Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.883
Filter
1.
Methods Mol Biol ; 2848: 169-186, 2025.
Article in English | MEDLINE | ID: mdl-39240523

ABSTRACT

The retinal explant culture system is a valuable tool for studying the pharmacological, toxicological, and developmental aspects of the retina. It is also used for translational studies such as gene therapy. While no photoreceptor-like cell lines are available for in vitro studies of photoreceptor cell biology, the retinal explant culture maintains the laminated retinal structure ex vivo for as long as a month. Human and nonhuman primate (NHP) postmortem retinal explants cut into small pieces offer the possibility of testing multiple conditions for safety and adeno-associated viral (AAV) vector optimization. In addition, the cone-enriched foveal area can be studied using the retinal explants. Here, we present a detailed working protocol for retinal explant isolation and culture from mouse, human, and NHP for testing drug efficacy and AAV transduction. Future applications of this protocol include combining live imaging and multiwell retinal explant culture for high-throughput drug screening systems in rodent and human retinal explants to identify new drugs against retinal degeneration.


Subject(s)
Dependovirus , Retina , Animals , Humans , Mice , Retina/cytology , Dependovirus/genetics , Primates , Genetic Vectors/genetics , Tissue Culture Techniques/methods , Transduction, Genetic
2.
Bull Exp Biol Med ; 177(4): 552-558, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39266922

ABSTRACT

A new gene-cell construct expressing nerve growth factor (NGF) has been developed. After obtaining engineered adenovectors Ad5-RGD-CAG-NGF and Ad5-RGD-CAG-EGFP, transduction efficiency and transgene expression were studied and multiplicity of infection was determined. The efficacy of transduced human olfactory ensheathing cells expressing NGF in restoring motor activity in rats has been shown in a limited period of time. Improved rat hindlimb mobility and cyst size reduction after gene-cell construct transplantation were more likely due to the cellular component of the construct.


Subject(s)
Cysts , Genetic Vectors , Nerve Growth Factor , Olfactory Mucosa , Animals , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Rats , Olfactory Mucosa/metabolism , Olfactory Mucosa/cytology , Humans , Cysts/therapy , Cysts/genetics , Cysts/pathology , Cysts/metabolism , Genetic Vectors/genetics , Transduction, Genetic , Genetic Therapy/methods , Adenoviridae/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
3.
Nat Commun ; 15(1): 7711, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231976

ABSTRACT

Baculovirus is an obligate parasitic virus of the phylum Arthropoda. Baculovirus including Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been widely used in the laboratory and industrial preparation of proteins or protein complexes. Due to its large packaging capacity and non-replicative and non-integrative natures in mammals, baculovirus has been proposed as a gene therapy vector for transgene delivery. However, the mechanism of baculovirus transduction in mammalian cells has not been fully illustrated. Here, we employed a cell surface protein-focused CRISPR screen to identify host dependency factors for baculovirus transduction in mammalian cells. The screening experiment uncovered a series of baculovirus host factors in human cells, including exostosin-like glycosyltransferase 3 (EXTL3) and NPC intracellular cholesterol transporter 1 (NPC1). Further investigation illustrated that EXTL3 affected baculovirus attachment and entry by participating in heparan sulfate biosynthesis. In addition, NPC1 promoted baculovirus transduction by mediating membrane fusion and endosomal escape. Moreover, in vivo, baculovirus transduction in Npc1-/+ mice showed that disruption of Npc1 gene significantly reduced baculovirus transduction in mouse liver. In summary, our study revealed the functions of EXTL3 and NPC1 in baculovirus attachment, entry, and endosomal escape in mammalian cells, which is useful for understanding baculovirus transduction in human cells.


Subject(s)
N-Acetylglucosaminyltransferases , Niemann-Pick C1 Protein , Nucleopolyhedroviruses , Animals , Nucleopolyhedroviruses/genetics , Nucleopolyhedroviruses/physiology , Humans , Mice , N-Acetylglucosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , HEK293 Cells , Endosomes/metabolism , Heparitin Sulfate/metabolism , Virus Internalization , Transduction, Genetic , Sf9 Cells , Liver/metabolism , Liver/virology , CRISPR-Cas Systems
4.
J Transl Med ; 22(1): 824, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237935

ABSTRACT

Highly efficient adeno associated viruses (AAVs) targeting the central nervous system (CNS) are needed to deliver safe and effective therapies for inherited neurological disorders. The goal of this study was to compare the organ-specific transduction efficiencies of two AAV capsids across three different delivery routes. We compared AAV9-CBA-fLucYFP to AAV-DJ-CBA-fLucYFP using the following delivery routes in mice: intracerebroventricular (ICV) 1 × 1012 vg/kg, intrathecal (IT) 1 × 1012 vg/kg, and intravenous (IV) 1 × 1013 vg/kg body weight. Our evaluations revealed that following ICV and IT administrations, AAV-DJ demonstrated significantly increased vector genome (vg) uptake throughout the CNS as compared to AAV9. Through the IV route, AAV9 demonstrated significantly increased vg uptake in the CNS. However, significantly fewer vgs were detected in the off-target organs (kidney and liver) following administration of AAV-DJ using the IT and IV delivery routes as compared to AAV9. Distributions of vgs correlate well with transgene transcript levels, luciferase enzyme activities, and immunofluorescence detection of YFP. Overall, between the two vectors, AAV-DJ resulted in better targeting and expression in CNS tissues paired with de-targeting and reduced expression in liver and kidneys. Our findings support further examination of AAV-DJ as a gene therapy capsid for the treatment of neurological disorders.


Subject(s)
Brain , Dependovirus , Genetic Vectors , Liver , Spinal Cord , Animals , Dependovirus/genetics , Liver/metabolism , Brain/metabolism , Genetic Vectors/administration & dosage , Spinal Cord/metabolism , Transgenes , Mice , Transduction, Genetic , Gene Transfer Techniques
5.
Breast Cancer Res ; 26(1): 131, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256827

ABSTRACT

BACKGROUND: Breast cancer is the second leading cause of death in women, with invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) as the two most common forms of invasive breast cancer. While estrogen receptor positive (ER+) IDC and ILC are treated similarly, the multifocality of ILC presents challenges in detection and treatment, worsening long-term clinical outcomes in patients. With increasing documentation of chemoresistance in ILC, additional treatment options are needed. Oncolytic adenoviral therapy may be a promising option, but cancer cells must express the coxsackievirus & adenovirus receptor (CAR) for adenoviral therapy to be effective. The present study aims to evaluate the extent to which CAR expression is observed in ILC in comparison to IDC, and how the levels of CAR expression correlate with adenovirus transduction efficiency. The effect of liposome encapsulation on transduction efficiency is also assessed. METHODS: To characterize CAR expression in invasive breast carcinoma, 36 formalin-fixed paraffin-embedded (FFPE) human breast tumor samples were assayed by CAR immunohistochemistry (IHC). Localization of CAR in comparison to other junctional proteins was performed using a multiplex immunofluorescence panel consisting of CAR, p120-catenin, and E-cadherin. ILC and IDC primary tumors and cell lines were transduced with E1- and E3-deleted adenovirus type 5 inserted with a GFP transgene (Ad-GFP) and DOTAP liposome encapsulated Ad-GFP (DfAd-GFP) at various multiplicities of infection (MOIs). Transduction efficiency was measured using a fluorescence plate reader. CAR expression in the human primary breast carcinomas and cell lines was also evaluated by IHC. RESULTS: We observed membranous CAR, p120-catenin and E-cadherin expression in IDC. In ILC, we observed cytoplasmic expression of CAR and p120-catenin, with absent E-cadherin. Adenovirus effectively transduced high-CAR IDC cell lines, at MOIs as low as 12.5. Ad-GFP showed similar transduction as DfAd-GFP in high-CAR IDC cell lines. Conversely, Ad-GFP transduction of ILC cell lines was observed only at MOIs of 50 and 100. Furthermore, Ad-GFP did not transduce CAR-negative IDC cell lines even at MOIs greater than 100. Liposome encapsulation (DfAd-GFP) improved transduction efficiency 4-fold in ILC and 17-fold in CAR-negative IDC cell lines. CONCLUSION: The present study demonstrates that oncolytic adenoviral therapy is less effective in ILC than IDC due to differences in spatial CAR expression. Liposome-enhanced delivery may be beneficial for patients with ILC and tumors with low or negative CAR expression to improve adenoviral therapeutic effectiveness.


Subject(s)
Adenoviridae , Breast Neoplasms , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Transduction, Genetic , Humans , Female , Breast Neoplasms/therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Adenoviridae/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Cell Line, Tumor , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/therapy , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Cadherins/metabolism , Cadherins/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Liposomes
6.
Nat Commun ; 15(1): 7965, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261465

ABSTRACT

Current adeno-associated virus (AAV) gene therapy using nature-derived AAVs is limited by non-optimal tissue targeting. In the treatment of muscular diseases (MD), high doses are often required but can lead to severe adverse effects. Here, we rationally design an AAV capsid that specifically targets skeletal muscle to lower treatment doses. We computationally integrate binding motifs of human integrin alphaV beta6, a skeletal muscle receptor, into a liver-detargeting capsid. Designed AAVs show higher productivity and superior muscle transduction compared to their parent. One variant, LICA1, demonstrates comparable muscle transduction to other myotropic AAVs with reduced liver targeting. LICA1's myotropic properties are observed across species, including non-human primate. Consequently, LICA1, but not AAV9, effectively delivers therapeutic transgenes and improved muscle functionality in two mouse MD models (male mice) at a low dose (5E12 vg/kg). These results underline the potential of our design method for AAV engineering and LICA1 variant for MD gene therapy.


Subject(s)
Dependovirus , Genetic Therapy , Muscle, Skeletal , Dependovirus/genetics , Animals , Humans , Muscle, Skeletal/metabolism , Mice , Genetic Therapy/methods , Male , Genetic Vectors/genetics , Integrins/metabolism , Integrins/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Muscular Diseases/therapy , Muscular Diseases/genetics , Transduction, Genetic , Liver/metabolism , Capsid/metabolism , Receptors, Vitronectin/metabolism , Receptors, Vitronectin/genetics , Disease Models, Animal , HEK293 Cells , Transgenes , Mice, Inbred C57BL , Antigens, Neoplasm
7.
J Dermatol Sci ; 115(3): 101-110, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127592

ABSTRACT

BACKGROUND: Local gene therapies, including in vivo genome editing, are highly anticipated for the treatment of genetic diseases in skin, especially the epidermis. While the adeno-associated virus (AAV) is a potent vector for in vivo gene delivery, the lack of efficient gene delivery methods has limited its clinical applications. OBJECTIVE: To optimize the AAV gene delivery system with higher gene delivery efficiency and specificity for epidermis and keratinocytes (KCs), using AAV capsid and promoter engineering technologies. METHODS: AAV variants with mutations in residues reported to be critical to determine the tropism of AAV2 for KCs were generated by site-directed mutagenesis of AAVDJ. The infection efficiency and specificity for KCs of these variants were compared with those of previously reported AAVs considered to be suitable for gene delivery to KCs in vitro and in vivo. Additionally, we generated an epidermis-specific promoter using the most recent short-core promoter and compared its specificity with existing promoters. RESULTS: A novel AAVDJ variant capsid termed AAVDJK2 was superior to the existing AAVs in terms of gene transduction efficiency and specificity for epidermis and KCs in vitro and in vivo. A novel tissue-specific promoter, termed the K14 SCP3 promoter, was superior to the existing promoters in terms of gene transduction efficiency and specificity for KCs. CONCLUSION: The combination of the AAVDJK2 capsid and K14 SCP3 promoter improves gene delivery to epidermis in vivo and KCs in vitro. The novel AAV system may benefit experimental research and development of new epidermis-targeted gene therapies.


Subject(s)
Dependovirus , Epidermis , Genetic Therapy , Genetic Vectors , Keratinocytes , Promoter Regions, Genetic , Transduction, Genetic , Dependovirus/genetics , Keratinocytes/metabolism , Keratinocytes/virology , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Humans , Animals , Promoter Regions, Genetic/genetics , Genetic Therapy/methods , Epidermis/metabolism , Mice , Mutagenesis, Site-Directed , Gene Transfer Techniques , Capsid Proteins/genetics , Capsid Proteins/metabolism
8.
Nat Commun ; 15(1): 6602, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39097583

ABSTRACT

Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.


Subject(s)
Capsid Proteins , Capsid , Dependovirus , Genetic Vectors , Liver , Dependovirus/genetics , Animals , Humans , Mice , Genetic Vectors/genetics , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Liver/metabolism , Transduction, Genetic , Gene Transfer Techniques , Machine Learning , Genetic Therapy/methods , Macaca , Hepatocytes/metabolism , HEK293 Cells , Genetic Engineering/methods
9.
Hum Gene Ther ; 35(17-18): 695-709, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39155805

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein. Although CF affects multiple organs, lung disease is the main cause of morbidity and mortality, and gene therapy is expected to provide a mutation-agnostic option for treatment. SP-101 is a recombinant adeno-associated virus (AAV) gene therapy vector carrying a human CFTR minigene, hCFTRΔR, and is being investigated as an inhalation treatment for people with CF. To further understand SP-101 activity, in vitro studies were performed in human airway epithelia (HAE) derived from multiple CF and non-CF donors. SP-101 restored CFTR-mediated chloride conductance, measured via Ussing chamber assay, at a multiplicity of infection (MOI) as low as 5E2 in the presence of doxorubicin, a small molecule known to augment AAV transduction. Functional correction of CF HAE increased with increasing MOI and doxorubicin concentration and correlated with increasing cell-associated vector genomes and hCFTRΔR mRNA expression. Tropism studies using a fluorescent reporter vector and single-cell mRNA sequencing of SP-101-mediated hCFTRΔR mRNA demonstrated broad expression in all cell types after apical transduction, including secretory, ciliated, and basal cells. In summary, SP-101, particularly in combination with doxorubicin, shows promise for a novel CF treatment strategy and strongly supports continued development.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Dependovirus , Genetic Therapy , Genetic Vectors , Respiratory Mucosa , Humans , Cystic Fibrosis/therapy , Cystic Fibrosis/genetics , Dependovirus/genetics , Genetic Therapy/methods , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Respiratory Mucosa/metabolism , Epithelial Cells/metabolism , Cells, Cultured , Transduction, Genetic , Doxorubicin/pharmacology
10.
Gene Ther ; 31(9-10): 489-498, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39134629

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors are currently the only proven vehicles for treating ophthalmological diseases through gene therapy. A wide range of gene therapy programs that target ocular diseases are currently being pursued. Nearly 20 years of research have gone into enhancing the efficacy of targeting retinal tissues and improving transgene delivery to specific cell types. The engineered AAV capsid, AAV2.7m8 is currently among the best capsids for transducing the retina following intravitreal (IVT) injection. However, adverse effects, including intraocular inflammation, have been reported following retinal administration of AAV2.7m8 vectors in clinical trials. Furthermore, we have consistently observed that AAV2.7m8 exhibits low packaging titers irrespective of the vector construct design. In this report, we found that AAV2.7m8 packages vector genomes with a higher degree of heterogeneity than AAV2. We also found that genome-loaded AAV2.7m8 stimulated the infiltration of microglia in mouse retinas following IVT administration, while the response to genome-loaded AAV2 and empty AAV2.7m8 capsids produced much milder responses. This finding suggests that IVT administration of AAV2.7m8 vectors may stimulate retinal immune responses in part because of its penchant to package and deliver non-unit length genomes.


Subject(s)
Capsid , Dependovirus , Genetic Therapy , Genetic Vectors , Retina , Dependovirus/genetics , Animals , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Mice , Retina/metabolism , Capsid/metabolism , Genetic Therapy/methods , Genome, Viral , Humans , Mice, Inbred C57BL , Transduction, Genetic/methods , Microglia/metabolism
11.
Viruses ; 16(8)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39205190

ABSTRACT

Transduction of producer cells during lentiviral vector (LVV) production causes the loss of 70-90% of viable particles. This process is called retro-transduction and it is a consequence of the interaction between the LVV envelope protein, VSV-G, and the LDL receptor located on the producer cell membrane, allowing lentiviral vector transduction. Avoiding retro-transduction in LVV manufacturing is crucial to improve net production and, therefore, the efficiency of the production process. Here, we describe a method for quantifying the transduction of producer cells and three different strategies that, focused on the interaction between VSV-G and the LDLR, aim to reduce retro-transduction.


Subject(s)
Genetic Vectors , Lentivirus , Receptors, LDL , Transduction, Genetic , Genetic Vectors/genetics , Lentivirus/genetics , Humans , Receptors, LDL/metabolism , Receptors, LDL/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , HEK293 Cells , Membrane Glycoproteins
12.
Viruses ; 16(8)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39205208

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) play a pivotal role in the treatment of genetic diseases. However, current production and purification processes yield AAV-based preparations that often contain unwanted empty, partially filled or damaged viral particles and impurities, including residual host cell DNA and proteins, plasmid DNA, and viral aggregates. To precisely understand the composition of AAV preparations, we systematically compared four different single-stranded AAV (ssAAV) and self-complementary (scAAV) fractions extracted from the CsCl ultracentrifugation gradient using established methods (transduction efficiency, analytical ultracentrifugation (AUC), quantitative and digital droplet PCR (qPCR and ddPCR), transmission electron microscopy (TEM) and enzyme-linked immunosorbent assay (ELISA)) alongside newer techniques (multiplex ddPCR, multi-angle light-scattering coupled to size-exclusion chromatography (SEC-MALS), multi-angle dynamic light scattering (MADLS), and high-throughput sequencing (HTS)). Suboptimal particle separation within the fractions resulted in unexpectedly similar infectivity levels. No single technique could simultaneously provide comprehensive insights in the presence of both bioactive particles and contaminants. Notably, multiplex ddPCR revealed distinct vector genome fragmentation patterns, differing between ssAAV and scAAV. This highlights the urgent need for innovative analytical and production approaches to optimize AAV vector production and enhance therapeutic outcomes.


Subject(s)
Dependovirus , Ultracentrifugation , Virion , Dependovirus/genetics , Dependovirus/isolation & purification , Humans , Virion/isolation & purification , Virion/genetics , Genetic Vectors/genetics , HEK293 Cells , Cesium/chemistry , Centrifugation, Density Gradient/methods , Transduction, Genetic , Chlorides
13.
J Gene Med ; 26(8): e3726, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39160647

ABSTRACT

BACKGROUND: Conventional adeno-associated viral (AAV) vectors, while highly effective in quiescent cells such as hepatocytes in the adult liver, confer less durable transgene expression in proliferating cells owing to episome loss. Sustained therapeutic success is therefore less likely in liver disorders requiring early intervention. We have previously developed a hybrid, dual virion approach, recombinant AAV (rAAV)/piggyBac transposon system capable of achieving stable gene transfer in proliferating hepatocytes at levels many fold above conventional AAV vectors. An alternative transposon system, Sleeping Beauty, has been widely used for ex vivo gene delivery; however liver-targeted delivery using a hybrid rAAV/Sleeping Beauty approach remains relatively unexplored. METHODS: We investigated the capacity of a Sleeping Beauty (SB)-based dual rAAV virion approach to achieve stable and efficient gene transfer to the newborn murine liver using transposable therapeutic cassettes encoding coagulation factor IX or ornithine transcarbamylase (OTC). RESULTS: At equivalent doses, rAAV/SB100X transduced hepatocytes with high efficiency, achieving stable expression into adulthood. Compared with conventional AAV, the proportion of hepatocytes transduced, and factor IX and OTC activity levels, were both markedly increased. The proportion of hepatocytes stably transduced increased 4- to 8-fold from <5%, and activity levels increased correspondingly, with markedly increased survival and stable urinary orotate levels in the OTC-deficient Spfash mouse following elimination of residual endogenous murine OTC. CONCLUSIONS: The present study demonstrates the first in vivo utility of a hybrid rAAV/SB100X transposon system to achieve stable long-term therapeutic gene expression following delivery to the highly proliferative newborn mouse liver. These results have relevance to the treatment of genetic metabolic liver diseases with neonatal onset.


Subject(s)
Animals, Newborn , DNA Transposable Elements , Dependovirus , Gene Transfer Techniques , Genetic Vectors , Hepatocytes , Liver , Transduction, Genetic , Animals , Dependovirus/genetics , DNA Transposable Elements/genetics , Liver/metabolism , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Factor IX/genetics , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Transposases/genetics , Transposases/metabolism , Humans , Transgenes , Genetic Therapy/methods , Mice, Inbred C57BL
14.
Hum Gene Ther ; 35(17-18): 754-766, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39046109

ABSTRACT

Rheumatoid arthritis (RA) is a chronic, inflammatory autoimmune disease marked by joint destruction and functional impairment. Tumor necrosis factor (TNF) plays a critical role in RA pathogenesis. Although TNF-targeting drugs are clinically effective, their need for frequent and long-term administration often results in poor patient adherence and suboptimal outcomes. This study developed a gene therapy approach using engineered adeno-associated virus (AAV) vectors to deliver an anti-TNF agent directly into the joint cavity of RA animal models. Animals receiving this therapy demonstrated sustained improvement in clinical scores, inflammatory markers, and joint tissue health. Immunofluorescence staining revealed that AAV vectors could transduce various cell types, including T cells, type A synoviocytes, and dendritic cells. Our results indicate that a single administration of this gene therapy provided long-term efficacy. This suggests that AAV-mediated anti-TNF gene therapy can offer prolonged relief from clinical symptoms and reduce inflammatory damage in a mouse model of RA. This innovative approach presents a promising new therapy with significant clinical prospects to treat patients with RA.


Subject(s)
Arthritis, Rheumatoid , Dependovirus , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Tumor Necrosis Factor-alpha , Animals , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Arthritis, Rheumatoid/therapy , Mice , Genetic Therapy/methods , Humans , Transduction, Genetic , Arthritis, Experimental/therapy , Injections, Intra-Articular
15.
Elife ; 132024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042440

ABSTRACT

Genetically encoded calcium indicators (GECIs) such as GCaMP are invaluable tools in neuroscience to monitor neuronal activity using optical imaging. The viral transduction of GECIs is commonly used to target expression to specific brain regions, can be conveniently used with any mouse strain of interest without the need for prior crossing with a GECI mouse line, and avoids potential hazards due to the chronic expression of GECIs during development. A key requirement for monitoring neuronal activity with an indicator is that the indicator itself minimally affects activity. Here, using common adeno-associated viral (AAV) transduction procedures, we describe spatially confined aberrant Ca2+ microwaves slowly travelling through the hippocampus following expression of GCaMP6, GCaMP7, or R-CaMP1.07 driven by the synapsin promoter with AAV-dependent gene transfer in a titre-dependent fashion. Ca2+ microwaves developed in hippocampal CA1 and CA3, but not dentate gyrus nor neocortex, were typically first observed at 4 wk after viral transduction, and persisted up to at least 8 wk. The phenomenon was robust and observed across laboratories with various experimenters and setups. Our results indicate that aberrant hippocampal Ca2+ microwaves depend on the promoter and viral titre of the GECI, density of expression, as well as the targeted brain region. We used an alternative viral transduction method of GCaMP which avoids this artefact. The results show that commonly used Ca2+-indicator AAV transduction procedures can produce artefactual Ca2+ responses. Our aim is to raise awareness in the field of these artefactual transduction-induced Ca2+ microwaves, and we provide a potential solution.


Subject(s)
Calcium , Dependovirus , Hippocampus , Synapsins , Animals , Dependovirus/genetics , Synapsins/metabolism , Synapsins/genetics , Calcium/metabolism , Hippocampus/metabolism , Mice , Genetic Vectors , Transduction, Genetic , Promoter Regions, Genetic , Mice, Inbred C57BL , Male
16.
Gene Ther ; 31(9-10): 455-466, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39039204

ABSTRACT

Different screening methods are being developed to generate adeno-associated viral vectors (AAV) with the ability to bypass the blood-brain barrier (BBB) upon intravenous administration. Recently, the AAV9P31 stood out as the most efficient version among a library of peptide-displaying capsids selected in C57BL/6 mice using RNA-driven biopanning. In this work we have characterized in detail its biodistribution in different mouse strains (C57BL/6 and Balb/c), as well as in Sprague Dawley rats and non-human primates (Macaca fascicularis). Using GFP and NanoLuc reporter genes, we confirmed homogeneous infection and transgene expression across the CNS of mice injected intravenously with AAV9P31. A more restricted pattern was observed upon either intracerebroventricular or intraparenchymal injection. Following intravenous delivery, region- and cell-specific differential patterns of transduction were observed in the mouse brain, including a preferential transduction of astrocytes and neurons in the cerebral cortex and striatum, whereas neurons were the only transduced cell type in subcortical locations across the hippocampus, thalamus, hypothalamus, mesencephalon, brainstem and cerebellum. Furthermore, transduced microglial cells were never found in any CNS location. Peripheral organs transduced upon intravenous administration included lung, liver, peritoneum, heart and skeletal muscle. However, a comparable performance of AAV9P31 to bypass the BBB in rats and macaques was not observed, although a more limited neuronal transduction was found in the brainstem of rats upon intravenous delivery. Finally, intracerebroventricular delivery in macaques resulted in neuronal transduction in cortical, subcortical structures and cerebellum following a patchy pattern. In conclusion, the widespread CNS transduction obtained in mice upon intravenous delivery of AAV9P31 represents a powerful tool for modeling a wide variety of neurological disorders as well as an appealing choice for the evaluation of gene therapy-based therapeutics.


Subject(s)
Blood-Brain Barrier , Brain , Dependovirus , Genetic Vectors , Rats, Sprague-Dawley , Transduction, Genetic , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Rats , Transduction, Genetic/methods , Mice , Blood-Brain Barrier/metabolism , Brain/metabolism , Mice, Inbred C57BL , Mice, Inbred BALB C , Macaca fascicularis , Male , Tissue Distribution , Genetic Therapy/methods
17.
Nucleic Acids Res ; 52(16): 9369-9383, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39077931

ABSTRACT

A fundamental challenge for cystic fibrosis (CF) gene therapy is ensuring sufficient transduction of airway epithelia to achieve therapeutic correction. Hypertonic saline (HTS) is frequently administered to people with CF to enhance mucus clearance. HTS transiently disrupts epithelial cell tight junctions, but its ability to improve gene transfer has not been investigated. Here, we asked if increasing the concentration of NaCl enhances the transduction efficiency of three gene therapy vectors: adenovirus, AAV, and lentiviral vectors. Vectors formulated with 3-7% NaCl exhibited markedly increased transduction for all three platforms, leading to anion channel correction in primary cultures of human CF epithelial cells and enhanced gene transfer in mouse and pig airways in vivo. The mechanism of transduction enhancement involved tonicity but not osmolarity or pH. Formulating vectors with a high ionic strength solution is a simple strategy to greatly enhance efficacy and immediately improve preclinical or clinical applications.


Subject(s)
Cystic Fibrosis , Genetic Therapy , Genetic Vectors , Transduction, Genetic , Animals , Humans , Genetic Vectors/genetics , Genetic Vectors/chemistry , Mice , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Osmolar Concentration , Swine , Genetic Therapy/methods , Adenoviridae/genetics , Dependovirus/genetics , Lentivirus/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Respiratory Mucosa/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Gene Transfer Techniques , Saline Solution, Hypertonic
18.
J Gene Med ; 26(7): e3716, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961849

ABSTRACT

BACKGROUND: Differentiation of pluripotent stem cells into desired lineages is the key aspect of regenerative medicine and cell-based therapy. Although RNA interference (RNAi) technology is exploited extensively for this, methods for long term silencing of the target genes leading to differentiation remain a challenge. Sustained knockdown of the target gene by RNAi is often inefficient as a result of low delivery efficiencies, protocol induced toxicity and safety concerns related to viral vectors. Earlier, we established octa-arginine functionalized hydroxyapatite nano vehicles (R8HNPs) for delivery of small interfering RNA (siRNA) against a pluripotency marker gene in mouse embryonic stem cells. Although we demonstrated excellent knockdown efficiency of the target gene, sustained gene silencing leading to differentiation was yet to be achieved. METHODS: To establish a sustained non-viral gene silencing protocol using R8HNP, we investigated various methods of siRNA delivery: double delivery of adherent cells (Adh-D), suspension delivery followed by adherent delivery (Susp + Adh), single delivery in suspension (Susp-S) and multiple deliveries in suspension (Susp-R). Sustained knockdown of a pluripotent marker gene followed by differentiation was analysed by reverse transcriptase-PCR, fluoresence-activated cell sorting and immunofluorescence techniques. Impact on cell viability as a result of repeated exposure of the R8HNP was also tested. RESULTS: Amongst the protocols tested, the most efficient knockdown of the target gene for a prolonged period of time was obtained by repeated suspension delivery of the R8HNP-siRNA conjugate. The long-term silencing of a pluripotency marker gene resulted in differentiation of R1 ESCs predominantly towards the extra embryonic and ectodermal lineages. Cells displayed excellent tolerance to repeated exposures of R8HNPs. CONCLUSIONS: The results demonstrate that R8HNPs are promising, biocompatible, non-viral alternatives for prolonged gene silencing and obtaining differentiated cells for therapeutics.


Subject(s)
Cell Differentiation , Durapatite , Mouse Embryonic Stem Cells , RNA, Small Interfering , Animals , Mice , Durapatite/chemistry , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/drug effects , RNA, Small Interfering/genetics , Gene Silencing , Biocompatible Materials/chemistry , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Nanoparticles/chemistry , Transduction, Genetic , RNA Interference , Gene Knockdown Techniques
19.
Hum Gene Ther ; 35(17-18): 734-753, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39001819

ABSTRACT

Effective use of adeno-associated viruses (AAVs) for clinical gene therapy is limited by their propensity to accumulate in and transduce the liver. This natural liver tropism is associated with severe adverse events at the high doses that can be necessary for achieving therapeutic transgene expression in extrahepatic tissues. To improve the safety and cost of AAV gene therapy, capsid engineering efforts are underway to redirect in vivo AAV biodistribution away from the liver toward disease-relevant peripheral organs such as the heart. Building on previous work, we generated a series of AAV libraries containing variations at three residues (Y446, N470, and W503) of the galactose-binding pocket of the AAV9 VP1 protein. Screening of this library in mice identified the XRH family of variants (Y446X, N470R, and W503H), the strongest of which, HRH, exhibited a 6-fold reduction in liver RNA expression and a 10-fold increase in cardiac RNA expression compared with wild-type AAV9 in the mouse. Screening of our library in a nonhuman primate (NHP) revealed reduced performance of AAV9 and two closely related vectors in the NHP liver compared with the mouse liver. Measurement of the galactose-binding capacity of our library further identified those same three vectors as the only strong galactose binders, suggesting an altered galactose presentation between the mouse and NHP liver. N-glycan profiling of these tissues revealed a 9% decrease in exposed galactose in the NHP liver compared with the mouse liver. In this work, we identified a novel family of AAV variants with desirable biodistribution properties that may be suitable for targeting extrahepatic tissues such as the heart. These data also provide important insights regarding species- and tissue-specific differences in glycan presentation that may have implications for the development and translation of AAV gene therapies.


Subject(s)
Dependovirus , Galactose , Genetic Therapy , Genetic Vectors , Polysaccharides , Dependovirus/genetics , Animals , Genetic Vectors/genetics , Galactose/metabolism , Genetic Therapy/methods , Mice , Polysaccharides/metabolism , Humans , Liver/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Species Specificity , Transduction, Genetic
20.
PLoS One ; 19(7): e0305742, 2024.
Article in English | MEDLINE | ID: mdl-39028743

ABSTRACT

In vivo gene delivery to tissues using adeno-associated vector (AAVs) has revolutionized the field of gene therapy. Yet, while sensorineural hearing loss is one of the most common sensory disorders worldwide, gene therapy applied to the human inner ear is still in its infancy. Recent advances in the development recombinant AAVs have significantly improved their cell tropism and transduction efficiency across diverse inner ear cell types to a level that renders this tool valuable for conditionally manipulating gene expression in the context of developmental biology studies of the mouse inner ear. Here, we describe a protocol for in utero micro-injection of AAVs into the embryonic inner ear, using the AAV-PHP.eB and AAV-DJ serotypes that respectively target the sensory hair cells and the supporting cells of the auditory sensory epithelium. We also aimed to standardize procedures for imaging acquisition and image analysis to foster research reproducibility and allow accurate comparisons between studies. We find that AAV-PHP.eB and AAV-DJ provide efficient and reliable tools for conditional gene expression targeting cochlear sensory and supporting cells in the mouse inner ear, from late embryonic stages on.


Subject(s)
Dependovirus , Ear, Inner , Gene Transfer Techniques , Genetic Vectors , Animals , Dependovirus/genetics , Mice , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Female , Transduction, Genetic/methods , Pregnancy , Genetic Therapy/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL