Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Virol ; 98(4): e0177123, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38440982

RESUMO

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.


Assuntos
Produtos do Gene env , Vírus da Leucemia Felina , Leucemia Felina , Animais , Gatos , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Vírus da Leucemia Felina/classificação , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Leucemia Felina/genética , Leucemia Felina/metabolismo , Leucemia Felina/virologia , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo , Receptores Virais/metabolismo , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Solubilidade , Feminino
2.
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
3.
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
4.
Viruses ; 14(4)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35458546

RESUMO

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.


Assuntos
Fármacos Anti-HIV , HIV-1 , Proteínas do Vírus da Imunodeficiência Humana , Proteínas Virais Reguladoras e Acessórias , Proteínas Viroporinas , Fármacos Anti-HIV/química , Antígeno 2 do Estroma da Médula Óssea/metabolismo , Proteínas Ligadas por GPI/metabolismo , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Vírus da Leucemia do Macaco Gibão/metabolismo , Bibliotecas de Moléculas Pequenas , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Viroporinas/antagonistas & inibidores
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.
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
7.
Metab Eng ; 20: 131-45, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24120735

RESUMO

Biopharmaceuticals derived from enveloped virus comprise an expanding market of vaccines, oncolytic vectors and gene therapy products. Thus, increased attention is given to the development of robust high-titer cell hosts for their manufacture. However, the knowledge on the physiological constraints modulating virus production is still scarce and the use of integrated strategies to improve hosts productivity and upstream bioprocess an under-explored territory. In this work, we conducted a functional genomics study, including the transcriptional profiling and central carbon metabolism analysis, following the metabolic changes in the transition 'parental-to-producer' of two human cell lines producing recombinant retrovirus. Results were gathered into three comprehensive metabolic maps, providing a broad and integrated overview of gene expression changes for both cell lines. Eight pathways were identified to be recruited in the virus production state: amino acid catabolism, carbohydrate catabolism and integration of the energy metabolism, nucleotide metabolism, glutathione metabolism, pentose phosphate pathway, polyamines biosynthesis and lipid metabolism. Their ability to modulate viral titers was experimentally challenged, leading to improved specific productivities of recombinant retrovirus up to 6-fold. Within recruited pathways in the virus production state, we sought for metabolic engineering gene targets in the low producing phenotypes. A mining strategy was used alternative to the traditional approach 'high vs. low producer' clonal comparison. Instead, 'high vs. low producer' from different genetic backgrounds (i.e. cell origins) were compared. Several genes were identified as limiting in the low-production phenotype, including two enzymes from cholesterol biosynthesis, two enzymes from glutathione biosynthesis and the regulatory machinery of polyamines biosynthesis. This is thus a frontier work, bridging fundamentals to technological research and contributing to enlarge our understanding of enveloped virus production dynamics in mammalian cell hosts.


Assuntos
Engenharia Celular , Vírus da Leucemia do Macaco Gibão/metabolismo , Vírus da Leucemia Murina/metabolismo , Infecções por Retroviridae/metabolismo , Animais , Células HEK293 , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia Murina/genética , Camundongos , Infecções por Retroviridae/genética
8.
PLoS One ; 8(6): e68507, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840857

RESUMO

HIV-1 forms infectious particles with Murine Leukemia virus (MLV) Env, but not with the closely related Gibbon ape Leukemia Virus (GaLV) Env. We have determined that the incompatibility between HIV-1 and GaLV Env is primarily caused by the HIV-1 accessory protein Vpu, which prevents GaLV Env from being incorporated into particles. We have characterized the 'Vpu sensitivity sequence' in the cytoplasmic tail domain (CTD) of GaLV Env using a chimeric MLV Env with the GaLV Env CTD (MLV/GaLV Env). Vpu sensitivity is dependent on an alpha helix with a positively charged face containing at least one Lysine. In the present study, we utilized functional complementation to address whether all the three helices in the CTD of an Env trimer have to contain the Vpu sensitivity motif for the trimer to be modulated by Vpu. Taking advantage of the functional complementation of the binding defective (D84K) and fusion defective (L493V) MLV and MLV/GaLV Env mutants, we were able to assay the activity of mixed trimers containing both MLV and GaLV CTDs. Mixed trimers containing both MLV and GaLV CTDs were functionally active and remained sensitive to Vpu. However, trimers containing an Env with the GaLV CTD and an Env with no CTD remained functional but were resistant to Vpu. Together these data suggest that the presence of at least one GaLV CTD is sufficient to make an Env trimer sensitive to Vpu, but only if it is part of a trimeric CTD complex.


Assuntos
Produtos do Gene env/genética , HIV-1/genética , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia Murina/genética , Estrutura Terciária de Proteína/genética , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Citoplasma/genética , Citoplasma/metabolismo , Produtos do Gene env/metabolismo , Células HEK293 , HIV-1/metabolismo , Humanos , Vírus da Leucemia do Macaco Gibão/metabolismo , Vírus da Leucemia Murina/metabolismo , Camundongos , Mutação , Proteínas do Envelope Viral/metabolismo
9.
Retrovirology ; 9: 51, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22691439

RESUMO

BACKGROUND: Both cell-free and cell-associated infection routes are important for retroviral dissemination. Regardless of the mechanism, the driving force of retroviral entry is the interaction between the viral envelope and its receptor. To date it remains unclear how decreased affinity of viruses for their receptors affects viral cell-free infection, cell-cell transmission, and spreading kinetics. We have previously characterized a mutant form of the amphotropic murine retrovirus receptor human phosphate transporter 2 (PiT2) wherein the single substitution of a glutamic acid for the lysine residue at position 522 of this receptor is sufficient to render it to function as a gibbon ape leukemia virus (GALV) receptor. RESULTS: In this study we analyzed the binding affinity of the mutant receptor PiT2K522E and determined that it has a 1000 fold decreased GALV envelope binding affinity compared to the GALV wild type receptor. The decreased affinity does not restrict the initiation of cell-free GALV infection. The diminished binding affinity does, however, correlate with a decrease in the ability of GALV to spread in cells expressing this mutant receptor. CONCLUSIONS: The reduced ability of GALV to subsequently spread among cells expressing PiT2K522E is likely resulted from reduced cell-cell transmission, the decreased ability of PiT2K522E-expressing cells to establish superinfection interference, and attendant cytopathic affects.


Assuntos
Vírus da Leucemia do Macaco Gibão/patogenicidade , Receptores Virais/metabolismo , Infecções por Retroviridae/virologia , Superinfecção/virologia , Interferência Viral , Ligação Viral , Animais , Células CHO , Técnicas de Cocultura , Cricetinae , Vetores Genéticos , Células Gigantes/virologia , Células HEK293 , Interações Hospedeiro-Parasita , Humanos , Vírus da Leucemia do Macaco Gibão/metabolismo , Camundongos , Receptores Virais/genética , Internalização do Vírus , Replicação Viral
10.
Biol Chem ; 392(10): 887-95, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21812756

RESUMO

Genetic modification of human embryonic stem cells (hESCs) using biophysical DNA transfection methods are hampered by the very low single cell survival rate and cloning efficiency of hESCs. Lentiviral gene transfer strategies are widely used to genetically modify hESCs but limited transduction efficiencies in the presence of feeder or stroma cells present problems, particularly if vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped viral particles are applied. Here, we investigated whether the recently described semen derived enhancer of virus infection (SEVI) and alternative viral envelope proteins derived from either Gibbon ape leukaemia virus (GALV) or feline leukaemia virus (RD114) are applicable for transducing hESCs during co-culture with feeder or stroma cells. Our first set of experiments demonstrates that SEVI has no toxic effect on murine or hESCs and that exposure to SEVI does not interfere with the pluripotency-associated phenotype. Focusing on hESCs, we were able to further demonstrate that SEVI increases the transduction efficiencies of GALV and RD114 pseudotyped lentiviral vectors. More importantly, aiming at targeted differentiation of hESCs into functional somatic cell types, GALV pseudotyped lentiviral particles could efficiently and exclusively transduce hESCs grown in co-culture with OP9-GFP stroma cells (which were often used to induce differentiation into haematopoietic derivatives).


Assuntos
Técnicas de Cocultura , Células-Tronco Embrionárias/metabolismo , Lentivirus/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Células Estromais/metabolismo , Transdução Genética/métodos , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Técnicas de Cocultura/métodos , Células-Tronco Embrionárias/citologia , Vetores Genéticos/genética , Humanos , Camundongos , Células Estromais/citologia
11.
J Biomed Sci ; 17: 91, 2010 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-21129221

RESUMO

The gibbon ape leukemia virus (GALV), the amphotropic murine leukemia virus (AMLV) and the human T-cell leukemia virus (HTLV) are retroviruses that specifically bind nutrient transporters with their envelope glycoproteins (Env) when entering host cells. Here, we used tagged ligands derived from GALV, AMLV, and HTLV Env to monitor the distribution of their cognate receptors, the inorganic phosphate transporters PiT1 and PiT2, and the glucose transporter GLUT1, respectively, in basal conditions and after acute energy deficiency. For this purpose, we monitored changes in the distribution of PiT1, PiT2 and GLUT1 in the cerebellum, the frontal cortex, the corpus callosum, the striatum and the substantia nigra (SN) of C57/BL6 mice after administration of 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridinium (MPTP), a mitochondrial complex I inhibitor which induces neuronal degeneration in the striato-nigral network.The PiT1 ligand stained oligodendrocytes in the corpus callosum and showed a reticular pattern in the SN. The PiT2 ligand stained particularly the cerebellar Purkinje cells, while GLUT1 labelling was mainly observed throughout the cortex, basal ganglia and cerebellar gray matter. Interestingly, unlike GLUT1 and PiT2 distributions which did not appear to be modified by MPTP intoxication, PiT1 immunostaining seemed to be more extended in the SN. The plausible reasons for this change following acute energy stress are discussed.These new ligands therefore constitute new metabolic markers which should help to unravel cellular adaptations to a wide variety of normal and pathologic conditions and to determine the role of specific nutrient transporters in tissue homeostasis.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Encéfalo/metabolismo , Transportador de Glucose Tipo 1/análise , Receptores Virais/análise , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/análise , Animais , Transporte Biológico , Biomarcadores/análise , Biomarcadores/metabolismo , Encéfalo/efeitos dos fármacos , Produtos do Gene env/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Receptores Virais/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo
12.
Hum Gene Ther ; 20(9): 966-74, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19453219

RESUMO

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.


Assuntos
Vetores Genéticos/fisiologia , Células-Tronco Hematopoéticas/virologia , Vírus da Leucemia do Macaco Gibão/fisiologia , Retroviridae/fisiologia , Transdução Genética , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Linhagem Celular , Meios de Cultura , Meios de Cultura Livres de Soro , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Linfócitos T/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus
13.
Hum Gene Ther ; 19(7): 744-52, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18557700

RESUMO

Low in vivo transduction efficiency and safety concerns have been hurdles for effective hematopoietic stem cell (HSC) gene therapy. Here, we investigate whether the safety and efficiency of retroviral gene transfer into HSCs can be improved by using human allogeneic umbilical cord blood (UCB)-derived supplements instead of fetal bovine serum (FBS). When CD34(+) cells were cultured ex vivo in UCB-derived serum (CBS) or plasma (CBP), comparable or higher maintenance of HSCs was observed than in FBS and serum-free substitution medium (SFM) as assessed by the frequency of positive engraftment and the level of engraftment in NOD/SCID mice after transplantation of cultured cells. CBS and CBP also exhibited higher level stabilization of retroviral particles than SFM during in vitro culture of retrovirus pseudotyped with gibbon ape leukemia virus or vesicular stomatitis virus glycoprotein. Retroviral gene transfer into CD34(+) cells performed with CBS or CBP resulted in increased gene transfer into CD34(+) cells and increased transduction of reconstituted bone marrow cells compared to transfers with SFM or FBS. The increased transduction of bone marrow cells was associated with a larger number of transduced progenitors in the recipient mice. Significant oligoclonality in the transduced progenitors, as determined by ligation-mediated polymerase chain reaction, suggested efficient retroviral targeting of multiple HSCs in the CBS- or CBP-supplemented media. Combined, our results show that allogeneic UCB-derived serum or plasma is a safe and easily accessible serum supplement that can support efficient retroviral gene transfer into HSCs for the clinical-grade manipulation of HSCs.


Assuntos
Sangue Fetal , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Retroviridae/genética , Transdução Genética , Transplante Homólogo , Animais , Antígenos CD34/metabolismo , Células da Medula Óssea , Bovinos , Células Cultivadas , Meios de Cultura/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Hematopoéticas/virologia , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Plasma , Soro , Proteínas do Envelope Viral
14.
J Gen Virol ; 87(Pt 6): 1577-1581, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16690921

RESUMO

The wild-type (wt) envelope (Env) proteins of spleen necrosis virus (SNV), together with the transmembrane (TM) protein fused to antibody domains (scFv), have been used for the generation of stable packaging cell lines releasing pseudotyped cell targeting vectors derived from SNV and Murine leukemia virus (MLV). As a first step towards assessing whether HIV-1(SNV/TM-scFv) packaging cells could be established for the production of lentiviral cell targeting vectors, it is reported here that infectious HIV-1-derived particles pseudotyped with wt SNV Env proteins could be generated. Using novel chimeric SNV-derived Env proteins encompassing wt and engineered cytoplasmic domains (C-tail) of the Gibbon ape leukemia virus (GaLV) TM protein, it was further shown that the wt C-tail not only excludes the GaLV TM protein from incorporation into HIV-1 particles, but confers this phenotype to other retroviral envelopes upon C-terminal fusion.


Assuntos
Vetores Genéticos , HIV-1/patogenicidade , Vírus da Leucemia do Macaco Gibão/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Animais , Linhagem Celular , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , HIV-1/genética , HIV-1/metabolismo , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , Proteínas do Envelope Viral/genética , Vírion/genética
15.
Cancer Res ; 66(9): 4835-42, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651439

RESUMO

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.


Assuntos
Fibrossarcoma/terapia , Flucitosina/farmacocinética , Vírus da Leucemia do Macaco Gibão/genética , Glicoproteínas de Membrana/genética , Terapia Viral Oncolítica/métodos , Simplexvirus/fisiologia , Animais , Biotransformação , Fusão Celular , Terapia Combinada , Citosina Desaminase/biossíntese , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/virologia , Flucitosina/farmacologia , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Humanos , Vírus da Leucemia do Macaco Gibão/metabolismo , Vírus da Leucemia do Macaco Gibão/fisiologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Pentosiltransferases/biossíntese , Pentosiltransferases/genética , Pentosiltransferases/metabolismo , Pró-Fármacos/farmacocinética , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Simplexvirus/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Mol Ther ; 11(3): 452-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15727942

RESUMO

A novel, stable human immunodeficiency virus type 1 vector packaging system, STAR, was tested for its ability to transduce human cord blood CD34+ progenitor cells assayed both in vitro and after transplantation into NOD/SCID mice. Vectors pseudotyped with three different gammaretrovirus envelopes were used: the amphotropic MLV envelope (MLV-A), a modified gibbon ape leukemia virus envelope (GALV+), and a modified feline endogenous virus RD114 envelope (RDpro). Gene transfer to freshly thawed CD34+ cells in the absence of cytokines was very low. Addition of cytokines increased gene transfer efficiency significantly and this was further augmented if the cells were prestimulated for 24 h. Concentration of the vectors (15-fold) by low-speed centrifugation increased gene transfer to CD34+ cells in vitro even further. More than 90% of cells were transduced with a single exposure to the RDpro vector as determined by GFP expression using flow cytometry. The two other pseudotypes transduced approximately 65-70% of the cells under the same conditions. Transplantation of CD34+ cells prestimulated for 24 h and then transduced with a single exposure to concentrated vector revealed that the RDpro vector transduced 55.1% of NOD/SCID repopulating human cells, which was significantly higher than the MLV-A (12.6%)- or GALV+ (25.1%)-pseudotyped vectors.


Assuntos
Antígenos CD34/imunologia , Vetores Genéticos , HIV-1 , Células-Tronco Hematopoéticas/metabolismo , Animais , Genes Reporter , HIV-1/genética , HIV-1/metabolismo , Células-Tronco Hematopoéticas/imunologia , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Vírus da Leucemia do Macaco Gibão/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
17.
Hum Gene Ther ; 14(5): 447-61, 2003 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-12691610

RESUMO

In previous studies we demonstrated that a modified human HSP70b promoter (HSE.70b) directs high levels of gene expression to tumor cells after mild hyperthermia treatment in the range of 41.5-44 degrees C. This transcriptional targeting system exhibits low basal activity at 37 degrees C, is highly induced (950-fold) after mild heat treatment (43 degrees C/30 min), and returns to basal activity levels within 12-24 hours of activation. Here we describe heat-directed targeting of an activated form of the Gibbon ape leukemia virus env protein (GALV FMG) to tumor cells. GALV FMG mediates cell-cell fusion, and when expressed in tumor cells can produce bystander effects of up to 1:200. Transient transfection of a HSE70b.GALV FMG minigene caused extensive syncytia formation in HeLa and HT-1080 cells following mild heat treatment (44 degrees C/30 min). Stable transfection into HT-1080 cells produced a cell line (HG5) that exhibits massive syncytia formation and a 60% reduction in viability relative to a vector-only control (CI1) following heat treatment in vitro. Mild hyperthermia also resulted in syncytia formation, necrosis, and complete macroscopic regression of HG5 xenograft tumors grown in the footpads of mice with severe combined immunodeficiency disorders (SCID). Median survival increased from 12.5 (in heated CI1 controls) to 52 days after a single heat treatment. Heat-directed tumor cell fusion may prove to be a highly beneficial adjunct to existing cancer treatment strategies that take advantage of the synergistic interaction between mild hyperthermia and radiation or chemotherapeutic drugs.


Assuntos
Temperatura Alta , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Experimentais/terapia , Adenoviridae/genética , Animais , Fusão Celular , Sobrevivência Celular , Vírus Defeituosos/genética , Feminino , Produtos do Gene env/genética , Vetores Genéticos , Proteínas de Choque Térmico HSP70/genética , Humanos , Vírus da Leucemia do Macaco Gibão/metabolismo , Camundongos , Camundongos SCID , Modelos Animais , Neoplasias Experimentais/patologia , Plasmídeos , Indução de Remissão , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Gene Med ; 4(6): 592-600, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12439851

RESUMO

BACKGROUND: In a strategy termed "Protease Targeting", retroviral vectors carrying an EGF infectivity-blocking domain fused to the N-terminus of the envelope SU via a MMP (matrix metalloproteinase)-cleavable linker were successfully used to target gene delivery to EGF receptor-(EGF-R-)positive tumour cells over-expressing MMPs. In the current study, we aimed to investigate whether this strategy could be applied to (a) limit the cytotoxic activity of a hyperfusogenic GALV therapeutic gene, and (b) enhance the immune-stimulatory properties of GALV via local, MMP-mediated release human granulocyte-macrophage colony stimulating factor (GM-CSF). METHODS: We generated GALV envelope expression constructs displaying EGF or GM-CSF blocking ligands at the N-terminus of GALV envelope SU via a non-cleavable, Factor Xa protease or MMP-cleavable linker and investigated their cytotoxicity on MMP-positive and negative cell lines. RESULTS: The unmodified hyperfusogenic GALV envelope was cytotoxic to all cell lines tested. The non-cleavable linker GALV envelope constructs caused no cytotoxicity, demonstrating efficient inhibition by the displayed domains. Moderate activation of fusion of the protease-cleavable linker constructs was observed in all cell lines, regardless of their level of MMP expression and of the specificity of the linker. High levels of the 'blocking domain' were detected in the cell supernatants due to dissociation of the surface unit (SU) from the transmembrane (TM) component of the GALV envelope glycoprotein TM. CONCLUSIONS: Unlike protease targeting in the context of retroviral vectors, protease activation of the cytotoxicity of GALV envelope by cleavage of a fusion blocking ligand at the cell surface does not appear to be specifically mediated by cell-surface MMPs. In addition, shedding of the SU-fusion protein from the TM limits the general applicability of this strategy for cancer gene therapy. Specificity of cell-cell fusion mediated by GALV envelope cannot be manipulated in the same fashion as virus-cell fusion.


Assuntos
Vírus da Leucemia do Macaco Gibão/metabolismo , Metaloproteinases da Matriz/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Fusão Celular , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Inibidores de Proteases/farmacologia , Especificidade por Substrato
19.
J Virol ; 76(15): 7683-93, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12097582

RESUMO

The mammalian gammaretroviruses gibbon ape leukemia virus (GALV) and feline leukemia virus subgroup B (FeLV-B) can use the same receptor, Pit1, to infect human cells. A highly polymorphic nine-residue sequence within Pit1, designated region A, has been proposed as the virus binding site, because mutations in this region abolish Pit1-mediated cellular infection by GALV and FeLV-B. However, a direct correlation between region A mutations deleterious for infection and loss of virus binding has not been established. We report that cells expressing a Pit1 protein harboring mutations in region A that abolish receptor function retain the ability to bind virus, indicating that Pit1 region A is not the virus binding site. Furthermore, we have now identified a second region in Pit1, comprising residues 232 to 260 (region B), that is required for both viral entry and virus binding. Epitope-tagged Pit1 proteins were used to demonstrate that mutations in region B result in improper orientation of Pit1 in the cell membrane. Compensatory mutations in region A can restore proper orientation and full receptor function to these region B mutants. Based on these results, we propose that region A of Pit1 confers competence for viral entry by influencing the topology of the authentic binding site in the membrane and hence its accessibility to a viral envelope protein. Based on glycosylation studies and results obtained by using N- and C-terminal epitope-tagged Pit1, region A and region B mutants, and the transmembrane helices predicted with the PHD PredictProtein algorithm, we propose a new Pit1 topology model.


Assuntos
Vírus da Leucemia Felina/patogenicidade , Vírus da Leucemia do Macaco Gibão/patogenicidade , Receptores Virais/química , Receptores Virais/metabolismo , Algoritmos , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Vírus da Leucemia Felina/metabolismo , Vírus da Leucemia do Macaco Gibão/metabolismo , Camundongos , Modelos Moleculares , Mutação
20.
Hum Gene Ther ; 13(8): 891-9, 2002 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12031122

RESUMO

In an effort to improve hematopoietic stem cell gene transfer rates using gibbon ape leukemia virus (GALV)-pseudotype retroviral vectors in baboons, we have studied preselection of transduced green fluorescent protein (GFP)-expressing CD34-enriched marrow cells. Three animals were transplanted with GFP-selected (GS) CD34-enriched marrow. To ensure engraftment, preselected GFP-positive cells were infused together with unselected neo-transduced cells. After transduction on fibronectin, cells were cultured for an additional 2 days to allow for expression of GFP. GFP-expressing cells were enriched by fluorescence-activated cell sorting and infused together with cells from the unselected fractions after myeloablative irradiation of the recipient. Three other animals were transplanted with GFP-transduced CD34-enriched cells without prior GFP selection (GU). At 4 weeks after transplant, the percentage of GFP-expressing white blood cells was significantly higher in the GS group (6.6%) than in the GU group (1.3%) (p < 0.002). The higher gene transfer levels in the animals transplanted with GS cells gradually declined, and by day 100 after transplant, gene transfer levels were similar in both groups. PCR analysis performed on genomic DNA isolated from peripheral blood cells demonstrated that the decline in GFP-positive cells was due to the loss of gene-marked cells and not due to loss of expression. These results show that transplantation of CD34-positive marrow cells selected for GFP-positive cells after transduction provides high levels of transduced granulocytes in the short term. However, using this experimental design with concomitant infusion of unselected cells and the use of oncoretroviral vectors, preenrichment of vector-expressing, transduced CD34-enriched cells does not improve long-term persistence and expression.


Assuntos
Antígenos CD34/genética , Células da Medula Óssea/metabolismo , Vetores Genéticos/metabolismo , Vírus da Leucemia do Macaco Gibão/metabolismo , Proteínas Luminescentes/genética , Transdução Genética , Animais , Transplante de Medula Óssea , Citometria de Fluxo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Cinética , Vírus da Leucemia do Macaco Gibão/genética , Proteínas Luminescentes/sangue , Camundongos , Microscopia de Fluorescência , Papio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA