Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Gene Ther ; 22(11): 856-65, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26125605

RESUMO

Helper-dependent adenoviral (HDA) vectors constitute excellent gene therapy tools for metabolic liver diseases. We have previously shown that an HDA vector encoding human porphobilinogen deaminase (PBGD) corrects acute intermittent porphyria mice. Now, six non-human primates were injected in the left hepatic lobe with the PBGD-encoding HDA vector to study levels and persistence of transgene expression. Intrahepatic administration of 5 × 10(12) viral particles kg(-1) (10(10) infective units kg(-1)) of HDA only resulted in transient (≈14 weeks) transgene expression in one out of three individuals. In contrast, a more prolonged 90-day immunosuppressive regimen (tacrolimus, mycophenolate, rituximab and steroids) extended meaningful transgene expression for over 76 weeks in two out of two cases. Transgene expression under immunosuppression (IS) reached maximum levels 6 weeks after HDA administration and gradually declined reaching a stable plateau within the therapeutic range for acute porphyria. The non-injected liver lobes also expressed the transgene because of vector circulation. IS controlled anticapsid T-cell responses and decreased the induction of neutralizing antibodies. Re-administration of HDA-hPBGD at week +78 achieved therapeutically meaningful transgene expression only in those animals receiving IS again at the time of this second vector exposure. Overall, immunity against adenoviral capsids poses serious hurdles for long-term HDA-mediated liver transduction, which can be partially circumvented by pharmacological IS.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vírus Auxiliares/genética , Imunossupressores/farmacologia , Fígado/fisiologia , Transgenes , Animais , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Hidroximetilbilano Sintase/biossíntese , Hidroximetilbilano Sintase/genética , Fígado/metabolismo , Macaca fascicularis , Masculino
2.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 55-63, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19656452

RESUMO

Acute intermittent porphyria (AIP), the most common acute hepatic porphyria, is an autosomal dominant disorder with low penetrance that results from a partial deficiency of hydroxymethylbilane synthase (HMBS), the third enzyme in the heme biosynthetic pathway. The disease is clinically characterized by acute neurovisceral attacks that are precipitated by several factors including certain drugs, steroid hormones, alcohol and fasting. Early diagnosis and counselling are essential to prevent attacks, being mutation analysis the most reliable method to identify asymptomatic carriers in AIP families. In this study we have investigated the molecular defect in 15 unrelated Spanish AIP patients. Mutation analysis of the HMBS gene revealed a total of fourteen mutations including six novel ones, two of them were on the same allele in one patient. The novel mutations were three missense (R26L, R173G and D178H), two frameshift (c.749_765dup and c.874insC) and one intronic deletion (IVS12+3_+11delAGGGCCTGT). RT-PCR and sequencing demonstrated that the intronic mutation caused abnormal splicing and exon 12 skipping. Prokaryotic expression of the novel missense mutations showed that only D178H had significant residual activity. These findings will facilitate the accurate identification of presymptomatic AIP carriers in these families and they further emphasize the molecular heterogeneity of AIP in Spain.


Assuntos
Hidroximetilbilano Sintase/genética , Porfiria Aguda Intermitente/genética , População Branca/genética , Adulto , Idoso , Alelos , Feminino , Mutação da Fase de Leitura , Deleção de Genes , Humanos , Hidroximetilbilano Sintase/química , Hidroximetilbilano Sintase/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Polimorfismo Genético , Porfiria Aguda Intermitente/enzimologia , Estabilidade Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Análise de Sequência de DNA , Espanha , Temperatura
4.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 29-37, 2009 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-19267999

RESUMO

Activation of the epidermal growth factor receptor (EGFR) plays an important role in liver regeneration and resistance to acute injury. However its chronic activation participates in the progression of liver disease, including fibrogenesis and malignant transformation. Hepatobiliary disease represents a constant feature in the clinically relevant Fechm1pas/Fechm1pas genetic model of erythropoietic protoporphyria (EPP). Similarly, chronic administration of griseofulvin to mice induces pathological changes similar to those found in patients with EPP-associated liver injury. We investigated the hepatic expression of the EGFR and its seven most relevant ligands in Fechm1pas/Fechm1pas mice bred in three different backgrounds, and in griseofulvin-induced protoporphyria. We observed that the expression of amphiregulin, betacellulin and epiregulin was significantly increased in young EPP mice when compared to aged-matched controls in all genetic backgrounds. The expression of these ligands was also tested in older (11 months) BALB/cJ EPP mice, and it was found to remain induced, while that of the EGFR was downregulated. Griseofulvin feeding also increased the expression of amphiregulin, betacellulin and epiregulin. Interestingly, protoporphyrin accumulation in cultured hepatic AML-12 cells readily elicited the expression of these three EGFR ligands. Our findings suggest that protoporphyrin could directly induce the hepatic expression of EGFR ligands, and that their chronic upregulation might participate in the pathogenesis of EPP-associated liver disease.


Assuntos
Receptores ErbB/agonistas , Receptores ErbB/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Protoporfiria Eritropoética/metabolismo , Anfirregulina , Animais , Betacelulina , Linhagem Celular , Família de Proteínas EGF , Fator de Crescimento Epidérmico/genética , Epigen , Epirregulina , Glicoproteínas/genética , Griseofulvina/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/genética , Hepatopatias/genética , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Protoporfiria Eritropoética/genética , Protoporfirinas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador alfa/genética
5.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 38-44, 2009 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-19268000

RESUMO

Erythropoietic Protoporphyria (EPP) is an inherited deficiency of ferrochelatase, the last enzyme of the heme pathway. Under general anaesthesia, some patients develop neurological dysfunction suggesting upregulation in heme biosynthesis similar to that described for acute porphyrias after xenobiotic administration. Our aim has been to evaluate whether Isoflurane induces alterations in the heme pathway in a mouse model for EPP. Administration of Isoflurane (a single dose of 2 ml/kg, i.p) to wild-type (+/+), heterozygous (+/Fechm1Pas) and homozygous (Fechm1Pas/Fechm1Pas) mice, was evaluated by measuring the activity of delta-aminolevulinic acid synthetase (ALA-S) and Porphobilinogen-deaminase (PBG-D) in different tissues, as well as Heme oxygenase (HO), cytochrome P-450, CYP2E1 and glutathione levels in liver. Porphyrin precursors were measured in 24 h-urine samples. Fechm1Pas/Fechm1Pas mice receiving anaesthesia show enhanced ALA-S and CYP2E1 activities in the liver and increased urinary excretion of porphyrin precursors. No alterations were found in either PBG-D or HO activities. Diminished glutathione levels suggest that anaesthesia may produce oxidative stress in these animals. In conclusion, Isoflurane induces ALA-S activity and increased excretion of porphyrin precursors in EPP mice. These findings appear to confirm our previous hypothesis and indicate that Isoflurane may be an unsafe anaesthetic not only for patients with acute porphyrias but also for individuals with non acute porphyrias.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Isoflurano/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Protoporfiria Eritropoética/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Glutationa/metabolismo , Heme Oxigenase (Desciclizante) , Hidroximetilbilano Sintase/metabolismo , Camundongos , Camundongos Mutantes , Estresse Oxidativo/efeitos dos fármacos
6.
Gene Ther ; 16(1): 136-41, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18668147

RESUMO

Non-invasive in vivo imaging of transgene expression is currently providing very important means to optimize gene therapy regimes. Results in non-human primates are considered the most predictive models for the outcome in patients. In this study, we have documented that tumour and primary cell lines from human and non-human primates are comparably gene-transduced in vitro by serotype 5 adenovirus expressing HSV1-thymidine kinase. Transgene expression can be quantified in human and monkey cultured cells by positron emission tomography (PET) imaging when transduced cells are incubated with a fluoride-18 labelled penciclovir analogue. In our hands, PET images of cell cultures estimate the number of transduced cells rather than intensity of transgene expression once a threshold of TK per cell is reached. Interestingly, in vivo systemic administration of a clinical grade recombinant adenovirus expressing TK into macaques gives rise to an intense retention of the radiotracer in the liver parenchyma, providing an experimental system to visualize transgene expression that ought to be similar in human and macaques. Such imaging methodology might contribute to improve strategies based on adenoviral vectors.


Assuntos
Terapia Genética/métodos , Herpesvirus Humano 1/enzimologia , Fígado/diagnóstico por imagem , Fígado/enzimologia , Tomografia por Emissão de Pósitrons , Timidina Quinase/genética , Aciclovir/análogos & derivados , Aciclovir/farmacologia , Adenoviridae/genética , Animais , Contagem de Células , Linhagem Celular Transformada , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Guanina , Humanos , Injeções Intravenosas , Macaca , Modelos Animais , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacologia , Transdução Genética/métodos , Transgenes
7.
Br J Dermatol ; 157(3): 501-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17627795

RESUMO

BACKGROUND: Porphyria cutanea tarda (PCT) results from decreased hepatic uroporphyrinogen decarboxylase (UROD) activity. In the majority of patients, the disease is sporadic (S-PCT or type I) and the enzyme deficiency is limited to the liver. Familial PCT (F-PCT or type II) is observed in 20-30% of patients in whom mutations on one allele of the UROD gene reduce UROD activity by approximately 50% in all tissues. Another variant of PCT (type III) is characterized by family history of the disease although it is biochemically indistinguishable from S-PCT. OBJECTIVES: To investigate the molecular basis of PCT in Spain and to compare enzymatic and molecular analysis for the identification of patients with F-PCT. METHODS: Erythrocyte UROD activity measurement and mutation analysis of the UROD gene were carried out in a cohort of 61 unrelated Spanish patients with PCT and 50 control individuals. Furthermore, each novel missense mutation identified was characterized by prokaryotic expression studies. RESULTS: Of these 61 patients, 40 (66%) were classified as having S-PCT, 16 (26%) as having F-PCT and five (8%) as having type III PCT. Discordant results between enzymatic and molecular analysis were observed in two patients with F-PCT. In total, 14 distinct mutations were found, including 10 novel mutations: five missense, one nonsense, three deletions and an insertion. Prokaryotic expression of the novel missense mutations demonstrated that each results in decreased enzyme activity or stability. CONCLUSIONS: These results confirm the high degree of molecular heterogeneity of F-PCT in Spain and emphasize the usefulness of molecular genetic analysis to distinguish between F-PCT and S-PCT.


Assuntos
Predisposição Genética para Doença , Mutação/genética , Porfiria Cutânea Tardia/genética , Uroporfirinogênio Descarboxilase/genética , Adulto , Idoso , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Porfiria Cutânea Tardia/classificação , Porfiria Cutânea Tardia/enzimologia , Fatores de Risco , Uroporfirinogênio Descarboxilase/deficiência
8.
Arch Environ Contam Toxicol ; 47(2): 276-80, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15386154

RESUMO

Childhood environmental lead exposure in the city of Antofagasta, Chile, was generated by the accumulation of recently removed lead stores derived from mining activities for a long period of time. Susceptibility to harmful lead effects may be associated with polymorphisms of delta-aminolevulinic acid dehydratase (ALAD) because of the differential binding of lead to the codified proteins. We assessed the associations and possible interactions among the following variables: blood lead levels, ALAD genotypes, and distance to the source of lead contamination in Chilean children exposed to lead contamination in Antofagasta, Chile. Ninety-three children were recruited from schools located near a lead- contaminated area. Lead blood levels were measured by atomic absorption spectrophotometry. ALAD genotypes were determined by polymerase chain reaction and restriction fragment-length polymorphism analysis. The frequency of the ALAD-2 allele was estimated at 0.054. Children with the ALAD-2 genotype had higher blood lead levels than noncarriers (p = 0.06). As expected, blood lead levels were inversely correlated with the distance from lead stores. Interestingly, ALAD-2 carriers were more frequent within the area defined by a distance of 200 m from lead deposits (27%) than in areas >200 m (5%) away. Children living within a maximum distance of 200 m from the lead stores showed higher blood lead levels in ALAD-2 carriers (geometric mean = 16.4 microg/dl, range 6 to 27) than in noncarriers (geometric mean = 12.1 microg/dl, range 0 to 26) without achieving statistical significance (p = 0.13). A trend for higher blood lead levels in ALAD-2 carriers compared with ALAD-1 homozygous children has been observed. Because ALAD-2 frequency was higher in subjects living within 200 m from the lead deposits, we hypothesized that a long-term selective pressure against the presence of the ALAD-1 allele is the cause of the overrepresentation of the ALAD-2 allele in children living in proximity to the recently removed lead stores.


Assuntos
Proteção da Criança , Exposição Ambiental , Poluentes Ambientais/sangue , Intoxicação por Chumbo/etiologia , Chumbo/sangue , Sintase do Porfobilinogênio/genética , Criança , Pré-Escolar , Chile , Feminino , Genótipo , Humanos , Masculino , Mineração , Instituições Acadêmicas
9.
Cell Mol Biol (Noisy-le-grand) ; 48(8): 845-52, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12699242

RESUMO

Erythrocyte uroporphyrinogen decarboxylase (UROD) activity was measured to classify 118 Spanish patients with porphyria cutanea tarda (PCT) into three subtypes: sporadic-, familial- and type III-PCT. Seventy-four patients (63%) had eythrocyte UROD activity within the normal range (74% to 126% of the mean activity of 43 healthy controls) and were classified as sporadic-PCT (47%) or as type III-PCT (16%) whenever a family history of PCT was documented. Forty-four patients (37%) had decreased UROD activity and were classified as familial-PCT. The frequency of both familial-PCT and type III-PCT was higher than reported in other countries. The clinical expression of PCT was associated with the coexistence of two or more risk factors in 80% of the sporadic-PCT patients and in 89% of the familial-PCT patients. Hepatitis C virus and alcohol abuse were risk factors frequently found in these patients, being unrelated to age of onset of skin lesions. A heavy alcohol intake was the main risk factor for type III-PCT. Estrogens appeared as a precipitating factor for women with familial-PCT. The H63D mutation in the hemochromatosis type 1 gene was more frequently found than the C282Y mutation. Both mutations appeared to play a role as precipitating factors in sporadic-PCT when associated with hepatitis C virus infection and alcohol abuse.


Assuntos
Porfiria Cutânea Tardia/diagnóstico , Porfiria Cutânea Tardia/genética , Adulto , Idade de Início , Consumo de Bebidas Alcoólicas , Alelos , Estrogênios/metabolismo , Saúde da Família , Feminino , Predisposição Genética para Doença , Hemocromatose/genética , Proteína da Hemocromatose , Hepatite C/complicações , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Mutação , Porfiria Cutânea Tardia/etiologia , Porfiria Cutânea Tardia/virologia , Fatores de Risco , Espanha , Uroporfirinogênio Descarboxilase/sangue
10.
Mol Ther ; 4(4): 331-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11592836

RESUMO

Successful treatment of blood disorders by gene therapy has several complications, one of which is the frequent lack of selective advantage of genetically corrected cells. Erythropoietic protoporphyria (EPP), caused by a ferrochelatase deficiency, is a good model of hematological genetic disorders with a lack of spontaneous in vivo selection. This disease is characterized by accumulation of protoporphyrin in red blood cells, bone marrow, and other organs, resulting in severe skin photosensitivity. Here we develop a self-inactivating lentiviral vector containing human ferrochelatase cDNA driven by the human ankyrin-1/beta-globin HS-40 chimeric erythroid promoter/enhancer. We collected bone marrow cells from EPP male donor mice for lentiviral transduction and injected them into lethally irradiated female EPP recipient mice. We observed a high transduction efficiency of hematopoietic stem cells resulting in effective gene therapy of primary and secondary recipient EPP mice without any selectable system. Skin photosensitivity was corrected for all secondary engrafted mice and was associated with specific ferrochelatase expression in the erythroid lineage. An erythroid-specific expression was sufficient to reverse most of the clinical and biological manifestations of the disease. This improvement in the efficiency of gene transfer with lentiviruses may contribute to the development of successful clinical protocols for erythropoietic diseases.


Assuntos
Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Terapia Genética/métodos , Lentivirus/genética , Porfiria Hepatoeritropoética/genética , Porfiria Hepatoeritropoética/terapia , Animais , Southern Blotting , Transplante de Medula Óssea , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Feminino , Ferroquelatase/genética , Ferroquelatase/metabolismo , Ferroquelatase/uso terapêutico , Expressão Gênica/genética , Vetores Genéticos/genética , Humanos , Lentivirus/fisiologia , Masculino , Camundongos , Especificidade de Órgãos , Porfiria Hepatoeritropoética/enzimologia , Porfiria Hepatoeritropoética/patologia , Porfirinas/metabolismo , Regiões Promotoras Genéticas/genética , Protoporfiria Eritropoética , Pele/patologia , Transdução Genética
11.
Gene Ther ; 8(8): 618-26, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11320408

RESUMO

Erythropoietic protoporphyria is characterized clinically by skin photosensitivity and biochemically by a ferrochelatase deficiency resulting in an excessive accumulation of photoreactive protoporphyrin in erythrocytes, plasma and other organs. The availability of the Fech(m1Pas)/Fech(m1Pas) murine model allowed us to test a gene therapy protocol to correct the porphyric phenotype. Gene therapy was performed by ex vivo transfer of human ferrochelatase cDNA with a retroviral vector to deficient hematopoietic cells, followed by re-injection of the transduced cells with or without selection in the porphyric mouse. Genetically corrected cells were separated by FACS from deficient ones by the absence of fluorescence when illuminated under ultraviolet light. Five months after transplantation, the number of fluorescent erythrocytes decreased from 61% (EPP mice) to 19% for EPP mice engrafted with low fluorescent selected BM cells. Absence of skin photosensitivity was observed in mice with less than 20% of fluorescent RBC. A partial phenotypic correction was found for animals with 20 to 40% of fluorescent RBC. In conclusion, a partial correction of bone marrow cells is sufficient to reverse the porphyric phenotype and restore normal hematopoiesis. This selection system represents a rapid and efficient procedure and an excellent alternative to the use of potentially harmful gene markers in retroviral vectors.


Assuntos
Separação Celular/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Porfiria Hepatoeritropoética/terapia , Animais , Linhagem Celular , DNA Complementar/genética , Modelos Animais de Doenças , Feminino , Ferroquelatase/genética , Citometria de Fluxo , Vetores Genéticos , Hematopoese , Interleucina-3/fisiologia , Hepatopatias/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Transtornos de Fotossensibilidade/terapia , Porfiria Hepatoeritropoética/fisiopatologia , Retroviridae/genética
12.
Mol Ther ; 3(3): 411-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11273784

RESUMO

Congenital erythropoietic porphyria (CEP) is an inherited disease due to a deficiency in the uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme pathway. It is characterized by accumulation of uroporphyrin I in the bone marrow, peripheral blood, and other organs. The onset of most cases occurs in infancy and the main symptoms are cutaneous photosensitivity and hemolysis. For severe transfusion-dependent cases, when allogeneic cell transplantation cannot be performed, autografting of genetically modified primitive/stem cells is the only alternative. In the present study, efficient mobilization of peripheral blood primitive CD34(+) cells was performed on a young adult CEP patient. Retroviral transduction of this cell population with the therapeutic human UROS (hUS) gene resulted in both enzymatic and metabolic correction of CD34(+)-derived cells, as demonstrated by the increase in UROS activity and by a 53% drop in porphyrin accumulation. A 10-24% gene transfer efficiency was achieved in the most primitive cells, as demonstrated by the expression of enhanced green fluorescent protein (EGFP) in long-term culture-initiating cells (LTC-IC). Furthermore, gene expression remained stable during in vitro erythroid differentiation. Therefore, these results are promising for the future treatment of CEP patients by gene therapy.


Assuntos
Antígenos CD34/metabolismo , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Porfiria Eritropoética/terapia , Retroviridae/genética , Uroporfirinogênio III Sintetase/genética , Antígenos CD34/genética , Medula Óssea/enzimologia , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Porfirinas/metabolismo , Transdução Genética , Células Tumorais Cultivadas
13.
J Viral Hepat ; 8(1): 41-7, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11155151

RESUMO

Previous studies have shown a high prevalence of hepatitis C virus (HCV) infection in patients with porphyria cutanea tarda (PCT). The aim of this study was to assess hepatic porphyrin concentrations (HPC) and hepatic uroporphyrinogen decarboxylase (UROD) activity in HCV-infected patients free of PCT. Thirty-two HCV-infected patients (20 M, 12 F, mean age 51 years) and seven control patients (4 M, 3 F, mean age 59 years) free of liver disease, were studied. Knodell's score was determined on liver biopsy by two independent anatomopathologists. Measurement of HPC and hepatic UROD activity levels were carried out on liver biopsy. Relative to controls, HCV-infected patients had high HPC levels (mean +/- SD: 47 +/- 20 vs. 17 +/- 6 pmol/mg protein, P < 0.001) and low hepatic UROD activity levels (514 +/- 95 vs. 619 +/- 125 pmol Copro/h/mg protein, P < 0.05). HPC was not correlated with hepatic UROD activity and the increase was due to coproporphyrin accumulation. No correlation was observed between HPC or hepatic UROD activity values and HCV-RNA concentrations, Knodell's score, hepatic fibrosis, periportal necrosis, periportal inflammation or hepatic iron content in HCV-infected patients. Hepatocellular necrosis was significantly correlated with HPC value (P < 0.005). Hence, in HCV-infected patients, HPC is significantly increased and hepatic UROD activity is very slightly decreased as compared to controls. HPC values and UROD activity are not correlated with HCV-RNA concentrations, hepatic iron content and hepatic fibrosis. The small increase in HPC values in hepatitis C infection is linked with hepatic injury and not with a direct effect on hepatic UROD enzyme.


Assuntos
Hepacivirus/fisiologia , Hepatite C/metabolismo , Fígado/metabolismo , Porfirinas/metabolismo , Uroporfirinogênio Descarboxilase/metabolismo , Adulto , Idoso , Feminino , Hepatite C/enzimologia , Hepatite C/virologia , Humanos , Fígado/enzimologia , Fígado/patologia , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue
14.
An. med. interna (Madr., 1983) ; 17(11): 609-613, nov. 2000. tab, graf
Artigo em Es | IBECS | ID: ibc-238

RESUMO

El diagnóstico y tipificación de las porfirias se consigue mediante una adecuada interrelación entre la sintomatología clínica y la detección de anomalías específicas del metabolismo del hemo. Las crisis de las porfirias se caracterizan por un frecuentemente confuso cuadro de síntomas abdomino-psiquico-neurológicos. El rasgo patognomónico de las porfirias agudas es un incremento en la eliminación urinaria de porfobilinógeno, que es detectado rápidamente mediante el sencillo test de Hoesch. Las lesiones dérmicas características de las porfirias cutáneas están asociadas a un acúmulo plasmático de porfirinas. Su presencia es fácil y rápidamente detectada mediante un barrido fluorimétrico de la muestra diluida en PBS (buffer fosfato salino). La caracterización de los defectos moleculares de los genes que codifican las enzimas implicadas en la síntesis del hemo es un complemento para el análisis bioquímico que permite clarificar los casos bioquímicamente inclasificables y facilita el diagnóstico antenatal en los casos homocigotos más severos (AU)


Assuntos
Humanos , Técnicas de Laboratório Clínico , Porfirias/diagnóstico , Porfirias/classificação , Algoritmos , Porfirias , Porfirias
15.
Hepatology ; 32(1): 73-81, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10869291

RESUMO

Erythropoietic protoporphyria (EPP) is characterized clinically by cutaneous photosensitivity and biochemically by the accumulation of excessive amounts of protoporphyrin in erythrocytes, plasma, feces, and other tissues, such as the liver. The condition is inherited as an autosomal dominant or recessive trait, with a deficiency of ferrochelatase activity. A major concern in EPP patients is the development of cholestasis with accumulation of protoporphyrin in hepatobiliary structures and progressive cellular damage, which can rapidly lead to fatal hepatic failure. The availability of a mouse model for the disease, the Fech(m1Pas)/Fech(m1Pas) mutant mouse, allowed us to test a cellular therapy protocol to correct the porphyric phenotype. When Fech/Fech mice received bone marrow cells from normal animals, the accumulation of protoporphyrin in red blood cells and plasma was reduced 10-fold but still remained 2.5 times above normal levels. Interestingly, in very young animals, bone marrow transplantation can prevent hepatobiliary complications as well as hepatocyte alterations and partially reverse protoporphyrin accumulation in the liver. Bone marrow transplantation may be an option for EPP patients who are at risk of developing hepatic complications.


Assuntos
Transplante de Medula Óssea , Fígado/patologia , Porfiria Eritropoética/terapia , Animais , Feminino , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Protoporfirinas/biossíntese
16.
An Med Interna ; 17(11): 609-13, 2000 Nov.
Artigo em Espanhol | MEDLINE | ID: mdl-11322037

RESUMO

The relationship between clinical features and specific alterations of heme metabolism allows the accurate diagnosis and classification of porphyrias. The acute porphyric attack is characterized by frequently confusing clinical pattern of abdominal-psiquic- and neurological symptoms. An increased urinary excretion of porphobilinogen, which can be quickly detected by the Hoesch test, confirms the diagnosis of this acute attack. Increased plasma porphyrin levels are associated with skin lesions, which are the characteristic features of the cutaneous porphyrias. Their presence is easily and rapidly detected by a fluorimetric scanning of PBS (phosphate buffer saline) diluted samples. Characterization of the molecular defects in genes coding for the enzymes involved in the heme synthetic pathway is complementary to the biochemical methods. Molecular analysis permits an accurate classification of those biochemically unclassified patients and allows prenatal diagnosis in those homozygotic cases where a severe prognosis is suspected.


Assuntos
Porfirias/diagnóstico , Algoritmos , Humanos , Porfirias/classificação
17.
Hum Mutat ; 14(3): 222-32, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10477430

RESUMO

The two porphyrias, familial porphyria cutanea tarda (fPCT) and hepatoerythropoietic porphyria (HEP), are associated with mutations in the gene encoding the enzyme uroporphyrinogen decarboxylase (UROD). Several mutations, most of which are private, have been identified in HEP and fPCT patients, confirming the heterogeneity of the underlying genetic defects of these diseases. We have established a denaturing gradient gel electrophoresis (DGGE) assay for mutation detection in the UROD gene, enabling the simultaneous screening for known and unknown mutations. The established assay has proved able to detect the underlying UROD mutation in 10 previously characterized DNA samples as well as a new mutation in each of six previously unexamined PCT patients. The six novel UROD mutations comprise three missense mutations (M01T, F229L, and M324T), two splice mutations (IVS3-2A-->T and IVS5-2A-->G) leading to exon skipping, and a 2-bp deletion (415-416delTA) resulting in a frameshift and the introduction of a premature stop codon. Heterologous expression and enzymatic studies of the mutant proteins demonstrate that the three mutations leading to shortening or truncation of the UROD protein have no residual catalytic activity, whereas the two missense mutants retained some residual activity. Furthermore, the missense mutants exhibited a considerable increase in thermolability. The six new mutations bring to a total of 29 the number of disease-related mutations in the UROD gene. The DGGE assay presented greatly improves the genetic diagnosis of fPCT and HEP, thereby facilitating the detection of familial UROD deficient patients as well as the discrimination between familial and sporadic PCT cases.


Assuntos
Testes Genéticos/métodos , Mutação de Sentido Incorreto , Porfiria Cutânea Tardia/enzimologia , Uroporfirinogênio Descarboxilase/genética , Alelos , Análise Mutacional de DNA , Eletroforese em Gel de Poliacrilamida , Estabilidade Enzimática , Éxons/genética , Expressão Gênica , Heterozigoto , Homozigoto , Humanos , Mutagênese Sítio-Dirigida , Splicing de RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Temperatura
18.
J Interferon Cytokine Res ; 19(5): 533-41, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10386866

RESUMO

Gene transduction into immature human hematopoietic cells collected from umbilical cord blood, bone marrow, or mobilized peripheral blood cells could be useful for the treatment of genetic and acquired disorders of the hematopoietic system. Immunodeficient mouse models have been used frequently as recipients to assay the growth and differentiation of human hematopoietic stem/progenitor cells. Indeed, high levels of human cell engraftment were first reported in human/murine chimeras using NOD/SCID mice, which now are considered as the standard for these types of experiments. However, NOD/SCID mice have some clear disadvantages (including spontaneous tumor formation) that limit their general use. We have developed a new immunodeficient mouse model by combining recombinase activating gene-2 (RAG2) and common cytokine receptor gamma chain (gamma c) mutations. The RAG2-/-/gamma c- double mutant mice are completely alymphoid (T-, B-, NK-), show no spontaneous tumor formation, and exhibit normal hematopoietic parameters. Interestingly, human cord blood cell engraftment in RAG2-/-/gamma c- mice was greatly enhanced by the exogenous administration of human cytokines interleukin-(IL-3) granulocyte-macrophage colony-stimulating factor, (GM-CSF), and erythropoietin in contrast to the NOD/SCID model. This unique feature of the RAG2-/-/gamma c- mouse model should be particularly well suited for assessing the role of different cytokines in human lymphopoiesis and stem/progenitor cell function in vivo.


Assuntos
Citocinas/farmacologia , Transplante de Células-Tronco Hematopoéticas , Fragmentos de Peptídeos/genética , Receptores de Citocinas/genética , Imunodeficiência Combinada Severa/genética , Animais , Antígenos CD34/sangue , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-3/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação , Proteínas Nucleares , Fenótipo , Receptores de Citocinas/química
19.
Blood ; 94(2): 465-74, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10397714

RESUMO

Hepatoerythropoietic porphyria (HEP) is an inherited metabolic disorder characterized by the accumulation of porphyrins resulting from a deficiency in uroporphyrinogen decarboxylase (UROD). This autosomal recessive disorder is severe, starting early in infancy with no specific treatment. Gene therapy would represent a great therapeutic improvement. Because hematopoietic cells are the target for somatic gene therapy in this porphyria, Epstein-Barr virus-transformed B-cell lines from patients with HEP provide a model system for the disease. Thus, retrovirus-mediated expression of UROD was used to restore enzymatic activity in B-cell lines from 3 HEP patients. The potential of gene therapy for the metabolic correction of the disease was demonstrated by a reduction of porphyrin accumulation to the normal level in deficient transduced cells. Mixed culture experiments demonstrated that there is no metabolic cross-correction of deficient cells by normal cells. However, the observation of cellular expansion in vitro and in vivo in immunodeficient mice suggested that genetically corrected cells have a competitive advantage. Finally, to facilitate future human gene therapy trials, we have developed a selection system based on the expression of the therapeutic gene. Genetically corrected cells are easily separated from deficient ones by the absence of fluorescence when illuminated under UV light.


Assuntos
Linfócitos B/enzimologia , Terapia Genética , Porfiria Hepatoeritropoética/enzimologia , Uroporfirinogênio Descarboxilase/deficiência , Animais , Linfócitos B/transplante , Linhagem Celular Transformada , Transformação Celular Viral , Técnicas de Cocultura , Citometria de Fluxo , Herpesvirus Humano 4 , Humanos , Masculino , Camundongos , Camundongos Mutantes , Microscopia de Fluorescência , Porfiria Hepatoeritropoética/genética , Porfiria Hepatoeritropoética/terapia , Seleção Genética , Transfecção , Raios Ultravioleta , Uroporfirinogênio Descarboxilase/genética
20.
Ecotoxicol Environ Saf ; 43(1): 98-102, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10330327

RESUMO

The heme biosynthetic pathway is a metabolic target of alcohol and lead poisoning. To analyze the interdependence of both xenobiotics on porphyrin metabolism, male Wistar rats (n=47) were divided into four groups and were fed Lieber-DeCarli semiliquid control or alcoholic diets containing or not containing lead acetate (160 mg/liter) for 8 weeks. After this period, hematocrit values and porphyrin concentration in liver and urine were similar in all groups, indicating that the goal of inducing only mild chronic intoxication was achieved. Compared with the control group, rats poisoned only with lead exhibited high levels of this metal in blood and liver, increased erythrocytic protoporphyrin, and hypoactivity of aminolevulinate dehydrase (ALA-D) in both blood and liver. Rats intoxicated only with alcohol exhibited mild hypoactivity of both hepatic and erythrocytic ALA-D, although such decreased enzymatic values did not achieve statistical significance. Rats receiving ethanol and lead simultaneously demonstrated abnormalities in heme biosynthesis similar to those in rats exposed to lead, although zinc hepatic levels decreased significantly only in animals exposed to both xenobiotics. Hepatic GSH and urinary ALA and porphyrin levels were maintained in a similar range in all groups.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Heme/metabolismo , Chumbo/toxicidade , Ácido Aminolevulínico/metabolismo , Ácido Aminolevulínico/urina , Animais , Sinergismo Farmacológico , Eritrócitos/enzimologia , Hematócrito , Heme/biossíntese , Intoxicação por Chumbo/sangue , Intoxicação por Chumbo/metabolismo , Intoxicação por Chumbo/urina , Fígado/enzimologia , Fígado/metabolismo , Masculino , Sintase do Porfobilinogênio/sangue , Sintase do Porfobilinogênio/metabolismo , Porfirinas/metabolismo , Porfirinas/urina , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...