Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.709
Filter
1.
Science ; 384(6703): ado7082, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38935715

ABSTRACT

Prion disease is caused by misfolding of the prion protein (PrP) into pathogenic self-propagating conformations, leading to rapid-onset dementia and death. However, elimination of endogenous PrP halts prion disease progression. In this study, we describe Coupled Histone tail for Autoinhibition Release of Methyltransferase (CHARM), a compact, enzyme-free epigenetic editor capable of silencing transcription through programmable DNA methylation. Using a histone H3 tail-Dnmt3l fusion, CHARM recruits and activates endogenous DNA methyltransferases, thereby reducing transgene size and cytotoxicity. When delivered to the mouse brain by systemic injection of adeno-associated virus (AAV), Prnp-targeted CHARM ablates PrP expression across the brain. Furthermore, we have temporally limited editor expression by implementing a kinetically tuned self-silencing approach. CHARM potentially represents a broadly applicable strategy to suppress pathogenic proteins, including those implicated in other neurodegenerative diseases.


Subject(s)
Brain , DNA Methylation , Dependovirus , Epigenesis, Genetic , Gene Silencing , Histones , Prion Proteins , Animals , Dependovirus/genetics , Mice , Brain/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Histones/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Prion Diseases/genetics , Prion Diseases/metabolism , Humans , Transgenes
2.
Curr Protoc ; 4(6): e1091, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923413

ABSTRACT

This article describes a step-by-step process of lumbar intrathecal injection of Evans blue dye and AAV9-EGFP in adult (2-month-old) and neonatal (postnatal day 10) mice. Intrathecal injection is a clinically translatable technique that has already been extensively applied in humans. In mice, intrathecal injection is considered a challenging procedure that requires a trained and experienced researcher. For both adult and neonatal mice, lumbar intrathecal injection is directed into the L5-L6 intervertebral space. Intrathecally injected material enters the cerebrospinal fluid (CSF) within the intrathecal space from where it can directly access the central nervous system (CNS) parenchyma. Simultaneously, intrathecally injected material exits the CSF with pressure gradient and enters the endoneurial fluid and ultimately the peripheral nerves. While in the CSF, the injectable material also enters the bloodstream and systemic circulation through the arachnoid villi. A successful lumbar intrathecal injection results in adequate biodistribution of the injectable material in the CNS, PNS, and peripheral organs. When correctly applied, this technique is considered as minimally invasive and non-disruptive and can be used for the lumbar delivery of any solute. © 2024 Wiley Periodicals LLC. Basic Protocol 1: C57BL/6 adult and P10 mice lumbar intrathecal injection Basic Protocol 2: Tissue collection and preparation for evaluating Evans blue dye diffusion Basic Protocol 3: Tissue collection and preparation for immunohistochemistry staining Basic Protocol 4: Tissue collection and vector genome copy number analysis.


Subject(s)
Animals, Newborn , Injections, Spinal , Mice, Inbred C57BL , Animals , Mice , Evans Blue/administration & dosage , Evans Blue/pharmacokinetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Dependovirus/genetics , Genetic Vectors/administration & dosage
3.
Methods Mol Biol ; 2810: 55-74, 2024.
Article in English | MEDLINE | ID: mdl-38926272

ABSTRACT

Here, we describe methods for the production of adeno-associated viral (AAV) vectors by transient transfection of HEK293 cells grown in serum-free medium using orbital shaken bioreactors and the subsequent purification of vector particles. The protocol for expression of AAV components is based on polyethyleneimine (PEI)-mediated transfection of a three-plasmid system and is specified for production in milliliter-to-liter scales. After PEI and plasmid DNA (pDNA) complex formation, the diluted cell culture is transfected without a prior concentration step or medium exchange. Following a 7-day batch process, cell cultures are further processed using a set of methods for cell lysis and vector recovery. Methods for the purification of viral particles are described, including immunoaffinity and anion-exchange chromatography, ultrafiltration, as well as digital PCR to quantify the concentration of vector particles.


Subject(s)
Dependovirus , Genetic Vectors , Transfection , Humans , Dependovirus/genetics , Dependovirus/isolation & purification , HEK293 Cells , Genetic Vectors/genetics , Genetic Vectors/isolation & purification , Transfection/methods , Plasmids/genetics , Plasmids/isolation & purification , Polyethyleneimine/chemistry , Bioreactors , Chromatography, Ion Exchange/methods , Virion/genetics , Virion/isolation & purification
4.
Genes (Basel) ; 15(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38927645

ABSTRACT

With the rapid development of gene therapy technology in recent years, its abuse as a method of sports doping in athletics has become a concern. However, there is still room for improvement in gene-doping testing methods, and a robust animal model needs to be developed. Therefore, the purposes of this study were to establish a model of gene doping using recombinant adeno-associated virus vector-9, including the human erythropoietin gene (rAAV9-hEPO), and to establish a relevant testing method. First, it was attempted to establish the model using rAAV9-hEPO on mice. The results showed a significant increase in erythrocyte volume accompanied by an increase in spleen weight, confirming the validity of the model. Next, we attempted to detect proof of gene doping by targeting DNA and RNA. Direct proof of gene doping was detected using a TaqMan-qPCR assay with certain primers/probes. In addition, some indirect proof was identified in RNAs through the combination of a TB Green qPCR assay with RNA sequencing. Taken together, these results could provide the foundation for an effective test for gene doping in human athletes in the future.


Subject(s)
Dependovirus , Doping in Sports , Erythropoietin , Genetic Vectors , Erythropoietin/genetics , Animals , Mice , Doping in Sports/methods , Dependovirus/genetics , Humans , Genetic Vectors/genetics , Male , Genetic Therapy/methods , Models, Animal
5.
Genes (Basel) ; 15(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38927656

ABSTRACT

Gene therapy holds promise as a transformative approach in the treatment landscape of age-related macular degeneration (AMD), diabetic retinopathy (DR), and diabetic macular edema (DME), aiming to address the challenges of frequent intravitreal anti-vascular endothelial growth factor (VEGF) injections. This manuscript reviews ongoing gene therapy clinical trials for these disorders, including ABBV-RGX-314, ixoberogene soroparvovec (ixo-vec), and 4D-150. ABBV-RGX-314 utilizes an adeno-associated virus (AAV) vector to deliver a transgene encoding a ranibizumab-like anti-VEGF antibody fragment, demonstrating promising results in Phase 1/2a and ongoing Phase 2b/3 trials. Ixo-vec employs an AAV2.7m8 capsid for intravitreal delivery of a transgene expressing aflibercept, showing encouraging outcomes in Phase 1 and ongoing Phase 2 trials. 4D-150 utilizes an evolved vector to express both aflibercept and a VEGF-C inhibitory RNAi, exhibiting positive interim results in Phase 1/2 studies. Other therapies reviewed include EXG102-031, FT-003, KH631, OLX10212, JNJ-1887, 4D-175, and OCU410. These therapies offer potential advantages of reduced treatment frequency and enhanced safety profiles, representing a paradigm shift in management towards durable and efficacious cellular-based biofactories. These advancements in gene therapy hold promise for improving outcomes in AMD and addressing the complex challenges of DME and DR, providing new avenues for the treatment of diabetic eye diseases.


Subject(s)
Diabetic Retinopathy , Genetic Therapy , Macular Degeneration , Humans , Diabetic Retinopathy/therapy , Diabetic Retinopathy/genetics , Genetic Therapy/methods , Macular Degeneration/therapy , Macular Degeneration/genetics , Genetic Vectors/genetics , Dependovirus/genetics , Vascular Endothelial Growth Factor A/genetics , Animals
6.
Nat Commun ; 15(1): 4531, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866749

ABSTRACT

Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.


Subject(s)
Autism Spectrum Disorder , CA1 Region, Hippocampal , Gene Editing , Memory , Mice, Knockout , Nerve Tissue Proteins , Social Behavior , Animals , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , CA1 Region, Hippocampal/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Mice , Memory/physiology , Neurons/metabolism , Dependovirus/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Inbred C57BL
8.
Nat Commun ; 15(1): 4728, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830864

ABSTRACT

Due to their exceptional solubility and stability, nanobodies have emerged as powerful building blocks for research tools and therapeutics. However, their generation in llamas is cumbersome and costly. Here, by inserting an engineered llama immunoglobulin heavy chain (IgH) locus into IgH-deficient mice, we generate a transgenic mouse line, which we refer to as 'LamaMouse'. We demonstrate that LamaMice solely express llama IgH molecules without association to Igκ or λ light chains. Immunization of LamaMice with AAV8, the receptor-binding domain of the SARS-CoV-2 spike protein, IgE, IgG2c, and CLEC9A enabled us to readily select respective target-specific nanobodies using classical hybridoma and phage display technologies, single B cell screening, and direct cloning of the nanobody-repertoire into a mammalian expression vector. Our work shows that the LamaMouse represents a flexible and broadly applicable platform for a facilitated selection of target-specific nanobodies.


Subject(s)
Camelids, New World , Immunoglobulin Heavy Chains , Mice, Transgenic , Single-Domain Antibodies , Spike Glycoprotein, Coronavirus , Animals , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Camelids, New World/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Immunoglobulin E/immunology , Humans , Dependovirus/genetics , Dependovirus/immunology , Immunoglobulin G/immunology , COVID-19/immunology , B-Lymphocytes/immunology
9.
Curr Protoc ; 4(6): e1068, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837274

ABSTRACT

Adeno-associated virus (AAV) vectors can efficiently transduce exogenous genes into various tissues in vivo. Owing to their convenience, high efficiency, long-term stable gene expression, and minimal side effects, AAV vectors have become one of the gold standards for investigating gene functions in vivo, especially in non-clinical studies. However, challenges persist in efficiently preparing a substantial quantity of high-quality AAV vectors. Commercial AAV vectors are typically associated with high costs. Further, in-laboratory production is hindered by the lack of specific laboratory equipment, such as ultracentrifuges. Therefore, a simple, quick, and scalable preparation method for AAV vectors is needed for proof-of-concept experiments. Herein, we present an optimized method for producing and purifying high-quality AAV serotype 9 (AAV9) vectors using standard laboratory equipment and chromatography. Using ceramic hydroxyapatite as a mixed-mode chromatography medium can markedly increase the quality of purified AAV vectors. Basic Protocols and optional methods for evaluating purified AAV vectors are also described. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Production of AAV9 vectors in 293EB cells Basic Protocol 2: Concentration and buffer exchange of AAV9 vectors from 293EB cell culture supernatants using tangential flow filtration Basic Protocol 3: Purification of AAV9 vectors from TFF samples using ceramic hydroxyapatite chromatography Basic Protocol 4: Analysis of the purified AAV9 vectors.


Subject(s)
Ceramics , Dependovirus , Durapatite , Genetic Vectors , Serogroup , Dependovirus/genetics , Dependovirus/isolation & purification , Genetic Vectors/isolation & purification , Genetic Vectors/genetics , Humans , Ceramics/chemistry , Durapatite/chemistry , Chromatography/methods , HEK293 Cells
10.
Protein Sci ; 33(7): e5074, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38888268

ABSTRACT

Adeno-associated virus (AAV), a widely used gene therapy vector, is a small, nonenveloped virus that contains a single-stranded DNA genome with a maximum length of 4.7 kb. Despite extensive biophysical and structural characterization, many aspects of AAV functions remain elusive. This knowledge gap is primarily due to a lack of structurally resolved dynamic information and the absence of structural coverage of functionally critical segments on the AAV capsid. Here, we developed a protocol to study AAV structural dynamics by hydrogen-deuterium exchange mass spectrometry (HDX-MS), a powerful method for monitoring protein structure stability and dynamics in solution. We performed HDX-MS measurements on AAVs without or with different DNA payloads of different sizes, and obtained detailed dynamic information on the entire AAV sequence including the two functionally important segments not previously structurally characterized. The unique N terminus of the capsid protein VP1 (VP1u) was found to adopt a highly dynamic and unstable conformation with low HDX protection across the entire region, whereas the presence of a DNA payload increased its protection. The VP1 and VP2 shared region (VP1/2) showed no measurable protection, with or without DNA. Differential HDX between empty and full capsid samples allowed us to identify potential new DNA-capsid interaction sites located primarily around the five-fold channel, which differ from the three-fold pocket binding site previously identified. Our HDX-MS method for characterizing AAV structural dynamics opens a new way for future efforts to understand AAV structure-function relationships and engineer next-generation AAV vectors with improved gene delivery properties.


Subject(s)
Capsid Proteins , Capsid , Dependovirus , Genetic Therapy , Genetic Vectors , Dependovirus/genetics , Dependovirus/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genetic Vectors/genetics , Genetic Therapy/methods , Capsid/chemistry , Capsid/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry , Protein Stability , Humans , Protein Conformation , Models, Molecular
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(5): 818-826, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38862439

ABSTRACT

OBJECTIVE: To investigate the effects of an adeno-associated virus (AAV2) vector expressing secretory transforming growth factor-ß (TGF-ß) type Ⅱ receptor (sTßRⅡ) extracellular domain-IgG2a Fc fusion protein (sTßRⅡ-Fc) on proliferation and migration of triple-negative murine breast cancer 4T1 cells in mice. METHODS: The pAAV-sTßRⅡ-Fc vector expressing sTßRⅡ-Fc fusion protein constructed by molecular cloning, the capsid protein-expressing vector pAAV2 and the helper vector were co-transfected into HEK 293T cells to prepare the recombinant AAV2-sTßRⅡ virus, which was purified by density gradient centrifugation with iodixanol. Western blotting was used to examine the effects of AAV-sTßRⅡ virus on Smad2/3 phosphorylation in 4T1 cells and on expression levels of E-cadherin, vimentin and p-Smad2/3 in 4T1 cell xenografts in mice. BALB/c mice bearing subcutaneous xenografts of luciferase-expressing 4T1 cells received intravenous injections of AAV-sTßRⅡ virus, AAV-GFP virus or PBS (n=6) through the tail vein, and the proliferation and migration of 4T1 cells were analyzed with in vivo imaging. Ki67 expression in the tumor tissues and sTßRⅡ protein expressions in mouse livers were detected with immunohistochemistry and immunofluorescence staining, and tumor metastases in the vital organs were examined with HE staining. RESULTS: The recombinant pAAV-sTßRⅡ-Fc vector successfully expressed sTßRⅡ in HEK 293T cells. Infection with AAV2-sTßRⅡ virus significantly reduced TGF-ß1-induced Smad2/3 phosphorylation in 4T1 cells and effectively inhibited proliferation and lung metastasis of 4T1 xenografts in mice (P<0.05). In the tumor-bearing mice, intravenous injection of AAV-sTßRⅡ virus significantly increased E-cadherin expression, reduced vimentin and Ki67 protein expressions and Smad2/3 phosphorylation level in the tumor tissues (P<0.05 or 0.01), and induced liver-specific sTßRⅡ expression without causing body weight loss or heart, liver, spleen or kidney pathologies. CONCLUSION: The recombinant AVV2 vector encoding sTßRⅡ extracellular domain is capable of blocking the TGF-ß signaling pathway to inhibit the proliferation and lung metastasis of 4T1 cells in mice.


Subject(s)
Cell Proliferation , Dependovirus , Genetic Vectors , Lung Neoplasms , Mice, Inbred BALB C , Receptor, Transforming Growth Factor-beta Type II , Animals , Mice , Dependovirus/genetics , Humans , HEK293 Cells , Lung Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Female , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cell Line, Tumor , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Cadherins/metabolism , Cadherins/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Cell Movement , Smad2 Protein/metabolism , Smad2 Protein/genetics
12.
J Cell Mol Med ; 28(11): e18460, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38864710

ABSTRACT

Haemophilic arthropathy (HA), a common comorbidity in haemophilic patients leads to joint pain, deformity and reduced quality of life. We have recently demonstrated that a long non-coding RNA, Neat1 as a primary regulator of matrix metalloproteinase (MMP) 3 and MMP13 activity, and its induction in the target joint has a deteriorating effect on articular cartilage. In the present study, we administered an Adeno-associated virus (AAV) 5 vector carrying an short hairpin (sh)RNA to Neat1 via intra-articular injection alone or in conjunction with systemic administration of a capsid-modified AAV8 (K31Q) vector carrying F8 gene (F8-BDD-V3) to study its impact on HA. AAV8K31Q-F8 vector administration at low dose, led to an increase in FVIII activity (16%-28%) in treated mice. We further observed a significant knockdown of Neat1 (~40 fold vs. untreated injured joint, p = 0.005) in joint tissue of treated mice and a downregulation of chondrodegenerative enzymes, MMP3, MMP13 and the inflammatory mediator- cPLA2, in mice receiving combination therapy. These data demonstrate that AAV mediated Neat1 knockdown in combination with F8 gene augmentation can potentially impact mediators of haemophilic joint disease.


Subject(s)
Dependovirus , Factor VIII , Genetic Vectors , Hemophilia A , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 3 , RNA, Long Noncoding , Animals , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia A/complications , Dependovirus/genetics , RNA, Long Noncoding/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Mice , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Factor VIII/genetics , Factor VIII/metabolism , Joint Diseases/therapy , Joint Diseases/genetics , Joint Diseases/etiology , Humans , Genetic Therapy/methods , Mice, Inbred C57BL , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Disease Models, Animal , Male
13.
Transl Vis Sci Technol ; 13(6): 11, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38888288

ABSTRACT

Purpose: To report on cases of unilateral perimacular atrophy after treatment with voretigene neparvovec-rzyl, in the setting of previous contralateral eye treatment with a different viral vector. Design: Single-center, retrospective chart review. Methods: In this case series, four patients between the ages of six and 11 years old with RPE65-related retinopathy were treated unilaterally with rAAV2-CB-hRPE65 as part of a gene augmentation clinical trial (NCT00749957). Six to 10 years later the contralateral eyes were treated with the Food and Drug Administration-approved drug, voretigene neparvovec-rzyl. Best-corrected visual acuity (BCVA), fundus photos, ocular coherence tomography, two-color dark-adapted perimetry, full field stimulus threshold testing (FST), and location of subretinal bleb and chorioretinal atrophy were evaluated. Results: Three out of four patients showed unilateral perimacular atrophy after treatment with voretigene, ranging from five to 22 months after treatment. Areas of robust visual field improvement were followed by areas of chorioretinal atrophy. Despite perimacular changes, BCVA, FST, and subjective improvements in vision and nyctalopia were maintained. Perimacular atrophy was not observed in the first eye treated with the previous viral vector. Conclusions: We observed areas of robust visual field improvement followed by perimacular atrophy in voretigene treated eyes, as compared to the initially treated contralateral eyes. Translational Relevance: Caution is advised when using two different viral vectors between eyes in gene therapy. This may become an important issue in the future with increasing gene therapy clinical trials for inherited retinal dystrophies.


Subject(s)
Genetic Therapy , Genetic Vectors , Tomography, Optical Coherence , Visual Acuity , cis-trans-Isomerases , Humans , Retrospective Studies , Genetic Vectors/genetics , Genetic Therapy/methods , Male , Female , Child , cis-trans-Isomerases/genetics , Dependovirus/genetics , Atrophy , Visual Fields
14.
Appl Microbiol Biotechnol ; 108(1): 385, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896252

ABSTRACT

Recombinant adeno-associated virus (rAAV) is a major gene delivery vehicle. We have constructed a stable rAAV producer cell line by integrating essential rAAV genome, viral and helper genes into the genome of HEK293 cell under the control of inducible promoters. Upon induction, the cell line produces transducing rAAV. To gain insight into enhancing rAAV productivity and vector quality, we performed a comparative transcriptomic and proteomic analysis of our synthetic cell line GX2 and two wild-type AAV (wtAAV) production systems, one by virus co-infection and the other by multi-plasmid transfection. The three systems had different kinetics in viral component synthesis but generated comparable copies of AAV genomes; however, the capsid titer of GX2 was an order of magnitude lower compared to those two wtAAV systems, indicating that its capsid production may be insufficient. The genome packaging efficiency was also lower in GX2 despite it produced higher levels of Rep52 proteins than either wtAAV systems, suggesting that Rep52 protein expression may not limit genome packaging. In the two wtAAV systems, VP were the most abundant AAV proteins and their levels continued to increase, while GX2 had high level of wasteful cargo gene expression. Furthermore, upregulated inflammation, innate immune responses, and MAPK signaling, as well as downregulated mitochondrial functions, were commonly observed in either rAAV or wtAAV systems. Overall, this comparative multi-omics study provided rich insights into host cell and viral factors that are potential targets for genetic and process intervention to enhance the productivity of synthetic rAAV producer cell lines. KEY POINTS: • wtAAV infection was more efficient in producing full viral particles than the synthetic cell GX2. • Capsid protein synthesis, genome replication, and packaging may limit rAAV production in GX2. • wtAAV infection and rAAV production in GX2 elicited similar host cell responses.


Subject(s)
Dependovirus , Proteomics , Dependovirus/genetics , Humans , HEK293 Cells , Transcriptome , Genetic Vectors/genetics , Kinetics , Genome, Viral , Gene Expression Profiling , Proteome
15.
Methods Mol Biol ; 2817: 115-132, 2024.
Article in English | MEDLINE | ID: mdl-38907151

ABSTRACT

Single-cell-type proteomics is an emerging field of research that combines cell-type specificity with the comprehensive proteome coverage offered by bulk proteomics. However, the extraction of single-cell-type proteomes remains a challenge, particularly for hard-to-isolate cells like neurons. In this chapter, we present an innovative technique for profiling single-cell-type proteomes using adeno-associated virus (AAV)-mediated proximity labeling (PL) and tandem-mass-tag (TMT) mass spectrometry. This technique eliminates the need for cell isolation and offers a streamlined workflow, including AAV delivery to express TurboID (an engineered biotin ligase) controlled by cell-type-specific promoters, biotinylated protein purification, on-bead digestion, TMT labeling, and liquid chromatography-mass spectrometry (LC-MS). We examined this method by analyzing distinct brain cell types in mice. Initially, recombinant AAVs were used to concurrently express TurboID and mCherry proteins driven by neuron- or astrocyte-specific promoters, which was validated through co-immunostaining with cellular markers. With biotin purification and TMT analysis, we successfully identified around 10,000 unique proteins from a few micrograms of protein samples with high reproducibility. Our statistical analyses revealed that these proteomes encompass cell-type-specific cellular pathways. By utilizing this technique, researchers can explore the proteomic landscape of specific cell types, paving the way for new insights into cellular processes, deciphering disease mechanisms, and identifying therapeutic targets in neuroscience and beyond.


Subject(s)
Brain , Dependovirus , Proteome , Proteomics , Tandem Mass Spectrometry , Dependovirus/genetics , Dependovirus/metabolism , Animals , Mice , Proteomics/methods , Proteome/analysis , Brain/metabolism , Tandem Mass Spectrometry/methods , Single-Cell Analysis/methods , Neurons/metabolism , Chromatography, Liquid/methods , Genetic Vectors/genetics , Biotinylation , Mass Spectrometry/methods , Astrocytes/metabolism
16.
STAR Protoc ; 5(2): 103117, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38857153

ABSTRACT

Studying synapses in vivo presents challenges due to the complexity of accurately targeting and visualizing specific synaptic proteins within the brain. Here, we present a protocol for in vivo analysis of pre- and post-synaptic protein function in mice. We describe steps for combining adeno-associated virus (AAV)-mediated gene transfer to manipulate specific neuron subtypes. We also describe immunofluorescence on artificial cerebrospinal fluid (ACSF)-perfused brain sections to enhance the visualization of synaptic proteins. For complete details on the use and execution of this protocol, please refer to Cramer et al.1.


Subject(s)
Dependovirus , Synapses , Animals , Mice , Synapses/metabolism , Dependovirus/genetics , Brain/metabolism , Neurons/metabolism , Gene Transfer Techniques , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics
17.
PLoS Pathog ; 20(6): e1012260, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885242

ABSTRACT

Adeno-associated virus (AAV) serotypes from primates are being developed and clinically used as vectors for human gene therapy. However, the evolutionary mechanism of AAV variants is far from being understood, except that genetic recombination plays an important role. Furthermore, little is known about the interaction between AAV and its natural hosts, human and nonhuman primates. In this study, natural AAV capsid genes were subjected to systemic evolutionary analysis with a focus on selection drives during the diversification of AAV lineages. A number of positively selected sites were identified from these AAV lineages with functional relevance implied by their localization on the AAV structures. The selection drives of the two AAV2 capsid sites were further investigated in a series of biological experiments. These observations did not support the evolution of the site 410 of the AAV2 capsid driven by selection pressure from the human CD4+ T-cell response. However, positive selection on site 548 of the AAV2 capsid was directly related to host humoral immunity because of the profound effects of mutations at this site on the immune evasion of AAV variants from human neutralizing antibodies at both the individual and population levels. Overall, this work provides a novel interpretation of the genetic diversity and evolution of AAV lineages in their natural hosts, which may contribute to their further engineering and application in human gene therapy.


Subject(s)
Capsid Proteins , Dependovirus , Evolution, Molecular , Selection, Genetic , Dependovirus/genetics , Dependovirus/immunology , Humans , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Variation , Genetic Therapy
18.
Acta Neuropathol Commun ; 12(1): 89, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38845058

ABSTRACT

The microtubule-associated protein Tau is a key player in various neurodegenerative conditions, including Alzheimer's disease (AD) and Tauopathies, where its hyperphosphorylation disrupts neuronal microtubular lattice stability. Glaucoma, a neurodegenerative disorder affecting the retina, leads to irreversible vision loss by damaging retinal ganglion cells and the optic nerve, often associated with increased intraocular pressure. Prior studies have indicated Tau expression and phosphorylation alterations in the retina in both AD and glaucoma, yet the causative or downstream nature of Tau protein changes in these pathologies remains unclear. This study investigates the impact of Tau protein modulation on retinal neurons under normal and experimental glaucoma conditions. Employing AAV9-mediated gene therapy for Tau overexpression and knockdown, both manipulations were found to adversely affect retinal structural and functional measures as well as neuroprotective Akt/Erk survival signalling in healthy conditions. In the experimental glaucoma model, Tau overexpression intensified inner retinal degeneration, while Tau silencing provided significant protection against these degenerative changes. These findings underscore the critical role of endogenous Tau protein levels in preserving retinal integrity and emphasize the therapeutic potential of targeting Tau in glaucoma pathology.


Subject(s)
Genetic Therapy , Glaucoma , tau Proteins , tau Proteins/metabolism , Animals , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/genetics , Genetic Therapy/methods , Proto-Oncogene Proteins c-akt/metabolism , Dependovirus/genetics , Disease Models, Animal , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , Retina/metabolism , Retina/pathology , MAP Kinase Signaling System/physiology , Signal Transduction/physiology , Mice , Mice, Inbred C57BL , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Phenotype
19.
Zool Res ; 45(4): 781-790, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38894521

ABSTRACT

Precise targeting of specific regions within the central nervous system (CNS) is crucial for both scientific research and gene therapy in the context of brain diseases. Adeno-associated virus 13 (AAV13) is known for its restricted diffusion range within the CNS, making it an ideal choice for precise labeling and administration within small brain regions. However, AAV13 mediates relatively low expression of target genes. Here, we introduced specifically engineered modifications to the AAV13 capsid protein to enhance its transduction efficiency. We first constructed AAV13-YF by mutating tyrosine to phenylalanine on the surface of the AAV13 capsid. We then inserted the 7m8 peptide, known to enhance cell transduction, into positions 587/588 and 585/586 of the AAV13 capsid, resulting in two distinct variants named AAV13-587-7m8 and AAV13-585-7m8, respectively. We found that AAV13-YF exhibited superior in vitro infectivity in HEK293T cells compared to AAV13, while AAV13-587-7m8 and AAV13-585-7m8 showed enhanced CNS infection capabilities in C57BL/6 mice, with AAV13-587-7m8 infection retaining a limited spread range. These modified AAV13 variants hold promising potential for applications in gene therapy and neuroscience research.


Subject(s)
Dependovirus , Mice, Inbred C57BL , Dependovirus/genetics , Animals , Humans , Mice , HEK293 Cells , Transduction, Genetic , Capsid Proteins/genetics , Capsid Proteins/metabolism
20.
Nat Commun ; 15(1): 4924, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858354

ABSTRACT

Targeted gene delivery to the brain is a critical tool for neuroscience research and has significant potential to treat human disease. However, the site-specific delivery of common gene vectors such as adeno-associated viruses (AAVs) is typically performed via invasive injections, which limit its applicable scope of research and clinical applications. Alternatively, focused ultrasound blood-brain-barrier opening (FUS-BBBO), performed noninvasively, enables the site-specific entry of AAVs into the brain from systemic circulation. However, when used in conjunction with natural AAV serotypes, this approach has limited transduction efficiency and results in substantial undesirable transduction of peripheral organs. Here, we use high throughput in vivo selection to engineer new AAV vectors specifically designed for local neuronal transduction at the site of FUS-BBBO. The resulting vectors substantially enhance ultrasound-targeted gene delivery and neuronal tropism while reducing peripheral transduction, providing a more than ten-fold improvement in targeting specificity in two tested mouse strains. In addition to enhancing the only known approach to noninvasively target gene delivery to specific brain regions, these results establish the ability of AAV vectors to be evolved for specific physical delivery mechanisms.


Subject(s)
Blood-Brain Barrier , Brain , Dependovirus , Gene Transfer Techniques , Genetic Vectors , Animals , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Dependovirus/genetics , Mice , Blood-Brain Barrier/metabolism , Brain/metabolism , Humans , Neurons/metabolism , Transduction, Genetic/methods , Mice, Inbred C57BL , Genetic Engineering/methods , Female , Male , HEK293 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...