Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.420
Filter
1.
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
2.
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
3.
STAR Protoc ; 5(3): 103200, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39028619

ABSTRACT

Patient-derived organoids (PDOs) are now used to study many diseases, including prostate cancer. Here, we present a protocol for the transduction of human epithelial prostate cells and PDOs. We describe the steps for producing lentiviruses and transducing PDOs with high efficiency to obtain either overexpression or knockdown of specific genes. More generally, this protocol represents an efficient lentiviral transduction technique to study cell biology using various organoid models.


Subject(s)
Epithelial Cells , Lentivirus , Organoids , Prostate , Humans , Organoids/cytology , Organoids/metabolism , Male , Prostate/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lentivirus/genetics , Transduction, Genetic/methods , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
4.
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
5.
Methods Mol Biol ; 2805: 19-30, 2024.
Article in English | MEDLINE | ID: mdl-39008172

ABSTRACT

Transformed lung organoids have extensive applications in lung cancer modeling and drug screening. Traditional two-dimensional (2D) cultures fail to propagate a large subpopulation of murine primary tumors in vitro. However, three-dimensional (3D) air-liquid interface (ALI) cultures, which are employed to grow normal lung organoids, can be used to efficiently culture cancerous lung tumor cells. Here, we detail a procedure for cultivating genetically modified lung organoids in 3D-ALI cultures. This protocol contains two parts. The first part describes how to transduce lung epithelial cells, which are either freshly sorted from lungs or from actively growing murine organoids, with virus in order to modify gene expression. The target lung cells are incubated with virus for 1-2 h for transduction. Then, the transduced cells are thoroughly washed and mixed with stromal support cells and Matrigel and are loaded into transwell inserts for culture and validated for genetic modifications through downstream assays. The second part describes how to isolate tumor cells growing orthotopically in genetically engineered mouse models to produce organoid cell lines that can be used for ex vivo drug discovery assays. For this protocol, tumors are isolated from lungs of mice, finely chopped and washed. Then, tumor chunks are mixed with Matrigel for 3D-ALI culture. Finally, organoids budding from tumor chunks are trypsinized and passaged to establish an organoid line. Together these two protocols provide a promising platform to study the genesis, progression, and treatment of lung cancer.


Subject(s)
Lung Neoplasms , Lung , Organoids , Organoids/cytology , Animals , Mice , Lung/cytology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Cell Culture Techniques, Three Dimensional/methods , Humans , Cell Culture Techniques/methods , Epithelial Cells/cytology , Transduction, Genetic/methods
6.
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
7.
Methods Mol Biol ; 2810: 29-53, 2024.
Article in English | MEDLINE | ID: mdl-38926271

ABSTRACT

Baculovirus-mediated gene expression in mammalian cells, BacMam, is a useful alternative to transient transfection for recombinant protein production in various types of mammalian cell lines. We decided to establish BacMam in our lab in order to streamline our workflows for gene expression in insect and mammalian cells, as it is straightforward to parallelize the baculovirus generation for both types of eukaryotic cells. This chapter provides a step-by-step description of the protocols we use for the generation of the recombinant BacMam viruses, the transduction of mammalian cell cultures, and optimization of the protein production conditions through small-scale expression and purification tests.


Subject(s)
Baculoviridae , Gene Expression , Recombinant Proteins , Baculoviridae/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , Animals , Humans , Genetic Vectors/genetics , Cell Line , Sf9 Cells , Transduction, Genetic/methods , Transfection/methods , Cell Culture Techniques/methods
8.
Methods Mol Biol ; 2810: 147-159, 2024.
Article in English | MEDLINE | ID: mdl-38926278

ABSTRACT

Lentiviral gene transfer represents a versatile and powerful method for genetic transduction of many cell lines and primary cells including "hard-to-transfect" cells. As a consequence of the integration of the recombinant lentiviral vector into the cellular genome, the transgene is stably maintained, and long-term producing cells are established. Here, we describe the current state of the art and give details for lab-scale production of lentiviral vectors as well as for infection and titration of the viral vectors.


Subject(s)
Genetic Vectors , Lentivirus , Transduction, Genetic , Transduction, Genetic/methods , Lentivirus/genetics , Genetic Vectors/genetics , Humans , Transgenes , Gene Expression , Cell Line , HEK293 Cells , Transfection/methods
9.
Analyst ; 149(15): 3980-3988, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38872436

ABSTRACT

To overcome limitations in the generalizability and efficiency of current AAV vectors, in this current study, we constructed an AAV variant library by the insertion of random heptapeptide sequences in the receptor-binding domain of the AAV9 capsid gene. We then applied a recently developed organ-on-a-chip in vitro model of the human blood-brain barrier (BBB) to iteratively enrich for variants that efficiently cross the BBB and transduce astrocyte cells. Through multiple rounds of screening, we obtained two candidate AAV variants, AAV-M6 and AAV-M8, which showed significantly higher BBB penetration efficiency than AAV9 or AAV-PHP.eB. Quantitative PCR (qPCR) assay showed that AAV-M6 could accumulate to a 5 times higher titer, while AAV-M8 reached a 3 times higher titer, than AAV-PHP.eB in the neural chamber of the model. The transduction assay further verified that the AAV-M6 candidate vector was able to infect HA-1800 cells after crossing the BBB, suggesting it could potentially transduce brain parenchymal cells after crossing the hCMEC/D3 layer at higher efficiency than AAV-PHP.eB. Molecular simulations suggested that the human receptor proteins, LY6D and M6PR, could bind the AAV-M6 heptapeptide insertion with high affinity. This study provides two promising candidate AAV vectors and demonstrates the use of this in vitro BBB model for scalable, high-throughput screening of gene therapies. These tools can drive investigations of the mechanisms underlying BBB permeability and the cell-type specificity of virus vectors.


Subject(s)
Blood-Brain Barrier , Dependovirus , Genetic Vectors , Humans , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Dependovirus/genetics , Dependovirus/chemistry , Genetic Vectors/genetics , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Microphysiological Systems , Transduction, Genetic/methods
10.
BMC Mol Cell Biol ; 25(1): 15, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741034

ABSTRACT

BACKGROUND: Transfection is an important analytical method for studying gene expression in the cellular environment. There are some barriers to efficient DNA transfection in host cells, including circumventing the plasma membrane, escaping endosomal compartmentalization, autophagy, immune sensing pathways, and translocating the nuclear envelope. Therefore, it would be very useful to introduce an optimum transfection approach to achieve a high transfection efficiency in the Vero cell line. The aim of this study was to compare various transfection techniques and introduce a highly efficient method for gene delivery in Vero cells. METHODS: In the current study, three transfection methods were used, including chemical transfection, electroporation, and lentiviral vector transduction, to obtain the optimum transfection conditions in the Vero cell line. Vero cells were cultured and transfected with chemical transfection reagents, electroporation, or HIV-1-based lentivectors under different experimental conditions. Transfection efficiency was assessed using flow cytometry and fluorescence microscopy to detect GFP-positive cells. RESULTS: Among the tested methods, TurboFect™ chemical transfection exhibited the highest efficiency. Optimal transfection conditions were achieved using 1 µg DNA and 4 µL TurboFect™ in 6 × 104 Vero cells. CONCLUSION: TurboFect™, a cationic polymer transfection reagent, demonstrated superior transfection efficiency in Vero cells compared with electroporation and lentivirus particles, and is the optimal choice for chemical transfection in the Vero cell line.


Subject(s)
Electroporation , Genetic Vectors , Transfection , Animals , Chlorocebus aethiops , Vero Cells , Electroporation/methods , Transfection/methods , Genetic Vectors/genetics , Lentivirus/genetics , Transduction, Genetic/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans
11.
Folia Neuropathol ; 62(1): 32-46, 2024.
Article in English | MEDLINE | ID: mdl-38741435

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a potential source of somatic cells for cell therapies due to their ability to self-renew and differentiate into various cells of the body. To date, the clinical application of hiPSCs has been limited due to safety issues. The present study aims to standardize the safety procedure of the derivation of GMP-compliant induced pluripotent stem cell (iPSC) lines from human fibroblasts. The hiPSC lines were generated using the nonintegrative Sendai virus method to incorporate Yamanaka reprogramming factors (OCT3/4, SOX2, KLF4 and c-MYC) into cells. A constant temperature was maintained during the cell culture, including all stages of the culture after transduction with Sendai virus. Pluripotency was proved in six independently generated hiPSC lines from adult female (47 years old) and male (57 years old) donors' derived fibroblasts via alkaline phosphatase live (ALP) staining, qPCR, and immunocytochemistry. The hiPSC lines showed a gradual decrease in the presence of the virus with each subsequent passage, and this reduction was specific to the hiPSC line. The frequency and probability of chromosomal aberrations in hiPSCs were dependent on both the iPSC clone identity and sex of the donor. In summary, the generation of hiPSC for clinical applications requires safety standards application (biosafety protocol, quality control of hiPSC lines, viral and genetic integrity screening) from the first stages of the clonal selection of hiPSC from the same donor.


Subject(s)
Induced Pluripotent Stem Cells , Kruppel-Like Factor 4 , Sendai virus , Humans , Female , Male , Middle Aged , Cell Line , Fibroblasts , Cell Differentiation/physiology , Transduction, Genetic/methods , Sex Factors
12.
Skelet Muscle ; 14(1): 9, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702726

ABSTRACT

BACKGROUND: Adeno-associated virus (AAV)-based gene therapy is a promising strategy to treat muscle diseases. However, this strategy is currently confronted with challenges, including a lack of transduction efficiency across the entire muscular system and toxicity resulting from off-target tissue effects. Recently, novel myotropic AAVs named MyoAAVs and AAVMYOs have been discovered using a directed evolution approach, all separately demonstrating enhanced muscle transduction efficiency and liver de-targeting effects. However, these newly discovered AAV variants have not yet been compared. METHODS: In this study, we performed a comparative analysis of these various AAV9-derived vectors under the same experimental conditions following different injection time points in two distinct mouse strains. RESULTS: We highlight differences in transduction efficiency between AAV9, AAVMYO, MyoAAV2A and MyoAAV4A that depend on age at injection, doses and mouse genetic background. In addition, specific AAV serotypes appeared more potent to transduce skeletal muscles including diaphragm and/or to de-target heart or liver. CONCLUSIONS: Our study provides guidance for researchers aiming to establish proof-of-concept approaches for preventive or curative perspectives in mouse models, to ultimately lead to future clinical trials for muscle disorders.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Mice, Inbred C57BL , Muscle, Skeletal , Transduction, Genetic , Animals , Dependovirus/genetics , Genetic Vectors/administration & dosage , Muscle, Skeletal/metabolism , Mice , Transduction, Genetic/methods , Genetic Therapy/methods , Male , Liver/metabolism , Mice, Inbred mdx
13.
EMBO Mol Med ; 16(6): 1427-1450, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684862

ABSTRACT

Lentiviral vectors (LV) are efficient vehicles for in vivo gene delivery to the liver. LV integration into the chromatin of target cells ensures their transmission upon proliferation, thus allowing potentially life-long gene therapy following a single administration, even to young individuals. The glycoprotein of the vesicular stomatitis virus (VSV.G) is widely used to pseudotype LV, as it confers broad tropism and high stability. The baculovirus-derived GP64 envelope protein has been proposed as an alternative for in vivo liver-directed gene therapy. Here, we perform a detailed comparison of VSV.G- and GP64-pseudotyped LV in vitro and in vivo. We report that VSV.G-LV transduced hepatocytes better than GP64-LV, however the latter showed improved transduction of liver sinusoidal endothelial cells (LSEC). Combining GP64-pseudotyping with the high surface content of the phagocytosis inhibitor CD47 further enhanced LSEC transduction. Coagulation factor VIII (FVIII), the gene mutated in hemophilia A, is naturally expressed by LSEC, thus we exploited GP64-LV to deliver a FVIII transgene under the control of the endogenous FVIII promoter and achieved therapeutic amounts of FVIII and correction of hemophilia A mice.


Subject(s)
Endothelial Cells , Factor VIII , Genetic Therapy , Genetic Vectors , Hemophilia A , Lentivirus , Liver , Animals , Hemophilia A/therapy , Hemophilia A/genetics , Genetic Vectors/genetics , Endothelial Cells/metabolism , Mice , Lentivirus/genetics , Genetic Therapy/methods , Liver/metabolism , Factor VIII/genetics , Factor VIII/metabolism , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Transduction, Genetic/methods , Hepatocytes/metabolism , Hepatocytes/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism
14.
Gene Ther ; 31(7-8): 378-390, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38684788

ABSTRACT

Allogeneic cell therapies, such as those involving macrophages or Natural Killer (NK) cells, are of increasing interest for cancer immunotherapy. However, the current techniques for genetically modifying these cell types using lenti- or gamma-retroviral vectors present challenges, such as required cell pre-activation and inefficiency in transduction, which hinder the assessment of preclinical efficacy and clinical translation. In our study, we describe a novel lentiviral pseudotype based on the Koala Retrovirus (KoRV) envelope protein, which we identified based on homology to existing pseudotypes used in cell therapy. Unlike other pseudotyped viral vectors, this KoRV-based envelope demonstrates remarkable efficiency in transducing freshly isolated primary human NK cells directly from blood, as well as freshly obtained monocytes, which were differentiated to M1 macrophages as well as B cells from multiple donors, achieving up to 80% reporter gene expression within three days post-transduction. Importantly, KoRV-based transduction does not compromise the expression of crucial immune cell receptors, nor does it impair immune cell functionality, including NK cell viability, proliferation, cytotoxicity as well as phagocytosis of differentiated macrophages. Preserving immune cell functionality is pivotal for the success of cell-based therapeutics in treating various malignancies. By achieving high transduction rates of freshly isolated immune cells before expansion, our approach enables a streamlined and cost-effective automated production of off-the-shelf cell therapeutics, requiring fewer viral particles and less manufacturing steps. This breakthrough holds the potential to significantly reduce the time and resources required for producing e.g. NK cell therapeutics, expediting their availability to patients in need.


Subject(s)
Genetic Vectors , Killer Cells, Natural , Lentivirus , Macrophages , Transduction, Genetic , Humans , Genetic Vectors/genetics , Killer Cells, Natural/immunology , Lentivirus/genetics , Transduction, Genetic/methods , Macrophages/immunology , Macrophages/metabolism , Gene Transfer Techniques , Monocytes/immunology , Monocytes/metabolism , Genetic Therapy/methods , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Animals
15.
Cytotherapy ; 26(9): 1084-1094, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38661611

ABSTRACT

BACKGROUND AIMS: Chimeric antigen receptor (CAR) T-cell products are commonly generated using lentiviral vector (LV) transduction. Optimal final formulation buffer (FFB) supporting LV stability during cryostorage is crucial for cost-effective manufacturing. METHODS: To identify the ideal LV FFB composition for ex vivo CAR-T production, primary human T cells were transduced with vesicular stomatitis virus G-protein (VSV-G) -pseudotyped LVs (encoding a reporter gene or an anti-CD19-CAR). The formulations included phosphate-buffered saline (PBS), HEPES, or X-VIVOTM 15, and stabilizing excipients. The functional and viral particle titers and vector copy number were measured after LV cryopreservation and up to 24 h post-thaw incubation. CAR-Ts were produced with LVs in selected FFBs, and the resulting cells were characterized. RESULTS: Post-cryopreservation, HEPES-based FFBs provided higher LV functional titers than PBS and X-VIVOTM 15, and 10% trehalose-20 mM MgCl2 improved LV transduction efficiency in PBS and 50 mM HEPES. Thawed vectors remained stable at +4°C, while a ≤ 25% median decrease in the functional titer occurred during 24 h at room temperature. Tested excipients did not enhance LV post-thaw stability. CAR-Ts produced using LVs cryopreserved in 10% trehalose- or sucrose-20 mM MgCl2 in 50 mM HEPES showed comparable transduction rates, cell yield, viability, phenotype, and in vitro functionality. CONCLUSION: A buffer consisting of 10% trehalose-20 mM MgCl2 in 50 mM HEPES provided a feasible FFB to cryopreserve a VSV-G -pseudotyped LV for CAR-T-cell production. The LVs remained relatively stable for at least 24 h post-thaw, even at ambient temperatures. This study provides insights into process development, showing LV formulation data generated using the relevant target cell type for CAR-T-cell manufacturing.


Subject(s)
Genetic Vectors , Lentivirus , Receptors, Chimeric Antigen , T-Lymphocytes , Transduction, Genetic , Humans , Lentivirus/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Transduction, Genetic/methods , Genetic Vectors/genetics , Cryopreservation/methods , Immunotherapy, Adoptive/methods , Antigens, CD19
16.
Biomolecules ; 14(4)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38672522

ABSTRACT

In this study, we introduce electrospun polydioxanone (PDO) nonwoven fabrics as a platform for the delivery of adeno-associated virus (AAV) vectors for transduction and genome editing by adhering them to organ surfaces, including the heart. AAV vectors were loaded onto the PDO fabrics by soaking the fabrics in a solution containing AAV vectors. In vitro, the amount of AAV vectors loaded onto the fabrics could be adjusted by changing their concentration in the solution, and the number of cells expressing the green fluorescent protein (GFP) encoded by the AAV vectors increased in correlation with the increasing amount of loaded AAV vectors. In vivo, both transduction and genome editing resulted in the observation of GFP expression around AAV vector-loaded PDO fabrics attached to the surfaces of mouse hearts, indicating effective transduction and expression at the target site. These results demonstrate the great potential of electrospun PDO nonwoven fabrics carrying therapeutic AAV vectors for gene therapy.


Subject(s)
Dependovirus , Gene Editing , Genetic Vectors , Polydioxanone , Dependovirus/genetics , Animals , Genetic Vectors/genetics , Polydioxanone/chemistry , Gene Editing/methods , Mice , Humans , Transduction, Genetic/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Genetic Therapy/methods , Myocardium/metabolism
17.
Cytotherapy ; 26(6): 586-591, 2024 06.
Article in English | MEDLINE | ID: mdl-38551525

ABSTRACT

BACKGROUND AIMS: Gene therapy using lentiviral vectors (LVs) that harbor a functional ß-globin gene provides a curative treatment for hemoglobinopathies including beta-thalassemia and sickle cell disease. Accurate quantification of the vector copy number (VCN) and/or the proportion of transduced cells is critical to evaluate the efficacy of transduction and stability of the transgene during treatment. Moreover, commonly used techniques for LV quantification, including real-time quantitative polymerase chain reaction (PCR) or fluorescence-activated cell sorting, require either a standard curve or expression of a reporter protein for the detection of transduced cells. In the present study, we describe a digital droplet PCR (ddPCR) technique to measure the lentiviral VCN in transduced hematopoietic stem and progenitor cells (HSPCs). METHODS: After HSPCs were transduced with an LV encoding the therapeutic ß-globin (ßA-T87Q) gene, the integrated lentiviral sequence in the host genome was amplified with primers that targeted a sequence within the vector and the human RPP30 gene. The dynamic range of ddPCR was between 5 × 10-3 ng and 5 × 10-6 ng of target copy per reaction. RESULTS: We found that the ddPCR-based approach was able to estimate VCN with high sensitivity and a low standard deviation. Furthermore, ddPCR-mediated quantitation of lentiviral copy numbers in differentiated erythroblasts correlated with the level of ßA-T87Q protein detected by reverse-phase high-performance liquid chromatography. CONCLUSIONS: Taken together, the ddPCR technique has the potential to precisely detect LV copy numbers in the host genome, which can be used for VCN estimation, calculation of infectious titer and multiplicity of infection for HSPC transduction in a clinical setting.


Subject(s)
Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cells , Lentivirus , Transduction, Genetic , beta-Globins , Humans , Lentivirus/genetics , Hematopoietic Stem Cells/metabolism , Genetic Vectors/genetics , beta-Globins/genetics , Transduction, Genetic/methods , Genetic Therapy/methods , beta-Thalassemia/therapy , beta-Thalassemia/genetics , Polymerase Chain Reaction/methods , Gene Dosage/genetics
18.
Gene Ther ; 31(5-6): 285-294, 2024 05.
Article in English | MEDLINE | ID: mdl-38374348

ABSTRACT

Manufacturing of recombinant adeno-associated virus (AAV) vectors produces three types of capsids: full, intermediate, and empty. While there are different opinions about the impact of intermediate and empty capsids on safety and efficacy of AAV products, they are generally considered impurities because they are not the intended fully intact vector product. The presence of these impurities could impact product efficacy due to potential competition with fully packaged AAVs for cellular transduction, as well as have potential implications to patient safety due to increased capsid load during dosing. To determine the impact of intermediate capsids on potency, an AAV preparation was separated into fractions enriched for full, intermediate, or empty capsids. Using a matrix of in vitro (infectivity, gene expression, biological activity) and in vivo potency assays to determine potency as a function of capsid content, our results indicate that while intermediate capsids contribute to the vector genome titer of the product and are equally as infectious as full capsids, they do not contribute to the potency of the AAV product. This study confirms the criticality of reducing and controlling the level of intermediate capsids to ensure a more efficacious AAV product.


Subject(s)
Capsid , Dependovirus , Genetic Vectors , Dependovirus/genetics , Capsid/metabolism , Genetic Vectors/genetics , Humans , Animals , Mice , Transduction, Genetic/methods , HEK293 Cells , Genetic Therapy/methods
19.
Oxid Med Cell Longev ; 2022: 5503575, 2022.
Article in English | MEDLINE | ID: mdl-35251476

ABSTRACT

Cellular senescence is characterized by cell cycle arrest and senescence-associated secretory phenotypes. Cellular senescence can be caused by various stress stimuli such as DNA damage, oxidative stress, and telomere attrition and is related to several chronic diseases, including atherosclerosis, Alzheimer's disease, and osteoarthritis. Chromobox homolog 4 (CBX4) has been shown to alleviate cellular senescence in human mesenchymal stem cells and is considered a possible target for senomorphic treatment. Here, we explored whether CBX4 expression is associated with replicative senescence in WI-38 fibroblasts, a classic human senescence model system. We also examined whether and how regulation of CBX4 modifies the senescence phenotype and functions as an antisenescence target in WI-38. During the serial culture of the WI-38 primary fibroblast cell line to a senescent state, we found increased expression of senescence markers, including senescence ß-galactosidase (SA-ßgal) activity, protein expression of p16, p21, and DPP4, and decreased proliferation marker EdU; moreover, CBX4 protein expression declined. With knockdown of CBX4, SA-ßgal activity and p16 protein expression increased, and EdU decreased. With the activation of CBX4, SA-ßgal activity, p16, and DPP4 protein decreased. In addition, CBX4 knockdown increased, while CBX4 activation decreased, gene expression of both CDKN2A (encoding the p16 protein) and DPP4. Genes related to DNA damage and cell cycle pathways were regulated by CBX4. These results demonstrate that CBX4 can regulate replicative senescence in a manner consistent with a senomorphic agent.


Subject(s)
Cellular Senescence/genetics , Fibroblasts/metabolism , Ligases/metabolism , Polycomb-Group Proteins/metabolism , Signal Transduction/genetics , Biomarkers/metabolism , Cell Cycle Checkpoints/genetics , Cell Line , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Damage/genetics , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation , Gene Knockdown Techniques/methods , Humans , Ligases/genetics , Oxidative Stress/genetics , Phenotype , Polycomb-Group Proteins/genetics , Transduction, Genetic/methods , beta-Galactosidase/metabolism
20.
Oxid Med Cell Longev ; 2022: 5635797, 2022.
Article in English | MEDLINE | ID: mdl-35154568

ABSTRACT

Osteoarthritis (OA) is a chronic degenerative disease featured by cartilage erosion and inflammation. Luteolin, a member of the flavonoid family, has been shown to exert anti-inflammatory and antioxidative activities. However, the potential biological effects and underlying mechanism of luteolin on chondrocytes and OA progression remain largely elusive. In this study, the potential effect and mechanism of luteolin on OA were investigated in vitro and in vivo. Our data revealed that luteolin inhibited H2O2-induced cell death, apoptosis, oxidative stress, programmed necrosis, and inflammatory mediator production in primary murine chondrocytes. In addition, luteolin could activate the AMPK and Nrf2 pathways, and AMPK serves as a positive upstream regulator of Nrf2. In vivo results demonstrated the therapeutic effects of luteolin on OA in the DMM mouse model. Collectively, our findings showed that luteolin might serve as a novel and effective treatment for OA and provided a new research direction for clinical OA therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antioxidants/administration & dosage , Biological Products/administration & dosage , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Progression , Hydrogen Peroxide/adverse effects , Luteolin/administration & dosage , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Oxidative Stress/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cartilage, Articular/cytology , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques/methods , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Osteoarthritis/pathology , Oxidative Stress/genetics , Transduction, Genetic/methods , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL