Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Virol ; 96(2): e0134821, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34705555

ABSTRACT

The surface envelope glycoproteins of nonprimate lentiviruses and betaretroviruses share sequence similarity with the inner proximal domain ß-sandwich of the human immunodeficiency virus type 1 (HIV-1) gp120 glycoprotein that faces the transmembrane glycoprotein as well as patterns of cysteine and glycosylation site distribution that points to a similar two-domain organization in at least some lentiviruses. Here, high-reliability models of the surface glycoproteins obtained with the AlphaFold algorithm are presented for the gp135 glycoprotein of the small ruminant caprine arthritis-encephalitis (CAEV) and visna lentiviruses and the betaretroviruses Jaagsiekte sheep retrovirus (JSRV), mouse mammary tumor virus (MMTV), and consensus human endogenous retrovirus type K (HERV-K). The models confirm and extend the inner domain structural conservation in these viruses and identify two outer domains with a putative receptor binding site in the CAEV and visna virus gp135. The location of that site is consistent with patterns of sequence conservation and glycosylation site distribution in gp135. In contrast, a single domain is modeled for the JSRV, MMTV, and HERV-K betaretrovirus envelope proteins that is highly conserved structurally in the proximal region and structurally diverse in apical regions likely to interact with cell receptors. The models presented here identify sites in small ruminant lentivirus and betaretrovirus envelope glycoproteins likely to be critical for virus entry and virus neutralization by antibodies and will facilitate their functional and structural characterization. IMPORTANCE Structural information on the surface envelope proteins of lentiviruses and related betaretroviruses is critical to understand mechanisms of virus-host interactions. However, experimental determination of these structures has been challenging, and only the structure of the human immunodeficiency virus type 1 gp120 has been determined. The advent of the AlphaFold artificial intelligence method for structure prediction allows high-quality modeling of the structures of small ruminant lentiviral and betaretroviral surface envelope proteins. The models are consistent with much of the previously described experimental data, show regions likely to interact with receptors, and identify domains that may be involved in mechanisms of antibody neutralization resistance in the small ruminant lentiviruses. The models will allow more precise design of mutants to further determine mechanisms of viral entry and immune evasion in this group of viruses and constructs for structural determination of these surface envelope proteins.


Subject(s)
Algorithms , Betaretrovirus/chemistry , Gene Products, env/chemistry , Lentivirus/chemistry , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Endogenous Retroviruses/chemistry , Gene Products, env/metabolism , Humans , Models, Molecular , Protein Binding , Protein Domains , Receptors, Virus/metabolism , Ruminants
2.
Biochem Biophys Res Commun ; 513(4): 933-939, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31003777

ABSTRACT

The lentiviral accessory protein Vpx enhances viral replication in macrophages, dendritic cells and resting CD4+ T cells by utilizing the host CRL4-DCAF1 E3 ligase to trigger the degradation of the intrinsic antiviral factor SAMHD1. Distinct from the species-specific recognition of either the N or C-terminus of SAMHD1 by Vpx proteins of different HIV-2 and SIV lineages, Vpx recruits SAMHD1 onto the same CRL4-DCAF1 complex. However, the determinants in DCAF1 that are required for Vpx-mediated SAMHD1 degradation have not been well characterized. Here, we demonstrate that the viral protein Vpx is resistant to suppression by a cellular inhibitor of the CRL4-DCAF1 E3 ligase, Merlin/NF2, through targeting a separate binding region in DCAF1. The Merlin binding-deficient DCAF1 truncation mutant (1-1417) is sufficient for Vpx-CRL4-DCAF1 E3 ligase assembly and SAMHD1 degradation. We found that the carboxyl-terminus ED-rich region (1312-1417) of DCAF1 is required for the nuclear localization of DCAF1 and for the Vpx-DCAF1 interaction. We identified the DCAF1 (1-1311) truncation mutant as a dominant negative mutant of wild-type DCAF1 that inhibits Vpx-mediated SAMHD1 degradation. These results suggest a unique strategy by which Vpx exploits DCAF1 to counteract this host restriction factor.


Subject(s)
Lentivirus/chemistry , Protein Serine-Threonine Kinases/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Adaptor Proteins, Signal Transducing , Binding Sites , Cell Line , Cell Nucleus/metabolism , Host-Pathogen Interactions , Humans , Mutant Proteins , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Virus Replication
3.
Arch Virol ; 164(4): 1193-1198, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30739201

ABSTRACT

Small-ruminant lentivirus (SRLV) infections are widespread in Poland, and circulation of subtypes A1, A12, A13, A16, A17, B1 and B2 has been documented. The aim of this study was to characterize the SRLV strains circulating in sheep and goats in mixed flocks in the Malopolska region, where the highest seroprevalence has been detected. Phylogenetic analysis revealed that most of the isolates from sheep belonged to subtype A13, suggesting that this subtype may be predominant in the Malopolska region. Furthermore, the existence of a new subtype, tentatively designated as A18, was described for the first time. This work extends the current knowledge on the distribution of SRLV subtypes in sheep and goats in Poland and provides further information on the genetic diversity of SRLV. The new data are important for both epidemiological studies and eradication programs and provide insight into the evolution of SRLV.


Subject(s)
Goat Diseases/virology , Lentivirus Infections/veterinary , Lentivirus/genetics , Lentivirus/isolation & purification , Sheep Diseases/virology , Amino Acid Sequence , Animals , Gene Products, gag/chemistry , Gene Products, gag/genetics , Goats , Lentivirus/chemistry , Lentivirus/classification , Lentivirus Infections/virology , Molecular Sequence Data , Phylogeny , Poland , Sequence Alignment , Sheep
4.
Methods ; 157: 100-105, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30067943

ABSTRACT

We describe a method for visualizing mRNAs in living mouse. Nascent transcripts and cytoplasmic mRNAs were labeled via lentiviral expression of MS2 coat protein (MCP) tagged with fluorescent protein (MCP-XFP) in knock-in mice whose ß-actin mRNAs contained MCP binding stem loops (MBS). Then the mRNA molecules were imaged in the live cerebral cortex through an optical cranial window by intravital two-photon microscopy. By means of the controlled expression of MCP-XFP, single mRNA particles could be detected differentially in the nucleus and cytoplasm of a specific cell type. Consequently, this method is useful for investigating the cell-type-dependent dynamics of mRNAs underlying the structure and function of the brain.


Subject(s)
Brain/ultrastructure , Cell Tracking/methods , In Situ Hybridization, Fluorescence/methods , RNA, Messenger/isolation & purification , Animals , Cell Lineage/genetics , Lentivirus/chemistry , Lentivirus/genetics , Mice , RNA, Messenger/ultrastructure
5.
J Pharm Sci ; 107(11): 2764-2774, 2018 11.
Article in English | MEDLINE | ID: mdl-30017889

ABSTRACT

A replication-deficient lentiviral vector encoding the tumor antigen gene NY-ESO-1 was characterized in terms of vector morphology, particle size range, concentration, and zeta potential using a variety of physical methods. Environmentally stressed vector samples were then evaluated in terms of viral vector particle size and concentration by nanoparticle tracking analysis (NTA). These NTA stability results correlated reasonably well with a quantitative polymerase chain reaction assay for quantitation of viral genome copy number (r2 = 0.80). Approximately 40 pharmaceutical excipients were examined for their ability to stabilize the vector against exposure to an adsorptive container surface (glass) as well as freeze-thaw cycling using NTA as the screening method. Stabilizing additives that inhibited viral vector particle loss under these conditions included proline, lactose, and mannitol. Several candidate frozen liquid formulations that contained a combination of these lead excipients and various buffering agents were further evaluated for their ability to stabilize the viral vector. The additional benefit of lowering the Tris buffer concentration was observed. This study highlights the use of physical particle assays such as NTA for initial screening of stabilizing excipients to minimize vector loss due to container adsorption and freeze-thaw cycling to facilitate early formulation development of viral vector candidates in frozen liquid formulations.


Subject(s)
Excipients/chemistry , Genetic Vectors/chemistry , Lentivirus/chemistry , Adsorption , Antigens, Neoplasm/genetics , Cryopreservation , Drug Compounding , Freezing , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Membrane Proteins/genetics , Particle Size
6.
J Neural Transm (Vienna) ; 125(1): 67-75, 2018 01.
Article in English | MEDLINE | ID: mdl-28058503

ABSTRACT

To understand the mechanisms underlying higher brain functions, we need to analyze the roles of specific neuronal pathways or cell types forming the complex neural networks. In the neuroscience field, the transgenic approach has provided a useful gene engineering tool for experimental studies of neural functions. The conventional transgenic technique requires the appropriate promoter regions that drive a neuronal type-specific gene expression, but the promoter sequences specifically functioning in each neuronal type are limited. Previously, we developed novel types of lentiviral vectors showing high efficiency of retrograde gene transfer in the central nervous system, termed highly efficient retrograde gene transfer (HiRet) vector and neuron-specific retrograde gene transfer (NeuRet) vector. The HiRet and NeuRet vectors enable genetical manipulation of specific neural pathways in diverse model animals in combination with conditional cell targeting, synaptic transmission silencing, and gene expression systems. These newly developed vectors provide powerful experimental strategies to investigate, more precisely, the machineries exerting various neural functions. In this review, we give an outline of the HiRet and NeuRet vectors and describe recent representative applications of these viral vectors for studies on neural circuits.


Subject(s)
Brain/metabolism , Gene Transfer Techniques , Genetic Vectors/metabolism , Lentivirus/metabolism , Nerve Net/metabolism , Animals , Brain Chemistry/physiology , Genetic Vectors/analysis , Genetic Vectors/genetics , Humans , Lentivirus/chemistry , Lentivirus/genetics , Nerve Net/chemistry
7.
Virology ; 514: 1-8, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29128752

ABSTRACT

The HIV-1 RNA genome contains complex structures with many structural elements playing regulatory roles during viral replication. A recent study has identified multiple RNA structures with unknown functions that are conserved among HIV-1 and two simian immunodeficiency viruses. To explore the roles of these conserved RNA structures, we introduced synonymous mutations into the HIV-1 genome to disrupt each structure. These mutants exhibited similar particle production, viral infectivity, and replication kinetics relative to the parent NL4-3 virus. However, when replicating in direct competition with the wild-type NL4-3 virus, mutations of RNA structures at inter-protein domain junctions can cause fitness defects. These findings reveal the ability of HIV-1 to tolerate changes in its sequences, even in apparently highly conserved structures, which permits high genetic diversity in HIV-1 population. Our results also suggest that some conserved RNA structures may function to fine-tune viral replication.


Subject(s)
HIV-1/genetics , Lentivirus/genetics , RNA, Viral/chemistry , Base Sequence , Conserved Sequence , HIV Infections/virology , HIV-1/chemistry , HIV-1/physiology , Humans , Inverted Repeat Sequences , Lentivirus/chemistry , Lentivirus/classification , Lentivirus/physiology , Lentivirus Infections/virology , Nucleic Acid Conformation , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication
8.
mBio ; 8(6)2017 12 12.
Article in English | MEDLINE | ID: mdl-29233896

ABSTRACT

We report a simple strategy for the creation of lentiviral vectors specific to any desired target cells. SpyTag is inserted into an engineered Sindbis virus envelope protein and displayed on the lentivirus surface to create Sindbis virus-SpyTag pseudoparticles (Sind-SpyTag-pp). The SpyTag serves as the covalent anchoring site for a target-cell-specific cell-binding protein (CBP) that is fused to a truncated SpyCatcher (SpyCatcherΔ). Target-cell-specific lentiviruses are created by mixing the Sind-SpyTag-pp and CBP-SpyCatcherΔ in vitro We first used a HER2-binding designed ankyrin repeat protein (DARPin.9.26) as the model CBP. The DARPin-conjugated lentivirus transduced HER2+ SKOV3 cells with an infectious titer of 5.2 × 106 IU/ml, >500-fold higher than the unfunctionalized "naked" virions (<104 IU/ml). The ability of the DARPin-conjugated lentivirus to transduce HER2+ cells correlated with the surface expression level of HER2. Furthermore, these lentiviruses preferentially transduced HER2+ cells in cocultures containing HER2+ and HER2- cells. To enable the use of commercially available monoclonal antibodies (MAbs) as the CBP, we developed a convenient click chemistry-based approach to conjugate MAb-derived Fab fragments to a variant SpyCatcherΔ protein containing a nonnatural amino acid, 4-azido-l-phenylalanine (AzF). Using the HER2-binding trastuzumab as a model cell-specific MAb, we created Fab-conjugated lentiviral vectors that transduced HER2+ SKOV3 cells with an infectious titer of 2.8 × 106 IU/ml, on par with the result achieved using the DARPin-SpyCatcherΔ fusion protein. The ability to create cell-specific lentiviral vectors through chemical conjugation of a CBP should make this approach generalizable to any antibody, giving it broad utility for a wide range of research and clinical applications.IMPORTANCE Lentiviral vectors hold great potential in gene therapy. However, it remains a major hurdle to robustly engineer cell-specific lentiviral vectors. This article reports a simple and effective strategy to functionalize lentiviral vectors with cell-binding proteins, thus retargeting these viruses to cells expressing the binding partner of the CBP. The CBP is genetically or chemically linked to the SpyCatcher. The SpyTag is displayed on the virion surface as a fusion to an engineered Sindbis virus envelope protein and is used as the anchorage site for SpyCatcher-linked CBP. Using this strategy, we created lentiviral vectors highly infectious toward HER2+ cancer cells. The ability to rapidly create cell-specific lentiviral vectors targeting a wide range of cell types should accelerate the development of custom lentiviral vectors for many research and clinical applications.


Subject(s)
Genetic Vectors , Lentivirus/chemistry , Lentivirus/genetics , Transduction, Genetic/methods , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Genes, erbB-2/genetics , Genes, erbB-2/immunology , Genetic Therapy , Humans , Lentivirus/metabolism , Sindbis Virus/genetics , Trastuzumab/immunology , Trastuzumab/metabolism , Viral Envelope Proteins/genetics
9.
Virology ; 488: 162-8, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26650691

ABSTRACT

While the envelope glycoprotein of vesicular stomatitis virus (VSV-G) is widely used for pseudotyping of lentiviral vectors, sub-optimal gene transfer into certain cell types and its sensitivity to inactivation by human complement hinders its broader applications. To find alternative candidates, here we evaluated two serologically distinct novel viral envelopes derived from Chandipura (CNV-G) and Piry (PRV-G) vesiculoviruses. Both permitted generation of high titer psuedotyped lentiviral vectors with a capacity for high efficiency gene transfer into various cell types from different species. In human lymphoid and hematopoietic stem cells, their transduction efficiency was significantly lower than that of VSV-G. However, both novel envelopes were found to be more resistant to inactivation by human serum complement compared to VSV-G. Thus CNV-G and PRV-G envelopes can be harnessed for multiple uses in the future based on the cell type that needs to be gene transduced and possibly for in vivo gene transfer.


Subject(s)
Cell Surface Display Techniques/methods , Gene Transfer Techniques , Lentivirus/chemistry , Transduction, Genetic , Vesiculovirus/chemistry , Viral Envelope Proteins/analysis , Cells, Cultured , Complement System Proteins , Humans , Lentivirus/genetics , Lentivirus/immunology , Stem Cells/virology , Vesiculovirus/genetics , Vesiculovirus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virus Inactivation
10.
Gene Ther ; 22(3): 280-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25608718

ABSTRACT

Lentiviral (LV) vectors have emerged as powerful tools for basic research and clinical applications because of their ability to stably transduce both dividing and nondividing cells. A wide range of viral envelope (Env) glycoproteins have the ability to associate with the membrane of LV vectors, a process that is referred to as pseudotyping. Pseudotyped vectors have the capacity to transduce specific cell types for specific applications. For example, LV vectors pseudotyped with the measles virus (MV)-derived hemagglutinin (H) and fusion (F) proteins have the ability to transduce quiescent lymphocytes. In addition, the MV H glycoprotein can be engineered allowing cell-specific targeting of LV vectors. One problem with MV glycoprotein-pseudotyped LV vectors is low titer during vector production. This results in the need to manufacture large volumes of the vectors and to concentrate them to appropriate titers. The commonly used centrifugation-based concentration techniques for LV vectors are not practical for large-scale vector manufacturing. Thus, there is a need for improved methods to concentrate LV vectors. In this study, we adapted an anion-exchange membrane chromatography method that we previously used in the context of LV vectors pseudotyped with the vesicular stomatitis virus glycoprotein to concentate MV glycoprotein-pseudotyped LV vectors. Up to 60% of the input vectors with an up to 5300-fold reduction in volume was achieved using this anion-exchange chromatography method in conjunction with a desalting/concentration step involving centrifugal filter units. This technique provides a rapid and scalable approach for concentrating MV-pseudotyped LV vectors that does not require an elaborate setup.


Subject(s)
Chromatography, Ion Exchange/methods , Genetic Vectors/chemistry , Lentivirus/chemistry , Measles virus/metabolism , Cell Line , Glycoproteins/metabolism , Humans
11.
Prog Mol Biol Transl Sci ; 129: 327-51, 2015.
Article in English | MEDLINE | ID: mdl-25595809

ABSTRACT

Infection is best described as a stochastic process. Whether a host becomes infected upon exposure has a strong random element. The same applies to cells exposed to virions. In this review, we show how the mathematical formalism for stochastic processes has been used to describe and understand the infection by the Human and Simian Immunodeficiency Virus on different levels. We survey quantitative studies on the establishment of infection in the host (the organismal level) and on the infection of target cells (the cellular and molecular level). We then discuss how a synthesis of the approaches across these levels could give rise to a predictive framework for assessing the efficacy of microbicides and vaccines.


Subject(s)
Host-Pathogen Interactions , Lentivirus Infections/virology , Lentivirus/chemistry , Lentivirus/pathogenicity , Primates/virology , Protein Subunits/metabolism , Animals , Humans , Virus Internalization
12.
Biomed Res Int ; 2014: 473823, 2014.
Article in English | MEDLINE | ID: mdl-25580433

ABSTRACT

This study aimed to develop optimal gelatin-based mucoadhesive nanocomposites as scaffolds for intravesical gene delivery to the urothelium. Hydrogels were prepared by chemically crosslinking gelatin A or B with glutaraldehyde. Physicochemical and delivery properties including hydration ratio, viscosity, size, yield, thermosensitivity, and enzymatic degradation were studied, and scanning electron microscopy (SEM) was carried out. The optimal hydrogels (H), composed of 15% gelatin A175, displayed an 81.5% yield rate, 87.1% hydration ratio, 42.9 Pa·s viscosity, and 125.8 nm particle size. The crosslinking density of the hydrogels was determined by performing pronase degradation and ninhydrin assays. In vitro lentivirus (LV) release studies involving p24 capsid protein analysis in 293T cells revealed that hydrogels containing lentivirus (H-LV) had a higher cumulative release than that observed for LV alone (3.7-, 2.3-, and 2.3-fold at days 1, 3, and 5, resp.). Lentivirus from lentivector constructed green fluorescent protein (GFP) was then entrapped in hydrogels (H-LV-GFP). H-LV-GFP showed enhanced gene delivery in AY-27 cells in vitro and to rat urothelium by intravesical instillation in vivo. Cystometrogram showed mucoadhesive H-LV reduced peak micturition and threshold pressure and increased bladder compliance. In this study, we successfully developed first optimal gelatin-based mucoadhesive nanocomposites as intravesical gene delivery scaffolds.


Subject(s)
Gelatin/administration & dosage , Gene Transfer Techniques , Nanocomposites/administration & dosage , Animals , Gelatin/chemistry , Glutaral/administration & dosage , Glutaral/chemistry , Humans , Hydrogels/administration & dosage , Hydrogels/chemistry , Lentivirus/chemistry , Lentivirus/genetics , Nanocomposites/chemistry , Rats , Urothelium/growth & development , Urothelium/physiopathology
13.
J Control Release ; 170(3): 421-9, 2013 Sep 28.
Article in English | MEDLINE | ID: mdl-23791981

ABSTRACT

Gene delivering biomaterials have increasingly been employed to modulate the cellular microenvironment to promote tissue regeneration, yet low transduction efficiency has been a persistent challenge for in vivo applications. In this report, we investigated the surface modification of poly(lactide-co-glycolide) (PLG) scaffolds with polysaccharides, which have been implicated in binding lentivirus but have not been used for delivery. Chitosan was directly conjugated onto PLG scaffolds, whereas heparin and hyaluronan were indirectly conjugated onto PLG scaffolds with multi-amine crosslinkers. The addition of chitosan and heparin onto PLG promoted the association of lentivirus to these scaffolds and enhanced their transduction efficiency in vitro relative to hyaluronan-conjugated and control scaffolds that had limited lentivirus association and transduction. Transduction efficiency in vitro was increased partly due to an enhanced retention of virus on the scaffold as well as an extended half-life of viral activity. Transduction efficiency was also evaluated in vivo using porous, multiple channel PLG bridges that delivered lentivirus to the injured mouse spinal cord. Transgene expression persisted for weeks after implantation, and was able to enhance axon growth and myelination. These studies support gene-delivering PLG scaffolds for in vivo regenerative medicine applications.


Subject(s)
Gene Transfer Techniques , Hyaluronic Acid/chemistry , Lentivirus/genetics , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Animals , Chitosan/chemistry , Female , Genetic Therapy , HEK293 Cells , Heparin/chemistry , Humans , Lactic Acid/chemistry , Lentivirus/chemistry , Luciferases/genetics , Mice , Mice, Inbred C57BL , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
14.
Virus Res ; 175(1): 1-11, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583684

ABSTRACT

Gene therapy utilizing lentiviral vectors (LVs) constitutes a real therapeutic alternative for many inherited monogenic diseases. Therefore, the generation of functional vectors using fast, non-laborious and cost-effective strategies is imperative. Among the available concentration methods for VSV-G pseudotyped lentiviruses to achieve high therapeutic titers, ultracentrifugation represents the most common approach. However, the procedure requires special handling and access to special instrumentation, it is time-consuming, and most importantly, it is cost-ineffective due to the high maintenance expenses and consumables of the ultracentrifuge apparatus. Here we describe an improved protocol in which vector stocks are prepared by transient transfection using standard cell culture media and are then concentrated by ultrafiltration, resulting in functional vector titers of up to 6×10(9) transducing units per millilitre (TU/ml) without the involvement of any purification step. Although ultrafiltration per se for concentrating viruses is not a new procedure, our work displays one major novelty; we characterized the nature and the constituents of the viral batches produced by ultrafiltration using peptide mass fingerprint analysis. We also determined the viral functional titer by employing flow cytometry and evaluated the actual viral particle size and concentration in real time by using laser-based nanoparticle tracking analysis based on Brownian motion. Vectors generated by this production method are contained in intact virions and when tested to transduce in vitro either murine total bone marrow or human CD34(+) hematopoietic stem cells, resulted in equal transduction efficiency and reduced toxicity, compared to lentiviral vectors produced using standard ultracentrifugation-based methods. The data from this study can eventually lead to the improvement of protocols and technical modifications for the clinical trials for gene therapy.


Subject(s)
Genetic Vectors/isolation & purification , Lentivirus/isolation & purification , Ultrafiltration/methods , Virology/methods , Animals , Genetic Therapy/methods , Genetic Vectors/chemistry , Hematopoietic Stem Cells/virology , Humans , Lentivirus/chemistry , Mice , Mice, Inbred C57BL , Peptide Mapping , Transduction, Genetic , Ultracentrifugation/methods
15.
ACS Nano ; 7(5): 3896-904, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23560365

ABSTRACT

Utilization of quantum dots (QDs) for single virus tracking has attracted growing interest. Through modification of viral surface proteins, viruses can be labeled with various functionalized QDs and used for tracking the routes of viral infections. However, incorporation of QDs on the viral surface may affect the efficiency of viral entry and alter virus-cell interactions. Here, we describe that QDs can be encapsulated into the capsid of vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped lentivirus (PTLV) in living cells without modification of the viral surface. QDs conjugated with modified genomic RNAs (gRNAs), which contain a packaging signal (Psi) sequence for viral genome encapsulation, can be packaged into virions together with the gRNAs. QD-containing PTLV demonstrated similar entry efficiency as the wild-type PTLV. After infection, QD signals entered the Rab5+ endosome and then moved to the microtubule organizing center of the infected cells in a microtubule-dependent manner. Findings in this study are consistent with previously reported infection routes of VSV and VSV-G pseudotyped lentivirus, indicating that our established QD packaging approach can be used for enveloped virus labeling and tracking.


Subject(s)
Lentivirus/chemistry , Lentivirus/physiology , Membrane Glycoproteins/chemistry , Quantum Dots , Staining and Labeling/methods , Viral Envelope Proteins/chemistry , Capsid/chemistry , Capsules , Cell Survival , HeLa Cells , Humans
17.
Methods Mol Biol ; 737: 183-209, 2011.
Article in English | MEDLINE | ID: mdl-21590398

ABSTRACT

Lentiviral vectors have evolved over the last decade as powerful, reliable, and safe tools for stable gene transfer in a wide variety of mammalian cells. Contrary to other vectors derived from oncoretroviruses, they allow for stable gene delivery into most nondividing primary cells. In particular, lentivectors (LVs) derived from HIV-1 have gradually evolved to display many desirable features aimed at increasing both their safety and their versatility. This is why lentiviral vectors are becoming the most useful and promising tools for genetic engineering, to generate cells that can be used for research, diagnosis, and therapy. This chapter describes protocols and guidelines, for production and titration of LVs, which can be implemented in a research laboratory setting, with an emphasis on standardization in order to improve transposability of results between laboratories. We also discuss latest designs in LV technology.


Subject(s)
Cell Culture Techniques , Genetic Therapy/methods , Genetic Vectors/isolation & purification , Lentivirus/growth & development , Virion/growth & development , Animals , Base Sequence , Cell Culture Techniques/standards , Cell Line , Centrifugation/methods , DNA Primers/chemistry , Flow Cytometry/methods , Genes, Viral , Genetic Engineering , Humans , Lentivirus/chemistry , Lentivirus/isolation & purification , Mice , Polymerase Chain Reaction/methods , Safety , Titrimetry/methods , Transfection/methods , Virion/chemistry , Virion/isolation & purification
18.
Cell Host Microbe ; 8(3): 248-59, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20833376

ABSTRACT

Lentiviruses are widespread in a variety of vertebrates, often associated with chronic disease states. However, until the recent discovery of the prehistoric endogenous lentiviruses in rabbits (RELIK) and lemurs (PSIV), it was thought that lentiviruses had no capacity for germline integration and were only spread horizontally in an exogenous fashion. The existence of RELIK and PSIV refuted these ideas, revealing lentiviruses to be present in a range of mammals, capable of germline integration, and far more ancient than previously thought. Using Gag sequences reconstructed from the remnants of these prehistoric lentiviruses, we have produced chimeric lentiviruses capable of infecting nondividing cells and determined structures of capsid domains from PSIV and RELIK. We show that the structures from these diverse viruses are highly similar, containing features found in modern-day lentiviruses, including a functional cyclophilin-binding loop. Together, these data provide evidence for an ancient capsid-cyclophilin interaction preserved throughout lentiviral evolution.


Subject(s)
Capsid Proteins/chemistry , Cyclophilin A/metabolism , Endogenous Retroviruses/chemistry , Endogenous Retroviruses/genetics , Evolution, Molecular , Lentivirus/chemistry , Lentivirus/genetics , Animals , Base Sequence , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Crystallography, X-Ray , Cyclophilin A/chemistry , DNA Methylation , Endogenous Retroviruses/physiology , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Genes, Viral , Genes, gag , Lemur/virology , Lentivirus/physiology , Lentiviruses, Primate/chemistry , Lentiviruses, Primate/genetics , Lentiviruses, Primate/physiology , Models, Molecular , Protein Structure, Tertiary , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Virion/metabolism
19.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(22): 1939-45, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20599176

ABSTRACT

Desthiobiotin-tagged lentiviral vectors have been metabolically produced by DBL producer cells in a 7,8-diaminopelargonic acid (7-DAPA) dependent manner for envelope independent, single-step affinity purification. 7-DAPA, which has little or no affinity for avidin/streptavidin, was synthesised and verified by NMR spectroscopy and mass spectrometry. By expressing the biotin acceptor, biotin ligase and desthiobiotin synthase bioD, DBL cells converted exogenous 7-DAPA into membrane-bound desthiobiotin. Desthiobiotin on the DBL cell surface was visualised by confocal microscopy and the desthiobiotin density was quantified by HABA-avidin assay. Desthiobiotin was then spontaneously incorporated onto the surface of lentiviral vectors produced by the DBL cells. It has been demonstrated by flow cytometry that the desthiobiotinylated lentiviruses were captured from the crude 7-DAPA-containing viral supernatant by Streptavidin Magnespheres and eluted by biotin solution efficiently whilst retaining infectivity. The practical, high yielding virus purification using Pierce monomeric avidin coated columns indicates a highly efficient biotin-dependent recovery of infectious lentiviruses at 68%. The recovered lentiviral vectors had a high purity and the majority were eluted within 45 min. This 7-DAPA mediated desthiobiotinylation technology can be applied in scalable production of viral vectors for clinical gene therapy.


Subject(s)
Amino Acids, Diamino/metabolism , Biotin/analogs & derivatives , Chromatography, Affinity/methods , Genetic Vectors/chemistry , Lentivirus/chemistry , Biotin/metabolism , Cell Line , Genetic Vectors/metabolism , Humans , Lentivirus/metabolism , Protein Binding , Streptavidin/chemistry
20.
J Virol ; 84(17): 8561-70, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20592083

ABSTRACT

Primate lentiviruses are unique in that they produce several accessory proteins to help in the establishment of productive viral infection. The major function of these proteins is to clear host resistance factors that inhibit viral replication. Vif is one of these proteins. It functions as an adaptor that binds to the cytidine deaminases APOBEC3G (A3G) and APOBEC3F (A3F) and bridges them to a cullin 5 (Cul5) and elongin (Elo) B/C E3 ubiquitin ligase complex for proteasomal degradation. So far, 11 discontinuous domains in Vif have been identified that regulate this degradation process. Here we report another domain, T(Q/D/E)x(5)ADx(2)(I/L), which is located at residues 96 to 107 in the human immunodeficiency virus type 1 (HIV-1) Vif protein. This domain is conserved not only in all HIV-1 subtypes but also in other primate lentiviruses, including HIV-2 and simian immunodeficiency virus (SIV), which infects rhesus macaques (SIVmac) and African green monkeys (SIVagm). Mutations of the critical residues in this motif seriously disrupted Vif's neutralizing activity toward both A3G and A3F. This motif regulates Vif interaction not only with A3G and A3F but also with Cul5. When this motif was inactivated in the HIV-1 genome, Vif failed to exclude A3G and A3F from virions, resulting in abortive HIV replication in nonpermissive human T cells. Thus, T(Q/D/E)x(5)ADx(2)(I/L) is a critical functional motif that directly supports the adaptor function of Vif and is an attractive target for inhibition of Vif function.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , HIV Infections/enzymology , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Cytidine Deaminase/genetics , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Evolution, Molecular , HIV Infections/genetics , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Lentivirus/chemistry , Lentivirus/enzymology , Lentivirus/genetics , Lentivirus Infections/virology , Molecular Sequence Data , Protein Binding , Sequence Alignment , vif Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...