Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Viruses ; 13(3)2021 03 12.
Article in English | MEDLINE | ID: mdl-33809082

ABSTRACT

The use of Natalizumab in Multiple Sclerosis (MS) can cause the reactivation of the polyomavirus JC (JCPyV); this may result in the development of progressive multifocal leukoencephalopathy (PML), a rare and usually lethal disease. JCPyV infection is highly prevalent in worldwide population, but the detection of anti-JCPyV antibodies is not sufficient to identify JCPyV infection, as PML can develop even in patients with negative JCPyV serology. Better comprehension of the JCPyV biology could allow a better understanding of JCPyV infection and reactivation, possibly reducing the risk of developing PML. Here, we investigated whether JCPyV miR-J1-5p-a miRNA that down-regulates the early phase viral protein T-antigen and promotes viral latency-could be detected and quantified by digital droplet PCR (ddPCR) in urine of 25 Natalizumab-treated MS patients. A 24-month study was designed: baseline, before the first dose of Natalizumab, and after 1 (T1), 12 (T12) and 24 months (T24) of therapy. miR-J1-5p was detected in urine of 7/25 MS patients (28%); detection was possible in three cases at T24, in two cases at T12, in one case at T1 and T12, and in the last case at baseline and T1. Two of these patients were seronegative for JCPyV Ab, and viral DNA was never found in either urine or blood. To note, only in one case miR-J1-5p was detected before initiation of Natalizumab. These results suggest that the measurement of miR-J1-5p in urine, could be a biomarker to monitor JCPyV infection and to better identify the possible risk of developing PML in Natalizumab-treated MS patients.


Subject(s)
JC Virus/growth & development , MicroRNAs/urine , Multiple Sclerosis/drug therapy , Natalizumab/therapeutic use , Virus Activation/drug effects , Antibodies, Viral/blood , Antigens, Viral, Tumor/biosynthesis , Biomarkers/urine , Down-Regulation/genetics , Female , Humans , Immunologic Factors/therapeutic use , JC Virus/drug effects , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/virology , Male , MicroRNAs/genetics , Virus Latency/genetics
2.
Jpn J Infect Dis ; 74(1): 48-53, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-32741932

ABSTRACT

JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the central nervous system affecting immunocompromised patients. The study of PML-type JCPyV in vitro has been limited owing to the inefficient propagation of the virus in cultured cells. In this study, we carried out long-term culture of COS-7 cells (designated as COS-IMRb cells) transfected with PML-type M1-IMRb, an adapted viral DNA with a rearranged non-coding control region (NCCR). The JCPyV derived from COS-IMRb cells were characterized by analyzing the viral replication, amount of virus by hemagglutination (HA), production of viral protein 1 (VP1), and structure of the NCCR. HA assays indicated the presence of high amounts of PML-type JCPyV in COS-IMRb cells. Immunostaining showed only a small population of JCPyV carrying COS-IMRb cells to be VP1-positive. Sequencing analysis of the NCCR of JCPyV after long-term culture revealed that the NCCR of M1-IMRb was conserved in COS-IMRb cells without any point mutation. The JCPyV genomic DNA derived from a clone of COS-IMRb-3 cells was detected, via Southern blotting, as a single band of approximately 5.1 kbp without deletion. These findings suggest the potential of using COS-IMRb-3 cells as a useful tool for screening anti-JCPyV drugs.


Subject(s)
JC Virus/growth & development , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Virus Cultivation/methods , Animals , Blotting, Southern/methods , COS Cells , Chlorocebus aethiops , DNA Replication , DNA, Viral/isolation & purification , Hemagglutination , Humans , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Environ Pollut ; 241: 234-239, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29857306

ABSTRACT

BACKGROUND: Human polyomaviruses (HPyVs) asymptomatically infect the human population during childhood and establish latency in the host. Viral reactivation and urinary excretion can occur when the immune system is impaired. Exposure to particulate air pollution, including the PM10/PM2.5 components, is a public health problem and has been linked to several disorders. Studies assessing the relationship between PM10/PM2.5 exposure and viral replication are lacking. OBJECTIVES: To investigate the relationship between HPyVs viruria and PM10/PM2.5 exposures. METHODS: Individual environmental exposure was assessed in 50 healthy adult volunteers using a chemical transport model (CTM) with a municipality resolution for daily PM10 and monitoring stations data for daily PM2.5 exposures. For each subject, a urine sample was collected, and HPyVs (JCPyV, BKPyV, MCPyV, HPyV6, HPyV7 and HPyV9) loads were determined. Zero-inflated negative binomial (ZINB) regression was used to model the count data, as it contained excessive zeros. Covariates were chosen by stepwise selection. RESULTS: HPyVs DNA was detected in 54% (median:87.6*105 copies/ml) of the urine samples. JCPyV was the prevalent (48%, (median viral load:126*105 copies/ml). Considering the load of the most frequently measured HPyVs, JCPyV, in the count-part of the ZINB model, every unitary in PM measured 2 days before urine collection (PM Day -2) was associated with an increase in JCPyV load (PM10: +4.0%, p-value = 0.002; PM2.5: +3.6%, p-value = 0.005). In the zero-part, the significant predictor was the PM10 measured 5 days before urine collection (+3%, p-value = 0.03). CONCLUSIONS: The environmental levels of PM10/PM2.5 increase the JCPyV viruria. Our findings emphasize the need for studies assessing the influence of air pollution exposure on the risk of viral reactivation.


Subject(s)
Air Pollutants/toxicity , Air Pollution/statistics & numerical data , Environmental Exposure/statistics & numerical data , JC Virus/growth & development , Particulate Matter/toxicity , Polyomavirus Infections/epidemiology , Adult , Air Pollutants/analysis , Air Pollution/analysis , Cities/epidemiology , Humans , Particulate Matter/analysis , Polyomavirus/growth & development , Prevalence
12.
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
13.
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
14.
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
15.
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
16.
Virus Res ; 231: 139-147, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27826043

ABSTRACT

The etiological role of human papillomavirus (HPV) in anogenital tract and head and neck cancers is well established. However, only a low percentage of HPV-positive women develop cancer, indicating that HPV is necessary but not sufficient in carcinogenesis. Several biological and environmental cofactors have been implicated in the development of HPV-associated carcinoma that include immune status, hormonal changes, parity, dietary habits, tobacco usage, and co-infection with other sexually transmissible agents. Such cofactors likely contribute to HPV persistent infection through diverse mechanisms related to immune control, efficiency of HPV infection, and influences on tumor initiation and progression. Conversely, HPV co-infection with other factors may also harbor anti-tumor effects. Here, we review epidemiological and experimental studies investigating human immunodeficiency virus (HIV), herpes simplex virus (HSV) 1 and 2, human cytomegalovirus (HCMV), Epstein-Barr virus (EBV), BK virus (BKV), JC virus (JCV), and adeno-associated virus (AAV) as viral cofactors in or therapeutic factors against the development of genital and oral HPV-associated carcinomas.


Subject(s)
Anus Neoplasms/virology , Genital Neoplasms, Female/virology , Head and Neck Neoplasms/virology , Papillomaviridae/pathogenicity , Papillomavirus Infections/virology , Anus Neoplasms/genetics , Anus Neoplasms/immunology , Anus Neoplasms/pathology , BK Virus/genetics , BK Virus/growth & development , BK Virus/pathogenicity , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Coinfection , Cytomegalovirus/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/pathogenicity , Dependovirus/genetics , Dependovirus/growth & development , Dependovirus/pathogenicity , Female , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/immunology , Genital Neoplasms, Female/pathology , HIV/genetics , HIV/growth & development , HIV/pathogenicity , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/growth & development , Herpesvirus 2, Human/pathogenicity , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Humans , JC Virus/genetics , JC Virus/growth & development , JC Virus/pathogenicity , Papillomaviridae/genetics , Papillomaviridae/growth & development , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Protective Factors , Risk Factors
17.
Annu Rev Virol ; 3(1): 517-532, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27501263

ABSTRACT

Mammalian polyomaviruses are characterized by establishing persistent infections in healthy hosts and generally causing clinical disease only in hosts whose immune systems are compromised. Despite the fact that these viruses were discovered decades ago, our knowledge of the mechanisms that govern viral persistence and reactivation is limited. Whereas mouse polyomavirus has been studied in a fair amount of detail, our understanding of the human viruses in particular is mostly inferred from experiments aimed at addressing other questions. In this review, we summarize the state of our current knowledge, draw conclusions when possible, and suggest areas that are in need of further study.


Subject(s)
BK Virus/growth & development , JC Virus/growth & development , Polyomavirus Infections/virology , Simian virus 40/growth & development , Tumor Virus Infections/virology , Animals , BK Virus/genetics , BK Virus/immunology , DNA, Viral/genetics , Humans , JC Virus/genetics , JC Virus/immunology , Mice , Simian virus 40/genetics , Simian virus 40/immunology , Virus Replication/genetics
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL