Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
J Virol ; 98(4): e0177123, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38440982

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancestral viral infections. Feline leukemia virus (FeLV) is an exogenous and endogenous retrovirus in domestic cats. It is classified into several subgroups (A, B, C, D, E, and T) based on viral receptor interference properties or receptor usage. ERV-derived molecules benefit animals, conferring resistance to infectious diseases. However, the soluble protein encoded by the defective envelope (env) gene of endogenous FeLV (enFeLV) functions as a co-factor in FeLV subgroup T infections. Therefore, whether the gene emerged to facilitate viral infection is unclear. Based on the properties of ERV-derived molecules, we hypothesized that the defective env genes possess antiviral activity that would be advantageous to the host because FeLV subgroup B (FeLV-B), a recombinant virus derived from enFeLV env, is restricted to viral transmission among domestic cats. When soluble truncated Env proteins from enFeLV were tested for their inhibitory effects against enFeLV and FeLV-B, they inhibited viral infection. Notably, this antiviral machinery was extended to infection with the Gibbon ape leukemia virus, Koala retrovirus A, and Hervey pteropid gammaretrovirus. Although these viruses used feline phosphate transporter 1 (fePit1) and phosphate transporter 2 as receptors, the inhibitory mechanism involved competitive receptor binding in a fePit1-dependent manner. The shift in receptor usage might have occurred to avoid the inhibitory effect. Overall, these findings highlight the possible emergence of soluble truncated Env proteins from enFeLV as a restriction factor against retroviral infection and will help in developing host immunity and antiviral defense by controlling retroviral spread.IMPORTANCERetroviruses are unique in using reverse transcriptase to convert RNA genomes into DNA, infecting germ cells, and transmitting to offspring. Numerous ancient retroviral sequences are known as endogenous retroviruses (ERVs). The soluble Env protein derived from ERVs functions as a co-factor that assists in FeLV-T infection. However, herein, we show that the soluble Env protein exhibits antiviral activity and provides resistance to mammalian retrovirus infection through competitive receptor binding. In particular, this finding may explain why FeLV-B transmission is not observed among domestic cats. ERV-derived molecules can benefit animals in an evolutionary arms race, highlighting the double-edged-sword nature of ERVs.


Subject(s)
Gene Products, env , Leukemia Virus, Feline , Leukemia, Feline , Animals , Cats , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Products, env/genetics , Gene Products, env/metabolism , Leukemia Virus, Feline/classification , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/metabolism , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Leukemia, Feline/genetics , Leukemia, Feline/metabolism , Leukemia, Feline/virology , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Receptors, Virus/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Solubility , Female
2.
Int J Mol Sci ; 25(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38473904

ABSTRACT

Cancer in dogs has increased in recent years and is a leading cause of death. We have developed a retroviral replicating vector (RRV) that specifically targets cancer cells for infection and replication. RRV carrying a suicide gene induced synchronized killing of cancer cells when administered with a prodrug after infection. In this study, we evaluated two distinct RRVs derived from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV) in canine tumor models both in vitro and in vivo. Despite low infection rates in normal canine cells, both RRVs efficiently infected and replicated within all the canine tumor cells tested. The efficient intratumoral spread of the RRVs after their intratumoral injection was also demonstrated in nude mouse models of subcutaneous canine tumor xenografts. When both RRVs encoded a yeast cytosine deaminase suicide gene, which converts the prodrug 5-fluorocytosine (5-FC) to the active drug 5-fluorouracil, they caused tumor-cell-specific 5-FC-induced killing of the canine tumor cells in vitro. Furthermore, in the AZACF- and AZACH-cell subcutaneous tumor xenograft models, both RRVs exerted significant antitumor effects. These results suggest that RRV-mediated suicide gene therapy is a novel therapeutic approach to canine cancers.


Subject(s)
Neoplasms , Prodrugs , Mice , Humans , Dogs , Animals , Genetic Therapy/methods , Cell Line, Tumor , Leukemia Virus, Gibbon Ape/genetics , Fluorouracil/pharmacology , Flucytosine/pharmacology , Prodrugs/pharmacology , Genetic Vectors , Cytosine Deaminase/genetics , Neoplasms/drug therapy
3.
Proc Natl Acad Sci U S A ; 121(6): e2220392121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38305758

ABSTRACT

Germline colonization by retroviruses results in the formation of endogenous retroviruses (ERVs). Most colonization's occurred millions of years ago. However, in the Australo-Papuan region (Australia and New Guinea), several recent germline colonization events have been discovered. The Wallace Line separates much of Southeast Asia from the Australo-Papuan region restricting faunal and pathogen dispersion. West of the Wallace Line, gibbon ape leukemia viruses (GALVs) have been isolated from captive gibbons. Two microbat species from China appear to have been infected naturally. East of Wallace's Line, the woolly monkey virus (a GALV) and the closely related koala retrovirus (KoRV) have been detected in eutherians and marsupials in the Australo-Papuan region, often vertically transmitted. The detected vertically transmitted GALV-like viruses in Australo-Papuan fauna compared to sporadic horizontal transmission in Southeast Asia and China suggest the GALV-KoRV clade originates in the former region and further models of early-stage genome colonization may be found. We screened 278 samples, seven bat and one rodent family endemic to the Australo-Papuan region and bat and rodent species found on both sides of the Wallace Line. We identified two rodents (Melomys) from Australia and Papua New Guinea and no bat species harboring GALV-like retroviruses. Melomys leucogaster from New Guinea harbored a genomically complete replication-competent retrovirus with a shared integration site among individuals. The integration was only present in some individuals of the species indicating this retrovirus is at the earliest stages of germline colonization of the Melomys genome, providing a new small wild mammal model of early-stage genome colonization.


Subject(s)
Chiroptera , Endogenous Retroviruses , Gammaretrovirus , Marsupialia , Animals , Leukemia Virus, Gibbon Ape/genetics , New Guinea , Gammaretrovirus/genetics , Murinae/genetics , Marsupialia/genetics , Germ Cells
4.
Anticancer Res ; 43(12): 5311-5317, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38030176

ABSTRACT

BACKGROUND/AIM: Retroviral replicating vectors (RRV) have exhibited efficient tumor transduction and improved therapeutic benefits in a variety of cancer models. In this study, we validated two RRV created from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV), which use different cell receptors for virus entry, in human ovarian cancer (OC) cells. MATERIALS AND METHODS: Expression levels of the receptors for AMLV (PiT-2) and GALV (PiT-1) in human OC cell lines (A2780, Caov3, RMG-1, SKOV-3), fibroblasts and HEK293 cells were evaluated using quantitative RT-PCR. In vitro RRV-GFP replication was monitored using flow cytometry, and cytotoxicity quantitated using AlamarBlue assay after 5-fluorocytosine treatment of OC cells transduced with RRV expressing the yeast cytosine deaminase prodrug activator gene. In vivo antitumor effect of RRV-mediated prodrug activator gene therapy was investigated in a SKOV-3 subcutaneous tumor model. RESULTS: Quantitative RT-PCR analysis revealed high expression levels of PiT-2 (AMLV receptor) and PiT-1 (GALV receptor) in the RMG-1 and SKOV3 OC cell lines, compared with their levels in non-malignant cells. In RMG-1 and SKOV3 cells, both RRV showed highly efficient RRV replication and spread leading to over 90% transduction by Days 10-13. Additionally, both RRV that express the yeast cytosine deaminase gene demonstrated effective cell killing of RMG-1 and SKOV-3 cells upon treatment with the prodrug 5-fluorocytosine. Notably, RRV-mediated prodrug activator gene therapy showed significant inhibition of subcutaneous SKOV-3 tumor growth in nude mice. CONCLUSION: RRV-mediated prodrug activator gene therapy may be used for treating PiT-expressing human OC.


Subject(s)
Ovarian Neoplasms , Prodrugs , Animals , Mice , Humans , Female , Cell Line, Tumor , Prodrugs/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/pharmacology , Mice, Nude , HEK293 Cells , Ovarian Neoplasms/therapy , Ovarian Neoplasms/drug therapy , Genetic Therapy , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Genetic Vectors/genetics
5.
Int J Mol Sci ; 24(19)2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37834271

ABSTRACT

Retroviral replicating vectors (RRVs) selectively replicate and can specifically introduce prodrug-activating genes into tumor cells, whereby subsequent prodrug administration induces the death of the infected tumor cells. We assessed the ability of two distinct RRVs generated from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV), which infect cells via type-III sodium-dependent phosphate transporters, PiT-2 and PiT-1, respectively, to infect human gastric cancer (GC) cells. A quantitative RT-PCR showed that all tested GC cell lines had higher expression levels of PiT-2 than PiT-1. Accordingly, AMLV, encoding a green fluorescent protein gene, infected and replicated more efficiently than GALV in most GC cell lines, whereas both RRVs had a low infection rate in human fibroblasts. RRV encoding a cytosine deaminase prodrug activator gene, which converts the prodrug 5-flucytosine (5-FC) to the active drug 5-fluorouracil, showed that AMLV promoted superior 5-FC-induced cytotoxicity compared with GALV, which correlated with the viral receptor expression level and viral spread. In MKN-74 subcutaneous xenograft models, AMLV had significant antitumor effects compared with GALV. Furthermore, in the MKN-74 recurrent tumor model in which 5-FC was discontinued, the resumption of 5-FC administration reduced the tumor volume. Thus, RRV-mediated prodrug activator gene therapy might be beneficial for treating human GC.


Subject(s)
Prodrugs , Stomach Neoplasms , Mice , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Prodrugs/pharmacology , Prodrugs/therapeutic use , Prodrugs/metabolism , Cell Line, Tumor , Genetic Therapy , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Genetic Vectors/genetics , Animals
6.
Viruses ; 14(4)2022 04 15.
Article in English | MEDLINE | ID: mdl-35458546

ABSTRACT

HIV-1 Vpu targets the host cell proteins CD4 and BST-2/Tetherin for degradation, ultimately resulting in enhanced virus spread and host immune evasion. The discovery and characterization of small molecules that antagonize Vpu would further elucidate the contribution of Vpu to pathogenesis and lay the foundation for the study of a new class of novel HIV-1 therapeutics. To identify novel compounds that block Vpu activity, we have developed a cell-based 'gain of function' assay that produces a positive signal in response to Vpu inhibition. To develop this assay, we took advantage of the viral glycoprotein, GaLV Env. In the presence of Vpu, GaLV Env is not incorporated into viral particles, resulting in non-infectious virions. Vpu inhibition restores infectious particle production. Using this assay, a high throughput screen of >650,000 compounds was performed to identify inhibitors that block the biological activity of Vpu. From this screen, we identified several positive hits but focused on two compounds from one structural family, SRI-41897 and SRI-42371. We developed independent counter-screens for off target interactions of the compounds and found no off target interactions. Additionally, these compounds block Vpu-mediated modulation of CD4, BST-2/Tetherin and antibody dependent cell-mediated toxicity (ADCC). Unfortunately, both SRI-41897 and SRI-42371 were shown to be specific to the N-terminal region of NL4-3 Vpu and did not function against other, more clinically relevant, strains of Vpu; however, this assay may be slightly modified to include more significant Vpu strains in the future.


Subject(s)
Anti-HIV Agents , HIV-1 , Human Immunodeficiency Virus Proteins , Viral Regulatory and Accessory Proteins , Viroporin Proteins , Anti-HIV Agents/chemistry , Bone Marrow Stromal Antigen 2/metabolism , GPI-Linked Proteins/metabolism , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Small Molecule Libraries , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/antagonists & inhibitors
7.
Viruses ; 13(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34452336

ABSTRACT

The Gibbon Ape Leukemia Virus envelope protein (GALV-Env) mediates efficient transduction of human cells, particularly primary B and T lymphocytes, and is therefore of great interest in gene therapy. Using internal domains from murine leukemia viruses (MLV), chimeric GALV-Env proteins such as GALV-C4070A were derived, which allow pseudotyping of lentiviral vectors. In order to improve expression efficiency and vector titers, we developed a codon-optimized (co) variant of GALV-C4070A (coGALV-Env). We found that coGALV-Env mediated efficient pseudotyping not only of γ-retroviral and lentiviral vectors, but also α-retroviral vectors. The obtained titers on HEK293T cells were equal to those with the classical GALV-Env, whereas the required plasmid amounts for transient vector production were significantly lower, namely, 20 ng coGALV-Env plasmid per 106 293T producer cells. Importantly, coGALV-Env-pseudotyped γ- and α-retroviral, as well as lentiviral vectors, mediated efficient transduction of primary human T cells. We propose that the novel chimeric coGALV-Env gene will be very useful for the efficient production of high-titer vector preparations, e.g., to equip human T cells with novel specificities using transgenic TCRs or CARs. The considerably lower amount of plasmid needed might also result in a significant cost advantage for good manufacturing practice (GMP) vector production based on transient transfection.


Subject(s)
Codon/genetics , Genetic Vectors/genetics , Lentivirus/genetics , Leukemia Virus, Gibbon Ape/genetics , Viral Envelope Proteins/genetics , Genetic Engineering , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Plasmids/genetics , Plasmids/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/virology , Transduction, Genetic , Viral Envelope Proteins/metabolism
8.
Arch Virol ; 165(5): 1089-1097, 2020 May.
Article in English | MEDLINE | ID: mdl-32146506

ABSTRACT

Gibbon ape leukemia virus (GALV) can infect a wide variety of cells but fails to infect most cells derived from laboratory mice. Transduction of human hematopoietic stem cells with GALV retroviral vectors is more efficient than with amphotropic vectors. In this study, a Moloney murine leukemia virus-gibbon ape leukemia virus (MoMLV-GALV) vector was constructed by replacing the natural env gene of the full-length Moloney MLV genome with the GALV env gene. To monitor viral transmission by green fluorescent protein (GFP) expression, internal ribosomal entry site-enhanced GFP (IRES-EGFP) was positioned between the GALV env gene and the 3' untranslated region (3' UTR) to obtain pMoMLV-GALV-EGFP. The MoMLV-GALV-EGFP vector was able to replicate with high titer in TE671 human rhabdomyosarcoma cells and U-87 human glioma cells. To evaluate the potential of the MoMLV-GALV vector as a therapeutic agent, the gene for the fusogenic envelope G glycoprotein of vesicular stomatitis virus (VSV-G) was incorporated into the vector. Infection with the resulting MoMLV-GALV-VSV-G vector resulted in lysis of the U-87 cells due to syncytium formation. Syncytium formation was also observed in the transfected human prostate cancer cell line LNCaP after extended cultivation of cells. In addition, we deleted the GALV env gene from the MoMLV-GALV-VSV-G vector to improve viral genome stability. This MoMLV-VSV-G vector is also replication competent and induces syncytium formation in 293T, HT1080, TE671 and U-87 cells. These results suggest that replication of the MoMLV-GALV-VSV-G vector or MoMLV-VSV-G vector may directly lead to cytotoxicity. Therefore, the vectors developed in this study are potentially useful tools for cancer gene therapy.


Subject(s)
Genetic Vectors , Leukemia Virus, Gibbon Ape/growth & development , Leukemia Virus, Murine/growth & development , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Cell Line , Genetic Therapy/methods , Humans , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Murine/genetics , Mice , Neoplasms/therapy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic
9.
Int J Mol Sci ; 21(4)2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32093290

ABSTRACT

Prodrug activator gene therapy mediated by murine leukemia virus (MLV)-based retroviral replicating vectors (RRV) was previously shown to be highly effective in killing glioma cells both in culture and in vivo. To avoid receptor interference and enable dual vector co-infection with MLV-RRV, we have developed another RRV based on gibbon ape leukemia virus (GALV) that also shows robust replicative spread in a wide variety of tumor cells. We evaluated the potential of GALV-based RRV as a cancer therapeutic agent by incorporating yeast cytosine deaminase (CD) and E. coli nitroreductase (NTR) prodrug activator genes into the vector. The expression of CD and NTR genes from GALV-RRV achieved highly efficient delivery of these prodrug activator genes to RG-2 glioma cells, resulting in enhanced cytotoxicity after administering their respective prodrugs 5-fluorocytosine and CB1954 in vitro. In an immune-competent intracerebral RG-2 glioma model, GALV-mediated CD and NTR gene therapy both significantly suppressed tumor growth with CB1954 administration after a single injection of vector supernatant. However, NTR showed greater potency than CD, with control animals receiving GALV-NTR vector alone (i.e., without CB1954 prodrug) showing extensive tumor growth with a median survival time of 17.5 days, while animals receiving GALV-NTR and CB1954 showed significantly prolonged survival with a median survival time of 30 days. In conclusion, GALV-RRV enabled high-efficiency gene transfer and persistent expression of NTR, resulting in efficient cell killing, suppression of tumor growth, and prolonged survival upon CB1954 administration. This validates the use of therapeutic strategies employing this prodrug activator gene to arm GALV-RRV, and opens the door to the possibility of future combination gene therapy with CD-armed MLV-RRV, as the latter vector is currently being evaluated in clinical trials.


Subject(s)
Aziridines/pharmacology , Brain Neoplasms/therapy , Flucytosine/pharmacology , Genetic Therapy , Genetic Vectors , Glioma/therapy , Neoplasms, Experimental/therapy , Prodrugs/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cytosine Deaminase/biosynthesis , Cytosine Deaminase/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Leukemia Virus, Gibbon Ape , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nitroreductases/biosynthesis , Nitroreductases/genetics , Rats, Inbred F344 , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics
10.
Hum Gene Ther ; 30(12): 1477-1493, 2019 12.
Article in English | MEDLINE | ID: mdl-31578886

ABSTRACT

Cell and gene therapies are finally becoming viable patient treatment options, with both T cell- and hematopoietic stem cell (HSC)-based therapies being approved to market in Europe. However, these therapies, which involve the use of viral vector to modify the target cells, are expensive and there is an urgent need to reduce manufacturing costs. One major cost factor is the viral vector production itself, therefore improving the gene modification efficiency could significantly reduce the amount of vector required per patient. This study describes the use of a transduction enhancing peptide, Vectofusin-1®, to improve the transduction efficiency of primary target cells using lentiviral and gammaretroviral vectors (LV and RV) pseudotyped with a variety of envelope proteins. Using Vectofusin-1 in combination with LV pseudotyped with viral glycoproteins derived from baboon endogenous retrovirus, feline endogenous virus (RD114), and measles virus (MV), a strongly improved transduction of HSCs, B cells and T cells, even when cultivated under low stimulation conditions, could be observed. The formation of Vectofusin-1 complexes with MV-LV retargeted to CD20 did not alter the selectivity in mixed cell culture populations, emphasizing the precision of this targeting technology. Functional, ErbB2-specific chimeric antigen receptor-expressing T cells could be generated using a gibbon ape leukemia virus (GALV)-pseudotyped RV. Using a variety of viral vectors and target cells, Vectofusin-1 performed in a comparable manner to the traditionally used surface-bound recombinant fibronectin. As Vectofusin-1 is a soluble peptide, it was possible to easily transfer the T cell transduction method to an automated closed manufacturing platform, where proof of concept studies demonstrated efficient genetic modification of T cells with GALV-RV and RD114-RV and the subsequent expansion of mainly central memory T cells to a clinically relevant dose.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Hematopoietic Stem Cells/drug effects , Peptides/pharmacology , Animals , Antigens, CD20/genetics , B-Lymphocytes/virology , Gammaretrovirus/genetics , Genetic Vectors/biosynthesis , Genetic Vectors/therapeutic use , Glycoproteins/genetics , Hematopoietic Stem Cells/virology , Humans , Lentivirus/genetics , Leukemia Virus, Gibbon Ape/genetics , Measles virus/genetics , Peptides/genetics , Retroviridae/genetics , T-Lymphocytes/virology , Transduction, Genetic , Viral Envelope Proteins/genetics
11.
Hum Gene Ther Methods ; 30(5): 153-171, 2019 10.
Article in English | MEDLINE | ID: mdl-31516018

ABSTRACT

Lentiviral vectors (LV) that are used in research and development as well as in clinical trials are in majority vesicular stomatitis virus G glycoprotein (VSVg) pseudotyped. The predominance of this pseudotype choice for clinical gene therapy studies is largely due to a lack of purification schemes for pseudotypes other than VSVg. In this study, we report for the first time the development of a new downstream process protocol allowing high-yield production of stable and infectious gibbon ape leukemia virus (GaLV)-TR-LV particles. We identified critical conditions in tangential flow filtration (TFF) and chromatographic steps for preserving the infectivity/functionality of LV during purification. This was carried out by identifying for each step, the critical parameters affecting LV infectivity, including pH, salinity, presence of stabilizers, temperature, and by defining the optimal order of these steps. A three-step process was developed for GaLV-TR-LV purification consisting of one TFF and two chromatographic steps (ion-exchange chromatography and size exclusion chromatography) permitting recoveries of >27% of infectious particles. With this process, purified GaLV-pseudotyped LV enabled the transduction of 70% human CD34+ cells in the presence of the Vectofusin-1 peptide, whereas in the same conditions nonpurified vector transduced only 9% of the cells (multiplicity of infection 20). Our protocol will allow for the first time the purification of GaLV-TR-LV that are biologically active, stable, and with sufficient recovery in the perspective of preclinical studies and clinical applications. Obviously, further optimizations are required to improve final vector yields.


Subject(s)
Lentivirus/isolation & purification , Leukemia Virus, Gibbon Ape/genetics , Chromatography, Gel , Chromatography, Ion Exchange , Genetic Vectors , Green Fluorescent Proteins/genetics , HCT116 Cells , HEK293 Cells , HIV-1 , Humans , Lentivirus/genetics , Transduction, Genetic
12.
Virus Genes ; 55(3): 421-424, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30877415

ABSTRACT

A novel gamma-retroviral sequence (7912 bp), inclusive of both partial 5' and 3' long terminal repeat regions, was identified from the brain of a black flying-fox (Pteropus alecto), Queensland, Australia. The sequence was distinct from other retroviral sequences identified in bats and showed greater identity to Koala, Gibbon ape leukaemia, Melomys burtoni and Woolly monkey retroviruses, forming their own phylogenetic clade. This finding suggests that these retroviruses may have an unknown common ancestor and that further investigation into the diversity of gamma-retroviruses in Australian Pteropus species may elucidate their evolutionary origins.


Subject(s)
Chiroptera/virology , Hylobates/virology , Phascolarctidae/virology , Retroviridae/genetics , Animals , Australia , Chiroptera/genetics , Hylobates/genetics , Leukemia Virus, Gibbon Ape/genetics , Phascolarctidae/genetics , Phylogeny , Sarcoma Virus, Woolly Monkey/genetics
13.
Cancer Gene Ther ; 26(1-2): 41-47, 2019 02.
Article in English | MEDLINE | ID: mdl-30042500

ABSTRACT

Retroviral replicating vectors (RRVs) have achieved efficient tumor transduction and enhanced therapeutic benefit in a wide variety of cancer models. Here, we evaluated two different RRVs derived from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV), which utilize different cellular receptors (PiT-2 and PiT-1, respectively) for viral entry, in human osteosarcoma cells. Quantitative RT-PCR showed that low levels of expression of both receptors were observed in normal and non-malignant cells. However, high PiT-2 (for AMLV) and low PiT-1 (for GALV) expression was observed in most osteosarcoma cell lines. Accordingly, AMLV expressing the green fluorescent protein gene infected and replicated more efficiently than GALV in most osteosarcoma cell lines. Furthermore, RRVs expressing the cytosine deaminase prodrug activator gene showed differential cytotoxicity that correlated with the results of viral spread. AMLV-RRV-mediated prodrug activator gene therapy achieved significant inhibition of subcutaneous MG-63 tumor growth over GALV in nude mice. These data indicate that AMLV vectors predominate over GALV in human osteosarcoma cells. Moreover, our findings support the potential utility of the two RRVs in personalized cancer virotherapy on the basis of receptor expression.


Subject(s)
Bone Neoplasms/therapy , Leukemia Virus, Gibbon Ape , Leukemia Virus, Murine , Oncolytic Virotherapy , Osteosarcoma/therapy , Receptors, Virus , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Cytosine Deaminase , Female , Humans , Mice , Mice, Nude , Osteosarcoma/metabolism , Prodrugs , Treatment Outcome , Xenograft Model Antitumor Assays
14.
Viruses ; 10(10)2018 10 19.
Article in English | MEDLINE | ID: mdl-30347660

ABSTRACT

The Human immunodeficiency virus-1 (HIV-1) accessory protein Vpu modulates numerous proteins, including the host proteins CD4 and BST-2/tetherin. Vpu interacts with the Skp, Cullin, F-Box (SCF) ubiquitin ligase through interactions with the F-Box protein ßTrCP (1 and/or 2). This interaction is dependent on phosphorylation of S52,56 in Vpu. Mutation of S52,56, or inhibition of the SCF, abolishes most Vpu activity against CD4 and partly reduces activity against BST-2/tetherin. Recently, Vpu has also been reported to interact with the clathrin adapter proteins AP-1 and AP-2, and these interactions were also found to be required for BST-2/tetherin antagonism in an S52,56 -dependent manner. In assays where HIV-1 is pseudotyped with gibbon ape leukemia virus (GaLV Env), Vpu has also been found to prevent GaLV Env from being incorporated into viral particles, but the mechanism for this antagonism is not fully understood. To clarify the role of the ßTrCPs in Vpu function we used CRISPR/Cas9 to generate a clonal cell line lacking both ßTrCP-1 and -2. Vpu activity against CD4 and GaLV Env was abolished in this cell line, and activity against BST-2/tetherin reduced significantly. Mutation of the S52,56 residues no longer affected Vpu activity against BST-2/tetherin in this cell line. These data suggest that the primary role of the S52,56 residues in antagonism of CD4, GaLV Env, and BST-2/tetherin is to recruit the SCF/ßTrCP ubiquitin ligase.


Subject(s)
Antigens, CD/metabolism , CD4 Antigens/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Viral Envelope Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Antigens, CD/genetics , CD4 Antigens/genetics , Cell Line , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/genetics , Humans , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Protein Binding , Viral Envelope Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , beta-Transducin Repeat-Containing Proteins/genetics
15.
Hum Gene Ther Methods ; 28(2): 78-90, 2017 04.
Article in English | MEDLINE | ID: mdl-28301970

ABSTRACT

Gammaretrovirus and lentivirus are the preferred viral vectors to genetically modify T and natural killer cells to be used in immune cell therapies. The transduction efficiency of hematopoietic and T cells is more efficient using gibbon ape leukemia virus (GaLV) pseudotyping. In this context gammaretroviral vector producer cells offer competitive higher titers than transient lentiviral vectors productions. The main aim of this work was to identify the key parameters governing GaLV-pseudotyped gammaretroviral vector productivity in stable producer cells, using a retroviral vector expression cassette enabling positive (facilitating cell enrichment) and negative cell selection (allowing cell elimination). The retroviral vector contains a thymidine kinase suicide gene fused with a ouabain-resistant Na+,K+-ATPase gene, a potential safer and faster marker. The establishment of retroviral vector producer cells is traditionally performed by randomly integrating the retroviral vector expression cassette codifying the transgene. More recently, recombinase-mediated cassette exchange methodologies have been introduced to achieve targeted integration. Herein we compared random and targeted integration of the retroviral vector transgene construct. Two retroviral producer cell lines, 293 OuaS and 293 FlexOuaS, were generated by random and targeted integration, respectively, producing high titers (on the order of 107 infectious particles·ml-1). Results showed that the retroviral vector transgene cassette is the key retroviral vector component determining the viral titers notwithstanding, single-copy integration is sufficient to provide high titers. The expression levels of the three retroviral constructs (gag-pol, GaLV env, and retroviral vector transgene) were analyzed. Although gag-pol and GaLV env gene expression levels should surpass a minimal threshold, we found that relatively modest expression levels of these two expression cassettes are required. Their levels of expression should not be maximized. We concluded, to establish a high producer retroviral vector cell line only the expression level of the genomic retroviral RNA, that is, the retroviral vector transgene cassette, should be maximized, both through (1) the optimization of its design (i.e., genetic elements composition) and (2) the selection of high expressing chromosomal locus for its integration. The use of methodologies identifying and promoting integration into high-expression loci, as targeted integration or high-throughput screening are in this perspective highly valuable.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Retroviridae/genetics , Virus Integration/genetics , Animals , Cell Line , Gene Expression , Genes, Transgenic, Suicide/genetics , Genetic Vectors/therapeutic use , Humans , Lentivirus/genetics , Leukemia Virus, Gibbon Ape/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Thymidine Kinase/genetics
16.
Virus Genes ; 53(2): 165-172, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28220345

ABSTRACT

Is the origin of gibbon ape leukemia virus (GALV) human after all? When GALV was discovered and found to cause neoplastic disease in gibbons, it stimulated a great deal of research including investigations into the origins of this virus. A number of publications have suggested that the GALV progenitor was a retrovirus present in one of several species of South East Asian rodents that had close contact with captive gibbons. However, there are no published retroviral sequences from any South East Asian species to support this view. Here we present an alternative hypothesis that the origin of GALV is a virus closely related to Melomys burtoni retrovirus, and that this virus infected human patients in Papua New Guinea from whom biological material was obtained or in some way contaminated these samples. This material we propose contained infectious MbRV-related virus that was then unwittingly introduced into gibbons which subsequently developed GALV infections.


Subject(s)
Hylobates/virology , Leukemia Virus, Gibbon Ape/genetics , RNA, Viral/genetics , Retroviridae Infections/genetics , Animals , Humans , Hylobates/genetics , Leukemia Virus, Gibbon Ape/pathogenicity , Phylogeny , Retroviridae/genetics , Retroviridae/pathogenicity , Retroviridae Infections/virology , Rodentia/virology
17.
J Virol ; 90(18): 8169-80, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27384662

ABSTRACT

UNLABELLED: Gibbon ape leukemia virus (GALV) and koala retrovirus (KoRV) most likely originated from a cross-species transmission of an ancestral retrovirus into koalas and gibbons via one or more intermediate as-yet-unknown hosts. A virus highly similar to GALV has been identified in an Australian native rodent (Melomys burtoni) after extensive screening of Australian wildlife. GALV-like viruses have also been discovered in several Southeast Asian species, although screening has not been extensive and viruses discovered to date are only distantly related to GALV. We therefore screened 26 Southeast Asian rodent species for KoRV- and GALV-like sequences, using hybridization capture and high-throughput sequencing, in the attempt to identify potential GALV and KoRV hosts. Only the individuals belonging to a newly discovered subspecies of Melomys burtoni from Indonesia were positive, yielding an endogenous provirus very closely related to a strain of GALV. The sequence of the critical receptor domain for GALV infection in the Indonesian M. burtoni subsp. was consistent with the susceptibility of the species to GALV infection. The second record of a GALV in M. burtoni provides further evidence that M. burtoni, and potentially other lineages within the widespread subfamily Murinae, may play a role in the spread of GALV-like viruses. The discovery of a GALV in the most western part of the Australo-Papuan distribution of M. burtoni, specifically in a transitional zone between Asia and Australia (Wallacea), may be relevant to the cross-species transmission to gibbons in Southeast Asia and broadens the known distribution of GALVs in wild rodents. IMPORTANCE: Gibbon ape leukemia virus (GALV) and the koala retrovirus (KoRV) are very closely related, yet their hosts neither are closely related nor overlap geographically. Direct cross-species infection between koalas and gibbons is unlikely. Therefore, GALV and KoRV may have arisen via a cross-species transfer from an intermediate host whose range overlaps those of both gibbons and koalas. Using hybridization capture and high-throughput sequencing, we have screened a wide range of rodent candidate hosts from Southeast Asia for KoRV- and GALV-like sequences. Only a Melomys burtoni subspecies from Wallacea (Indonesia) was positive for GALV. We report the genome sequence of this newly identified GALV, the critical domain for infection of its potential cellular receptor, and its phylogenetic relationships with the other previously characterized GALVs. We hypothesize that Melomys burtoni, and potentially related lineages with an Australo-Papuan distribution, may have played a key role in cross-species transmission to other taxa.


Subject(s)
Leukemia Virus, Gibbon Ape/isolation & purification , Murinae/virology , Retroviridae Infections/veterinary , Rodent Diseases/virology , Animals , High-Throughput Nucleotide Sequencing , Indonesia , Leukemia Virus, Gibbon Ape/genetics , Nucleic Acid Hybridization , Proviruses/genetics , Proviruses/isolation & purification , Retroviridae Infections/virology , Sequence Analysis, DNA
18.
Methods Mol Biol ; 1448: 41-8, 2016.
Article in English | MEDLINE | ID: mdl-27317171

ABSTRACT

Gene transfer vectors based on retroviridae are increasingly becoming a tool of choice for biomedical research and for the development of biotherapies in rare diseases or cancers. To meet the challenges of preclinical and clinical production, different steps of the production process of self-inactivating γ-retroviral (RVs) and lentiviral vectors (LVs) have been improved (e.g., transfection, media optimization, cell culture conditions). However, the increasing need for mass production of such vectors is still a challenge and could hamper their availability for therapeutic use. Recently, we observed that the use of a neutral pH during vector production is not optimal. The use of mildly acidic pH conditions (pH 6) can increase by two- to threefold the production of RVs and LVs pseudotyped with the vesicular stomatitis virus G (VSV-G) or gibbon ape leukemia virus (GALV) glycoproteins. Here, we describe the production protocol in mildly acidic pH conditions of GALVTR- and VSV-G-pseudotyped LVs using the transient transfection of HEK293T cells and the production protocol of GALV-pseudotyped RVs produced from a murine producer cell line. These protocols should help to achieve higher titers of vectors, thereby facilitating experimental research and therapeutic applications.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Lentivirus/genetics , Leukemia Virus, Murine/genetics , Membrane Glycoproteins/genetics , Viral Envelope Proteins/genetics , Acids/chemistry , Animals , Glycoproteins/genetics , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Leukemia Virus, Gibbon Ape/genetics , Mice , Transduction, Genetic/methods , Transfection
19.
J Virol ; 90(4): 1757-72, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26637454

ABSTRACT

UNLABELLED: Gibbon ape leukemia viruses (GALVs) are part of a larger group of pathogenic gammaretroviruses present across phylogenetically diverse host species of Australasian mammals. Despite the biomedical utility of GALVs as viral vectors and in cancer gene therapy, full genome sequences have not been determined for all of the five identified GALV strains, nor has a comprehensive evolutionary analysis been performed. We therefore generated complete genomic sequences for each GALV strain using hybridization capture and high-throughput sequencing. The four strains of GALV isolated from gibbons formed a monophyletic clade that was closely related to the woolly monkey virus (WMV), which is a GALV strain that likely originated in a gibbon host. The GALV-WMV clade in turn formed a sister group to the koala retroviruses (KoRVs). Genomic signatures of episodic diversifying selection were detected among the gammaretroviruses with concentration in the env gene across the GALV strains that were particularly oncogenic and KoRV strains that were potentially exogenous, likely reflecting their adaptation to the host immune system. In vitro studies involving vectors chimeric between GALV and KoRV-B established that variable regions A and B of the surface unit of the envelope determine which receptor is used by a viral strain to enter host cells. IMPORTANCE: The gibbon ape leukemia viruses (GALVs) are among the most medically relevant retroviruses due to their use as viral vectors for gene transfer and in cancer gene therapy. Despite their importance, full genome sequences have not been determined for the majority of primate isolates, nor has comprehensive evolutionary analysis been performed, despite evidence that the viruses are facing complex selective pressures associated with cross-species transmission. Using hybridization capture and high-throughput sequencing, we report here the full genome sequences of all the GALV strains and demonstrate that diversifying selection is acting on them, particularly in the envelope gene in functionally important domains, suggesting that host immune pressure is shaping GALV evolution.


Subject(s)
Evolution, Molecular , Hylobates/virology , Leukemia Virus, Gibbon Ape/genetics , Selection, Genetic , Animals , Australasia , Cluster Analysis , Gene Products, env/genetics , Genetic Vectors , Genome, Viral , High-Throughput Nucleotide Sequencing , Molecular Sequence Data , Phascolarctidae , Phylogeny , RNA, Viral/genetics , Recombination, Genetic , Sequence Analysis, DNA , Sequence Homology , Virus Internalization
20.
J Biol Chem ; 291(5): 2161-9, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26668323

ABSTRACT

Gene delivery into hCD34+ hematopoietic stem/progenitor cells (HSPCs) using human immunodeficiency virus, type 1-derived lentiviral vectors (LVs) has several promising therapeutic applications. Numerous clinical trials are currently underway. However, the efficiency, safety, and cost of LV gene therapy could be ameliorated by enhancing target cell transduction levels and reducing the amount of LV used on the cells. Several transduction enhancers already exist, such as fibronectin fragments or cationic compounds. Recently, we discovered Vectofusin-1, a new transduction enhancer, also called LAH4-A4, a short histidine-rich amphipathic peptide derived from the LAH4 family of DNA transfection agents. Vectofusin-1 enhances the infectivity of lentiviral and γ-retroviral vectors pseudotyped with various envelope glycoproteins. In this study, we compared a family of Vectofusin-1 isomers and showed that Vectofusin-1 remains the lead peptide for HSPC transduction enhancement with LVs pseudotyped with vesicular stomatitis virus glycoproteins and also with modified gibbon ape leukemia virus glycoproteins. By comparing the capacity of numerous Vectofusin-1 variants to promote the modified gibbon ape leukemia virus glycoprotein-pseudotyped lentiviral vector infectivity of HSPCs, the lysine residues on the N-terminal extremity of Vectofusin-1, a hydrophilic angle of 140° formed by the histidine residues in the Schiffer-Edmundson helical wheel representation, hydrophobic residues consisting of leucine were all found to be essential and helped to define a minimal active sequence. The data also show that the critical determinants necessary for lentiviral transduction enhancement are partially different from those necessary for efficient antibiotic or DNA transfection activity of LAH4 derivatives. In conclusion, these results help to decipher the action mechanism of Vectofusin-1 in the context of hCD34+ cell-based gene therapy.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/cytology , Lentivirus , Amino Acid Sequence , Antigens, CD34/metabolism , DNA/chemistry , Genetic Therapy/methods , Glycoproteins/chemistry , HCT116 Cells , HEK293 Cells , HIV-1/metabolism , HeLa Cells , Histidine/chemistry , Humans , Leukemia Virus, Gibbon Ape , Molecular Sequence Data , Peptides/chemistry , Sequence Homology, Amino Acid , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...