Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Int J Mol Sci ; 25(5)2024 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-38473904

RESUMO

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.


Assuntos
Neoplasias , Pró-Fármacos , Camundongos , Humanos , Cães , Animais , Terapia Genética/métodos , Linhagem Celular Tumoral , Vírus da Leucemia do Macaco Gibão/genética , Fluoruracila/farmacologia , Flucitosina/farmacologia , Pró-Fármacos/farmacologia , Vetores Genéticos , Citosina Desaminase/genética , Neoplasias/tratamento farmacológico
2.
Proc Natl Acad Sci U S A ; 121(6): e2220392121, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38305758

RESUMO

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.


Assuntos
Quirópteros , Retrovirus Endógenos , Gammaretrovirus , Marsupiais , Animais , Vírus da Leucemia do Macaco Gibão/genética , Nova Guiné , Gammaretrovirus/genética , Murinae/genética , Marsupiais/genética , Células Germinativas
3.
Anticancer Res ; 43(12): 5311-5317, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38030176

RESUMO

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.


Assuntos
Neoplasias Ovarianas , Pró-Fármacos , Animais , Camundongos , Humanos , Feminino , Linhagem Celular Tumoral , Pró-Fármacos/farmacologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Flucitosina/farmacologia , Camundongos Nus , Células HEK293 , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/tratamento farmacológico , Terapia Genética , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Vetores Genéticos/genética
4.
Int J Mol Sci ; 24(19)2023 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-37834271

RESUMO

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.


Assuntos
Pró-Fármacos , Neoplasias Gástricas , Camundongos , Humanos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Pró-Fármacos/metabolismo , Linhagem Celular Tumoral , Terapia Genética , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Vetores Genéticos/genética , Animais
5.
Viruses ; 13(8)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34452336

RESUMO

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.


Assuntos
Códon/genética , Vetores Genéticos/genética , Lentivirus/genética , Vírus da Leucemia do Macaco Gibão/genética , Proteínas do Envelope Viral/genética , Engenharia Genética , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Lentivirus/metabolismo , Vírus da Leucemia do Macaco Gibão/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/virologia , Transdução Genética , Proteínas do Envelope Viral/metabolismo
6.
Arch Virol ; 165(5): 1089-1097, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32146506

RESUMO

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.


Assuntos
Vetores Genéticos , Vírus da Leucemia do Macaco Gibão/crescimento & desenvolvimento , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vesiculovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Linhagem Celular , Terapia Genética/métodos , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia Murina/genética , Camundongos , Neoplasias/terapia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinação Genética
7.
Hum Gene Ther ; 30(12): 1477-1493, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31578886

RESUMO

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.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Antígenos CD20/genética , Linfócitos B/virologia , Gammaretrovirus/genética , Vetores Genéticos/biossíntese , Vetores Genéticos/uso terapêutico , Glicoproteínas/genética , Células-Tronco Hematopoéticas/virologia , Humanos , Lentivirus/genética , Vírus da Leucemia do Macaco Gibão/genética , Vírus do Sarampo/genética , Peptídeos/genética , Retroviridae/genética , Linfócitos T/virologia , Transdução Genética , Proteínas do Envelope Viral/genética
8.
Hum Gene Ther Methods ; 30(5): 153-171, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31516018

RESUMO

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.


Assuntos
Lentivirus/isolamento & purificação , Vírus da Leucemia do Macaco Gibão/genética , Cromatografia em Gel , Cromatografia por Troca Iônica , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Células HCT116 , Células HEK293 , HIV-1 , Humanos , Lentivirus/genética , Transdução Genética
9.
Virus Genes ; 55(3): 421-424, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30877415

RESUMO

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.


Assuntos
Quirópteros/virologia , Hylobates/virologia , Phascolarctidae/virologia , Retroviridae/genética , Animais , Austrália , Quirópteros/genética , Hylobates/genética , Vírus da Leucemia do Macaco Gibão/genética , Phascolarctidae/genética , Filogenia , Vírus do Sarcoma do Macaco-Barrigudo/genética
10.
Viruses ; 10(10)2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30347660

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Antígenos CD4/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Antígenos CD/genética , Antígenos CD4/genética , Linhagem Celular , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Ligação Proteica , Proteínas do Envelope Viral/genética , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Contendo Repetições de beta-Transducina/genética
11.
Hum Gene Ther Methods ; 28(2): 78-90, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28301970

RESUMO

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.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Retroviridae/genética , Integração Viral/genética , Animais , Linhagem Celular , Expressão Gênica , Genes Transgênicos Suicidas/genética , Vetores Genéticos/uso terapêutico , Humanos , Lentivirus/genética , Vírus da Leucemia do Macaco Gibão/genética , ATPase Trocadora de Sódio-Potássio/genética , Timidina Quinase/genética
12.
Virus Genes ; 53(2): 165-172, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28220345

RESUMO

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.


Assuntos
Hylobates/virologia , Vírus da Leucemia do Macaco Gibão/genética , RNA Viral/genética , Infecções por Retroviridae/genética , Animais , Humanos , Hylobates/genética , Vírus da Leucemia do Macaco Gibão/patogenicidade , Filogenia , Retroviridae/genética , Retroviridae/patogenicidade , Infecções por Retroviridae/virologia , Roedores/virologia
13.
J Virol ; 90(18): 8169-80, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27384662

RESUMO

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.


Assuntos
Vírus da Leucemia do Macaco Gibão/isolamento & purificação , Murinae/virologia , Infecções por Retroviridae/veterinária , Doenças dos Roedores/virologia , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Indonésia , Vírus da Leucemia do Macaco Gibão/genética , Hibridização de Ácido Nucleico , Provírus/genética , Provírus/isolamento & purificação , Infecções por Retroviridae/virologia , Análise de Sequência de DNA
14.
Methods Mol Biol ; 1448: 41-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27317171

RESUMO

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.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Lentivirus/genética , Vírus da Leucemia Murina/genética , Glicoproteínas de Membrana/genética , Proteínas do Envelope Viral/genética , Ácidos/química , Animais , Glicoproteínas/genética , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Vírus da Leucemia do Macaco Gibão/genética , Camundongos , Transdução Genética/métodos , Transfecção
15.
J Virol ; 90(4): 1757-72, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26637454

RESUMO

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.


Assuntos
Evolução Molecular , Hylobates/virologia , Vírus da Leucemia do Macaco Gibão/genética , Seleção Genética , Animais , Australásia , Análise por Conglomerados , Produtos do Gene env/genética , Vetores Genéticos , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Dados de Sequência Molecular , Phascolarctidae , Filogenia , RNA Viral/genética , Recombinação Genética , Análise de Sequência de DNA , Homologia de Sequência , Internalização do Vírus
16.
J Immunother ; 38(3): 127-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751502

RESUMO

The successful genetic engineering of patient T cells with γ-retroviral vectors expressing chimeric antigen receptors or T-cell receptors for phase II clinical trials and beyond requires the large-scale manufacture of high-titer vector stocks. The production of retroviral vectors from stable packaging cell lines using roller bottles or 10- to 40-layer cell factories is limited by a narrow harvest window, labor intensity, open-system operations, and the requirement for significant incubator space. To circumvent these shortcomings, we optimized the production of vector stocks in a disposable fixed-bed bioreactor using good manufacturing practice-grade packaging cell lines. High-titer vector stocks were harvested over 10 days, representing a much broader harvest window than the 3-day harvest afforded by cell factories. For PG13 and 293Vec packaging cells, the average vector titer and the vector stocks' yield in the bioreactor were higher by 3.2- to 7.3-fold, and 5.6- to 13.1-fold, respectively, than those obtained in cell factories. The vector production was 10.4 and 18.6 times more efficient than in cell factories for PG13 and 293Vec cells, respectively. Furthermore, the vectors produced from the fixed-bed bioreactors passed the release test assays for clinical applications. Therefore, a single vector lot derived from 293Vec is suitable to transduce up to 500 patients cell doses in the context of large clinical trials using chimeric antigen receptors or T-cell receptors. These findings demonstrate for the first time that a robust fixed-bed bioreactor process can be used to produce γ-retroviral vector stocks scalable up to the commercialization phase.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Técnicas de Cultura Celular por Lotes/normas , Reatores Biológicos , Vetores Genéticos/genética , Vetores Genéticos/normas , Retroviridae/genética , Animais , Linhagem Celular Transformada , Células HEK293 , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Linfócitos T/metabolismo , Transdução Genética
17.
PLoS One ; 9(9): e106954, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25251014

RESUMO

Gibbon ape leukaemia virus (GALV) and koala retrovirus (KoRV) share a remarkably close sequence identity despite the fact that they occur in distantly related mammals on different continents. It has previously been suggested that infection of their respective hosts may have occurred as a result of a species jump from another, as yet unidentified vertebrate host. To investigate possible sources of these retroviruses in the Australian context, DNA samples were obtained from 42 vertebrate species and screened using PCR in order to detect proviral sequences closely related to KoRV and GALV. Four proviral partial sequences totalling 2880 bases which share a strong similarity with KoRV and GALV were detected in DNA from a native Australian rodent, the grassland melomys, Melomys burtoni. We have designated this novel gammaretrovirus Melomys burtoni retrovirus (MbRV). The concatenated nucleotide sequence of MbRV shares 93% identity with the corresponding sequence from GALV-SEATO and 83% identity with KoRV. The geographic ranges of the grassland melomys and of the koala partially overlap. Thus a species jump by MbRV from melomys to koalas is conceivable. However the genus Melomys does not occur in mainland South East Asia and so it appears most likely that another as yet unidentified host was the source of GALV.


Assuntos
Produtos do Gene pol/genética , Murinae/virologia , Retroviridae/genética , Animais , Austrália , Células Cultivadas , Chlorocebus aethiops , DNA Viral/química , DNA Viral/genética , Regulação Viral da Expressão Gênica , Produtos do Gene env/genética , Hylobates/virologia , Vírus da Leucemia do Macaco Gibão/genética , Masculino , Dados de Sequência Molecular , Phascolarctidae/virologia , Filogenia , Reação em Cadeia da Polimerase , RNA Viral/genética , Retroviridae/classificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Células Vero
18.
Cell Biochem Biophys ; 70(1): 321-6, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24687598

RESUMO

The aim of the study was to successfully construct three plasmids, which include the GALV.fus gene plasmid regulated by the herpes simplex virus type 1 (HSV-1) late expression gene-UL38 promoter and induced by HSV-1 (HSV-UL38P-GALV.fus), the cytomegalovirus promoter without tumor specificity (CMVP) GALV.fus plasmid (HSV-CMVP-GALV.fus), and the control plasmid in which the GALV.fus gene fragment was replaced by the enhanced green fluorescent protein (EGFP) gene fragment (HSV-CMVP-EGFP). The three constructed plasmids were all packaged and named as Synco-2, Synco-1, and Baco-1. The plasmids were amplified in coliform bacterium and transfected into Vero cells using lipofectamine. These recombinant HSV-1 were amplified in Vero cells and purified by conventional methods of cesium chloride, TCID50 method is used to measure virus titers. The total RNA was then extracted from the HepG2 cells transfected by Synco-1 and Synco-2, and the expression of GALV.fus mRNA was detected by RT-PCR. The three recombinant HSV-1 vectors were propagated in Vero cells and purified by cesium chloride density gradient centrifugation, titrated by TCID50 method, and packaged. The titers of Baco-1, Synco-1, and Synco-2 were 3 × 10(10), 1 × 10(11), and 4 × 10(10) pfu/ml. The GALV.fus gene was identified in the infected HepG2 cells by RT-PCR method.


Assuntos
DNA Recombinante/genética , Engenharia Genética/métodos , Herpesvirus Humano 1/genética , Vírus da Leucemia do Macaco Gibão/genética , Técnicas de Amplificação de Ácido Nucleico , Proteína FUS de Ligação a RNA/genética , Montagem de Vírus , Animais , Chlorocebus aethiops , Terapia Genética , Células Hep G2 , Herpesvirus Humano 1/fisiologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Plasmídeos/genética , RNA/genética , RNA/isolamento & purificação , Transfecção , Células Vero
19.
Gene Ther ; 21(5): 533-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24694535

RESUMO

Adoptive T-cell transfer for cancer immunotherapy requires genetic modification of T cells with recombinant T-cell receptors (TCRs). Amphotropic retroviral vectors (RVs) used for TCR transduction for this purpose are considered safe in principle. Despite this, TCR-coding and packaging vectors could theoretically recombine to produce replication competent vectors (RCVs), and transduced T-cell preparations must be proven free of RCV. To eliminate the need for RCV testing, we transduced human T cells with ecotropic RVs so potential RCV would be non-infectious for human cells. We show that transfection of synthetic messenger RNA encoding murine cationic amino-acid transporter 1 (mCAT-1), the receptor for murine retroviruses, enables efficient transient ecotropic transduction of human T cells. mCAT-1-dependent transduction was more efficient than amphotropic transduction performed in parallel, and preferentially targeted naive T cells. Moreover, we demonstrate that ecotropic TCR transduction results in antigen-specific restimulation of primary human T cells. Thus, ecotropic RVs represent a versatile, safe and potent tool to prepare T cells for the adoptive transfer.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Transportador 1 de Aminoácidos Catiônicos/genética , Receptores de Antígenos de Linfócitos T/genética , Retroviridae/genética , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Linhagem Celular , Eletroporação , Vetores Genéticos , Células HEK293 , Humanos , Imunoterapia Adotiva , Células Jurkat , Vírus da Leucemia do Macaco Gibão/genética , Glicoproteínas de Membrana/genética , Camundongos , Plasmídeos/genética , RNA Mensageiro/genética , Transdução Genética , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética
20.
Cell Biochem Biophys ; 69(3): 583-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24510538

RESUMO

The aim of this study is to observe the in vitro-targeted destruction of lung adenocarcinoma using recombinant Type I herpes simplex virus (HSV-I)-mediated gibbon ape leukemia virus envelope glycoprotein (GALV.fus), controlled by UL38 promoter and cytomegalovirus promoter (CMVP). A recombinant HSV-I plasmid encoding the GALV.fus was transfected into green monkey kidney cells, the lung adenocarcinoma line A549, and the human fetal fibroblast cell line HFL-I GNHu5 in various doses. The effects and expression of in vitro GALV.fus were observed using an inverted microscope. Enhanced green fluorescence protein expression served as the contro1 for GALV.fus. Recombinant HSV-I virus was produced. Fusogenic recombinant virus infection led to cell fusions in A549 in a dose-dependent manner. Nonfusogenic viruses only produced conventional cytotoxic effects. Recombinant HSV-I with the CMVP initiated cell fusions in HFL-1 GNHu5 cells with arrested cell cycles or as quiescence. HSV-I regulated by UL38p caused cell fusion only in growing cells. Protein expression of GALV.fus was confirmed by Western Blot in infected A549 and HFL-1 GNHu5. Delivery and tumor-specific expression of GALV.fus gene can selectively and safely target lung cancer in vitro, and may prove to be a novel gene therapy for lung cancer.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/terapia , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Proteínas Virais de Fusão/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , DNA Recombinante/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Herpesvirus Humano 1/fisiologia , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Pulmonares/patologia , Terapia Viral Oncolítica , Plasmídeos/genética , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...