Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurovirol ; 26(3): 347-357, 2020 06.
Article in English | MEDLINE | ID: mdl-32124265

ABSTRACT

The gliomagenesis remains not fully established and their etiological factors still remain obscure. Polyomaviruses were detected and involved in several human tumors. Their potential implication in gliomas has been not yet surveyed in Africa and Arab World. Herein, we investigated the prevalence of six polyomaviruses (SV40, JCPyV, BKPyV, MCPyV, KIPyV, and WUPyV) in 112 gliomas from Tunisian patients. The DNA sequences of polyomaviruses were examined by PCR assays. Viral infection was confirmed by DNA in situ hybridization (ISH) and/or immunohistochemistry (IHC). The relationships between polyomavirus infection and tumor features were evaluated. Specific SV40 Tag, viral regulatory, and VP1 regions were identified in 12 GBM (10.7%). DNA ISH targeting the whole SV40 genome and SV40 Tag IHC confirmed the PCR findings. Five gliomas yielded JCPyV positivity by PCR and DNA ISH (2.7%). However, no BKPyV, KIPyV, and WUPyV DNA sequences were identified in all samples. MCPyV DNA was identified in 30 gliomas (26.8%). For GBM samples, MCPyV was significantly related to patient age (p = 0.037), tumor recurrence (p = 0.024), and SV40 (p = 0.045) infection. No further significant association was identified with the remaining tumor features (p > 0.05) and patient survival (Log Rank, p > 0.05). Our study indicates the presence of SV40, JCPyV, and MCPyV DNA in Tunisian gliomas. Further investigations are required to more elucidate the potential involvement of polyomaviruses in these destructive malignancies.


Subject(s)
Brain Neoplasms/virology , Glioma/virology , JC Virus/genetics , Merkel cell polyomavirus/genetics , Neoplasm Recurrence, Local/virology , Polyomavirus Infections/virology , Simian virus 40/genetics , Adult , Age Factors , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Capsid Proteins/genetics , Capsid Proteins/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Female , Follow-Up Studies , Glioma/genetics , Glioma/mortality , Glioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization , JC Virus/growth & development , JC Virus/pathogenicity , Male , Merkel cell polyomavirus/growth & development , Merkel cell polyomavirus/pathogenicity , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Polyomavirus Infections/genetics , Polyomavirus Infections/mortality , Polyomavirus Infections/pathology , Simian virus 40/growth & development , Simian virus 40/pathogenicity , Survival Analysis , Viral Load
2.
J Neurovirol ; 26(3): 437-441, 2020 06.
Article in English | MEDLINE | ID: mdl-31807988

ABSTRACT

Idelalisib, a selective phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor, is a newly approved second-line drug for patients with chronic lymphocytic leukemia. Recent clinical trials have suggested a possible association between idelalisib treatment and development of progressive multifocal leukoencephalopathy (PML) due to John Cunningham virus (JCV) reactivation. Nevertheless, clinical course and radiological and pathological features of idelalisib-induced PML still need to be clarified. We provide here the first clinicopathological description of idelalisib-associated PML in a patient who developed epilepsia partialis continua (EPC) as the first manifestation of the disease. Since EPC could present without electroencephalogram alterations, it is crucial to recognize the clinical features of this epileptic condition. EPC is characterized by the presence of repetitive, irregular, clonic jerking, often associated with hemiparesis and involvement of distal rather than proximal muscle groups. Moreover, we highlight the importance of brain biopsy in selected cases when there is a high clinical suspicion of PML, despite negative JCV testing in the cerebrospinal fluid. The pathological finding of prominent inflammatory infiltrate observed here was consistent with a diagnosis of immune reconstitution inflammatory syndrome (IRIS). IRIS is often associated with PML as a paradoxical worsening of clinical symptoms due to an overreacting immune response, in the context of previous immunosuppression. The unprecedented pathologic observation of IRIS in idelalisib-associated PML provides further insights into the pathogenesis of this rare neurological side effect.


Subject(s)
Antineoplastic Agents/adverse effects , Epilepsia Partialis Continua/diagnosis , Immune Reconstitution Inflammatory Syndrome/diagnosis , JC Virus/drug effects , Leukoencephalopathy, Progressive Multifocal/diagnosis , Purines/adverse effects , Quinazolinones/adverse effects , Antineoplastic Agents/administration & dosage , Epilepsia Partialis Continua/pathology , Epilepsia Partialis Continua/virology , Female , Humans , Immune Reconstitution Inflammatory Syndrome/pathology , Immune Reconstitution Inflammatory Syndrome/virology , JC Virus/growth & development , JC Virus/pathogenicity , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Middle Aged , Purines/administration & dosage , Quinazolinones/administration & dosage , Virus Activation/drug effects
3.
J Neurovirol ; 26(3): 452-455, 2020 06.
Article in English | MEDLINE | ID: mdl-32394398

ABSTRACT

Although there are several case reports of progressive multifocal leukoencephalopathy (PML) in multiple myeloma (MM), there are few reports of cases associated with pomalidomide. Here, we report the case of a 69-year-old female who had received 41 cycles of pomalidomide and dexamethasone treatment for relapsed/refractory IgG-κ MM presented with right-hand weakness; she was diagnosed as pomalidomide-associated PML. Fluid-attenuated inversion recovery (FLAIR) on admission showed high signals in the bilateral front-parietal lobe white matter, with multiple punctate lesions in the vicinity of the main lesions. These punctate pattern findings on FLAIR were similar to that of natalizumab-associated PML. Susceptibility weighted imaging (SWI) showed hypointense rims within the cortex at unaffected sites, in the initial stages. Subsequently, the clinical manifestations deteriorated, and the FLAIR images showed new hyperintense white matter lesions at the sites where cortical SWI hypointense rims were detected on the initial MRI examination. Our patient's serial MRI findings suggest that cortical SWI hypointense rims appear prior to the visible demyelinating white matter lesions in patients with PML.


Subject(s)
Immunologic Factors/adverse effects , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Multiple Myeloma/diagnostic imaging , Neoplasm Recurrence, Local/diagnostic imaging , Thalidomide/analogs & derivatives , Aged , Clinical Deterioration , Dexamethasone/adverse effects , Female , Humans , JC Virus/growth & development , Leukoencephalopathy, Progressive Multifocal/etiology , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Magnetic Resonance Imaging , Multiple Myeloma/pathology , Multiple Myeloma/virology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/virology , Parietal Lobe/diagnostic imaging , Parietal Lobe/pathology , Parietal Lobe/virology , Thalidomide/adverse effects , White Matter/diagnostic imaging , White Matter/pathology , White Matter/virology
4.
J Neurovirol ; 26(6): 961-963, 2020 12.
Article in English | MEDLINE | ID: mdl-32910430

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease affecting the central nervous system as a result of reactivation of the John Cunningham (JC) polyomavirus and occurs almost exclusively in immunosuppressed individuals. The disease course of PML is variable but usually progressive and often fatal. Treatment is predominantly focused on immune restoration, although this is difficult to do outside of human immunodeficiency virus-associated PML. A recent case series demonstrated a potential role for programmed cell death protein 1 (PD-1) inhibitors, such as pembrolizumab, to contain and/or clear JC virus. Herein, we discuss the first reported Australian case of a 61-year-old female with PML secondary to chemoimmunotherapy demonstrating complete clearance of JC virus as well as clinical and radiological stabilisation following pembrolizumab treatment.


Subject(s)
Agammaglobulinemia/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Hypertension/drug therapy , Leukoencephalopathy, Progressive Multifocal/drug therapy , Lymphoma/drug therapy , Agammaglobulinemia/diagnostic imaging , Agammaglobulinemia/immunology , Agammaglobulinemia/virology , Brain/diagnostic imaging , Brain/drug effects , Brain/immunology , Brain/virology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Female , Humans , Hypertension/diagnostic imaging , Hypertension/immunology , Hypertension/virology , JC Virus/drug effects , JC Virus/growth & development , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/virology , Lymphocyte Activation/drug effects , Lymphoma/diagnostic imaging , Lymphoma/immunology , Lymphoma/virology , Magnetic Resonance Imaging , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
5.
Molecules ; 25(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854369

ABSTRACT

The arbocyclic nucleosides aristeromycin and neplanocin have been studied as a source for new antiviral agents. A convenient synthesis of C-5'-truncated 3-deaza-1',6'-isoneplanocin, which combines the features of antiviral candidates 5'-noraristeromycin and 3-deaza-1',6'-isoneplanocin is reported from (-)-cyclopentenone to give the two C-4' epimers of 5'-nor-3-deaza isoneplanocin. Antiviral assays showed activity against the JC virus (EC50 = 1.12 µM for (4'R)-8; EC50 = 59.14 µM for (4'S)-7) and inactivity of both compounds against several DNA and RNA viruses. Both compounds lacked cytotoxicity.


Subject(s)
Adenosine , Antiviral Agents , JC Virus/growth & development , RNA Viruses/growth & development , Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Adenosine/chemistry , Adenosine/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Humans
6.
J Neurovirol ; 25(1): 133-136, 2019 02.
Article in English | MEDLINE | ID: mdl-30414049

ABSTRACT

Long-term treatment of multiple sclerosis with natalizumab (NTZ) carries the risk of a devastating complication in the form of an encephalopathy caused by a reactivation of a latent John Cunningham virus infection (progressive multifocal leucoencephalopathy, PML). Early diagnosis is associated with considerably better prognosis. Quantitative EEG as an objective, rater-independent technique provides high sensitivity (88%) and specificity (82%) for the diagnosis of NTZ-PML. Combination of diagnostic modalities addressing static morphological (brain MRI) as well as functional (EEG) pathologic changes may improve risk management programmes.


Subject(s)
Electroencephalography/methods , Immunologic Factors/adverse effects , JC Virus/drug effects , Leukoencephalopathy, Progressive Multifocal/diagnosis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/adverse effects , Adult , Female , Humans , Immunologic Factors/administration & dosage , JC Virus/growth & development , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/chemically induced , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/pathology , Natalizumab/administration & dosage , Prognosis , Retrospective Studies , Virus Activation/drug effects
7.
J Neurovirol ; 25(2): 284-287, 2019 04.
Article in English | MEDLINE | ID: mdl-30864100

ABSTRACT

Therapy for progressive multifocal leukoencephalopathy (PML) remains challenging since there are no antiviral therapies available for JC virus. Immune reconstitution has improved the prognosis in many settings where PML occurs, but it often is not possible in PML patients with hematologic malignancies. We describe the first biopsy proven PML case where the PD-1 inhibitor nivolumab appears to have stimulated immune activation resulting in effective control of PML in a patient with hematologic malignancy. This report supports further investigation of the utility of checkpoint inhibitors for treating PML where other immune reconstitution options are not available.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hodgkin Disease/drug therapy , Immune Reconstitution Inflammatory Syndrome/chemically induced , Leukoencephalopathy, Progressive Multifocal/drug therapy , Nivolumab/therapeutic use , Aged , Biopsy , Female , Gene Expression , Hodgkin Disease/diagnostic imaging , Hodgkin Disease/immunology , Hodgkin Disease/virology , Humans , Immune Reconstitution Inflammatory Syndrome/immunology , Immune Reconstitution Inflammatory Syndrome/virology , JC Virus/drug effects , JC Virus/growth & development , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/virology , Magnetic Resonance Imaging , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism
8.
J Cell Physiol ; 233(3): 2343-2359, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28722139

ABSTRACT

Agnoprotein (Agno) is an important regulatory protein of JC virus (JCV), BK virus (BKV) and simian virus 40 (SV40) and these viruses are unable to replicate efficiently in the absence of this protein. Recent 3D-NMR structural data revealed that Agno contains two alpha-helices (a minor and a major) while the rest of the protein adopts an unstructured conformation (Coric et al., 2017, J Cell Biochem). Previously, release of the JCV Agno from the Agno-positive cells was reported. Here, we have further mapped the regions of Agno responsible for its release by a structure-based systematic mutagenesis approach. Results revealed that amino acid residues (Lys22, Lys23, Phe31, Glu34, and Asp38) located either on or adjacent to the hydrophilic surface of the major alpha-helix domain of Agno play critical roles in release. Additionally, Agno was shown to strongly interact with unidentified components of the cell surface when cells are treated with Agno, suggesting additional novel roles for Agno during the viral infection cycle.


Subject(s)
JC Virus/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , JC Virus/genetics , JC Virus/growth & development , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Sequence Deletion , Structure-Activity Relationship , Surface Properties , Transfection , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics
9.
Microbiol Immunol ; 62(8): 524-530, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29932215

ABSTRACT

JC polyomavirus (JCPyV) is the causative agent of progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the central nervous system in immunocompromised patients. Archetype JCPyV circulates in the human population. There have been several reports of archetype JCPyV replication in cultured cells, in which propagation was not enough to produce high titers of archetype JCPyV. In this study, we carried out cultivation of the transfected cells with archetype JCPyV DNA MY for more than 2 months to establish COS-7 cells (designated COS-JC cells) persistently producing archetype JCPyV. Moreover, JCPyV derived from COS-JC cells was characterized by analyzing the viral propagation, size of the viral genome, amount of viral DNA, production of viral protein, and structure of the non-coding control region (NCCR). Southern blotting using a digoxigenin-labeled JCPyV probe showed two different sizes of the JCPyV genome in COS-JC cells. For molecular cloning, four of five clones showed a decrease in the size of complete JCPyV genome. Especially, clone No. 10 was generated the large deletion within the Large T antigen. On the other hand, the archetype structure of the NCCR was maintained in COS-JC cells, although a few point mutations occurred. Quantitative PCR analysis of viral DNA in COS-JC cells indicated that a high copy number of archetype JCPyV DNA was replicated in COS-JC cells. These findings suggest that COS-JC cells could efficiently propagate archetype JCPyV MY and offer a useful tool to study persistent infection of archetype JCPyV in a kidney-derived system.


Subject(s)
JC Virus/growth & development , JC Virus/genetics , Transfection , Virus Cultivation , Virus Replication/genetics , Animals , Antigens, Viral, Tumor/genetics , Base Sequence , COS Cells , Capsid Proteins/genetics , Capsid Proteins/isolation & purification , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA Replication , DNA, Viral/genetics , DNA, Viral/isolation & purification , Genome, Viral , Humans , Leukoencephalopathy, Progressive Multifocal/virology , Point Mutation , Viral Load , Viral Proteins/genetics , Viral Proteins/metabolism
10.
J Neurovirol ; 23(2): 226-238, 2017 04.
Article in English | MEDLINE | ID: mdl-27812788

ABSTRACT

Natalizumab is effective against multiple sclerosis (MS), but is associated with progressive multifocal leukoencephalopathy (PML), fatal disease caused by the JCV polyomavirus. The SF2/ASF (splicing factor2/alternative splicing factor) inhibits JCV in glial cells. We wondered about SF2/ASF modulation in the blood of natalizumab-treated patients and if this could influence JCV reactivation. Therefore, we performed a longitudinal study of MS patients under natalizumab, in comparison to patients under fingolimod and to healthy blood donors. Blood samples were collected at time intervals. The expression of SF2/ASF and the presence and expression of JCV in PBMC were analyzed. A bell-shaped regulation of SF2/ASF was observed in patients treated with natalizumab, increased in the first year of therapy, and reduced in the second one, while slightly changed, if any, in patients under fingolimod. Notably, SF2/ASF was up-regulated, during the first year, only in JCV DNA-positive patients, or with high anti-JCV antibody response; the expression of the JCV T-Ag protein in circulating B cells was inversely related to SF2/ASF protein expression. The SF2/ASF reduction, parallel to JCV activation, during the second year of therapy with natalizumab, but not with fingolimod, may help explain the increased risk of PML after the second year of treatment with natalizumab, but not with fingolimod. We propose that SF2/ASF has a protective role against JCV reactivation in MS patients. This study suggests new markers of disease behavior and, possibly, help in re-evaluations of therapy protocols.


Subject(s)
Host-Pathogen Interactions , Immunologic Factors/therapeutic use , Leukoencephalopathy, Progressive Multifocal/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/therapeutic use , Serine-Arginine Splicing Factors/genetics , Adult , Antibodies, Viral/blood , Dose-Response Relationship, Drug , Female , Fingolimod Hydrochloride/therapeutic use , Gene Expression Regulation , Humans , JC Virus/drug effects , JC Virus/genetics , JC Virus/growth & development , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/virology , Longitudinal Studies , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/virology , Neuroglia/drug effects , Neuroglia/immunology , Neuroglia/virology , Serine-Arginine Splicing Factors/immunology , Signal Transduction , Virus Activation/drug effects
11.
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
12.
Arch Virol ; 162(12): 3745-3752, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28884263

ABSTRACT

John Cunningham virus (JCPyV) is an ubiquitous human pathogen that causes disease in immunocompromised patients. The JCPyV genome is composed of an early region and a late region, which are physically separated by the non-coding control region (NCCR). The DNA sequence of the NCCR distinguishes two forms of JCPyV, the designated archetype and the prototype, which resulted from a rearrangement of the archetype sequence. To date, the cell culture systems for propagating JCPyV archetype have been very limited in their availability and robustness. Prior to this study, it was demonstrated that JCPyV archetype DNA replicates in COS-7 simian kidney cells expressing SV40 TAg and COS-7 cells expressing HIV-1 Tat. Based on these observations, the present study was conducted to reproduce an in vitro model in COS-7 cells transfected with the JCPyV archetype strain in order to study JCPyV DNA replication and analyze NCCR rearrangements during the viral life cycle. The efficiency of JCPyV replication was evaluated by quantitative PCR (Q-PCR) and by hemagglutination (HA) assay after transfection. In parallel, sequence analysis of JCPyV NCCR was performed. JCPyV efficiently replicated in kidney-derived COS-7 cells, as demonstrated by a progressive increase in viral load and virion particle production after transfection. The archetypal structure of NCCR was maintained during the viral cycle, but two characteristic point mutations were detected 28 days after transfection. This model is a useful tool for analyzing NCCR rearrangements during in vitro replication in cells that are sites of viral persistence, such as tubular epithelial cells of the kidney.


Subject(s)
Adaptation, Biological , Gene Rearrangement , JC Virus/growth & development , JC Virus/genetics , Animals , COS Cells , Chlorocebus aethiops , Hemagglutination Tests , Humans , Point Mutation , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Transfection , Virus Cultivation
13.
Clin Infect Dis ; 62(8): 986-94, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26743090

ABSTRACT

BACKGROUND: Progressive multifocal leukoencephalopathy (PML) is a rare, severe, otherwise fatal viral infection of the white matter of the brain caused by the polyomavirus JC virus, which typically occurs only in immunocompromised patients. One patient with dominant gain-of-function (GOF) mutation in signal transducer and activator of transcription 1 (STAT1) with chronic mucocutaneous candidiasis and PML was reported previously. We aim to identify the molecular defect in 3 patients with PML and to review the literature on PML in primary immune defects (PIDs). METHODS: STAT1 was sequenced in 3 patients with PML. U3C cell lines were transfected with STAT1 and assays to search for STAT1 phosphorylation, transcriptional response, and target gene expression were performed. RESULTS: We identified 3 new unrelated cases of PML in patients with GOF STAT1 mutations, including the novel STAT1 mutation, L400Q. These STAT1 mutations caused delayed STAT1 dephosphorylation and enhanced interferon-gamma-driven responses. In our review of the literature regarding PML in primary immune deficiencies we found 26 cases, only 54% of which were molecularly characterized, the remainder being syndromically diagnosed only. CONCLUSIONS: The occurrence of PML in 4 cases of STAT1 GOF suggests that STAT1 plays a critical role in the control of JC virus in the central nervous system.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Mutation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , Adult , Brain/diagnostic imaging , Cell Line, Tumor , Female , Gene Expression Regulation , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/diagnostic imaging , Interferon-gamma/pharmacology , JC Virus/growth & development , Leukoencephalopathy, Progressive Multifocal/complications , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/immunology , Male , Middle Aged , Sequence Analysis, DNA , Transcriptional Activation , Viral Load , Young Adult
14.
J Gen Virol ; 97(7): 1597-1603, 2016 07.
Article in English | MEDLINE | ID: mdl-27100458

ABSTRACT

The non-coding control region (NCCR) of polyomaviruses includes the promoters for early and late genes, a transcription enhancer and the origin of DNA replication. Particularly virulent variants of the human pathogens BKPyV and JCPyV, as well as of simian virus 40 (SV40), occur in vitro and in vivo. These strains often harbour rearrangements in their NCCR, typically deletions of some DNA segment(s) and/or duplications of others. Using an SV40-based model system we provide evidence that duplications of enhancer elements, whether from SV40 itself or from the related BKPyV and JCPyV, increase early gene transcription and replicative capacity. SV40 harbouring subsegments of the strong cytomegalovirus (HCMV) enhancer replicated better than the common 'wild-type' SV40 in the human cell lines HEK293 and U2OS. In conclusion, replacing the SV40 enhancer with heterologous enhancers can profoundly influence SV40's infective capacity, underscoring the potential of small DNA viruses to overcome cell type and species barriers.


Subject(s)
BK Virus/genetics , DNA, Viral/genetics , Enhancer Elements, Genetic/genetics , JC Virus/genetics , Simian virus 40/genetics , Viral Tropism/genetics , Animals , BK Virus/growth & development , BK Virus/physiology , Base Sequence , Cell Line , Chlorocebus aethiops , Cytomegalovirus/genetics , DNA Replication/genetics , HEK293 Cells , Hep G2 Cells , Humans , JC Virus/growth & development , JC Virus/physiology , Mice , Promoter Regions, Genetic/genetics , Simian virus 40/growth & development , Simian virus 40/physiology , Transcription, Genetic/genetics , Viral Tropism/physiology
15.
J Virol ; 89(7): 3910-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25609820

ABSTRACT

UNLABELLED: JC polyomavirus (JCPyV) infection of immunocompromised individuals results in the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML). The viral capsid of JCPyV is composed primarily of the major capsid protein virus protein 1 (VP1), and pentameric arrangement of VP1 monomers results in the formation of a pore at the 5-fold axis of symmetry. While the presence of this pore is conserved among polyomaviruses, its functional role in infection or assembly is unknown. Here, we investigate the role of the 5-fold pore in assembly and infection of JCPyV by generating a panel of mutant viruses containing amino acid substitutions of the residues lining this pore. Multicycle growth assays demonstrated that the fitness of all mutants was reduced compared to that of the wild-type virus. Bacterial expression of VP1 pentamers containing substitutions to residues lining the 5-fold pore did not affect pentamer assembly or prevent association with the VP2 minor capsid protein. The X-ray crystal structures of selected pore mutants contained subtle changes to the 5-fold pore, and no other changes to VP1 were observed. Pore mutant pseudoviruses were not deficient in assembly, packaging of the minor capsid proteins, or binding to cells or in transport to the host cell endoplasmic reticulum. Instead, these mutant viruses were unable to expose VP2 upon arrival to the endoplasmic reticulum, a step that is critical for infection. This study demonstrated that the 5-fold pore is an important structural feature of JCPyV and that minor modifications to this structure have significant impacts on infectious entry. IMPORTANCE: JCPyV is an important human pathogen that causes a severe neurological disease in immunocompromised individuals. While the high-resolution X-ray structure of the major capsid protein of JCPyV has been solved, the importance of a major structural feature of the capsid, the 5-fold pore, remains poorly understood. This pore is conserved across polyomaviruses and suggests either that these viruses have limited structural plasticity in this region or that this pore is important in infection or assembly. Using a structure-guided mutational approach, we showed that modulation of this pore severely inhibits JCPyV infection. These mutants do not appear deficient in assembly or early steps in infectious entry and are instead reduced in their ability to expose a minor capsid protein in the host cell endoplasmic reticulum. Our work demonstrates that the 5-fold pore is an important structural feature for JCPyV.


Subject(s)
Capsid Proteins/metabolism , Capsid/physiology , JC Virus/physiology , Protein Multimerization , Virus Assembly , Virus Internalization , Amino Acid Substitution , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Crystallography, X-Ray , Humans , JC Virus/chemistry , JC Virus/genetics , JC Virus/growth & development , Macromolecular Substances/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation
16.
J Neurovirol ; 22(4): 533-5, 2016 08.
Article in English | MEDLINE | ID: mdl-26843383

ABSTRACT

The risk algorithm for natalizumab-associated PML was first established in 2012 using the observations that JC virus antibody status, prolonged duration of natalizumab therapy (>2 years), and prior exposure to immunosuppressive therapy increased the risk for the disease. Prior to the publication of Biogen's algorithm, a risk algorithm was created by Fox and Rudick using an Excel spreadsheet in order to address the concerns of their patients. Applying the most recently available data regarding natalizumab-associated PML, the risk assessments for PML were recalculated. The current numbers indicate substantially higher risks for PML in 2015 than in 2012. Our calculations suggest that an individual having all three risk factors has an approximately 1 in 44 chance of developing PML.


Subject(s)
JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/diagnosis , Natalizumab/adverse effects , Seroconversion/drug effects , Algorithms , Humans , JC Virus/drug effects , JC Virus/growth & development , Leukoencephalopathy, Progressive Multifocal/chemically induced , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/virology , Natalizumab/administration & dosage , Risk Assessment , Risk Factors
17.
J Neurovirol ; 22(5): 597-606, 2016 10.
Article in English | MEDLINE | ID: mdl-26951564

ABSTRACT

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.


Subject(s)
Culture Media, Conditioned/pharmacology , Host-Pathogen Interactions , JC Virus/drug effects , Neuroglia/drug effects , Serine-Arginine Splicing Factors/genetics , Virus Replication/drug effects , Fetus , Gene Expression Regulation , Humans , JC Virus/genetics , JC Virus/growth & development , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Neuroglia/cytology , Neuroglia/immunology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serine-Arginine Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/immunology , Signal Transduction , Transcription, Genetic/drug effects
18.
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
19.
J Pept Sci ; 21(3): 236-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522925

ABSTRACT

The JC polyomavirus (JCPyV) infects approximately 50% of the human population. In healthy individuals, the infection remains dormant and asymptomatic, but in immuno-suppressed patients, it can cause progressive multifocal leukoencephalopathy (PML), a potentially fatal demyelinating disease. Currently, there are no drugs against JCPyV infection nor for the treatment of PML. Here, we report the development of small-molecule inhibitors of JCPyV that target the initial interaction between the virus and host cell and thereby block viral entry. Utilizing a combination of computational and NMR-based screening techniques, we target the LSTc tetrasaccharide binding site within the VP1 pentameric coat protein of JCPyV. Four of the compounds from the screen effectively block viral infection in our in vitro assays using SVG-A cells. For the most potent compound, we used saturation transfer difference NMR to determine the mode of binding to purified pentamers of JCPyV VP1. Collectively, these results demonstrate the viability of this class of compounds for eventual development of JCPyV-antiviral therapeutics.


Subject(s)
Antiviral Agents/chemistry , Capsid Proteins/antagonists & inhibitors , JC Virus/drug effects , Small Molecule Libraries/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/chemical synthesis , Binding Sites , Biological Assay , COS Cells , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line, Transformed , Chlorocebus aethiops , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , JC Virus/growth & development , JC Virus/metabolism , Molecular Docking Simulation , Neuroglia/drug effects , Neuroglia/virology , Protein Binding/drug effects , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
20.
J Gen Virol ; 93(Pt 3): 651-661, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22071512

ABSTRACT

JC virus (JCV) is the aetiological agent of the demyelinating disease progressive multifocal leukoencephalopathy, an AIDS defining illness and serious complication of mAb therapies. Initial infection probably occurs in childhood. In the working model of dissemination, virus persists in the kidney and lymphoid tissues until immune suppression/modulation causes reactivation and trafficking to the brain where JCV replicates in oligodendrocytes. JCV infection is regulated through binding of host factors such as Spi-B to, and sequence variation in the non-coding control region (NCCR). Although NCCR sequences differ between sites of persistence and pathogenesis, evidence suggests that the virus that initiates infection in the brain disseminates via B-cells derived from latently infected haematopoietic precursors in the bone marrow. Spi-B binds adjacent to TATA boxes in the promoter/enhancer of the PML-associated JCV Mad-1 and Mad-4 viruses but not the non-pathogenic, kidney-associated archetype. The Spi-B-binding site of Mad-1/Mad-4 differs from that of archetype by a single nucleotide, AAAAGGGAAGGGA to AAAAGGGAAGGTA. Point mutation of the Mad-1 Spi-B site reduced early viral protein large T-antigen expression by up to fourfold. Strikingly, the reverse mutation in the archetype NCCR increased large T-antigen expression by 10-fold. Interestingly, Spi-B protein binds the NCCR sequence flanking the viral promoter/enhancer, but these sites are not essential for early viral gene expression. The effect of mutating Spi-B-binding sites within the JCV promoter/enhancer on early viral gene expression strongly suggests a role for Spi-B binding to the viral promoter/enhancer in the activation of early viral gene expression.


Subject(s)
Astrocytes/virology , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Viral , JC Virus/growth & development , Promoter Regions, Genetic , TATA Box , Transcription Factors/metabolism , Binding Sites , Cells, Cultured , Humans , JC Virus/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL