Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 8161, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289368

RESUMEN

Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on co-infection of cells by wild-type and engineered viruses. Here, we report on a viral gene drive against human herpes simplex virus 1 (HSV-1) and show that it propagates efficiently in cell culture and during HSV-1 infection in mice. We describe high levels of co-infection and gene drive-mediated recombination in neuronal tissues during herpes encephalitis as the infection progresses from the site of inoculation to the peripheral and central nervous systems. In addition, we show evidence that a superinfecting gene drive virus could recombine with wild-type viruses during latent infection. These findings indicate that HSV-1 achieves high rates of co-infection and recombination during viral infection, a phenomenon that is currently underappreciated. Overall, this study shows that a viral gene drive could spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Animales , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Ratones , Herpes Simple/virología , Herpes Simple/genética , Humanos , Coinfección/virología , Tecnología de Genética Dirigida/métodos , Femenino , Células Vero , Chlorocebus aethiops , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/virología , Ratones Endogámicos C57BL , Recombinación Genética/genética , Genes Virales/genética
2.
Nat Commun ; 15(1): 4018, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740820

RESUMEN

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Asunto(s)
Dependovirus , Edición Génica , Herpes Simple , Herpesvirus Humano 1 , Carga Viral , Esparcimiento de Virus , Animales , Edición Génica/métodos , Femenino , Dependovirus/genética , Ratones , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Herpes Simple/genética , Herpes Simple/virología , Herpes Simple/terapia , Modelos Animales de Enfermedad , Latencia del Virus/genética , Humanos , Vectores Genéticos/genética , Células Vero , Terapia Genética/métodos , Herpes Genital/terapia , Herpes Genital/virología , ADN Viral/genética
4.
Gene Ther ; 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165032

RESUMEN

Over 15 years after hepatotoxicity was first observed following administration of an adeno-associated virus (AAV) vector during a hemophilia B clinical trial, recent reports of treatment-associated neurotoxicity in animals and humans have brought the potential impact of AAV-associated toxicity back to prominence. In both pre-clinical studies and clinical trials, systemic AAV administration has been associated with neurotoxicity in peripheral nerve ganglia and spinal cord. Neurological signs have also been seen following direct AAV injection into the brain, both in non-human primates and in a clinical trial for late infantile Batten disease. Neurotoxic events appear variable across species, and preclinical animal studies do not fully predict clinical observations. Accumulating data suggest that AAV-associated neurotoxicity may be underdiagnosed and may differ between species in terms of frequency and/or severity. In this review, we discuss the different animal models that have been used to demonstrate AAV-associated neurotoxicity, its potential causes and consequences, and potential approaches to blunt AAV-associated neurotoxicity.

5.
Microbiol Spectr ; 11(3): e0517622, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37199630

RESUMEN

Hepatitis B virus (HBV) is a pathogen of major public health importance that is largely incurable once a chronic infection is established. Only humans and great apes are fully permissive to HBV infection, and this species restriction has impacted HBV research by limiting the utility of small animal models. To combat HBV species restrictions and enable more in vivo studies, liver-humanized mouse models have been developed that are permissive to HBV infection and replication. Unfortunately, these models can be difficult to establish and are expensive commercially, which has limited their academic use. As an alternative mouse model to study HBV, we evaluated liver-humanized NSG-PiZ mice and showed that they are fully permissive to HBV. HBV selectively replicates in human hepatocytes within chimeric livers, and HBV-positive (HBV+) mice secrete infectious virions and hepatitis B surface antigen (HBsAg) into blood while also harboring covalently closed circular DNA (cccDNA). HBV+ mice develop chronic infections lasting at least 169 days, which should enable the study of new curative therapies targeting chronic HBV, and respond to entecavir therapy. Furthermore, HBV+ human hepatocytes in NSG-PiZ mice can be transduced by AAV3b and AAV.LK03 vectors, which should enable the study of gene therapies that target HBV. In summary, our data demonstrate that liver-humanized NSG-PiZ mice can be used as a robust and cost-effective alternative to existing chronic hepatitis B (CHB) models and may enable more academic research labs to study HBV disease pathogenesis and antiviral therapy. IMPORTANCE Liver-humanized mouse models have become the gold standard for the in vivo study of hepatitis B virus (HBV), yet their complexity and cost have prohibited widespread use of existing models in research. Here, we show that the NSG-PiZ liver-humanized mouse model, which is relatively inexpensive and simple to establish, can support chronic HBV infection. Infected mice are fully permissive to hepatitis B, supporting both active replication and spread, and can be used to study novel antiviral therapies. This model is a viable and cost-effective alternative to other liver-humanized mouse models that are used to study HBV.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Ratones , Humanos , Animales , Hepatitis B Crónica/tratamiento farmacológico , Virus de la Hepatitis B/genética , Hepatitis B/tratamiento farmacológico , Antígenos de Superficie de la Hepatitis B , Antivirales/uso terapéutico , ADN Circular/uso terapéutico , ADN Viral/genética
6.
Hepatol Commun ; 7(2): e0009, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074875

RESUMEN

BACKGROUND AND AIMS: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. APPROACH AND RESULTS: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. CONCLUSIONS: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


Asunto(s)
Cápside , Dependovirus , Animales , Ratones , Femenino , Masculino , Humanos , Cápside/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Distribución Tisular , Macaca mulatta/genética , Macaca mulatta/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Terapia Genética/métodos
7.
Mol Ther Methods Clin Dev ; 20: 258-275, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33473359

RESUMEN

Chronic hepatitis B virus (HBV) infection is a major public health problem. New treatment approaches are needed because current treatments do not target covalently closed circular DNA (cccDNA), the template for HBV replication, and rarely clear the virus. We harnessed adeno-associated virus (AAV) vectors and CRISPR-Staphylococcus aureus (Sa)Cas9 to edit the HBV genome in liver-humanized FRG mice chronically infected with HBV and receiving entecavir. Gene editing was detected in livers of five of eight HBV-specific AAV-SaCas9-treated mice, but not control mice, and mice with detectable HBV gene editing showed higher levels of SaCas9 delivery to HBV+ human hepatocytes than those without gene editing. HBV-specific AAV-SaCas9 therapy significantly improved survival of human hepatocytes, showed a trend toward decreasing total liver HBV DNA and cccDNA, and was well tolerated. This work provides evidence for the feasibility and safety of in vivo gene editing for chronic HBV infections, and it suggests that with further optimization, this approach may offer a plausible way to treat or even cure chronic HBV infections.

8.
Hum Gene Ther ; 32(1-2): 96-112, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32998579

RESUMEN

Adeno-associated virus (AAV) vectors such as AAV6, which shows tropism for primary human CD4+ T cells in vitro, are being explored for delivery of anti-HIV therapeutic modalities in vivo. However, pre-existing immunity and sequestration in nontarget organs can significantly hinder their performance. To overcome these challenges, we investigated whether immunosuppression would allow gene delivery by AAV6 or targeted AAV6 derivatives in seropositive rhesus macaques. Animals were immune suppressed with rapamycin before intravenous (IV) or subcutaneous (SC) delivery of AAV, and we monitored vector biodistribution, gene transfer, and safety. Macaques received phosphate-buffered saline, AAV6 alone, or an equal dose of AAV6 and an AAV6-55.2 vector retargeted to CD4 through a direct ankyrin repeat protein (DARPin). AAV6 and AAV6-55.2 vector genomes were found in peripheral blood mononuclear cells and most organs up to 28 days postadministration, with the highest levels seen in liver, spleen, lymph nodes (LNs), and muscle, suggesting that retargeting did not prevent vector sequestration. Despite vector genome detection, gene expression from AAV6-55.2 was not detected in any tissue. SC injection of AAV6 facilitated efficient gene expression in muscle adjacent to the injection site, plus low-level gene expression in spleen, LNs, and liver, whereas gene expression following IV injection of AAV6 was predominantly seen in the spleen. AAV vectors were well tolerated, although elevated liver enzymes were detected in three of four AAV-treated animals 14 days after rapamycin withdrawal. One SC-injected animal had muscle inflammation proximal to the injection site, plus detectable T cell responses against transgene and AAV6 capsid at study finish. Overall, our data suggest that rapamycin treatment may offer a possible strategy to express anti-HIV therapeutics such as broadly neutralizing antibodies from muscle. This study provides important safety and efficacy data that will aid study design for future anti-HIV gene therapies.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Proteínas de Repetición de Anquirina Diseñadas , Vectores Genéticos/genética , Humanos , Leucocitos Mononucleares , Macaca mulatta , Sirolimus/uso terapéutico , Distribución Tisular
9.
Nat Commun ; 11(1): 4148, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811834

RESUMEN

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.


Asunto(s)
Desoxirribonucleasas/genética , Dependovirus/genética , Infecciones del Ojo/terapia , Edición Génica/métodos , Herpes Simple/terapia , Herpesvirus Humano 1/genética , Latencia del Virus/genética , Animales , Sistemas CRISPR-Cas/genética , Células Cultivadas , Chlorocebus aethiops , Infecciones del Ojo/genética , Infecciones del Ojo/virología , Femenino , Células HEK293 , Herpes Simple/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Ratones , Neuronas/metabolismo , Neuronas/virología , RNA-Seq , Análisis de la Célula Individual , Ganglio Cervical Superior/metabolismo , Ganglio Cervical Superior/virología , Células Vero
10.
ACS Infect Dis ; 4(6): 871-880, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29522311

RESUMEN

Chronic viral infections remain a major public health issue affecting millions of people worldwide. Highly active antiviral treatments have significantly improved prognosis and infection-related morbidity and mortality but have failed to eliminate persistent viral forms. Therefore, new strategies to either eradicate or control these viral reservoirs are paramount to allow patients to stop antiretroviral therapy and realize a cure. Viral genome disruption based on gene editing by programmable endonucleases is one promising curative gene therapy approach. Recent findings on RNA-guided human immunodeficiency virus 1 (HIV-1) genome cleavage by Cas9 and other gene-editing enzymes in latently infected cells have shown high levels of site-specific genome disruption and potent inhibition of virus replication. However, HIV-1 can readily develop resistance to genome editing at a single antiviral target site. Current data suggest that cellular repair associated with DNA double-strand breaks can accelerate the emergence of resistance. On the other hand, a combination antiviral target strategy can exploit the same repair mechanism to functionally cure HIV-1 infection in vitro while avoiding the development of resistance. This perspective summarizes recent findings on the biology of resistance to genome editing and discusses the significance of viral genetic diversity on the application of gene editing strategies toward cure.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Genoma Viral , Virosis/terapia , Virosis/virología , Virus/genética , Animales , Portador Sano , Terapia Combinada , Farmacorresistencia Viral , Terapia Genética , Variación Genética , Humanos , Cuasiespecies/genética , ARN Guía de Kinetoplastida , ARN Viral , Replicación Viral/genética , Virus/efectos de los fármacos
11.
Sci Rep ; 7(1): 927, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28424485

RESUMEN

The ability to genetically manipulate trigeminal ganglion (TG) neurons would be useful in the study of the craniofacial nervous system and latent alphaherpesvirus infections. We investigated adeno-associated virus (AAV) vectors for gene delivery to the TG after intradermal whiskerpad delivery in mice. We demonstrated that AAV vectors of serotypes 1, 7, 8, and 9 trafficked from the whiskerpad into TG neurons and expressed transgenes within cell bodies and axons of sensory neurons in all three branches of the TG. Gene expression was highest with AAV1, and steadily increased over time up to day 28. Both constitutive and neuronal-specific promoters were able to drive transgene expression in TG neurons. Levels of vector genomes in the TG increased with input dose, and multiple transgenes could be co-delivered to TG neurons by separate AAV vectors. In conclusion, AAV1 vectors are suitable for gene delivery to TG sensory neurons following intradermal whiskerpad injection.


Asunto(s)
Dependovirus/genética , Células Receptoras Sensoriales/virología , Transgenes , Ganglio del Trigémino/virología , Animales , Células Cultivadas , Chlorocebus aethiops , Dependovirus/inmunología , Terapia Genética , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Inyecciones Intradérmicas , Ratones , Modelos Animales , Serogrupo , Transducción Genética , Células Vero
12.
JCI Insight ; 1(14)2016 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-27642635

RESUMEN

A large portion of the global population carries latent herpes simplex virus (HSV), which can periodically reactivate, resulting in asymptomatic shedding or formation of ulcerative lesions. Current anti-HSV drugs do not eliminate latent virus from sensory neurons where HSV resides, and therefore do not eliminate the risk of transmission or recurrent disease. Here, we report the ability of HSV-specific endonucleases to induce mutations of essential HSV genes both in cultured neurons and in latently infected mice. In neurons, viral genomes are susceptible to endonuclease-mediated mutagenesis, regardless of the time of treatment after HSV infection, suggesting that both HSV lytic and latent forms can be targeted. Mutagenesis frequency after endonuclease exposure can be increased nearly 2-fold by treatment with a histone deacetylase (HDAC) inhibitor. Using a mouse model of latent HSV infection, we demonstrate that a targeted endonuclease can be delivered to viral latency sites via an adeno-associated virus (AAV) vector, where it is able to induce mutation of latent HSV genomes. These data provide the first proof-of-principle to our knowledge for the use of a targeted endonuclease as an antiviral agent to treat an established latent viral infection in vivo.

13.
Antiviral Res ; 126: 90-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26718067

RESUMEN

Incurable chronic viral infections are a major cause of morbidity and mortality worldwide. One potential approach to cure persistent viral infections is via the use of targeted endonucleases. Nevertheless, a potential concern for endonuclease-based antiviral therapies is the emergence of treatment resistance. Here we detect for the first time an endonuclease-resistant infectious virus that is found with high frequency after antiviral endonuclease therapy. While testing the activity of HIV pol-specific zinc finger nucleases (ZFNs) alone or in combination with three prime repair exonuclease 2 (Trex2), we identified a treatment-resistant and infectious mutant virus that was derived from a ZFN-mediated disruption of reverse transcriptase (RT). Although gene disruption of HIV protease, RT and integrase could inhibit viral replication, a chance single amino acid insertion within the thumb domain of RT produced a virus that could actively replicate. The endonuclease-resistant virus could replicate in primary CD4(+) T cells, but remained susceptible to treatment with antiretroviral RT inhibitors. When secondary ZFN-derived mutations were introduced into the mutant virus's RT or integrase domains, replication could be abolished. Our observations suggest that caution should be exercised during endonuclease-based antiviral therapies; however, combination endonuclease therapies may prevent the emergence of resistance.


Asunto(s)
Fármacos Anti-VIH/farmacología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/genética , Inhibidores de la Transcriptasa Inversa/farmacología , Dedos de Zinc , Secuencia de Bases , Línea Celular , ADN Viral/genética , Farmacorresistencia Viral , Endonucleasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/farmacología , Productos del Gen pol/genética , Productos del Gen pol/metabolismo , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/terapia , Proteasa del VIH/genética , Proteasa del VIH/metabolismo , Transcriptasa Inversa del VIH/genética , Transcriptasa Inversa del VIH/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacología , Transducción Genética , Replicación Viral/efectos de los fármacos
14.
PLoS One ; 9(5): e97579, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24827459

RESUMEN

Despite an existing effective vaccine, hepatitis B virus (HBV) remains a major public health concern. There are effective suppressive therapies for HBV, but they remain expensive and inaccessible to many, and not all patients respond well. Furthermore, HBV can persist as genomic covalently closed circular DNA (cccDNA) that remains in hepatocytes even during otherwise effective therapy and facilitates rebound in patients after treatment has stopped. Therefore, the need for an effective treatment that targets active and persistent HBV infections remains. As a novel approach to treat HBV, we have targeted the HBV genome for disruption to prevent viral reactivation and replication. We generated 3 zinc finger nucleases (ZFNs) that target sequences within the HBV polymerase, core and X genes. Upon the formation of ZFN-induced DNA double strand breaks (DSB), imprecise repair by non-homologous end joining leads to mutations that inactivate HBV genes. We delivered HBV-specific ZFNs using self-complementary adeno-associated virus (scAAV) vectors and tested their anti-HBV activity in HepAD38 cells. HBV-ZFNs efficiently disrupted HBV target sites by inducing site-specific mutations. Cytotoxicity was seen with one of the ZFNs. scAAV-mediated delivery of a ZFN targeting HBV polymerase resulted in complete inhibition of HBV DNA replication and production of infectious HBV virions in HepAD38 cells. This effect was sustained for at least 2 weeks following only a single treatment. Furthermore, high specificity was observed for all ZFNs, as negligible off-target cleavage was seen via high-throughput sequencing of 7 closely matched potential off-target sites. These results show that HBV-targeted ZFNs can efficiently inhibit active HBV replication and suppress the cellular template for HBV persistence, making them promising candidates for eradication therapy.


Asunto(s)
Antivirales/metabolismo , Endonucleasas/genética , Productos del Gen pol/genética , Marcación de Gen , Virus de la Hepatitis B/genética , Transactivadores/genética , Proteínas del Núcleo Viral/genética , Antivirales/química , Secuencia de Bases , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Viral/genética , Dependovirus/genética , Endonucleasas/química , Endonucleasas/metabolismo , Productos del Gen pol/antagonistas & inhibidores , Productos del Gen pol/química , Vectores Genéticos , Células HEK293 , Virus de la Hepatitis B/química , Hepatocitos/virología , Humanos , Datos de Secuencia Molecular , Ingeniería de Proteínas , Transactivadores/antagonistas & inhibidores , Transactivadores/química , Proteínas del Núcleo Viral/antagonistas & inhibidores , Proteínas del Núcleo Viral/química , Proteínas Reguladoras y Accesorias Virales , Replicación Viral/genética , Dedos de Zinc/genética
15.
Virology ; 454-455: 353-61, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24485787

RESUMEN

Treatment for most persistent viral infections consists of palliative drug options rather than curative approaches. This is often because long-lasting viral DNA in infected cells is not affected by current antivirals, providing a source for viral persistence and reactivation. Targeting latent viral DNA itself could therefore provide a basis for novel curative strategies. DNA cleavage enzymes can be used to induce targeted mutagenesis of specific genes, including those of exogenous viruses. Although initial in vitro and even in vivo studies have been carried out using DNA cleavage enzymes targeting various viruses, many questions still remain concerning the feasibility of these strategies as they transition into preclinical research. Here, we review the most recent findings on DNA cleavage enzymes for human viral infections, consider the most relevant animal models for several human viral infections, and address issues regarding safety and enzyme delivery. Results from well-designed in vivo studies will ideally provide answers to the most urgent remaining questions, and allow continued progress toward clinical application.


Asunto(s)
ADN Viral/genética , ADN Viral/metabolismo , Virosis/terapia , Latencia del Virus , Fenómenos Fisiológicos de los Virus , Animales , División del ADN , Humanos
16.
Mol Ther Nucleic Acids ; 3: e146, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24496438

RESUMEN

Following acute infection, herpes simplex virus (HSV) establishes latency in sensory neurons, from which it can reactivate and cause recurrent disease. Available antiviral therapies do not affect latent viral genomes; therefore, they do not prevent reactivation following therapy cessation. One possible curative approach involves the introduction of DNA double strand breaks in latent HSV genomes by rare-cutting endonucleases, leading to mutagenesis of essential viral genes. We tested this approach in an in vitro HSV latency model using the engineered homing endonuclease (HE) HSV1m5, which recognizes a sequence in the HSV-1 gene UL19, encoding the virion protein VP5. Coexpression of the 3'-exonuclease Trex2 with HEs increased HE-mediated mutagenesis frequencies up to sixfold. Following HSV1m5/Trex2 delivery with adeno-associated viral (AAV) vectors, the target site was mutated in latent HSV genomes with no detectable cell toxicity. Importantly, HSV production by latently infected cells after reactivation was decreased after HSV1m5/Trex2 exposure. Exposure to histone deacetylase inhibitors prior to HSV1m5/Trex2 treatment increased mutagenesis frequencies of latent HSV genomes another two- to fivefold, suggesting that chromatin modification may be a useful adjunct to gene-targeting approaches. These results support the continuing development of HEs and other nucleases (ZFNs, TALENs, CRISPRs) for cure of chronic viral infections.Molecular Therapy-Nucleic Acids (2014) 3, e1; doi:10.1038/mtna.2013.75; published online 4 February 2014.

17.
J Virol ; 86(17): 8920-36, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22718830

RESUMEN

Human immunodeficiency virus type 1 (HIV-1), hepatitis B virus (HBV), and herpes simplex virus (HSV) have been incurable to date because effective antiviral therapies target only replicating viruses and do not eradicate latently integrated or nonreplicating episomal viral genomes. Endonucleases that can target and cleave critical regions within latent viral genomes are currently in development. These enzymes are being engineered with high specificity such that off-target binding of cellular DNA will be absent or minimal. Imprecise nonhomologous-end-joining (NHEJ) DNA repair following repeated cleavage at the same critical site may permanently disrupt translation of essential viral proteins. We discuss the benefits and drawbacks of three types of DNA cleavage enzymes (zinc finger endonucleases, transcription activator-like [TAL] effector nucleases [TALENs], and homing endonucleases [also called meganucleases]), the development of delivery vectors for these enzymes, and potential obstacles for successful treatment of chronic viral infections. We then review issues regarding persistence of HIV-1, HBV, and HSV that are relevant to eradication with genome-altering approaches.


Asunto(s)
ADN Viral/genética , Endonucleasas/antagonistas & inhibidores , Mutagénesis , Virosis/tratamiento farmacológico , Virosis/virología , Virus/genética , Animales , ADN Viral/metabolismo , Humanos , Virus/efectos de los fármacos , Virus/metabolismo
18.
PLoS One ; 6(2): e16825, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21399673

RESUMEN

Current antiviral therapy does not cure HIV-infected individuals because the virus establishes lifelong latent infection within long-lived memory T cells as integrated HIV proviral DNA. Here, we report a new therapeutic approach that aims to cure cells of latent HIV infection by rendering latent virus incapable of replication and pathogenesis via targeted cellular mutagenesis of essential viral genes. This is achieved by using a homing endonuclease to introduce DNA double-stranded breaks (dsb) within the integrated proviral DNA, which is followed by triggering of the cellular DNA damage response and error-prone repair. To evaluate this concept, we developed an in vitro culture model of viral latency, consisting of an integrated lentiviral vector with an easily evaluated reporter system to detect targeted mutagenesis events. Using this system, we demonstrate that homing endonucleases can efficiently and selectively target an integrated reporter lentivirus within the cellular genome, leading to mutation in the proviral DNA and loss of reporter gene expression. This new technology offers the possibility of selectively disabling integrated HIV provirus within latently infected cells.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Marcación de Gen/métodos , Genes Reporteros/genética , Integrasa de VIH/metabolismo , Lentivirus/genética , Secuencia de Bases , Células Cultivadas , Eficiencia , Endodesoxirribonucleasas/genética , Infecciones por VIH/genética , Infecciones por VIH/terapia , Integrasa de VIH/genética , VIH-1/genética , Humanos , Células Jurkat , Datos de Secuencia Molecular , Mutagénesis Insercional/métodos , Mutagénesis Insercional/fisiología , Ingeniería de Proteínas/métodos , Homología de Secuencia de Ácido Nucleico , Integración Viral/fisiología , Latencia del Virus/genética , Latencia del Virus/fisiología
19.
J Virol ; 83(12): 6171-83, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19339346

RESUMEN

The virological synapse (VS) is a specialized molecular structure that facilitates the transfer of certain lymphotropic viruses into uninfected T cells. However, the role of the VS in the transfer of nonlymphotropic viruses into T cells is unknown. Herpes simplex virus (HSV) has been shown in vitro to infect T cells and modulate T-cell receptor function, thereby suppressing T-cell antiviral function. However, whether such infection of T cells occurs in vivo is unknown. Here, we examined whether T-cell infection could be observed in human HSV disease and investigated the mechanism of HSV entry into T cells. We found that HSV-infected T cells were readily detectable during human disease, suggesting that infection and modulation of T-cell function plays a role in human immunopathology. HSV infection of both CD4(+) and CD8(+) T cells occurred much more efficiently via direct cell-to-cell spread from infected fibroblasts than by cell-free virus. Activation of T cells increased their permissivity to HSV infection. Cell-to-cell spread to T cells did not require HSV glycoproteins E and I (gE and gI), which are critical for cell-to-cell spread between epithelial cells. Transfer of HSV to T cells required gD, and the four known entry receptors appear to be contributing to viral entry, with a dominant role for the herpesvirus entry mediator and nectin-1. VS-like structures enriched in activated lymphocyte function-associated antigen 1 (LFA-1) were observed at the point of contact between HSV-infected fibroblasts and T cells. Consistent with spread occurring via the VS, transfer of HSV was increased by activation of LFA-1, and cell-to-cell spread could be inhibited by antibodies to LFA-1 or gD. Taken together, these results constitute the first demonstration of VS-dependent cell-to-cell spread for a predominantly nonlymphotropic virus. Furthermore, they support an important role for infection and immunomodulation of T cells in clinical human disease. Targeting of the VS might allow selective immunopotentiation during infections with HSV or other nonlymphotropic viruses.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Linfocitos T/virología , Internalización del Virus , Animales , Moléculas de Adhesión Celular/metabolismo , Chlorocebus aethiops , Fibroblastos/virología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Humanos , Células Jurkat , Activación de Linfocitos , Ratones , Nectinas , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Células Vero , Proteínas del Envoltorio Viral/metabolismo
20.
Methods Mol Biol ; 414: 47-56, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18175811

RESUMEN

Apoptosis (programmed cell death) is an active process that plays a critical role in multiple biologic processes from embryologic development, to lymphocyte development and selection, and homeostasis. The two major mechanisms of cell death are referred to as the intrinsic and extrinsic pathways. These pathways lead to a cascade of events that ultimately converge to the activation of an effector enzyme, caspase-3. Caspase-3 is a cysteine protease with aspartic specificity and a well-characterized effector of apoptosis or programmed cell death signaling. The pro-form of caspase-3 (p32 caspase-3) is sequestered as a zymogen, where upon proteolysis at a conserved DEVD sequence, is converted to the active (p17 caspase-3) enzyme capable of disassembling the cell. Cell death can become disregulated under various conditions and multiple disease states (e.g., viral infection, carcinogenesis, and metastasis). Sensitive and reproducible detection of active caspase-3 is critical to advance the understanding of cellular functions and multiple pathologies of various etiologies. Here, we provide two simple and reproducible methods to measure active caspase-3 in multiple cell types and conditions using a flow cytometric-based analysis.


Asunto(s)
Caspasa 3/análisis , Citometría de Flujo/métodos , Animales , Apoptosis/fisiología , Caspasa 3/metabolismo , Activación Enzimática , Humanos , Modelos Biológicos , Coloración y Etiquetado/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA