Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Virology ; 587: 109866, 2023 10.
Article in English | MEDLINE | ID: mdl-37741199

ABSTRACT

We previously reported the discovery and characterization of two novel proteins (ORF1 and ORF2) generated by the alternative splicing of the JC virus (JCV) late coding region. Here, we report the discovery and partial characterization of three additional novel ORFs from the same coding region, ORF3, ORF4 and ORF5, which potentially encode 70, 173 and 265 amino acid long proteins respectively. While ORF3 protein exhibits a uniform distribution pattern throughout the cells, we were unable to detect ORF5 expression. Surprisingly, ORF4 protein was determined to be the only JCV protein specifically targeting the promyelocytic leukemia nuclear bodies (PML-NBs) and inducing their reorganization in nucleus. Although ORF4 protein has a modest effect on JCV replication, it is implicated to play major roles during the JCV life cycle, perhaps by regulating the antiviral response of PML-NBs against JCV infections and thus facilitating the progression of the JCV-induced disease in infected individuals.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , Polyomavirus , Humans , JC Virus/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Open Reading Frames , Promyelocytic Leukemia Nuclear Bodies
2.
Int J Mol Sci ; 23(17)2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36077131

ABSTRACT

Protein quality control allows eukaryotes to maintain proteostasis under the stress of constantly changing conditions. In this review, we discuss the current literature on PQC, highlighting flaws that must exist for malignancy to occur. At the nidus of PQC, the expression of BAG1-6 reflects the cell environment; each isoform directs proteins toward different, parallel branches of the quality control cascade. The sum of these branches creates a net shift toward either homeostasis or apoptosis. With an established role in ALP, Bag3 is necessary for cell survival in stress conditions including those of the cancerous niche (i.e., hypoxia, hypermutation). Evidence suggests that excessive Bag3-HSP70 activity not only sustains, but also propagates cancers. Its role is anti-apoptotic-which allows malignant cells to persist-and intercellular-with the production of infectious 'oncosomes' enabling cancer expansion and recurrence. While Bag3 has been identified as a key prognostic indicator in several cancer types, its investigation is limited regarding glioblastoma. The cochaperone HSP70 has been strongly linked with GBM, while ALP inhibitors have been shown to improve GBM susceptibility to chemotherapeutics. Given the highly resilient, frequently recurrent nature of GBM, the targeting of Bag3 is a necessary consideration for the successful and definitive treatment of GBM.


Subject(s)
Glioblastoma , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy , Glioblastoma/drug therapy , HSP70 Heat-Shock Proteins/metabolism , Humans , Transcription Factors/metabolism
3.
Viruses ; 13(10)2021 09 26.
Article in English | MEDLINE | ID: mdl-34696366

ABSTRACT

Polyomavirus JC (JCPyV) causes the demyelinating disease progressive multifocal leukoencephalopathy (PML). JCPyV infection is very common in childhood and, under conditions of severe immunosuppression, JCPyV may reactivate to cause PML. JC viral proteins expression is regulated by the JCPyV non-coding control region (NCCR), which contains binding sites for cellular transcriptional factors which regulate JCPyV transcription. Our earlier studies suggest that JCPyV reactivation occurs within glial cells due to cytokines such as TNF-α which stimulate viral gene expression. In this study, we examined interferon-α (IFNα) or ß (IFNß) which have a negative effect on JCPyV transcriptional regulation. We also showed that these interferons induce the endogenous liver inhibitory protein (LIP), an isoform of CAAT/enhancer binding protein beta (C/EBPß). Treatment of glial cell line with interferons increases the endogenous level of C/EBPß-LIP. Furthermore, we showed that the negative regulatory role of the interferons in JCPyV early and late transcription and viral replication is more pronounced in the presence of C/EBPß-LIP. Knockdown of C/EBPß-LIP by shRNA reverse the inhibitory effect on JCPyV viral replication. Therefore, IFNα and IFNß negatively regulate JCPyV through induction of C/EBPß-LIP, which together with other cellular transcriptional factors may control the balance between JCPyV latency and activation.


Subject(s)
Interferon-alpha/metabolism , Interferon-beta/metabolism , JC Virus/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , DNA, Viral/genetics , Gene Expression/genetics , Gene Expression Regulation, Viral/genetics , Humans , Interferon-alpha/immunology , Interferon-beta/immunology , JC Virus/genetics , JC Virus/immunology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Neuroglia , Protein Isoforms , Virus Replication/genetics
4.
Methods Mol Biol ; 2311: 155-160, 2021.
Article in English | MEDLINE | ID: mdl-34033083

ABSTRACT

Lentiviruses are a very reliable class of viral vectors wildly used in gene therapy. In this chapter, we described a general method for the construction of lentiviral delivery system by using a derived HIV-1 based lentivirus expression vector pKLV-Puro containing a monomeric blue fluorescent protein mammalian codon-optimized (TagBFP). HIV-1 based lentivirus particles are prepared by transfection of four plasmids into 293 T cells using the Fugene 6 transfection reagent. In this case, the target cells for transduction are human primary fetal astrocytes but the method is applicable to any primary cell culture from the CNS or other tissue.


Subject(s)
Astrocytes/metabolism , Genetic Vectors , HIV-1/genetics , Luminescent Proteins/metabolism , Transduction, Genetic , Fetus , Genes, Reporter , Gestational Age , HEK293 Cells , Humans , Lipids/chemistry , Luminescent Proteins/genetics , Primary Cell Culture
5.
Int J Mol Sci ; 21(17)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872288

ABSTRACT

Cancer is the second leading cause of mortality worldwide. The study of DNA tumor-inducing viruses and their oncoproteins as a causative agent in cancer initiation and tumor progression has greatly enhanced our understanding of cancer cell biology. The initiation of oncogenesis is a complex process. Specific gene mutations cause functional changes in the cell that ultimately result in the inability to regulate cell differentiation and proliferation effectively. The human neurotropic Polyomavirus JC (JCV) belongs to the family Polyomaviridae and it is the causative agent of progressive multifocal leukoencephalopathy (PML), which is a fatal neurodegenerative disease in an immunosuppressed state. Sero-epidemiological studies have indicated JCV infection is prevalent in the population (85%) and that initial infection usually occurs during childhood. The JC virus has small circular, double-stranded DNA that includes coding sequences for viral early and late proteins. Persistence of the virus in the brain and other tissues, as well as its potential to transform cells, has made it a subject of study for its role in brain tumor development. Earlier observation of malignant astrocytes and oligodendrocytes in PML, as well as glioblastoma formation in non-human primates inoculated with JCV, led to the hypothesis that JCV plays a role in central nervous system (CNS) tumorigenesis. Some studies have reported the presence of both JC viral DNA and its proteins in several primary brain tumor specimens. The discovery of new Polyomaviruses such as the Merkel cell Polyomavirus, which is associated with Merkel cell carcinomas in humans, ignited our interest in the role of the JC virus in CNS tumors. The current evidence known about JCV and its effects, which are sufficient to produce tumors in animal models, suggest it can be a causative factor in central nervous system tumorigenesis. However, there is no clear association between JCV presence in CNS and its ability to initiate CNS cancer and tumor formation in humans. In this review, we will discuss the correlation between JCV and tumorigenesis of CNS in animal models, and we will give an overview of the current evidence for the JC virus's role in brain tumor formation.


Subject(s)
Central Nervous System Neoplasms/virology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/complications , Animals , Disease Progression , Gene Expression Regulation, Viral , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Mutation , Viral Proteins/genetics
6.
Mol Ther ; 27(12): 2067-2079, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31653397

ABSTRACT

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.


Subject(s)
Anti-HIV Agents/pharmacology , Brain/drug effects , Receptor, Interferon alpha-beta/physiology , Rilpivirine/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Animals , Brain/virology , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , Zika Virus Infection/pathology , Zika Virus Infection/virology
7.
J Neuroimmune Pharmacol ; 14(4): 578-594, 2019 12.
Article in English | MEDLINE | ID: mdl-31512166

ABSTRACT

Virus-induced diseases or neurological complications are huge socio-economic burden to human health globally. The complexity of viral-mediated CNS pathology is exacerbated by reemergence of new pathogenic neurotropic viruses of high public relevance. Although the central nervous system is considered as an immune privileged organ and is mainly protected by barrier system, there are a vast majority of neurotropic viruses capable of gaining access and cause diseases. Despite continued growth of the patient population and a number of treatment strategies, there is no successful viral specific therapy available for viral induced CNS diseases. Therefore, there is an urgent need for a clear alternative treatment strategy that can effectively target neurotropic viruses of DNA or RNA genome. To address this need, rapidly growing gene editing technology based on CRISPR/Cas9, provides unprecedented control over viral genome editing and will be an effective, highly specific and versatile tool for targeting CNS viral infection. In this review, we discuss the application of this system to control CNS viral infection and associated neurological disorders and future prospects. Graphical Abstract CRISPR/Cas9 technology as agent control over CNS viral infection.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Central Nervous System Diseases/genetics , Central Nervous System Diseases/therapy , Gene Editing/trends , Genetic Therapy/trends , Animals , CRISPR-Associated Protein 9/metabolism , Central Nervous System Diseases/metabolism , Gene Editing/methods , Genetic Therapy/methods , Humans
8.
J Cell Physiol ; 233(5): 4137-4155, 2018 05.
Article in English | MEDLINE | ID: mdl-29044559

ABSTRACT

Although the human neurotropic polyomavirus, JC virus (JCV), was isolated almost a half century ago, understanding the molecular mechanisms governing its biology remains highly elusive. JCV infects oligodendrocytes and astrocytes in the central nervous system (CNS) and causes a rare fatal brain disease known as progressive multifocal leukoencephalopathy (PML) in immunocompromised individuals including AIDS. It has a small circular DNA genome (∼5 kb) and generates two primary transcripts from its early and late coding regions, producing several predicted alternatively spliced products mainly by cis-splicing. Here, we report the discovery and characterization of two novel open reading frames (ORF1 and ORF2) associated with JCV late transcripts, generated by an unusual splicing process called trans-splicing. These ORFs result from (i) the trans-splicing of two different lengths of the 5'-short coding region of VP1 between the coding regions of agnoprotein and VP2 after replacing the intron located between these two coding regions and (ii) frame-shifts occurring within the VP2 coding sequences terminated by a stop codon. ORF1 and ORF2 are capable of encoding 58 and 72 aa long proteins respectively and are expressed in infected cells and PML patients. Each ORF protein shares a common coding region with VP1 and has a unique coding sequence of their own. When the expression of the unique coding regions of ORFs is blocked by a stop codon insertion in the viral background, the mutant virus replicates less efficiently when compared to wild-type, suggesting that the newly discovered ORFs play critical roles in the JCV life cycle.


Subject(s)
JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Polyomavirus/genetics , Trans-Splicing/genetics , Brain/virology , Codon, Terminator/genetics , DNA, Viral/classification , DNA, Viral/genetics , Exons/genetics , Gene Expression Regulation, Viral , Genome, Viral/genetics , Humans , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Open Reading Frames , Polyomavirus/pathogenicity , Virus Replication/genetics
9.
Virol J ; 14(1): 31, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28202068

ABSTRACT

BACKGROUND: Infection of glial cells by human neurotropic polyomavirus JC (JCV), the causative agent of the CNS demyelinating disease progressive multifocal leukoencephalopathy (PML), rapidly inflicts damage to cellular DNA. This activates DNA damage response (DDR) signaling including induction of expression of DNA repair factor Rad51. We previously reported that Rad51 co-operates with the transcription factor NF-κB p65 to activate JCV early transcription. Thus Rad51 induction by JCV infection may provide positive feedback for viral activation early in JCV infection. DDR is also known to stimulate NF-κB activity, a phenomenon known as nucleus to cytoplasm or "inside-out" NF-κB signaling, which is initiated by Ataxia telangiectasia mutated (ATM) protein, a serine/threonine kinase recruited and activated by DNA double-strand breaks. Downstream of ATM, there occurs a series of post-translational modifications of NF-κB essential modulator (NEMO), the γ regulatory subunit of inhibitor of NF-κB (IκB) kinase (IKK), resulting in NF-κB activation. METHODS: We analyzed the effects of downstream pathways in the DDR by phosphospecific Western blots and analysis of the subcellular distribution of NEMO by cell fractionation and immunocytochemistry. The role of DDR in JCV infection was analyzed using a small molecule inhibitor of ATM (KU-55933). NEMO sumoylation was investigated by Western and association of ATM and NEMO by immunoprecipitation/Western blots. RESULTS: We show that JCV infection caused phosphorylation and activation of ATM while KU-55933 inhibited JCV replication. JCV infection caused a redistribution of NEMO from cytoplasm to nucleus. Co-expression of JCV large T-antigen and FLAG-tagged NEMO showed the occurrence of sumoylation of NEMO, while co-expression of ATM and FLAG-NEMO demonstrated physical association between ATM and NEMO. CONCLUSIONS: We propose a model where JCV infection induces both overexpression of Rad51 protein and activation of the nucleus to cytoplasm NF-κB signaling pathway, which then act together to enhance JCV gene expression.


Subject(s)
DNA Damage , Host-Pathogen Interactions , JC Virus/growth & development , NF-kappa B/metabolism , Neuroglia/virology , Signal Transduction , Stress, Physiological , Blotting, Western , Cell Fractionation , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Viral , Humans , I-kappa B Kinase/analysis , Immunohistochemistry , JC Virus/genetics , Models, Biological , Protein Transport , Rad51 Recombinase/metabolism , Transcription, Genetic
10.
Ann Neurol ; 80(4): 479-89, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27464346

ABSTRACT

The emergence of Zika virus in the Americas has followed a pattern that is familiar from earlier epidemics of other viruses, where a new disease is introduced into a human population and then spreads rapidly with important public health consequences. In the case of Zika virus, an accumulating body of recent evidence implicates the virus in the etiology of serious pathologies of the human nervous system, that is, the occurrence of microcephaly in neonates and Guillain-Barré syndrome in adults. Zika virus is an arbovirus (arthropod-borne virus) and a member of the family Flaviviridae, genus Flavivirus. Zika virions are enveloped and icosahedral, and contain a nonsegmented, single-stranded, positive-sense RNA genome, which encodes 3 structural and 7 nonstructural proteins that are expressed as a single polyprotein that undergoes cleavage. Zika genomic RNA replicates in the cytoplasm of infected host cells. Zika virus was first detected in 1947 in the blood of a febrile monkey in Uganda's Zika Forest and in crushed suspensions of the Aedes mosquito, which is one of the vectors for Zika virus. The virus remained obscure, with a few human cases confined to Africa and Asia. There are two lineages of the Zika virus, African and Asian, with the Asian strain causing outbreaks in Micronesia in 2007 and French Polynesia in 2013-2014. From here, the virus spread to Brazil with the first report of autochthonous Zika transmission in the Americas in March 2015. The rapid advance of the virus in the Americas and its likely association with microcephaly and Guillain-Barré syndrome make Zika an urgent public health concern. Ann Neurol 2016;80:479-489.


Subject(s)
Guillain-Barre Syndrome/etiology , Microcephaly/etiology , Zika Virus Infection/complications , Zika Virus/genetics , Zika Virus/pathogenicity , Adult , History, 20th Century , History, 21st Century , Humans , Infant, Newborn , Zika Virus/classification , Zika Virus Infection/epidemiology , Zika Virus Infection/history
11.
Sci Rep ; 6: 23146, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27064617

ABSTRACT

HSV-1 induced illness affects greater than 85% of adults worldwide with no permanent curative therapy. We used RNA-guided CRISPR/Cas9 gene editing to specifically target for deletion of DNA sequences of the HSV-1 genome that span the region directing expression of ICP0, a key viral protein that stimulates HSV-1 gene expression and replication. We found that CRISPR/Cas9 introduced InDel mutations into exon 2 of the ICP0 gene profoundly reduced HSV-1 infectivity in permissive human cell culture models and protected permissive cells against HSV-1 infection. CRISPR/Cas9 mediated targeting ICP0 prevented HSV-1-induced disintegration of promonocytic leukemia (PML) nuclear bodies, an intracellular event critical to productive HSV-1 infection that is initiated by interaction of the ICP0 N-terminus with PML. Combined treatment of cells with CRISPR targeting ICP0 plus the immediate early viral proteins, ICP4 or ICP27, completely abrogated HSV-1 infection. We conclude that RNA-guided CRISPR/Cas9 can be used to develop a novel, specific and efficacious therapeutic and prophylactic platform for targeted viral genomic ablation to treat HSV-1 diseases.


Subject(s)
Gene Editing/methods , Genes, Viral , Herpesvirus 1, Human/physiology , Virus Replication , Animals , CRISPR-Cas Systems , Cell Line , Chlorocebus aethiops , DNA, Viral/genetics , Herpesvirus 1, Human/genetics , Humans , INDEL Mutation , Sequence Deletion
12.
J Neurovirol ; 22(5): 615-625, 2016 10.
Article in English | MEDLINE | ID: mdl-27007123

ABSTRACT

Brd4 is an epigenetic reader protein and a member of the BET (bromodomain and extra terminal domain) family of proteins with two bromodomains that recognize acetylated lysine residues. Brd4 specifically binds to acetylated transcription factor NF-κB p65 and coactivates transcription. Polyomavirus JC (JCV) is regulated by a noncoding control region (NCCR) containing promoter/enhancer elements for viral gene expression including a binding site for NF-κB, which responds to proinflammatory cytokines such as TNF-α, the DNA damage response, calcium signaling and acetylation of the NF-κB p65 subunit on lysine residues K218 and K221. Earlier studies indicated that NF-κB is involved in the reactivation of persistent/latent JCV in glial cells to cause progressive multifocal leukoencephalopathy (PML), a severe demyelinating disease of the brain caused by replication of JCV in glial cells. To investigate the mechanism of action of NF-κB acetylation on JCV transcription, we examined Brd4 and found that JCV early transcription was stimulated by Brd4 via the JCV NF-κB site and that p65 K218 and K221 were involved. Treatment with the Brd4 inhibitor JQ1(+) or mutation of either K218 or K221 to glutamine (K218R or K221) inhibited this stimulation and decreased the proportion of p65 in the nucleus. We conclude that Brd4 is involved in the regulation of the activation status of JCV in glial cells.


Subject(s)
Host-Pathogen Interactions , JC Virus/drug effects , Nuclear Proteins/genetics , Transcription Factor RelA/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , Virus Replication/drug effects , Acetylation , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Epigenesis, Genetic , Genes, Reporter , Humans , JC Virus/genetics , JC Virus/growth & development , Luciferases/genetics , Luciferases/metabolism , Mutation , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Signal Transduction , Transcription Factor RelA/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Virus Activation
13.
Curr HIV Res ; 14(1): 47-53, 2016.
Article in English | MEDLINE | ID: mdl-26531763

ABSTRACT

INTRODUCTION: Progressive multifocal leukoencephalopathy (PML) is a debilitating demyelinating disease of the CNS caused by the infection and destruction of glial cells by JC virus (JCV) and is an AIDS-defining disease. Infection with JCV is common and most people acquire antibodies early in life. After initial infection, JCV remains in an asymptomatic persistent state and can be detected by PCR in many tissues including brain. A major question in PML pathogenesis is how the virus reactivates from persistence in HIV-1/AIDS. Our studies with primary cultures of glial cells have implicated transcription factors NF-κB and NFAT4, which bind to a unique site in the JCV noncoding control region and stimulate viral gene expression. Furthermore, these transcription factors are controlled by pathways downstream of proinflammatory cytokines, e.g., TNF-α activates NF-κB and stimulates JCV transcription. OBJECTIVES: We hypothesize that HIV-1/PML initiation may involve reactivation of JCV by cytokine disturbances in the brain such as occur in HIV-1/AIDS. In this study, the objective was to evaluate HIV-1/PML clinical samples for expression of TNF-α and its receptors and subcellular localization of NF-κB p65 and NFAT4 compared to non-PML controls. METHODS: We evaluated HIV-1/PML clinical samples and non-PML controls for expression of TNF-α and its receptors and subcellular localization of NF-κB p65 and NFAT4 using Western blot and immunohistochemistry. RESULTS: Consistent with our hypothesis, compared to non-PML controls, HIV-1/PML tissue has high levels of TNF-α and TNFR1 expression and NF-κB and NFAT4 were preferentially localized to the nucleus. CONCLUSION: The involvement of TNF-α/NF-κB/NFAT4 signaling in JCV regulation that we reported from experiments in cultured human glial cells may be clinically relevant in PML.


Subject(s)
HIV Infections/metabolism , Leukoencephalopathy, Progressive Multifocal/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Neuroglia/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Blotting, Western , Brain/metabolism , Case-Control Studies , Cell Line , Female , HIV Infections/complications , HIV-1 , Humans , Immunohistochemistry , Male , Middle Aged , Protein Subunits/metabolism , Virus Activation/physiology
14.
PLoS One ; 10(9): e0136046, 2015.
Article in English | MEDLINE | ID: mdl-26360417

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease of the central nervous system (CNS) caused by reactivation of the human polyomavirus JCV gene expression and its replication in oligodendrocytes, the myelin producing cells in the brain. Once a rare disease seen in patients with lymphotproliferative and myeloproliferative disorders, PML has been seen more frequently in HIV-1 positive/AIDS patients as well as patients undergoing immunomodulatory therapy due for autoimmune disorders including multiple sclerosis, rheumatoid arthritis, and others. As of now there is no cure for PML and in most cases disease progression leads to death within two years. Similar to other polyomaviruses, the JCV genome is small circular double stranded DNA that includes coding sequences for the viral early protein, T-antigen, which is critical for directing viral reactivation and lytic infection. Here, we employ a newly developed gene editing strategy, CRISPR/Cas9, to introduce mutations in the viral genome and, by inactivating the gene encoding T-antigen, inhibit viral replication. We first used bioinformatics screening and identified several potential targets within the JCV T-antigen gene that can serve as sites for the creation of guide RNAs (gRNAs) for positioning the Cas9 nuclease on the designated area of the viral genome for editing. Results from a series of integrated genetic and functional studies showed that transient or conditional expression of Cas9 and gRNAs specifically targets the DNA sequences corresponding to the N-terminal region of T-antigen, and by introducing mutation, interferes with expression and function of of the viral protein, hence suppressing viral replication in permissive cells. Results from SURVEYOR assay revealed no off-target effects of the JCV-specific CRISPR/Cas9 editing apparatus. These observations provide the first evidence for the employment of a gene editing strategy as a promising tool for the elimination of the JCV genome and a potential cure for PML.


Subject(s)
CRISPR-Cas Systems , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Antigens, Viral, Tumor/genetics , Base Sequence , Cell Line, Tumor , Gene Expression , Gene Knockdown Techniques , Gene Targeting , Genetic Therapy/methods , Genome, Viral , Humans , Leukoencephalopathy, Progressive Multifocal/therapy , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , RNA Editing , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Sequence Alignment , Virus Replication
15.
Cell Cycle ; 14(13): 2075-9, 2015.
Article in English | MEDLINE | ID: mdl-26017382

ABSTRACT

Endoplasmic reticulum (ER) stress is caused by the accumulation of misfolded or unfolded proteins in the lumen of the endoplasmic reticulum. CCAAT/enhancer binding proteins are one of the cellular proteins whose expression is upregulated during ER stress. Previously, we have identified C/EBPbeta isoforms, especially LIP, as a negative regulator of polyomavirus JC (JCV), the causative agent of the demyelinating disease progressive multifocal leukoencephalopathy (PML). Here, we show that the induction of ER stress by thapsigargin increase the expression of endogenous LIP and the degradation of JCV T-antigen in a JCV-transgenic mouse tumor cell line. Our results also revealed that overexpression of LIP significantly reduced the level of T-Ag and this effect is reversed upon siRNA-mediated silencing of LIP. Immunoprecipitation/Western blot experiments indicated that LIP interacts with T-antigen directly. Treatment of cells that overexpress LIP with MG115, a proteasome inhibitor, partially rescued LIP-mediated degradation of T-antigen. Our observations point to a role of LIP in ER stress regulation of T-antigen stability and may open a new avenue to study host-virus interaction during ER stress.


Subject(s)
Antigens, Viral, Tumor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carrier Proteins/metabolism , Endoplasmic Reticulum Stress/physiology , JC Virus/metabolism , Steroid Isomerases/metabolism , Animals , Antigens, Viral, Tumor/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Carrier Proteins/immunology , Cell Line, Tumor , JC Virus/immunology , Mice , Mice, Transgenic , Protein Isoforms/immunology , Protein Isoforms/metabolism , Steroid Isomerases/immunology
16.
J Neurovirol ; 21(6): 679-87, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25791343

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease caused by neurotropic polyomavirus, JC virus (JCV), a virus that causes lytic infection of CNS glial cells. After primary infection, JCV is controlled by the immune system but virus persists asymptomatically. Rarely, when immune function is impaired, it can reemerge to cause PML. The mechanisms of JCV persistence and reactivation are not well understood but our earlier work implicated epigenetic control by protein acetylation since histone deacetylase inhibitors such as trichostatin A (TSA) strongly stimulate JCV transcription. Since both TNF-α and TSA activate JCV transcription via the same unique NF-κB site in the JCV control region, we investigated a role for acetylation of NF-κB in JCV regulation. A site-directed mutagenesis strategy was employed targeting the known lysine acetylation sites of NF-κB p65: K218, K221, and K310. We individually mutated each lysine to arginine, which cannot be acetylated and retains a positive charge like lysine. K218R and K221R impaired transactivation of JCV early promoter transcription either alone or combined with TSA treatment or coexpression of acetyltransferase transcriptional coactivator p300 but K310R was largely without effect. Mutation of lysine to glutamine gives mutants with a negative charge like acetyllysine. However, K218Q and K221Q showed impaired activity and only K310Q showed enhanced transactivation. NF-κB acetylation can regulate several aspects of the process of activation including complex formation with IκB, translocation to the nucleus, and DNA binding and transcriptional activation. Cell fractionation studies revealed that the mutants had no defect in translocation to the nucleus whereas gel shift studies revealed reduced binding to the JCV NF-κB site. Thus, acetylation regulates NF-κB p65 activity toward JCV at the level of p65 binding to the JCV control region and activation of JCV transcription.


Subject(s)
Epigenesis, Genetic/genetics , JC Virus/genetics , Transcription Factor RelA/metabolism , Transcriptional Activation/genetics , Virus Activation/genetics , Acetylation , Blotting, Western , Cell Line, Tumor , Humans , Lysine/metabolism , Mutagenesis, Site-Directed , Oligodendroglia/virology , Transfection
17.
Ann Neurol ; 77(4): 560-70, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25623836

ABSTRACT

Many neurological diseases of the central nervous system (CNS) are underpinned by malfunctions of the immune system, including disorders involving opportunistic infections. Progressive multifocal leukoencephalopathy (PML) is a lethal CNS demyelinating disease caused by the human neurotropic polyomavirus JC (JCV) and is found almost exclusively in individuals with immune disruption, including patients with human immunodeficiency virus/acquired immunodeficiency syndrome, patients receiving therapeutic immunomodulatory monoclonal antibodies to treat conditions such as multiple sclerosis, and transplant recipients. Thus, the public health significance of this disease is high, because of the number of individuals constituting the at-risk population. The incidence of PML is very low, whereas seroprevalence for the virus is high, suggesting infection by the virus is very common, and so it is thought that the virus is restrained but it persists in an asymptomatic state that can only occasionally be disrupted to lead to viral reactivation and PML. When JCV actively replicates in oligodendrocytes and astrocytes of the CNS, it produces cytolysis, leading to formation of demyelinated lesions with devastating consequences. Defining the molecular nature of persistence and events leading to reactivation of the virus to cause PML has proved to be elusive. In this review, we examine the current state of knowledge of the JCV life cycle and mechanisms of pathogenesis. We will discuss the normal course of the JCV life cycle including transmission, primary infection, viremia, and establishment of asymptomatic persistence as well as pathogenic events including migration of the virus to the brain, reactivation from persistence, viral infection, and replication in the glial cells of the CNS and escape from immunosurveillance.


Subject(s)
JC Virus/metabolism , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/transmission , Animals , Humans , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/diagnosis , Seroepidemiologic Studies
18.
PLoS One ; 9(10): e110122, 2014.
Article in English | MEDLINE | ID: mdl-25310191

ABSTRACT

The human neurotropic polyomavirus JC (JCV) causes the fatal CNS demyelinating disease progressive multifocal leukoencephalopathy (PML). JCV infection is very common and after primary infection, the virus is able to persist in an asymptomatic state. Rarely, and usually only under conditions of immune impairment, JCV re-emerges to actively replicate in the astrocytes and oligodendrocytes of the brain causing PML. The regulatory events involved in the reactivation of active viral replication in PML are not well understood but previous studies have implicated the transcription factor NF-κB acting at a well-characterized site in the JCV noncoding control region (NCCR). NF-κB in turn is regulated in a number of ways including activation by cytokines such as TNF-α, interactions with other transcription factors and epigenetic events involving protein acetylation--all of which can regulate the transcriptional activity of JCV. Active JCV infection is marked by the occurrence of rapid and extensive DNA damage in the host cell and the induction of the expression of cellular proteins involved in DNA repair including Rad51, a major component of the homologous recombination-directed double-strand break DNA repair machinery. Here we show that increased Rad51 expression activates the JCV early promoter. This activation is co-operative with the stimulation caused by NF-κB p65, abrogated by mutation of the NF-κB binding site or siRNA to NFκB p65 and enhanced by the histone deacetylase inhibitor sodium butyrate. These data indicate that the induction of Rad51 resulting from infection with JCV acts through NF-κB via its binding site to stimulate JCV early transcription. We suggest that this provides a novel positive feedback mechanism to enhance viral gene expression during the early stage of JCV infection.


Subject(s)
JC Virus/genetics , Rad51 Recombinase/metabolism , Transcription, Genetic , Butyric Acid/pharmacology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Fractionation , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/pharmacology , DNA, Intergenic/genetics , Genes, Dominant , Genes, Reporter , Humans , I-kappa B Proteins/metabolism , JC Virus/drug effects , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
19.
PLoS One ; 9(5): e98304, 2014.
Article in English | MEDLINE | ID: mdl-24847939

ABSTRACT

Transcription from the HIV-1 promoter is controlled by a series of ubiquitous and inducible cellular proteins with the ability to enter the nucleus and interact with the promoter. A DNA sequence spanning nucleotides -120 to -80, which supports the association of the inducible NF-κB transcription factor, has received much attention. Here we demonstrate that the interplay between Rad51, a key regulator of the homologous recombination pathway of DNA repair and whose level is induced upon HIV-1 infection, with the NF-κB pathway, augments transcription of the viral promoter. Evidently, stimulation of the NF-κB pathway by PMA and/or TSA promotes association of Rad51 with the LTR DNA sequence and that the p65 subunit of NF-κB is important for this event. Our results also demonstrate that, similar to p65, Rad51 utilizes the NF-κB pathway to position itself in the nucleus as ectopic expression of an IκB mutant impedes its nuclear appearance and transcriptional activity upon the HIV-1 LTR. Treatment of peripheral blood mononuclear cells with small molecules that inhibit Rad51 activity results in greater than 50% decrease in the HIV-1 infection of cells. These observations provide evidence for the involvement of DNA repair factors in control of HIV-1 gene activation and offer a new avenue for the development of anti-viral therapeutics that affect viral gene transcription in latently infected cells.


Subject(s)
HIV-1/physiology , Rad51 Recombinase/metabolism , Transcription Factor RelA/metabolism , Astrocytes/cytology , Astrocytes/virology , Brain/embryology , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Regulation, Viral , HIV Infections/metabolism , HIV Long Terminal Repeat , Humans , Leukocytes, Mononuclear/cytology , Mutation , Promoter Regions, Genetic , Rad51 Recombinase/genetics , Recombination, Genetic , Transcriptional Activation
20.
Virol J ; 10: 264, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23971673

ABSTRACT

BACKGROUND: Polyomavirus JC (JCV) causes the CNS demyelinating disease progressive multifocal leukoencephalopathy (PML), which occurs almost exclusively in people with immune deficiencies, such as HIV-1/AIDS patients. JCV infection is very common and usually occurs early in life. After primary infection, virus is controlled by the immune system but, rarely when immune function is impaired, it can re-emerge and multiply in the astrocytes and oligodendrocytes in the brain and cause PML. Thus a central question in PML pathogenesis is the nature of the molecular mechanisms maintaining JCV in a latent state and then allowing reactivation. METHODS: Since transcription can be regulated by epigenetic mechanisms including DNA methylation and histone acetylation, we investigated their role in JCV regulation by employing inhibitors of epigenetic events. RESULTS: The histone deacetylase inhibitors trichostatin A (TSA) and sodium butyrate powerfully stimulated JCV early and late transcription while the DNA methylation inhibitor 5-azacytidine had no effect. Analysis of JCV mutants showed that this effect was mediated by the KB element of the JCV control region, which binds transcription factors NF-κB p65, NFAT4 and C/EBPß and mediates stimulation by TNF-α. Stimulation of transcription by p65 was additive with TSA as was cotransfection with transcriptional coactivators/acetyltransferase p300 whereas depletion of endogenous p65 by RNA interference inhibited the effect of TSA. EMSA with a KB oligonucleotide showed p65 expression, TNF-α stimulation or TSA treatment each caused a gel shift that was further shifted by antibody to p65. CONCLUSIONS: We conclude that JCV is regulated epigenetically by protein acetylation events and that these involve the NF-κB p65 binding site in the JCV control region.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Viral , JC Virus/genetics , Cell Line , Host-Pathogen Interactions , Humans , JC Virus/physiology , Virus Activation , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL