Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cell ; 186(2): 446-460.e19, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638795

ABSTRACT

Precise targeting of large transgenes to T cells using homology-directed repair has been transformative for adoptive cell therapies and T cell biology. Delivery of DNA templates via adeno-associated virus (AAV) has greatly improved knockin efficiencies, but the tropism of current AAV serotypes restricts their use to human T cells employed in immunodeficient mouse models. To enable targeted knockins in murine T cells, we evolved Ark313, a synthetic AAV that exhibits high transduction efficiency in murine T cells. We performed a genome-wide knockout screen and identified QA2 as an essential factor for Ark313 infection. We demonstrate that Ark313 can be used for nucleofection-free DNA delivery, CRISPR-Cas9-mediated knockouts, and targeted integration of large transgenes. Ark313 enables preclinical modeling of Trac-targeted CAR-T and transgenic TCR-T cells in immunocompetent models. Efficient gene targeting in murine T cells holds great potential for improved cell therapies and opens avenues in experimental T cell immunology.


Subject(s)
Dependovirus , Genetic Engineering , T-Lymphocytes , Animals , Mice , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Gene Targeting , Genetic Engineering/methods
2.
J Virol ; 95(19): e0058721, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34232726

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Dependovirus/metabolism , Directed Molecular Evolution , Receptors, Virus/metabolism , Amino Acid Motifs , CRISPR-Cas Systems , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cryoelectron Microscopy , Dependovirus/chemistry , Dependovirus/ultrastructure , Genetic Variation , Glycosaminoglycans/metabolism , Humans , Integrin beta1/chemistry , Integrin beta1/metabolism , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/chemistry , Virus Internalization
3.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817219

ABSTRACT

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.


Subject(s)
Calcium-Transporting ATPases/genetics , Calcium/metabolism , Dependovirus/genetics , Genetic Vectors/metabolism , Golgi Apparatus/metabolism , Animals , Animals, Newborn , Biological Transport/drug effects , Brain/drug effects , Brain/metabolism , CRISPR-Cas Systems , Calcium-Transporting ATPases/deficiency , Cell Line, Tumor , Chelating Agents/pharmacology , Dependovirus/drug effects , Dependovirus/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Gene Deletion , Genetic Vectors/chemistry , Golgi Apparatus/drug effects , Golgi Apparatus/virology , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Injections, Intraventricular , Ionomycin/pharmacology , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Inbred C57BL , Stereotaxic Techniques , Transduction, Genetic , Vesiculovirus/genetics , Vesiculovirus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...