Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.480
Filter
Add more filters

Publication year range
1.
Cell ; 187(13): 3236-3248.e21, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38772369

ABSTRACT

Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.


Subject(s)
Gene Regulatory Networks , Single-Cell Analysis , Animals , Female , Humans , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Genetic Vectors/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/cytology , Single-Cell Analysis/methods , Transcriptome/genetics , Cell Line , Transcription, Genetic
2.
Cell ; 187(1): 44-61.e17, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38134932

ABSTRACT

Cytokines employ downstream Janus kinases (JAKs) to promote chronic inflammatory diseases. JAK1-dependent type 2 cytokines drive allergic inflammation, and patients with JAK1 gain-of-function (GoF) variants develop atopic dermatitis (AD) and asthma. To explore tissue-specific functions, we inserted a human JAK1 GoF variant (JAK1GoF) into mice and observed the development of spontaneous AD-like skin disease but unexpected resistance to lung inflammation when JAK1GoF expression was restricted to the stroma. We identified a previously unrecognized role for JAK1 in vagal sensory neurons in suppressing airway inflammation. Additionally, expression of Calcb/CGRPß was dependent on JAK1 in the vagus nerve, and CGRPß suppressed group 2 innate lymphoid cell function and allergic airway inflammation. Our findings reveal evolutionarily conserved but distinct functions of JAK1 in sensory neurons across tissues. This biology raises the possibility that therapeutic JAK inhibitors may be further optimized for tissue-specific efficacy to enhance precision medicine in the future.


Subject(s)
Dermatitis, Atopic , Immunity, Innate , Lung , Sensory Receptor Cells , Animals , Humans , Mice , Cytokines , Dermatitis, Atopic/immunology , Inflammation , Lung/immunology , Lymphocytes , Sensory Receptor Cells/enzymology
3.
Cell ; 186(24): 5394-5410.e18, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37922901

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disorder. Its symptoms are typically treated with levodopa or dopamine receptor agonists, but its action lacks specificity due to the wide distribution of dopamine receptors in the central nervous system and periphery. Here, we report the development of a gene therapy strategy to selectively manipulate PD-affected circuitry. Targeting striatal D1 medium spiny neurons (MSNs), whose activity is chronically suppressed in PD, we engineered a therapeutic strategy comprised of a highly efficient retrograde adeno-associated virus (AAV), promoter elements with strong D1-MSN activity, and a chemogenetic effector to enable precise D1-MSN activation after systemic ligand administration. Application of this therapeutic approach rescues locomotion, tremor, and motor skill defects in both mouse and primate models of PD, supporting the feasibility of targeted circuit modulation tools for the treatment of PD in humans.


Subject(s)
Genetic Therapy , Parkinson Disease , Animals , Humans , Mice , Corpus Striatum/metabolism , Levodopa/therapeutic use , Levodopa/genetics , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/therapy , Primates , Receptors, Dopamine D1/metabolism , Disease Models, Animal
4.
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
5.
Cell ; 184(19): 4919-4938.e22, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34506722

ABSTRACT

Replacing or editing disease-causing mutations holds great promise for treating many human diseases. Yet, delivering therapeutic genetic modifiers to specific cells in vivo has been challenging, particularly in large, anatomically distributed tissues such as skeletal muscle. Here, we establish an in vivo strategy to evolve and stringently select capsid variants of adeno-associated viruses (AAVs) that enable potent delivery to desired tissues. Using this method, we identify a class of RGD motif-containing capsids that transduces muscle with superior efficiency and selectivity after intravenous injection in mice and non-human primates. We demonstrate substantially enhanced potency and therapeutic efficacy of these engineered vectors compared to naturally occurring AAV capsids in two mouse models of genetic muscle disease. The top capsid variants from our selection approach show conserved potency for delivery across a variety of inbred mouse strains, and in cynomolgus macaques and human primary myotubes, with transduction dependent on target cell expressed integrin heterodimers.


Subject(s)
Capsid/metabolism , Dependovirus/metabolism , Directed Molecular Evolution , Gene Transfer Techniques , Muscle, Skeletal/metabolism , Amino Acid Sequence , Animals , Capsid/chemistry , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Integrins/metabolism , Macaca fascicularis , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/therapy , Protein Multimerization , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/therapeutic use , RNA, Guide, Kinetoplastida/metabolism , Recombination, Genetic/genetics , Species Specificity , Transgenes
6.
Cell ; 184(21): 5465-5481.e16, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34582787

ABSTRACT

In vivo cell fate conversions have emerged as potential regeneration-based therapeutics for injury and disease. Recent studies reported that ectopic expression or knockdown of certain factors can convert resident astrocytes into functional neurons with high efficiency, region specificity, and precise connectivity. However, using stringent lineage tracing in the mouse brain, we show that the presumed astrocyte-converted neurons are actually endogenous neurons. AAV-mediated co-expression of NEUROD1 and a reporter specifically and efficiently induces reporter-labeled neurons. However, these neurons cannot be traced retrospectively to quiescent or reactive astrocytes using lineage-mapping strategies. Instead, through a retrograde labeling approach, our results reveal that endogenous neurons are the source for these viral-reporter-labeled neurons. Similarly, despite efficient knockdown of PTBP1 in vivo, genetically traced resident astrocytes were not converted into neurons. Together, our results highlight the requirement of lineage-tracing strategies, which should be broadly applied to studies of cell fate conversions in vivo.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Cell Lineage , Neurons/cytology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/pathology , Brain Injuries/pathology , Cell Line, Tumor , Cellular Reprogramming , Dependovirus/metabolism , Down-Regulation , Gene Expression Regulation , Genes, Reporter , Glial Fibrillary Acidic Protein/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homeodomain Proteins/metabolism , Humans , Integrases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
7.
Annu Rev Neurosci ; 45: 447-469, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35440143

ABSTRACT

Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types.


Subject(s)
Dependovirus , Genetic Vectors , Brain , Capsid/metabolism , Dependovirus/genetics , Gene Transfer Techniques
8.
Trends Biochem Sci ; 49(5): 457-469, 2024 May.
Article in English | MEDLINE | ID: mdl-38531696

ABSTRACT

Gene delivery vehicles based on adeno-associated viruses (AAVs) are enabling increasing success in human clinical trials, and they offer the promise of treating a broad spectrum of both genetic and non-genetic disorders. However, delivery efficiency and targeting must be improved to enable safe and effective therapies. In recent years, considerable effort has been invested in creating AAV variants with improved delivery, and computational approaches have been increasingly harnessed for AAV engineering. In this review, we discuss how computationally designed AAV libraries are enabling directed evolution. Specifically, we highlight approaches that harness sequences outputted by next-generation sequencing (NGS) coupled with machine learning (ML) to generate new functional AAV capsids and related regulatory elements, pushing the frontier of what vector engineering and gene therapy may achieve.


Subject(s)
Dependovirus , Gene Transfer Techniques , Dependovirus/genetics , Humans , Genetic Therapy/methods , Genetic Vectors/metabolism , Genetic Engineering , Animals , Computational Biology/methods
9.
Immunity ; 50(3): 567-575.e5, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30850342

ABSTRACT

Long-term delivery of anti-HIV monoclonal antibodies (mAbs) using adeno-associated virus (AAV) vectors holds promise for the prevention and treatment of HIV infection. We describe a therapy trial in which four rhesus monkeys were infected with SHIV-AD8 for 86 weeks before receiving the AAV-encoded mAbs 3BNC117, 10-1074, and 10E8. Although anti-drug antibody (ADA) responses restricted mAb delivery, one monkey successfully maintained 50-150 µg/mL of 3BNC117 and 10-1074 for over 2 years. Delivery of these two mAbs to this monkey resulted in an abrupt decline in plasma viremia, which remained undetectable for 38 successive measurements over 3 years. We generated two more examples of virologic suppression using AAV delivery of a cocktail of four mAbs in a 12-monkey study. Our results provide proof of concept for AAV-delivered mAbs to produce a "functional cure." However, they also serve as a warning that ADAs may be a problem for practical application of this approach in humans.


Subject(s)
Antibodies, Monoclonal/immunology , Dependovirus/immunology , HIV Infections/immunology , HIV-1/immunology , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Broadly Neutralizing Antibodies , Cell Line , HEK293 Cells , HIV Antibodies/immunology , Humans , Macaca mulatta , Viremia/immunology
10.
EMBO J ; 42(23): e114188, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37916874

ABSTRACT

Hyper IgM1 is an X-linked combined immunodeficiency caused by CD40LG mutations, potentially treatable with CD4+ T-cell gene editing with Cas9 and a "one-size-fits-most" corrective template. Contrary to established gene therapies, there is limited data on the genomic alterations following long-range gene editing, and no consensus on the relevant assays. We developed drop-off digital PCR assays for unbiased detection of large on-target deletions and found them at high frequency upon editing. Large deletions were also common upon editing different loci and cell types and using alternative Cas9 and template delivery methods. In CD40LG edited T cells, on-target deletions were counter-selected in culture and further purged by enrichment for edited cells using a selector coupled to gene correction. We then validated the sensitivity of optical genome mapping for unbiased detection of genome wide rearrangements and uncovered on-target trapping of one or more vector copies, which do not compromise functionality, upon editing using an integrase defective lentiviral donor template. No other recurring events were detected. Edited patient cells showed faithful reconstitution of CD40LG regulated expression and function with a satisfactory safety profile. Large deletions and donor template integrations should be anticipated and accounted for when designing and testing similar gene editing strategies.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Gene Editing/methods , Genome , T-Lymphocytes , CD4-Positive T-Lymphocytes
11.
Hum Mol Genet ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38757200

ABSTRACT

Gaucher Disease (GD) is an inherited metabolic disorder caused by mutations in the GBA1 gene. It can manifest with severe neurodegeneration and visceral pathology. The most acute neuronopathic form (nGD), for which there are no curative therapeutic options, is characterised by devastating neuropathology and death during infancy. In this study, we investigated the therapeutic benefit of systemically delivered AAV9 vectors expressing the human GBA1 gene at two different doses comparing a neuronal-selective promoter with ubiquitous promoters. Our results highlight the importance of a careful evaluation of the promoter sequence used in gene delivery vectors, suggesting a neuron-targeted therapy leading to high levels of enzymatic activity in the brain but lower GCase expression in the viscera, might be the optimal therapeutic strategy for nGD.

12.
Am J Hum Genet ; 110(10): 1648-1660, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37673065

ABSTRACT

X-linked myotubular myopathy (XLMTM) is a severe congenital disease characterized by profound muscle weakness, respiratory failure, and early death. No approved therapy for XLMTM is currently available. Adeno-associated virus (AAV)-mediated gene replacement therapy has shown promise as an investigational therapeutic strategy. We aimed to characterize the transcriptomic changes in muscle biopsies of individuals with XLMTM who received resamirigene bilparvovec (AT132; rAAV8-Des-hMTM1) in the ASPIRO clinical trial and to identify potential biomarkers that correlate with therapeutic outcome. We leveraged RNA-sequencing data from the muscle biopsies of 15 study participants and applied differential expression analysis, gene co-expression analysis, and machine learning to characterize the transcriptomic changes at baseline (pre-dose) and at 24 and 48 weeks after resamirigene bilparvovec dosing. As expected, MTM1 expression levels were significantly increased after dosing (p < 0.0001). Differential expression analysis identified upregulated genes after dosing that were enriched in several pathways, including lipid metabolism and inflammatory response pathways, and downregulated genes were enriched in cell-cell adhesion and muscle development pathways. Genes involved in inflammatory and immune pathways were differentially expressed between participants exhibiting ventilator support reduction of either greater or less than 6 h/day after gene therapy compared to pre-dosing. Co-expression analysis identified similarly regulated genes, which were grouped into modules. Finally, the machine learning model identified five genes, including MTM1, as potential RNA biomarkers to monitor the progress of AAV gene replacement therapy. These findings further extend our understanding of AAV-mediated gene therapy in individuals with XLMTM at the transcriptomic level.


Subject(s)
Myopathies, Structural, Congenital , Transcriptome , Humans , Biomarkers/metabolism , Gene Expression Profiling , Genetic Therapy , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Myopathies, Structural, Congenital/pathology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA/metabolism , Transcriptome/genetics
13.
Annu Rev Neurosci ; 41: 323-348, 2018 07 08.
Article in English | MEDLINE | ID: mdl-29709207

ABSTRACT

Recombinant viruses allow for targeted transgene expression in specific cell populations throughout the nervous system. The adeno-associated virus (AAV) is among the most commonly used viruses for neuroscience research. Recombinant AAVs (rAAVs) are highly versatile and can package most cargo composed of desired genes within the capsid's ∼5-kb carrying capacity. Numerous regulatory elements and intersectional strategies have been validated in rAAVs to enable cell type-specific expression. rAAVs can be delivered to specific neuronal populations or globally throughout the animal. The AAV capsids have natural cell type or tissue tropism and trafficking that can be modified for increased specificity. Here, we describe recently engineered AAV capsids and associated cargo that have extended the utility of AAVs in targeting molecularly defined neurons throughout the nervous system, which will further facilitate neuronal circuit interrogation and discovery.


Subject(s)
Central Nervous System/physiology , Genetic Engineering , Peripheral Nervous System/physiology , Animals , Dependovirus/genetics , Humans
14.
Proc Natl Acad Sci U S A ; 120(35): e2302997120, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37603759

ABSTRACT

Tissue macrophages, including microglia, are notoriously resistant to genetic manipulation. Here, we report the creation of Adeno-associated viruses (AAV) variants that efficiently and widely transduce microglia and tissue macrophages in vivo following intravenous delivery, with transgene expression of up to 80%. We use this technology to demonstrate manipulation of microglia gene expression and microglial ablation, thereby providing invaluable research tools for the study of these important cells.


Subject(s)
Dependovirus , Microglia , Dependovirus/genetics , Capsid , Transgenes , Macrophages
15.
Proc Natl Acad Sci U S A ; 120(23): e2303509120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252952

ABSTRACT

Vectored immunoprophylaxis was first developed as a means of establishing engineered immunity to HIV using an adenoassociated viral vector expressing a broadly neutralizing antibody. We applied this concept to establish long-term prophylaxis against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a mouse model using adenoassociated virus and lentiviral vectors expressing a high-affinity angiotensin-converting enzyme 2 (ACE2) decoy. Administration of decoy-expressing (adenoassociated virus) AAV2.retro and AAV6.2 vectors by intranasal instillation or intramuscular injection protected mice against high-titered SARS-CoV-2 infection. AAV and lentiviral vectored immunoprophylaxis was durable and was active against SARS-CoV-2 Omicron subvariants. The AAV vectors were also effective therapeutically when administered postinfection. Vectored immunoprophylaxis could be of value for immunocompromised individuals for whom vaccination is not practical and as a means to rapidly establish protection from infection. Unlike monoclonal antibody therapy, the approach is expected to remain active despite continued evolution viral variants.


Subject(s)
COVID-19 , Animals , Mice , COVID-19/prevention & control , SARS-CoV-2/genetics , Immunization , Immunotherapy , Vaccination , Dependovirus/genetics , Spike Glycoprotein, Coronavirus , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use
16.
J Biol Chem ; 300(3): 105732, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336290

ABSTRACT

The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.


Subject(s)
Amino Acid Substitution , Cation Transport Proteins , Dependovirus , Liver , Manganese , Mutation , Animals , Mice , Body Weight , Brain/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Dependovirus/genetics , Erythrocytes , Genome-Wide Association Study , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Manganese/metabolism , Manganese Poisoning/metabolism , Phenotype , Promoter Regions, Genetic , Thyroxine-Binding Globulin/genetics
17.
Annu Rev Med ; 74: 231-247, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36103998

ABSTRACT

In vivo gene therapy is rapidly emerging as a new therapeutic paradigm for monogenic disorders. For almost three decades, hemophilia A (HA) and hemophilia B (HB) have served as model disorders for the development of gene therapy. This effort is soon to bear fruit with completed pivotal adeno-associated viral (AAV) vector gene addition trials reporting encouraging results and regulatory approval widely anticipated in the near future for the current generation of HA and HB AAV vectors. Here we review the clinical development of AAV gene therapy for HA and HB and examine outstanding questions that have recently emerged from AAV clinical trials for hemophilia and other monogenic disorders.


Subject(s)
Hemophilia A , Hemophilia B , Humans , Hemophilia A/genetics , Hemophilia A/therapy , Dependovirus/genetics , Factor IX/genetics , Genetic Vectors , Hemophilia B/genetics , Hemophilia B/therapy , Genetic Therapy/methods
18.
J Virol ; 98(6): e0063324, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38775479

ABSTRACT

Adeno-associated viruses (AAVs) package a single-stranded (ss) DNA genome of 4.7 kb in their capsid of ~20 nm in diameter. AAV replication requires co-infection of a helper virus, such as adenovirus. During the optimization of recombinant AAV production, a small viral nonstructural protein, membrane-associated accessory protein (MAAP), was identified. However, the function of the MAAP in the context of AAV infection remains unknown. Here, we investigated the expression strategy and function of the MAAP during infection of both AAV2 and AAV5 in human embryonic kidney (HEK)293 cells. We found that AAV2 MAAP2 and AAV5 MAAP5 are expressed from the capsid gene (cap)-transcribing mRNA spliced from the donor to the second splice site that encodes VP2 and VP3. Thus, this AAV cap gene transcribes a multicistronic mRNA that can be translated to four viral proteins, MAAP, VP2, AAP, and VP3 in order. In AAV2 infection, MAAP2 predominantly localized in the cytoplasm, alongside the capsid, near the nuclear and plasma membranes, but a fraction of MAAP2 exhibited nuclear localization. In AAV5 infection, MAAP5 revealed a distinct pattern, predominantly localizing within the nucleus. In the cells infected with an MAAP knockout mutant of AAV2 or AAV5, both viral DNA replication and virus replication increased, whereas virus egress decreased, and the decrease in virus egress can be restored by providing MAAP in trans. In summary, MAAP, a novel AAV nonstructural protein translated from a multicistronic viral cap mRNA, not only facilitates cellular egress of AAV but also likely negatively affects viral DNA replication during infection. IMPORTANCE: Recombinant adeno-associated virus (rAAV) has been used as a gene delivery vector in clinical gene therapy. In current gene therapies employing rAAV, a high dose of the vector is required. Consequently, there is a high demand for efficient and high-purity vector production systems. In this study, we demonstrated that membrane-associated accessory protein (MAAP), a small viral nonstructural protein, is translated from the same viral mRNA transcript encoding VP2 and VP3. In AAV-infected cells, apart from its prevalent expression in the cytoplasm with localization near the plasma and nuclear membranes, the MAAP also exhibits notable localization within the nucleus. During AAV infection, MAAP expression increases the cellular egress of progeny virions and decreases viral DNA replication and progeny virion production. Thus, the choice of MAAP expression has pros and cons during AAV infection, which could provide a guide to rAAV production.


Subject(s)
Dependovirus , Parvoviridae Infections , Viral Nonstructural Proteins , Humans , Capsid Proteins/genetics , Dependovirus/genetics , Dependovirus/metabolism , Dependovirus/physiology , HEK293 Cells , Parvoviridae Infections/virology , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Virus Replication , Genes, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
19.
J Pathol ; 263(1): 22-31, 2024 05.
Article in English | MEDLINE | ID: mdl-38332723

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive scarring disease of the lung that leads rapidly to respiratory failure. Novel approaches to treatment are urgently needed. The bioactive lipid sphingosine-1-phosphate (S1P) is increased in IPF lungs and promotes proinflammatory and profibrotic TGF-ß signaling. Hence, decreasing lung S1P represents a potential therapeutic strategy for IPF. S1P is degraded by the intracellular enzyme S1P lyase (SPL). Here we find that a knock-in mouse with a missense SPL mutation mimicking human disease resulted in reduced SPL activity, increased S1P, increased TGF-ß signaling, increased lung fibrosis, and higher mortality after injury compared to wild type (WT). We then tested adeno-associated virus 9 (AAV9)-mediated overexpression of human SGPL1 (AAV-SPL) in mice as a therapeutic modality. Intravenous treatment with AAV-SPL augmented lung SPL activity, attenuated S1P levels within the lungs, and decreased injury-induced fibrosis compared to controls treated with saline or only AAV. We confirmed that AAV-SPL treatment led to higher expression of SPL in the epithelial and fibroblast compartments during bleomycin-induced lung injury. Additionally, AAV-SPL decreased expression of the profibrotic cytokines TNFα and IL1ß as well as markers of fibroblast activation, such as fibronectin (Fn1), Tgfb1, Acta2, and collagen genes in the lung. Taken together, our results provide proof of concept for the use of AAV-SPL as a therapeutic strategy for the treatment of IPF. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Dependovirus , Idiopathic Pulmonary Fibrosis , Lysophospholipids , Sphingosine/analogs & derivatives , Humans , Mice , Animals , Dependovirus/genetics , Lung/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/therapy , Idiopathic Pulmonary Fibrosis/metabolism , Bleomycin , Models, Animal , Genetic Therapy , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism
20.
Brain ; 147(4): 1457-1473, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38066620

ABSTRACT

Acyl-CoA binding domain containing 5 (ACBD5) is a critical player in handling very long chain fatty acids (VLCFA) en route for peroxisomal ß-oxidation. Mutations in ACBD5 lead to the accumulation of VLCFA and patients present retinal dystrophy, ataxia, psychomotor delay and a severe leukodystrophy. Using CRISPR/Cas9, we generated and characterized an Acbd5 Gly357* mutant allele. Gly357* mutant mice recapitulated key features of the human disorder, including reduced survival, impaired locomotion and reflexes, loss of photoreceptors, and demyelination. The ataxic presentation of Gly357* mice involved the loss of cerebellar Purkinje cells and a giant axonopathy throughout the CNS. Lipidomic studies provided evidence for the extensive lipid dysregulation caused by VLCFA accumulation. Following a proteomic survey, functional studies in neurons treated with VLCFA unravelled a deregulated cytoskeleton with reduced actin dynamics and increased neuronal filopodia. We also show that an adeno-associated virus-mediated gene delivery ameliorated the gait phenotypes and the giant axonopathy, also improving myelination and astrocyte reactivity. Collectively, we established a mouse model with significance for VLCFA-related disorders. The development of relevant neuropathological outcomes enabled the understanding of mechanisms modulated by VLCFA and the evaluation of the efficacy of preclinical therapeutic interventions.


Subject(s)
Adrenoleukodystrophy , Fatty Acids , Humans , Mice , Animals , Fatty Acids/metabolism , Dependovirus/genetics , Proteomics , Ataxia , Genetic Therapy , Adrenoleukodystrophy/genetics
SELECTION OF CITATIONS
SEARCH DETAIL