Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Ther ; 27(3): 611-622, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30772143

RESUMEN

Adeno-associated virus (AAV) has emerged as a promising gene delivery vector because of its non-pathogenicity, simple structure and genome, and low immunogenicity compared to other viruses. However, its adoption as a safe and effective delivery vector for certain diseases relies on altering its tropism to deliver transgenes to desired cell populations. To this end, we have developed a protease-activatable AAV vector, named provector, that responds to elevated extracellular protease activity commonly found in diseased tissue microenvironments. The AAV9-based provector is initially inactive, but then it can be switched on by matrix metalloproteinases (MMP)-2 and -9. Cryo-electron microscopy and image reconstruction reveal that the provector capsid is structurally similar to that of AAV9, with a flexible peptide insertion at the top of the 3-fold protrusions. In an in vivo model of myocardial infarction (MI), the provector is able to deliver transgenes site specifically to high-MMP-activity regions of the damaged heart, with concomitant decreased delivery to many off-target organs, including the liver. The AAV provector may be useful in the future for enhanced delivery of transgenes to sites of cardiac damage.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Animales , Anticuerpos Neutralizantes/metabolismo , Circulación Sanguínea/fisiología , Microscopía por Crioelectrón , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Miocardio/metabolismo , Miocardio/patología
2.
Lab Invest ; 99(8): 1233-1244, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30728464

RESUMEN

Genetically engineered mouse models (GEMMs) that recapitulate the major genetic drivers in pancreatic ductal adenocarcinoma (PDAC) have provided unprecedented insights into the pathogenesis of this lethal neoplasm. Nonetheless, generating an autochthonous model is an expensive, time consuming and labor intensive process, particularly when tissue specific expression or deletion of compound alleles are involved. In addition, many of the current PDAC GEMMs cause embryonic, pancreas-wide activation or loss of driver alleles, neither of which reflects the cognate human disease scenario. The advent of CRISPR/Cas9 based gene editing can potentially circumvent many of the aforementioned shortcomings of conventional breeding schema, but ensuring the efficiency of gene editing in vivo remains a challenge. Here we have developed a pipeline for generating PDAC GEMMs of complex genotypes with high efficiency using a single "workhorse" mouse strain expressing Cas9 in the adult pancreas under a p48 promoter. Using adeno-associated virus (AAV) mediated delivery of multiplexed guide RNAs (sgRNAs) to the adult murine pancreas of p48-Cre; LSL-Cas9 mice, we confirm our ability to express an oncogenic Kras G12D allele through homology-directed repair (HDR), in conjunction with CRISPR-induced disruption of cooperating alleles (Trp53, Lkb1 and Arid1A). The resulting GEMMs demonstrate a spectrum of precursor lesions (pancreatic intraepithelial neoplasia [PanIN] or Intraductal papillary mucinous neoplasm [IPMN] with eventual progression to PDAC. Next generation sequencing of the resulting murine PDAC confirms HDR of oncogenic KrasG12D allele at the endogenous locus, and insertion deletion ("indel") and frameshift mutations of targeted tumor suppressor alleles. By using a single "workhorse" mouse strain and optimal AAV serotype for in vivo gene editing with combination of driver alleles, we present a facile autochthonous platform for interrogation of the PDAC genome.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Neoplasias Experimentales , Neoplasias Pancreáticas , Recombinación Genética/genética , Animales , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Guía de Kinetoplastida/genética
3.
Virology ; 563: 107-115, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34509702

RESUMEN

Adeno-associated virus (AAV) is a promising gene therapy vector, but questions remain regarding mechanisms of basic viral functions. We previously showed that a serine/threonine (S/T) triplet motif and its flanking residues, located in the overlapping N-terminus of VP1/VP2 and highly conserved across most AAV serotypes, are critical for viral transcript production in vitro. Here we generate a panel of S/T triplet mutants in AAV serotypes 2, 4, and 9 and characterize their behaviors in vitro and in vivo using next generation sequencing. We show that S/T triplet mutations can significantly hinder some stages of transduction in a serotype-dependent manner in vitro. Interestingly, these defects are largely overcome in C57BL/6 mice, with only one mutant displaying altered behavior in vivo. Taken together, our results identify a short N-terminal capsid motif with diverse roles across several AAV serotypes which better informs engineering efforts to improve AAV as a vector for gene therapy.


Asunto(s)
Proteínas de la Cápside/metabolismo , Dependovirus/clasificación , Dependovirus/fisiología , Regulación Viral de la Expresión Génica/fisiología , Serogrupo , Secuencia de Aminoácidos , Animales , Células COS , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Chlorocebus aethiops , Clonación Molecular , Dependovirus/genética , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación
4.
Virology ; 546: 127-132, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32452411

RESUMEN

Adeno-associated virus (AAV) is one of the most researched, clinically utilized gene therapy vectors. Though clinical success has been achieved, transgene delivery and expression may be hindered by cellular and tissue barriers. Understanding the role of receptor binding, entry, endosomal escape, cytoplasmic and nuclear trafficking, capsid uncoating, and viral transcription in therapeutic efficacy is paramount. Previous studies have shown that N-terminal regions of the AAV capsid proteins are responsible for endosomal escape and nuclear trafficking, however the mechanisms remain unknown. We identified a highly-conserved three-residue serine/threonine (S/T) motif in the capsid N-terminus, previously uncharacterized in its role in intracellular trafficking and transduction. Using alanine scanning mutagenesis, we found S155 and the flanking residues, D154 and G158, are essential for AAV2 transduction efficiency. Remarkably, specific capsid mutants show a 5 to 9-fold decrease in viral mRNA transcripts, highlighting a potential role of the S/T motif in transcription of the viral genome.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Dependovirus/genética , Regulación Viral de la Expresión Génica , Secuencias de Aminoácidos , Proteínas de la Cápside/genética , Dependovirus/química , Dependovirus/fisiología , Vectores Genéticos/química , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Ensamble de Virus , Replicación Viral
5.
Tissue Eng Part C Methods ; 21(8): 808-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25603445

RESUMEN

Currently, curative therapies for heart valve diseases do not exist, thus motivating the need for new therapeutics, regenerative and tissue-engineered valves, and further basic research into pathological mechanisms. For studying valve diseases and developing valve therapies, effective methods to manipulate gene expression in primary valvular interstitial cells (VICs), which promote calcification in disease, would be valuable. Unfortunately, there is little information reported about effective gene delivery methods for VICs. Adeno-associated virus (AAV) is a clinically proven gene delivery vector capable of transducing many cell types and tissues, but has not yet been reported to infect valvular cells. In this study, AAV serotypes 1-9 were tested for their ability to deliver a green fluorescent protein (GFP) reporter into VICs in vitro. Flow cytometry results indicate AAV2 and AAV3 are capable of transducing VICs more efficiently than other serotypes. Furthermore, transduction efficiencies can be optimized by increasing the multiplicity of infection (MOI) and using self-complementary, double-stranded genomes, yielding up to 98% successfully transduced cells. Transduction of VICs by AAV2 or AAV3 in the presence of competing soluble heparin significantly reduces delivery efficiencies, suggesting heparan sulfate proteoglycans act as the primary VIC receptors of these two serotypes. Overall, this study establishes AAV2 and AAV3 as efficient gene delivery vehicles for primary VICs. Such effective delivery vectors for valve cells may be broadly useful for numerous applications, including the study of valvular cell biology, development of valve disease therapies, and regulation of genes for tissue engineering heart valves.


Asunto(s)
Válvula Aórtica/metabolismo , Dependovirus , Vectores Genéticos , Transducción Genética/métodos , Animales , Válvula Aórtica/citología , Porcinos
6.
ACS Nano ; 8(5): 4740-6, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24796495

RESUMEN

We explored the unique signal integration properties of the self-assembling 60-mer protein capsid of adeno-associated virus (AAV), a clinically proven human gene therapy vector, by engineering proteolytic regulation of virus-receptor interactions such that processing of the capsid by proteases is required for infection. We find the transfer function of our engineered protease-activatable viruses (PAVs), relating the degree of proteolysis (input) to PAV activity (output), is highly nonlinear, likely due to increased polyvalency. By exploiting this dynamic polyvalency, in combination with the self-assembly properties of the virus capsid, we show that mosaic PAVs can be constructed that operate under a digital AND gate regime, where two different protease inputs are required for virus activation. These results show viruses can be engineered as signal-integrating nanoscale nodes whose functional properties are regulated by multiple proteolytic signals with easily tunable and predictable response surfaces, a promising development toward advanced control of gene delivery.


Asunto(s)
Dependovirus/química , Nanopartículas/química , Nanotecnología/métodos , Péptido Hidrolasas/química , Virus/química , Secuencia de Aminoácidos , Cápside/química , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Proteínas Fluorescentes Verdes/química , Células HEK293 , Humanos , Microscopía Electrónica , Datos de Secuencia Molecular , Nanomedicina/métodos , Plásmidos/química , Ingeniería de Proteínas/métodos , Propiedades de Superficie , Temperatura , Transgenes
7.
ACS Synth Biol ; 2(12): 724-33, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23899192

RESUMEN

Adeno-associated virus (AAV) recombination can result in chimeric capsid protein subunits whose ability to assemble into an oligomeric capsid, package a genome, and transduce cells depends on the inheritance of sequence from different AAV parents. To develop quantitative design principles for guiding site-directed recombination of AAV capsids, we have examined how capsid structural perturbations predicted by the SCHEMA algorithm correlate with experimental measurements of disruption in seventeen chimeric capsid proteins. In our small chimera population, created by recombining AAV serotypes 2 and 4, we found that protection of viral genomes and cellular transduction were inversely related to calculated disruption of the capsid structure. Interestingly, however, we did not observe a correlation between genome packaging and calculated structural disruption; a majority of the chimeric capsid proteins formed at least partially assembled capsids and more than half packaged genomes, including those with the highest SCHEMA disruption. These results suggest that the sequence space accessed by recombination of divergent AAV serotypes is rich in capsid chimeras that assemble into 60-mer capsids and package viral genomes. Overall, the SCHEMA algorithm may be useful for delineating quantitative design principles to guide the creation of libraries enriched in genome-protecting virus nanoparticles that can effectively transduce cells. Such improvements to the virus design process may help advance not only gene therapy applications but also other bionanotechnologies dependent upon the development of viruses with new sequences and functions.


Asunto(s)
Cápside/química , Quimera/genética , Dependovirus/genética , Ingeniería de Proteínas/métodos , Recombinación Genética/genética , Algoritmos , Quimera/metabolismo , Biología Computacional , Dependovirus/química , Dependovirus/metabolismo , Genoma Viral , Heparina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA