Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
2.
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
3.
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
4.
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
5.
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
6.
Cancer Gene Ther ; 15(4): 241-51, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18202716

RESUMO

Xeroderma pigmentosum type C (XPC) is a rare autosomal recessive disorder that occurs due to inactivation of the XPC protein, an important DNA damage recognition protein involved in DNA nucleotide excision repair (NER). This defect, which prevents removal of a wide array of direct and indirect DNA lesions, is associated with a decrease in catalase activity. To test the hypothesis of a novel photoprotective approach, we irradiated epidermis reconstructed with XPC human keratinocytes sustainably overexpressing lentivirus-mediated catalase enzyme. Following UVB irradiation, there was a marked decrease in sunburn cell formation, caspase-3 activation and p53 accumulation in human XPC-reconstructed epidermis overexpressing catalase. Moreover, XPC-reconstructed epidermis was more resistant to UVB-induced apoptosis than normal reconstructed epidermis. While not correcting the gene defect, indirect gene therapy using antioxidant enzymes may be of help in limiting photosensitivity in XPC and probably in other monogenic/polygenic photosensitive disorders characterized by ROS accumulation.


Assuntos
Apoptose/efeitos da radiação , Catalase/genética , Epiderme/patologia , Raios Ultravioleta , Xeroderma Pigmentoso/patologia , Células Cultivadas , Epiderme/enzimologia , Vetores Genéticos , Humanos , Lentivirus/genética , Xeroderma Pigmentoso/enzimologia
7.
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
8.
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
9.
Mol Imaging ; 2(1): 11-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12926233

RESUMO

Spatial and temporal control of transgene expression is one of the major prerequisites of efficient gene therapy. Recently, a noninvasive, physical approach has been presented based on local heat in combination with a heat-sensitive promoter. This strategy requires tight temperature control in vivo. Here, we use MRI-guided focused ultrasound (MRI-FUS) with real-time feedback control on a whole-body clinical MRI system for a completely automatic execution of a predefined temperature-time trajectory in the focal point. Feasibility studies on expression control were carried out on subcutaneously implanted rat tumors. A stable modified C6 glioma cell line was used carrying a fused gene coding for thymidine kinase (TK) and green fluorescent protein (GFP) under control of the human heat-shock protein 70 (HSP70) promoter. In vitro studies showed strong induction of the TK-GFP gene expression upon heat shock under various conditions and localization of the protein product in the nucleus. In vivo tumors were subjected to a 3-min temperature elevation using MRI-FUS with a constant temperature, and were analysed 24 hr after the heat shock with respect to GFP fluorescence. Preliminary results showed strong local induction in regions heated above 40 degrees C, and a good correspondence between temperature maps at the end of the heating period and elevated expression of TK-GFP.


Assuntos
Expressão Gênica , Terapia Genética , Imageamento por Ressonância Magnética/métodos , Animais , Proteínas de Fluorescência Verde , Proteínas de Choque Térmico HSP70/genética , Humanos , Técnicas In Vitro , Proteínas Luminescentes/genética , Imageamento por Ressonância Magnética/instrumentação , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/genética , Temperatura , Timidina Quinase/genética , Transfecção , Células Tumorais Cultivadas
10.
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
11.
J Gene Med ; 5(4): 333-42, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12692867

RESUMO

BACKGROUND: Among the techniques used to induce and control gene expression, a non-invasive, physical approach based on local heat in combination with a heat-sensitive promoter represents a promising alternative but requires accurate temperature control in vivo. MRI-guided focused ultrasound (MRI-FUS) with real-time feedback control allows automatic execution of a predefined temperature-time trajectory. The purpose of this study was to demonstrate temporal and spatial control of transgene expression based on a well-defined local hyperthermia generated by MRI-FUS. METHODS: Expression of the green fluorescent protein (GFP) marker gene was used. Two cell lines were derived from C6 glioma cells. The GFP expression of the first one is under the control of the CMV promoter, whereas it is under the control of the HSP70 promoter in the second one and thus inducible by heat. Subcutaneous tumours were generated by injection in immuno-deficient mice and rats. Tumours were subjected to temperatures varying from 42 to 50 degrees C for 3 to 25 min controlled by MRI-FUS and analyzed 24 h after the heat-shock. Endogenous HSP70 expression and C6 cell distribution were also analyzed. RESULTS: The results demonstrate strong expression at 50 degrees C applied during a short time period (3 min) without affecting cell viability. Induced expression was also clearly shown for temperature in the range 44-48 degrees C but not at 42 degrees C. CONCLUSIONS: Heating with MRI-FUS allows a tight and non-invasive control of transgene expression in a tumour.


Assuntos
Regulação da Expressão Gênica , Temperatura Alta , Imageamento por Ressonância Magnética/métodos , Regiões Promotoras Genéticas/genética , Ultrassonografia/métodos , Animais , Glioma/genética , Glioma/patologia , Proteínas de Choque Térmico HSP70/genética , Resposta ao Choque Térmico/genética , Humanos , Hipertermia Induzida , Camundongos , Camundongos Mutantes , Neoplasias de Tecido Conjuntivo/genética , Neoplasias de Tecido Conjuntivo/patologia , Neoplasias de Tecido Conjuntivo/secundário , Ratos , Ratos Wistar , Fatores de Tempo , Transgenes , Células Tumorais Cultivadas
12.
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
13.
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
14.
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
15.
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
16.
J Interferon Cytokine Res ; 20(6): 577-87, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10888114

RESUMO

Interferon (IFN) is an effective treatment for chronic myeloid leukemia (CML) in chronic phases, and a number of in vitro antileukemic effects of IFN on CML cells have been reported. The transfer of cytokine genes into tumor cells is reportedly a valuable approach to improve the antitumor activity of cytokines in various models. We first investigated the possibility of transducing CML cells with the retroviral vectors LIalpha2SN and LIgammaSN, encoding the IFN-alpha2 and IFN-gamma genes, respectively, and with the bicistronic vector LIalpha2IrIgammaSN coexpressing the IFN-alpha2 and IFN-gamma genes. We then analyzed the effects of IFN-alpha2 and IFN-gamma produced alone or simultaneously on the proliferation of CML cells. We optimized the transduction efficiency by using the CML-derived K562 cell line. We then introduced IFN genes into CML CD34+ cells. Secretion of IFN-alpha and IFN-gamma was demonstrated in K562 and CML CD34+ cells transduced with the different vectors. The MHC class I antigens were overexpressed in both K562 and CML CD34+ transduced cells. Inhibition of the proliferation of LIalpha2IrIgammaSN-transduced CML cells was greater than with the LIalpha2SN and the LIgammaSN-transduced CML cells. We demonstrate an additive effect of IFN-alpha and IFN-gamma on the inhibition of K562 and CML CD34+ cell proliferation.


Assuntos
Interferon Tipo I/genética , Interferon gama/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Antígenos CD34/metabolismo , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Primers do DNA/genética , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/farmacologia , Interferon gama/biossíntese , Interferon gama/farmacologia , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Proteínas Recombinantes , Retroviridae/genética , Transdução Genética , Ensaio Tumoral de Célula-Tronco
17.
J Hepatol ; 32(5): 805-11, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10845668

RESUMO

BACKGROUND/AIMS: Histological and biochemical iron overload has been reported in non-tumoral liver of most patients presenting an hepatocellular carcinoma (HCC) developed in non-cirrhotic liver (NCL). The aim of our study was to investigate HFE mutations in patients with HCC in NCL. METHODS: Thirty-five patients with HCC in NCL were included either retrospectively or prospectively. Clinical data, iron and viral status, and HFE gene mutations were compared between groups with (I+, n = 19) or without histological iron overload (I-, n = 16). RESULTS: Twenty per cent of patients were HBV or HCV positive. Fifty-four per cent had hepatocytic iron overload at histology. Mean hepatic iron concentration was 100.2 +/- 14.6 micromol/g in I+ versus 23.2 +/- 2.1 micromol/g in I- (p<0.001). Among the 19 I+ patients, eight mutations were found: two C282Y/C282Y, three C282Y/WT, two C282Y/H63D and one H63D/H63D. None of these mutations was found in the I- group. There was no significant difference concerning the H63D heterozygous mutation between I+ or I- patients. CONCLUSIONS: In patients with HCC in NCL, HBV and HCV markers are rare (20%), and mild iron overload is frequent (54%). In patients with HCC in NCL and iron overload, C282Y mutations are frequent (36.8% of cases) and significantly increased (p<0.009) compared to HCC in NCL without iron overload; these mutations are mostly heterozygous. H63D heterozygosity is not associated with liver iron overload. Because of the small size of the series, HFE C282Y mutation should be investigated on a larger scale in patients with HCC in NCL with iron overload in order to confirm this association.


Assuntos
Carcinoma Hepatocelular/genética , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe I/genética , Sobrecarga de Ferro/genética , Neoplasias Hepáticas/genética , Proteínas de Membrana , Idoso , Carcinoma Hepatocelular/patologia , Feminino , Frequência do Gene , Genes MHC Classe I , Proteína da Hemocromatose , Heterozigoto , Humanos , Incidência , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Mutação
18.
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
19.
J Gene Med ; 2(2): 89-96, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10809142

RESUMO

BACKGROUND: Local production of therapeutic proteins, e.g. for cancer treatments, is based on gene therapy approaches and requires tight spatial and temporal control of gene expression. Here we demonstrate the use of local hyperthermia of varying intensity and duration to control the expression of a transgene under control of the thermoinducible hsp70 (heat shock protein) promoter. METHODS: Heat-induced expression of the EGFP (green fluorescent protein) reporter gene was characterized using a stably transfected glioma C6 cell line expressing the EGFP gene under control of the heat inducible minimal hsp70 promoter both in vitro and in vivo for subcutaneous tumors in immunodeficient mice. RESULTS: A heat shock of 20-30 min at 43 degrees C in cell culture led to a maximum EGFP concentration at about 24 h. Heat treatments at higher temperature (up to 48 degrees C) but with shorter durations (down to 30 s) also induced strong EGFP expression. Local heating in situ led to gradients in EGFP expression which decreased with increasing distance from the heat source. CONCLUSION: Local hyperthermia, in combination with a heat sensitive promoter, represents a method for the spatial and temporal control of transgene expression.


Assuntos
Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Luminescentes/metabolismo , Regiões Promotoras Genéticas , Animais , Western Blotting , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde , Calefação , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos , Ratos , Transfecção , Células Tumorais Cultivadas
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA