Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Folia Biol (Praha) ; 61(6): 219-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26789143

RESUMO

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.


Assuntos
Alelos , Predisposição Genética para Doença , Mutação/genética , Porfiria Hepatoeritropoética/enzimologia , Porfiria Hepatoeritropoética/genética , Uroporfirinogênio Descarboxilase/genética , Adolescente , Sequência de Aminoácidos , Sequência de Bases , Criança , Cicatriz/complicações , Análise Mutacional de DNA , Egito , Família , Feminino , Humanos , Hipertricose/complicações , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Taxa de Mutação , Linhagem , Porfiria Hepatoeritropoética/complicações , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Uroporfirinogênio Descarboxilase/química
3.
Br J Dermatol ; 167(5): 1165-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22612618

RESUMO

BACKGROUND: Loss of function FLG alleles were first identified as causative of ichthyosis vulgaris (IV) and were subsequently found to be major predisposing factors for atopic dermatitis (AD) and atopic disorders. OBJECTIVES: To identify independent factors associated with the clinical IV phenotype in adult caucasian patients with AD and to assess the performance of a global clinical severity score of IV in predicting common FLG null mutations. PATIENTS AND METHODS: This was a prospective study conducted from January 2007 to June 2008. Adult patients attending the department of dermatology with a diagnosis of AD with or without IV were eligible to participate. For each patient, five clinical signs of IV were scored from 0 to 3 - diffuse xerosis, hyperlinearity of palms, scales on legs, scalp desquamation and keratosis pilaris - and a global IV clinical severity score was derived (0-15). Age of onset of AD, SCORAD (SCORing of Atopic Dermatitis), family and personal history for other signs of atopy, and total immunoglobulin E were recorded. Genotyping was performed for R501X and 2282del4. Univariate and multivariate analysis for factors associated with AD or AD + IV were conducted. RESULTS: In univariate analysis, family history of atopy, global clinical severity scoring and 2282del4 FLG mutation were positively correlated with the AD + IV phenotype. Using multivariate analysis, SCORAD for AD (OR 0·94, P = 0·01) and global clinical severity scoring for AD + IV (OR 2·62, P < 0·0001) were found to be independent factors. CONCLUSIONS: The 2282del4 FLG mutation was confirmed as a good marker of early-onset disease. Moreover, our global clinical severity score yielded a good negative predictive value of common caucasian null FLG mutations.


Assuntos
Dermatite Atópica/genética , Ictiose Vulgar/genética , Proteínas de Filamentos Intermediários/genética , Adulto , Idade de Início , Estudos Transversais , Dermatite Atópica/etiologia , Feminino , Proteínas Filagrinas , Predisposição Genética para Doença , Genótipo , Humanos , Ictiose Vulgar/complicações , Modelos Logísticos , Masculino , Mutação , Valor Preditivo dos Testes , Estudos Prospectivos , Índice de Gravidade de Doença , População Branca
4.
Br J Dermatol ; 167(4): 888-900, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22804244

RESUMO

BACKGROUND: Congenital erythropoietic porphyria (CEP) is an autosomal recessive photomutilating porphyria with onset usually in childhood, where haematological complications determine prognosis. Due to its extreme rarity and clinical heterogeneity, management decisions in CEP are often difficult. OBJECTIVES: To develop a management algorithm for patients with CEP based on data from carefully characterized historical cases. METHODS: A single investigator collated data related to treatments and their outcomes in 29 patients with CEP from the U.K., France, Germany and Switzerland. RESULTS: Six children were treated with bone marrow transplantation (BMT); five have remained symptomatically cured up to 11.5 years post-transplantation. Treatments such as oral charcoal, splenectomy and chronic hypertransfusion were either of no benefit or were associated with complications and negative impact on health-related quality of life. Lack of consistent genotype-phenotype correlation meant that this could not be used to predict disease prognosis. The main poor prognostic factors were early age of disease onset and severity of haematological manifestations. CONCLUSIONS: A management algorithm is proposed where every patient, irrespective of disease severity at presentation, should receive a comprehensive, multidisciplinary clinical assessment and should then be reviewed at intervals based on their predicted prognosis, and the rate of onset of complications. A BMT should be considered in those with progressive, symptomatic haemolytic anaemia and/or thrombocytopenia. Uroporphyrinogen III synthase genotypes associated with poor prognosis would additionally justify consideration for a BMT. Rigorous photoprotection of the skin and eyes from visible light is essential in all patients.


Assuntos
Porfiria Eritropoética/terapia , Índice de Gravidade de Doença , Adolescente , Adulto , Algoritmos , Transfusão de Sangue/métodos , Transplante de Medula Óssea/métodos , Carvão Vegetal/administração & dosagem , Criança , Pré-Escolar , Estudos de Coortes , Europa (Continente) , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Porfiria Eritropoética/genética , Roupa de Proteção , Esplenectomia/métodos , Adulto Jovem , beta Caroteno/administração & dosagem
5.
Br J Dermatol ; 167(4): 901-13, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22816431

RESUMO

BACKGROUND: Congenital erythropoietic porphyria (CEP) is an autosomal recessive cutaneous porphyria caused by decreased activity of uroporphyrinogen III synthase (UROS). Its predominant characteristics include bullous cutaneous photosensitivity to visible light from early infancy, progressive photomutilation and chronic haemolytic anaemia. Due to its rarity and genetic heterogeneity, clinical phenotypes are unclear and its impact on health-related quality of life (HRQoL) has not been previously assessed. OBJECTIVES: To define comprehensively CEP phenotypes and assess their impact on HRQoL, and to correlate these factors with laboratory parameters. METHODS: A single observer assessed patients with CEP from four European countries. RESULTS: Twenty-seven unrelated patients with CEP, aged between 7.6 and 65 years, participated in the study. The patients came from the U.K. (17), France (4), Switzerland (4) and Germany (2). Additional data were obtained for two deceased patients. Newly characterized features of CEP include acute-onset cutaneous and noncutaneous symptoms immediately following sunlight exposure, and pink erythematous facial papules. There was a lack of consistent genotype-phenotype correlation in CEP. The main poor prognostic factors in CEP are the early age of disease onset and haematological complications. CONCLUSIONS: CEP is a multisystem disease; cutaneous, ocular, oral and skeletal manifestations also contribute to disease severity and impact on HRQoL, in addition to the haematological complications. The rarity of the disease can lead to delayed diagnosis. The lack of consistent genotype-phenotype correlation in CEP suggests a contribution to phenotype from other factors, such as environment, patients' photoprotective behaviour and genes other than UROS. There is currently an unmet need for multidisciplinary management of patients with CEP.


Assuntos
Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , Adolescente , Adulto , Criança , Estudos de Coortes , Europa (Continente) , Feminino , Estudos de Associação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Porfiria Eritropoética/fisiopatologia , Qualidade de Vida , Adulto Jovem
6.
Dermatology ; 222(4): 336-41, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21701148

RESUMO

BACKGROUND: Atopic dermatitis (AD) is significantly associated with keratoconus (KC). An inherited component for KC has been suggested. Filaggrin (FLG) mutations are a strong genetic risk factor for AD. Since filaggrin is also expressed in the corneal epithelium, we hypothesized a common aetiology for ichthyosis vulgaris (IV), AD and KC. OBJECTIVES: We examined the prevalence of AD and IV in a KC population. We also studied the expression of filaggrin in normal and KC cornea and analysed 2 prevalent loss-of-function FLG alleles (R501X and 2282del4) in a KC population. Finally we examined whether the population with KC and FLG mutations had specific clinical characteristics. RESULTS: Of 89 KC patients, 38 had current or a history of AD and/or IV. Five patients were carriers of at least 1 FLG mutant allele and had a clinical diagnosis of AD and IV with a severer KC. CONCLUSION: The low frequency of FLG mutations is surprising since 42.7% of our KC population had AD associated or not with IV; the expected frequency would have been 12-15%, based on our previous studies. Further studies are required to look at other possible FLG mutations or other candidate genes.


Assuntos
Dermatite Atópica/genética , Proteínas de Filamentos Intermediários/genética , Ceratocone/genética , Mutação , Adolescente , Adulto , Criança , Dermatite Atópica/epidemiologia , Feminino , Proteínas Filagrinas , Predisposição Genética para Doença/epidemiologia , Humanos , Ceratocone/epidemiologia , Masculino , Pessoa de Meia-Idade , Prevalência , Estudos Prospectivos , Adulto Jovem
7.
Ann Dermatol Venereol ; 137(10): 635-9, 2010 Oct.
Artigo em Francês | MEDLINE | ID: mdl-20932444

RESUMO

BACKGROUND: Congenital erythropoietic porphyria (CEP) is a genodermatosis associated uroporphyrinogen III synthase deficit that results in porphyrin accumulation in various organs, particularly the skin. It is the most severe form of porphyria associated with haemolytic anaemia and cutaneous phototoxicity. We report a severe case of CEP treated by allogeneic bone marrow transplantation. CASE REPORT: A one-year-old child presented erythrodontia and scarring on exposed areas. The diagnosis of CEP was confirmed by the decline of uroporphyrinogen III synthase activity. Demonstration of p.Cys73Arg mutation confirmed the severity of the disease. Allogeneic bone marrow transplantation resulted in persistent resolution of clinical signs 25 months after grafting. DISCUSSION: Symptomatic treatment is ineffective in this serious disease associated with early mortality. 11 of the 13 patients treated by allogeneic hematopoietic stem cell graft, including our patient, continued to be asymptomatic an average of seven years after transplantation. CONCLUSION: This new case confirms the role of allogeneic hematopoietic stem cell grafting in the treatment of congenital erythropoietic porphyria.


Assuntos
Transplante de Medula Óssea/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Porfiria Eritropoética/terapia , Alelos , Diagnóstico Diferencial , Feminino , Triagem de Portadores Genéticos , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/etiologia , Humanos , Imunossupressores/uso terapêutico , Lactente , Porfiria Eritropoética/diagnóstico , Porfiria Eritropoética/genética , Descoloração de Dente/diagnóstico , Descoloração de Dente/genética , Descoloração de Dente/terapia , Uroporfirinogênio III Sintetase/genética
8.
Science ; 234(4777): 732-4, 1986 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-3775362

RESUMO

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.


Assuntos
Carboxiliases/genética , Porfirias/genética , Uroporfirinogênio Descarboxilase/genética , Sequência de Aminoácidos , Clonagem Molecular , DNA/genética , Humanos , Hepatopatias/genética , Mutação , Dermatopatias/genética , Relação Estrutura-Atividade , Uroporfirinogênio Descarboxilase/deficiência , Uroporfirinogênio Descarboxilase/metabolismo
9.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 53-60, 2009 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-19268002

RESUMO

High quality genotype/phenotype analysis is a difficult issue in rare genetic diseases such as congenital erythropoietic porphyria (CEP) or Günther's disease, a heme biosynthesis defect due to uroporphyrinogen III synthase deficiency. The historical background and the main phenotypic features of the disease are depicted together with an update of published mutants and genotype/phenotype correlations. General rules concerning the prediction of disease severity are drawn as a guide for patient management and therapeutic choices. The phenotypic heterogeneity of the disease is presented in relation with a likely influence of modifying factors, either genetic or acquired.


Assuntos
Mutação/fisiologia , Fenótipo , Porfiria Eritropoética/genética , Genótipo , Humanos , Mutação/genética , Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/genética , Uroporfirinogênio III Sintetase/fisiologia
10.
J Clin Invest ; 72(3): 1139-49, 1983 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6886003

RESUMO

Three siblings with intense jaundice and hemolytic anemia at birth were found to excrete a high level of coproporphyrin in their urine and feces; the pattern of fecal porphyrin excretion was atypical for hereditary coproporphyria because the major porphyrin was harderoporphyrin (greater than 60%; normal value is less than 20%). The lymphocyte coproporphyrinogen III oxidase activity of each patient was 10% of control values, which suggests a homozygous state. Both parents showed only mild abnormalities in porphyrin excretion and lymphocyte coproporphyrinogen III oxidase activity decreased to 50% of normal values, as is expected in heterozygous cases of hereditary coproporphyria. Kinetic parameters of coproporphyrinogen III oxidase from these patients were clearly modified, with a Michaelis constant 15-20-fold higher than normal values when using coproporphyrinogen or harderoporphyrinogen as substrates. Maximal velocity was half the normal value, and we also observed a marked sensitivity to thermal denaturation. The possibility that a mutation affecting the enzyme on the active center which is specifically involved in the second decarboxylation (from harderoporphyrinogen to protoporphyrinogen) was eliminated by experiments on rat liver that showed that coproporphyrinogen and harderoporphyrinogen were metabolized at the same active center. The pattern of porphyrin excretion and the coproporphyrinogen oxidase from the three patients exhibited abnormalities that were different from the abnormalities found in another recently described homozygous case of hereditary coproporphyria. We suggest naming this variant of coproporphyrinogen oxidase defect "harderoporphyria."


Assuntos
Hepatopatias/genética , Mutação , Porfirias/genética , Porfirinogênios/metabolismo , Animais , Coproporfirinogênio Oxidase/sangue , Coproporfirinogênio Oxidase/genética , Coproporfirinogênio Oxidase/metabolismo , Descarboxilação , Fezes/análise , Feminino , Humanos , Recém-Nascido , Cinética , Hepatopatias/diagnóstico , Hepatopatias/metabolismo , Linfócitos/enzimologia , Masculino , Porfirias/diagnóstico , Porfirias/metabolismo , Porfirinogênios/análise , Porfirinas/biossíntese , Porfirinas/sangue , Porfirinas/urina , Ratos
11.
J Clin Invest ; 77(2): 431-5, 1986 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3753711

RESUMO

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.


Assuntos
Carboxiliases/deficiência , Eritropoese , Hepatopatias , Porfirias/genética , Uroporfirinogênio Descarboxilase/deficiência , Linhagem Celular , DNA/genética , Meia-Vida , Humanos , Linfócitos/metabolismo , Hibridização de Ácido Nucleico , Porfirias/enzimologia , Biossíntese de Proteínas , RNA Mensageiro/genética , Uroporfirinogênio Descarboxilase/genética
12.
Respir Med Case Rep ; 20: 64-67, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28053854

RESUMO

Alpha-1-antitrypsin deficiency (A1ATD) is a genetic condition caused by SERPINA1 mutations, which results into decreased protease inhibitor activity in the serum and predisposes to emphysema and/or to liver disease due to accumulation of the abnormal protein in the hepatic cells. In most cases the clinical manifestations of A1ATD are associated with PIZZ (p.Glu366Lys; p.Glu366Lys (p.Glu342Lys; p.Glu342Lys)) or PISZ (p.Glu288Val; p.Glu366Lys (p.Glu264Val; p.Glu342Lys)) genotype, less frequently, deficient or null alleles may be present in compound heterozygous or homozygous A1AT deficient patients. We report the identification of a novel alpha1-antitrypsin variant in a 64-year old woman presenting with dyspnea on exertion. Imaging revealed bilateral bronchiectasis associated with moderate panacinar emphysema. The pulmonary function tests (PFTs) were subnormal but hypoxemia was noticed and A1AT quantitative analysis revealed a severe deficiency. DNA sequencing showed compound heterozygosity for the PIZ variant and a novel missense variant p.Phe232Leu (p.Phe208Leu). No specific treatment was proposed since PFTs were within the normal range at this stage of the disease. Close follow-up of pulmonary and hepatic parameters was recommended.

13.
Leukemia ; 31(1): 65-74, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27220663

RESUMO

Although tyrosine kinase inhibitors (TKIs) efficiently cure chronic myeloid leukemia (CML), they can fail to eradicate CML stem cells (CML-SCs). The mechanisms responsible for CML-SC survival need to be understood for designing therapies. Several previous studies suggest that TKIs could modulate CML-SC quiescence. Unfortunately, CML-SCs are insufficiently available. Induced pluripotent stem cells (iPSCs) offer a promising alternative. In this work, we used iPSCs derived from CML patients (Ph+). Ph+ iPSC clones expressed lower levels of stemness markers than normal iPSCs. BCR-ABL1 was found to be involved in stemness regulation and ERK1/2 to have a key role in the signaling pathway. TKIs unexpectedly promoted stemness marker expression in Ph+ iPSC clones. Imatinib also retained quiescence and induced stemness gene expression in CML-SCs. Our results suggest that TKIs might have a role in residual disease and confirm the need for a targeted therapy different from TKIs that could overcome the stemness-promoting effect caused by TKIs. Interestingly, a similar pro-stemness effect was observed in normal iPSCs and hematopoietic SCs. These findings could help to explain CML resistance mechanisms and the teratogenic side-effects of TKIs in embryonic cells.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas de Fusão bcr-abl/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Neoplásicas/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Células Tumorais Cultivadas
14.
Biochim Biophys Acta ; 611(1): 174-86, 1980 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-7350915

RESUMO

Several kinetic properties of uroporphyrinogen decarboxylase (uroporphyrinogen-III carboxy-lyase, EC 4.1.1.37) from human hemoglobin-free hemolysates were studied, using substrates of both isomeric series I and III (uroporphyrinogen, hepta and pentacarboxyl porphyrinogens). Enzyme affinity for series II isomers was always found to be higher than for corresponding series I isomers. Mixed substrate experiments using porphyrinogen (both labelled with 14C and unlabelled) showed: (a) a reciprocal inhibition of decarboxylation of series III porphyrinogens by series I porphyrinogens with the same number of carboxylic groups; (b) no inhibition of hepta- and pentacarboxylic series III porphyrinogens decarboxylation by uroporphyrinogen III. It is demonstrated that porphyrinogens of both isomeric series with the same number of carboxylic groups are decarboxylated at the same active center; in contrast, the sequential decarboxylation of uroporphyrinogen III to coproporphyrinogen III occurs at four different active centers. Relationship between the kinetic properties of uroporphyrinogen decarboxylase and biological data of porphyria cutanea are discussed.


Assuntos
Carboxiliases/sangue , Eritrócitos/enzimologia , Uroporfirinogênio Descarboxilase/sangue , Humanos , Cinética , Porfirias/enzimologia , Relação Estrutura-Atividade , Especificidade por Substrato , Uroporfirinogênios/farmacologia
15.
Biochim Biophys Acta ; 882(3): 384-8, 1986 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-3460638

RESUMO

Porphobilinogen deaminase is induced during the dimethyl sulfoxide-mediated differentiation of Friend erythroleukemia cells. We have previously shown that when succinylacetone, a potent inhibitor of porphobilinogen formation, is present during the differentiation process, the induction of the enzyme is apparently suppressed. Here, we provide evidence that, in this condition, porphobilinogen deaminase is synthesized normally but does not accumulate as a consequence of an accelerated turnover. The normal half-life of the protein is 24 h but decreases to 10 h when the formation of its substrate is impaired by succinylacetone. We propose that when the enzyme is covalently bound to its substrate, a normal step in this enzymatic reaction, it is protected from proteolytic degradation, and we show that this new finding is relevant to the human disorder acute intermittent porphyria.


Assuntos
Amônia-Liases/metabolismo , Hidroximetilbilano Sintase/metabolismo , Leucemia Eritroblástica Aguda/enzimologia , Diferenciação Celular/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Fluorometria , Vírus da Leucemia Murina de Friend , Heptanoatos/farmacologia , Leucemia Eritroblástica Aguda/ultraestrutura , Metionina/metabolismo , Peso Molecular , Fatores de Tempo
16.
Biochim Biophys Acta ; 629(3): 577-86, 1980 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-7417513

RESUMO

Partial deficiencies in enzymes activity of the heme biosynthesis pathway have been demonstrated in cultured skin fibroblasts and other tissues from patients suffering from congenital erythropoietic porphyria and hereditary coproporphyria. Using a new fluorimetric method, we have assessed quantitatively porphyrin biosynthesis from added delta-aminolevulinic acid in cultured fibroblasts of two congenital erythropoietic porphyria patients and one homozygous case of hereditary corproporphyria. The results were compared with those of the patients' parents and those of normal controls. All the porphyrins synthesized remained within the cells of normal subjects and of patients with congenital erythropoietic porphyria; these porphyrins were mostly (95%) protoporphyrin. The fibroblasts of the patient with homozygous hereditary coproporphyria synthesized the same amount of porphyrins, but only 25% were found within the cells, whereas 75% were found in the medium. The porphyrins found within the cells were coproporphyrin (25%) and protoporphyrin (75%); in the medium, only coproporphyrin was identified.


Assuntos
Fibroblastos/metabolismo , Porfirias/metabolismo , Porfirinas/biossíntese , Células Cultivadas , Criança , Coproporfirinas/biossíntese , Homozigoto , Humanos , Porfirias/congênito , Porfirias/genética , Protoporfirinas/biossíntese , Pele/metabolismo , Uroporfirinas/biossíntese
17.
Biochim Biophys Acta ; 1090(3): 311-6, 1991 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-1954253

RESUMO

Changes in the level of transcripts encoding enzymes of the heme biosynthetic pathway as well as those encoding ubiquitous proteins were examined in murine Friend virus-transformed erythroleukemia cells during erythroid cell differentiation induced by chemicals including dimethyl sulfoxide (DMSO). Early changes following DMSO treatment were marked decreases in mRNAs for three ubiquitous proteins, i.e., a 70 kDa heat shock protein (less than 6 h), heme oxygenase and nonspecific delta-aminolevulinate synthase (ALAS) (less than 12 h). These changes were followed by sequential increases in mRNAs for enzymes in the heme biosynthetic pathway. Namely, mRNAs for the erythroid-specific ALAS, delta-aminolevulinate dehydratase, porphobilinogen deaminase and uroporphyrinogen decarboxylase started to increase at 12, 18, 18-24 and 24 h, respectively. Nuclear runoff studies revealed that these changes are largely transcriptional. Treatments with other inducers of erythroid differentiation, e.g., hexamethylene bisacetamide, n-butyric acid and N'-methylnicotinamide, also showed similar effects on mRNAs as those following DMSO. These findings suggest that both suppression of ubiquitous genes and activation of heme pathway enzyme genes are associated with erythroid differentiation, and the former occurs preceding changes in the latter.


Assuntos
Eritrócitos/citologia , Vírus da Leucemia Murina de Friend/fisiologia , Regulação Enzimológica da Expressão Gênica , Heme/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Animais , Northern Blotting , Diferenciação Celular , Transformação Celular Viral , Cinética , Camundongos , RNA Mensageiro/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
18.
J Mol Med (Berl) ; 81(5): 310-20, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12721665

RESUMO

Congenital erythropoietic porphyria (CEP) is an inherited disease due to a deficiency in the uroporphyrinogen III synthase, the fourth enzyme of the heme biosynthesis pathway. It is characterized by accumulation of uroporphyrin I in the bone marrow, peripheral blood and other organs. The prognosis of CEP is poor, with death often occurring early in adult life. For severe transfusion-dependent cases, when allogeneic cell transplantation cannot be performed, the autografting of genetically modified primitive/stem cells may be the only alternative. In vitro gene transfer experiments have documented the feasibility of gene therapy via hematopoietic cells to treat this disease. In the present study lentiviral transduction of porphyric cell lines and primary CD34(+) cells with the therapeutic human uroporphyrinogen III synthase (UROS) cDNA resulted in both enzymatic and metabolic correction, as demonstrated by the increase in UROS activity and the suppression of porphyrin accumulation in transduced cells. Very high gene transfer efficiency (up to 90%) was achieved in both cell lines and CD34(+) cells without any selection. Expression of the transgene remained stable over long-term liquid culture. Furthermore, gene expression was maintained during in vitro erythroid differentiation of CD34(+) cells. Therefore the use of lentiviral vectors is promising for the future treatment of CEP patients by gene therapy.


Assuntos
Terapia Genética , Lentivirus/genética , Porfiria Eritropoética/terapia , Uroporfirinogênio III Sintetase/genética , Adulto , Técnicas de Cultura de Células , Diferenciação Celular , Eritroblastos/metabolismo , Fluorescência , Expressão Gênica , Vetores Genéticos , Humanos , Fenótipo , Porfiria Eritropoética/genética , Transdução Genética , Replicação Viral
19.
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
20.
Eur J Hum Genet ; 4(5): 274-82, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8946173

RESUMO

Congenital erythropoietic porphyria (CEP) or Günther's disease is an inborn error of heme biosynthesis, transmitted as an autosomal recessive trait and characterized by a profound deficiency of uroporphyrinogen III synthase activity (UROIIIS). The molecular defects observed in CEP are mainly heterogeneous, except for one missense mutation, C73R (Cys to Arg substitution at codon 73) which represents nearly 40% of the disease alleles. A convenient strategy was designed to establish a rapid diagnosis at the genetic level in samples from patients with CEP. As a first step, the most frequent mutation is searched for by restriction analysis from genomic. DNA amplified by PCR. Next, the nine coding exons and intron-exon boundaries are sequenced from genomic DNA. As an alternative, the mutation can be determined by sequencing the UROIIIS cDNA of the patient, using the RT-PCR technique on RNAs when a lymphoblastoid cell line can be established. Finally, for each new mutation in UROIIIS coding sequence, the corresponding mutant protein is expressed in Escherichia coli, in order to demonstrate the pathological significance of the mutation. This work describes the analysis of UROIIIS gene mutations in 10 new families with CEP and summarizes the data from 20 unrelated families studied in our laboratory. Three new missense mutations of UROIIIS coding sequence (H173Y, Q187P and P248Q) have been observed together with 8 known mutations. The significance of three intronic base changes (476 -31 T-->C; 562 -4 A-->T; 562 -23 A-->G) is discussed. In 6 alleles out of 40 (15%), the mutation remains undetermined.


Assuntos
Mutação , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , Adolescente , Alelos , Linhagem Celular Transformada , Criança , Pré-Escolar , DNA Complementar , Éxons , Feminino , Heterozigoto , Humanos , Recém-Nascido , Masculino , Porfiria Eritropoética/enzimologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa