX (PPIX) to form heme. Patients with erythropoietic protoporphyria (EPP), an inherited deficiency in FECH, often show signs of iron deficiency in addition to phototoxicity which is caused by PPIX accumulation. However, iron supplementation often leads to exacerbation of phototoxicity. We report three EPP patients who had reduced erythrocytic PPIX concentrations when they were iron-deficient and their microcytic and hypochromic anemia deteriorated. Additionally, we found a significant increase in the amount of ALAS2 mRNA and protein among EPP patients. To verify the connection between FECH deficiency and ALAS2 over-expression, we inhibited FECH in cultured cells and found a subsequent increase in ALAS2 mRNA. We conclude that the primary deficiency in ferrochelatase leads to a secondary increase in ALAS2 expression. The combined action of these two enzymes within the heme biosynthetic pathway contributes to the accumulation of PPIX. Furthermore, we hypothesize that EPP patients may benefit from a mild iron deficiency since it would limit PPIX production by restricting ALAS2 over-expression.


Assuntos
5-Aminolevulinato Sintetase/biossíntese , Eritrócitos/enzimologia , Regulação Enzimológica da Expressão Gênica , Ferro/metabolismo , Protoporfiria Eritropoética/enzimologia , Protoporfirinas/metabolismo , Adolescente , Adulto , Eritrócitos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Protoporfiria Eritropoética/patologia , RNA Mensageiro/biossíntese
4.
Eur J Dermatol ; 21(4): 479-83, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21659066

RESUMO

Erythropoietic protoporphyria (EPP) is a rare inherited disorder of heme biosynthesis mostly caused by a deficient activity of the enzyme ferrochelatase (FECH), and consequent accumulation of protoporphyrin (PP) in various tissues. Clinical manifestations include a childhood onset, cutaneous photosensitivity and, sometimes, hepatobiliary disease. We report a 16-year-old male with EPP characterized by acute episodes of painful photosensitivity since early infancy, permanent changes in the photoexposed skin, microcytic anemia, thrombocytopenia, and mild hepatic dysfunction. His 18-year-old sister presented less acute symptoms with no chronic changes. Lesional biopsy disclosed perivascular deposition of PAS positive hyaline material. Rimington-Cripps test was positive and PP erythrocyte levels were >9,000 µg/L (N<1,600), but normal in their parents and younger brother. Genetic studies in both patients and their mother revealed heterozygosity for a novel mutation (c.1052delA) in FECH gene of both children, and heterozygosity for the hypomorphic allele IVS3-48T>C in all of them. This confirms the "pseudodominant" inheritance pattern usually observed, explained by the combined presence of a disabling FECH mutation and a common intronic polymorphism affecting the counterpart allele (IVS3-48T>C). Phenotypic heterogeneity for this genotype explains the divergent clinical presentation. This is the first description of a Portuguese family with EPP characterized at the molecular level.


Assuntos
Ferroquelatase/genética , Mutação , Polimorfismo de Nucleotídeo Único/genética , Protoporfiria Eritropoética/genética , Adolescente , Biópsia , Feminino , Humanos , Masculino , Portugal , Protoporfiria Eritropoética/diagnóstico , Protoporfiria Eritropoética/enzimologia
5.
J Hepatol ; 55(1): 162-70, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21145811

RESUMO

BACKGROUND & AIMS: Erythropoietic protoporphyria (EPP) is an inherited disorder of heme biosynthesis caused by partial ferrochelatase deficiency, resulting in protoporphyrin IX (PPIX) accumulation in erythrocytes, responsible for skin photosensitivity. In some EPP patients, the development of cholestatic liver injury due to PPIX accumulation can lead to hepatic failure. In adult EPP mice, bone marrow transplantation (BMT) leads to skin photosensitivity correction but fails to reverse liver damages, probably because of the irreversible nature of liver fibrosis. Our aim was to determine the time course of liver disease progression in EPP mice and to evaluate the protective effect of BMT into neonates. METHODS: We studied the development of liver disease from birth in EPP mice, in relation with erythroid and hepatic PPIX accumulation. To prevent the development of liver disease, BMT was performed into newborn mice using a novel busulfan-mediated preconditioning assay. RESULTS: We showed that hepatic PPIX accumulates during the first 2 weeks and correlates with the onset of a progressive liver fibrosis in 12-day-old EPP mice. Transplantation of normal congenic hematopoietic stem cells into EPP neonates led to long-term donor hematopoiesis recovery. A full correction of erythroid PPIX accumulation and skin photosensitivity was obtained. Furthermore, five months after neonatal BMT, liver damage was almost completely prevented. CONCLUSIONS: We demonstrated for the first time that BMT could be successfully used to prevent liver disease in EPP mice and suggested that BMT would be an attractive therapeutic option to prevent severe liver dysfunction in EPP patients.


Assuntos
Transplante de Medula Óssea , Hepatopatias/prevenção & controle , Protoporfiria Eritropoética/complicações , Protoporfiria Eritropoética/terapia , Animais , Animais Recém-Nascidos , Bussulfano/administração & dosagem , Modelos Animais de Doenças , Progressão da Doença , Ferroquelatase/genética , Humanos , Fígado/metabolismo , Fígado/patologia , Hepatopatias/etiologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Falência Hepática/prevenção & controle , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Agonistas Mieloablativos/administração & dosagem , Protoporfiria Eritropoética/enzimologia , Protoporfiria Eritropoética/genética , Protoporfirinas/metabolismo , Condicionamento Pré-Transplante
7.
Blood ; 112(10): 4308-13, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18780836

RESUMO

Mouse models have proven invaluable for understanding erythropoiesis. Here, we describe an autosomal recessive, inherited anemia in the mouse mutant hem6. Hematologic and transplantation analyses reveal a mild, congenital, hypochromic, microcytic anemia intrinsic to the hematopoietic system that is associated with a decreased red blood cell zinc protoporphyrin to heme ratio, indicative of porphyrin insufficiency. Intercross matings show that hem6 can suppress the porphyric phenotype of mice with erythropoietic protoporphyria (EPP). Furthermore, iron uptake studies in hem6 reticulocytes demonstrate defective incorporation of iron into heme that can be partially corrected by the addition of porphyrin precursors. Gene expression and enzymatic assays indicate that erythroid 5-aminolevulinic acid synthase (Alas2) is decreased in hem6 animals, suggesting a mechanism that could account for the anemia. Overall, these data lead to the hypothesis that hem6 encodes a protein that directly or indirectly regulates the expression of Alas2.


Assuntos
5-Aminolevulinato Sintetase/biossíntese , Anemia Hipocrômica/enzimologia , Regulação Enzimológica da Expressão Gênica , Hematopoese , Protoporfiria Eritropoética/enzimologia , Reticulócitos/enzimologia , 5-Aminolevulinato Sintetase/genética , Anemia Hipocrômica/genética , Anemia Hipocrômica/patologia , Animais , Cruzamentos Genéticos , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Hematopoese/genética , Heme/biossíntese , Heme/genética , Ferro/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/patologia , Reticulócitos/patologia
8.
Blood ; 110(12): 4108-10, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17804693

RESUMO

Erythropoietic protoporphyria (EPP) results from deficiency of ferrochelatase (FECH). Accumulation of protoporphyrin IX causes life-long acute photosensitivity. Microcytic anemia occurs in 20% to 60% of patients. We investigated 178 patients with dominant EPP confirmed by molecular analysis. Erythropoiesis was impaired in all patients; all had a downward shift in hemoglobin (Hb), and the mean decreased in males by 12 g/L (1.2 g/dL). By World Health Organization criteria, 48% of women and 33% of men were anemic. Iron stores, assessed by serum ferritin (sFn), were decreased by two-thirds, but normal serum soluble transferrin receptor-1 and iron concentrations suggested that erythropoiesis was not limited by iron supply. FECH deficiency in EPP appears to lead to a steady state in which decreased erythropoiesis is matched by reduced iron absorption and supply. This response may in part be mediated by protoporphyrin, but we found no correlation between erythrocyte protoporphyrin and Hb, sFn, total iron-binding capacity, or transferrin saturation.


Assuntos
Antígenos CD/sangue , Eritropoese , Ferritinas/sangue , Hemoglobinas/análise , Ferro/sangue , Protoporfiria Eritropoética/sangue , Protoporfirinas/sangue , Receptores da Transferrina/sangue , Anemia/sangue , Anemia/enzimologia , Estudos Transversais , Eritrócitos/enzimologia , Eritrócitos/metabolismo , Feminino , Ferroquelatase , Humanos , Masculino , Transtornos de Fotossensibilidade/sangue , Transtornos de Fotossensibilidade/enzimologia , Protoporfiria Eritropoética/enzimologia , Fatores Sexuais
9.
Gene Ther ; 11(22): 1638-47, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15284838

RESUMO

Erythropoietic protoporphyria (EPP) is an inherited defect of the ferrochelatase (FECH) gene characterized by the accumulation of toxic protoporphyrin in the liver and bone marrow resulting in severe skin photosensitivity. We previously described successful gene therapy of an animal model of the disease with erythroid-specific lentiviral vectors in the absence of preselection of corrected cells. However, the high-level of gene transfer obtained in mice is not translatable to large animal models and humans if there is no selective advantage for genetically modified hematopoietic stem cells (HSCs) in vivo. We used bicistronic SIN-lentiviral vectors coexpressing EGFP or FECH and the G156A-mutated O6-methylguanine-DNA-methyltransferase (MGMT) gene, which allowed efficient in vivo selection of transduced HSCs after O6-benzylguanine and BCNU treatment. We demonstrate for the first time that the correction and in vivo expansion of deficient transduced HSC population can be obtained by this dual gene therapy, resulting in a progressive increase of normal RBCs in EPP mice and a complete correction of skin photosensitivity. Finally, we developed a novel bipromoter SIN-lentiviral vector with a constitutive expression of MGMT gene to allow the selection of HSCs and with an erythroid-specific expression of the FECH therapeutic gene.


Assuntos
Terapia Genética/métodos , Guanina/análogos & derivados , O(6)-Metilguanina-DNA Metiltransferase/genética , Protoporfiria Eritropoética/terapia , Transplante de Células-Tronco , Animais , Antineoplásicos/uso terapêutico , Carmustina/uso terapêutico , Feminino , Ferroquelatase/genética , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Guanina/uso terapêutico , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Protoporfiria Eritropoética/enzimologia , Células-Tronco/enzimologia , Transdução Genética/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA