Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 152(5): 1021-36, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452851

RESUMEN

Histone modifications regulate chromatin-dependent processes, yet the mechanisms by which they contribute to specific outcomes remain unclear. H3K4me3 is a prominent histone mark that is associated with active genes and promotes transcription through interactions with effector proteins that include initiation factor TFIID. We demonstrate that H3K4me3-TAF3 interactions direct global TFIID recruitment to active genes, some of which are p53 targets. Further analyses show that (1) H3K4me3 enhances p53-dependent transcription by stimulating preinitiation complex (PIC) formation; (2) H3K4me3, through TAF3 interactions, can act either independently or cooperatively with the TATA box to direct PIC formation and transcription; and (3) H3K4me3-TAF3/TFIID interactions regulate gene-selective functions of p53 in response to genotoxic stress. Our findings indicate a mechanism by which H3K4me3 directs PIC assembly for the rapid induction of specific p53 target genes.


Asunto(s)
Código de Histonas , Histonas/metabolismo , Factor de Transcripción TFIID/metabolismo , Iniciación de la Transcripción Genética , Línea Celular Tumoral , Humanos , Lisina/metabolismo , Metilación , TATA Box , Factores Asociados con la Proteína de Unión a TATA , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Factores de Transcripción p300-CBP/metabolismo
2.
Nature ; 559(7714): 405-409, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29995861

RESUMEN

Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. The need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells using homology-directed repair5,6. Here we developed a CRISPR-Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.


Asunto(s)
Reprogramación Celular/genética , Edición Génica , Genoma Humano/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Autoinmunidad/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Masculino , Ratones , Trasplante de Neoplasias , Ingeniería de Proteínas , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/citología
3.
Genes Dev ; 29(21): 2287-97, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26545813

RESUMEN

The host chromatin-binding factor LEDGF/p75 interacts with HIV-1 integrase and directs integration to active transcription units. To understand how LEDGF/p75 recognizes transcription units, we sequenced 1 million HIV-1 integration sites isolated from cultured HEK293T cells. Analysis of integration sites showed that cancer genes were preferentially targeted, raising concerns about using lentivirus vectors for gene therapy. Additional analysis led to the discovery that introns and alternative splicing contributed significantly to integration site selection. These correlations were independent of transcription levels, size of transcription units, and length of the introns. Multivariate analysis with five parameters previously found to predict integration sites showed that intron density is the strongest predictor of integration density in transcription units. Analysis of previously published HIV-1 integration site data showed that integration density in transcription units in mouse embryonic fibroblasts also correlated strongly with intron number, and this correlation was absent in cells lacking LEDGF. Affinity purification showed that LEDGF/p75 is associated with a number of splicing factors, and RNA sequencing (RNA-seq) analysis of HEK293T cells lacking LEDGF/p75 or the LEDGF/p75 integrase-binding domain (IBD) showed that LEDGF/p75 contributes to splicing patterns in half of the transcription units that have alternative isoforms. Thus, LEDGF/p75 interacts with splicing factors, contributes to exon choice, and directs HIV-1 integration to transcription units that are highly spliced.


Asunto(s)
VIH-1/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Integración Viral/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Intrones/genética , Unión Proteica , Estructura Terciaria de Proteína , Empalme del ARN
4.
Antimicrob Agents Chemother ; 66(1): e0150021, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34723625

RESUMEN

Resistance to anti-HIV drugs has been a problem from the beginning of antiviral drug treatments. The recent expansion of combination antiretroviral therapy worldwide has led to an increase in resistance to antiretrovirals; understanding the mechanisms of resistance is increasingly important. In this study, we analyzed reverse transcriptase (RT) variants based on sequences derived from an individual who had low-level rebound viremia while undergoing therapy with abacavir, azidothymidine (AZT) (zidovudine), and (-)-l-2',3'-dideoxy-3'-thiacytidine (3TC) (lamivudine). The RT had mutations at positions 64, 67, 70, 184, and 219 and a threonine insertion after amino acid 69 in RT. The virus remained partially susceptible to the nucleoside RT inhibitor (NRTI) regimen. We show how these mutations affect the ability of NRTIs to inhibit DNA synthesis by RT. The presence of the inserted threonine reduced the susceptibility of the RT mutant to inhibition by tenofovir.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , VIH-1 , Aminoácidos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Infecciones por VIH/tratamiento farmacológico , Transcriptasa Inversa del VIH/metabolismo , VIH-1/genética , VIH-1/metabolismo , Humanos , Lamivudine/farmacología , Mutación/genética , Inhibidores de la Transcriptasa Inversa/química , Zidovudina/farmacología
5.
PLoS Pathog ; 15(7): e1007869, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31291371

RESUMEN

Clonal expansion of HIV infected cells plays an important role in the formation and persistence of the reservoir that allows the virus to persist, in DNA form, despite effective antiretroviral therapy. We used integration site analysis to ask if there is a similar clonal expansion of SIV infected cells in macaques. We show that the distribution of HIV and SIV integration sites in vitro is similar and that both viruses preferentially integrate in many of the same genes. We obtained approximately 8000 integration sites from blood samples taken from SIV-infected macaques prior to the initiation of ART, and from blood, spleen, and lymph node samples taken at necropsy. Seven clones were identified in the pre-ART samples; one persisted for a year on ART. An additional 100 clones were found only in on-ART samples; a number of these clones were found in more than one tissue. The timing and extent of clonal expansion of SIV-infected cells in macaques and HIV-infected cells in humans is quite similar. This suggests that SIV-infected macaques represent a useful model of the clonal expansion of HIV infected cells in humans that can be used to evaluate strategies intended to control or eradicate the viral reservoir.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Animales , Antirretrovirales/uso terapéutico , Linfocitos T CD4-Positivos/virología , Reservorios de Enfermedades/virología , Infecciones por VIH/patología , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Técnicas In Vitro , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Carga Viral/efectos de los fármacos , Integración Viral/genética , Integración Viral/fisiología , Replicación Viral/efectos de los fármacos
6.
J Virol ; 88(13): 7589-601, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24760888

RESUMEN

UNLABELLED: The genetic variation in HIV-1 in patients is due to the high rate of viral replication, the high viral load, and the errors made during viral replication. Some of the mutations in reverse transcriptase (RT) that alter the deoxynucleoside triphosphate (dNTP)-binding pocket, including those that confer resistance to nucleoside/nucleotide analogs, affect dNTP selection during replication. The effects of mutations in RT on the spectrum (nature, position, and frequency) of errors made in vivo are poorly understood. We previously determined the mutation rate and the frequency of different types of mutations and identified hot spots for mutations in a lacZα (the α complementing region of lacZ) reporter gene carried by an HIV-1 vector that replicates using wild-type RT. We show here that four mutations (Y115F, M184V, M184I, and Q151M) in the dNTP-binding pocket of RT that had relatively small effects on the overall HIV-1 mutation rate (less than 3-fold compared to the wild type) significantly increased mutations at some specific positions in the lacZα reporter gene. We also show that changes in a sequence that flanks the reporter gene can affect the mutations that arise in the reporter. These data show that changes either in HIV-1 RT or in the sequence of the nucleic acid template can affect the spectrum of mutations made during viral replication. This could, by implication, affect the generation of drug-resistant mutants and immunological-escape mutants in patients. IMPORTANCE: RT is the viral enzyme that converts the RNA genome of HIV into DNA. Errors made during replication allow the virus to escape from the host's immune system and to develop resistance to the available anti-HIV drugs. We show that four different mutations in RT which are known to be associated with resistance to anti-RT drugs modestly increased the overall frequency of errors made during viral replication. However, the increased errors were not uniformly distributed; the additional errors occurred at a small number of positions (hot spots). Moreover, some of the RT mutations preferentially affected the nature of the errors that were made (some RT mutations caused an increase in insertion and deletion errors; others caused an increase in substitution errors). We also show that sequence changes in a region adjacent to a target gene can affect the errors made within the target gene.


Asunto(s)
Farmacorresistencia Viral/genética , Infecciones por VIH/virología , Transcriptasa Inversa del VIH/genética , VIH-1/fisiología , Operón Lac/genética , Mutación/genética , Replicación Viral/genética , Células Cultivadas , ADN Viral/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Humanos , Inhibidores de la Transcriptasa Inversa/farmacología
7.
J Virol ; 88(8): 4504-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24501411

RESUMEN

UNLABELLED: Retroviral vectors have been used in successful gene therapies. However, in some patients, insertional mutagenesis led to leukemia or myelodysplasia. Both the strong promoter/enhancer elements in the long terminal repeats (LTRs) of murine leukemia virus (MLV)-based vectors and the vector-specific integration site preferences played an important role in these adverse clinical events. MLV integration is known to prefer regions in or near transcription start sites (TSS). Recently, BET family proteins were shown to be the major cellular proteins responsible for targeting MLV integration. Although MLV integration sites are significantly enriched at TSS, only a small fraction of the MLV integration sites (<15%) occur in this region. To resolve this apparent discrepancy, we created a high-resolution genome-wide integration map of more than one million integration sites from CD34(+) hematopoietic stem cells transduced with a clinically relevant MLV-based vector. The integration sites form ∼60,000 tight clusters. These clusters comprise ∼1.9% of the genome. The vast majority (87%) of the integration sites are located within histone H3K4me1 islands, a hallmark of enhancers. The majority of these clusters also have H3K27ac histone modifications, which mark active enhancers. The enhancers of some oncogenes, including LMO2, are highly preferred targets for integration without in vivo selection. IMPORTANCE: We show that active enhancer regions are the major targets for MLV integration; this means that MLV preferentially integrates in regions that are favorable for viral gene expression in a variety of cell types. The results provide insights for MLV integration target site selection and also explain the high risk of insertional mutagenesis that is associated with gene therapy trials using MLV vectors.


Asunto(s)
Elementos de Facilitación Genéticos , Vectores Genéticos/fisiología , Virus de la Leucemia Murina/fisiología , Integración Viral , Animales , Células Cultivadas , Terapia Genética , Vectores Genéticos/genética , Genoma Humano , Células Madre Hematopoyéticas/virología , Histonas/genética , Histonas/metabolismo , Humanos , Virus de la Leucemia Murina/genética , Ratones , Mutagénesis Insercional
8.
J Virol ; 87(1): 648-58, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097450

RESUMEN

Retroviruses integrate into cellular DNA nonrandomly. Lentiviruses such as human immunodeficiency virus type 1 (HIV-1) favor the bodies of active genes and gene-enriched transcriptionally active regions of chromosomes. The interaction between lentiviral integrase and the cellular protein lens epithelium-derived growth factor (LEDGF)/p75 underlies the targeting of gene bodies, whereas recent research has highlighted roles for the HIV-1 capsid (CA) protein and cellular factors implicated in viral nuclear import, including transportin 3 (TNPO3) and nucleoporin 358 (NUP358), in the targeting of gene-dense regions of chromosomes. Here, we show that CA mutations, which include the substitution of Asp for Asn74 (N74D), significantly reduce the dependency of HIV-1 on LEDGF/p75 during infection and that this difference correlates with the efficiency of viral DNA integration. The distribution of integration sites mapped by Illumina sequencing confirms that the N74D mutation reduces integration into gene-rich regions of chromosomes and gene bodies and reveals previously unrecognized roles for NUP153 (another HIV-1 cofactor implicated in viral nuclear import) and LEDGF/p75 in the targeting of the viral preintegration complex to gene-dense regions of chromatin. A role for the CA protein in determining the dependency of HIV-1 on LEDGF/p75 during infection highlights a connection between the viral capsid and chromosomal DNA integration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Cápside/metabolismo , ADN Viral/metabolismo , VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Proteínas de Complejo Poro Nuclear/metabolismo , Factores de Transcripción/metabolismo , Integración Viral , Animales , Proteínas de la Cápside/genética , Línea Celular , Humanos , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense
9.
Nucleic Acids Res ; 40(22): 11518-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23042676

RESUMEN

The binding of integrase (IN) to lens epithelium-derived growth factor (LEDGF)/p75 in large part determines the efficiency and specificity of HIV-1 integration. However, a significant residual preference for integration into active genes persists in Psip1 (the gene that encodes for LEDGF/p75) knockout (KO) cells. One other cellular protein, HRP2, harbors both the PWWP and IN-binding domains that are important for LEDGF/p75 co-factor function. To assess the role of HRP2 in HIV-1 integration, cells generated from Hdgfrp2 (the gene that encodes for HRP2) and Psip1/Hdgfrp2 KO mice were infected alongside matched control cells. HRP2 depleted cells supported normal infection, while disruption of Hdgfrp2 in Psip1 KO cells yielded additional defects in the efficiency and specificity of integration. These deficits were largely restored by ectopic expression of either LEDGF/p75 or HRP2. The double-KO cells nevertheless supported residual integration into genes, indicating that IN and/or other host factors contribute to integration specificity in the absence of LEDGF/p75 and HRP2. Psip1 KO significantly increased the potency of an allosteric inhibitor that binds the LEDGF/p75 binding site on IN, a result that was not significantly altered by Hdgfrp2 disruption. These findings help to rule out the host factor-IN interactions as the primary antiviral targets of LEDGF/p75-binding site IN inhibitors.


Asunto(s)
Inhibidores de Integrasa VIH/farmacología , VIH-1/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Integración Viral , Acetatos/farmacología , Animales , Sitios de Unión , Línea Celular , Fibroblastos/metabolismo , Fibroblastos/virología , VIH-1/efectos de los fármacos , VIH-1/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Noqueados , Quinolinas/farmacología
10.
Proc Natl Acad Sci U S A ; 107(7): 3135-40, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133638

RESUMEN

Lens epithelium-derived growth factor (LEDGF) fusion proteins can direct HIV-1 DNA integration to novel sites in the host genome. The C terminus of LEDGF contains an integrase binding domain (IBD), and the N terminus binds chromatin. LEDGF normally directs integrations to the bodies of expressed genes. Replacing the N terminus of LEDGF with chromatin binding domains (CBDs) from other proteins changes the specificity of HIV-1 DNA integration. We chose two well-characterized CBDs: the plant homeodomain (PHD) finger from ING2 and the chromodomain from heterochromatin binding protein 1alpha (HP1alpha). The ING2 PHD finger binds H3K4me3, a histone mark that is associated with the transcriptional start sites of expressed genes. The HP1alpha chromodomain binds H3K9me2,3, histone marks that are widely distributed throughout the genome. A fusion protein in which the ING2 PHD finger was linked to the LEDGF IBD directed integrations near the start sites of expressed genes. A similar fusion protein in which the HP1alpha chromodomain was linked to the LEDGF IBD directed integrations to sites that differed from both the PHD finger fusion-directed and LEDGF-directed integration sites. The ability to redirect HIV-1 DNA integration may help solve the problems associated with the activation of oncogenes when retroviruses are used in gene therapy.


Asunto(s)
ADN Viral/metabolismo , VIH-1 , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Integración Viral/fisiología , Animales , Sitios de Unión/genética , Línea Celular , Cromatina/metabolismo , Biología Computacional , ADN Viral/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Terapia Genética/métodos , Integrasa de VIH/metabolismo , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Análisis de Secuencia de ADN , Proteínas Supresoras de Tumor/genética
11.
J Virol ; 84(19): 9864-78, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660205

RESUMEN

There is considerable HIV-1 variation in patients. The extent of the variation is due to the high rate of viral replication, the high viral load, and the errors made during viral replication. Mutations can arise from errors made either by host DNA-dependent RNA polymerase II or by HIV-1 reverse transcriptase (RT), but the relative contributions of these two enzymes to the mutation rate are unknown. In addition, mutations in RT can affect its fidelity, but the effect of mutations in RT on the nature of the mutations that arise in vivo is poorly understood. We have developed an efficient system, based on existing technology, to analyze the mutations that arise in an HIV-1 vector in a single cycle of replication. A lacZalpha reporter gene is used to identify viral DNAs that contain mutations which are analyzed by DNA sequencing. The forward mutation rate in this system is 1.4 x 10(-5) mutations/bp/cycle, equivalent to the retroviral average. This rate is about 3-fold lower than previously reported for HIV-1 in vivo and is much lower than what has been reported for purified HIV-1 RT in vitro. Although the mutation rate was not affected by the orientation of lacZalpha, the sites favored for mutations (hot spots) in lacZalpha depended on which strand of lacZalpha was present in the viral RNA. The pattern of hot spots seen in lacZalpha in vivo did not match any of the published data obtained when purified RT was used to copy lacZalpha in vitro.


Asunto(s)
VIH-1/genética , VIH-1/fisiología , Mutación , Replicación Viral/genética , Secuencia de Bases , Línea Celular , ADN Viral/genética , Mutación del Sistema de Lectura , Genes Reporteros , Genes Virales , Vectores Genéticos , Transcriptasa Inversa del VIH/genética , Humanos , Mutación INDEL , Operón Lac , Datos de Secuencia Molecular , Mutagénesis Insercional , Plásmidos/genética , Mutación Puntual , Eliminación de Secuencia
12.
Cell Res ; 25(3): 288-305, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25675981

RESUMEN

Complexes containing INTS3 and either NABP1 or NABP2 were initially characterized in DNA damage responses, but their biochemical function remained unknown. Using affinity purifications and HIV Integration targeting-sequencing (HIT-Seq), we find that these complexes are part of the Integrator complex, which binds RNA Polymerase II and regulates specific target genes. Integrator cleaves snRNAs as part of their processing to their mature form in a mechanism that is intimately coupled with transcription termination. However, HIT-Seq reveals that Integrator also binds to the 3' end of replication-dependent histones and promoter proximal regions of genes with polyadenylated transcripts. Depletion of Integrator subunits results in transcription termination failure, disruption of histone mRNA processing, and polyadenylation of snRNAs and histone mRNAs. Furthermore, promoter proximal binding of Integrator negatively regulates expression of genes whose transcripts are normally polyadenylated. Integrator recruitment to all three gene classes is DSIF-dependent, suggesting that Integrator functions as a termination complex at DSIF-dependent RNA Polymerase II pause sites.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Mitocondriales/metabolismo , ARN Polimerasa II/metabolismo , Terminación de la Transcripción Genética/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Histonas/biosíntesis , Histonas/genética , Humanos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Nucleares/genética , Poliadenilación/genética , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/genética , Procesamiento Postranscripcional del ARN , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Elongación Transcripcional/genética
13.
J Virol ; 79(16): 10258-67, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16051819

RESUMEN

Previous attempts to extend the host range of the avian sarcoma/leukosis virus (ASLV)-based RCASBP vectors produced two viral vectors, RCASBP M2C (4070A) and RCASBP M2C (797-8), which replicate using the amphotropic murine leukemia virus 4070A Env protein (2). Both viruses were adapted to replicate efficiently in the avian cell line DF-1, but RCASBP M2C (4070A) caused extensive cytopathic effects (CPE) in DF-1 cells whereas RCASBP M2C (797-8) induced low levels of CPE. The two viruses differed only at amino acid 242 of the polyproline-rich region in the surface (SU) subunit of the Env protein. In RCASBP M2C (4070A), an isoleucine replaced the wild-type proline residue, whereas a threonine residue was found in RCASBP M2C (797-8). In the present study, we show that other amino acid substitutions at position 242 strongly influence the CPE and replication rate of the chimeric viruses. There was a correlation between the amount of unintegrated linear retroviral DNA present in infected DF-1 cells and the level of CPE. This suggests that there may be a role for superinfection in the CPE. The treatment of RCASBP M2C (4070A)-infected cells with dantrolene, which inhibits the release of calcium from the endoplasmic reticulum (ER), reduced the amount of CPE seen during infection with the highly cytotoxic virus. Dantrolene treatment did not appear to affect virus production, suggesting that Ca2+ release from the ER had a role in the CPE caused by these viruses.


Asunto(s)
Efecto Citopatogénico Viral , Productos del Gen env/química , Orthoreovirus Aviar/fisiología , Replicación Viral , Animales , Calcio/metabolismo , Línea Celular , Embrión de Pollo , ADN Viral/análisis , Dantroleno/farmacología , Retículo Endoplásmico/metabolismo , Productos del Gen env/fisiología , Vectores Genéticos , Orthoreovirus Aviar/patogenicidad , Péptidos
14.
J Virol ; 76(4): 1762-8, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11799171

RESUMEN

We constructed two versions of an RCASBP-based retroviral shuttle vector, RSVP (RCASBP shuttle vector plasmid), containing either the zeocin or blasticidin resistance gene. In this vector, the drug resistance gene is expressed in avian cells from the long terminal repeat (LTR) promoter, whereas in bacteria the resistance gene is expressed from a bacterial promoter. The vector contains a bacterial origin of replication (ColE1) to allow circular viral DNA to replicate as a plasmid in bacteria. The vector also contains the lac operator sequence, which binds to the lac repressor protein, providing a simple and rapid way to purify the vector DNA. The RSVP plasmid contains the following sequence starting with the 5" end: LTR, gag, pol, env, drug resistance gene, lac operator, ColE1, LTR. After this plasmid was transfected into DF-1 cells, we were able to rescue the circularized unintegrated viral DNA from RSVP simply by transforming the Hirt DNA into Escherichia coli. Furthermore, we were able to rescue the integrated provirus. DNA from infected cells was digested with an appropriate restriction enzyme (ClaI) and the vector-containing segments were enriched using lac repressor protein and then self-ligated. These enriched fractions were used to transform E. coli. The transformation was successful and we did recover integration sites, but higher-efficiency rescue was obtained with electroporation. The vector is relatively stable upon passage in avian cells. Southern blot analyses of genomic DNAs derived from successive viral passages under nonselective conditions showed that the cassette (drug resistance gene-lac operator-ColE1) insert was present in the vector up to the third viral passage for both resistance genes, which suggests that the RSVP vectors are stable for approximately three viral passages. Together, these results showed that RSVP vectors are useful tools for cloning unintegrated or integrated viral DNAs.


Asunto(s)
Vectores Genéticos , Plásmidos/genética , Retroviridae/genética , Replicación Viral , Animales , Secuencia de Bases , Células Cultivadas , Embrión de Pollo , ADN Viral/genética , Escherichia coli/genética , Retroviridae/fisiología , Análisis de Secuencia de ADN , Transfección , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA