Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Biologicals ; 71: 1-8, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34039532

RESUMO

Xenogenic cell-based therapeutic products are expected to alleviate the chronic shortage of human donor organs. For example, porcine islet cell products are currently under development for the treatment of human diabetes. As porcine cells possess endogenous retrovirus (PERV), which can replicate in human cells in vitro, the potential transmission of PERV has raised concerns in the case of products that use living pig cells as raw materials. Although several PERV sequences exist in the porcine genome, not all have the ability to infect human cells. Therefore, polymerase chain reaction analysis, which amplifies a portion of the target gene, may not accurately assess the infection risk. Here, we determined porcine genome sequences and evaluated the infectivity of PERVs using high-throughput sequencing technologies. RNA sequencing was performed on both PERV-infected human cells and porcine cells, and reads mapped to PERV sequences were examined. The normalized number of the reads mapped to PERV regions was able to predict the infectivity of PERVs, indicating that it would be useful for evaluation of the PERV infection risk prior to transplantation of porcine products.


Assuntos
Retrovirus Endógenos , Gammaretrovirus , Sequenciamento de Nucleotídeos em Larga Escala , Animais , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Ilhotas Pancreáticas/virologia , Suínos , Transplante Heterólogo
2.
Retrovirology ; 17(1): 34, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33008414

RESUMO

BACKGROUND: Koalas are infected with the koala retrovirus (KoRV) that exists as exogenous or endogenous viruses. KoRV is genetically diverse with co-infection with up to ten envelope subtypes (A-J) possible; KoRV-A is the prototype endogenous form. KoRV-B, first found in a small number of koalas with an increased leukemia prevalence at one US zoo, has been associated with other cancers and increased chlamydial disease. To better understand the molecular epidemiology of KoRV variants and the effect of increased viral loads (VLs) on transmissibility and pathogenicity we developed subtype-specific quantitative PCR (qPCR) assays and tested blood and tissue samples from koalas at US zoos (n = 78), two Australian zoos (n = 27) and wild-caught (n = 21) in Australia. We analyzed PCR results with available clinical, demographic, and pedigree data. RESULTS: All koalas were KoRV-A-infected. A small number of koalas (10.3%) at one US zoo were also infected with non-A subtypes, while a higher non-A subtype prevalence (59.3%) was found in koalas at Australian zoos. Wild koalas from one location were only infected with KoRV-A. We observed a significant association of infection and plasma VLs of non-A subtypes in koalas that died of leukemia/lymphoma and other neoplasias and report cancer diagnoses in KoRV-A-positive animals. Infection and VLs of non-A subtypes was not associated with age or sex. Transmission of non-A subtypes occurred from dam-to-offspring and likely following adult-to-adult contact, but associations with contact type were not evaluated. Brief antiretroviral treatment of one leukemic koala infected with high plasma levels of KoRV-A, -B, and -F did not affect VL or disease progression. CONCLUSIONS: Our results show a significant association of non-A KoRV infection and plasma VLs with leukemia and other cancers. Although we confirm dam-to-offspring transmission of these variants, we also show other routes are possible. Our validated qPCR assays will be useful to further understand KoRV epidemiology and its zoonotic transmission potential for humans exposed to koalas because KoRV can infect human cells.


Assuntos
Gammaretrovirus/genética , Phascolarctidae/virologia , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/veterinária , Animais , Animais Selvagens , Animais de Zoológico , Austrália/epidemiologia , Feminino , Gammaretrovirus/classificação , Gammaretrovirus/isolamento & purificação , Gammaretrovirus/patogenicidade , Variação Genética , Masculino , Epidemiologia Molecular , Reação em Cadeia da Polimerase/veterinária , Prevalência , RNA Viral/genética , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/transmissão , Infecções Tumorais por Vírus/virologia , Estados Unidos/epidemiologia , Carga Viral
3.
Cell ; 179(3): 632-643.e12, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31607510

RESUMO

Antisense Piwi-interacting RNAs (piRNAs) guide silencing of established transposons during germline development, and sense piRNAs drive ping-pong amplification of the antisense pool, but how the germline responds to genome invasion is not understood. The KoRV-A gammaretrovirus infects the soma and germline and is sweeping through wild koalas by a combination of horizontal and vertical transfer, allowing direct analysis of retroviral invasion of the germline genome. Gammaretroviruses produce spliced Env mRNAs and unspliced transcripts encoding Gag, Pol, and the viral genome, but KoRV-A piRNAs are almost exclusively derived from unspliced genomic transcripts and are strongly sense-strand biased. Significantly, selective piRNA processing of unspliced proviral transcripts is conserved from insects to placental mammals. We speculate that bypassed splicing generates a conserved molecular pattern that directs proviral genomic transcripts to the piRNA biogenesis machinery and that this "innate" piRNA response suppresses transposition until antisense piRNAs are produced, establishing sequence-specific adaptive immunity.


Assuntos
Gammaretrovirus/genética , Phascolarctidae/genética , RNA Interferente Pequeno/genética , Animais , Elementos de DNA Transponíveis , Gammaretrovirus/metabolismo , Gammaretrovirus/patogenicidade , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Produtos do Gene pol/genética , Produtos do Gene pol/metabolismo , Genoma , Células Germinativas/metabolismo , Células Germinativas/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Phascolarctidae/virologia , Splicing de RNA , RNA Antissenso/genética , RNA Antissenso/metabolismo , RNA Interferente Pequeno/metabolismo
4.
Arch Virol ; 164(11): 2735-2745, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31486907

RESUMO

Koala retrovirus (KoRV) is unique among endogenous retroviruses because its endogenization is still active. Two major KoRV subtypes, KoRV-A and B, have been described, and KoRV-B is associated with disease and poses a health threat to koalas. Here, we investigated the co-prevalence of KoRV-A and KoRV-B, detected by type-specific PCR and sequencing, and their impact on the health of koalas in three Japanese zoos. We also investigated KoRV proviral loads and found varying amounts of genomic DNA (gDNA) in peripheral blood mononuclear cells (PBMCs). We found that 100% of the koalas examined were infected with KoRV-A and 60% (12/20) were coinfected with KoRV-B. The KoRV-A sequence was highly conserved, whereas the KoRV-B sequence varied among individuals. Interestingly, we observed possible vertical transmission of KoRV-B in one offspring in which the KoRV-B sequence was similar to that of the father but not the mother. Moreover, we characterized the KoRV growth patterns in concanavalin-A-stimulated PBMCs isolated from KoRV-B-coinfected or KoRV-B-uninfected koalas. We quantified the KoRV provirus in gDNA and the KoRV RNA copy numbers in cells and culture supernatants by real-time PCR at days 4, 7, and 14 post-seeding. As the study population is housed in captivity, a longitudinal study of these koalas may provide an opportunity to study the transmission mode of KoRV-B. In addition, we characterized KoRV isolates by infecting tupaia cells. The results suggested that tupaia may be used as an infection model for KoRV. Thus, this study may enhance our understanding of KoRV-B coinfection and transmission in the captive koalas.


Assuntos
Retrovirus Endógenos/genética , Gammaretrovirus/patogenicidade , Phascolarctidae/virologia , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/veterinária , Animais , Animais de Zoológico/virologia , Linhagem Celular , Coinfecção/veterinária , Coinfecção/virologia , Retrovirus Endógenos/classificação , Retrovirus Endógenos/isolamento & purificação , Feminino , Gammaretrovirus/classificação , Gammaretrovirus/genética , Gammaretrovirus/isolamento & purificação , Japão/epidemiologia , Masculino , Provírus/genética , Infecções por Retroviridae/virologia , Tupaia/virologia , Carga Viral
5.
APMIS ; 124(1-2): 20-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26818259

RESUMO

A virus first perspective is presented as an alternative hypothesis to explain the role of various endogenized retroviruses in the origin of the mammalian placenta. It is argued that virus-host persistence is a key determinant of host survival and the various ERVs involved have directly affected virus-host persistence.


Assuntos
Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Gammaretrovirus/genética , Placenta/fisiologia , Placenta/virologia , Animais , Evolução Molecular , Feminino , Gammaretrovirus/patogenicidade , Gammaretrovirus/fisiologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Gravidez
6.
Uirusu ; 66(1): 21-30, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-28484175

RESUMO

RD-114 virus is a feline endogenous retrovirus (ERV) isolated from human rhabdomyosarcoma in 1971 and classified as endogenous gammaretrovirus in domestic cats (Felis catus). Based on the previous reports in 70's, it has been considered that a horizontal, infectious event occurred to transfer the virus from ancient baboon species to ancient cat species, whereupon it became endogenous in the cat species about several million years ago in Mediterranean Basin. Although it has been believed that all domestic cats harbor infectious RD-114 provirus in their genome, we revealed that cats do not have infectious RD-114 viral loci, but infectious RD-114 virus is resurrected by recombination between uninfectious RD-114 virus-related ERVs [here we designated them as RD-114-related sequences (RDRSs)]. Further, we also revealed the RDRSs which would potentially be resurrected as RD-114 virus (here we refer to them as ''new'' RDRSs) had entered the genome of the domestic cat after domestication of the cat around 10 thousand years ago. The fractions and positions of RDRSs in the cat genome differed in Western and Eastern cat populations and cat breeds. Our study revealed that RDRS would be a useful tool for elucidating the world travel routes of domestic cats after domestication.


Assuntos
Gatos/genética , Gatos/virologia , Gammaretrovirus , Animais , Domesticação , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Genoma , Humanos , Filogenia
7.
Retrovirology ; 12: 68, 2015 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-26253512

RESUMO

BACKGROUND: Koala retrovirus (KoRV) is an endogenous and exogenous retrovirus of koalas that may cause lymphoma. As for many other gammaretroviruses, the KoRV genome can potentially encode an alternate form of Gag protein, glyco-gag. RESULTS: In this study, a convenient assay for assessing KoRV infectivity in vitro was employed: the use of DERSE cells (initially developed to search for infectious xenotropic murine leukemia-like viruses). Using infection of DERSE and other human cell lines (HEK293T), no evidence for expression of glyco-gag by KoRV was found, either in expression of glyco-gag protein or changes in infectivity when the putative glyco-gag reading frame was mutated. Since glyco-gag mediates resistance of Moloney murine leukemia virus to the restriction factor APOBEC3, the sensitivity of KoRV (wt or putatively mutant for glyco-gag) to restriction by murine (mA3) or human APOBEC3s was investigated. Both mA3 and hA3G potently inhibited KoRV infectivity. Interestingly, hA3G restriction was accompanied by extensive G → A hypermutation during reverse transcription while mA3 restriction was not. Glyco-gag status did not affect the results. CONCLUSIONS: These results indicate that the mechanisms of APOBEC3 restriction of KoRV by hA3G and mA3 differ (deamination dependent vs. independent) and glyco-gag does not play a role in the restriction.


Assuntos
Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Gammaretrovirus/fisiologia , Produtos do Gene gag/metabolismo , Phascolarctidae/virologia , Replicação Viral , Desaminases APOBEC , Sequência de Aminoácidos , Animais , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Produtos do Gene gag/química , Produtos do Gene gag/genética , Células HEK293 , Humanos , Camundongos , Fases de Leitura Aberta , Transcrição Reversa , Alinhamento de Sequência
8.
Trends Biochem Sci ; 40(2): 108-16, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555456

RESUMO

To achieve productive infection, retroviruses such as HIV stably integrate their reverse transcribed RNA genome into a host chromosome. Each retroviral family preferentially integrates near a unique subset of genomic features. HIV integrase (IN) is targeted to the body of active transcription units through interaction with lens epithelium-derived growth factor (LEDGF/p75). We describe the successful effort to develop inhibitors of the interaction between IN and LEDGF/p75, referred to as LEDGINs. Gammaretroviruses display a distinct integration pattern. Recently, BET (bromo- and extraterminal domain) proteins were identified as the LEDGF/p75 counterparts that target the integration of gammaretroviruses. The identification of the chromatin-readers LEDGF/p75 and BET as cellular cofactors that orchestrate lentiviral or gammaretroviral integration opens new avenues to developing safer viral vectors for gene therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por HIV/tratamento farmacológico , Inibidores de Integrase de HIV/administração & dosagem , Fatores de Transcrição/metabolismo , Integração Viral/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Cromatina/efeitos dos fármacos , Gammaretrovirus/efeitos dos fármacos , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Infecções por HIV/virologia , Integrase de HIV/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Lentivirus/efeitos dos fármacos , Lentivirus/genética , Lentivirus/patogenicidade , Fatores de Transcrição/genética , Integração Viral/efeitos dos fármacos
10.
J Virol ; 85(3): 1205-13, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084477

RESUMO

A novel gammaretrovirus, xenotropic murine leukemia virus-related virus (XMRV), has been identified in patients with prostate cancer and in patients with chronic fatigue syndromes. Standard Mus musculus laboratory mice lack a functional XPR1 receptor for XMRV and are therefore not a suitable model for the virus. In contrast, Gairdner's shrew-mice (Mus pahari) do express functional XPR1. To determine whether Mus pahari could serve as a model for XMRV, primary Mus pahari fibroblasts and mice were infected with cell-free XMRV. Infection of cells in vitro resulted in XMRV Gag expression and the production of XMRV virions. After intraperitoneal injection of XMRV into Mus pahari mice, XMRV proviral DNA could be detected in spleen, blood, and brain. Intravenous administration of a green fluorescent protein (GFP) vector pseudotyped with XMRV produced GFP(+) CD4(+) T cells and CD19(+) B cells. Mice mounted adaptive immune responses against XMRV, as evidenced by the production of neutralizing and Env- and Gag-specific antibodies. Prominent G-to-A hypermutations were also found in viral genomes isolated from the spleen, suggesting intracellular restriction of XMRV infection by APOBEC3 in vivo. These data demonstrate infection of Mus pahari by XMRV, potential cell tropism of the virus, and immunological and intracellular restriction of virus infection in vivo. These data support the use of Mus pahari as a model for XMRV pathogenesis and as a platform for vaccine and drug development against this potential human pathogen.


Assuntos
Modelos Animais de Doenças , Gammaretrovirus/patogenicidade , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Infecções por Retroviridae/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos CD19/análise , Linfócitos B/química , Linfócitos B/virologia , Sangue/virologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Fibroblastos/virologia , Gammaretrovirus/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/patologia , Baço/virologia , Coloração e Rotulagem/métodos , Tropismo Viral , Receptor do Retrovírus Politrópico e Xenotrópico
11.
J Virol ; 84(24): 12841-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943975

RESUMO

Laboratory mouse strains carry endogenous copies of the xenotropic mouse leukemia viruses (X-MLVs), named for their inability to infect cells of the laboratory mouse. This resistance to exogenous infection is due to a nonpermissive variant of the XPR1 gammaretrovirus receptor, a resistance that also limits in vivo expression of germ line X-MLV proviruses capable of producing infectious virus. Because laboratory mice vary widely in their proviral contents and in their virus expression patterns, we screened inbred strains for sequence and functional variants of the XPR1 receptor. We also typed inbred strains and wild mouse species for an endogenous provirus, Bxv1, that is capable of producing infectious X-MLV and that also contributes to the generation of pathogenic recombinant MLVs. We identified the active Bxv1 provirus in many common inbred strains and in some Japanese Mus molossinus mice but in none of the other wild mouse species that carry X-MLVs. Our screening for Xpr1 variants identified the permissive Xpr1(sxv) allele in 7 strains of laboratory mice, including a Bxv1-positive strain, F/St, which is characterized by lifelong X-MLV viremia. Cells from three strains carrying Xpr1(sxv), namely, SWR, SJL, and SIM.R, were shown to be infectable by X-MLV and XMRV; these strains carry different alleles at Fv1 and vary in their sensitivities to specific X/P-MLV isolates and XMRV. Several strains with Xpr1(sxv) lack the active Bxv1 provirus or other endogenous X-MLVs and may provide a useful model system to evaluate the in vivo spread of these gammaretroviruses and their disease potential in their natural host.


Assuntos
Suscetibilidade a Doenças , Gammaretrovirus/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Camundongos Endogâmicos/virologia , Provírus/genética , Viremia/genética , Animais , Fibroblastos , Humanos , Camundongos , Camundongos Endogâmicos/genética , Células NIH 3T3 , Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Receptores Virais/genética , Receptor do Retrovírus Politrópico e Xenotrópico
13.
Nat Rev Urol ; 7(7): 392-402, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20517289

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is an authentic, newly recognized human retrovirus first identified in prostate cancer tissues from men with a deficiency in the innate immunity gene RNASEL. At present, studies have detected XMRV at widely different rates in prostate cancer cases (0-27%) and in patients with chronic fatigue syndrome (CFS; 0-67%). Indirect or direct modes of carcinogenesis by XMRV have been suggested depending on whether the virus was found in stroma or malignant epithelium. Viral replication in the prostate might be affected by androgens, which stimulate XMRV through a transcriptional enhancer site in viral DNA. By contrast, host restriction factors, such as APOBEC3 and tetherin, inhibit virus replication. Immune dysfunction mediated by XMRV has been suggested as a possible factor in CFS. Recent studies show that some existing antiretroviral drugs suppress XMRV infections and diagnostic assays are under development. Although other retroviruses of the same genus as XMRV (gammaretroviruses) cause cancer and neurological disease in animals, whether XMRV is a cause of either prostate cancer or CFS remains unknown. Emerging science surrounding XMRV is contributing to our knowledge of retroviral infections while focusing intense interest on two major human diseases.


Assuntos
Síndrome de Fadiga Crônica/virologia , Neoplasias da Próstata/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Síndrome de Fadiga Crônica/imunologia , Síndrome de Fadiga Crônica/terapia , Feminino , Gammaretrovirus/efeitos dos fármacos , Gammaretrovirus/imunologia , Gammaretrovirus/patogenicidade , Humanos , Vírus da Leucemia Murina/efeitos dos fármacos , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/patogenicidade , Masculino , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Retroviridae/efeitos dos fármacos , Retroviridae/imunologia , Retroviridae/patogenicidade , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/terapia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/terapia
14.
Virology ; 399(1): 23-30, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20110097

RESUMO

Recently, the first human infection with an exogenous gammaretrovirus (XMRV) was reported. In its initial description, XMRV was confined to prostate stromal fibroblasts, although subsequent reports demonstrated XMRV protein expression in prostate epithelial cells. Most recently, XMRV has been detected in blood cells of patients with chronic fatigue syndrome. The aim of this study was to elucidate the transmission routes and tissue tropism of XMRV by comparing its host range, receptor usage and LTR functionality with other MLV isolates. We demonstrate using pseudotype experiments that XMRV Env mediates efficient infection of cells from different species. We show that replication competent XMRV infects various human cell types, including hematopoietic cell lines and prostate stromal fibroblasts. XMRV-LTR activity is significantly higher in the prostate cancer cell line LNCaP and in prostate stromal fibroblasts, compared to other cell types tested and could be one factor contributing to efficient viral spread in prostate tissue.


Assuntos
Gammaretrovirus/patogenicidade , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Gammaretrovirus/fisiologia , Humanos , Masculino , Camundongos , Filogenia , Próstata/virologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Sequências Repetidas Terminais/fisiologia , Proteínas do Envelope Viral/fisiologia , Replicação Viral/fisiologia , Receptor do Retrovírus Politrópico e Xenotrópico
15.
J Virol ; 84(5): 2556-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20015990

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a novel human gammaretrovirus discovered in association with human prostate tumors. XMRV was first identified in prostate stromal cells surrounding the tumors of patients carrying a mutation in the HPC1 gene locus. To determine the tropism of XMRV in cell culture, we tested the ability of XMRV to spread and replicate in various prostate and nonprostate cell lines. We found that although the expression of XMRV viral proteins and the spread of infectious virus were minimal in a variety of cell lines, XMRV displayed robust expression and infection in LNCaP prostate tumor cells. The transcriptional activity of the XMRV long terminal repeat (LTR) was found to be higher than the Moloney murine leukemia virus LTRs in both LNCaP and WPMY-1 (simian virus 40-transformed prostate stromal cells). The U3 promoter of XMRV and a glucocorticoid response element (GRE) within the U3 were required for the transcriptional activity in LNCaP cells. Coexpression of the androgen receptor and stimulation with dihydrotestosterone stimulated XMRV-LTR-dependent transcription in 293T cells, and the GRE was required for this activity. These data suggest that XMRV may replicate more efficiently in LNCaP cells in part due to the transcriptional environment in LNCaP cells.


Assuntos
Gammaretrovirus , Próstata , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/virologia , Transcrição Gênica , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Gammaretrovirus/patogenicidade , Genes Reporter , Humanos , Masculino , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/metabolismo , Regiões Promotoras Genéticas , Próstata/citologia , Próstata/patologia , Próstata/virologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Internalização do Vírus
16.
J Clin Invest ; 119(4): 964-75, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19307726

RESUMO

gamma-Retroviral vectors (gammaRVs), which are commonly used in gene therapy, can trigger oncogenesis by insertional mutagenesis. Here, we have dissected the contribution of vector design and viral integration site selection (ISS) to oncogenesis using an in vivo genotoxicity assay based on transplantation of vector-transduced tumor-prone mouse hematopoietic stem/progenitor cells. By swapping genetic elements between gammaRV and lentiviral vectors (LVs), we have demonstrated that transcriptionally active long terminal repeats (LTRs) are major determinants of genotoxicity even when reconstituted in LVs and that self-inactivating (SIN) LTRs enhance the safety of gammaRVs. By comparing the genotoxicity of vectors with matched active LTRs, we were able to determine that substantially greater LV integration loads are required to approach the same oncogenic risk as gammaRVs. This difference in facilitating oncogenesis is likely to be explained by the observed preferential targeting of cancer genes by gammaRVs. This integration-site bias was intrinsic to gammaRVs, as it was also observed for SIN gammaRVs that lacked genotoxicity in our model. Our findings strongly support the use of SIN viral vector platforms and show that ISS can substantially modulate genotoxicity.


Assuntos
Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Animais , Sequência de Bases , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Primers do DNA/genética , Genes p16 , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Lentivirus/genética , Lentivirus/patogenicidade , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/etiologia , Neoplasias Experimentais/genética , Sequências Repetidas Terminais
17.
J Virol ; 83(1): 336-46, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945767

RESUMO

Although transcription factors of the basic helix-loop-helix family have been shown to regulate enhancers of lymphomagenic gammaretroviruses through E-box motifs, the overlap of an E-box motif (Egre) with the glucocorticoid response element (GRE) has obscured their function in vivo. We report here that Egre, but not the GRE, affects disease induction by the murine T-lymphomagenic SL3-3 virus. Mutating all three copies of Egre prolonged the tumor latency period from 60 to 109 days. Further mutating an E-box motif (Ea/s) outside the enhancer prolonged the latency period to 180 days, suggesting that Ea/s works as a backup site for Egre. While wild-type SL3-3 and GRE and Ea/s mutants exclusively induced T-cell lymphomas with wild-type latencies mainly of the CD4(+) CD8(-) phenotype, Egre as well as the Egre and Ea/s mutants induced B-cell lymphomas and myeloid leukemia in addition to T-cell lymphomas. T-cell lymphomas induced by the two Egre mutants had the same phenotype as those induced by wild-type SL3-3, indicating the incomplete disruption of T-cell lymphomagenesis, which is in contrast to previous findings for a Runx site mutant of SL3-3. Mutating the Egre site or Egre and Ea/s triggered several tumor phenotype-associated secondary enhancer changes encompassing neighboring sites, none of which led to the regeneration of an E-box motif. Taken together, our results demonstrate a role for the E-box but not the GRE in T lymphomagenesis by SL3-3, unveil an inherent broader disease specificity of the virus, and strengthen the notion of selection for more potent enhancer variants of mutated viruses during tumor development.


Assuntos
Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Linfoma/virologia , Elementos de Resposta , Animais , DNA Viral/genética , Incidência , Camundongos , Mutação
19.
Cell Mol Life Sci ; 65(21): 3399-412, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18818871

RESUMO

Xenotransplantation is defined by the PHS as any procedure that involves the transplantation, implantation or infusion into a human recipient of either (a) live cells, tissues or organs from a nonhuman animal source, or (b) human body fluids, cells, tissues or organs that have had ex vivo contact with live nonhuman animal cells, tissues or organs (Public Health Service Guideline on Infectious Disease Issues in Xenotransplantation). Use of pigs for human xenotransplantation raises concerns about the risks of transfer of infectious agents from the pig cells to xenotransplantation recipients. The observation that the porcine germline harbors genetic loci encoding porcine endogenous retroviruses (PERVs) that are in some cases infectious for human cells has resulted in renewed scientific interest in PERVs. However, in spite of the past 10 years of investigation, the actual risk for PERV infection, replication, and pathogenic outcome in human recipients of xenotransplantation products is still undefined. (Part of a multi-author review).


Assuntos
Retrovirus Endógenos/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Suínos/virologia , Transplante Heterólogo/efeitos adversos , Animais , Linhagem Celular/virologia , Retrovirus Endógenos/classificação , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Evolução Molecular , Gammaretrovirus/classificação , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Gammaretrovirus/fisiologia , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Humanos , Modelos Animais , Receptores Virais/fisiologia , Recombinação Genética , Infecções por Retroviridae/prevenção & controle , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/veterinária , Infecções por Retroviridae/virologia , Especificidade da Espécie , Sus scrofa/genética , Sus scrofa/virologia , Suínos/genética , Suínos/imunologia , Doenças dos Suínos/transmissão , Doenças dos Suínos/virologia , Transplante Heterólogo/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Infecções Tumorais por Vírus/transmissão , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia , Virulência , Replicação Viral
20.
J Mol Biol ; 369(5): 1214-29, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17498744

RESUMO

Retroviruses are well known for their ability to incorporate envelope (Env) proteins from other retroviral strains and genera, and even from other virus families. This characteristic has been widely exploited for the generation of replication-defective retroviral vectors, including those derived from murine leukemia virus (MLV), bearing heterologous Env proteins. We investigated the possibility of "genetically pseudotyping" replication-competent MLV by replacing the native env gene in a full-length viral genome with that of another gammaretrovirus. Earlier, we developed replication-competent versions of MLV that stably transmit and express transgenes inserted into the 3' untranslated region of the viral genome. In one such tagged MLV expressing green fluorescent protein, we replaced the native env sequence with that of gibbon ape leukemia virus (GALV). Although the GALV Env protein is commonly used to make high-titer pseudotypes of MLV vectors, we found that the env replacement greatly attenuated viral replication. However, extended cultivation of cells exposed to the chimeric virus resulted in selection of mutants exhibiting rapid replication kinetics and different variants arose in different infections. Two of these variants had acquired mutations at or adjacent to the splice acceptor site, and three others had acquired dual mutations within the long terminal repeat. Analysis of the levels of unspliced and spliced viral RNA produced by the parental and adapted viruses showed that the mutations gained by each of these variants functioned to reverse an imbalance in splicing caused by the env gene substitution. Our results reveal the presence of previously unknown cis-acting sequences in MLV that modulate splicing of the viral transcript and demonstrate that tagging of the retroviral genome with an easily assayed transgene can be combined with in vitro evolution as an approach to efficiently generating and screening for replicating mutants of replication-impaired recombinant viruses.


Assuntos
Evolução Molecular , Gammaretrovirus/genética , Splicing de RNA , Sequências Reguladoras de Ácido Ribonucleico , Adaptação Biológica , Animais , Sequência de Bases , Células Cultivadas , Replicação do DNA , Gammaretrovirus/patogenicidade , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sequências Repetidas Terminais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...