Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Biotechnol ; 40(2): 194-197, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34462587

RESUMEN

CRISPR-Cas13 systems have been developed for precise RNA editing, and can potentially be used therapeutically when temporary changes are desirable or when DNA editing is challenging. We have identified and characterized an ultrasmall family of Cas13b proteins-Cas13bt-that can mediate mammalian transcript knockdown. We have engineered compact variants of REPAIR and RESCUE RNA editors by functionalizing Cas13bt with adenosine and cytosine deaminase domains, and demonstrated packaging of the editors within a single adeno-associated virus.


Asunto(s)
Sistemas CRISPR-Cas , ARN , Adenosina/genética , Adenosina Desaminasa/genética , Animales , Sistemas CRISPR-Cas/genética , Edición Génica , Mamíferos/genética , ARN/genética , Edición de ARN/genética
2.
Nat Methods ; 18(8): 965-974, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34341582

RESUMEN

CRISPR-Cas9 technologies have dramatically increased the ease of targeting DNA sequences in the genomes of living systems. The fusion of chromatin-modifying domains to nuclease-deactivated Cas9 (dCas9) has enabled targeted epigenome editing in both cultured cells and animal models. However, delivering large dCas9 fusion proteins to target cells and tissues is an obstacle to the widespread adoption of these tools for in vivo studies. Here, we describe the generation and characterization of two conditional transgenic mouse lines for epigenome editing, Rosa26:LSL-dCas9-p300 for gene activation and Rosa26:LSL-dCas9-KRAB for gene repression. By targeting the guide RNAs to transcriptional start sites or distal enhancer elements, we demonstrate regulation of target genes and corresponding changes to epigenetic states and downstream phenotypes in the brain and liver in vivo, and in T cells and fibroblasts ex vivo. These mouse lines are convenient and valuable tools for facile, temporally controlled, and tissue-restricted epigenome editing and manipulation of gene expression in vivo.


Asunto(s)
Sistemas CRISPR-Cas , Epigénesis Genética , Epigenoma , Edición Génica/métodos , Regulación de la Expresión Génica , Animales , Encéfalo/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Linfocitos T/metabolismo
3.
J Virol ; 94(21)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32817219

RESUMEN

Adeno-associated viruses (AAVs) are dependoparvoviruses that have proven useful for therapeutic gene transfer; however, our understanding of host factors that influence AAV trafficking and transduction is still evolving. Here, we investigated the role of cellular calcium in the AAV infectious pathway. First, we demonstrated a critical role for the host Golgi compartment-resident ATP-powered calcium pump (secretory pathway calcium ATPase 1 [SPCA1]) encoded by the ATP2C1 gene in AAV infection. CRISPR-based knockout (KO) of ATP2C1 decreases transduction by different AAV serotypes. ATP2C1 KO does not appear to inhibit AAV binding, cellular uptake, or nuclear entry; however, capsids within ATP2C1 KO cells demonstrate dispersed and punctate trafficking distinct from the perinuclear, trans-Golgi pattern observed in normal cells. In addition, we observed a defect in the ability of AAV capsids to undergo conformational changes and support efficient vector genome transcription in ATP2C1 KO cells. The calcium chelator BAPTA-AM, which reduces cytosolic calcium, rescues the defective ATP2C1 KO phenotype and AAV transduction in vitro Conversely, the calcium ionophore ionomycin, which disrupts calcium gradients, blocks AAV transduction. Further, we demonstrated that modulating calcium in the murine brain using BAPTA-AM augments AAV gene expression in vivo Taking these data together, we postulate that the maintenance of an intracellular calcium gradient by the calcium ATPase and processing within the Golgi compartment are essential for priming the capsid to support efficient AAV genome transcription.IMPORTANCE Adeno-associated viruses (AAVs) have proven to be effective gene transfer vectors. However, our understanding of how the host cell environment influences AAV transduction is still evolving. In the present study, we investigated the role of ATP2C1, which encodes a membrane calcium transport pump, SPCA1, essential for maintaining cellular calcium homeostasis on AAV transduction. Our results indicate that cellular calcium is essential for efficient intracellular trafficking and conformational changes in the AAV capsid that support efficient genome transcription. Further, we show that pharmacological modulation of cellular calcium levels can potentially be applied to improve the AAV gene transfer efficiency.


Asunto(s)
ATPasas Transportadoras de Calcio/genética , Calcio/metabolismo , Dependovirus/genética , Vectores Genéticos/metabolismo , Aparato de Golgi/metabolismo , Animales , Animales Recién Nacidos , Transporte Biológico/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Sistemas CRISPR-Cas , ATPasas Transportadoras de Calcio/deficiencia , Línea Celular Tumoral , Quelantes/farmacología , Dependovirus/efectos de los fármacos , Dependovirus/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Eliminación de Gen , Vectores Genéticos/química , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/virología , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inyecciones Intraventriculares , Ionomicina/farmacología , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas Estereotáxicas , Transducción Genética , Vesiculovirus/genética , Vesiculovirus/metabolismo
4.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31391273

RESUMEN

Adeno-associated viruses (AAV) are helper-dependent parvoviruses that have been developed into promising gene therapy vectors. Many studies, including a recent unbiased genomic screen, have identified host factors essential for AAV cell entry, but no genome-wide screens that address inhibitory host factors have been reported. Here, we utilize a novel CRISPR screen to identify AAV restriction factors in a human hepatocyte cell line. The major hit from our gain-of-function screen is the apical polarity determinant Crumbs 3 (Crb3). Knockout (KO) of Crb3 enhances AAV transduction, while overexpression exerts the opposite effect. Further, Crb3 appears to restrict AAV transduction in a serotype- and cell type-specific manner. Particularly, for AAV serotype 9 and a rationally engineered AAV variant, we demonstrate that increased availability of galactosylated glycans on the surfaces of Crb3 KO cells, but not the universal AAV receptor, leads to increased capsid attachment and enhanced transduction. We postulate that Crb3 could serve as a key molecular determinant that restricts the availability of AAV glycan attachment factors on the cell surface by maintaining apical-basal polarity and tight junction integrity.IMPORTANCE Adeno-associated viruses (AAVs) have recently emerged at the forefront as gene therapy vectors; however, our understanding of host factors that influence AAV transduction in different cell types is still evolving. In the present study, we perform a genome-scale CRISPR knockout screen to identify cellular host factors that restrict AAV infection in hepatocyte cultures. We discover that Crumbs 3, which determines cellular polarity, also influences the distribution of certain carbohydrate attachment factors on the cell surface. This in turn affects the ability of virions to bind and enter the cells. This study underscores the importance of cell polarity in AAV transduction and provides a potential molecular basis for the differential infectious mechanism(s) in cell culture versus organ systems.


Asunto(s)
Dependovirus/fisiología , Hepatocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Infecciones por Parvoviridae/virología , Sistemas CRISPR-Cas , Cápside/metabolismo , Línea Celular , Membrana Celular/metabolismo , Polaridad Celular , Claudinas/genética , Claudinas/metabolismo , Dependovirus/genética , Expresión Génica , Técnicas de Inactivación de Genes , Hepatocitos/fisiología , Hepatocitos/virología , Humanos , Glicoproteínas de Membrana/genética , Infecciones por Parvoviridae/metabolismo , Polisacáridos/metabolismo , Serogrupo , Uniones Estrechas , Transducción Genética , Acoplamiento Viral
5.
PLoS Pathog ; 15(8): e1007988, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31386698

RESUMEN

Adeno-associated viruses (AAV) are Dependoparvoviruses that have shown promise as recombinant vectors for gene therapy. While infectious pathways of AAV are well studied, gaps remain in our understanding of host factors affecting vector genome expression. Here, we map the role of ring finger protein 121 (RNF121), an E3 ubiquitin ligase, as a key regulator of AAV genome transcription. CRISPR-mediated knockout of RNF121 (RNF121 KO) in different cells markedly decreased AAV transduction regardless of capsid serotype or vector dose. Recombinant AAV transduction is partially rescued by overexpressing RNF121, but not by co-infection with helper Adenovirus. Major steps in the AAV infectious pathway including cell surface binding, cellular uptake, nuclear entry, capsid uncoating and second strand synthesis are unaffected. While gene expression from transfected plasmids or AAV genomes is unaffected, mRNA synthesis from AAV capsid-associated genomes is markedly decreased in RNF121 KO cells. These observations were attributed to transcriptional arrest as corroborated by RNAPol-ChIP and mRNA half-life measurements. Although AAV capsid proteins do not appear to be direct substrates of RNF121, the catalytic domain of the E3 ligase appears essential. Inhibition of ubiquitin-proteasome pathways revealed that blocking Valosin Containing Protein (VCP/p97), which targets substrates to the proteasome, can selectively and completely restore AAV-mediated transgene expression in RNF121 KO cells. Expanding on this finding, transcriptomic and proteomic analysis revealed that the catalytic subunit of DNA PK (DNAPK-Cs), a known activator of VCP, is upregulated in RNF121 KO cells and that the DNA damage machinery is enriched at sites of stalled AAV genome transcription. We postulate that a network of RNF121, VCP and DNA damage response elements function together to regulate transcriptional silencing and/or activation of AAV vector genomes.


Asunto(s)
Carcinoma Hepatocelular/virología , Proteína Quinasa Activada por ADN/metabolismo , Dependovirus/genética , Genoma Viral , Proteínas de la Membrana/metabolismo , Transducción Genética , Proteína que Contiene Valosina/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteína Quinasa Activada por ADN/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteoma , Transcriptoma , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitinación , Proteína que Contiene Valosina/genética , Internalización del Virus
6.
Mol Ther ; 26(2): 510-523, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29175157

RESUMEN

Effective gene delivery to the CNS by intravenously administered adeno-associated virus (AAV) vectors requires crossing the blood-brain barrier (BBB). To achieve therapeutic CNS transgene expression, high systemic vector doses are often required, which poses challenges such as scale-up costs and dose-dependent hepatotoxicity. To improve the specificity and efficiency of CNS gene transfer, a better understanding of the structural features that enable AAV transit across the BBB is needed. We generated a combinatorial domain swap library using AAV1, a serotype that does not traverse the vasculature, and AAVrh.10, which crosses the BBB in mice. We then screened individual variants by phylogenetic and structural analyses and subsequently conducted systemic characterization in mice. Using this approach, we identified key clusters of residues on the AAVrh.10 capsid that enabled transport across the brain vasculature and widespread neuronal transduction in mice. Through rational design, we mapped a minimal footprint from AAVrh.10, which, when grafted onto AAV1, confers the aforementioned CNS phenotype while diminishing vascular and hepatic transduction through an unknown mechanism. Functional mapping of this capsid surface footprint provides a roadmap for engineering synthetic AAV capsids for efficient CNS gene transfer with an improved safety profile.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/virología , Dependovirus/fisiología , Dependovirus/ultraestructura , Animales , Transporte Biológico , Encéfalo/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Dependovirus/clasificación , Expresión Génica , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Modelos Moleculares , Miocardio/metabolismo , Especificidad de Órganos , Filogenia , Unión Proteica , Distribución Tisular , Transducción Genética , Transgenes
7.
Proc Natl Acad Sci U S A ; 114(24): E4812-E4821, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28559317

RESUMEN

Preexisting neutralizing antibodies (NAbs) against adeno-associated viruses (AAVs) pose a major, unresolved challenge that restricts patient enrollment in gene therapy clinical trials using recombinant AAV vectors. Structural studies suggest that despite a high degree of sequence variability, antibody recognition sites or antigenic hotspots on AAVs and other related parvoviruses might be evolutionarily conserved. To test this hypothesis, we developed a structure-guided evolution approach that does not require selective pressure exerted by NAbs. This strategy yielded highly divergent antigenic footprints that do not exist in natural AAV isolates. Specifically, synthetic variants obtained by evolving murine antigenic epitopes on an AAV serotype 1 capsid template can evade NAbs without compromising titer, transduction efficiency, or tissue tropism. One lead AAV variant generated by combining multiple evolved antigenic sites effectively evades polyclonal anti-AAV1 neutralizing sera from immunized mice and rhesus macaques. Furthermore, this variant displays robust immune evasion in nonhuman primate and human serum samples at dilution factors as high as 1:5, currently mandated by several clinical trials. Our results provide evidence that antibody recognition of AAV capsids is conserved across species. This approach can be applied to any AAV strain to evade NAbs in prospective patients for human gene therapy.


Asunto(s)
Dependovirus/genética , Dependovirus/inmunología , Evolución Molecular Dirigida/métodos , Evasión Inmune/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/inmunología , Variación Antigénica/genética , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Dependovirus/clasificación , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos , Células HEK293 , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Serotipificación
8.
Curr Opin Virol ; 18: 89-96, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27262111

RESUMEN

Adeno-associated viruses (AAV) are currently at the forefront of human gene therapy clinical trials as recombinant vectors. Significant progress has been made in elucidating the structure, biology and tropisms of different naturally occurring AAV isolates in the past decade. In particular, a spectrum of AAV capsid interactions with host receptors have been identified and characterized. These studies have enabled a better understanding of key determinants of AAV cell recognition and entry in different hosts. This knowledge is now being applied toward engineering new, lab-derived AAV capsids with favorable transduction profiles. The current review conveys a structural perspective of capsid-glycan interactions and provides a roadmap for generating synthetic strains by engineering AAV receptor footprints.


Asunto(s)
Proteínas de la Cápside/metabolismo , Dependovirus/genética , Dependovirus/fisiología , Ingeniería Genética/métodos , Terapia Genética , Receptores Virales/genética , Receptores Virales/metabolismo , Cápside/química , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Vectores Genéticos , Interacciones Huésped-Patógeno , Humanos , Polisacáridos/química , Polisacáridos/metabolismo
9.
PLoS One ; 8(9): e73493, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039960

RESUMEN

Nerve damage is a major morbidity associated with numerous surgical interventions. Yet, nerve visualization continues to challenge even the most experienced surgeons. A nerve-specific fluorescent contrast agent, especially one with near-infrared (NIR) absorption and emission, would be of immediate benefit to patients and surgeons. Currently, there are only three classes of small molecule organic fluorophores that penetrate the blood nerve barrier and bind to nerve tissue when administered systemically. Of these three classes, the distyrylbenzenes (DSBs) are particularly attractive for further study. Although not presently in the NIR range, DSB fluorophores highlight all nerve tissue in mice, rats, and pigs after intravenous administration. The purpose of the current study was to define the pharmacophore responsible for nerve-specific uptake and retention, which would enable future molecules to be optimized for NIR optical properties. Structural analogs of the DSB class of small molecules were synthesized using combinatorial solid phase synthesis and commercially available building blocks, which yielded more than 200 unique DSB fluorophores. The nerve-specific properties of all DSB analogs were quantified using an ex vivo nerve-specific fluorescence assay on pig and human sciatic nerve. Results were used to perform quantitative structure-activity relationship (QSAR) modeling and to define the nerve-specific pharmacophore. All DSB analogs with positive ex vivo fluorescence were tested for in vivo nerve specificity in mice to assess the effect of biodistribution and clearance on nerve fluorescence signal. Two new DSB fluorophores with the highest nerve to muscle ratio were tested in pigs to confirm scalability.


Asunto(s)
Medios de Contraste/metabolismo , Colorantes Fluorescentes/metabolismo , Imagen Óptica/métodos , Nervio Ciático/metabolismo , Estirenos/metabolismo , Adulto , Animales , Medios de Contraste/química , Medios de Contraste/farmacocinética , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Humanos , Ratones , Ratas , Nervio Ciático/química , Espectrometría de Fluorescencia , Espectroscopía Infrarroja Corta , Relación Estructura-Actividad , Estirenos/química , Estirenos/farmacocinética , Porcinos , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA