Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 454
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 95(18): e0063421, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34190600

RESUMEN

The host transmembrane protein SERINC5 is incorporated into viral particles and restricts infection by certain retroviruses. However, what motif of SERINC5 mediates this process remains elusive. By conducting mutagenesis analyses, we found that the substitution of phenylalanine with alanine at position 412 (F412A) resulted in a >75-fold reduction in SERINC5's restriction function. The F412A substitution also resulted in the loss of SERINC5's function to sensitize HIV-1 neutralization by antibodies recognizing the envelope's membrane proximal region. A series of biochemical analyses revealed that F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into secreted virus particles to a greater extent than in the wild type. Furthermore, introduction of several amino acid mutations at this position revealed that the aromatic side chains, including phenylalanine, tyrosine, and tryptophan, were required to maintain SERINC5 functions to impair the virus-cell fusion process and virion infectivity. Moreover, the wild-type SERINC5 restricted infection of lentiviruses pseudotyped with envelopes of murine leukemia viruses, simian immunodeficiency virus, and HIV-2, and F412A abrogated this function. Taken together, our results highlight the importance of the aromatic side chain at SERINC5 position 412 to maintain its restriction function against diverse retrovirus envelopes. IMPORTANCE The host protein SERINC5 is incorporated into progeny virions of certain retroviruses and restricts the infectivity of these viruses or sensitizes the envelope glycoprotein to a class of neutralizing antibodies. However, how and which part of SERINC5 engages with the diverse array of retroviral envelopes and exerts its antiretroviral functions remain elusive. During mutagenesis analyses, we eventually found that the single substitution of phenylalanine with alanine, but not with tyrosine or tryptophan, at position 412 (F412A) resulted in the loss of SERINC5's functions toward diverse retroviruses, whereas F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into progeny virions to a greater extent than the wild type. Results suggest that the aromatic side chain at position 412 of SERINC5 plays a critical role in mediating antiviral functions toward various retroviruses, thus providing additional important information regarding host and retrovirus interaction.


Asunto(s)
Aminoácidos Aromáticos/genética , Membrana Celular/metabolismo , Infecciones por VIH/virología , VIH-1/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Proteínas de la Membrana/metabolismo , Mutación , Células HEK293 , Infecciones por VIH/genética , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Virus de la Leucemia Murina/genética , Proteínas de la Membrana/genética , Virulencia
2.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32321818

RESUMEN

RNA modifications play diverse roles in regulating RNA function, and viruses co-opt these pathways for their own benefit. While recent studies have highlighted the importance of N6-methyladenosine (m6A)-the most abundant mRNA modification-in regulating retrovirus replication, the identification and function of other RNA modifications in viral biology have been largely unexplored. Here, we characterized the RNA modifications present in a model retrovirus, murine leukemia virus (MLV), using mass spectrometry and sequencing. We found that 5-methylcytosine (m5C) is highly enriched in viral genomic RNA relative to uninfected cellular mRNAs, and we mapped at single-nucleotide resolution the m5C sites, which are located in multiple clusters throughout the MLV genome. Further, we showed that the m5C reader protein ALYREF plays an important role in regulating MLV replication. Together, our results provide a complete m5C profile in a virus and its function in a eukaryotic mRNA.IMPORTANCE Over 130 modifications have been identified in cellular RNAs, which play critical roles in many cellular processes, from modulating RNA stability to altering translation efficiency. One such modification, 5-methylcytosine, is relatively abundant in mammalian mRNAs, but its precise location and function are not well understood. In this study, we identified unexpectedly high levels of m5C in the murine leukemia virus RNA, precisely mapped its location, and showed that ALYREF, a reader protein that specifically recognizes m5C, regulates viral production. Together, our findings provide a high-resolution atlas of m5C in murine leukemia virus and reveal a functional role of m5C in viral replication.


Asunto(s)
5-Metilcitosina/metabolismo , Virus de la Leucemia Murina/genética , 5-Metilcitosina/fisiología , Animales , Metilación de ADN/genética , Genoma Viral/genética , Células HEK293 , Humanos , Virus de la Leucemia Murina/metabolismo , Virus de la Leucemia Murina/patogenicidad , Metiltransferasas/metabolismo , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Retroviridae/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética , Replicación Viral/genética
3.
J Virol ; 91(21)2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794032

RESUMEN

Ecotropic, xenotropic, and polytropic mouse leukemia viruses (E-, X-, and P-MLVs) exist in mice as infectious viruses and endogenous retroviruses (ERVs) inserted into mouse chromosomes. All three MLV subgroups are linked to leukemogenesis, which involves generation of recombinants with polytropic host range. Although P-MLVs are deemed to be the proximal agents of disease induction, few biologically characterized infectious P-MLVs have been sequenced for comparative analysis. We analyzed the complete genomes of 16 naturally occurring infectious P-MLVs, 12 of which were typed for pathogenic potential. We sought to identify ERV progenitors, recombinational hot spots, and segments that are always replaced, never replaced, or linked to pathogenesis or host range. Each P-MLV has an E-MLV backbone with P- or X-ERV replacements that together cover 100% of the recombinant genomes, with different substitution patterns for X- and P-ERVs. Two segments are always replaced, both coding for envelope (Env) protein segments: the N terminus of the surface subunit and the cytoplasmic tail R peptide. Viral gag gene replacements are influenced by host restriction genes Fv1 and Apobec3 Pathogenic potential maps to the env transmembrane subunit segment encoding the N-heptad repeat (HR1). Molecular dynamics simulations identified three novel interdomain salt bridges in the lymphomagenic virus HR1 that could affect structural stability, entry or sensitivity to host immune responses. The long terminal repeats of lymphomagenic P-MLVs are differentially altered by recombinations, duplications, or mutations. This analysis of the naturally occurring, sometimes pathogenic P-MLV recombinants defines the limits and extent of intersubgroup recombination and identifies specific sequence changes linked to pathogenesis and host interactions.IMPORTANCE During virus-induced leukemogenesis, ecotropic mouse leukemia viruses (MLVs) recombine with nonecotropic endogenous retroviruses (ERVs) to produce polytropic MLVs (P-MLVs). Analysis of 16 P-MLV genomes identified two segments consistently replaced: one at the envelope N terminus that alters receptor choice and one in the R peptide at the envelope C terminus, which is removed during virus assembly. Genome-wide analysis shows that nonecotropic replacements in the progenitor ecotropic MLV genome are more extensive than previously appreciated, covering 100% of the genome; contributions from xenotropic and polytropic ERVs differentially alter the regions responsible for receptor determination or subject to APOBEC3 and Fv1 restriction. All pathogenic viruses had modifications in the regulatory elements in their long terminal repeats and differed in a helical segment of envelope involved in entry and targeted by the host immune system. Virus-induced leukemogenesis thus involves generation of complex recombinants, and specific replacements are linked to pathogenesis and host restrictions.


Asunto(s)
Especificidad del Huésped/genética , Virus de la Leucemia Murina/clasificación , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/virología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Molecular , Genoma Viral , Virus de la Leucemia Murina/genética , Ratones , Simulación de Dinámica Molecular , Conformación Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , Homología de Secuencia , Secuencias Repetidas Terminales , Proteínas Virales/química , Proteínas Virales/metabolismo
4.
J Virol ; 90(7): 3385-99, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764005

RESUMEN

UNLABELLED: Certain murine leukemia viruses (MLVs) are capable of inducing fatal progressive spongiform motor neuron disease in mice that is largely mediated by viral Env glycoprotein expression within central nervous system (CNS) glia. While the etiologic mechanisms and the glial subtypes involved remain unresolved, infection of NG2 glia was recently observed to correlate spatially and temporally with altered neuronal physiology and spongiogenesis. Since one role of NG2 cells is to serve as oligodendrocyte (OL) progenitor cells (OPCs), we examined here whether their infection by neurovirulent (FrCasE) or nonneurovirulent (Fr57E) ecotropic MLVs influenced their viability and/or differentiation. Here, we demonstrate that OPCs, but not OLs, are major CNS targets of both FrCasE and Fr57E. We also show that MLV infection of neural progenitor cells (NPCs) in culture did not affect survival, proliferation, or OPC progenitor marker expression but suppressed certain glial differentiation markers. Assessment of glial differentiation in vivo using transplanted transgenic NPCs showed that, while MLVs did not affect cellular engraftment or survival, they did inhibit OL differentiation, irrespective of MLV neurovirulence. In addition, in chimeric brains, where FrCasE-infected NPC transplants caused neurodegeneration, the transplanted NPCs proliferated. These results suggest that MLV infection is not directly cytotoxic to OPCs but rather acts to interfere with OL differentiation. Since both FrCasE and Fr57E viruses restrict OL differentiation but only FrCasE induces overt neurodegeneration, restriction of OL maturation alone cannot account for neuropathogenesis. Instead neurodegeneration may involve a two-hit scenario where interference with OPC differentiation combined with glial Env-induced neuronal hyperexcitability precipitates disease. IMPORTANCE: A variety of human and animal retroviruses are capable of causing central nervous system (CNS) neurodegeneration manifested as motor and cognitive deficits. These retroviruses infect a variety of CNS cell types; however, the specific role each cell type plays in neuropathogenesis remains to be established. The NG2 glia, whose CNS functions are only now emerging, are a newly appreciated viral target in murine leukemia virus (MLV)-induced neurodegeneration. Since one role of NG2 glia is that of oligodendrocyte progenitor cells (OPCs), we investigated here whether their infection by the neurovirulent MLV FrCasE contributed to neurodegeneration by affecting OPC viability and/or development. Our results show that both neurovirulent and nonneurovirulent MLVs interfere with oligodendrocyte differentiation. Thus, NG2 glial infection could contribute to neurodegeneration by preventing myelin formation and/or repair and by suspending OPCs in a state of persistent susceptibility to excitotoxic insult mediated by neurovirulent virus effects on other glial subtypes.


Asunto(s)
Virus de la Leucemia Murina/patogenicidad , Enfermedad de la Neurona Motora/virología , Células-Madre Neurales/virología , Neurogénesis/fisiología , Neuroglía/virología , Infecciones por Retroviridae/complicaciones , Células 3T3 , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Femenino , Productos del Gen env/biosíntesis , Masculino , Ratones , Ratones Transgénicos , Oligodendroglía/citología , Oligodendroglía/virología
5.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226373

RESUMEN

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Asunto(s)
Glutatión/deficiencia , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Murino/etiología , Infecciones por Retroviridae/complicaciones , Células Th2/inmunología , Infecciones Tumorales por Virus/complicaciones , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Activación de Linfocitos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/patología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/virología , Células Th2/metabolismo , Células Th2/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
6.
J Virol ; 89(1): 155-64, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320301

RESUMEN

UNLABELLED: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine that is secreted in response to inflammasome activation by innate microbe-sensing pathways. Although some retroviruses can trigger IL-1ß secretion through the DNA-sensing molecule IFI16, the effect of IL-1ß on the course of infection is unknown. To test whether IL-1ß secretion affects retroviral replication in vivo, I constructed a novel murine leukemia virus strain (FMLV-IL-1ß) that encodes the mature form of IL-1ß. This virus replicated with kinetics similar to that of wild-type virus in tissue culture but caused a dramatically more aggressive infection of both C57BL/6 and BALB/c mice. By 7 days postinfection (PI), mice infected with FMLV-IL-1ß exhibited splenomegaly and viral loads 300-fold higher than those in mice infected with wild-type FMLV. Furthermore, the enlarged spleens of FMLV-IL-1ß-infected mice correlated with a large expansion of Gr-1(+) CD11b(+) myeloid-derived suppressor cells, as well as elevated levels of immune activation. Although FMLV-IL-1ß infection was controlled by C57BL/6 mice by 14 days p.i., FMLV-IL-1ß was able to establish a significant persistent infection and immune activation in BALB/c mice. These results demonstrate that IL-1ß secretion is a powerful positive regulator of retroviral infection and that FMLV-IL-1ß represents a new model of proinflammatory retroviral infection. IMPORTANCE: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine released in response to activation of innate pathogen-sensing pathways during microbial infection. To examine the potential impact of IL-1ß on retroviral replication in vivo, I constructed a novel mouse retrovirus strain (FMLV-IL-1ß) that encodes IL-1ß and promotes abundant IL-1ß secretion from infected cells. This virus replicates with normal kinetics in cultured cells but displays a dramatically enhanced ability to replicate in mice and caused persistent infection and immune activation in the BALB/c strain of mice. These results establish IL-1ß as a positive regulator of retroviral replication and suggest that targeting this pathway may have therapeutic benefits in infections with proinflammatory retroviruses. This virus can also be used to further study the impact of inflammatory pathways on retroviral infection.


Asunto(s)
Interleucina-1beta/metabolismo , Virus de la Leucemia Murina/inmunología , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/patología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología , Animales , Interacciones Huésped-Patógeno , Interleucina-1beta/genética , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/fisiología , Leucemia Experimental/virología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecciones por Retroviridae/virología , Esplenomegalia/patología , Infecciones Tumorales por Virus/virología , Carga Viral , Virulencia , Replicación Viral
7.
Biologicals ; 43(4): 256-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25997567

RESUMEN

Infectivity and reverse transcriptase quantitative real-time polymerase chain reaction (qRT-PCR) assays have been optimized and validated for xenotropic murine leukemia virus (X-MuLV) detection. We have evaluated the assays systematically with regard to specificity, linearity, lower limit of detection (LLOD), lower limit of quantification (LLOQ), and precision. Both assays are specific for X-MuLV detection, with a linear detection range of 0.6-5.6 log(10) TCID(50)/mL for the infectivity assay, and 1.4-6.5 log(10) particles/mL for the qRT-PCR assay. The LLOD and LLOQ of the infectivity and the qRT-PCR assays are determined as 0.5 and 1.0 log(10)/mL, and 1.4 and 2.2 log(10)/mL. The inter-assay repeatability of qRT-PCR assay (4.2% coefficient of variation [CV]) is higher than the infectivity assay (7.9% CV). We have shown that both assays are closely correlated (r = 0.85, P < 0.05, n = 22). The particle/infectivity ratio is determined as 66. Both assays were applied to evaluate virus removal using virus clearance samples of chromatographic and filtration processes. Here, we have demonstrated that the qRT-PCR assay is much faster in testing and is approximately 8-fold more sensitive than the infectivity assay. Therefore, the qRT-PCR assay can replace the infectivity assay in many cases, but both assays are complementary in elucidating the mechanism of virus inactivation and removal in virus clearance validation.


Asunto(s)
Virus de la Leucemia Murina/patogenicidad , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Virulencia , Animales , Gatos , Línea Celular , Virus de la Leucemia Murina/aislamiento & purificación , Límite de Detección , Reproducibilidad de los Resultados
8.
J Virol ; 87(15): 8372-87, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23698308

RESUMEN

The envelope protein (Env) from the CasBrE murine leukemia virus (MLV) can cause acute spongiform neurodegeneration analogous to that induced by prions. Upon central nervous system (CNS) infection, Env is expressed as multiple isoforms owing to differential asparagine (N)-linked glycosylation. Because N-glycosylation can affect protein folding, stability, and quality control, we explored whether unique CasBrE Env glycosylation features could influence neurovirulence. CasBrE Env possesses 6/8 consensus MLV glycosylation sites (gs) but is missing gs3 and gs5 and contains a putative site (gs*). Twenty-nine mutants were generated by modifying these three sites, individually or in combination, to mimic the amino acid sequence in the nonneurovirulent Friend 57 MLV. Three basic viral phenotypes were observed: replication defective (dead; titer < 1 focus-forming unit [FFU]/ml), replication compromised (RC) (titer = 10(2) to 10(5) FFU/ml); and wild-type-like (WTL) (titer > 10(5) FFU/ml). Env protein was undetectable in dead mutants, while RC and WTL mutants showed variations in Env expression, processing, virus incorporation, virus entry, and virus spread. The newly introduced gs3 and gs5 sites were glycosylated, whereas gs* was not. Six WTL mutants tested in mice showed no clear attenuation in disease onset or severity versus controls. Furthermore, three RC viruses tested by neural stem cell (NSC)-mediated brainstem dissemination also induced acute spongiosis. Thus, while unique N-glycosylation affected structural features of Env involved in protein stability, proteolytic processing, and virus assembly and entry, these changes had minimal impact on CasBrE Env neurotoxicity. These findings suggest that the Env protein domains responsible for spongiogenesis represent highly stable elements upon which the more variable viral functional domains have evolved.


Asunto(s)
Productos del Gen env/metabolismo , Virus de la Leucemia Murina/fisiología , Virus de la Leucemia Murina/patogenicidad , Procesamiento Proteico-Postraduccional , Animales , Enfermedad de Canavan/patología , Enfermedad de Canavan/virología , Análisis Mutacional de ADN , Productos del Gen env/genética , Glicosilación , Virus de la Leucemia Murina/genética , Ratones , Virulencia , Replicación Viral
9.
J Virol ; 87(5): 2496-507, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23255787

RESUMEN

Poly(ADP-ribose) polymerase 1 (PARP-1) is a cellular enzyme with a fundamental role in DNA repair and the regulation of chromatin structure, processes involved in the cellular response to retroviral DNA integration. However, the function of PARP-1 in retroviral DNA integration is controversial, probably due to the functional redundancy of the PARP family in mammalian cells. We evaluated the function of PARP-1 in retroviral infection using the chicken B lymphoblastoid cell line DT40. These cells lack significant PARP-1 functional redundancy and efficiently support the postentry early events of the mammalian-retrovirus replication cycle. We observed that DT40 PARP-1(-/-) cells were 9- and 6-fold more susceptible to infection by human immunodeficiency virus type 1 (HIV-1)- and murine leukemia virus (MLV)-derived viral vectors, respectively, than cells expressing PARP-1. Production of avian Rous-associated virus type 1 was also impaired by PARP-1. However, the susceptibilities of these cell lines to infection by the nonretrovirus vesicular stomatitis virus were indistinguishable. Real-time PCR analysis of the HIV-1 life cycle demonstrated that PARP-1 did not impair reverse transcription, nuclear import of the preintegration complex, or viral DNA integration, suggesting that PARP-1 regulates a postintegration step. In support of this hypothesis, pharmacological inhibition of the epigenetic mechanism of transcriptional silencing increased retroviral expression in PARP-1-expressing cells, suppressing the differences observed. Further analysis of the implicated molecular mechanism indicated that PARP-1-mediated retroviral silencing requires the C-terminal region, but not the enzymatic activity, of the protein. In sum, our data indicate a novel role of PARP-1 in the transcriptional repression of integrated retroviruses.


Asunto(s)
Poli(ADP-Ribosa) Polimerasas/metabolismo , Retroviridae/genética , Transcripción Genética , Replicación Viral/genética , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/patogenicidad , Virus de la Leucosis Aviar/fisiología , Línea Celular , Pollos , Regulación Viral de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , VIH-1/genética , VIH-1/patogenicidad , VIH-1/fisiología , Humanos , Células Jurkat , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Virus de la Leucemia Murina/fisiología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Retroviridae/patogenicidad , Retroviridae/fisiología , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Virus de la Estomatitis Vesicular Indiana/fisiología , Integración Viral/genética
10.
Transgenic Res ; 23(3): 539-56, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24474164

RESUMEN

Draft genome sequences for the human schistosomes, Schistosoma japonicum, S. mansoni and S. haematobium are now available. The schistosome genome contains ~11,000 protein encoding genes for which the functions of few are well understood. Nonetheless, the newly described gene products and novel non-coding RNAs represent potential intervention targets, and molecular tools are being developed to determine their importance. Over the past decade, noteworthy advances has been reported towards development of tools for gene manipulation of schistosomes, including gene expression perturbation by RNAi, and transient and stable transfection including transgenesis mediated by genome integration competent vectors. Retrovirus-mediated transgenesis is an established functional genomic approach for model species. It offers the means to establish gain- or loss-of-function phenotypes, supports vector-based RNA interference, and represents a powerful forward genetics tool for insertional mutagenesis. Murine leukemia virus (MLV) pseudotyped with vesicular stomatitis virus glycoprotein mediates somatic transgenesis in S. mansoni, and vertical transmission of integrated transgenes in S. mansoni has been demonstrated, leading the establishment of transgenic lines. In addition, MLV transgenes encoding antibiotic resistance allow the selection of MLV-transduced parasites with the appropriate antibiotics. Here we describe detailed methods to produce and quantify pseudotyped MLV particles for use in transducing developmental stages of schistosomes. Approaches to analyze MLV-transduced schistosomes, including qPCR and high throughput approaches to verify and map genome integration of transgenes are also presented. We anticipate these tools should find utility in genetic investigations in other laboratories and for other helminth pathogens of important neglected tropical diseases.


Asunto(s)
Técnicas de Transferencia de Gen , Genoma , Virus de la Leucemia Murina/genética , Schistosoma mansoni/genética , Animales , Células Germinativas/virología , Humanos , Virus de la Leucemia Murina/patogenicidad , Ratones , Ratones Transgénicos/genética , Mutagénesis Insercional , Transducción Genética
11.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22821964

RESUMEN

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Asunto(s)
Inmunidad Adaptativa , Regulación hacia Abajo/inmunología , Interferón gamma/fisiología , Virus de la Leucemia Murina/inmunología , Infecciones por Retroviridae/inmunología , Inmunidad Adaptativa/genética , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Virus de la Leucemia Murina de Friend/patogenicidad , Interferón gamma/deficiencia , Interferón gamma/genética , Virus Elevador de Lactato Deshidrogenasa/inmunología , Virus Elevador de Lactato Deshidrogenasa/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/genética , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Ratones , Ratones Congénicos , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/patología , Virus Formadores de Foco en el Bazo/inmunología , Virus Formadores de Foco en el Bazo/patogenicidad , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
12.
Blood ; 117(6): 1899-910, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21135260

RESUMEN

The Graffi murine leukemia virus induces a large spectrum of leukemias in mice and thus provides a good model to compare the transcriptome of all types of leukemias. We analyzed the gene expression profiles of both T and B leukemias induced by the virus with DNA microarrays. Given that we considered that a 4-fold change in expression level was significant, 388 probe sets were associated to B, to T, or common to both leukemias. Several of them were not yet associated with lymphoid leukemia. We confirmed specific deregulation of Fmn2, Arntl2, Bfsp2, Gfra2, Gpm6a, and Gpm6b in B leukemia, of Nln, Fbln1, and Bmp7 in T leukemias, and of Etv5 in both leukemias. More importantly, we show that the mouse Fmn2 induced an anchorage-independent growth, a drastic modification in cell shape with a concomitant disruption of the actin cytoskeleton. Interestingly, we found that human FMN2 is overexpressed in approximately 95% of pre-B acute lymphoblastic leukemia with the highest expression levels in patients with a TEL/AML1 rearrangement. These results, surely related to the role of FMN2 in meiotic spindle maintenance, suggest its important role in leukemogenesis. Finally, we propose a new panel of genes potentially involved in T and/or B leukemias.


Asunto(s)
Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/genética , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Oncogenes , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Adulto , Animales , Biomarcadores de Tumor/genética , Niño , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Forminas , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Leucemia Experimental/metabolismo , Leucemia de Células T/genética , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Infecciones por Retroviridae/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Tumorales por Virus/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(20): 9364-9, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20439730

RESUMEN

Optimal infectivity of HIV-1 virions requires synthesis of the HIV-1 regulatory protein Nef in some producer cells but not others. A survey of 18 lymphoid cell lines found that Nef was dispensable in three, each of which harbored gammaretroviruses. Nef-dependent cell lines were rendered Nef-independent by a cell-free supernatant from the independent lines or by transfection of cloned murine leukemia virus (MLV). Analysis of MLV deletion mutations identified glycosylated gag (glycogag) as the factor that rescues Nef-defective HIV-1 virions. Glycogag was also demonstrated to be required for the infectivity of MLV virions produced in lymphoid cells. Direct comparison of Nef and glycogag revealed identical dependence for activity on Env-pseudotype and producer cell type. The two proteins colocalize within cells, and both increase the yield of viral cDNA in target cells. The functional similarity of Nef and glycogag is a compelling example of convergent evolution in which two structurally unrelated proteins provide a function necessary for virion infectivity in lymphoid cells.


Asunto(s)
Productos del Gen gag/metabolismo , VIH-1/patogenicidad , Virus de la Leucemia Murina/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/deficiencia , Western Blotting , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida , Evolución Molecular , Glicosilación , VIH-1/metabolismo , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Retrovirology ; 9: 50, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22691411

RESUMEN

BACKGROUND: APOBEC3 proteins are host factors that restrict infection by retroviruses like HIV, MMTV, and MLV and are variably expressed in hematopoietic and non-hematopoietic cells, such as macrophages, lymphocytes, dendritic, and epithelia cells. Previously, we showed that APOBEC3 expressed in mammary epithelia cells function to limit milk-borne transmission of the beta-retrovirus, mouse mammary tumor virus. In this present study, we used APOBEC3 knockout mice and their wild type counterpart to query the role of APOBEC3 in sexual transmission of LP-BM5 MLV - the etiological agent of murine AIDs (mAIDs). RESULTS: We show that mouse APOBEC3 is expressed in murine genital tract tissues and gametes and that genital tract tissue of APOBEC3-deficient mice are more susceptible to infection by LP-BM5 virus. APOBEC3 expressed in genital tract tissues most likely plays a role in decreasing virus transmission via the sexual route, since mice deficient in APOBEC3 gene have higher genitalia and seminal plasma virus load and sexually transmit the virus more efficiently to their partners compared to APOBEC3+ mice. Moreover, we show that female mice sexually infected with LP-BM5 virus transmit the virus to their off-spring in APOBEC3-dependent manner. CONCLUSION: Our data indicate that genital tissue intrinsic APOBEC3 restricts genital tract infection and limits sexual transmission of LP-BM5 virus.


Asunto(s)
Citidina Desaminasa/metabolismo , Genitales/virología , Virus de la Leucemia Murina/patogenicidad , Síndrome de Inmunodeficiencia Adquirida del Murino/transmisión , Enfermedades Virales de Transmisión Sexual/transmisión , Animales , Citidina Desaminasa/genética , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/virología , Femenino , Genitales/metabolismo , Células Germinativas/metabolismo , Células Germinativas/virología , Virus de la Leucemia Murina/metabolismo , Masculino , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Enfermedades Virales de Transmisión Sexual/metabolismo , Esplenomegalia/virología , Carga Viral
15.
J Virol ; 85(7): 3498-506, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21270153

RESUMEN

BHK cells remain resistant to xenotropic murine retrovirus-related virus (XMRV) or gibbon ape leukemia virus (GALV) infection, even when their respective receptors, Xpr1 or PiT1, are expressed. We set out to determine the stage at which viral infection is blocked and whether this block is mediated by a dominant-negative factor or the absence of a requisite ancillary factor. BHK cells bind neither XMRV nor GALV envelope proteins. BHK cells expressing the appropriate receptors bind XMRV or GALV envelope proteins. BHK cells can be infected by NZB-XMV(New Zealand Black mouse xenotropic murine virus)-enveloped vectors, expressing an envelope derived from a xenotropic retrovirus that, like XMRV, employs Xpr1 as a receptor, and also by vectors bearing the envelope of 10A1 murine leukemia virus (MLV), a murine retrovirus that can use PiT1 as a receptor. The retroviral vectors used in these analyses differ solely in their viral envelope proteins, suggesting that the block to XMRV and GALV infection is mediated at the level of envelope-receptor interactions. N-linked glycosylation of the receptors was not found to mediate resistance of receptor-expressing BHK cells to GALV or XMRV, as shown by tunicamycin treatment and mutation of the specific glycosylation site of the PiT1 receptor. Hybrid cells produced by fusing BHKXpr1 or BHKPiT1 to XMRV- or GALV-resistant cells, respectively, can mediate efficient XMRV or GALV infection. These findings indicate that BHK cells lack a factor that is required for infection by primate xenotropic viruses. This factor is not required for viruses that use the same receptors but were directly isolated from mice.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Leucemia del Gibón/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Receptores Virales/metabolismo , Acoplamiento Viral , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/patogenicidad , Animales , Línea Celular , Cricetinae , Ratones , Receptores Virales/genética , Receptor de Retrovirus Xenotrópico y Politrópico
16.
J Virol ; 85(24): 13322-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21957284

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) Gag is the main structural protein driving assembly and release of virions from infected cells. Gag alone is capable of self-assembly in vitro, but host factors have been shown to play a role in efficient viral replication and particle morphogenesis within the living cell. In a series of affinity purification experiments, we identified the cellular protein Lyric to be an HIV-1 Gag-interacting protein. Lyric was previously described to be an HIV-inducible gene and is involved in various signaling pathways. Gag interacts with endogenous Lyric via its matrix (MA) and nucleocapsid (NC) domains. This interaction requires Gag multimerization and Lyric amino acids 101 to 289. Endogenous Lyric is incorporated into HIV-1 virions and is cleaved by the viral protease. Gag-Lyric interaction was also observed for murine leukemia virus and equine infectious anemia virus, suggesting that it represents a conserved feature among retroviruses. Expression of the Gag binding domain of Lyric increased Gag expression levels and viral infectivity, whereas expression of a Lyric mutant lacking the Gag binding site resulted in lower Gag expression and decreased viral infectivity. The results of the current study identify Lyric to be a cellular interaction partner of HIV-1 Gag and hint at a potential role in regulating infectivity. Further experiments are needed to elucidate the precise role of this interaction.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Mapeo de Interacción de Proteínas , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Virus de la Anemia Infecciosa Equina/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Proteínas de la Membrana , Unión Proteica , Proteínas de Unión al ARN
17.
J Virol ; 85(5): 2060-78, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191010

RESUMEN

Certain murine leukemia viruses (MLVs) can induce progressive noninflammatory spongiform neurodegeneration similar to that caused by prions. The primary MLV determinants responsible have been mapped to within the env gene; however, it has remained unclear how env mediates disease, whether non-Env viral components are required, and what central nervous system (CNS) cells constitute the critical CNS targets. To address these questions, we examined the effect of transplanting engraftable C17.2 neural stem cells engineered to pseudotype, disseminate, and trans-complement neurovirulent (CasBrE, CasE, and CasES) or non-neurovirulent (Friend and SFF-FE) env sequences (SU or SU/TM) within the CNS using either the "non-neurovirulent" amphotropic helper virus, 4070A, or pgag-polgpt (a nonpackaged vector encoding Gag-Pol). These studies revealed that acute MLV-induced spongiosis results from two separable activities of Env. First, Env causes neuropathology through unique viral targeting within the CNS, which was efficiently mediated by ecotropic Envs (CasBrE and Friend), but not 4070A amphotropic Env. Second, Env induces spongiosis through a toxin activity that is MLV-receptor independent and does not require the coexpression of other viral structural proteins. CasBrE and 4070A Envs possess the toxin activity, whereas Friend Env does not. Although the identity of the critical viral target cell(s) remains unresolved, our results appear to exclude microglia and oligodendrocyte lineage cells, while implicating viral entry into susceptible neurons. Thus, MLV-induced disease parallels prionopathies in that a single protein, Env, mediates both the CNS targeting and the toxicity of the infectious agent that manifests itself as progressive vacuolar neurodegeneration.


Asunto(s)
Virus de la Leucemia Murina/metabolismo , Enfermedades Neurodegenerativas/virología , Infecciones por Retroviridae/virología , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/metabolismo , Humanos , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Ratones , Degeneración Nerviosa , Células-Madre Neurales/patología , Células-Madre Neurales/virología , Enfermedades Neurodegenerativas/patología , Infecciones por Retroviridae/patología , Proteínas del Envoltorio Viral/genética , Virulencia
18.
PLoS Biol ; 7(7): e1000163, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19636361

RESUMEN

We have investigated the underlying mechanism by which direct cell-cell contact enhances the efficiency of cell-to-cell transmission of retroviruses. Applying 4D imaging to a model retrovirus, the murine leukemia virus, we directly monitor and quantify sequential assembly, release, and transmission events for individual viral particles as they happen in living cells. We demonstrate that de novo assembly is highly polarized towards zones of cell-cell contact. Viruses assembled approximately 10-fold more frequently at zones of cell contact with no change in assembly kinetics. Gag proteins were drawn to adhesive zones formed by viral Env glycoprotein and its cognate receptor to promote virus assembly at cell-cell contact. This process was dependent on the cytoplasmic tail of viral Env. Env lacking the cytoplasmic tail while still allowing for contact formation, failed to direct virus assembly towards contact sites. Our data describe a novel role for the viral Env glycoprotein in establishing cell-cell adhesion and polarization of assembly prior to becoming a fusion protein to allow virus entry into cells.


Asunto(s)
Virus de la Leucemia Murina/metabolismo , Animales , Células COS , Adhesión Celular , Células Cultivadas , Chlorocebus aethiops , Citoplasma/metabolismo , Productos del Gen gag/metabolismo , Humanos , Cinética , Virus de la Leucemia Murina/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral
19.
Rev Med Virol ; 21(4): 227-39, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21560181

RESUMEN

Viruses usually infect a restricted set of host species, and only in rare cases does productive infection occur outside the natural host range. Infection of a new host species can manifest as a distinct disease. In this respect, the use of non-human viruses in clinical therapy may be a cause for concern. It could provide the opportunity for the viruses to adapt to the new host and be transferred to the recipient's relatives or medical caretakers, or even to the normal host species. Such environmental impact is evidently undesirable. To forecast future clinical use of non-human viruses, a literature study was performed to identify the viruses that are being considered for application as therapeutic agents for use in humans. Twenty-seven non-human virus species were identified that are in (pre)clinical development, mainly as oncolytic agents. For risk management, it is essential that the potential environmental consequences are assessed before initiating clinical use, even if the virus is not formally classified as a genetically modified organism. To aid such assessment, each of these viruses was classified in one of five relative environmental risk categories, ranging from "Negligible" to "Very High". Canary pox virus and the Autographa californica baculovirus were assigned a "Negligible" classification, and Seneca Valley virus, murine leukemia virus, and Maraba virus to the "High" category. A complicating factor in the classification is the scarcity of publicly available information on key aspects of virus biology in some species. In such cases the relative environmental risk score was increased as a precaution.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/patogenicidad , Animales , Ensayos Clínicos como Asunto , Humanos , Virus de la Leucemia Murina/patogenicidad , Picornaviridae/patogenicidad , Rhabdoviridae/patogenicidad , Virosis/transmisión
20.
Cell Rep ; 38(3): 110279, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045303

RESUMEN

Murine leukemia virus (MLV)-presenting cells form stable intercellular contacts with target cells during infection of lymphoid tissue, indicating a role of cell-cell contacts in retrovirus dissemination. Whether host cell adhesion proteins are required for retrovirus spread in vivo remains unknown. Here, we demonstrate that the lymphocyte-function-associated-antigen-1 (LFA1) and its ligand intercellular-adhesion-molecule-1 (ICAM1) are important for cell-contact-dependent transmission of MLV between leukocytes. Infection experiments in LFA1- and ICAM1-deficient mice demonstrate a defect in MLV spread within lymph nodes. Co-culture of primary leukocytes reveals a specific requirement for ICAM1 on donor cells and LFA1 on target cells for cell-contact-dependent spread through trans- and cis-infection. Importantly, adoptive transfer experiments combined with a newly established MLV-fusion assay confirm that the directed LFA1-ICAM1 interaction is important for retrovirus fusion and transmission in vivo. Taken together, our data provide insights on how retroviruses exploit host proteins and the biology of cell-cell interactions for dissemination.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/virología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Infecciones por Retroviridae/virología , Animales , Interacciones Huésped-Patógeno/fisiología , Linfocitos/virología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/transmisión , Infecciones Tumorales por Virus/transmisión , Infecciones Tumorales por Virus/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA