Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Retrovirology ; 9: 83, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23035841

ABSTRACT

BACKGROUND: The Moloney murine leukaemia virus (Mo-MLV) gag gene encodes three main structural proteins, matrix, capsid and nucleocapsid and a protein called p12. In addition to its role during the late stages of infection, p12 has an essential, but undefined, function during early post-entry events. As these stages of retroviral infection remain poorly understood, we set out to investigate the function of p12. RESULTS: Examination of the infectivity of Mo-MLV virus-like particles containing a mixture of wild type and mutant p12 revealed that the N- and C-terminal regions of p12 are sequentially acting domains, both required for p12 function, and that the N-terminal activity precedes the C-terminal activity in the viral life cycle. By creating a panel of p12 mutants in other gammaretroviruses, we showed that these domains are conserved in this retroviral genus. We also undertook a detailed mutational analysis of each domain, identifying residues essential for function. These data show that different regions of the N-terminal domain are necessary for infectivity in different gammaretroviruses, in stark contrast to the C-terminal domain where the same region is essential for all viruses. Moreover, chimeras between the p12 proteins of Mo-MLV and gibbon ape leukaemia virus revealed that the C-terminal domains are interchangeable whereas the N-terminal domains are not. Finally, we identified potential functions for each domain. We observed that particles with defects in the N-terminus of p12 were unable to abrogate restriction factors, implying that their cores were impaired. We further showed that defects in the C-terminal domain of p12 could be overcome by introducing a chromatin binding motif into the protein. CONCLUSIONS: Based on these data, we propose a model for p12 function where the N-terminus of p12 interacts with, and stabilizes, the viral core, allowing the C-terminus of p12 to tether the preintegration complex to host chromatin during mitosis, facilitating integration.


Subject(s)
Gene Products, gag/genetics , Gene Products, gag/metabolism , Moloney murine leukemia virus/physiology , Virus Replication , DNA Mutational Analysis , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/physiology , Moloney murine leukemia virus/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism
2.
Oncol Rep ; 28(1): 21-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22552490

ABSTRACT

Gene therapy mediated by murine leukemia virus (MLV)-based replicating retrovirus vector (RRV) was previously proven to be highly effective in tumor cell killing, resulting in significant suppression of tumor growth in vivo. Recently, we developed a different form of RRV which is derived from another retrovirus, gibbon ape leukemia virus (GALV), as a cancer therapeutic agent. We compared the gene delivery efficiency and antitumor effects in the two types of RRV in experimental hepatocellular carcinoma (HCC). Our results show that both RRVs can efficiently spread throughout entire HCC cell populations in vitro and achieve high transduction efficiency in HCC xenografts in vivo, while GALV RRV, in general, exhibited more rapid replication kinetics in the tumors. In vitro, substantial HCC cell killing was achieved even when initially only 1% of the HCC cells were producing RRVs that express the yeast cytosine deaminase suicide gene, indicating that the high efficiency of gene transfer by replicative spread of RRVs greatly increased suicide gene toxicity. In vivo, GALV RRV-mediated suicide gene therapy efficiently suppressed HCC tumor growth and no detectable RRV signals were observed in extratumoral tissues, showing promise in using GALV RRV as a cancer therapeutic agent.


Subject(s)
Carcinoma, Hepatocellular/therapy , Leukemia Virus, Gibbon Ape/genetics , Liver Neoplasms, Experimental/therapy , Oncolytic Viruses/genetics , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biotransformation , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cytosine Deaminase/biosynthesis , Cytosine Deaminase/genetics , Flucytosine/metabolism , Flucytosine/pharmacology , Flucytosine/therapeutic use , Fungal Proteins/biosynthesis , Fungal Proteins/genetics , Genetic Therapy , Hep G2 Cells , Humans , Leukemia Virus, Gibbon Ape/enzymology , Leukemia Virus, Gibbon Ape/physiology , Liver Neoplasms, Experimental/pathology , Mice , Mice, Nude , Oncolytic Virotherapy , Oncolytic Viruses/enzymology , Oncolytic Viruses/physiology , Prodrugs/metabolism , Prodrugs/pharmacology , Prodrugs/therapeutic use , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transduction, Genetic , Tumor Burden/drug effects , Virus Replication , Xenograft Model Antitumor Assays
3.
Retrovirology ; 8: 53, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21729311

ABSTRACT

BACKGROUND: Over the last several decades it has been noted, using a variety of different methods, that cells infected by a specific gammaretrovirus are resistant to infection by other retroviruses that employ the same receptor; a phenomenon termed receptor interference. Receptor masking is thought to provide an earlier means of blocking superinfection, whereas receptor down regulation is generally considered to occur in chronically infected cells. RESULTS: We used replication-competent GFP-expressing viruses containing either an amphotropic murine leukemia virus (A-MLV) or the gibbon ape leukemia virus (GALV) envelope. We also constructed similar viruses containing fluorescence-labeled Gag proteins for the detection of viral particles. Using this repertoire of reagents together with a wide range of antibodies, we were able to determine the presence and availability of viral receptors, and detect viral envelope proteins and particles presence on the cell surface of chronically infected cells. CONCLUSIONS: A-MLV or GALV receptors remain on the surface of chronically infected cells and are detectable by respective antibodies, indicating that these receptors are not downregulated in these infected cells as previously proposed. We were also able to detect viral envelope proteins on the infected cell surface and infected cells are unable to bind soluble A-MLV or GALV envelopes indicating that receptor binding sites are masked by endogenously expressed A-MLV or GALV viral envelope. However, receptor masking does not completely prevent A-MLV or GALV superinfection.


Subject(s)
Host-Pathogen Interactions , Leukemia Virus, Gibbon Ape/physiology , Leukemia Virus, Murine/physiology , Receptors, Virus/biosynthesis , Animals , Cattle , Cell Line , Cricetinae , Cricetulus , Down-Regulation , Genes, Reporter , Humans , Leukemia Virus, Gibbon Ape/growth & development , Leukemia Virus, Murine/growth & development , Mice
4.
Cancer Gene Ther ; 17(4): 256-65, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19893593

ABSTRACT

Fusogenic membrane glycoproteins (FMGs) are viral envelope proteins, which bind surface receptors and induce fusion of the cell membrane. An FMG-transfected cell will fuse with neighbor cells, thus forming syncytia that die within 5 days. In this report, plasmids encoding for FMGs from Human Endogenous Retrovirus-W (HERV-W) was compared with Gibbon Ape Leukemia Virus (GALV) and feline endogenous virus RD-114 (RD). These plasmids were transfected in human non-small-cell lung cancer (NSCLC) cells in vitro or directly injected into tumors in mice. All FMGs induced the formation of syncytia containing around 50 cells. HERV-W or GALV FMGs decreased up to 80% of cell viability in vitro and inhibited tumor growth in vivo (60-70% reduction). In contrast, RD FMG was not efficient. Apoptosis played a role in the death of the syncytia, but addition of the caspase inhibitor Z-VAD-fmk had no effect, suggesting that apoptosis is not the only mechanism responsible for FMG-induced cell death. Altogether, our results demonstrate that even at very low transfection efficiency, the antitumor activity of HERV-W FMG is as effective as that of GALV in vitro and in vivo for the treatment of human lung tumors.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Giant Cells/metabolism , Lung Neoplasms/therapy , Oncolytic Virotherapy , Viral Fusion Proteins/metabolism , Animals , Bystander Effect , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/virology , Cats , Endogenous Retroviruses/physiology , Female , Genetic Vectors/therapeutic use , Giant Cells/virology , Humans , In Vitro Techniques , Leukemia Virus, Feline/physiology , Leukemia Virus, Gibbon Ape/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/virology , Mice , Plasmids/genetics , Promoter Regions, Genetic , Transfection , Tumor Cells, Cultured , Viral Fusion Proteins/genetics , Xenograft Model Antitumor Assays
5.
Hum Gene Ther ; 20(9): 966-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19453219

ABSTRACT

Retroviral vectors derived from the Moloney murine leukemia virus have been used in successful and promising gene therapy clinical trials. However, platforms for their large-scale production must be further developed. As a proof of principle, we reported the generation of a packaging cell line that produces amphotropic retroviral vectors in suspension and serum-free medium (SFM). In the present study, we have constructed and characterized two retroviral packaging cell lines designed for gene transfer in hematopoietic cells. These cell lines grow in suspension and SFM, and produce high-titer RD114- and gibbon ape leukemia virus (GALV)-pseudotyped vectors for a 3-month culture period. Viral particles released are as robust during repeated freeze-thaw cycles and on thermal inactivation at 37 degrees C as their counterparts produced in cells cultured adherently with serum. We also show that RD114- and GALV-pseudotyped vectors produced in suspension and SFM efficiently transduce human lymphocytes and hematopoietic stem cells. As these retroviral packaging cell lines distinctively maintain high vector titers while growing in suspension and SFM, we conclude that these cell lines are uniquely suitable for large-scale clinical-grade vector production for late-phase clinical trials involving gene transfer into hematopoietic cells.


Subject(s)
Genetic Vectors/physiology , Hematopoietic Stem Cells/virology , Leukemia Virus, Gibbon Ape/physiology , Retroviridae/physiology , Transduction, Genetic , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Line , Culture Media , Culture Media, Serum-Free , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Retroviridae/genetics , Retroviridae/metabolism , T-Lymphocytes/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Assembly
6.
J Immunother ; 32(3): 272-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242373

ABSTRACT

The Food and Drug Administration/Center for Biologics Evaluation and Research has defined that for retroviral gene therapy, the vector-producing cell, the vector preparation, and the ex vivo gene-transduced cells have to be tested for absence of replication-competent retrovirus (RCR) if the transduced cells are cultured for >4 days. We assessed the sensitivity of the "extended PG4(S+L-) assay" to detect gibbon ape leukemia virus (GALV) RCR, and applied this assay to measure GALV RCR spread in retrovirally transduced T cells. To this end, T cells were expanded for 12 days after transduction with a GALV-envelope pseudotyped retroviral vector expressing single chain variable fragment (anticarbonic anhydrase IX) in presence or absence of GALV RCR. Results showed that: (1) the "extended PG4(S+L-) assay" detects 1 focus-forming unit (ffu) GALV RCR and thus is applicable and sufficiently sensitive to screen human T-cell cultures for absence of infectious GALV RCR; (2) although GALV RCR infect human T cells, it very poorly replicate in T cells; (3) GALV RCR, when present at low levels immediately upon gene transduction (ie, 100 ffu/20x10 T cells in 100 mL), did not spread during a 12-day T-cell culture at clinical scale. Our observation that GALV RCR poorly spreads in primary human T-cell cultures questions the relevance of testing T-cell transductants for RCR on top of testing the vector-producing cells and the clinical vector batch for RCR and warrants evaluation of the current policy for safety testing of ex vivo retrovirally transduced T lymphocytes for GALV RCR.


Subject(s)
Genetic Vectors/physiology , Leukemia Virus, Gibbon Ape/physiology , T-Lymphocytes/virology , Virus Replication , Animals , Biological Assay , Genetic Vectors/genetics , Humans , Leukemia Virus, Gibbon Ape/genetics , Sensitivity and Specificity , Transduction, Genetic
7.
Gene Ther ; 15(17): 1240-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18509378

ABSTRACT

Fusogenic membrane glycoproteins (FMGs) may enhance the cytotoxicity of conditionally replicative adenoviruses. However, expression at early stages of infection impairs virus replication. We have inserted the hyperfusogenic form of the gibbon ape leukemia virus (GALV) envelope glycoprotein as a new splice unit of the major late promoter (MLP) to generate a replication-competent adenovirus expressing this protein. At high multiplicity of infection (MOI), this virus replicated efficiently forming clumps of fused cells and showing a faster release. In contrast, at low MOI, infected cells formed syncytia where only one nucleus contained virus DNA, decreasing total virus production but increasing cytotoxicity.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Giant Cells/physiology , Leukemia Virus, Gibbon Ape/physiology , Oncolytic Virotherapy/methods , Viral Fusion Proteins/genetics , Cell Line, Tumor , Gene Expression , Genetic Engineering , Humans , Leukemia Virus, Gibbon Ape/genetics , Promoter Regions, Genetic , Transgenes , Virus Replication
8.
Cancer Res ; 66(9): 4835-42, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651439

ABSTRACT

We have previously developed an oncolytic herpes simplex virus-1 based on a clinical virus isolate, which was deleted for ICP34.5 to provide tumor selected replication and ICP47 to increase antigen presentation as well as tumor selective virus replication. A phase I/II clinical trial using a version of this virus expressing granulocyte macrophage colony-stimulating factor has shown promising results. The work reported here aimed to develop a version of this virus in which local tumor control was further increased through the combined expression of a highly potent prodrug activating gene [yeast cytosine deaminase/uracil phospho-ribosyltransferase fusion (Fcy::Fur)] and the fusogenic glycoprotein from gibbon ape leukemia virus (GALV), which it was hoped would aid the spread of the activated prodrug through the tumor. Viruses expressing the two genes individually or in combination were constructed and tested, showing (a) GALV and/or Fcy::Fur expression did not affect virus growth; (b) GALV expression causes cell fusion and increases the tumor cell killing at least 30-fold in vitro and tumor shrinkage 5- to 10-fold in vivo; (c) additional expression of Fcy::Fur combined with 5-fluorocytosine administration improves tumor shrinkage further. These results indicate, therefore, that the combined expression of the GALV protein and Fcy::Fur provides a highly potent oncolytic virus with improved capabilities for local tumor control. It is intended to enter the GALV/Fcy::Fur expressing virus into clinical development for the treatment of tumor types, such as pancreatic or lung cancer, where local control would be anticipated to be clinically advantageous.


Subject(s)
Fibrosarcoma/therapy , Flucytosine/pharmacokinetics , Leukemia Virus, Gibbon Ape/genetics , Membrane Glycoproteins/genetics , Oncolytic Virotherapy/methods , Simplexvirus/physiology , Animals , Biotransformation , Cell Fusion , Combined Modality Therapy , Cytosine Deaminase/biosynthesis , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/virology , Flucytosine/pharmacology , Fluorouracil/pharmacokinetics , Fluorouracil/pharmacology , Humans , Leukemia Virus, Gibbon Ape/metabolism , Leukemia Virus, Gibbon Ape/physiology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Pentosyltransferases/biosynthesis , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Prodrugs/pharmacokinetics , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simplexvirus/genetics , Virus Replication , Xenograft Model Antitumor Assays
9.
J Virol ; 79(14): 9332-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15994829

ABSTRACT

Gammaretroviruses that enter cells via binding to a surface receptor use one of two fundamental mechanisms. In the first, binding of the virus particle to its cognate receptor is followed by fusion and internalization. The second, less common mechanism requires the addition of an accessory protein in order to achieve fusion and entry into the target cells; this protein is usually the soluble form of the envelope protein containing the receptor-binding domain (RBD). For some viruses, such as amphotropic murine leukemia virus (A-MLV), particles with fusion-defective envelope proteins can enter cells in the presence of their own RBD or that of another viral envelope, regardless of its cognate receptor, suggesting that these viruses share a common entry mechanism. A notable exception is gibbon ape leukemia virus (GALV). Fusion-impaired GALV envelope mutants can be trans-activated for infectivity only by GALV RBDs. Using dually functional GALV/A-MLV receptors, we examined the role of receptor with respect to which RBD could overcome fusion impaired virus entry.


Subject(s)
Leukemia Virus, Gibbon Ape/physiology , Receptors, Virus/physiology , Symporters/physiology , Animals , Membrane Fusion , Mice , Sodium-Phosphate Cotransporter Proteins
10.
Rev Clin Exp Hematol ; 7(4): 329-35, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15129646

ABSTRACT

A study of the growth of primate/human T cells led to mechanisms for temporary laboratory culture of these cells (discovery of interleukin-2) and also their continuous culture (by immortalization after infection with human T-cell lymphotropic virus type 1 or 2 (HTLV-1 or 2)). Cultures of lymphocytes also led us to isolate five persisting T-tropic viruses: 1. the Hall's Island strain of gibbon ape leukemia virus, 2. HTLV-1, 3. HTLV-2, 4. human immunodeficiency virus and 5. human herpes virus-6 (HHV-6). This report is a brief synopsis of the discoveries of the first human retroviruses, the HTLV.


Subject(s)
Medical Oncology/history , Retroviridae Infections/virology , Retroviridae/physiology , T-Lymphocytes/virology , Virology/history , Animals , Cell Transformation, Viral , Cells, Cultured/virology , Deltaretrovirus/isolation & purification , Deltaretrovirus/physiology , HIV/isolation & purification , HIV/physiology , Herpesvirus 6, Human/isolation & purification , Herpesvirus 6, Human/physiology , History, 20th Century , Humans , Leukemia/etiology , Leukemia/virology , Leukemia Virus, Gibbon Ape/isolation & purification , Leukemia Virus, Gibbon Ape/physiology , Lymphoma/etiology , Lymphoma/virology , Primates , Retroviridae Infections/history
11.
Acta Biochim Pol ; 49(2): 333-9, 2002.
Article in English | MEDLINE | ID: mdl-12362974

ABSTRACT

Recombinant retroviral vectors are still the most common gene delivery vehicles for gene therapy purposes, especially for construction of genetically modified tumor vaccines (GMTV). However, these vehicles are characterized by relatively low titre and in the case of many tumor cell lines, low transduction efficiency. We constructed bicistronic retroviral vector pseudotypes of amphotropic murine leukemia virus (A-MuLV) and gibbon ape leukemia virus (GaLV), encoding enhanced green fluorescent protein (EGFP) as a rapid and easily detectable reporter gene. Transduction of five different human melanoma and four renal carcinoma cell lines by these two virus pseudotypes revealed differences in transduction efficiency, which wase markedly lower for the renal carcinoma cell lines. Stimulation of retroviral receptor expression (PiT1 and PiT2) by phosphate depletion induced a limited increase of receptor mRNA levels, but did not improve the gene transfer efficiency. In contrast, simultaneous transduction with both vector pseudotypes markedly increased the transduction efficiency, compared to GaLV or A-MuLV alone. The same effect could be achieved by several repeated exposures of target cells to fresh vector preparation. Overexpression of GaLV receptor (PiT1) in target cells significantly increased the transduction rate and enabled retrovirus mediated gene transfer into the cells which normally are not transducible by GaLV pseudotypes. We demonstrated that, using different transduction strategies, the relatively inefficient, widely used retroviral vector systems could be significantly improved.


Subject(s)
Receptors, Virus/genetics , Receptors, Virus/metabolism , Retroviridae/genetics , Retroviridae/physiology , Transduction, Genetic/methods , Carcinoma, Renal Cell/genetics , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/physiology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Melanoma/genetics , Phosphates/pharmacology , Polymerase Chain Reaction , Tumor Cells, Cultured
12.
Hum Gene Ther ; 12(1): 61-70, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11177543

ABSTRACT

The potential pathogenicity of replication-competent retroviruses (RCR) requires vigilant testing to exclude inadvertent contamination of clinical gene therapy vector products with RCR. Pseudotyped vectors using the gibbon ape leukemia virus (GALV) envelope have entered into clinical trials but specific recommendations regarding methods for screening of vector product and analysis of clinical samples have not been set forth. Unfortunately, current screening assays used for detecting amphotropic RCR are not suitable for GALV-pseudotyped RCR. We modified the extended S+/L- assay for RCR detection by using human 293 cells for virus amplification. Of five cell lines tested, 293 cells were selected because they combined a high transduction efficiency and an ability to generate RCR at high titer. After optimizing the amplification assay, a dilution of GALV virus could consistently be detected at a dilution of 10(-6). In coculture experiments, one GALV-infected cell could be consistently detected in 10(6) uninfected cells. A PCR-based assay was developed that was capable of detecting 100 copies of a GALV envelope containing plasmid diluted in 1 microg of DNA obtained from uninfected cells. PCR was also able to detect one GALV-infected cell in 10(6) uninfected cells. These assays will be suitable for testing of vector preparations and for monitoring of clinical samples from patients treated in clinical gene therapy protocols. The assays developed are similar in methodology and sensitivity to those currently used for certification of amphotropic retroviral vectors.


Subject(s)
Leukemia Virus, Gibbon Ape/physiology , Retroviridae/physiology , Virus Replication , Biological Assay , Cell Transformation, Viral , DNA Primers/chemistry , DNA, Viral/analysis , Genetic Vectors , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Polymerase Chain Reaction
13.
J Virol ; 74(20): 9797-801, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000257

ABSTRACT

Chinese hamster ovary (CHO) cells are resistant to infections by gibbon ape leukemia virus (GALV) and amphotropic murine leukemia virus (A-MLV) unless they are pretreated with tunicamycin, an inhibitor of N-linked glycosylation. These viruses use the related sodium-phosphate symporters Pit1 and Pit2, respectively, as receptors in nonhamster cells, and evidence has suggested that the corresponding transporters of CHO cells may be masked by tunicamycin-sensitive secreted inhibitors. Although the E36 line of Chinese hamster cells was reported to secrete the putative Pit2 inhibitor and to be sensitive to the inhibitory CHO factors, E36 cells are highly susceptible to both GALV and A-MLV in the absence of tunicamycin. Moreover, expression of E36 Pit2 in CHO cells conferred tunicamycin-independent susceptibilities to both viruses. Based on the latter results, it was suggested that E36 Pit2 must functionally differ from the endogenous Pit2 of CHO cells. To test these ideas, we analyzed the receptor properties of CHO Pit1 and Pit2 in CHO cells. Surprisingly, and counterintuitively, transfection of a CHO Pit2 expression vector into CHO cells conferred strong susceptibility to both GALV and A-MLV, and similar overexpression of CHO Pit1 conferred susceptibility to GALV. Thus, CHO Pit2 is a promiscuous functional receptor for both viruses, and CHO Pit1 is a functional receptor for GALV. Similarly, we found that the natural resistance of Mus dunni tail fibroblasts to subgroup C feline leukemia viruses (FeLV-C) was eliminated simply by overexpression of the endogenous FeLV-C receptor homologue. These results demonstrate a novel and simple method to unmask latent retroviral receptor activities that occur in some cells. Specifically, resistances to retroviruses that are caused by subthreshold levels of receptor expression or by stoichiometrically limited masking or interference mechanisms can be efficiently overcome simply by overexpressing the endogenous receptors in the same cells.


Subject(s)
Leukemia Virus, Feline/physiology , Leukemia Virus, Gibbon Ape/physiology , Receptors, Virus/analysis , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Mice , Molecular Sequence Data , Receptors, Virus/physiology , Tunicamycin/pharmacology
14.
J Virol ; 73(4): 2916-20, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074140

ABSTRACT

The Chinese hamster cell lines E36 and CHOK1 dramatically differ in susceptibility to amphotropic murine leukemia virus (A-MuLV) and gibbon ape leukemia virus (GALV); E36 cells are highly susceptible to both viruses, CHOK1 cells are not. We have previously shown that GALV can infect E36 cells by using both its own receptor, HaPit1, and the A-MuLV receptor, HaPit2. Given that the two cell lines are from the same species, the loss of function of both of these receptors in CHOK1 cells is surprising. Other studies have shown that CHOK1 cells secrete proteins that block A-MuLV entry into CHOK1 as well as E36, suggesting the two A-MuLV receptors are functionally identical. However, CHOK1 conditioned medium does not block GALV entry into E36, indicating the secreted inhibitors do not block HaPit1. HaPit1 and ChoPit1 therefore differ as receptors for GALV; ChoPit1 is either inactivated by secreted factors or intrinsically nonfunctional. To determine why GALV cannot infect CHOK1, we cloned and sequenced ChoPit1 and ChoPit2. ChoPit2 is almost identical to HaPit2, which explains why CHOK1 conditioned medium blocks A-MuLV entry via both receptors. Although ChoPit1 and HaPit1 are 91% identical, a notable difference is at position 550 in the fourth extracellular region, shown by several studies to be crucial for GALV infection. Pit1 and HaPit1 have aspartate at 550, whereas ChoPit1 has threonine at this position. We assessed the significance of this difference for GALV infection by replacing the aspartate 550 in Pit1 with threonine. This single substitution rendered Pit1 nonfunctional for GALV and suggests that threonine at 550 inactivates ChoPit1 as a GALV receptor. Whether native ChoPit1 functions for GALV was determined by interference assays using Lec8, a glycosylation-deficient derivative of CHOK1 that is susceptible to both viruses and that has the same receptors as CHOK1. Unlike with E36, GALV and A-MuLV exhibited reciprocal interference when infecting Lec8, suggesting that they use the same receptor. We conclude both viruses can use ChoPit2 in the absence of the inhibitors secreted by CHOK1 and ChoPit1 is nonfunctional.


Subject(s)
Leukemia Virus, Gibbon Ape/physiology , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cricetinae , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Virus/genetics , Sequence Alignment , Signal Transduction , Virus Replication
15.
J Virol ; 71(10): 7619-22, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9311843

ABSTRACT

Pit1, the receptor for gibbon ape leukemia virus (GALV), is proposed to be an integral membrane protein with five extracellular loops. Chimeras made between Pit1 homologs differing in permissivity for infection and between Pit1 and the related protein Pit2 have shown that the fourth extracellular loop plays a critical role in infection. However, further elucidation of the roles of the extracellular loops in infection is hampered by the high level of sequence similarity among these proteins. The sodium-dependent phosphate transporter, Pho-4, from the filamentous fungus Neurospora crassa is distantly related to Pit1 and -2, showing an amino acid identity of only 35% to Pit1 in the putative extracellular loops. We show here that Pho-4 itself does not function as a receptor for GALV. Introduction of 12 Pit1-specific amino acid residues in the putative fourth extracellular loop of Pho-4 resulted in a functional GALV receptor. Therefore, the presence of a Pit1 loop 4-specific sequence is sufficient to confer receptor function for the mammalian retrovirus GALV on the fungal phosphate transporter Pho-4.


Subject(s)
Leukemia Virus, Gibbon Ape/physiology , Membrane Transport Proteins/metabolism , Phosphate Transport Proteins , Receptors, Virus/physiology , 3T3 Cells , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Hylobates , Membrane Transport Proteins/chemistry , Mice , Molecular Sequence Data , Neurospora crassa/metabolism , Protein Biosynthesis , Receptors, Virus/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription, Genetic , Transfection
16.
Blood ; 90(3): 952-7, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9242523

ABSTRACT

It is essential for the study of T-cell function and the improvement of adoptive cell therapies to efficiently generate large populations of human primary T cells that reliably express foreign genes. This goal is achieved by using recombinant retroviruses pseudotyped with either the gibbon ape leukemia virus (GaLV) envelope or the vesicular stomatitis virus G (VSV-G) glycoprotein. We show here that both retroviral particles mediate stable gene transfer in CD4+ and in CD8+ peripheral blood lymphocytes cultured under optimized conditions. However, VSV-G-pseudotyped virions may cause transduction artifacts that must be carefully excluded. The VSV-G virions require 10- to 100-fold higher concentrations of infectious particles to achieve levels of gene transfer comparable to GaLV-virions. Nonetheless, the physical stability of VSV-G-coated particles enables the concentration of viral stocks to 10(9) infectious particles per milliliter or more.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Gene Products, env/physiology , Genetic Vectors/physiology , Leukemia Virus, Gibbon Ape/physiology , Membrane Glycoproteins , Receptors, Nerve Growth Factor/genetics , Transduction, Genetic , Transfection , Vesicular stomatitis Indiana virus/physiology , Viral Envelope Proteins/physiology , 3T3 Cells , Animals , Artifacts , DNA, Complementary/genetics , False Positive Reactions , Genetic Vectors/chemistry , Genetic Vectors/ultrastructure , HeLa Cells , Humans , Leukemia Virus, Gibbon Ape/chemistry , Lung Neoplasms/pathology , Mice , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/chemistry
17.
Hum Gene Ther ; 7(12): 1415-22, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8844200

ABSTRACT

Gene-modified lymphocytes have a potential role in the therapy of cancer, infectious diseases, and genetic disorders of the immune system. Current gene therapy protocols involving gene transfer into lymphocytes utilize retroviruses with amphotropic envelope proteins. However, transduction efficiencies in lymphocytes using these viruses are relatively low. A potential strategy to improve gene transfer efficiency is the utilization of alternative retroviral envelopes that target unique receptors on the cell surface. One such alternative retroviral envelope, the gibbon ape leukemia virus (GALV) envelope, targets a distinct surface receptor (GLVR-1) that is 60% homologous but not cross-reactive to the amphotropic receptor (GLVR-2/RAM-1). Understanding the relationship between receptor expression and transduction efficiency is important for designing new strategies to improve gene transfer. Therefore, we compared GLVR-1 and GLVR-2 mRNA levels in lymphocytes and found that GLVR-1 was expressed 8- to 19-fold higher than GLVR-2. We then analyzed whether this enhanced expression of GLVR-1 correlated with increased infectivity of lymphocytes by retroviral vectors that utilize the GALV envelope compared to those that use the amphotropic envelope. We evaluated retroviral vectors packaged with either PA317 or PG13, which express the amphotropic and GALV envelopes, respectively. Lymphocyte transduction with PG13-packaged vectors was 4- to 18-fold higher than that with PA317-packaged vectors. These findings suggest that receptor expression level is an important factor in retroviral-target interactions and that gene transfer into human T lymphocytes should be performed with retroviruses that use the GALV envelope as opposed to retroviruses that use the amphotropic envelope.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/physiology , Leukemia Virus, Gibbon Ape/physiology , Lymphocytes, Tumor-Infiltrating/virology , Moloney murine leukemia virus/genetics , Phosphate Transport Proteins , Receptors, Virus/metabolism , Symporters , T-Lymphocytes/virology , Viral Envelope Proteins/physiology , Cells, Cultured , DNA, Complementary/genetics , Genetic Vectors/genetics , Humans , Leukemia Virus, Gibbon Ape/genetics , Lymphocytes, Tumor-Infiltrating/metabolism , Moloney murine leukemia virus/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Virus/genetics , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type III , T-Lymphocytes/metabolism , Viral Envelope Proteins/genetics
18.
Blood ; 86(6): 2379-87, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7662985

ABSTRACT

Leukocyte adherence deficiency (LAD) is an inherited immunodeficiency disease caused by defects in the CD18 leukocyte integrin subunit. Transduction of CD18 into hematopoietic cells from children with LAD represents a potential therapy for this disorder. In an attempt to maximize transfer and expression of CD18, we evaluated retroviral vectors with and without the neomycin selectable marker, with a modified tRNA primer binding site designed to prevent inhibition of gene expression, and with two different viral envelope proteins produced by using the amphotropic retrovirus packaging cell line PA317 or the gibbon ape leukemia virus packaging cell line PG13. The vectors were tested using transducing K562/CD11b cells and LAD Epstein-Barr virus (EBV) B cells and measuring levels of cell-surface CD11/CD18 expression by fluorescence-activated cell sorter analysis. The best results were obtained with vectors made using PG13 packaging cells, for which about 25% of the K562 cells exposed once to the vectors expressed surface CD11b/CD18 and about 25% of the LAD EBV B cells exposed three times over a 3-day period to the vectors expressed surface CD11a/CD18. In contrast, transduction of cells under similar conditions with retroviral vectors produced using PA317 producer cells yielded less than 2% of the K562 cells and less than 4% of the LAD EBV B cells expressing the CD11/CD18 heterodimer on the cell surface. The presence or absence of the neomycin resistance gene or the modified tRNA primer had no effect on CD18 gene transfer rate or expression level. The increase in transduction with PG13 vectors correlated with Northern blotting and reverse transcription-polymerase chain reaction studies that indicated that both K562 cells and the LAD EBV B cells express transcripts for the gibbon ape leukemia virus receptor at higher levels than for the amphotropic virus receptor. These findings indicate that the transduction efficiency of retroviral packaging cell lines correlates with receptor gene expression in the target cells and that vectors made using PG13 cells may be efficacious for gene therapy for LAD and other diseases in which gene transfer to hematopoietic cells is required.


Subject(s)
CD18 Antigens/genetics , Defective Viruses/physiology , Genetic Vectors/physiology , Hematopoietic Stem Cells , Leukemia Virus, Gibbon Ape/physiology , Leukocyte-Adhesion Deficiency Syndrome/pathology , Viral Envelope Proteins/physiology , B-Lymphocytes/metabolism , Base Sequence , CD11 Antigens/analysis , CD18 Antigens/biosynthesis , Cell Line, Transformed , Child , Defective Viruses/genetics , Genes, Reporter , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Herpesvirus 4, Human , Humans , Kanamycin Kinase , Leukemia Virus, Gibbon Ape/genetics , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/biosynthesis , Retroviridae/genetics , Retroviridae/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL