Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Transl Vis Sci Technol ; 13(8): 37, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39177995

ABSTRACT

Purpose: Adeno-associated virus (AAV) demonstrates promise in delivering therapeutic genes to retinal ganglion cells (RGCs). Delivery of neuroprotective genes is constrained by packaging size and/or cell selectivity. This study compares the ability of the RGC-selective gamma-synuclein (SNCG) promoter and the smaller RGC-selective neurofilament heavy chain (NEFH) promoter, as well as portions of the RGC-selective atonal bHLH transcription factor 7 (ATOH7) enhancer, to drive gene expression in RGCs. Methods: AAV2 constructs with green fluorescent protein (GFP) or human sirtuin 1 (hSIRT1) driven by cytomegalovirus (CMV) enhancer and NEFH promoter (AAV2-eCMV-NEFH) or distal active sequences of the ATOH7 enhancer (DiATOH7) with the SNCG promoter (AAV2-DiATOH7-SNCG) were intravitreally injected into C57BL/6J mice. RGCs were immunolabeled with Brn3a antibodies and counted. AAV constructs with the utmost transduction efficiency were used to test the therapeutic efficacy of the hSIRT1 gene in 12-week-old C57BL/6J mice subjected to microbead (MB)-induced intraocular pressure (IOP) elevation. Visual function was measured using optokinetic responses (OKRs). Results: The eGFP transduction efficiency of AAV2-eCMV-NEFH was similar to that of AAV2-eCMV-SNCG and AAV2-DiATOH7-SNCG. When combined with the SNCG promoter, a larger ATOH7 enhancer was less efficient than the shorter DiATOH7 enhancer. Similarly, the hSIRT1 efficiency of AAV2-eCMV-NEFH was similar to that of AAV2-eCMV-SNCG. The latter two vectors were equally efficient in increasing RGC survival and improving visual function in the mouse model of MB-induced IOP elevation. Conclusions: SNCG and NEFH promoters represent two equally efficient and comparable RGC selective promoter sequences; however, the NEFH promoter offers a smaller packaging size. Translational Relevance: Smaller enhancer-promoter combinations can be used to deliver larger genes in human cells and for treatment in optic neuropathies including glaucoma.


Subject(s)
Dependovirus , Disease Models, Animal , Glaucoma , Mice, Inbred C57BL , Promoter Regions, Genetic , Retinal Ganglion Cells , Sirtuin 1 , gamma-Synuclein , Animals , Sirtuin 1/genetics , Sirtuin 1/metabolism , Mice , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Humans , Glaucoma/genetics , Glaucoma/therapy , Glaucoma/metabolism , Dependovirus/genetics , gamma-Synuclein/genetics , gamma-Synuclein/metabolism , Intravitreal Injections , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Intraocular Pressure/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation
2.
EBioMedicine ; 106: 105266, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39094262

ABSTRACT

The Chimeric Antigen Receptor (CAR) T cell therapy has emerged as a ground-breaking immunotherapeutic approach in cancer treatment. To overcome the complexity and high manufacturing cost associated with current ex vivo CAR T cell therapy products, alternative strategies to produce CAR T cells directly in the body have been developed in recent years. These strategies involve the direct infusion of CAR genes via engineered nanocarriers or viral vectors to generate CAR T cells in situ. This review offers a comprehensive overview of recent advancements in the development of T cell-targeted CAR generation in situ. Additionally, it identifies the challenges associated with in vivo CAR T method and potential strategies to overcome these issues.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/genetics , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
3.
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
4.
Nat Commun ; 15(1): 7193, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39168991

ABSTRACT

Durable factor VIII expression that normalizes hemostasis is an unrealized goal of hemophilia A adeno-associated virus-mediated gene therapy. Trials with initially normal factor VIII activity observed unexplained year-over-year declines in expression while others reported low-level, stable expression inadequate to restore normal hemostasis. Here we demonstrate that male mice recapitulate expression-level-dependent loss of factor VIII levels due to declines in vector copy number. We show that an enhanced function factor VIII variant (factor VIII-R336Q/R562Q), resistant to activated protein C-mediated inactivation, normalizes hemostasis at below-normal expression without evidence of prothrombotic risk in male hemophilia A mice. These data support that factor VIII-R336Q/R562Q may restore normal factor VIII function at low levels of expression to permit durability using low vector doses to minimize dose-dependent adeno-associated virus toxicities. This work informs the mechanism of factor VIII durability after gene transfer and supports that factor VIII-R336Q/R562Q may safely overcome current hemophilia A gene therapy limitations.


Subject(s)
Dependovirus , Factor VIII , Genetic Therapy , Genetic Vectors , Hemophilia A , Animals , Hemophilia A/therapy , Hemophilia A/genetics , Factor VIII/genetics , Factor VIII/metabolism , Genetic Therapy/methods , Male , Mice , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Humans , Disease Models, Animal , Mice, Inbred C57BL , Hemostasis
5.
CNS Neurosci Ther ; 30(8): e14919, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39123298

ABSTRACT

BACKGROUND: Sanfilippo syndrome (mucopolysaccharidosis type IIIA; MPS IIIA) is a childhood dementia caused by inherited mutations in the sulfamidase gene. At present, there is no treatment and children with classical disease generally die in their late teens. Intravenous or intra-cerebrospinal fluid (CSF) injection of AAV9-gene replacement is being examined in human clinical trials; evaluation of the impact on brain disease is an intense focus; however, MPS IIIA patients also experience profound, progressive photoreceptor loss, leading to night blindness. AIM: To compare the relative efficacy of the two therapeutic approaches on retinal degeneration in MPS IIIA mice. METHODS: Neonatal mice received i.v. or intra-CSF AAV9-sulfamidase or vehicle and after 20 weeks, biochemical and histological evaluation of neuroretina integrity was carried out. RESULTS: Both treatments improved central retinal thickness; however, in peripheral retina, outer nuclear layer thickness and photoreceptor cell length were only significantly improved by i.v. gene replacement. Further, normalization of endo-lysosomal compartment size and microglial morphology was only observed following intravenous gene delivery. CONCLUSIONS: Confirmatory studies are needed in adult mice; however, these data indicate that i.v. AAV9-sulfamidase infusion leads to superior outcomes in neuroretina, and cerebrospinal fluid-delivered AAV9 may need to be supplemented with another therapeutic approach for optimal patient quality of life.


Subject(s)
Dependovirus , Genetic Therapy , Mucopolysaccharidosis III , Retina , Animals , Mucopolysaccharidosis III/therapy , Mucopolysaccharidosis III/genetics , Genetic Therapy/methods , Dependovirus/genetics , Retina/pathology , Mice , Disease Models, Animal , Hydrolases/genetics , Animals, Newborn , Mice, Inbred C57BL , Dementia/genetics , Dementia/therapy , Genetic Vectors/administration & dosage , Injections, Intravenous
6.
Int J Nanomedicine ; 19: 7691-7708, 2024.
Article in English | MEDLINE | ID: mdl-39099791

ABSTRACT

Gene therapy aims to add, replace or turn off genes to help treat disease. To date, the US Food and Drug Administration (FDA) has approved 14 gene therapy products. With the increasing interest in gene therapy, feasible gene delivery vectors are necessary for inserting new genes into cells. There are different kinds of gene delivery vectors including viral vectors like lentivirus, adenovirus, retrovirus, adeno-associated virus et al, and non-viral vectors like naked DNA, lipid vectors, polymer nanoparticles, exosomes et al, with viruses being the most commonly used. Among them, the most concerned vector is adeno-associated virus (AAV) because of its safety, natural ability to efficiently deliver gene into cells and sustained transgene expression in multiple tissues. In addition, the AAV genome can be engineered to generate recombinant AAV (rAAV) containing transgene sequences of interest and has been proven to be a safe gene vector. Recently, rAAV vectors have been approved for the treatment of various rare diseases. Despite these approvals, some major limitations of rAAV remain, namely nonspecific tissue targeting and host immune response. Additional problems include neutralizing antibodies that block transgene delivery, a finite transgene packaging capacity, high viral titer used for per dose and high cost. To deal with these challenges, several techniques have been developed. Based on differences in engineering methods, this review proposes three strategies: gene engineering-based capsid modification (capsid modification), capsid surface tethering through chemical conjugation (surface tethering), and other formulations loaded with AAV (virus load). In addition, the major advantages and limitations encountered in rAAV engineering strategies are summarized.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Transgenes , Dependovirus/genetics , Humans , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Genetic Therapy/methods , Immune Evasion , Animals , Genetic Engineering/methods , Gene Transfer Techniques , Viral Tropism
7.
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
8.
Methods Mol Biol ; 2842: 309-321, 2024.
Article in English | MEDLINE | ID: mdl-39012603

ABSTRACT

Modern neuroscience research is increasingly discovering that alterations in epigenetic states within key brain cells is correlated with brain diseases. These epigenetic alterations may include changes in histone post-translational modifications and/or DNA modifications, all of which affect transcription and other gene expression programs within the brain cells that comprise central brain regions. However, the exact causal contribution of these epigenome changes to brain disease cannot be elucidated in the absence of direct in vivo manipulations in the implicated brain areas. Combining the design and creation of epigenetic editing constructs, gene delivery strategies, and stereotaxic surgery enables neuroscience researchers to target and manipulate the epigenetic state of the brain cells of laboratory rodents in a locus-specific manner and test its causal contribution to disease-related pathology and behaviors. Here, we describe the surgical protocol utilized by our group and others, which is optimized for herpes simplex virus delivery into the mouse brain, although the protocol outlined herein could be applied for delivery of adeno-associated viruses, lentiviruses, or nonviral gene-delivery methods in both mice and rats. The method allows for the overexpression of engineered DNA-binding proteins for direct and targeted epigenome editing in rodent brain with excellent spatiotemporal control. Nearly any brain region of interest can be targeted in rodents at every stage of postnatal life. Owing to the versatility, reproducibility, and utility of this technique, it is an important method for any laboratory interested in studying the cellular, circuit, and behavioral consequences of manipulating the brain epigenome in laboratory rodents.


Subject(s)
Brain , Epigenesis, Genetic , Gene Editing , Gene Transfer Techniques , Stereotaxic Techniques , Animals , Mice , Gene Editing/methods , Brain/metabolism , Rats , Genetic Vectors/genetics , Genetic Vectors/administration & dosage
9.
Sci Rep ; 14(1): 16487, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39019902

ABSTRACT

Cerebral dopamine neurotrophic factor (CDNF) and its close structural relative, mesencephalic astrocyte-derived neurotrophic factor (MANF), are proteins with neurotrophic properties. CDNF protects and restores the function of dopamine (DA) neurons in rodent and non-human primate (NHP) toxin models of Parkinson's disease (PD) and therefore shows promise as a drug candidate for disease-modifying treatment of PD. Moreover, CDNF was found to be safe and to have some therapeutic effects on PD patients in phase 1/2 clinical trials. However, the mechanism underlying the neurotrophic activity of CDNF is unknown. In this study, we delivered human CDNF (hCDNF) to the brain using an adeno-associated viral (AAV) vector and demonstrated the neurotrophic effect of AAV-hCDNF in an acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. AAV-hCDNF resulted in the expression of hCDNF in the striatum (STR) and substantia nigra (SN), and no toxic effects on the nigrostriatal pathway were observed. Intrastriatal injection of AAV-hCDNF reduced motor impairment and partially alleviated gait dysfunction in the acute MPTP mouse model. In addition, gene therapy with AAV-hCDNF had significant neuroprotective effects on the nigrostriatal pathway and decreased the levels of interleukin 1beta (IL-1ß) and complement 3 (C3) in glial cells in the acute MPTP mouse model. Moreover, AAV-hCDNF reduced C/EBP homologous protein (CHOP) and glucose regulatory protein 78 (GRP78) expression in astroglia. These results suggest that the neuroprotective effects of CDNF may be mediated at least in part through the regulation of neuroinflammation and the UPR pathway in a mouse MPTP model of PD in vivo.


Subject(s)
Dependovirus , Disease Models, Animal , Dopaminergic Neurons , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Genetic Vectors , Nerve Growth Factors , Animals , Dopaminergic Neurons/metabolism , Dependovirus/genetics , Mice , Humans , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Gene Transfer Techniques , Male , Parkinson Disease/therapy , Parkinson Disease/metabolism , Parkinson Disease/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Inflammation/metabolism , Genetic Therapy/methods , Mice, Inbred C57BL , Corpus Striatum/metabolism , MPTP Poisoning/therapy , MPTP Poisoning/metabolism , Substantia Nigra/metabolism
10.
Skelet Muscle ; 14(1): 17, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39044305

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron (MN) degeneration, leading to neuromuscular junction (NMJ) dismantling and severe muscle atrophy. The nuclear receptor interaction protein (NRIP) functions as a multifunctional protein. It directly interacts with calmodulin or α-actinin 2, serving as a calcium sensor for muscle contraction and maintaining sarcomere integrity. Additionally, NRIP binds with the acetylcholine receptor (AChR) for NMJ stabilization. Loss of NRIP in muscles results in progressive motor neuron degeneration with abnormal NMJ architecture, resembling ALS phenotypes. Therefore, we hypothesize that NRIP could be a therapeutic factor for ALS. METHODS: We used SOD1 G93A mice, expressing human SOD1 with the ALS-linked G93A mutation, as an ALS model. An adeno-associated virus vector encoding the human NRIP gene (AAV-NRIP) was generated and injected into the muscles of SOD1 G93A mice at 60 days of age, before disease onset. Pathological and behavioral changes were measured to evaluate the therapeutic effects of AAV-NRIP on the disease progression of SOD1 G93A mice. RESULTS: SOD1 G93A mice exhibited lower NRIP expression than wild-type mice in both the spinal cord and skeletal muscle tissues. Forced NRIP expression through AAV-NRIP intramuscular injection was observed in skeletal muscles and retrogradely transduced into the spinal cord. AAV-NRIP gene therapy enhanced movement distance and rearing frequencies in SOD1 G93A mice. Moreover, AAV-NRIP increased myofiber size and slow myosin expression, ameliorated NMJ degeneration and axon terminal denervation at NMJ, and increased the number of α-motor neurons (α-MNs) and compound muscle action potential (CMAP) in SOD1 G93A mice. CONCLUSIONS: AAV-NRIP gene therapy ameliorates muscle atrophy, motor neuron degeneration, and axon terminal denervation at NMJ, leading to increased NMJ transmission and improved motor functions in SOD1 G93A mice. Collectively, AAV-NRIP could be a potential therapeutic drug for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Dependovirus , Disease Models, Animal , Genetic Therapy , Mice, Transgenic , Motor Neurons , Muscular Atrophy , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Genetic Therapy/methods , Muscular Atrophy/genetics , Muscular Atrophy/therapy , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Dependovirus/genetics , Mice , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Genetic Vectors/administration & dosage , Nerve Degeneration/genetics , Nerve Degeneration/therapy , Male , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
11.
J Gene Med ; 26(7): e3720, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39041639

ABSTRACT

BACKGROUND: A novel adeno-associated virus 2 (AAV2)-carried multi-characteristic opsin (MCO) (MCO-010) is undergoing several clinical trials as a novel therapeutic modality for the treatment of degenerative retinal diseases including retinitis pigmentosa and Stargardt disease. The present study aimed to determine the ocular and systemic safety of MCO-010 and the AAV2 vehicle in adult Beagle dogs following intravitreal (IVT) injection. METHODS: The current safety/toxicology studies spanning 13 weeks described here utilized well-documented techniques to assess the effects of IVT injection of MCO-010 up to 2.2 × 1011 genome copies (gc) per eye, or the AAV2 capsid (vehicle control) on gross behavioral and immunogenic changes, alterations in body weights, blood biochemistry, hematology, blood coagulation, gross necropsy lesions, organ weight changes and histopathology in the dogs (n = 4 per group; two males and two females per group). Immunohistochemical and functional electroretinogram studies were also conducted to determine MCO expression in the retina and determine any retinal toxicity associated with MCO-010. RESULTS: There were no significant deleterious effects of the MCO-010 (or the AAV2 at the tested doses) on any of the examined parameters, including the absence of any severe ocular or systemic adverse events. However, as expected, inflammation after IVT delivery of AAV2 and MCO-010 was observed in the conjunctivae of all groups of animals, although this self-resolved within 1 week post-injection. Quantitative immunohistochemical analyses of MCO-010-associated mCherry revealed successful delivery of the gene therapy within the inner retina. CONCLUSIONS: In summary, MCO-010 demonstrated a favorable safety profile when administered to the eyes of adult Beagle dogs of both sexes at dose levels up to 2.2 × 1011 gc per eye, with no adverse effects observed. This dose was identified as the No Observed Adverse Effect Level (i.e. NOAEL) and guided selection of safe doses for human clinical trials.


Subject(s)
Dependovirus , Genetic Vectors , Intravitreal Injections , Opsins , Retina , Animals , Dogs , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Female , Male , Retina/metabolism , Opsins/genetics , Opsins/metabolism , Genetic Therapy/methods , Electroretinography
12.
Hum Gene Ther ; 35(15-16): 564-575, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38970425

ABSTRACT

Choroideremia, an incurable, progressive retinal degeneration primarily affecting young men, leads to sight loss. GEMINI was a multicenter, open-label, prospective, two-period, interventional Phase II study assessing the safety of bilateral sequential administration of timrepigene emparvovec, a gene therapy, in adult males with genetically confirmed choroideremia (NCT03507686, ClinicalTrials.gov). Timrepigene emparvovec is an adeno-associated virus serotype 2 vector encoding the cDNA of Rab escort protein 1, augmented by a downstream woodchuck hepatitis virus post-transcriptional regulatory element. Up to 0.1 mL of timrepigene emparvovec, containing 1 × 1011 vector genomes, was administered by subretinal injection following vitrectomy and retinal detachment. The second eye was treated after an intrasurgery window of <6, 6-12, or >12 months. Each eye was followed at up to nine visits over 12 months. Overall, 66 participants received timrepigene emparvovec, and 53 completed the study. Visual acuity (VA) was generally maintained in both eyes, independent of intrasurgery window duration, even after bilateral retinal detachment and subretinal injection. Bilateral treatment was well tolerated, with predominantly mild or moderate treatment-emergent adverse events (TEAEs) and a low rate of serious surgical complications (7.6%). Retinal inflammation TEAEs were reported in 45.5% of participants, with similar rates in both eyes; post hoc analyses found that these were not associated with clinically significant vision loss at month 12 versus baseline. Two participants (3.0%) reported serious noninfective retinitis. Prior timrepigene emparvovec exposure did not increase the risk of serious TEAEs or serious ocular TEAEs upon injection of the second eye; furthermore, no systemic immune reaction or inoculation effect was observed. Presence of antivector neutralizing antibodies at baseline was potentially associated with a higher percentage of TEAEs related to ocular inflammation or reduced VA after injection of the first eye. The GEMINI study results may inform decisions regarding bilateral sequential administration of other gene therapies for retinal diseases.


Subject(s)
Choroideremia , Dependovirus , Genetic Therapy , Genetic Vectors , Choroideremia/therapy , Choroideremia/genetics , Humans , Genetic Therapy/adverse effects , Genetic Therapy/methods , Male , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Genetic Vectors/genetics , Adult , Middle Aged , Dependovirus/genetics , Retina/pathology , Retina/metabolism , Visual Acuity , Adaptor Proteins, Signal Transducing/genetics , Prospective Studies , Treatment Outcome , Young Adult
13.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000316

ABSTRACT

We aimed to produce a mouse model of spinocerebellar ataxia type 3 (SCA3) using the mouse blood-brain barrier (BBB)-penetrating adeno-associated virus (AAV)-PHP.B. Four-to-five-week-old C57BL/6 mice received injections of high-dose (2.0 × 1011 vg/mouse) or low-dose (5.0 × 1010 vg/mouse) AAV-PHP.B encoding a SCA3 causative gene containing abnormally long 89 CAG repeats [ATXN3(Q89)] under the control of the ubiquitous chicken ß-actin hybrid (CBh) promoter. Control mice received high doses of AAV-PHP.B encoding ATXN3 with non-pathogenic 15 CAG repeats [ATXN3(Q15)] or phosphate-buffered saline (PBS) alone. More than half of the mice injected with high doses of AAV-PHP.B encoding ATXN3(Q89) died within 4 weeks after the injection. No mice in other groups died during the 12-week observation period. Mice injected with low doses of AAV-PHP.B encoding ATXN3(Q89) exhibited progressive motor uncoordination starting 4 weeks and a shorter stride in footprint analysis performed at 12 weeks post-AAV injection. Immunohistochemistry showed thinning of the molecular layer and the formation of nuclear inclusions in Purkinje cells from mice injected with low doses of AAV-PHP.B encoding ATXN3(Q89). Moreover, ATXN3(Q89) expression significantly reduced the number of large projection neurons in the cerebellar nuclei to one third of that observed in mice expressing ATXN3(Q15). This AAV-based approach is superior to conventional methods in that the required number of model mice can be created simply by injecting AAV, and the expression levels of the responsible gene can be adjusted by changing the amount of AAV injected. Moreover, this method may be applied to produce SCA3 models in non-human primates.


Subject(s)
Ataxin-3 , Dependovirus , Disease Models, Animal , Genetic Vectors , Machado-Joseph Disease , Mice, Inbred C57BL , Animals , Dependovirus/genetics , Machado-Joseph Disease/genetics , Machado-Joseph Disease/therapy , Machado-Joseph Disease/metabolism , Machado-Joseph Disease/pathology , Mice , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Ataxin-3/genetics , Ataxin-3/metabolism , Injections, Intravenous , Blood-Brain Barrier/metabolism , Promoter Regions, Genetic
14.
Nat Commun ; 15(1): 5871, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997283

ABSTRACT

There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability.


Subject(s)
Chemokine CXCL9 , Dependovirus , Genetic Therapy , Glioblastoma , Immune Checkpoint Inhibitors , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Glioblastoma/therapy , Glioblastoma/immunology , Dependovirus/genetics , Tumor Microenvironment/immunology , Animals , Humans , Immune Checkpoint Inhibitors/therapeutic use , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Mice , Genetic Therapy/methods , Female , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Cell Line, Tumor , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics
15.
Theranostics ; 14(10): 3827-3842, 2024.
Article in English | MEDLINE | ID: mdl-38994027

ABSTRACT

Rationale: In male mammals, many developmental-stage-specific RNA transcripts (both coding and noncoding) are preferentially or exclusively expressed in the testis, where they play important roles in spermatogenesis and male fertility. However, a reliable platform for efficiently depleting various types of RNA transcripts to study their biological functions during spermatogenesis in vivo has not been developed. Methods: We used an adeno-associated virus serotype nine (AAV9)-mediated CRISPR-CasRx system to knock down the expression of exogenous and endogenous RNA transcripts in the testis. Virus particles were injected into the seminiferous tubules via the efferent duct. Using an autophagy inhibitor, 3-methyladenine (3-MA), we optimized the AAV9 transduction efficiency in germ cells in vivo. Results: AAV9-mediated delivery of CRISPR-CasRx effectively and specifically induces RNA transcripts (both coding and noncoding) knockdown in the testis in vivo. In addition, we showed that the co-microinjection of AAV9 and 3-MA into the seminiferous tubules enabled long-term transgene expression in the testis. Finally, we found that a promoter of Sycp1 gene induced CRISPR-CasRx-mediated RNA transcript knockdown in a germ-cell-type-specific manner. Conclusion: Our results demonstrate the efficacy and versatility of the AAV9-mediated CRISPR-CasRx system as a flexible knockdown platform for studying gene function during spermatogenesis in vivo. This approach may advance the development of RNA-targeting therapies for conditions affecting reproductive health.


Subject(s)
CRISPR-Cas Systems , Dependovirus , Gene Knockdown Techniques , Spermatogenesis , Testis , Male , Animals , Dependovirus/genetics , CRISPR-Cas Systems/genetics , Mice , Testis/metabolism , Gene Knockdown Techniques/methods , Spermatogenesis/genetics , RNA/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage
16.
Mol Ther ; 32(8): 2624-2640, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38956871

ABSTRACT

Chronic pancreatitis (CP) is marked by progressive fibrosis and the activation of pancreatic stellate cells (PSCs), accompanied by the destruction of pancreatic parenchyma, leading to the loss of acinar cells (ACs). Few research studies have explored the mechanism by which damaged ACs (DACs) contribute to PSCs activation and pancreatic fibrosis. Currently, there are no effective drugs for curing CP or limiting the progression of pancreatic fibrosis. In this research, co-culture with intact acinar cells (IACs) suppressed PSC activation, while co-culture with DACs did the opposite. Krüppel-like factor 4 (KLF4) was significantly upregulated in DACs and was established as the key molecule that switches ACs from PSCs-suppressor to PSCs-activator. We revealed the exosomes of IACs contributed to the anti-activated function of IACs-CS on PSCs. MiRNome profiling showed that let-7 family is significantly enriched in IAC-derived exosomes (>30% miRNome), which partially mediates IACs' suppressive impacts on PSCs. Furthermore, it has been observed that the enrichment of let-7 in exosomes was influenced by the expression level of KLF4. Mechanistic studies demonstrated that KLF4 in ACs upregulated Lin28A, thereby decreasing let-7 levels in AC-derived exosomes, and thus promoting PSCs activation. We utilized an adeno-associated virus specifically targeting KLF4 in ACs (shKLF4-pAAV) to suppress PSCs activation in CP, resulting in reduced pancreatic fibrosis. IAC-derived exosomes hold potential as potent weapons against PSCs activation via let-7s, while activated KLF4/Lin28A signaling in DACs diminished such functions. ShKLF4-pAAV holds promise as a novel therapeutic approach for CP.


Subject(s)
Acinar Cells , Exosomes , Fibrosis , Kruppel-Like Factor 4 , MicroRNAs , Pancreatic Stellate Cells , Pancreatitis, Chronic , Kruppel-Like Factor 4/metabolism , Animals , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Exosomes/metabolism , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , MicroRNAs/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Dependovirus/genetics , Mice , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Male , Coculture Techniques , Pancreas/metabolism , Pancreas/pathology , Genetic Therapy/methods
17.
Methods Mol Biol ; 2837: 207-218, 2024.
Article in English | MEDLINE | ID: mdl-39044087

ABSTRACT

Mice infected with a recombinant adeno-associated virus carrying a replication-competent hepatitis B virus genome (rAAV-HBV) via the intravenous route establish a persistent HBV replication in hepatocytes and develop immune tolerance. They serve as models to evaluate antiviral immunity and to assess potential therapeutic approaches for chronic HBV infection. Combining selected HBV variants and different mouse genotypes allows for addressing a broad spectrum of research questions. This chapter describes the basic principles of the rAAV-HBV mouse model, rAAV-HBV production and purification methods, and finally, the in vivo application.


Subject(s)
Dependovirus , Disease Models, Animal , Genetic Vectors , Hepatitis B virus , Virus Replication , Animals , Dependovirus/genetics , Dependovirus/isolation & purification , Hepatitis B virus/genetics , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Humans , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/immunology , Hepatitis B/virology , Hepatitis B/immunology
18.
PLoS One ; 19(7): e0306719, 2024.
Article in English | MEDLINE | ID: mdl-38976688

ABSTRACT

Previously, we reported the development of a human Aγ-globin gene lentivirus (LV), GbG, which expresses high levels of HbF to correct the sickle cell anemia (SCA) phenotype in the Berkeley SCA mouse model, and then modified the γ-globin gene by substituting glycine at codon 16 with aspartic acid in the Aγ-globin gene to generate GbGM LV. In the present study, we evaluated the long-term safety of human Aγ-globin gene carrying GbGM LV in wild-type mice after primary and secondary transplants of GbGM-modified hematopoietic stem cells (HSC) over 18 months. The safety of the GbGM bone marrow transplant was assessed by monitoring the effects on body weight, hematology, histopathology, malignancy formation, and survival. Mice transplanted with Mock-transduced and spleen focus forming virus (SFFV) γ-retroviral vector (RV)-transduced HSC served as negative and positive controls, respectively. The mean donor-cell engraftment was comparable across Mock, GbGM LV, and SFFV RV groups. There were no significant differences in body weight, clinical signs, immunophenotype, or histopathology in the GbGM-treated mice compared to controls. Four SFFV RV-treated mice, but none of the GbGM-treated mice, developed donor-derived, vector-positive lymphomas as demonstrated by flow cytometry analysis and in situ hybridization. These results highlight the safety of the administration of GbGM LV-modified HSC with long-term follow-up after primary and secondary transplants in mice. This data supported the initiation of phase 1/2 first-in-human SCA clinical trial in the United States.


Subject(s)
Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Hemoglobinopathies , Lentivirus , gamma-Globins , Animals , Lentivirus/genetics , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Mice , Humans , gamma-Globins/genetics , Hemoglobinopathies/therapy , Hemoglobinopathies/genetics , Hematopoietic Stem Cells/metabolism , Transplantation, Autologous , Disease Models, Animal
19.
Sci Rep ; 14(1): 14325, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38906906

ABSTRACT

Diabetic retinopathy (DR) is a multifactorial disease displaying vascular-associated pathologies, including vascular leakage and neovascularization, ultimately leading to visual impairment. However, animal models accurately reflecting these pathologies are lacking. Vascular endothelial growth factor A (VEGF-A) is an important factor in the development of micro- and macro-vascular pathology in DR. In this study, we evaluated the feasibility of using a cumate-inducible lentivirus (LV) mediated expression of vegf-a to understand DR pathology in vitro and in vivo. Retinal pigment epithelial cells (ARPE-19) were transduced with cumate-inducible LV expressing vegf-a, with subsequent analysis of vegf-a expression and its impact on cell proliferation, viability, motility, and permeability. Cumate tolerability in adult Wistar rat eyes was assessed as an initial step towards a potential DR animal model development, by administering cumate via intravitreal injections (IVT) and evaluating consequent effects by spectral domain optical coherence tomography (SD-OCT), flash electroretinography (fERG), ophthalmic examination (OE), and immunohistochemistry. Transduction of ARPE-19 cells with cumate-inducible LV resulted in ~ 2.5-fold increase in vegf-a mRNA and ~ threefold increase in VEGF-A protein secretion. Transduced cells displayed enhanced cell proliferation, viability, permeability, and migration in tube-like structures. However, IVT cumate injections led to apparent retinal toxicity, manifesting as retinal layer abnormalities, haemorrhage, vitreous opacities, and significant reductions in a- and b-wave amplitudes, along with increased microglial activation and reactive gliosis. In summary, while cumate-inducible LV-mediated vegf-a expression is valuable for in vitro mechanistic studies in cellular drug discovery, its use is not a feasible approach to model DR in in vivo studies due to cumate-induced retinal toxicity.


Subject(s)
Diabetic Retinopathy , Lentivirus , Retinal Pigment Epithelium , Vascular Endothelial Growth Factor A , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Diabetic Retinopathy/pathology , Diabetic Retinopathy/metabolism , Lentivirus/genetics , Rats , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Humans , Rats, Wistar , Cell Proliferation , Disease Models, Animal , Cell Line , Intravitreal Injections , Male , Cell Movement , Cell Survival , Tomography, Optical Coherence , Genetic Vectors/administration & dosage , Genetic Vectors/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL