Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nat Commun ; 14(1): 4072, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37429857

RESUMEN

The CRISPR-Cas9 system has revolutionized our ability to precisely modify the genome and has led to gene editing in clinical applications. Comprehensive analysis of gene editing products at the targeted cut-site has revealed a complex spectrum of outcomes. ON-target genotoxicity is underestimated with standard PCR-based methods and necessitates appropriate and more sensitive detection methods. Here, we present two complementary Fluorescence-Assisted Megabase-scale Rearrangements Detection (FAMReD) systems that enable the detection, quantification, and cell sorting of edited cells with megabase-scale loss of heterozygosity (LOH). These tools reveal rare complex chromosomal rearrangements caused by Cas9-nuclease and show that LOH frequency depends on cell division rate during editing and p53 status. Cell cycle arrest during editing suppresses the occurrence of LOH without compromising editing. These data are confirmed in human stem/progenitor cells, suggesting that clinical trials should consider p53 status and cell proliferation rate during editing to limit this risk by designing safer protocols.


Asunto(s)
Sistemas CRISPR-Cas , Proteína p53 Supresora de Tumor , Humanos , Sistemas CRISPR-Cas/genética , Proteína p53 Supresora de Tumor/genética , Puntos de Control del Ciclo Celular/genética , División Celular , Separación Celular , ARN
2.
Nat Commun ; 12(1): 4922, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34389729

RESUMEN

CRISPR-Cas9 is a promising technology for gene therapy. However, the ON-target genotoxicity of CRISPR-Cas9 nuclease due to DNA double-strand breaks has received little attention and is probably underestimated. Here we report that genome editing targeting globin genes induces megabase-scale losses of heterozygosity (LOH) from the globin CRISPR-Cas9 cut-site to the telomere (5.2 Mb). In established lines, CRISPR-Cas9 nuclease induces frequent terminal chromosome 11p truncations and rare copy-neutral LOH. In primary hematopoietic progenitor/stem cells, we detect 1.1% of clones (7/648) with acquired megabase LOH induced by CRISPR-Cas9. In-depth analysis by SNP-array reveals the presence of copy-neutral LOH. This leads to 11p15.5 partial uniparental disomy, comprising two Chr11p15.5 imprinting centers (H19/IGF2:IG-DMR/IC1 and KCNQ1OT1:TSS-DMR/IC2) and impacting H19 and IGF2 expression. While this genotoxicity is a safety concern for CRISPR clinical trials, it is also an opportunity to model copy-neutral-LOH for genetic diseases and cancers.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Globinas/genética , Células Madre Hematopoyéticas/metabolismo , Pérdida de Heterocigocidad/genética , Eliminación de Secuencia , Células Cultivadas , Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Metilación de ADN , Expresión Génica , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante/genética
3.
Biochem Biophys Res Commun ; 569: 23-28, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34216994

RESUMEN

Intravenous injections of human hematopoietic stem cells (hHSCs) is routinely used in clinic and for modeling hematopoiesis in mice. However, unspecific dilution in vascular system and non-hematopoietic organs challenges engraftment efficiency. Although spleen is capable of extra medullar hematopoiesis, its ability to support human HSC transplantation has never been evaluated. We demonstrate that intra-splenic injection results in high and sustained engraftment of hHSCs into immune-deficient mice, with higher chimerisms than with intravenous or intra-femoral injections. Our results support that spleen microenvironment provides a niche for HSCs amplification and offers a new route for efficient HSC transplantation.


Asunto(s)
Supervivencia de Injerto/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Bazo/citología , Animales , Antígenos CD34/metabolismo , Femenino , Citometría de Flujo/métodos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inyecciones , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Bazo/metabolismo , Quimera por Trasplante , Trasplante Heterólogo
4.
Leukemia ; 31(1): 65-74, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27220663

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Fusión bcr-abl/fisiología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Sistema de Señalización de MAP Quinasas/fisiología , Células Madre Neoplásicas/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Células Tumorales Cultivadas
5.
Diabetologia ; 52(8): 1608-17, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19513688

RESUMEN

AIMS/HYPOTHESIS: Wingless and iNT-1 (WNT) pathway members are critical for pancreatic development and exocrine tissue formation. Recently, much attention has focused on delineating the roles of beta-catenin in pancreatic organogenesis. However, little is known about the involvement of beta-catenin in the endocrine or exocrine function of the mature pancreas. We report for the first time the impact of beta-catenin deletion in the pancreatic beta cells. METHODS: We targeted the deletion of the beta-catenin gene in pancreatic beta cells by crossing a floxed beta-catenin mouse strain with a RIP-Cre mouse strain. RESULTS: Surprisingly, the majority of the mutant mice died shortly after birth and had deregulated glucose and insulin levels. The newborn mutant pancreases demonstrated increased insulin content, reflecting a defect in insulin release confirmed in vitro. Moreover, there was a reduction in total endocrine tissue at birth, while cellularity in islets was greater, suggesting that lack of beta-catenin affects beta cell size. Some newborns survived beta-catenin deletion and showed a milder phenotype during adulthood. CONCLUSIONS/INTERPRETATION: The deletion of beta-catenin in the maturing beta cells negatively impacts on islet morphology and function. This work reveals that lack of beta-catenin in early life is related to severe deregulation of glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Islotes Pancreáticos/patología , beta Catenina/deficiencia , Animales , Animales Recién Nacidos , Cruzamientos Genéticos , ADN/genética , ADN/aislamiento & purificación , Eliminación de Gen , Hiperglucemia/genética , Hiperinsulinismo/genética , Hipoglucemia/genética , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Endogámicos , Reacción en Cadena de la Polimerasa , beta Catenina/genética
6.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 53-60, 2009 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-19268002

RESUMEN

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.


Asunto(s)
Mutación/fisiología , Fenotipo , Porfiria Eritropoyética/genética , Genotipo , Humanos , Mutación/genética , Porfiria Eritropoyética/enzimología , Uroporfirinógeno III Sintetasa/genética , Uroporfirinógeno III Sintetasa/fisiología
7.
Leukemia ; 23(4): 679-85, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158834

RESUMEN

Imatinib is an effective first-line therapy for chronic myelogenous leukemia (CML) that acts by targeting the tyrosine kinase activity of BCR-ABL. To overcome resistance, second-generation inhibitors of BCR-ABL have been developed. Among these, nilotinib is more potent against BCR-ABL than imatinib, and is effective against many imatinib-resistant BCR-ABL mutants. In this study, an in vitro flow cytometry assay to analyze imatinib- and nilotinib-induced apoptosis in CML cells has been developed. Both the drugs induced significant apoptosis in CD34+ cells from 36 CML bone marrow samples (P<10(-4)), whereas CD34+ cells from BCR-ABL negative samples were unaffected. When the experiments were carried out in the presence of a cocktail of cytokines, nilotinib- but not imatinib-induced apoptosis was inhibited. This differential inhibition was confirmed on K562 cells. A blocking anti-CD117 antibody alleviated the antiapoptotic effect of cytokines against nilotinib. Moreover, using short hairpin RNA against BCR-ABL, we showed that K562 cells were not dependent on BCR-ABL signaling as long as the stem cell factor (SCF) receptor pathway was activated. We conclude that the c-KIT pathway may substitute for BCR-ABL tyrosine kinase to activate survival signals, and that c-KIT must be inhibited besides Bcr-Abl to allow apoptosis of CML cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Benzamidas , Citocinas/farmacología , Humanos , Mesilato de Imatinib , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Pirimidinas/farmacología , Factor de Células Madre/antagonistas & inhibidores , Células Tumorales Cultivadas
8.
Leukemia ; 21(1): 93-101, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17109025

RESUMEN

Imatinib is an effective therapy for chronic myeloid leukemia (CML), a myeloproliferative disorder characterized by the expression of the recombinant oncoprotein Bcr-Abl. In this investigation, we studied an imatinib-resistant cell line (K562-r) generated from the K562 cell line in which none of the previously described mechanisms of resistance had been detected. A threefold increase in the expression of the heat-shock protein 70 (Hsp70) was detected in these cells. This increase was not associated to heat-shock transcription factor-1 (HSF-1) overexpression or activation. RNA silencing of Hsp70 decreased dramatically its expression (90%), and was accompanied by a 34% reduction in cell viability. Overexpression of Hsp70 in the imatinib-sensitive K562 line induced resistance to imatinib as detected by a large reduction in cell death in the presence of 1 muM of imatinib. Hsp70 level was also increased in blast cells of CML patients resistant to imatinib, whereas the level remained low in responding patients. Taken together, the results demonstrate that overexpression of Hsp70 can lead to both in vitro and in vivo resistance to imatinib in CML cells. Moreover, the overexpression of Hsp70 detected in imatinib-resistant CML patients supports this mechanism and identifies potentially a marker and a therapeutic target of CML evolution.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas HSP70 de Choque Térmico/biosíntesis , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Regulación hacia Arriba , Biomarcadores de Tumor , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/genética , Regulación Neoplásica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo
9.
Genomics ; 87(1): 84-92, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16314073

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Mutación Missense , Porfiria Eritropoyética/enzimología , Uroporfirinógeno III Sintetasa/genética , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Terapia Genética , Ratones , Ratones Transgénicos , Porfiria Eritropoyética/patología , Porfiria Eritropoyética/terapia , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinas/metabolismo
10.
Gene Ther ; 11(22): 1638-47, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15284838

RESUMEN

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.


Asunto(s)
Terapia Genética/métodos , Guanina/análogos & derivados , O(6)-Metilguanina-ADN Metiltransferasa/genética , Protoporfiria Eritropoyética/terapia , Trasplante de Células Madre , Animales , Antineoplásicos/uso terapéutico , Carmustina/uso terapéutico , Femenino , Ferroquelatasa/genética , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Guanina/uso terapéutico , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Protoporfiria Eritropoyética/enzimología , Células Madre/enzimología , Transducción Genética/métodos
11.
J Mol Med (Berl) ; 81(5): 310-20, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12721665

RESUMEN

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.


Asunto(s)
Terapia Genética , Lentivirus/genética , Porfiria Eritropoyética/terapia , Uroporfirinógeno III Sintetasa/genética , Adulto , Técnicas de Cultivo de Célula , Diferenciación Celular , Eritroblastos/metabolismo , Fluorescencia , Expresión Génica , Vectores Genéticos , Humanos , Fenotipo , Porfiria Eritropoyética/genética , Transducción Genética , Replicación Viral
12.
Mol Ther ; 4(4): 331-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11592836

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Terapia Genética/métodos , Lentivirus/genética , Porfiria Hepatoeritropoyética/genética , Porfiria Hepatoeritropoyética/terapia , Animales , Southern Blotting , Trasplante de Médula Ósea , Línea Celular , Elementos de Facilitación Genéticos/genética , Femenino , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Ferroquelatasa/uso terapéutico , Expresión Génica/genética , Vectores Genéticos/genética , Humanos , Lentivirus/fisiología , Masculino , Ratones , Especificidad de Órganos , Porfiria Hepatoeritropoyética/enzimología , Porfiria Hepatoeritropoyética/patología , Porfirinas/metabolismo , Regiones Promotoras Genéticas/genética , Protoporfiria Eritropoyética , Piel/patología , Transducción Genética
13.
Blood ; 98(9): 2664-72, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11675336

RESUMEN

Use of oncoretroviral vectors in gene therapy for hemoglobinopathies has been impeded by low titer vectors, genetic instability, and poor expression. Fifteen self- inactivating (SIN) lentiviral vectors using 4 erythroid promoters in combination with 4 erythroid enhancers with or without the woodchuck hepatitis virus postregulatory element (WPRE) were generated using the enhanced green fluorescent protein as a reporter gene. Vectors with high erythroid-specific expression in cell lines were tested in primary human CD34(+) cells and in vivo in the murine bone marrow (BM) transplantation model. Vectors containing the ankyrin-1 promoter showed high-level expression and stable proviral transmission. Two vectors containing the ankyrin-1 promoter and 2 erythroid enhancers (HS-40 plus GATA-1 or HS-40 plus 5-aminolevulinate synthase intron 8 [I8] enhancers) and WPRE expressed at levels higher than the HS2/beta-promoter vector in bulk unilineage erythroid cultures and individual erythroid blast-forming units derived from human BM CD34(+) cells. Sca1(+)/lineage(-) Ly5.1 mouse hematopoietic cells, transduced with these 2 ankyrin-1 promoter vectors, were injected into lethally irradiated Ly5.2 recipients. Eleven weeks after transplantation, high-level expression was seen from both vectors in blood (63%-89% of red blood cells) and erythroid cells in BM (70%-86% engraftment), compared with negligible expression in myeloid and lymphoid lineages in blood, BM, spleen, and thymus (0%-4%). The I8/HS-40-containing vector encoding a hybrid human beta/gamma-globin gene led to 43% to 113% human gamma-globin expression/copy of the mouse alpha-globin gene. Thus, modular use of erythroid-specific enhancers/promoters and WPRE in SIN-lentiviral vectors led to identification of high-titer, stably transmitted vectors with high-level erythroid-specific expression for gene therapy of red cell diseases.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Vectores Genéticos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Transducción Genética/métodos , Animales , Antígenos CD34 , Células de la Médula Ósea , Trasplante de Médula Ósea , Elementos de Facilitación Genéticos , Expresión Génica , Regulación Viral de la Expresión Génica , Vectores Genéticos/normas , Proteínas Fluorescentes Verdes , Virus de la Hepatitis B de la Marmota/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Modelos Animales , Regiones Promotoras Genéticas , Procesamiento Postranscripcional del ARN/genética , Transducción Genética/normas , gammaglobulinas/genética , gammaglobulinas/metabolismo
14.
Gene Ther ; 8(8): 618-26, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11320408

RESUMEN

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.


Asunto(s)
Separación Celular/métodos , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Porfiria Hepatoeritropoyética/terapia , Animales , Línea Celular , ADN Complementario/genética , Modelos Animales de Enfermedad , Femenino , Ferroquelatasa/genética , Citometría de Flujo , Vectores Genéticos , Hematopoyesis , Interleucina-3/fisiología , Hepatopatías/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Fenotipo , Trastornos por Fotosensibilidad/terapia , Porfiria Hepatoeritropoyética/fisiopatología , Retroviridae/genética
15.
Mol Ther ; 3(3): 411-7, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11273784

RESUMEN

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.


Asunto(s)
Antígenos CD34/metabolismo , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Porfiria Eritropoyética/terapia , Retroviridae/genética , Uroporfirinógeno III Sintetasa/genética , Antígenos CD34/genética , Médula Ósea/enzimología , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Lentivirus/genética , Porfirinas/metabolismo , Transducción Genética , Células Tumorales Cultivadas
16.
Blood ; 94(2): 465-74, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10397714

RESUMEN

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.


Asunto(s)
Linfocitos B/enzimología , Terapia Genética , Porfiria Hepatoeritropoyética/enzimología , Uroporfirinógeno Descarboxilasa/deficiencia , Animales , Linfocitos B/trasplante , Línea Celular Transformada , Transformación Celular Viral , Técnicas de Cocultivo , Citometría de Flujo , Herpesvirus Humano 4 , Humanos , Masculino , Ratones , Ratones Mutantes , Microscopía Fluorescente , Porfiria Hepatoeritropoyética/genética , Porfiria Hepatoeritropoyética/terapia , Selección Genética , Transfección , Rayos Ultravioleta , Uroporfirinógeno Descarboxilasa/genética
17.
J Interferon Cytokine Res ; 19(5): 533-41, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10386866

RESUMEN

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.


Asunto(s)
Citocinas/farmacología , Trasplante de Células Madre Hematopoyéticas , Fragmentos de Péptidos/genética , Receptores de Citocinas/genética , Inmunodeficiencia Combinada Grave/genética , Animales , Antígenos CD34/sangre , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-3/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Proteínas Nucleares , Fenotipo , Receptores de Citocinas/química
18.
Ann Biol Clin (Paris) ; 57(1): 43-50, 1999.
Artículo en Francés | MEDLINE | ID: mdl-9920966

RESUMEN

Gene transfer in hematopoietic cells is intended to treat patients with malignant disease and inherited monogenic (hematological, immunological, and metabolic) disorders. Hematopoietic progenitor or stem cells are a favoured target for gene therapy because these cells are easily withdrawn from the patient, expanded and genetically modified ex vivo and then reinjected into the organism. Retroviral vectors allow an efficient transfer of the genes of interest. Transduction of stem cells leads to a stable expression of the transgene for long periods of time. However, we are at the beginning of this new therapeutic application, the technique was being already successful in very few cases. Problems to be solved are mainly in the understanding of the physiology of the hematopoietic stem cell and in the improvement of technical qualities of the vectors for a targeted gene transfer in vivo.


Asunto(s)
Terapia Genética/métodos , Virus Defectuosos/genética , Expresión Génica , Técnicas de Transferencia de Gen , Enfermedades Genéticas Congénitas/terapia , Marcadores Genéticos , Vectores Genéticos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Neoplasias/terapia , Retroviridae
19.
J Gene Med ; 1(5): 322-30, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10738549

RESUMEN

BACKGROUND: Congenital erythropoietic porphyria (CEP) is an inherited disease caused by a deficiency of uroporphyrinogen III synthase, the fourth enzyme of the haem 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 occurring in early adult life and available treatments are only symptomatic and unsatisfactory. In vitro gene transfer experiments have documented the feasibility of gene therapy via haematopoietic stem cells to treat this disease. To facilitate future ex vivo gene therapy in humans, the design of efficient selection procedures to increase the frequency of genetically corrected cells prior to autologous transplantation is a critical step. METHODS: An alternative selection procedure based upon expression of a transferred gene was performed on a lymphoblastoid (LB) cell line from a patient with congenital erythropoietic porphyria to obtain high frequencies of genetically modified cells. The presence of exogeneous delta-aminolevulinic acid (ALA), a haem precursor, induces an increase in porphyrin accumulation in LB deficient cells. Porphyrins exhibit a specific fluorescent emission and can be detected by cytofluorimetry under ultraviolet excitation. RESULTS: In genetically modified cells, the restored metabolic flow from ALA to haem led to a lesser accumulation of porphyrins in the cells, which were easily separated from the deficient cells by flow cytometry cell sorting. CONCLUSION: This selection process represents a rapid and efficient procedure and an excellent alternative to the use of potentially harmful gene markers in retroviral vectors.


Asunto(s)
Terapia Genética , Vectores Genéticos , Porfiria Eritropoyética/terapia , Retroviridae/genética , Transducción Genética , Adulto , Ácido Aminolevulínico/farmacología , Línea Celular , Separación Celular , Citometría de Flujo , Humanos , Linfocitos , Melatonina/farmacología , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/metabolismo , Porfirinas/metabolismo
20.
Gene Ther ; 5(4): 556-62, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9614582

RESUMEN

We have developed an efficient and rapid method to analyze transduction in human hematopoietic cells and to select them. We constructed two retroviral vectors using the recombinant humanized S65T green fluorescent protein (rHGFP) gene. Transduced cells appeared with specific green fluorescence on microscopy or fluorescence-activated cell sorting (FACS) analysis. The rHGFP gene was placed under the control of two different retroviral promotors (LTR) in the LGSN vector and in the SF-GFP vector. Amphotropic retroviruses were tested on NIH/3T3 fibroblasts or human hematopoietic (K562, TF-1) cell lines. Then CD34+ cells isolated from cord blood were infected three times after a 48-h prestimulation with IL-3, IL-6, SCF or with IL-3, IL-6, SCF, GM-CSF, Flt3-L and TPO. After 6 days of expansion, a similar number of total CD34(+)-derived cells, CD34+ cells and CFC was obtained in non-transduced and transduced cells, demonstrating the absence of toxicity of the GFP. A transduction up to 46% in total CD34(+)-derived cells and 21% of CD34+ cells was shown by FACS analysis. These results were confirmed by fluorescence of colonies in methyl-cellulose (up to 36% of CFU-GM and up to 25% of BFU-E). The FACS sorting of GFP cells led to 83-100% of GFP-positive colonies after 2 weeks of methyl-cellulose culture. Moreover, a mean gene transfer efficiency of 8% was also demonstrated in longterm culture initiating cells (LTC-IC). This rapid and efficient method represents a substantial improvement to monitor gene transfer and retroviral expression of various vectors in characterized human hematopoietic cells.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , Células Madre Hematopoyéticas , Retroviridae , Transfección , Células Cultivadas , Citometría de Flujo , Expresión Génica , Marcadores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Microscopía Fluorescente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA