Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Neurovirol ; 26(3): 371-381, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144727

RESUMEN

Zika virus (ZIKV) is an emerging virus belonging to the genus Flavivirus. ZIKV infection is a significant health concern, with increasing numbers of reports of microcephaly cases in fetuses and Guillain-Barré syndrome (GBS) in adults. Interestingly, chemosensory disturbances are also reported as one of the manifestations of GBS. ZIKV infects several human tissues and cell types in vitro and in vivo. However, there is no study demonstrating ZIKV infection and replication in chemosensory cells, including olfactory and taste cells. Taste papilla and olfactory cells are chemosensory receptor cells with unique histological, molecular, and physiological characteristics. Here we examined ZIKV infection (PRVABC59) in cultured human olfactory epithelial cells (hOECs) and fungiform taste papilla (HBO) cells in vitro, as well as in vivo mouse taste and olfactory epithelial and olfactory bulb tissues. Interestingly, while HBO cells showed resistance to ZIKV replication, hOECs were highly susceptible for ZIKV infection and replication. Further, we demonstrated the presence of ZIKV particles and expression of viral proteins in olfactory epithelium, as well as in olfactory bulb, but not in taste papillae, of immunocompromised mice (ifnar/-) infected with the PRVABC59 strain of ZIKV. These observations suggest that chemosensory cells in the olfactory neuroepithelium and olfactory bulb may be important tissues for ZIKV replication and dissemination.


Asunto(s)
Células Quimiorreceptoras/virología , Receptor de Interferón alfa y beta/inmunología , Replicación Viral/fisiología , Infección por el Virus Zika/virología , Virus Zika/patogenicidad , Animales , Línea Celular , Células Quimiorreceptoras/inmunología , Células Quimiorreceptoras/patología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Olfato/fisiología , Gusto/fisiología , Virus Zika/crecimiento & desarrollo , Virus Zika/inmunología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/patología
2.
Mol Ther ; 27(12): 2067-2079, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31653397

RESUMEN

Zika virus (ZIKV) infection is associated with microcephaly in neonates and Guillain-Barré syndrome in adults. ZIKV produces a class of nonstructural (NS) regulatory proteins that play a critical role in viral transcription and replication, including NS5, which possesses RNA-dependent RNA polymerase (RdRp) activity. Here we demonstrate that rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV-1 infection, inhibits the enzymatic activity of NS5 and suppresses ZIKV infection and replication in primary human astrocytes. Similarly, other members of the NNRTI family, including etravirine and efavirenz, showed inhibitory effects on viral infection of brain cells. Site-directed mutagenesis identified 14 amino acid residues within the NS5 RdRp domain (AA265-903), which are important for the RPV interaction and the inhibition of NS5 polymerase activity. Administration of RPV to ZIKV-infected interferon-alpha/beta receptor (IFN-A/R) knockout mice improved the clinical outcome and prevented ZIKV-induced mortality. Histopathological examination of the brains from infected animals revealed that RPV reduced ZIKV RNA levels in the hippocampus, frontal cortex, thalamus, and cerebellum. Repurposing of NNRTIs, such as RPV, for the inhibition of ZIKV replication offers a possible therapeutic strategy for the prevention and treatment of ZIKV-associated disease.


Asunto(s)
Fármacos Anti-VIH/farmacología , Encéfalo/efectos de los fármacos , Receptor de Interferón alfa y beta/fisiología , Rilpivirina/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Animales , Encéfalo/virología , Humanos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Conformación Proteica , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología
3.
Alcohol Clin Exp Res ; 41(10): 1715-1724, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28800142

RESUMEN

BACKGROUND: Heavy and chronic ethanol (EtOH) exposure can cause significant structural and functional damage to the adult brain. The most devastating consequence of EtOH exposure is the neurotoxicity associated with the depletion of neurons. Regulation of splice variants in the brain can modulate protein functions, which may ultimately affect behaviors associated with alcohol dependence and EtOH-mediated neurotoxicity. As alcohol consumption is associated with neurotoxicity, it is possible that altered splicing of survival and pro-survival factors during the development of alcoholism may contribute to the neurotoxicity. METHODS: Primary human neurons and a neuroblastoma cell line were exposed to different concentrations of EtOH for various time periods. Cell viability and neuronal marker expression were analyzed by MTT assay and immunoblotting, respectively. Effect of EtOH exposure on splicing regulatory protein expression and alternative splicing of candidate genes was analyzed by a biochemical approach. Transcriptional activity of serine/arginine-rich splicing factor 1 (SRSF1) gene was determined by reporter gene analysis. RESULTS: Our results suggest that EtOH exposure to neuronal cells at 25 mM and higher concentrations are detrimental. In addition, EtOH exposure caused a dramatic reduction in SRSF1 expression levels. Furthermore, EtOH exposure led to pre-mRNA missplicing of Mcl-1, a pro-survival member of the Bcl-2 family, by down-regulating the expression levels of SRSF1. Moreover, ectopic expression of both SRSF1 and Mcl-1L isoform was able to recover EtOH-mediated neurotoxicity. CONCLUSIONS: Our results suggest that EtOH exposure can lead to pre-mRNA missplicing of Mcl-1 in neuronal cells. Our results indicate that EtOH exposure of neurons leads to a decrease in the ratio of Mcl-1L/Mcl-1S by favoring pro-apoptotic Mcl-1S splicing over anti-apoptotic Mcl-1L isoform suggesting that Mcl-1S may play a crucial role in neurotoxicity associated with alcohol consumption.


Asunto(s)
Empalme Alternativo/fisiología , Etanol/toxicidad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Neuronas/fisiología , Precursores del ARN/genética , Empalme Alternativo/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/biosíntesis , Neuronas/efectos de los fármacos , Precursores del ARN/biosíntesis
4.
J Neurovirol ; 22(5): 597-606, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26951564

RESUMEN

Progressive multifocal leukoemcephalopathy (PML) is a fatal demyelinating disease caused by the human neurotropic JC virus (JCV). JCV infects the majority of the human population during childhood and establishes a latent/persistent life-long infection. The virus reactivates under immunosuppressive conditions by unknown mechanisms, resulting in productive infection of oligodendrocytes in the central nervous system (CNS). Given the fact that the natural occurrence of PML is strongly associated with immunosuppression, the functional and molecular interaction between glial cells and neuroimmune signaling mediated by soluble immune mediators is likely to play a major role in reactivation of JCV and the progression of the lytic viral life cycle leading to the development of PML. In order to explore the effect of soluble immune mediators secreted by peripheral blood mononuclear cells (PBMCs) on JCV transcription, primary human fetal glial (PHFG) cells were treated with conditioned media from PBMCs. We observed a strong suppression of JCV early as well as late gene transcription in cells treated with conditioned media from induced PBMCs. Using a variety of virological and molecular biological approaches, we demonstrate that immune mediators secreted by PBMCs induce the expression of SRSF1, a strong inhibitor of JCV gene expression, and inhibit the replication of JCV. Our results show that downregulation of SRSF1 in glial cells overcomes the suppression of JCV gene expression and its replication mediated by soluble immune mediators. These findings suggest the presence of a novel immune signaling pathway between glial cells and PBMCs that may control JCV gene expression during the course of viral reactivation.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Interacciones Huésped-Patógeno , Virus JC/efectos de los fármacos , Neuroglía/efectos de los fármacos , Factores de Empalme Serina-Arginina/genética , Replicación Viral/efectos de los fármacos , Feto , Regulación de la Expresión Génica , Humanos , Virus JC/genética , Virus JC/crecimiento & desarrollo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Neuroglía/citología , Neuroglía/inmunología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Empalme Serina-Arginina/antagonistas & inhibidores , Factores de Empalme Serina-Arginina/inmunología , Transducción de Señal , Transcripción Genética/efectos de los fármacos
5.
J Cell Physiol ; 230(4): 831-41, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25204229

RESUMEN

Autophagy is an evolutionarily conserved, selective degradation pathway of cellular components that is important for cell homeostasis under healthy and pathologic conditions. Here we demonstrate that an increase in the level of BAG3 results in stimulation of autophagy in glioblastoma cells. BAG3 is a member of a co-chaperone family of proteins that associates with Hsp70 through a conserved BAG domain positioned near the C-terminus of the protein. Expression of BAG3 is induced by a variety of environmental changes that cause stress to cells. Our results show that BAG3 overexpression induces autophagy in glioma cells. Interestingly, inhibition of the proteasome caused an increase in BAG3 levels and induced autophagy. Further analysis using specific siRNA against BAG3 suggests that autophagic activation due to proteosomal inhibition is mediated by BAG3. Analyses of BAG3 domain mutants suggest that the WW domain of BAG3 is crucial for the induction of autophagy. BAG3 overexpression also increased the interaction between Bcl2 and Beclin-1, instead of disrupting them, suggesting that BAG3 induced autophagy is Beclin-1 independent. These observations reveal a novel role for the WW domain of BAG3 in the regulation of autophagy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Glioma/metabolismo , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Beclina-1 , Células Cultivadas , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo
6.
Virol J ; 12: 196, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26596376

RESUMEN

BACKGROUND: Human polyomavirus JCV is the etiologic agent of progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease characterized by lytic infection of glial cells in the central nervous system. PML is seen primarily in immunosuppressed patients and is mainly classified as an AIDS-defining disease. In addition to structural capsid proteins, JCV encodes multiple regulatory proteins, including T-antigen and agnoprotein, which are required for functional lytic infection. Previous studies have suggested that molecular interaction between viral proteins and host factors play an important role in reactivation of JCV and progression of the viral life cycle in glial cells. Recently, serine/arginine rich splicing factor 1 (SRSF1), a cellular alternative splicing factor, was identified as a strong negative regulator of JCV in glial cells. SRSF1 inhibits JCV gene expression and viral replication by directly interacting with viral promoter sequences. Here, we have investigated possible impact of JCV regulatory proteins, T-antigen and agnoprotein, on SRSF1-mediated suppression of JCV gene expression in glial cells. RESULTS: Reporter gene analysis has suggested that T-antigen rescues viral transcriptional suppression mediated by SRSF1. Further analyses have revealed that T-antigen promotes viral gene expression by suppressing SRSF1 gene transcription in glial cells. A subsequent ChIP analysis revealed that T-antigen associates with the promoter region of SRSF1 to induce the transcriptional suppression. CONCLUSIONS: These findings have revealed a molecular interplay between cellular SRSF1 and viral T-antigen in controlling JCV gene expression, and may suggest a novel mechanism of JCV reactivation in patients who are at risk of developing PML.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Regulación Viral de la Expresión Génica , Virus JC/inmunología , Virus JC/fisiología , Neuroglía/inmunología , Neuroglía/virología , Factores de Empalme Serina-Arginina/metabolismo , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune
7.
Virol J ; 10: 147, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23672192

RESUMEN

BACKGROUND: Patients undergoing immune modulatory therapies for the treatment of autoimmune diseases such as multiple sclerosis, and individuals with an impaired-immune system, most notably AIDS patients, are in the high risk group of developing progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease of the white matter caused by human neurotropic polyomavirus, JC virus. It is now widely accepted that pathologic strains of JCV shows unique rearrangements consist of deletions and insertions within viral NCCR. While these kinds of rearrangements are related to viral tropism and pathology of the disease, their roles in molecular regulation of JCV gene expression and replication are unclear. We have previously identified SF2/ASF as a negative regulator of JCV gene expression in glial cells. This negative impact of SF2/ASF was dependent on its ability to bind a specific region mapped to the tandem repeat within viral promoter. In this report, functional role of SF2/ASF binding region in viral gene expression and replication was investigated by using deletion mutants of viral regulatory sequences. RESULTS: The second 98-base-pair tandem repeat on Mad1 strain was first mutated by deletion and named Mad1-(1X98). In addition to this mutant, the CR3 region which served the binding side for SF2/ASF was also mutated and named Mad1-ΔCR3 (1X73). Both mutations were tested for SF2/ASF binding by ChIP assay. While SF2/ASF was associated with Mad1-WT and Mad1-(1X98), its interaction was completely abolished on Mad1-ΔCR3 (1X73) construct as expected. Surprisingly, reporter gene analysis of Mad1-(1X98) and Mad1-ΔCR3 (1X73) early promoter sequences showed two and three fold increase in promoter activities, respectively. The impact of "CR3" region on JCV propagation was also tested on the viral background. While replication of Mad1-(1X98) strain in glial cells was similar to Mad1-WT strain, propagation of Mad1-ΔCR3 (1X73) was less productive. Further analysis of the transcription mediated by Mad1-ΔCR3 (1X73) NCCR revealed that late gene expression was significantly affected. CONCLUSIONS: The results of this study reveal a differential role of CR3 region within JCV NCCR in expression of JCV early and late genes.


Asunto(s)
ADN Viral/metabolismo , Interacciones Huésped-Patógeno , Virus JC/genética , Neuroglía/virología , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Inmunoprecipitación de Cromatina , Análisis Mutacional de ADN , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Factores de Empalme Serina-Arginina
8.
Methods Mol Biol ; 2311: 147-153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033082

RESUMEN

Efficient transfection of genes into the neurons is a crucial step for the study of neuronal cell biology and functions. These include but not limited to investigating gene function by overexpression of target proteins via expression plasmids and knocking down the expression levels of neuronal genes by RNA interference (RNAi). In addition, reporter gene constructs are widely used to investigate the promoter activities of neuronal genes. Numerous transfection techniques have been established to deliver genes into the cells. However, efficient transfection of postmitotic cells, including neurons, still remains a challenging task. Here, we overview the advantages and disadvantages of various techniques for the transfection of primary neurons, and provide an optimized protocol for FuGENE-6 (Promega) which allows for a suitable transfection efficiency of primary neuronal cultures.


Asunto(s)
Lípidos/química , Neuronas/fisiología , Transfección , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Neuronas/metabolismo , Cultivo Primario de Células
9.
J Neuroimmune Pharmacol ; 14(4): 649-660, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31452013

RESUMEN

With the advent of immunomodulatory therapies and the HIV epidemic, the impact of JC Virus (JCV) on the public health system has grown significantly due to the increased incidence of Progressive Multifocal Leukoencephalopathy (PML). Currently, there are no pharmaceutical agents targeting JCV infection for the treatment and the prevention of viral reactivation leading to the development of PML. As JCV primarily reactivates in immunocompromised patients, it is proposed that the immune system (mainly the cellular-immunity component) plays a key role in the regulation of JCV to prevent productive infection and PML development. However, the exact mechanism of JCV immune regulation and reactivation is not well understood. Likewise, the impact of host factors on JCV regulation and reactivation is another understudied area. Here we discuss the current literature on host factor-mediated and immune factor-mediated regulation of JCV gene expression with the purpose of developing a model of the factors that are bypassed during JCV reactivation, and thus are potential targets for the development of therapeutic interventions to suppress PML initiation. Graphical Abstract.


Asunto(s)
Interacciones Microbiota-Huesped/fisiología , Virus JC/inmunología , Virus JC/metabolismo , Leucoencefalopatía Multifocal Progresiva/inmunología , Leucoencefalopatía Multifocal Progresiva/metabolismo , Activación Viral/fisiología , Animales , Humanos , Huésped Inmunocomprometido/fisiología , Inmunoterapia/métodos , Inmunoterapia/tendencias , Leucoencefalopatía Multifocal Progresiva/terapia
10.
Cell Death Dis ; 10(6): 447, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31171771

RESUMEN

Alternative splicing and expression of splice variants of genes in the brain may lead to the modulation of protein functions, which may ultimately influence behaviors associated with alcohol dependence and neurotoxicity. We recently showed that ethanol exposure can lead to pre-mRNA missplicing of Mcl-1, a pro-survival member of the Bcl-2 family, by downregulating the expression levels of serine/arginine rich splicing factor 1 (SRSF1). Little is known about the physiological expression of these isoforms in neuronal cells and their role in toxicity induced by alcohol exposure during the developmental period. In order to investigate the impact of alcohol exposure on alternative splicing of Mcl-1 pre-mRNA and its role in neurotoxicity, we developed a unique primary human neuronal culture model where neurospheres (hNSPs), neural progenitors (hNPCs), immature neurons, and mature neurons were cultured from the matching donor fetal brain tissues. Our data suggest that neural progenitors and immature neurons are highly sensitive to the toxic effects of ethanol, while mature neuron cultures showed resistance to ethanol exposure. Further analysis of Mcl-1 pre-mRNA alternative splicing by semi-quantitative and quantitative analysis revealed that ethanol exposure causes a significant decrease in Mcl-1L/Mcl-1S ratio in a dose and time dependent manner in neural progenitors. Interestingly, ectopic expression of Mcl-1L isoform in neural progenitors was able to recover the viability loss and apoptosis induced by alcohol exposure. Altogether, these observations suggest that alternative splicing of Mcl-1 may play a crucial role in neurotoxicity associated with alcohol exposure in the developing fetal brain.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Etanol/toxicidad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Apoptosis/genética , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/metabolismo , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Organoides/efectos de los fármacos , Organoides/metabolismo , Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
11.
J Cell Physiol ; 215(2): 309-19, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18022798

RESUMEN

The regulatory proteins of polyomaviruses, including small and large T antigens, play important roles, not only in the viral life cycle but also in virus-induced cell transformation. Unlike many other tumor viruses, the transforming proteins of polyomaviruses have no cellular homologs but rather exert their effects mostly by interacting with cellular proteins that control fundamental processes in the regulation of cell proliferation and the cell cycle. Thus, they have proven to be valuable tools to identify specific signaling pathways involved in tumor progression. Elucidation of these pathways using polyomavirus transforming proteins as tools is critically important in understanding fundamental regulatory mechanisms and hence to develop effective therapeutic strategies against cancer. In this short review, we will focus on the structural and functional features of one polyomavirus transforming protein, that is, the small t-antigen of the human neurotropic JC virus (JCV) and the simian virus, SV40.


Asunto(s)
Antígenos Virales de Tumores/fisiología , Transformación Celular Viral/fisiología , Virus JC/fisiología , Virus 40 de los Simios/fisiología , Secuencia de Aminoácidos , Animales , Antígenos Virales de Tumores/genética , Humanos , Virus JC/inmunología , Chaperonas Moleculares/fisiología , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína/fisiología , Sistemas de Lectura , Fase S/fisiología , Virus 40 de los Simios/inmunología , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
J Neuroimmune Pharmacol ; 13(2): 126-142, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29159704

RESUMEN

JC virus (JCV) is a human polyomavirus and the etiologic agent of the demyelinating disease progressive multifocal leukoencephalopathy (PML). PML is observed in patients with underlying immunocompromising conditions, suggesting that neuro-immune interactions between peripheral immune cells and neuro-glia play an important role in controlling viral reactivation in the brain. There is little known about the immunobiology of JCV reactivation in glial cells and the role of immune, glial, and viral players in this regulation. We have previously showed that agnoprotein, a small JCV regulatory protein, is released from infected cells and internalized by neighboring bystander cells. Here we have investigated the possible role of extracellular and intracellular agnoprotein in the neuroimmune response to JC virus. Our findings suggest that glial cells exposed to agnoprotein secrete significantly less GM-CSF, which is mediated by agnoprotein induced suppression of GM-CSF transcription. Likewise, monocytes treated with agnoprotein showed altered differentiation and maturation. In addition, monocytes and microglial cells exposed to agnoprotein showed a significant reduction in their phagocytic activities. Moreover, when an in vitro blood-brain barrier model was used, agnoprotein treatment resulted in decreased monocyte migration through the endothelial cell layer in response to activated astrocytes. All together, these results have revealed a novel immunomodulatory function of agnoprotein during JCV infection within theCNS and open a new avenue of research to better understand the mechanisms associated with JCV reactivation in patients who are at risk of developing PML.


Asunto(s)
Leucoencefalopatía Multifocal Progresiva/inmunología , Monocitos/inmunología , Monocitos/virología , Neuroinmunomodulación/inmunología , Proteínas Reguladoras y Accesorias Virales/inmunología , Activación Viral/inmunología , Línea Celular , Humanos , Virus JC/inmunología
13.
Genes Cancer ; 9(3-4): 130-141, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30108683

RESUMEN

The human neurotropic polyomavirus JC, JC virus (JCV), infects the majority of human population during early childhood and establishes a latent/persistent infection for the rest of the life. JCV is the etiologic agent of the fatal demyelinating disease of the central nervous system, progressive multifocal leukoencephalopathy (PML) that is seen primarily in immunocompromised individuals. In addition to the PML, JCV has also been shown to transform cells in culture systems and cause a variety of tumors in experimental animals. Moreover, JCV genomic DNA and tumor antigen expression have been shown in a variety of human tumors with CNS origin. Similar to all polyomaviruses, JCV encodes for several tumor antigens from a single transcript of early coding region via alternative splicing. There is little known regarding the characteristics of JCV induced tumors and impact of DNA damage induced by radiation on viral tumor antigen expression and growth of these cells. Here we analyzed the possible impact of ionizing radiation on transformed phenotype and tumor antigen expression by utilizing a mouse medulloblastoma cell line (BSB8) obtained from a mouse transgenic for JCV tumor antigens. Our results suggest that a small subset of BSB8 cells survives and shows radiation resistance. Further analysis of the transformed phenotype of radiation resistant BSB8 cells (BSB8-RR) have revealed that they are capable of forming significantly higher numbers and sizes of colonies under anchorage dependent and independent conditions with reduced viral tumor antigen expression. Moreover, BSB8-RR cells show an increased rate of double-strand DNA break repair by homologous recombination (HR). More interestingly, knockout studies of JCV tumor antigens by utilizing CRISPR/Cas9 gene editing reveal that unlike parental BSB8 cells, BSB8-RR cells are no longer required the expression of viral tumor antigens in order to maintain transformed phenotype.

14.
Artículo en Inglés | MEDLINE | ID: mdl-30406039

RESUMEN

Autophagy, a highly conserved process, serves to maintain cellular homeostasis in response to an extensive variety of internal and external stimuli. The classic, or canonical, pathway of autophagy involves the coordinated degradation and recycling of intracellular components and pathogenic material. Proper regulation of autophagy is critical to maintain cellular health, as alterations in the autophagy pathway have been linked to the progression of a variety of physiological and pathological conditions in humans, namely in aging and in viral infection. In addition to its canonical role as a degradative pathway, a more unconventional and non-degradative role for autophagy has emerged as an area of increasing interest. This process, known as secretory autophagy, is gaining widespread attention as many viruses are believed to use this pathway as a means to release and spread viral particles. Moreover, secretory autophagy has been found to intersect with other intracellular pathways, such as the biogenesis and secretion of extracellular vesicles (EVs). Here, we provide a review of the current landscape surrounding both degradative autophagy and secretory autophagy in relation to both aging and viral infection. We discuss their key features, while describing their interplay with numerous different viruses (i.e. hepatitis B and C viruses, Epstein-Barr virus, SV40, herpesviruses, HIV, chikungunya virus, dengue virus, Zika virus, Ebola virus, HTLV, Rift Valley fever virus, poliovirus, and influenza A virus), and compare secretory autophagy to other pathways of extracellular vesicle release. Lastly, we highlight the need for, and emphasize the importance of, more thorough methods to study the underlying mechanisms of these pathways to better advance our understanding of disease progression.


Asunto(s)
Envejecimiento/fisiología , Autofagia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/virología , Virosis/inmunología , Liberación del Virus , Virus/inmunología , Animales , Humanos
15.
Cell Death Dis ; 8(1): e2542, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28079886

RESUMEN

Human immunodeficiency virus-associated neurological disorders (HANDs) affect the majority of AIDS patients and are a significant problem among HIV-1-infected individuals who live longer because of combined anti-retroviral therapies. HIV-1 utilizes a number of viral proteins and subsequent cytokine inductions to unleash its toxicity on neurons. Among HIV-1 viral proteins, Nef is a small protein expressed abundantly in astrocytes of HIV-1-infected brains and has been suggested to have a role in the pathogenesis of HAND. In order to explore its effect in the central nervous system, HIV-1 Nef was expressed in primary human fetal astrocytes (PHFAs) using an adenovirus. Our results revealed that HIV-1 Nef is released in extracellular vesicles (EVs) derived from PHFA cells expressing the protein. Interestingly, HIV-1 Nef release in EVs was enriched significantly when the cells were treated with autophagy activators perifosine, tomaxifen, MG-132, and autophagy inhibitors LY294002 and wortmannin suggesting a novel role of autophagy signaling in HIV-1 Nef release from astrocytes. Next, Nef-carrying EVs were purified from astrocyte cultures and neurotoxic effects on neurons were analyzed. We observed that HIV-1 Nef-containing EVs were readily taken up by neurons as demonstrated by immunocytochemistry and immunoblotting. Furthermore, treatment of neurons with Nef-carrying EVs induced oxidative stress as evidenced by a decrease in glutathione levels. To further investigate its neurotoxic effects, we expressed HIV-1 Nef in primary neurons by adenoviral transduction. Intracellular expression of HIV-1 Nef caused axonal and neurite degeneration of neurons. Furthermore, expression of HIV-1 Nef decreased the levels of phospho-tau while enhancing total tau in primary neurons. In addition, treatment of primary neurons with Nef-carrying EVs suppressed functional neuronal action potential assessed by multielectrode array studies. Collectively, these data suggested that HIV-1 Nef can be a formidable contributor to neurotoxicity along with other factors, which leads to HAND in HIV-1-infected AIDS patients.


Asunto(s)
Vesículas Extracelulares/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Síndromes de Neurotoxicidad/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/virología , Autofagia/efectos de los fármacos , Autofagia/genética , Vesículas Extracelulares/virología , Feto/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/virología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/virología , Cultivo Primario de Células , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/administración & dosificación
16.
PLoS One ; 11(6): e0156819, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27257867

RESUMEN

OBJECTIVE: PML is a rare and fatal demyelinating disease of the CNS caused by the human polyomavirus, JC virus (JCV), which occurs in AIDS patients and those on immunosuppressive monoclonal antibody therapies (mAbs). We sought to identify mechanisms that could stimulate reactivation of JCV in a cell culture model system and targeted pathways which could affect early gene transcription and JCV T-antigen production, which are key steps of the viral life cycle for blocking reactivation of JCV. Two important regulatory partners we have previously identified for T-antigen include Pur-alpha and SRSF1 (SF2/ASF). SRSF1, an alternative splicing factor, is a potential regulator of JCV whose overexpression in glial cells strongly suppresses viral gene expression and replication. Pur-alpha has been most extensively characterized as a sequence-specific DNA- and RNA-binding protein which directs both viral gene transcription and mRNA translation, and is a potent inducer of the JCV early promoter through binding to T-antigen. METHODS AND RESULTS: Pur-alpha and SRSF1 both act directly as transcriptional regulators of the JCV promoter and here we have observed that Pur-alpha is capable of ameliorating SRSF1-mediated suppression of JCV gene expression and viral replication. Interestingly, Pur-alpha exerted its effect by suppressing SRSF1 at both the protein and mRNA levels in glial cells suggesting this effect can occur independent of T-antigen. Pur-alpha and SRSF1 were both localized to oligodendrocyte inclusion bodies by immunohistochemistry in brain sections from patients with HIV-1 associated PML. Interestingly, inclusion bodies were typically positive for either Pur-alpha or SRSF1, though some cells appeared to be positive for both proteins. CONCLUSIONS: Taken together, these results indicate the presence of an antagonistic interaction between these two proteins in regulating of JCV gene expression and viral replication and suggests that they play an important role during viral reactivation leading to development of PML.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neuroglía/metabolismo , Neuroglía/virología , Factores de Empalme Serina-Arginina/metabolismo , Factores de Transcripción/metabolismo , Antígenos Virales de Tumores/genética , Southern Blotting , Células Cultivadas , Proteínas de Unión al ADN/genética , Humanos , Inmunohistoquímica , Virus JC/genética , Virus JC/patogenicidad , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Empalme Serina-Arginina/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Transcripción Genética/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
17.
Cell Cycle ; 14(18): 2899-904, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26176554

RESUMEN

Viruses often exploit autophagy, a common cellular process of degradation of damaged proteins, organelles, and pathogens, to avoid destruction. HIV-1 dysregulates this process in several cell types by means of Nef protein. Nef is a small HIV-1 protein which is expressed abundantly in astrocytes of HIV-1-infected brains and has been suggested to have a role in the pathogenesis of HIV-Associated Neurocognitive Disorders (HAND). In order to explore its effect in the CNS with respect to autophagy, HIV-1 Nef was expressed in primary human fetal astrocytes (PHFA) using an adenovirus vector (Ad-Nef). We observed that Nef expression triggered the accumulation of autophagy markers, ATG8/LC3 and p62 (SQSMT1). Similar results were obtained with Bafilomycin A1, an autophagy inhibitor which blocks the fusion of autophagosome to lysosome. Furthermore co-expression of tandem LC3 vector (mRFP-EGFP-LC3) and Ad-Nef in these cells produced mainly yellow puncta (mRFP+, EGFP+) strongly suggesting that autophagosome fusion to lysosome is blocked in PHFA cells in the presence of Nef. Together these data indicate that HIV-1 Nef mimics Bafilomycin A1 and blocks the last step of autophagy thereby helping HIV-1 virus to avoid autophagic degradation in human astrocytes.


Asunto(s)
Astrocitos/patología , Autofagia , VIH-1/patogenicidad , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/virología , Familia de las Proteínas 8 Relacionadas con la Autofagia , Biomarcadores/metabolismo , Humanos , Macrólidos/farmacología , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Unión al ARN/metabolismo
18.
PLoS One ; 10(6): e0129694, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26061652

RESUMEN

OBJECTIVE: Patients undergoing immune modulatory therapies for the treatment of autoimmune diseases such as multiple sclerosis, and individuals with an impaired-immune system, most notably AIDS patients, are in the high risk group of developing progressive multifocal leukoencephalopathy (PML), an often lethal disease of the brain characterized by lytic infection of oligodendrocytes in the central nervous system (CNS) with JC virus (JCV). The immune system plays an important regulatory role in controlling JCV reactivation from latent sites by limiting viral gene expression and replication. However, little is known regarding the molecular mechanisms responsible for this regulation. METHODS AND RESULTS: Here, we investigated the impact of soluble immune mediators secreted by activated PBMCs on viral replication and gene expression by cell culture models and molecular virology techniques. Our data revealed that viral gene expression and viral replication were suppressed by soluble immune mediators. Further studies demonstrated that soluble immune mediators secreted by activated PBMCs inhibit viral replication induced by T-antigen, the major viral regulatory protein, by suppressing its expression in glial cells. This unexpected suppression of T-antigen was mainly associated with the suppression of translational initiation. Cytokine/chemokine array studies using conditioned media from activated PBMCs revealed several candidate cytokines with possible roles in this regulation. Among them, only IFN-γ showed a robust inhibition of T-antigen expression. While potential roles for IFN-ß, and to a lesser extent IFN-α have been described for JCV, IFN-γ has not been previously implicated. Further analysis of IFN-γ signaling pathway revealed a novel role of Jak1 signaling in control of viral T-antigen expression. Furthermore, IFN-γ suppressed JCV replication and viral propagation in primary human fetal glial cells, and showed a strong anti-JCV activity. CONCLUSIONS: Our results suggest a novel role for IFN-γ in the regulation of JCV gene expression via downregulation of the major viral regulatory protein, T-antigen, and provide a new avenue of research to understand molecular mechanisms for downregulation of viral reactivation that may lead to development of novel strategies for the treatment of PML.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Antivirales/farmacología , Interferón gamma/farmacología , Virus JC/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antígenos Virales de Tumores/genética , Línea Celular Tumoral , Humanos , Virus JC/inmunología , Virus JC/fisiología , Neuroglía/virología
19.
Virology ; 468-470: 88-95, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25151063

RESUMEN

Poliomavirus JC replicates in glial cells in the brain, and causes the fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML). PML is usually seen in patients with underlying immunocompromised conditions, notably among AIDS patients and those on chronic immunosuppressive regimens. The late leader sequence of JC virus contains an open reading frame encoding a small regulatory protein called agnoprotein. Agnoprotein contributes to progressive viral infection by playing significant roles in viral replication cycle. Here, we demonstrate that agnoprotein can be detected in cell-free fractions of glial cultures infected with JCV, transfected with expression plasmids or transduced with an adenovirus expression system. We also provide evidence that extracellular agnoprotein can be taken up by uninfected neighboring cells. These studies have revealed a novel phenomenon of agnoprotein during the viral life cycle with a potential of developing diagnostic and therapeutic interventions.


Asunto(s)
Virus JC/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Adenoviridae , Línea Celular Tumoral , Sistema Libre de Células , Exosomas , Humanos , Virus JC/genética , Plásmidos , Proteínas Reguladoras y Accesorias Virales/genética , Replicación Viral/fisiología
20.
Cell Cycle ; 13(23): 3640-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483098

RESUMEN

BAG3 protein has been described as an anti-apoptotic and pro-autophagic factor in several neoplastic and normal cells. We previously demonstrated that BAG3 expression is elevated upon HIV-1 infection of glial and T lymphocyte cells. Among HIV-1 proteins, Tat is highly involved in regulating host cell response to viral infection. Therefore, we investigated the possible role of Tat protein in modulating BAG3 protein levels and the autophagic process itself. In this report, we show that transfection with Tat raises BAG3 levels in glioblastoma cells. Moreover, BAG3 silencing results in highly reducing Tat- induced levels of LC3-II and increasing the appearance of sub G0/G1 apoptotic cells, in keeping with the reported role of BAG3 in modulating the autophagy/apoptosis balance. These results demonstrate for the first time that Tat protein is able to stimulate autophagy through increasing BAG3 levels in human glial cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Reguladoras de la Apoptosis/biosíntesis , Autofagia/efectos de los fármacos , Neuroglía/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Neuroglía/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA