Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Ther ; 26(11): 2592-2603, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30122422

RESUMEN

Deficiency of basement membrane heterotrimeric laminin 332 component, coded by LAMA3, LAMB3, and LAMC2 genes, causes junctional epidermolysis bullosa (JEB), a severe skin adhesion defect. Herein, we report the first application of CRISPR/Cas9-mediated homology direct repair (HDR) to in situ restore LAMB3 expression in JEB keratinocytes in vitro and in immunodeficient mice transplanted with genetically corrected skin equivalents. We packaged an adenovector carrying Cas9/guide RNA (gRNA) tailored to the intron 2 of LAMB3 gene and an integration defective lentiviral vector bearing a promoterless quasi-complete LAMB3 cDNA downstream a splice acceptor site and flanked by homology arms. Upon genuine HDR, we exploited the in vitro adhesion advantage of laminin 332 production to positively select LAMB3-expressing keratinocytes. HDR and restored laminin 332 expression were evaluated at single-cell level. Notably, monoallelic-targeted integration of LAMB3 cDNA was sufficient to in vitro recapitulate the adhesive property, the colony formation typical of normal keratinocytes, as well as their cell growth. Grafting of genetically corrected skin equivalents onto immunodeficient mice showed a completely restored dermal-epidermal junction. This study provides evidence for efficient CRISPR/Cas9-mediated in situ restoration of LAMB3 expression, paving the way for ex vivo clinical application of this strategy to laminin 332 deficiency.


Asunto(s)
Sistemas CRISPR-Cas/genética , Moléculas de Adhesión Celular/genética , Epidermólisis Ampollosa de la Unión/terapia , Terapia Genética , Animales , Membrana Basal/patología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/deficiencia , Reparación del ADN/genética , ADN Complementario/genética , Epidermólisis Ampollosa de la Unión/genética , Epidermólisis Ampollosa de la Unión/patología , Regulación de la Expresión Génica , Humanos , Intrones/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Laminina/genética , Lentivirus/genética , Ratones , Mutación , Edición de ARN/genética , Kalinina
2.
Blood ; 121(4): 573-84, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23160470

RESUMEN

Long-living memory stem T cells (T(SCM)) with the ability to self-renew and the plasticity to differentiate into potent effectors could be valuable weapons in adoptive T-cell therapy against cancer. Nonetheless, procedures to specifically target this T-cell population remain elusive. Here, we show that it is possible to differentiate in vitro, expand, and gene modify in clinically compliant conditions CD8(+) T(SCM) lymphocytes starting from naive precursors. Requirements for the generation of this T-cell subset, described as CD62L(+)CCR7(+)CD45RA(+)CD45R0(+)IL-7Rα(+)CD95(+), are CD3/CD28 engagement and culture with IL-7 and IL-15. Accordingly, T(SCM) accumulates early after hematopoietic stem cell transplantation. The gene expression signature and functional phenotype define this population as a distinct memory T-lymphocyte subset, intermediate between naive and central memory cells. When transplanted in immunodeficient mice, gene-modified naive-derived T(SCM) prove superior to other memory lymphocytes for the ability to expand and differentiate into effectors able to mediate a potent xenogeneic GVHD. Furthermore, gene-modified T(SCM) are the only T-cell subset able to expand and mediate GVHD on serial transplantation, suggesting self-renewal capacity in a clinically relevant setting. These findings provide novel insights into the origin and requirements for T(SCM) generation and pave the way for their clinical rapid exploitation in adoptive cell therapy.


Asunto(s)
Memoria Inmunológica , Interleucina-15/metabolismo , Interleucina-7/metabolismo , Células Precursoras de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Interleucina-15/genética , Interleucina-7/genética , Selectina L/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Precursoras de Linfocitos T/citología , Células Precursoras de Linfocitos T/trasplante , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/trasplante , Receptor fas/metabolismo
3.
J Infect Dis ; 208(2): 235-43, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23559464

RESUMEN

BACKGROUND: The genotoxicity of zidovudine has been established in experimental models. The objective of the study was to identify genotoxicity markers in cord blood cells from newborns exposed in utero to antiretroviral (ARV) combinations containing zidovudine. METHODS: Cells were investigated by karyotyping and gene expression analysis of the CD34(+) hematopoietic stem/progenitor cell (HPC) compartment. RESULTS: Karyotyping of the cord blood cells from 15 ARV-exposed newborns and 12 controls revealed a higher proportion of aneuploid cells in the exposed group (median, 18.8% [interquartile range, 10.0%-26.7%] vs 6.6% [interquartile range, 3.1%-11.7%]; P < .001). All chromosomes were involved, with a random distribution of these alterations. Gene expression profiling of CD34(+) HPCs from 7 ARV-exposed and 6 control newborns revealed that >300 genes were significantly upregulated or downregulated by at least 1.5-fold in the exposed group (P < .05 for all comparisons). Significant alterations of genes involved in cell cycle control, mitotic checkpoints, and DNA repair were identified. Although this study does not allow discrimination between the roles of each of the 3 drugs, both cytogenetic and transcriptional findings are similar to those in cellular experiments that used zidovudine alone. CONCLUSIONS: The cord blood cells, including hematopoietic stem cells, from newborns exposed in utero to a zidovudine-based ARV combination present cytogenetic and transcriptional abnormalities compatible with DNA damage.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Sangre Fetal/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Zidovudina/efectos adversos , Adulto , Antígenos CD34/genética , Antígenos CD34/metabolismo , Ciclo Celular/genética , Reparación del ADN/genética , Combinación de Medicamentos , Femenino , Sangre Fetal/citología , Sangre Fetal/fisiología , Perfilación de la Expresión Génica/métodos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Cariotipificación/métodos , Intercambio Materno-Fetal/fisiología , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Efectos Tardíos de la Exposición Prenatal , Células Madre/metabolismo , Transcriptoma/genética , Adulto Joven , Zidovudina/farmacocinética
4.
Dev Biol ; 317(2): 497-507, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18407260

RESUMEN

5' HoxD genes are required for the correct formation of limb skeletal elements. Hoxd13, the most 5'-located HoxD gene, is important for patterning the most distal limb region, and its mutation causes human limb malformation syndromes. The mechanisms underlying the control of developmental processes by Hoxd13, and by Hox genes in general, are still elusive, due to the limited knowledge on their direct downstream target genes. We identified by ChIP-on-chip 248 known gene loci bound invivo by Hoxd13. Genes relevant to limb patterning and skeletogenesis were further analysed. We found that Hoxd13 binds invivo, in developing limbs, the loci of Hand2, a gene crucial to limb AP axis patterning, of Meis1 and Meis2, involved in PD patterning, of the Sfrp1, Barx1, and Fbn1 genes, involved in skeletogenesis, and of the Dach1, Bmp2, Bmp4, andEmx2 genes. We show that Hoxd13 misexpression in developing chick limbs alters the expression of the majority of these genes, supporting the conclusion that Hoxd13 directly regulates their transcription. Our results indicate that 5' Hox proteins regulate directly both key genes for early limb AP and PD axis patterning and genes involved, at later stages, in skeletal patterning.


Asunto(s)
Desarrollo Óseo/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Desarrollo Óseo/fisiología , Línea Celular , Embrión de Pollo , Inmunoprecipitación de Cromatina , Humanos , Ratones , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Mol Cell Biol ; 25(9): 3737-51, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15831478

RESUMEN

In response to DNA damage, p53 activates G(1)/S blocking and apoptotic genes through sequence-specific binding. p53 also represses genes with no target site, such as those for Cdc2 and cyclin B, key regulators of the G(2)/M transition. Like most G(2)/M promoters, they rely on multiple CCAAT boxes activated by NF-Y, whose binding to DNA is temporally regulated during the cell cycle. NF-Y associates with p53 in vitro and in vivo through the alphaC helix of NF-YC (a subunit of NF-Y) and a region close to the tetramerization domain of p53. Chromatin immunoprecipitation experiments indicated that p53 is associated with cyclin B2, CDC25C, and Cdc2 promoters in vivo before and after DNA damage, requiring DNA-bound NF-Y. Following DNA damage, p53 is rapidly acetylated at K320 and K373 to K382, histones are deacetylated, and the release of PCAF and p300 correlates with the recruitment of histone deacetylases (HDACs)-HDAC1 before HDAC4 and HDAC5-and promoter repression. HDAC recruitment requires intact NF-Y binding sites. In transfection assays, PCAF represses cyclin B2, and a nonacetylated p53 mutant shows a complete loss of repression potential, despite its abilities to bind NF-Y and to be recruited on G(2)/M promoters. These data (i) detail a strategy of direct p53 repression through associations with multiple NF-Y trimers that is independent of sequence-specific binding of p53 and that requires C-terminal acetylation, (ii) suggest that p53 is a DNA damage sentinel of the G(2)/M transition, and (iii) delineate a new role for PCAF in cell cycle control.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Daño del ADN/fisiología , Regiones Promotoras Genéticas/fisiología , Proteínas Represoras/fisiología , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Acetiltransferasas/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular , Inmunoprecipitación de Cromatina , Daño del ADN/genética , Regulación hacia Abajo , Fase G2 , Histona Acetiltransferasas , Histona Desacetilasas/metabolismo , Histona Desacetilasas/fisiología , Humanos , Ratones , Mutación/genética , Regiones Promotoras Genéticas/genética , Mapeo de Interacción de Proteínas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Factores de Transcripción p300-CBP
6.
J Vis Exp ; (131)2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29364270

RESUMEN

The Sleeping Beauty (SB) transposon is a non-viral integrating system with proven efficacy for gene transfer and functional genomics. To optimize the SB transposon machinery, a transcriptionally regulated hyperactive transposase (SB100X) and T2-based transposon are employed. Typically, the transposase and transposon are provided transiently by plasmid transfection and SB100X expression is driven by a constitutive promoter. Here, we describe an efficient method to deliver the SB components to human cells that are resistant to several physical and chemical transfection methods, to control SB100X expression and stably integrate a gene of interest (GOI) through a "cut and paste" SB mechanism. The expression of hyperactive transposase is tightly controlled by the Tet-ON system, widely used to control gene expression since 1992. The gene of interest is flanked by inverted repeats (IR) of the T2 transposon. Both SB components are packaged in integration defective lentiviral vectors transiently produced in HEK293T cells. Human cells, either cell lines or primary cells from human tissue, are in vitro transiently transduced with viral vectors. Upon addition of doxycycline (dox, tetracycline analog) into the culture medium, a fine-tuning of transposase expression is measured and results in a long-lasting integration of the gene of interest in the genome of the treated cells. This method is efficient and applicable to the cell line (e.g., HeLa cells) and primary cells (e.g., human primary keratinocytes), and thus represents a valuable tool for genetic engineering and therapeutic gene transfer.


Asunto(s)
Elementos Transponibles de ADN , Vectores Genéticos/genética , Lentivirus/genética , Transposasas/genética , Animales , Células HEK293 , Células HeLa , Humanos , Transfección
7.
J Invest Dermatol ; 137(4): 836-844, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28027893

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by defects in type-VII collagen (C7), a protein encoded by the COL7A1 gene and essential for anchoring fibril formation at the dermal-epidermal junction. Gene therapy of RDEB is based on transplantation of autologous epidermal grafts generated from gene-corrected keratinocytes sustaining C7 deposition at the dermal-epidermal junction. Transfer of the COL7A1 gene is complicated by its very large size and repetitive sequence. This article reports a gene delivery approach based on the Sleeping beauty transposon, which allows integration of a full-length COL7A1 cDNA and secretion of C7 at physiological levels in RDEB keratinocytes without rearrangements or detrimental effects on their clonogenic potential. Skin equivalents derived from gene-corrected RDEB keratinocytes were tested in a validated preclinical model of xenotransplantation on immunodeficient mice, where they showed normal deposition of C7 at the dermal-epidermal junction and restoration of skin adhesion properties. These results indicate the feasibility and efficacy of a transposon-based gene therapy approach to RDEB.


Asunto(s)
Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/terapia , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Queratinocitos/trasplante , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Mutantes , Mutación , Distribución Aleatoria , Reproducibilidad de los Resultados , Medición de Riesgo , Trasplante Heterólogo/métodos , Resultado del Tratamiento
8.
Mol Ther Methods Clin Dev ; 3: 16038, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27574698

RESUMEN

The Sleeping Beauty (SB) transposase and, in particular, its hyperactive variant SB100X raises increasing interest for gene therapy application, including genome modification and, more recently, induced pluripotent stem cells (iPS) reprogramming. The documented cytotoxicity of the transposase, when constitutively expressed by an integrating retroviral vector (iRV), has been circumvented by the transient delivery of SB100X using retroviral mRNA transfer. In this study, we developed an alternative, safe, and efficient transposase delivery system based on a tetracycline-ON regulated expression cassette and the rtTA2(S)-M2 transactivator gene transiently delivered by integration-defective lentiviral vectors (IDLVs). Compared with iRV-mediated delivery, expression of tetracycline-induced SB100X delivered by an IDLV results in more efficient integration of a GFP transposon and reduced toxicity. Tightly regulated expression and reactivation of the transposase was achieved in HeLa cells as wells as in human primary keratinocytes. Based on these properties, the regulated transposase-IDLV vectors may represent a valuable tool for genetic engineering and therapeutic gene transfer.

9.
Mol Ther Nucleic Acids ; 5(11): e389, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27874856

RESUMEN

The bacterial CRISPR/Cas system has proven to be an efficient tool for genetic manipulation in various organisms. Here we show the application of CRISPR-Cas9 technology to edit the human Rhodopsin (RHO) gene in a mouse model for autosomal dominant Retinitis Pigmentosa. We designed single or double sgRNAs to knock-down mutant RHO expression by targeting exon 1 of the RHO gene carrying the P23H dominant mutation. By delivering Cas9 and sgRNAs in a single plasmid we induced an efficient gene editing in vitro, in HeLa cells engineered to constitutively express the P23H mutant RHO allele. Similarly, after subretinal electroporation of the CRISPR/Cas9 plasmid expressing two sgRNAs into P23H RHO transgenic mice, we scored specific gene editing as well as significant reduction of the mutant RHO protein. Successful in vivo application of the CRISPR/Cas9 system confirms its efficacy as a genetic engineering tool in photoreceptor cells.

10.
Sci Rep ; 6: 24724, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27095295

RESUMEN

Genome-wide approaches allow investigating the molecular circuitry wiring the genetic and epigenetic programs of human somatic stem cells. Hematopoietic stem/progenitor cells (HSPC) give rise to the different blood cell types; however, the molecular basis of human hematopoietic lineage commitment is poorly characterized. Here, we define the transcriptional and epigenetic profile of human HSPC and early myeloid and erythroid progenitors by a combination of Cap Analysis of Gene Expression (CAGE), ChIP-seq and Moloney leukemia virus (MLV) integration site mapping. Most promoters and transcripts were shared by HSPC and committed progenitors, while enhancers and super-enhancers consistently changed upon differentiation, indicating that lineage commitment is essentially regulated by enhancer elements. A significant fraction of CAGE promoters differentially expressed upon commitment were novel, harbored a chromatin enhancer signature, and may identify promoters and transcribed enhancers driving cell commitment. MLV-targeted genomic regions co-mapped with cell-specific active enhancers and super-enhancers. Expression analyses, together with an enhancer functional assay, indicate that MLV integration can be used to identify bona fide developmentally regulated enhancers. Overall, this study provides an overview of transcriptional and epigenetic changes associated to HSPC lineage commitment, and a novel signature for regulatory elements involved in cell identity.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Epigénesis Genética , Regulación Viral de la Expresión Génica , Células Madre Hematopoyéticas/citología , Secuencias Reguladoras de Ácidos Nucleicos , Retroviridae/genética , Transcriptoma , Secuencia de Bases , Secuencia de Consenso , Elementos de Facilitación Genéticos , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Humanos , Células Madre Multipotentes/citología , Especificidad de Órganos , Posición Específica de Matrices de Puntuación , Regiones Promotoras Genéticas , Iniciación de la Transcripción Genética
11.
Mol Ther Methods Clin Dev ; 2: 15038, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26509184

RESUMEN

Globoid cell leukodystrophy (GLD) is a demyelinating lysosomal storage disease due to the deficiency of the galactocerebrosidase (GALC) enzyme. The favorable outcome of hematopoietic stem and progenitor cell (HSPC)-based approaches in GLD and other similar diseases suggests HSPC gene therapy as a promising therapeutic option for patients. The path to clinical development of this strategy was hampered by a selective toxicity of the overexpressed GALC in the HSPC compartment. Here, we presented the optimization of a lentiviral vector (LV) in which miR-126 regulation was coupled to codon optimization of the human GALC cDNA to obtain a selective and enhanced enzymatic activity only upon transduced HSPCs differentiation. The safety of human GALC overexpression driven by this LV was extensively demonstrated in vitro and in vivo on human HSPCs from healthy donors. No perturbation in the content of proapoptotic sphingolipids, gene expression profile, and capability of engraftment and mutlilineage differentiation in chimeric mice was observed. The therapeutic potential of this LV was then assessed in a severe GLD murine model that benefited from transplantation of corrected HSPCs with longer survival and ameliorated phenotype as compared to untreated siblings. This construct has thus been selected as a candidate for clinical translation.

12.
Stem Cell Reports ; 2(1): 1-8, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24511464

RESUMEN

We report a long-term follow-up (6.5 years) of a phase I/II clinical trial envisaging the use of autologous genetically modified cultured epidermal stem cells for gene therapy of junctional epidermolysis bullosa, a devastating genetic skin disease. The critical goals of the trial were to evaluate the safety and long-term persistence of genetically modified epidermis. A normal epidermal-dermal junction was restored and the regenerated transgenic epidermis was found to be fully functional and virtually indistinguishable from a normal control. The epidermis was sustained by a discrete number of long-lasting, self-renewing transgenic epidermal stem cells that maintained the memory of the donor site, whereas the vast majority of transduced transit-amplifying progenitors were lost within the first few months after grafting. These data pave the way for the safe use of epidermal stem cells in combined cell and gene therapy for genetic skin diseases.


Asunto(s)
Células Epidérmicas , Epidermólisis Ampollosa de la Unión/terapia , Terapia Genética , Trasplante de Células Madre , Células Madre/citología , Células Cultivadas , Epidermis/metabolismo , Epidermis/patología , Estudios de Seguimiento , Humanos , Integrina alfa6beta4/metabolismo , Queratina-14/metabolismo , Laminina/metabolismo , Precursores de Proteínas/metabolismo , Regeneración , Células Madre/metabolismo
13.
J Clin Invest ; 122(5): 1653-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22523069

RESUMEN

Retroviral vectors integrate in genes and regulatory elements and may cause transcriptional deregulation of gene expression in target cells. Integration into transcribed genes also has the potential to deregulate gene expression at the posttranscriptional level by interfering with splicing and polyadenylation of primary transcripts. To examine the impact of retroviral vector integration on transcript splicing, we transduced primary human cells or cultured cells with HIV-derived vectors carrying a reporter gene or a human ß-globin gene under the control of a reduced-size locus-control region (LCR). Cells were randomly cloned and integration sites were determined in individual clones. We identified aberrantly spliced, chimeric transcripts in more than half of the targeted genes in all cell types. Chimeric transcripts were generated through the use of constitutive and cryptic splice sites in the HIV 5ι long terminal repeat and gag gene as well as in the ß-globin gene and LCR. Compared with constitutively spliced transcripts, most aberrant transcripts accumulated at a low level, at least in part as a consequence of nonsense-mediated mRNA degradation. A limited set of cryptic splice sites caused the majority of aberrant splicing events, providing a strategy for recoding lentiviral vector backbones and transgenes to reduce their potential posttranscriptional genotoxicity.


Asunto(s)
Empalme Alternativo , Genoma Humano , Infecciones por Lentivirus/genética , Lentivirus/fisiología , Isoformas de Proteínas/genética , Integración Viral , Secuencia de Bases , Células Cultivadas , Genes Reporteros , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Interacciones Huésped-Patógeno , Humanos , Intrones , Lentivirus/genética , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Cultivo Primario de Células , Isoformas de Proteínas/metabolismo , Sitios de Empalme de ARN , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transgenes
14.
PLoS One ; 5(12): e15688, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21203516

RESUMEN

The infusion of donor lymphocytes transduced with a retroviral vector expressing the HSV-TK suicide gene in patients undergoing hematopoietic stem cell transplantation for leukemia/lymphoma promotes immune reconstitution and prevents infections and graft-versus-host disease. Analysis of the clonal dynamics of genetically modified lymphocytes in vivo is of crucial importance to understand the potential genotoxic risk of this therapeutic approach. We used linear amplification-mediated PCR and pyrosequencing to build a genome-wide, high-definition map of retroviral integration sites in the genome of peripheral blood T cells from two different donors and used gene expression profiling and bioinformatics to associate integration clusters to transcriptional activity and to genetic and epigenetic features of the T cell genome. Comparison with matched random controls and with integrations obtained from CD34(+) hematopoietic stem/progenitor cells showed that integration clusters occur within chromatin regions bearing epigenetic marks associated with active promoters and regulatory elements in a cell-specific fashion. Analysis of integration sites in T cells obtained ex vivo two months after infusion showed no evidence of integration-related clonal expansion or dominance, but rather loss of cells harboring integration events interfering with RNA post-transcriptional processing. The study shows that high-definition maps of retroviral integration sites are a powerful tool to analyze the fate of genetically modified T cells in patients and the biological consequences of retroviral transduction.


Asunto(s)
Retroviridae/genética , Linfocitos T/citología , Antígenos CD34/biosíntesis , Mapeo Cromosómico/métodos , Análisis por Conglomerados , Biología Computacional/métodos , Epigénesis Genética , Perfilación de la Expresión Génica , Enfermedad Injerto contra Huésped/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Humanos , Depleción Linfocítica , Reacción en Cadena de la Polimerasa/métodos , Análisis de Secuencia de ADN
15.
J Biol Chem ; 278(2): 1336-45, 2003 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-12401788

RESUMEN

The heterotrimeric transcription factor NF-Y recognizes with high specificity and affinity the CCAAT regulatory element that is widely represented in promoters and enhancer regions. The CCAAT box acts in concert with neighboring elements, and its bending by NF-Y is thought to be a major mechanism required for transcription activation. We have solved the structure of the NF-YC/NF-YB subcomplex of NF-Y, which shows that the core domains of both proteins interact through histone fold motifs. This histone-like pair is closely related to the H2A/H2B and NC2alpha/NC2beta families, with features that are both common to this class of proteins and unique to NF-Y. The structure together with the modeling of the nonspecific interaction of NF-YC/NF-YB with DNA and the full NF-Y/CCAAT box complex highlight important structural features that account for different and possibly similar biological functions of the transcriptional regulators NF-Y and NC2. In particular, it emphasizes the role of the newly described alphaC helix of NF-YC, which is both important for NF-Y trimerization and a target for regulatory proteins, such as MYC and p53.


Asunto(s)
Factor de Unión a CCAAT/química , Factor de Unión a CCAAT/fisiología , Proteínas de Unión al ADN/química , ADN/metabolismo , Factores de Transcripción/química , Transcripción Genética , Secuencia de Aminoácidos , Secuencia de Bases , Cristalización , Dimerización , Datos de Secuencia Molecular , Estructura Secundaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA