Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Virol ; 95(19): e0058721, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34232726

RESUMO

Adeno-associated viruses utilize different glycans and the AAV receptor (AAVR) for cellular attachment and entry. Directed evolution has yielded new AAV variants; however, structure-function correlates underlying their improved transduction are generally overlooked. Here, we report that infectious cycling of structurally diverse AAV surface loop libraries yields functionally distinct variants. Newly evolved variants show enhanced cellular binding, uptake, and transduction, but through distinct mechanisms. Using glycan-based and genome-wide CRISPR knockout screens, we discover that one AAV variant acquires the ability to recognize sulfated glycosaminoglycans, while another displays receptor switching from AAVR to integrin ß1 (ITGB1). A previously evolved variant, AAVhum.8, preferentially utilizes the ITGB1 receptor over AAVR. Visualization of the AAVhum.8 capsid by cryoelectron microscopy at 2.49-Å resolution localizes the newly acquired integrin recognition motif adjacent to the AAVR footprint. These observations underscore the new finding that distinct AAV surface epitopes can be evolved to exploit different cellular receptors for enhanced transduction. IMPORTANCE Understanding how viruses interact with host cells through cell surface receptors is central to discovery and development of antiviral therapeutics, vaccines, and gene transfer vectors. Here, we demonstrate that distinct epitopes on the surface of adeno-associated viruses can be evolved by infectious cycling to recognize different cell surface carbohydrates and glycoprotein receptors and solve the three-dimensional structure of one such newly evolved AAV capsid, which provides a roadmap for designing viruses with improved attributes for gene therapy applications.


Assuntos
Dependovirus/genética , Dependovirus/metabolismo , Evolução Molecular Direcionada , Receptores Virais/metabolismo , Motivos de Aminoácidos , Sistemas CRISPR-Cas , Capsídeo/química , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Microscopia Crioeletrônica , Dependovirus/química , Dependovirus/ultraestrutura , Variação Genética , Glicosaminoglicanos/metabolismo , Humanos , Integrina beta1/química , Integrina beta1/metabolismo , Polissacarídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/química , Internalização do Vírus
2.
J Virol ; 94(19)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669336

RESUMO

Adeno-associated viruses (AAV) are composed of nonenveloped, icosahedral protein shells that can be adapted to package and deliver recombinant therapeutic DNA. Approaches to engineer recombinant capsids for gene therapy applications have focused on rational design or library-based approaches that can address one or two desirable attributes; however, there is an unmet need to comprehensively improve AAV vector properties. Such cannot be achieved by utilizing sequence data alone but requires harnessing the three-dimensional (3D) structural properties of AAV capsids. Here, we solve the structures of a natural AAV isolate complexed with antibodies using cryo-electron microscopy and harness this structural information to engineer AAV capsid libraries through saturation mutagenesis of different antigenic footprints. Each surface loop was evolved by infectious cycling in the presence of a helper adenovirus to yield a new AAV variant that then serves as a template for evolving the next surface loop. This stepwise process yielded a humanized AAV8 capsid (AAVhum.8) displaying nonnatural surface loops that simultaneously display tropism for human hepatocytes, increased gene transfer efficiency, and neutralizing antibody evasion. Specifically, AAVhum.8 can better evade neutralizing antisera from multiple species than AAV8. Further, AAVhum.8 displays robust transduction in a human liver xenograft mouse model with expanded tropism for both murine and human hepatocytes. This work supports the hypothesis that critical properties, such as AAV capsid antibody evasion and tropism, can be coevolved by combining rational design and library-based evolution for clinical gene therapy.IMPORTANCE Clinical gene therapy with recombinant AAV vectors has largely relied on natural capsid isolates. There is an unmet need to comprehensively improve AAV tissue tropism, transduction efficiency, and antibody evasion. Such cannot be achieved by utilizing capsid sequence data alone but requires harnessing the 3D structural properties of AAV capsids. Here, we combine rational design and library-based evolution to coevolve multiple, desirable properties onto AAV by harnessing 3D structural information.


Assuntos
Proteínas do Capsídeo/imunologia , Capsídeo/imunologia , Dependovirus/imunologia , Tropismo , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Microscopia Crioeletrônica , Dependovirus/genética , Terapia Genética , Hepatócitos/metabolismo , Humanos , Camundongos , Simulação de Acoplamento Molecular
3.
Nat Protoc ; 18(11): 3413-3459, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37735235

RESUMO

Over the past 5 years, our laboratory has systematically developed a structure-guided library approach to evolve new adeno-associated virus (AAV) capsids with altered tissue tropism, higher transduction efficiency and the ability to evade pre-existing humoral immunity. Here, we provide a detailed protocol describing two distinct evolution strategies using structurally divergent AAV serotypes as templates, exemplified by improving CNS gene transfer efficiency in vivo. We outline four major components of our strategy: (i) structure-guided design of AAV capsid libraries, (ii) AAV library production, (iii) library cycling in single versus multiple animal models, followed by (iv) evaluation of lead AAV vector candidates in vivo. The protocol spans ~95 d, excluding gene expression analysis in vivo, and can vary depending on user experience, resources and experimental design. A distinguishing attribute of the current protocol is the focus on providing biomedical researchers with 3D structural information to guide evolution of precise 'hotspots' on AAV capsids. Furthermore, the protocol outlines two distinct methods for AAV library evolution consisting of adenovirus-enabled infectious cycling in a single species and noninfectious cycling in a cross-species manner. Notably, our workflow can be seamlessly merged with other RNA transcript-based library strategies and tailored for tissue-specific capsid selection. Overall, the procedures outlined herein can be adapted to expand the AAV vector toolkit for genetic manipulation of animal models and development of human gene therapies.


Assuntos
Capsídeo , Dependovirus , Animais , Humanos , Capsídeo/química , Dependovirus/genética , Terapia Genética/métodos , Técnicas de Transferência de Genes , Proteínas do Capsídeo/genética , Vetores Genéticos , Transdução Genética
4.
Nat Commun ; 13(1): 5947, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36210364

RESUMO

Recombinant adeno-associated viral (AAV) vectors are a promising gene delivery platform, but ongoing clinical trials continue to highlight a relatively narrow therapeutic window. Effective clinical translation is confounded, at least in part, by differences in AAV biology across animal species. Here, we tackle this challenge by sequentially evolving AAV capsid libraries in mice, pigs and macaques. We discover a highly potent, cross-species compatible variant (AAV.cc47) that shows improved attributes benchmarked against AAV serotype 9 as evidenced by robust reporter and therapeutic gene expression, Cre recombination and CRISPR genome editing in normal and diseased mouse models. Enhanced transduction efficiency of AAV.cc47 vectors is further corroborated in macaques and pigs, providing a strong rationale for potential clinical translation into human gene therapies. We envision that ccAAV vectors may not only improve predictive modeling in preclinical studies, but also clinical translatability by broadening the therapeutic window of AAV based gene therapies.


Assuntos
Dependovirus , Edição de Genes , Animais , Dependovirus/metabolismo , Terapia Genética , Vetores Genéticos/genética , Humanos , Macaca/genética , Camundongos , Suínos , Transdução Genética
5.
Nat Commun ; 12(1): 6239, 2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34716331

RESUMO

Adeno-associated viruses (AAV) rely on helper viruses to transition from latency to lytic infection. Some AAV serotypes are secreted in a pre-lytic manner as free or extracellular vesicle (EV)-associated particles, although mechanisms underlying such are unknown. Here, we discover that the membrane-associated accessory protein (MAAP), expressed from a frameshifted open reading frame in the AAV cap gene, is a novel viral egress factor. MAAP contains a highly conserved, cationic amphipathic domain critical for AAV secretion. Wild type or recombinant AAV with a mutated MAAP start site (MAAPΔ) show markedly attenuated secretion and correspondingly, increased intracellular retention. Trans-complementation with MAAP restored secretion of multiple AAV/MAAPΔ serotypes. Further, multiple processing and analytical methods corroborate that one plausible mechanism by which MAAP promotes viral egress is through AAV/EV association. In addition to characterizing a novel viral egress factor, we highlight a prospective engineering platform to modulate secretion of AAV vectors or other EV-associated cargo.


Assuntos
Dependovirus/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Liberação de Vírus , Membrana Celular/química , Dependovirus/patogenicidade , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microrganismos Geneticamente Modificados/metabolismo , Domínios Proteicos , Proteínas Virais/química , Proteínas Virais/genética
6.
JCI Insight ; 5(19)2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32941184

RESUMO

Preexisting humoral immunity to recombinant adeno-associated virus (AAV) vectors restricts the treatable patient population and efficacy of human gene therapies. Approaches to clear neutralizing antibodies (NAbs), such as plasmapheresis and immunosuppression, are either ineffective or cause undesirable side effects. Here, we describe a clinically relevant strategy to rapidly and transiently degrade NAbs before AAV administration using an IgG-degrading enzyme (IdeZ). We demonstrate that recombinant IdeZ efficiently cleaved IgG in dog, monkey, and human antisera. Prophylactically administered IdeZ cleaved circulating human IgG in mice and prevented AAV neutralization in vivo. In macaques, a single intravenous dose of IdeZ rescued AAV transduction by transiently reversing seropositivity. Importantly, IdeZ efficiently cleaved NAbs and rescued AAV transduction in mice passively immunized with individual human donor sera representing a diverse population. Our antibody clearance approach presents a potentially new paradigm for expanding the prospective patient cohort and improving efficacy of AAV gene therapy.


Assuntos
Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Dependovirus/genética , Soros Imunes/química , Imunoglobulina G/metabolismo , Transdução Genética/métodos , Animais , Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Dependovirus/imunologia , Cães , Terapia Genética/métodos , Humanos , Soros Imunes/metabolismo , Imunidade Inata , Imunização Passiva , Imunoglobulina G/química , Macaca/imunologia , Camundongos , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa