Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther ; 32(7): 2080-2093, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38715362

ABSTRACT

Systemic dosing of adeno-associated viral (AAV) vectors poses potential risk of adverse side effects including complement activation triggered by anti-capsid immunity. Due to the multifactorial nature of toxicities observed in this setting, a wide spectrum of immune modulatory regimens are being investigated in the clinic. Here, we discover an IgM cleaving enzyme (IceM) that degrades human IgM, a key trigger in the anti-AAV immune cascade. We then engineer a fusion enzyme (IceMG) with dual proteolytic activity against human IgM and IgG. IceMG cleaves B cell surface antigen receptors and inactivates phospholipase gamma signaling in vitro. Importantly, IceMG is more effective at inhibiting complement activation compared with an IgG cleaving enzyme alone. Upon IV dosing, IceMG rapidly and reversibly clears circulating IgM and IgG in macaques. Antisera from these animals treated with IceMG shows decreased ability to neutralize AAV and activate complement. Consistently, pre-conditioning with IceMG restores AAV transduction in mice passively immunized with human antisera. Thus, IgM cleaving enzymes show promise in simultaneously addressing multiple aspects of anti-AAV immunity mediated by B cells, circulating antibodies and complement. These studies have implications for improving safety of AAV gene therapies and possibly broader applications including organ transplantation and autoimmune diseases.


Subject(s)
Complement Activation , Dependovirus , Genetic Vectors , Immunoglobulin G , Immunoglobulin M , Dependovirus/genetics , Dependovirus/immunology , Animals , Immunoglobulin M/immunology , Humans , Immunoglobulin G/immunology , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Antibodies, Neutralizing/immunology , Transduction, Genetic , Gene Transfer Techniques , Antibodies, Viral/immunology , Proteolysis , Genetic Therapy/methods , Protein Engineering
2.
Cell Rep ; 43(3): 113902, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38431840

ABSTRACT

Adeno-associated virus (AAV) is a member of the genus Dependoparvovirus, which infects a wide range of vertebrate species. Here, we observe that, unlike most primate AAV isolates, avian AAV is transcriptionally silenced in human cells. By swapping the VP1 N terminus from primate AAVs (e.g., AAV8) onto non-mammalian isolates (e.g., avian AAV), we identify a minimal component of the AAV capsid that controls viral transcription and unlocks robust transduction in both human cells and mouse tissue. This effect is accompanied by increased AAV genome chromatin accessibility and altered histone methylation. Proximity ligation analysis reveals that host factors are selectively recruited by the VP1 N terminus of AAV8 but not avian AAV. Notably, these include AAV essential factors implicated in the nuclear factor κB pathway, chromatin condensation, and histone methylation. We postulate that the AAV capsid has evolved mechanisms to recruit host factors to its genome, allowing transcriptional activation in a species-specific manner.


Subject(s)
Capsid , Dependovirus , Humans , Animals , Mice , Capsid/metabolism , Dependovirus/metabolism , Histones/metabolism , Viral Transcription , Genetic Vectors , Capsid Proteins/genetics , Capsid Proteins/metabolism , Primates , Host Specificity , Chromatin/metabolism
3.
Nat Commun ; 15(1): 2325, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38485709

ABSTRACT

Type VI CRISPR enzymes have been developed as programmable RNA-guided Cas proteins for eukaryotic RNA editing. Notably, Cas13 has been utilized for site-targeted single base edits, demethylation, RNA cleavage or knockdown and alternative splicing. However, the ability to edit large stretches of mRNA transcripts remains a significant challenge. Here, we demonstrate that CRISPR-Cas13 systems can be repurposed to assist trans-splicing of exogenous RNA fragments into an endogenous pre-mRNA transcript, a method termed CRISPR Assisted mRNA Fragment Trans-splicing (CRAFT). Using split reporter-based assays, we evaluate orthogonal Cas13 systems, optimize guide RNA length and screen for optimal trans-splicing site(s) across a range of intronic targets. We achieve markedly improved editing of large 5' and 3' segments in different endogenous mRNAs across various mammalian cell types compared to other spliceosome-mediated trans-splicing methods. CRAFT can serve as a versatile platform for attachment of protein tags, studying the impact of multiple mutations/single nucleotide polymorphisms, modification of untranslated regions (UTRs) or replacing large segments of mRNA transcripts.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , RNA, Guide, CRISPR-Cas Systems , RNA, Messenger/genetics , Trans-Splicing , RNA/genetics , Mammals/genetics
4.
Nat Commun ; 15(1): 1955, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38438373

ABSTRACT

Clinical translation of AAV-mediated gene therapy requires preclinical development across different experimental models, often confounded by variable transduction efficiency. Here, we describe a human liver chimeric transgene-free Il2rg-/-/Rag2-/-/Fah-/-/Aavr-/- (TIRFA) mouse model overcoming this translational roadblock, by combining liver humanization with AAV receptor (AAVR) ablation, rendering murine cells impermissive to AAV transduction. Using human liver chimeric TIRFA mice, we demonstrate increased transduction of clinically used AAV serotypes in primary human hepatocytes compared to humanized mice with wild-type AAVR. Further, we demonstrate AAV transduction in human teratoma-derived primary cells and liver cancer tissue, displaying the versatility of the humanized TIRFA mouse. From a mechanistic perspective, our results support the notion that AAVR functions as both an entry receptor and an intracellular receptor essential for transduction. The TIRFA mouse should allow prediction of AAV gene transfer efficiency and the study of AAV vector biology in a preclinical human setting.


Subject(s)
Dependovirus , Liver , Humans , Animals , Mice , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Hepatocytes
5.
Sci Transl Med ; 15(692): eadf4086, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37075130

ABSTRACT

Glutaric aciduria type I (GA-1) is an inborn error of metabolism with a severe neurological phenotype caused by the deficiency of glutaryl-coenzyme A dehydrogenase (GCDH), the last enzyme of lysine catabolism. Current literature suggests that toxic catabolites in the brain are produced locally and do not cross the blood-brain barrier. In a series of experiments using knockout mice of the lysine catabolic pathway and liver cell transplantation, we uncovered that toxic GA-1 catabolites in the brain originated from the liver. Moreover, the characteristic brain and lethal phenotype of the GA-1 mouse model was rescued by two different liver-directed gene therapy approaches: Using an adeno-associated virus, we replaced the defective Gcdh gene or we prevented flux through the lysine degradation pathway by CRISPR deletion of the aminoadipate-semialdehyde synthase (Aass) gene. Our findings question the current pathophysiological understanding of GA-1 and reveal a targeted therapy for this devastating disorder.


Subject(s)
Glutaryl-CoA Dehydrogenase , Lysine , Animals , Mice , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Lysine/metabolism , Mice, Knockout , Liver/metabolism
6.
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
7.
Sci Rep ; 10(1): 4345, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32132565

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Sci Rep ; 9(1): 15857, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676833

ABSTRACT

HIV-associated nephropathy (HIVAN) is a rapidly progressive kidney disease that is caused by HIV infection of renal epithelial cells with subsequent expression of viral genes, including vpr. Antiretroviral therapy ameliorates HIVAN without eradicating HIV from the kidneys and the mechanism by which it protects kidneys is poorly understood. Since HIV protease inhibitors have "off target" cellular effects, we studied whether darunavir, the most commonly prescribed protease inhibitor, protects kidneys from HIV-induced injury via mechanisms independent of HIV protease and viral replication. Renal epithelial cells were transduced with lentiviruses encoding HIV (lacking protease and reverse transcriptase), Vpr, or vector control. Darunavir attenuated HIV and Vpr-induced activation of Stat3, Src, Erk, and cytokines, which are critical for HIVAN pathogenesis. We then studied HIV-transgenic mice, which develop HIVAN in the absence of HIV protease or reverse transcriptase. Mice were treated with darunavir, zidovudine, darunavir + zidovudine, or control. Darunavir and darunavir + zidovudine reduced albuminuria and histologic kidney injury and normalized expression of dysregulated proteins. RNA-seq analyses demonstrated that darunavir suppressed HIV-induced upregulation of immune response genes in human kidney cells. These data demonstrate that darunavir protects against HIV-induced renal injury via mechanisms that are independent of inhibition of HIV protease.


Subject(s)
AIDS-Associated Nephropathy/prevention & control , Darunavir/pharmacology , HIV Protease Inhibitors/pharmacology , HIV-1/metabolism , Kidney/metabolism , MAP Kinase Signaling System/drug effects , AIDS-Associated Nephropathy/metabolism , AIDS-Associated Nephropathy/pathology , Animals , Cell Line , Humans , Kidney/pathology , Kidney/virology , Mice , Mice, Transgenic , Zidovudine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...