Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Int J Mol Sci ; 22(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34947988

ABSTRACT

Recently, we have shown the molecular basis for lactate sensing by cervical epithelial cells resulting in enhanced DNA repair processes through DNA-PKcs regulation. Interestingly, DNA-PKcs is indispensable for proper retroviral DNA integration in the cell host genome. According to recent findings, the mucosal epithelium can be efficiently transduced by retroviruses and play a pivotal role in regulating viral release by cervical epithelial cells. This study examined the effects of lactate on lentiviral transduction in cervical cancer cells (HeLa, CaSki, and C33A) and model glioma cell lines (DNA-PKcs proficient and deficient). Our study showed that L- and D-lactate enhanced DNA-PKcs presence in nuclear compartments by between 38 and 63%, which corresponded with decreased lentiviral transduction rates by between 15 and 36%. Changes in DNA-PKcs expression or its inhibition with NU7441 also greatly affected lentiviral transduction efficacy. The stimulation of cells with either HCA1 agonist 3,5-DHBA or HDAC inhibitor sodium butyrate mimicked, in part, the effects of L-lactate. The inhibition of lactate flux by BAY-8002 enhanced DNA-PKcs nuclear localization which translated into diminished lentiviral transduction efficacy. Our study suggests that L- and D-lactate present in the uterine cervix may play a role in the mitigation of viral integration in cervical epithelium and, thus, restrict the viral oncogenic and/or cytopathic potential.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Glioma/virology , Lactic Acid/pharmacology , Lentivirus/physiology , Uterine Cervical Neoplasms/virology , Benzoates/pharmacology , Butyric Acid/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Chromones/pharmacology , Female , Glioma/metabolism , HeLa Cells , Humans , Lentivirus/drug effects , Morpholines/pharmacology , Transduction, Genetic , Uterine Cervical Neoplasms/metabolism
2.
Food Funct ; 12(22): 11537-11546, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34708225

ABSTRACT

Glycolipids are a group of sugar-containing lipids with versatile functions. In this study, a natural glycolipid product was obtained from soy lecithin, and its emulsifying, oil-gelling, antibacterial and antiviral properties were investigated. A silica-based extraction method on a preparative scale was used to recover the glycolipid product (GLP) from soy lecithin. The GLP consisted of three different glycolipid classes: acylated sterol glucoside (64.16%), sterol glucoside (25.57%) and cerebroside (6.71%). As an emulsifier, the GLP was able to form a stable water-in-oil emulsion. The GLP exhibited a good oil-gelling property, capable of gelling rapeseed oil at a concentration of 6%. For the investigated microorganisms (Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus), the GLP did not show any antibacterial effects. The GLP exerted antiviral activity against lentivirus, but not adenovirus. The results of this study help in enriching the knowledge on the properties of naturally occurring glycolipids, which may find potential applications in the food, pharmaceutical and related industries.


Subject(s)
Anti-Infective Agents , Biological Products , Glycolipids , Surface-Active Agents , Adenoviridae/drug effects , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Biological Products/chemistry , Biological Products/pharmacology , Emulsifying Agents/chemistry , Emulsifying Agents/pharmacology , Glycolipids/chemistry , Glycolipids/pharmacology , Lentivirus/drug effects , Rapeseed Oil/chemistry , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacology
3.
Virology ; 561: 6-16, 2021 09.
Article in English | MEDLINE | ID: mdl-34089997

ABSTRACT

Based on the previously reported 13-residue antibacterial peptide analog, brevinin-1EMa (FLGWLFKVASKVL, peptide B), we attempted to design a novel class of antiviral peptides. For this goal, we synthesized three peptides with different stapling positions (B-2S, B-8S, and B-5S). The most active antiviral peptide with the specific stapling position (B-5S) was further modified in combination with either cysteine (B-5S3C, B-5S7C, and B-5S10C) or hydrophilic amino acid substitution (Bsub and Bsub-5S). Overall, B, B-5S, and Bsub-5S peptides showed superior antiviral activities against enveloped viruses such as retrovirus, lentivirus, hepatitis C virus, and herpes simplex virus with EC50 values of 1-5 µM. Murine norovirus, a non-enveloped virus, was not susceptible to the virucidal actions of these peptides, suggesting the virus membrane disruption as their main antiviral mechanisms of action. We believe that these three novel peptides could serve as promising candidates for further development of membrane-targeting antiviral drugs in the future.


Subject(s)
Antiviral Agents/pharmacology , Ion Channels/chemistry , Ion Channels/pharmacology , Peptides/pharmacology , Virus Internalization/drug effects , Viruses/drug effects , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Bacteria/drug effects , Cell Line , Drug Design , Hepacivirus/drug effects , Hepacivirus/physiology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Ion Channels/metabolism , Lentivirus/drug effects , Lentivirus/physiology , Microbial Sensitivity Tests , Norovirus/drug effects , Norovirus/physiology , Peptides/chemistry , Peptides/metabolism , Retroviridae/drug effects , Retroviridae/physiology , Virus Physiological Phenomena
4.
J Gen Virol ; 102(4)2021 04.
Article in English | MEDLINE | ID: mdl-33830908

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has caused a pandemic with tens of millions of cases and more than a million deaths. The infection causes COVID-19, a disease of the respiratory system of divergent severity. No treatment exists. Epigallocatechin-3-gallate (EGCG), the major component of green tea, has several beneficial properties, including antiviral activities. Therefore, we examined whether EGCG has antiviral activity against SARS-CoV-2. EGCG blocked not only the entry of SARS-CoV-2, but also MERS- and SARS-CoV pseudotyped lentiviral vectors and inhibited virus infections in vitro. Mechanistically, inhibition of the SARS-CoV-2 spike-receptor interaction was observed. Thus, EGCG might be suitable for use as a lead structure to develop more effective anti-COVID-19 drugs.


Subject(s)
Antiviral Agents/pharmacology , Catechin/analogs & derivatives , SARS-CoV-2/drug effects , Tea/chemistry , Animals , Betacoronavirus/drug effects , Betacoronavirus/physiology , COVID-19/prevention & control , COVID-19/virology , Catechin/pharmacology , Cell Survival/drug effects , Chlorocebus aethiops , HEK293 Cells , Humans , Lentivirus/drug effects , Lentivirus/genetics , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/genetics , Vero Cells , Virus Attachment/drug effects , Virus Replication/drug effects
5.
Transgenic Res ; 30(1): 63-76, 2021 02.
Article in English | MEDLINE | ID: mdl-33394315

ABSTRACT

Taking advantage of their ability to integrate their genomes into the host genome, lentiviruses have been used to rapidly produce transgenic mice in biomedical research. In most cases, transgenes delivered by lentiviral vectors have resisted silencing mediated by epigenetic modifications in mice. However, some studies revealed that methylation caused decreased transgene expression in mice. Therefore, there is conflicting evidence regarding the methylation-induced silencing of transgenes delivered by lentiviral transduction in mice. In this study, we present evidence that the human TTR transgene was silenced by DNA methylation in the liver of a transgenic mouse model generated by lentiviral transduction. The density of methylation on the transgene was increased during reproduction, and the expression of the transgene was completely silenced in mice of the F2 generation. Interestingly, 5-azacytidine (5-AzaC), a methyltransferase inhibitor, potently reactivated the silenced genes in neonatal mice whose hepatocytes were actively proliferating and led to stable transgene expression during development. However, 5-AzaC did not rescue liver transgene expression when administered to adult mice. Moreover, 5-AzaC at the given dose had low developmental toxicity in the newborn mice. In summary, we demonstrate the methylation-induced silencing of an exogenous gene in the liver of a mouse model generated by lentiviral transduction and show that the silenced transgene can be safely and efficiently reactivated by 5-AzaC treatment, providing an alternative way to obtain progeny with stable transgene expression in the case of the methylation of exogenous genes in transgenic mice generated by lentiviral transduction.


Subject(s)
DNA Methylation/genetics , Lentivirus/genetics , Prealbumin/genetics , Transgenes/genetics , Animals , Animals, Newborn , Azacitidine/pharmacology , DNA Methylation/drug effects , Gene Expression Regulation, Developmental/drug effects , Genetic Vectors/drug effects , Humans , Lentivirus/drug effects , Mice , Mice, Transgenic/genetics
8.
J Virol Methods ; 276: 113768, 2020 02.
Article in English | MEDLINE | ID: mdl-31704112

ABSTRACT

Lentiviral vectors and lentiviruses are important tools for basic and applied biomedical research. Yet, biosafety regulations from legal authorities have to be fulfilled when transferring BSL-2 to -3 vectors/viruses to facilities with lower biosafety level. Here, we (re-)evaluated different chemical and thermal approaches to inactivate vesicular stomatitis virus G-protein (VSV-G) pseudotyped lentiviral vectors and either wildtype or VSV-G pseudotyped human immunodeficiency viruses (HIV). Aldehydes, detergents and alcohols were as effective as thermal inactivation procedures to efficiently inactivate purified lentiviral vectors and replication-competent HIV. In addition, no residual infectivity was detected when inactivating HIV-infected TZM-bl reporter cells with selected detergents and aldehydes. Thus, our established inactivation protocols can be used by other laboratories working with lentiviral vectors or infectious lentiviruses and provide a template for viruses with similar physicochemical properties.


Subject(s)
Genetic Vectors/drug effects , HIV/drug effects , Lentivirus/drug effects , Virus Inactivation/drug effects , Alcohols/pharmacology , Aldehydes/pharmacology , Detergents/pharmacology , HEK293 Cells , HIV/pathogenicity , Hot Temperature , Humans , Lentivirus/physiology
9.
Viruses ; 11(10)2019 10 09.
Article in English | MEDLINE | ID: mdl-31600877

ABSTRACT

Sterile alpha motif and histidine/aspartic domain-containing protein 1 (SAMHD1) is a protein with anti-viral, anti-neoplastic, and anti-inflammatory properties. By degrading cellular dNTPs to constituent deoxynucleoside and free triphosphate, SAMHD1 limits viral DNA synthesis and prevents replication of HIV-1 and some DNA viruses such as HBV, vaccinia, and HSV-1. Recent findings suggest SAMHD1 is broadly active against retroviruses in addition to HIV-1, such as HIV-2, FIV, BIV, and EIAV. Interferons are cytokines produced by lymphocytes and other cells that induce a wide array of antiviral proteins, including some with activity again lentiviruses. Here we evaluated the role of IFNs on SAMHD1 gene expression, transcription, and post-translational modification in a feline CD4+ T cell line (FeTJ) and in primary feline CD4+ T lymphocytes. SAMHD1 mRNA in FetJ cells increased in a dose-related manner in response to IFNγ treatment concurrent with increased nuclear localization and phosphorylation. IFNα treatment induced SAMHD1 mRNA but did not significantly alter SAMHD1 protein detection, phosphorylation, or nuclear translocation. In purified primary feline CD4+ lymphocytes, IL2 supplementation increased SAMHD1 expression, but the addition of IFNγ did not further alter SAMHD1 protein expression or nuclear localization. Thus, the effect of IFNγ on SAMHD1 expression is cell-type dependent, with increased translocation to the nucleus and phosphorylation in FeTJ but not primary CD4+ lymphocytes. These findings imply that while SAMH1 is inducible by IFNγ, overall activity is cell type and compartment specific, which is likely relevant to the establishment of lentiviral reservoirs in quiescent lymphocyte populations.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , SAM Domain and HD Domain-Containing Protein 1/drug effects , Animals , Antiviral Agents/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cats , Cell Line , Gene Expression/drug effects , Interleukin-2/metabolism , Lentivirus/drug effects , Lentivirus/growth & development , Phosphorylation/drug effects , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Virus Replication/drug effects
10.
Cell Stem Cell ; 23(6): 820-832.e9, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30416070

ABSTRACT

Innate immune factors may restrict hematopoietic stem cell (HSC) genetic engineering and contribute to broad individual variability in gene therapy outcomes. Here, we show that HSCs harbor an early, constitutively active innate immune block to lentiviral transduction that can be efficiently overcome by cyclosporine H (CsH). CsH potently enhances gene transfer and editing in human long-term repopulating HSCs by inhibiting interferon-induced transmembrane protein 3 (IFITM3), which potently restricts VSV glycoprotein-mediated vector entry. Importantly, individual variability in endogenous IFITM3 levels correlated with permissiveness of HSCs to lentiviral transduction, suggesting that CsH treatment will be useful for improving ex vivo gene therapy and standardizing HSC transduction across patients. Overall, our work unravels the involvement of innate pathogen recognition molecules in immune blocks to gene correction in primary human HSCs and highlights how these roadblocks can be overcome to develop innovative cell and gene therapies.


Subject(s)
Cyclosporine/pharmacology , Gene Editing , Hematopoietic Stem Cells/drug effects , Immunity, Innate/drug effects , Lentivirus/drug effects , Lentivirus/genetics , Transduction, Genetic , Animals , Cell Line , Female , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/immunology , Mice , Mice, Inbred NOD , Mice, Knockout
11.
Methods Mol Biol ; 1836: 139-158, 2018.
Article in English | MEDLINE | ID: mdl-30151572

ABSTRACT

Our understanding of infection biology is based on experiments in which pathogen or host proteins are perturbed by small compound inhibitors, mutation, or depletion. This approach has been remarkably successful, as, for example, demonstrated by the independent identification of the endosomal membrane protein Niemann-Pick C1 as an essential factor for Ebola virus infection in both small compound and insertional mutagenesis screens (Côté, Nature 477:344-348, 2011; Carette et al., Nature 477:340-343, 2011). However, many aspects of host-pathogen interactions are poorly understood because we cannot target all of the involved molecules with small molecules, or because we cannot deplete essential proteins. Single domain antibody fragments expressed in the cytosol or other organelles constitute a versatile alternative to perturb the function of any given protein by masking protein-protein interaction interfaces, by stabilizing distinct conformations, or by directly interfering with enzymatic activities. The variable domains of heavy chain-only antibodies (VHHs) from camelid species can be cloned from blood samples of animals immunized with the desired target molecules. We can thus exploit the ability of the camelid immune system to generate affinity-matured single domain antibody fragments to obtain highly specific tools. Interesting VHH candidates are typically identified based on their affinity toward immobilized antigens using techniques such as phage display.The phenotypical screening approach described here allows the direct identification of VHHs that prevent infection of cells with influenza A virus (IAV) or other pathogens. The VHH repertoire is cloned into a lentiviral vector, which is used to generate pseudo-typed lentivirus particles. Target cells are transduced with the lentivirus, so that every cell inducibly expresses a different VHH. This cell collection is then challenged with a lethal dose of virus. Only the cells which express a VHH that prevents infection by targeting virus proteins or host cell components essential for infection will survive. We can thus identify critical target molecules including vulnerable epitopes and conformations, render target molecules accessible to informative perturbation studies, and stabilize intermediates of virus entry for detailed analysis.


Subject(s)
Anti-Retroviral Agents/pharmacology , Lentivirus/drug effects , Phenotype , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Cell Line , Drug Evaluation, Preclinical/methods , Gene Library , Genetic Vectors/genetics , Humans , Influenza A virus/genetics , Lentivirus/genetics , Lentivirus Infections/drug therapy , Lentivirus Infections/virology , Microbial Sensitivity Tests , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics
12.
J Vis Exp ; (131)2018 01 15.
Article in English | MEDLINE | ID: mdl-29364266

ABSTRACT

The efficient transduction of specific genes into natural killer (NK) cells has been a major challenge. Successful transductions are critical to defining the role of the gene of interest in the development, differentiation, and function of NK cells. Recent advances related to chimeric antigen receptors (CARs) in cancer immunotherapy accentuate the need for an efficient method to deliver exogenous genes to effector lymphocytes. The efficiencies of lentiviral-mediated gene transductions into primary human or mouse NK cells remain significantly low, which is a major limiting factor. Recent advances using cationic polymers, such as polybrene, show an improved gene transduction efficiency in T cells. However, these products failed to improve the transduction efficiencies of NK cells. This work shows that dextran, a branched glucan polysaccharide, significantly improves the transduction efficiency of human and mouse primary NK cells. This highly reproducible transduction methodology provides a competent tool for transducing human primary NK cells, which can vastly improve clinical gene delivery applications and thus NK cell-based cancer immunotherapy.


Subject(s)
Dextrans/pharmacology , Genetic Therapy/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lentivirus/drug effects , Lentivirus/genetics , Animals , Humans , Mice
13.
Biotechniques ; 63(3): 136-138, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911318

ABSTRACT

To enable simple and effective high titer recombinant lentivirus production, we examined key parameters for the generation of lentivirus including: transfection optimization, media change, incubation time and DNA vector selection. These results illustrate the importance of optimizing transfection processes for high titer recombinant lentivirus production.


Subject(s)
Lentivirus/growth & development , Viral Load , Virus Cultivation/methods , Culture Media/pharmacology , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Lentivirus/drug effects , Lentivirus/genetics , Time Factors , Transfection , Viral Load/drug effects , Viral Proteins/metabolism
14.
Hum Gene Ther Methods ; 28(6): 330-339, 2017 12.
Article in English | MEDLINE | ID: mdl-28826344

ABSTRACT

Lentiviral vectors (LV) represent a key tool for gene and cell therapy applications. The production of these vectors in sufficient quantities for clinical applications remains a hurdle, prompting the field toward developing suspension processes that are conducive to large-scale production. This study describes a LV production strategy using a stable inducible producer cell line. The HEK293 cell line employed grows in suspension, thus offering direct scalability, and produces a green fluorescent protein (GFP)-expressing lentiviral vector in the 106 transduction units (TU)/mL range without optimization. The stable producer cell line, called clone 92, was derived by stable transfection from a packaging cell line with a plasmid encoding the transgene GFP. The packaging cell line expresses all the other necessary components to produce LV upon induction with cumate and doxycycline. First, the study demonstrated that LV production using clone 92 is scalable from 20 mL shake flasks to 3 L bioreactors. Next, two strategies were developed for high-yield LV production in perfusion mode using acoustic cell filter technology in 1-3 L bioreactors. The first approach uses a basal commercial medium and perfusion mode both pre- and post-induction for increasing cell density and LV recovery. The second approach makes use of a fortified medium formulation to achieve target cell density for induction in batch mode, followed by perfusion mode after induction. Using these perfusion-based strategies, the titer was improved to 3.2 × 107 TU/mL. As a result, cumulative functional LV titers were increased by up to 15-fold compared to batch mode, reaching a cumulative total yield of 8 × 1010 TU/L of bioreactor culture. This approach is easily amenable to large-scale production and commercial manufacturing.


Subject(s)
Biotechnology/methods , Cell Culture Techniques/methods , Genetic Vectors/genetics , Lentivirus/physiology , Transduction, Genetic/methods , Virus Cultivation/methods , Benzoates/pharmacology , Bioreactors , Doxycycline/pharmacology , HEK293 Cells , Humans , Lentivirus/drug effects , Lentivirus/genetics
15.
Peptides ; 88: 87-96, 2017 02.
Article in English | MEDLINE | ID: mdl-28012857

ABSTRACT

Antimicrobial peptides are currently considered as promising antiviral compounds. Current assays to evaluate the effectivity of peptides against enveloped viruses based on liposomes or hemolysis are encumbered by the artificial nature of liposomes or distinctive membrane composition of used erythrocytes. We propose a novel assay system based on enzymatic Ebola virus-like particles containing sensitive luciferase reporter. The assay was validated with several cationic and anionic peptides and compared with lentivirus inactivation and hemolytic assays. The assay is sensitive and easy to perform in standard biosafety level laboratory with potential for high-throughput screens. The use of virus-like particles in the assay provides a system as closely related to the native viruses as possible eliminating some issues associated with other more artificial set ups. We have identified CAM-W (KWKLWKKIEKWGQGIGAVLKWLTTWL) as a peptide with the greatest antiviral activity against infectious lentiviral vectors and filoviral virus-like particles.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/prevention & control , Peptides/pharmacology , Anions , Antiviral Agents/pharmacology , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Humans , Lentivirus/drug effects , Lentivirus/genetics , Liposomes/chemistry , Vaccines, Virus-Like Particle
16.
Nat Chem Biol ; 12(5): 361-6, 2016 May.
Article in English | MEDLINE | ID: mdl-27018887

ABSTRACT

Broad-spectrum antiviral drugs targeting host processes could potentially treat a wide range of viruses while reducing the likelihood of emergent resistance. Despite great promise as therapeutics, such drugs remain largely elusive. Here we used parallel genome-wide high-coverage short hairpin RNA (shRNA) and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 screens to identify the cellular target and mechanism of action of GSK983, a potent broad-spectrum antiviral with unexplained cytotoxicity. We found that GSK983 blocked cell proliferation and dengue virus replication by inhibiting the pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase (DHODH). Guided by mechanistic insights from both genomic screens, we found that exogenous deoxycytidine markedly reduced GSK983 cytotoxicity but not antiviral activity, providing an attractive new approach to improve the therapeutic window of DHODH inhibitors against RNA viruses. Our results highlight the distinct advantages and limitations of each screening method for identifying drug targets, and demonstrate the utility of parallel knockdown and knockout screens for comprehensive probing of drug activity.


Subject(s)
Antiviral Agents/pharmacology , CRISPR-Cas Systems/genetics , Carbazoles/pharmacology , Lentivirus/drug effects , RNA, Small Interfering/genetics , Carbazoles/chemistry , Cell Line, Tumor , Cloning, Molecular , Humans , Lentivirus/physiology
17.
Antiviral Res ; 124: 122-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26542648

ABSTRACT

Amiodarone and other cationic amphiphilic drugs (CADs) inhibit cell entry by diverse human pathogenic viruses including Filoviruses, Dengue virus and Japanese encephalitis virus. They are thus considered potential broad spectrum antiviral agents. Here we report the unexpected finding that amiodarone and other CADs markedly enhance rabies virus (RABV) glycoprotein- (GP-) mediated cell entry of pseudotyped lentiviruses into non-neuronal cells but not in neuronal cells. Increased cell entry can also be elicited when CADs are added several hours after pseudoviral attachment. Perturbing endosomal processing with phosphoinosite-3-kinase inhibitors wortmannin and LY294002 mimics the effects of CADs on RABV GP-mediated cell entry. Thus, CADs may enhance RABV GP-mediated cell entry of pseudotyped lentiviruses by promoting a late step of the pseudoviral cell entry process, possibly release from an endosomal compartment into the cytosol. In contrast to the pseudotyped lentiviruses, infection by fully infectious RABV was not enhanced by CADs, indicating, that the observed stimulation of RABV GP mediated lentivirus entry also depended on the used lentivirus vector backbone. In conclusion, we show that while CADs inhibit cell entry of diverse viruses they can also have a paradoxical enhancing effect on the ability of a viral glycoprotein to mediate cell entry depending on the cellular and viral context. Although, we show CAD-mediated enhancement of entry only for pseudoviruses, but not fully infectious RABV, the potential to unexpectedly enhance viral entry should be taken into account when considering use of CADs as antiviral agents.


Subject(s)
Antiviral Agents/pharmacology , Glycoproteins/metabolism , Lentivirus/drug effects , Rabies virus/physiology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Caco-2 Cells , Cell Line , Endosomes/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Rabies/drug therapy , Rabies/virology , Rabies virus/drug effects , Receptors, Adrenergic/metabolism , Vision, Ocular
18.
Trends Biochem Sci ; 40(2): 108-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25555456

ABSTRACT

To achieve productive infection, retroviruses such as HIV stably integrate their reverse transcribed RNA genome into a host chromosome. Each retroviral family preferentially integrates near a unique subset of genomic features. HIV integrase (IN) is targeted to the body of active transcription units through interaction with lens epithelium-derived growth factor (LEDGF/p75). We describe the successful effort to develop inhibitors of the interaction between IN and LEDGF/p75, referred to as LEDGINs. Gammaretroviruses display a distinct integration pattern. Recently, BET (bromo- and extraterminal domain) proteins were identified as the LEDGF/p75 counterparts that target the integration of gammaretroviruses. The identification of the chromatin-readers LEDGF/p75 and BET as cellular cofactors that orchestrate lentiviral or gammaretroviral integration opens new avenues to developing safer viral vectors for gene therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , HIV Infections/drug therapy , HIV Integrase Inhibitors/administration & dosage , Transcription Factors/metabolism , Virus Integration/genetics , Adaptor Proteins, Signal Transducing/genetics , Chromatin/drug effects , Gammaretrovirus/drug effects , Gammaretrovirus/genetics , Gammaretrovirus/pathogenicity , HIV Infections/virology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/pathogenicity , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Lentivirus/drug effects , Lentivirus/genetics , Lentivirus/pathogenicity , Transcription Factors/genetics , Virus Integration/drug effects
20.
Blood ; 124(6): 913-23, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24914132

ABSTRACT

Transplantation of genetically modified hematopoietic stem cells (HSCs) is a promising therapeutic strategy for genetic diseases, HIV, and cancer. However, a barrier for clinical HSC gene therapy is the limited efficiency of gene delivery via lentiviral vectors (LVs) into HSCs. We show here that rapamycin, an allosteric inhibitor of the mammalian target of rapamycin complexes, facilitates highly efficient lentiviral transduction of mouse and human HSCs and dramatically enhances marking frequency in long-term engrafting cells in mice. Mechanistically, rapamycin enhanced postbinding endocytic events, leading to increased levels of LV cytoplasmic entry, reverse transcription, and genomic integration. Despite increasing LV copy number, rapamycin did not significantly alter LV integration site profile or chromosomal distribution in mouse HSCs. Rapamycin also enhanced in situ transduction of mouse HSCs via direct intraosseous infusion. Collectively, rapamycin strongly augments LV transduction of HSCs in vitro and in vivo and may prove useful for therapeutic gene delivery.


Subject(s)
Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Lentivirus/drug effects , Lentivirus/genetics , Sirolimus/pharmacology , Transduction, Genetic/methods , Animals , Genetic Vectors/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/virology , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Virus Internalization/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...