Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Biomed Sci ; 31(1): 79, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138523

ABSTRACT

Gene therapy has made considerable strides in recent years. More than 4000 protein-coding genes have been implicated in more than 6000 genetic diseases; next-generation sequencing has dramatically revolutionized the diagnosis of genetic diseases. Most genetic diseases are considered very rare or ultrarare, defined here as having fewer than 1:100,000 cases, but only one of the 12 approved gene therapies (excluding RNA therapies) targets an ultrarare disease. This article explores three gene supplementation therapy approaches suitable for various rare genetic diseases: lentiviral vector-modified autologous CD34+ hematopoietic stem cell transplantation, systemic delivery of adeno-associated virus (AAV) vectors to the liver, and local AAV delivery to the cerebrospinal fluid and brain. Together with RNA therapies, we propose a potential business model for these gene therapies.


Subject(s)
Dependovirus , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Genetic Therapy/methods , Humans , Dependovirus/genetics , Genetic Vectors , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/genetics , Rare Diseases/therapy , Rare Diseases/genetics , Lentivirus/genetics
2.
Methods Mol Biol ; 2845: 161-175, 2024.
Article in English | MEDLINE | ID: mdl-39115665

ABSTRACT

The purpose of this protocol is to provide a comprehensive, stepwise guide for assessing mitophagy flux utilizing a live-cell mt-KEIMA approach. The proposed protocol is sensitive, reproducible, quantitative, and easy to perform. While mitophagy has been extensively studied, current methodologies primarily focus on terminal measurements, neglecting the dynamic aspect of this process. Hence, the introduction of this straightforward live-cell mitophagy tracing protocol enables real-time monitoring of the dynamics of mitochondrial selective autophagy, thereby enhancing the ability to draw conclusions regarding key regulators and the reversibility of the process. The assay employs a lentiviral approach to induce mt-KEIMA expression in primary or immortalized cell lines. Subsequently, the respective mitophagy reporter cells are observed using a live-cell imaging system at specific time intervals, and further quantification allows the detection of mitophagy flux. This protocol has proven efficacious in investigating mitophagy flux, including responses to chemical inducers or genetically modified cells over time. Notably, this approach is well-suited for large throughput screening of chemicals or appropriate gene-editing libraries that may influence mitophagy responses in cells.


Subject(s)
Mitochondria , Mitophagy , Humans , Mitochondria/metabolism , Cell Line , Lentivirus/genetics
3.
J Gene Med ; 26(7): e3717, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967915

ABSTRACT

BACKGROUND: Synaptic Ras GTPase activating protein 1 (SYNGAP1)-related non-specific intellectual disability is a neurodevelopmental disorder caused by an insufficient level of SynGAP1 resulting in a dysfunction of neuronal synapses and presenting with a wide array of clinical phenotypes. Hematopoietic stem cell gene therapy has the potential to deliver therapeutic levels of functional SynGAP1 to affected neurons upon transduction of hematopoietic stem and progenitor cells with a lentiviral vector. METHODS: As a novel approach toward the treatment of SYNGAP1, we have generated a lentiviral vector expressing a modified form of SynGAP1 for transduction of human CD34+ hematopoietic stem and progenitor cells. The gene-modified cells were then transplanted into adult immunodeficient SYNGAP1+/- heterozygous mice and evaluated for improvement of SYNGAP1-related clinical phenotypes. Expression of SynGAP1 was also evaluated in the brain tissue of transplanted mice. RESULTS: In our proof-of-concept study, we have demonstrated significant improvement of SYNGAP1-related phenotypes including an improvement in motor abilities observed in mice transplanted with the vector transduced cells because they displayed decreased hyperactivity in an open field assay and an increased latency to fall in a rotarod assay. An increased level of SynGAP1 was also detected in the brains of these mice. CONCLUSIONS: These early-stage results highlight the potential of this stem cell gene therapy approach as a treatment strategy for SYNGAP1.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Intellectual Disability , Animals , Humans , Mice , Brain/metabolism , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Intellectual Disability/therapy , Intellectual Disability/genetics , Lentivirus/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Transduction, Genetic
4.
Int Immunopharmacol ; 138: 112645, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38972208

ABSTRACT

BACKGROUND: Pulmonary fibrosis is a progressive disease with high incidence and poor prognosis. It is urgent to explore new therapeutic methods for pulmonary fibrosis. As a new treatment method, gene therapy has attracted more and more attention. CCDC59 is a transcriptional coactivator of SP-B and SP-C. Our study mainly aims to explore the effect of overexpression of CCDC59 gene in pulmonary fibrosis of mice. METHODS: CCDC59 overexpressing lentivirus was constructed and then concentrated. RT-qPCR, Western blotting, and immunofluorescence assays were used to detect the expression of CCDC59, SP-B and SP-C protein in cell line and lung tissues after infected with lentivirus. Immunohistochemical staining and hematoxylin-eosin staining assays were used to assess the degree of fibrosis and ELISA assay was used to detect the concentrations of inflammatory factors, SP-B, and SP-C in bronchoalveolar lavage fluid of mice. Dynamic changes of mice lung function at various time points were assessed by lung function test assay. HIPPO pathway and proliferation capacity of alveolar type II epithelial cells were evaluated by immunofluorescence staining and Western blotting. RESULTS: Results showed that endotracheal instillation of CCDC59 overexpressed lentivirus significantly alleviated bleomycin-induced inflammation and pulmonary fibrosis in mice. Overexpression of CCDC59 protein in type II alveolar epithelial cells can enhance the expression of SP-B and SP-C. Overexpression of CCDC59 protein significantly protected against pulmonary inflammatory response and improved lung function of mice. Overexpression of CCDC59 protein significantly alleviated the hyperactivation of HIPPO pathway and increased the proliferative capacity of type II alveolar epithelial cells in lung. CONCLUSION: CCDC59 can alleviate inflammation and pulmonary fibrosis in mice by upregulating the expression of SP-B and SP-C in type II alveolar epithelial cells and alleviating the hyperactivation of HIPPO pathway. Our study offers a new potential treatment for pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , Pulmonary Surfactant-Associated Protein C , Animals , Humans , Male , Mice , Bleomycin , Disease Models, Animal , Lentivirus/genetics , Lung/pathology , Lung/metabolism , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/therapy , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/therapy , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism
5.
Cell Mol Life Sci ; 81(1): 305, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012348

ABSTRACT

Lentiviral vectors have markedly enhanced gene therapy efficiency in treating congenital diseases, but their long-term safety remains controversial. Most gene therapies for congenital eye diseases need to be carried out at early ages, yet the assessment of related risks to ocular development posed by lentiviral vectors is challenging. Utilizing single-cell transcriptomic profiling on human retinal organoids, this study explored the impact of lentiviral vectors on the retinal development and found that lentiviral vectors can cause retinal precursor cells to shift toward photoreceptor fate through the up-regulation of key fate-determining genes such as PRDM1. Further investigation demonstrated that the intron and intergenic region of PRDM1 was bound by PHLDA1, which was also up-regulated by lentiviral vectors exposure. Importantly, knockdown of PHLDA1 successfully suppressed the lentivirus-induced differentiation bias of photoreceptor cells. The findings also suggest that while lentiviral vectors may disrupt the fate determination of retinal precursor cells, posing risks in early-stage retinal gene therapy, these risks could potentially be reduced by inhibiting the PHLDA1-PRDM1 axis.


Subject(s)
Cell Differentiation , Genetic Vectors , Lentivirus , Retina , Stem Cells , Transcription Factors , Humans , Retina/metabolism , Retina/cytology , Lentivirus/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Genetic Vectors/metabolism , Genetic Vectors/genetics , Cell Differentiation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Organoids/metabolism , Organoids/cytology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Genetic Therapy/methods
6.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2282-2293, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044591

ABSTRACT

The ubiquitin/proteasome system (UPS) plays a crucial role in maintaining cellular protein homeostasis. The catalytic activity of proteasome in the UPS is regulated by ß1 (PSMB6), ß2 (PSMB7), and ß5 (PSMB5) subunits. Interferon (IFN)-γ, tumor necrosis factor (TNF)-α, inflammation, and oxidative stress can induce the replacement of ß1, ß2, and ß5 with their respective immuno-subunits ß1i (PSMB9), ß2i (PSMB10), and ß5i (PSMB8), which can be assembled into the immunoproteasome. Compared with the standard proteasome, the immunoproteasome exerts enhanced regulatory effects on immune responses, such as processing and presenting MHC class Ⅰ antigens, production of pro-inflammatory cytokines, and T cell differentiation and proliferation. Abnormal aggregation of immunoproteasomes can cause neurodegenerative diseases like Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. To explore the function of PSMB9 after bacterial infection, we constructed a lentivirus plasmid overexpressing PSMB9-eGFP-His and transfected the plasmid into HEK293T cells for packaging by using a triple-plasmid system in this study. After screening with puromycin, we obtained a stable human leukemia monocytic THP-1 cell line expressing the fusion protein of PSMB9. Western blotting (WB) and fluorescence microscopy verified the expression of the fusion protein in the stable THP-1 cells. Quantitative PCR (qPCR) was employed to measure the copies of PSMB9-eGFP in THP-1 cells. Immunofluorescence results found that eGFP-His did not affect the subcellular localization of PSMB9. The purification with nickel affinity chromatography confirmed that the fusion protein could be assembled into the 20S immunoproteasome and exhibited cleaving activity for fluorescent peptide substrates. These results indicated that the PSMB9-eGFP fusion gene was integrated into the chromosome, and could be stably expressed in the constructed THP-1 cell line. This cell line can be utilized for the research on subcellular localization, dynamic expression, and activity of PSMB9 in live cells at different infection conditions and disease stages. It also provides a model for the stable cell lines construction of other immunoproteasome subunits PSMB8 and PSMB10.


Subject(s)
Green Fluorescent Proteins , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , THP-1 Cells , Lentivirus/genetics , Recombinant Fusion Proteins/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism
7.
Clin Exp Med ; 24(1): 155, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003408

ABSTRACT

Knowledge of the molecular pathogenesis of acute myeloid leukemia has advanced in recent years. Despite novel treatment options, acute myeloid leukemia remains a survival challenge for elderly patients. We have recently shown that the triphosphohydrolase SAMHD1 is one of the factors determining resistance to Ara-C treatment. Here, we designed and tested novel and simpler virus-like particles incorporating the lentiviral protein Vpx to efficiently and transiently degrade SAMHD1 and increase the efficacy of Ara-C treatment. The addition of minute amounts of lentiviral Rev protein during production enhanced the generation of virus-like particles. In addition, we found that our 2nd generation of virus-like particles efficiently targeted and degraded SAMHD1 in AML cell lines with high levels of SAMHD1, thereby increasing Ara-CTP levels and response to Ara-C treatment. Primary AML blasts were generally less responsive to VLP treatment. In summary, we have been able to generate novel and simpler virus-like particles that can efficiently deliver Vpx to target cells.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/drug therapy , Cytarabine/pharmacology , Cytarabine/therapeutic use , SAM Domain and HD Domain-Containing Protein 1/metabolism , SAM Domain and HD Domain-Containing Protein 1/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Cell Line, Tumor , Lentivirus/genetics
8.
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
9.
PLoS One ; 19(6): e0297817, 2024.
Article in English | MEDLINE | ID: mdl-38833479

ABSTRACT

Lentiviral vectors derived from human immunodeficiency virus type I are widely used to deliver functional gene copies to mammalian cells for research and gene therapies. Post-transcriptional splicing of lentiviral vector transgene in transduced host and transfected producer cells presents barriers to widespread application of lentiviral vector-based therapies. The present study examined effects of indole derivative compound IDC16 on splicing of lentiviral vector transcripts in producer cells and corresponding yield of infectious lentiviral vectors. Indole IDC16 was shown previously to modify alternative splicing in human immunodeficiency virus type I. Human embryonic kidney 293T cells were transiently transfected by 3rd generation backbone and packaging plasmids using polyethyleneimine. Reverse transcription-quantitative polymerase chain reaction of the fraction of unspliced genomes in human embryonic kidney 293T cells increased up to 31% upon the indole's treatment at 2.5 uM. Corresponding yield of infectious lentiviral vectors decreased up to 4.5-fold in a cell transduction assay. Adjusting timing and duration of IDC16 treatment indicated that the indole's disruption of early stages of transfection and cell cycle had a greater effect on exponential time course of lentiviral vector production than its reduction of post-transcriptional splicing. Decrease in transfected human embryonic kidney 293T proliferation by IDC16 became significant at 10 uM. These findings indicated contributions by early-stage transfection, cell proliferation, and post-transcriptional splicing in transient transfection of human embryonic kidney 293T cells for lentiviral vector production.


Subject(s)
Alternative Splicing , Cell Proliferation , Genetic Vectors , Indoles , Lentivirus , Transfection , Humans , Indoles/pharmacology , Cell Proliferation/drug effects , Genetic Vectors/genetics , Lentivirus/genetics , Transfection/methods , HEK293 Cells
10.
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
11.
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
12.
Cells ; 13(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38920646

ABSTRACT

Dopaminergic neurons are the predominant brain cells affected in Parkinson's disease. With the limited availability of live human brain dopaminergic neurons to study pathological mechanisms of Parkinson's disease, dopaminergic neurons have been generated from human-skin-cell-derived induced pluripotent stem cells. Originally, induced pluripotent stem-cell-derived dopaminergic neurons were generated using small molecules. These neurons took more than two months to mature. However, the transcription-factor-mediated differentiation of induced pluripotent stem cells has revealed quicker and cheaper methods to generate dopaminergic neurons. In this study, we compared and contrasted three protocols to generate induced pluripotent stem-cell-derived dopaminergic neurons using transcription-factor-mediated directed differentiation. We deviated from the established protocols using lentivirus transduction to stably integrate different transcription factors into the AAVS1 safe harbour locus of induced pluripotent stem cells. We used different media compositions to generate more than 90% of neurons in the culture, out of which more than 85% of the neurons were dopaminergic neurons within three weeks. Therefore, from our comparative study, we reveal that a combination of transcription factors along with small molecule treatment may be required to generate a pure population of human dopaminergic neurons.


Subject(s)
Cell Differentiation , Dopaminergic Neurons , Induced Pluripotent Stem Cells , Transcription Factors , Humans , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Lentivirus/genetics , Lentivirus/metabolism
13.
Viruses ; 16(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38932120

ABSTRACT

A gene delivery system utilizing lentiviral vectors (LVs) requires high transduction efficiency for successful application in human gene therapy. Pseudotyping allows viral tropism to be expanded, widening the usage of LVs. While vesicular stomatitis virus G (VSV-G) single-pseudotyped LVs are commonly used, dual-pseudotyping is less frequently employed because of its increased complexity. In this study, we examined the potential of phenotypically mixed heterologous dual-pseudotyped LVs with VSV-G and Sendai virus hemagglutinin-neuraminidase (SeV-HN) glycoproteins, termed V/HN-LV. Our findings demonstrated the significantly improved transduction efficiency of V/HN-LV in various cell lines of mice, cynomolgus monkeys, and humans compared with LV pseudotyped with VSV-G alone. Notably, V/HN-LV showed higher transduction efficiency in human cells, including hematopoietic stem cells. The efficient incorporation of wild-type SeV-HN into V/HN-LV depended on VSV-G. SeV-HN removed sialic acid from VSV-G, and the desialylation of VSV-G increased V/HN-LV infectivity. Furthermore, V/HN-LV acquired the ability to recognize sialic acid, particularly N-acetylneuraminic acid on the host cell, enhancing LV infectivity. Overall, VSV-G and SeV-HN synergistically improve LV transduction efficiency and broaden its tropism, indicating their potential use in gene delivery.


Subject(s)
Genetic Vectors , HN Protein , Lentivirus , Sendai virus , Transduction, Genetic , Viral Envelope Proteins , Animals , Humans , Genetic Vectors/genetics , Lentivirus/genetics , Sendai virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Mice , HN Protein/genetics , HN Protein/metabolism , Cell Line , Macaca fascicularis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Viral Tropism , HEK293 Cells , Gene Transfer Techniques , Genetic Therapy/methods
14.
J Nanobiotechnology ; 22(1): 329, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858736

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) play a vital role in the occurrence, maintenance, and recurrence of solid tumors. Although, miR-145-5p can inhibit CSCs survival, poor understanding of the underlying mechanisms hamperes further therapeutic optimization for patients. Lentivirus with remarkable transduction efficiency is the most commonly used RNA carrier in research, but has shown limited tumor-targeting capability. METHODS: We have applied liposome to decorate lentivirus surface thereby yielding liposome-lentivirus hybrid-based carriers, termed miR-145-5p-lentivirus nanoliposome (MRL145), and systematically analyzed their potential therapeutic effects on liver CSCs (LCSCs). RESULTS: MRL145 exhibited high delivery efficiency and potent anti-tumor efficacy under in vitro and in vivo. Mechanistically, the overexpressed miR-145-5p can significantly suppress the self-renewal, migration, and invasion abilities of LCSCs by targeting Collagen Type IV Alpha 3 Chain (COL4A3). Importantly, COL4A3 can promote phosphorylating GSK-3ß at ser 9 (p-GSK-3ß S9) to inactivate GSK3ß, and facilitate translocation of ß-catenin into the nucleus to activate the Wnt/ß-catenin pathway, thereby promoting self-renewal, migration, and invasion of LCSCs. Interestingly, COL4A3 could attenuate the cellular autophagy through modulating GSK3ß/Gli3/VMP1 axis to promote self-renewal, migration, and invasion of LCSCs. CONCLUSIONS: These findings provide new insights in mode of action of miR-145-5p in LCSCs therapy and indicates that liposome-virus hybrid carriers hold great promise in miRNA delivery.


Subject(s)
Lentivirus , Liposomes , MicroRNAs , Neoplastic Stem Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Liposomes/chemistry , Humans , Animals , Mice , Lentivirus/genetics , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Mice, Nude , Liver Neoplasms/therapy , Mice, Inbred BALB C , Cell Movement , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/metabolism , Wnt Signaling Pathway
15.
J Neurosci Methods ; 409: 110197, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38878976

ABSTRACT

BACKGROUND: Trigeminal ganglion (TG) plays an important role in the process of orthodontic pain. It's necessary to design an accurate, precise and minimally invasive trigeminal ganglion injection guide plate to study TG. METHODS: Micro-CT was used to obtain the Dicom format data, and three-dimensional (3D) software (mimics and magics23.03) was used to reconstruct 3D head models. Design and modifications of the TG injection guide plate were performed in Magic 23.03 software, and the guide plate was produced by a 3D stereolithography printer. X-ray, micro-CT, Evans blue, and virus transduction were used to demonstrate the accuracy of the guide-assisted injection. Pain levels were evaluated after using the injection guide by a bite force test and Von Frey test. RESULTS: X-ray and micro-CT tests confirmed that the injection needle reached the bilateral trigeminal ganglia fossa. The Evans blue test and virus transduction proved that the injected drug could be accurately injected into the bilateral trigeminal ganglion and the lentivirus could be successfully transfected. The percentage of accurate injection was 10/10 (bilateral trigeminal ganglia). Orofacial pain induced by the trigeminal ganglion injection was mild and returned to baseline within seven days. CONCLUSION: The injection guide described in this study is viable and reliable for the delivery of drugs and virus transduction into the trigeminal ganglia.


Subject(s)
Trigeminal Ganglion , Trigeminal Ganglion/diagnostic imaging , Animals , X-Ray Microtomography/methods , Male , Rats, Sprague-Dawley , Imaging, Three-Dimensional/methods , Injections , Printing, Three-Dimensional , Rats , Facial Pain/diagnostic imaging , Lentivirus/genetics , Software
16.
Biotechnol Bioeng ; 121(9): 2936-2951, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38874319

ABSTRACT

The demand for Lentiviral Vector (LV) drug substance is increasing. However, primary capture using convective anion-exchange chromatography remains a significant manufacturing challenge. This stems from a poor understanding of the complex adsorption behaviors linked to LVs intricate and variable structure, such as high binding heterogeneity which is typically characterized by a gradient elution profile consisting of two peaks. Understanding which LV structural components drive these phenomena is therefore crucial for rational process design. This work identifies the key LV envelope components responsible for binding to quaternary-amine membrane adsorbents. Eliminating the pseudotype protein (Vesicular Stomatitis Virus G glycoprotein [VSV-G]) did not impact the heterogenous two-peak elution profile, suggesting it is not a major binding species. Digestion of envelope glycosaminoglycans (GAGs), present on proteoglycans, leads to a dramatic reduction in the proportion of vector eluted in peak 2, decreasing from 50% to 3.1%, and a threefold increase in peak 1 maximum. Data from reinjection experiments point towards interparticle envelope heterogeneity from discrete LV populations, where the two-peak profile emerges from a subpopulation of LVs interacting via highly charged GAGs (peak 2) along with a weaker binding population likely interacting through the phospholipid membrane and envelope protein (peak 1).


Subject(s)
Genetic Vectors , Lentivirus , Chromatography, Ion Exchange/methods , Lentivirus/genetics , Genetic Vectors/genetics , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
17.
Cells ; 13(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38891052

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare genetic disorder affecting the bone marrow's ability to produce red blood cells, leading to severe anemia and various physical abnormalities. Approximately 75% of DBA cases involve heterozygous mutations in ribosomal protein (RP) genes, classifying it as a ribosomopathy, with RPS19 being the most frequently mutated gene. Non-RP mutations, such as in GATA1, have also been identified. Current treatments include glucocorticosteroids, blood transfusions, and hematopoietic stem cell transplantation (HSCT), with HSCT being the only curative option, albeit with challenges like donor availability and immunological complications. Gene therapy, particularly using lentiviral vectors and CRISPR/Cas9 technology, emerges as a promising alternative. This review explores the potential of gene therapy, focusing on lentiviral vectors and CRISPR/Cas9 technology in combination with non-integrating lentiviral vectors, as a curative solution for DBA. It highlights the transformative advancements in the treatment landscape of DBA, offering hope for individuals affected by this condition.


Subject(s)
Anemia, Diamond-Blackfan , Genetic Therapy , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/therapy , Genetic Therapy/methods , Humans , CRISPR-Cas Systems/genetics , Genetic Vectors , Lentivirus/genetics , Animals , Ribosomal Proteins/genetics , Mutation/genetics , Gene Editing/methods
18.
Front Immunol ; 15: 1404668, 2024.
Article in English | MEDLINE | ID: mdl-38903492

ABSTRACT

Heart transplantation is associated with major hurdles, including the limited number of available organs for transplantation, the risk of rejection due to genetic discrepancies, and the burden of immunosuppression. In this study, we demonstrated the feasibility of permanent genetic engineering of the heart during ex vivo perfusion. Lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin (shß2m) and class II transactivator (shCIITA) were delivered to the graft during two hours of normothermic EVHP. Highly efficient genetic engineering was indicated by stable reporter gene expression in endothelial cells and cardiomyocytes. Remarkably, swine leucocyte antigen (SLA) class I and SLA class II expression levels were decreased by 66% and 76%, respectively, in the vascular endothelium. Evaluation of lactate, troponin T, and LDH levels in the perfusate and histological analysis showed no additional cell injury or tissue damage caused by lentiviral vectors. Moreover, cytokine secretion profiles (IL-6, IL-8, and TNF-α) of non-transduced and lentiviral vector-transduced hearts were comparable. This study demonstrated the ex vivo generation of genetically engineered hearts without compromising tissue integrity. Downregulation of SLA expression may contribute to reduce the immunogenicity of the heart and support graft survival after allogeneic or xenogeneic transplantation.


Subject(s)
Genetic Vectors , Heart Transplantation , Histocompatibility Antigens Class I , Lentivirus , Animals , Lentivirus/genetics , Heart Transplantation/methods , Genetic Vectors/genetics , Swine , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Perfusion/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , beta 2-Microglobulin/genetics , Cytokines/metabolism , Genetic Engineering , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Humans , RNA, Small Interfering/genetics , Graft Survival/immunology , Graft Survival/genetics , Endothelial Cells/metabolism , Endothelial Cells/immunology , Nuclear Proteins , Trans-Activators
19.
Biomed Phys Eng Express ; 10(4)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38772344

ABSTRACT

Lentiviral transduction is widely used in research, has shown promise in clinical trials involving gene therapy and has been approved for CAR-T cell immunotherapy. However, most modifications are doneex vivoand rely on systemic administration of large numbers of transduced cells for clinical applications. A novel approach utilizingin situbiomaterial-based gene delivery can reduce off-target side effects while enhancing effectiveness of the manipulation process. In this study, poly(ethylene glycol) diacrylate (PEGDA)-based scaffolds were developed to enablein situlentivirus-mediated transduction. Compared to other widely popular biomaterials, PEGDA stands out due to its robustness and cost-effectiveness. These scaffolds, prepared via cryogelation, are capable of flowing through surgical needles in bothin vitroandin vivoconditions, and promptly regain their original shape. Modification with poly(L-lysine) (PLL) enables lentivirus immobilization while interconnected macroporous structure allows cell infiltration into these matrices, thereby facilitating cell-virus interaction over a large surface area for efficient transduction. Notably, these preformed injectable scaffolds demonstrate hemocompatibility, cell viability and minimally inflammatory response as shown by ourin vitroandin vivostudies involving histology and immunophenotyping of infiltrating cells. This study marks the first instance of using preformed injectable scaffolds for delivery of lentivectors, which offers a non-invasive and localized approach for delivery of factors enablingin situlentiviral transduction suitable for both tissue engineering and immunotherapeutic applications.


Subject(s)
Cryogels , Gene Transfer Techniques , Lentivirus , Polyethylene Glycols , Polyethylene Glycols/chemistry , Cryogels/chemistry , Humans , Lentivirus/genetics , Animals , Cell Survival/drug effects , Tissue Scaffolds/chemistry , Transduction, Genetic , Mice , Biocompatible Materials/chemistry , Genetic Therapy/methods , Surface Properties , Injections , Polylysine/chemistry
20.
BMC Vet Res ; 20(1): 195, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741095

ABSTRACT

Small ruminant lentiviruses (SRLVs) are widespread and infect goats and sheep. Several reports also suggest that SRLVs can infect wild ruminants. The presence of specific antibodies against SRLVs has been identified in wild ruminants from Poland, but no studies have been conducted to detect proviral DNA of SRLVs in these animals. Therefore, the purpose of this study was to examine samples from Polish wild ruminants to determine whether these animals can serve as reservoirs of SRLVs under natural conditions. A total of 314 samples were tested from red deer (n = 255), roe deer (n = 52) and fallow deer (n = 7) using nested real-time PCR. DNA from positive real-time PCR samples was subsequently used to amplify a CA fragment (625 bp) of the gag gene, a 1.2 kb fragment of the pol gene and an LTR-gag fragment. Three samples (0.95%) were positive according to nested real-time PCR using primers and probe specific for CAEV (SRLV group B). All the samples were negative for the primers and probe specific for MVV (SRLV A group). Only SRLV LTR-gag sequences were obtained from two red deer. Phylogenetic analysis revealed that these sequences were more closely related to CAEV than to MVV. Our results revealed that deer can carry SRLV proviral sequences and therefore may play a role in the epidemiology of SRLVs. To our knowledge, this is the first study describing SRLV sequences from red deer.


Subject(s)
DNA, Viral , Deer , Lentivirus Infections , Proviruses , Animals , Deer/virology , Poland/epidemiology , Proviruses/genetics , Lentivirus Infections/veterinary , Lentivirus Infections/virology , Lentivirus Infections/epidemiology , DNA, Viral/genetics , Lentivirus/isolation & purification , Lentivirus/genetics , Lentivirus/classification , Phylogeny , Real-Time Polymerase Chain Reaction/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL