Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 228(7): 829-833, 2023 10 03.
Article in English | MEDLINE | ID: mdl-36988117

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a severe neurological condition caused by reactivation of JC polyomavirus (JCPyV) in immunosuppression. Asymptomatic JCPyV persists in peripheral tissues. Upon reactivation, neurotropic rearrangements may emerge, and the virus gains access to the brain. To assess the mechanisms of PML pathogenesis, brain tissue material from PML patients was collected for small RNA sequencing. Upregulation of 8 microRNAs (miRNAs) in PML brain was validated using quantitative microRNA polymerase chain reaction (PCR). Bioinformatics tools were utilized to identify major associations of the upregulated miRNAs: neuroinflammation and blood-brain barrier disruption. The results indicate involvement of human miRNA regulation in PML pathogenesis.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , MicroRNAs , Humans , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/pathology , JC Virus/genetics , MicroRNAs/genetics , Brain/pathology , Base Sequence
2.
PLoS Pathog ; 16(4): e1008523, 2020 04.
Article in English | MEDLINE | ID: mdl-32324824

ABSTRACT

JC polyomavirus (JCPyV, JCV) causes progressive multifocal leukoencephalopathy (PML) in immunocompromised hosts. JCPyV replicates in oligodendrocytes within the brain tissue of patients with PML. The JCPyV genome encodes a microRNA (miRNA) in the region encoding the large T antigen. JCPyV-encoded miRNA (miR-J1) has been detected in the tissue and cerebrospinal fluid samples of patients with PML; however, there are no reports describing the localization of polyomavirus-encoded miRNA in histological samples of patients with virus-associated diseases. In the present study, we detected high miR-J1 expression in the nuclei of JCPyV-infected cells in PML tissue samples via in situ hybridization. Additionally, in situ hybridization also revealed the expression of BK polyomavirus (BKPyV, BKV)-encoded miRNA in lesions of BKPyV-associated nephropathy. In situ hybridization for miR-J1-5p and -3p showed positive signals in 24/25 (96%) of PML tissues that were positive for JCPyV by immunohistochemistry. Higher copy numbers of miR-J1 were detected in PML tissues than in non-PML tissues by real-time reverse transcription PCR. Next generation sequencing showed that miR-J1-5p, a mature miRNA of primary miRNA, was predominant in the lesions compared with miR-J1-3p, another mature miRNA. Deletion or mutation of miR-J1 in recombinant JCPyV promoted the production of JCPyV-encoded proteins in cells transfected with JCPyV DNA, suggesting that polyomavirus-encoded miRNA may have a repressive role in viral replication in PML tissues. In situ hybridization for viral miRNA may be a useful diagnostic tool for PML.


Subject(s)
JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Adult , Antigens, Viral, Tumor , BK Virus/genetics , DNA, Viral/genetics , Female , Gene Expression/genetics , Gene Expression Regulation, Viral/genetics , Genome , Humans , Male , MicroRNAs/genetics , Middle Aged , Oligodendroglia/metabolism , Polyomavirus/genetics , Polyomavirus Infections/genetics , RNA, Viral/genetics , Transfection , Viral Load , Virus Replication
3.
J Neurovirol ; 28(4-6): 616-618, 2022 12.
Article in English | MEDLINE | ID: mdl-35976539

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a rare and potentially fatal demyelinating disease of the central nervous system (CNS) caused by JC virus; it was previously seen predominantly in immunocompromised patients and those under intense immune suppression. Here, we report the case of a patient with PML with hypogammaglobulinemia and a heterozygous mutation in the TCF3 gene. As the TCF3 gene has been demonstrated to play an important role in the B cell differentiation process and the patient had no other medical history of the immune system, he was diagnosed with common variable immunodeficiency (CVID). To our knowledge, this is the first case of patient with a TCF3 gene deficiency and hypogammaglobulinemia who developed PML.


Subject(s)
Agammaglobulinemia , Common Variable Immunodeficiency , JC Virus , Leukoencephalopathy, Progressive Multifocal , Male , Humans , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/genetics , Agammaglobulinemia/complications , Agammaglobulinemia/genetics , JC Virus/genetics , Mutation , Basic Helix-Loop-Helix Transcription Factors/genetics
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(1): 78-82, 2021 Jan 10.
Article in Zh | MEDLINE | ID: mdl-33423265

ABSTRACT

OBJECTIVE: To detect variant of the CD40L gene and infection of Jamestown Canyon virus (JCV) in a 7-year-and-9-month-old boy with co-commitment progressive multifocal leukoencephalopathy (PML) and X-linked hyper IgM syndrome (XHIGM). METHODS: Peripheral blood samples of the child and his parents were collected for the extraction of genomic DNA. The 5 exons and exon/intronic boundaries of the CD40L gene were subjected to PCR amplification and sequencing. Suspected variants were analyzed by using bioinformatic software. The JCV gene was amplified from genomic DNA by nested PCR and sequenced. RESULTS: The child was found to harbor a hemizygous c.506 A>C (p.Y169S) missense variant in exon 5 of the CD40L gene. The variant may affect the TNFH domain of the CD40L protein and result in structural instability and loss of hydrophobic interaction between CD40L and CD40. As predicted by PolyPhen2 and SIFT software, the variant was probably damaging (score = 1.00) and deleterious (score= -8.868). His mother was found to be a heterozygous carrier, while the same variant was not found in his father. Gel electrophoresis of the nested PCR product revealed presence of target JCV band, which was confirmed to be 99% identical with the JCV gene by sequencing. CONCLUSION: The patient was diagnosed with co-commitment XHIGM and PML based on the testing of the CD40L gene and JCV infection.


Subject(s)
CD40 Ligand , Hyper-IgM Immunodeficiency Syndrome, Type 1 , Leukoencephalopathy, Progressive Multifocal , Adult , CD40 Ligand/genetics , Child , Exons/genetics , Female , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Male , Mutation, Missense , Polymerase Chain Reaction
5.
J Cell Physiol ; 234(6): 8295-8315, 2019 06.
Article in English | MEDLINE | ID: mdl-30390301

ABSTRACT

Polyomavirus family consists of a highly diverse group of small DNA viruses. The founding family member (MPyV) was first discovered in the newborn mouse in the late 1950s, which induces solid tumors in a wide variety of tissue types that are the epithelial and mesenchymal origin. Later, other family members were also isolated from a number of mammalian, avian and fish species. Some of these viruses significantly contributed to our current understanding of the fundamentals of modern biology such as transcription, replication, splicing, RNA editing, and cell transformation. After the discovery of first two human polyomaviruses (JC virus [JCV] and BK virus [BKV]) in the early 1970s, there has been a rapid expansion in the number of human polyomaviruses in recent years due to the availability of the new technologies and brought the present number to 14. Some of the human polyomaviruses cause considerably serious human diseases, including progressive multifocal leukoencephalopathy, polyomavirus-associated nephropathy, Merkel cell carcinoma, and trichodysplasia spinulosa. Emerging evidence suggests that the expression of the polyomavirus genome is more complex than previously thought. In addition to encoding universally expressed regulatory and structural proteins (LT-Ag, Sm t-Ag, VP1, VP2, and VP3), some polyomaviruses express additional virus-specific regulatory proteins and microRNAs. This review summarizes the recent advances in polyomavirus genome expression with respect to the new viral proteins and microRNAs other than the universally expressed ones. In addition, a special emphasis is devoted to the recent structural and functional discoveries in the field of polyomavirus agnoprotein which is expressed only by JCV, BKV, and simian virus 40 genomes.


Subject(s)
Genome, Viral/genetics , MicroRNAs/genetics , Polyomavirus/genetics , Viral Regulatory and Accessory Proteins/genetics , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/virology , DNA, Viral/genetics , Gene Expression Regulation, Viral/genetics , Humans , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Polyomavirus/pathogenicity , Virus Replication/genetics
6.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29321332

ABSTRACT

The human JC polyomavirus (JCPyV) infects the majority of the population worldwide and presents as an asymptomatic, persistent infection in the kidneys. In individuals who are immunocompromised, JCPyV can become reactivated and cause a lytic infection in the central nervous system resulting in the fatal, demyelinating disease progressive multifocal leukoencephalopathy (PML). Infection is initiated by interactions between the capsid protein viral protein 1 (VP1) and the α2,6-linked sialic acid on lactoseries tetrasaccharide c (LSTc), while JCPyV internalization is facilitated by 5-hydroxytryptamine 2 receptors (5-HT2Rs). The mechanisms by which the serotonin receptors mediate virus entry and the signaling cascades required to drive viral infection remain poorly understood. JCPyV was previously shown to induce phosphorylation of extracellular signal-regulated kinase (ERK), a downstream target of the mitogen-activated protein kinase (MAPK) pathway, upon virus entry. However, it remained unclear whether ERK activation was required for JCPyV infection. Both ERK-specific small interfering RNA (siRNA) and ERK inhibitor treatments resulted in significantly diminished JCPyV infection in both kidney and glial cells yet had no effect on the infectivity of the polyomavirus simian virus 40 (SV40). Experiments characterizing the role of ERK during steps in the viral life cycle indicate that ERK activation is required for viral transcription, as demonstrated by a significant reduction in production of large T antigen (TAg), a key viral protein associated with the initiation of viral transcription and viral replication. These findings delineate the role of the MAPK-ERK signaling pathway in JCPyV infection, elucidating how the virus reprograms the host cell to promote viral pathogenesis.IMPORTANCE Viral infection is dependent upon host cell factors, including the activation of cellular signaling pathways. These interactions between viruses and host cells are necessary for infection and play an important role in viral disease outcomes. The focus of this study was to determine how the human JC polyomavirus (JCPyV), a virus that resides in the kidney of the majority of the population and can cause the fatal, demyelinating disease progressive multifocal leukoencephalopathy (PML) in the brains of immunosuppressed individuals, usurps a cellular signaling pathway to promote its own infectious life cycle. We demonstrated that the activation of extracellular signal-regulated kinase (ERK), a component of the mitogen-activated protein kinase (MAPK) pathway, promotes JCPyV transcription, which is required for viral infection. Our findings demonstrate that the MAPK-ERK signaling pathway is a key determinant of JCPyV infection, elucidating new information regarding the signal reprogramming of host cells by a pathogenic virus.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , JC Virus/metabolism , Leukoencephalopathy, Progressive Multifocal/metabolism , MAP Kinase Signaling System , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/pathology
7.
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
8.
Clin Immunol ; 179: 1-7, 2017 06.
Article in English | MEDLINE | ID: mdl-28216420

ABSTRACT

Here we describe novel mutations in recombination activation gene 1 (RAG1) in a compound heterozygous male patient with combined T and B cell immunodeficiency (CID). Clinical manifestations besides antibody deficiency included airway infections, granulomatosis and autoimmune features. He died at the age of 37 due to PML caused by JC virus infection. By targeted next-generation sequencing we detected post mortem in this patient three mutations in RAG1. One allele harbored two novel mutations (c.1123C>G, p.H375D and c.1430delC, p.F478Sfs*14), namely a missense variant and a frameshift deletion, of which the latter leads to a truncated RAG1 protein. The other allele revealed a previously described missense mutation (c.1420C>T, p.R474C, rs199474678). Functional analysis of the p.R474C variant in an in vitro V(D)J recombination assay exhibited reduced recombination activity compared to a wild-type control. Our findings suggest that mutations in RAG1, specifically the p.R474C variant, can be associated with relatively mild clinical symptoms or delayed occurrence of T cell and B cell deficiencies but may predispose to PML.


Subject(s)
Homeodomain Proteins/genetics , Immunologic Deficiency Syndromes/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Adult , B-Lymphocytes/immunology , Biopsy , Brain/diagnostic imaging , Brain/pathology , Cell Proliferation , Humans , Immunoglobulins/blood , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/diagnostic imaging , Immunologic Deficiency Syndromes/immunology , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/immunology , Lymphocyte Count , Magnetic Resonance Imaging , Male , Mutation , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , V(D)J Recombination
9.
Brain ; 139(Pt 3): 782-94, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26912632

ABSTRACT

This study focused on the molecular characterization of patients with leukoencephalopathy associated with a specific biochemical defect of mitochondrial respiratory chain complex III, and explores the impact of a distinct magnetic resonance imaging pattern of leukoencephalopathy to detect biallelic mutations in LYRM7 in patients with biochemically unclassified leukoencephalopathy. 'Targeted resequencing' of a custom panel including genes coding for mitochondrial proteins was performed in patients with complex III deficiency without a molecular genetic diagnosis. Based on brain magnetic resonance imaging findings in these patients, we selected additional patients from a database of unclassified leukoencephalopathies who were scanned for mutations in LYRM7 by Sanger sequencing. Targeted sequencing revealed homozygous mutations in LYRM7, encoding mitochondrial LYR motif-containing protein 7, in four patients from three unrelated families who had a leukoencephalopathy and complex III deficiency. Two subjects harboured previously unreported variants predicted to be damaging, while two siblings carried an already reported pathogenic homozygous missense change. Sanger sequencing performed in the second cohort of patients revealed LYRM7 mutations in three additional patients, who were selected on the basis of the magnetic resonance imaging pattern. All patients had a consistent magnetic resonance imaging pattern of progressive signal abnormalities with multifocal small cavitations in the periventricular and deep cerebral white matter. Early motor development was delayed in half of the patients. All patients but one presented with subacute neurological deterioration in infancy or childhood, preceded by a febrile infection, and most patients had repeated episodes of subacute encephalopathy with motor regression, irritability and stupor or coma resulting in major handicap or death. LYRM7 protein was strongly reduced in available samples from patients; decreased complex III holocomplex was observed in fibroblasts from a patient carrying a splice site variant; functional studies in yeast confirmed the pathogenicity of two novel mutations. Mutations in LYRM7 were previously found in a single patient with a severe form of infantile onset encephalopathy. We provide new molecular, clinical, and neuroimaging data allowing us to characterize more accurately the molecular spectrum of LYRM7 mutations highlighting that a distinct and recognizable magnetic resonance imaging pattern is related to mutations in this gene. Inter- and intrafamilial variability exists and we observed one patient who was asymptomatic by the age of 6 years.


Subject(s)
Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/genetics , Magnetic Resonance Imaging , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , Mutation/genetics , Adolescent , Amino Acid Sequence , Child , Child, Preschool , Female , Humans , Infant , Magnetic Resonance Imaging/methods , Male , Molecular Sequence Data , Saccharomyces cerevisiae
10.
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
11.
J Neurovirol ; 22(6): 736-746, 2016 12.
Article in English | MEDLINE | ID: mdl-27170332

ABSTRACT

Estimating the individual risk for the development of progressive multifocal leukoencephalopathy (PML) in anti-John Cunningham virus (JCV) antibody-negative patients with multiple sclerosis (MS) treated with natalizumab is a major challenge. A serological conversion occurring under treatment from anti-JCV antibody-negative to positive status may significantly increase this risk. We investigated changes in peripheral blood cells' gene expression induced by natalizumab treatment in anti-JCV antibody-negative MS patients and tested blood transcriptional profile that characterizes patients predisposed to antibody switch under natalizumab treatment. After 3 years of natalizumab treatment, 24.6 % of anti-JCV antibody-negative MS patients switched to become anti-JCV antibody-positive (JCV switchers). Natalizumab induced 946 and 1186 significantly differentiating genes in JCV switchers and non-switchers, respectively. In JCV switchers, the signature was enriched by over-expression of genes associated with the first stages of viral entry to host cells including macropinocytosis (p = 1.82E-06), virus entry via endocytosis (p = 1.60E-06), clathrin-mediated endocytosis (p = 1.13E-04), and caveolar-mediated endocytosis (p = 4.50E-04) pathways. Further analysis to identify pre-existing transcriptional differences that characterize future JCV switchers prior to treatment initiation also demonstrated a transcriptional signature enriched by similar viral entry mechanisms. These findings, verified in an additional independent cohort of natalizumab-treated patients, could lead to future identification of patients that remain anti-JCV antibody-negative thus allowing safe continuation of treatment, as well as the development of future targeted therapeutic interventions to reduce the risk of PML.


Subject(s)
Host-Pathogen Interactions , Immunologic Factors/therapeutic use , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/immunology , Multiple Sclerosis/immunology , Natalizumab/therapeutic use , Seroconversion/drug effects , Adult , Antibodies, Viral/biosynthesis , Endocytosis/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Integrins/genetics , Integrins/immunology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/etiology , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Male , Multiple Sclerosis/complications , Multiple Sclerosis/genetics , Multiple Sclerosis/virology , Pinocytosis/drug effects , Prospective Studies , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Risk Factors , Seroconversion/genetics , Signal Transduction , Virus Internalization/drug effects
12.
J Cell Physiol ; 230(12): 2869-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26041694

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a severe demyelinating disease of the CNS caused by the human polyomavirus JC (JCV). JCV replication occurs only in human cells and investigation of PML has been severely hampered by the lack of an animal model. The common feature of PML is impairment of the immune system. The key to understanding PML is working out the complex mechanisms that underlie viral entry and replication within the CNS and the immunosurveillance that suppresses the virus or allows it to reactivate. Early models involved the simple inoculation of JCV into animals such as monkeys, hamsters, and mice. More recently, mouse models transgenic for the gene encoding the JCV early protein, T-antigen, a protein thought to be involved in the disruption of myelin seen in PML, have been employed. These animal models resulted in tumorigenesis rather than demyelination. Another approach is to use animal polyomaviruses that are closely related to JCV but able to replicate in the animal such as mouse polyomavirus and SV40. More recently, novel models have been developed that involve the engraftment of human cells into the animal. Here, we review progress that has been made to establish an animal model for PML, the advances and limitations of different models and weigh future prospects.


Subject(s)
Brain/virology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Animals , Animals, Genetically Modified , Brain/immunology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Host-Pathogen Interactions , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Myelin Sheath/metabolism , Phenotype , Species Specificity , Virus Replication
13.
J Clin Immunol ; 35(1): 92-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25388448

ABSTRACT

PURPOSE: A 30 year-old man with a history of recurrent skin infections as well as elevated serum IgE and eosinophils developed neurological symptoms and had T2-hyperintense lesions observed in cerebral MRI. The immune symptoms were attributed to Hyper IgE syndrome (HIES) and the neurological symptoms with presence of JC virus in cerebrospinal fluid were diagnosed as Progressive Multifocal Leukoencephalopathy (PML). The patient was negative for STAT3 mutations. To determine if other mutations explain HIES and/or PML in this subject, his DNA was analyzed by whole genome sequencing. METHODS: Whole genome sequencing was completed to 30X coverage, and whole genome SNP typing was used to complement these data. The methods revealed single nucleotide variants, structural variants, and copy number variants across the genome. Genome-wide data were analyzed for homozygous or compound heterozygous null mutations for all protein coding genes. Mutations were confirmed by PCR and/or Sanger sequencing. RESULTS: Whole genome analysis revealed deletions near the telomere of both copies of chromosome 9p. Several genes, including DOCK8, were impacted by the deletions but it was unclear whether each chromosome had identical or distinct deletions. PCR across the impacted region combined with Sanger sequencing of selected fragments confirmed a homozygous deletion from position 10,211 to 586,751. CONCLUSION: While several genes are impacted by the deletion, DOCK8 deficiency is the most probable cause of HIES in this patient. DOCK8 deficiency may have also predisposed the patient to develop PML.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Job Syndrome/genetics , Job Syndrome/immunology , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/immunology , Adult , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/immunology , DNA Mutational Analysis , Gene Deletion , Homozygote , Humans , Male , Telomere/genetics
14.
J Virol ; 88(9): 5177-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24554653

ABSTRACT

Progressive multifocal leukoencephalopathy (PML)-derived noncoding control region (NCCR) sequences permitted greater early viral gene expression than kidney-associated NCCR sequences. This was driven in part by binding of the transcription factor Spi-B to unique PML-associated Spi-B binding sites. Spi-B is upregulated in developing B cells in response to natalizumab therapy, a known risk factor for PML. Naturally occurring JCV sequence variation, together with drug treatment-induced cellular changes, may synergize to create an environment leading to an increased risk of PML.


Subject(s)
Gene Expression , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Lymphocytes/immunology , Regulatory Sequences, Nucleic Acid , DNA-Binding Proteins/biosynthesis , Genetic Association Studies , Humans , Molecular Sequence Data , Risk Assessment , Sequence Analysis, DNA , Transcription Factors/biosynthesis
15.
J Virol ; 87(24): 13490-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089568

ABSTRACT

The human JC polyomavirus (JCPyV) causes the rapidly progressing demyelinating disease progressive multifocal leukoencephalopathy (PML). The disease occurs most often in individuals with AIDS but also occurs in individuals receiving immunomodulatory therapies for immune-related diseases such as multiple sclerosis. JCPyV infection of host cells requires the pentasaccharide lactoseries tetrasaccharide c (LSTc) and the serotonin receptor 5-hydroxytryptamine (5-HT) receptor 5-HT2AR. While LSTc is involved in the initial attachment of virus to cells via interactions with VP1, the mechanism by which 5-HT2AR contributes to infection is not clear. To further define the roles of serotonin receptors in infection, HEK293A cells, which are poorly permissive to JCPyV, were transfected with 14 different isoforms of serotonin receptor. Only 5-HT2 receptors were found to support infection by JCPyV. None of the other 11 isoforms of serotonin receptor supported JCPyV infection. Expression of 5-HT2 receptors did not increase binding of JCPyV to cells, but this was not unexpected, given that the cells uniformly expressed the major attachment receptor, LSTc. Infection of these cells remained sensitive to inhibition with soluble LSTc, confirming that LSTc recognition is required for JCPyV infection. Virus internalization into HEK293A cells was significantly and specifically enhanced when 5HT2 receptors were expressed. Taken together, these data confirm that the carbohydrate LSTc is the attachment receptor for JCPyV and that the type 2 serotonin receptors contribute to JCPyV infection by facilitating entry.


Subject(s)
JC Virus/physiology , Leukoencephalopathy, Progressive Multifocal/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Virus Internalization , HEK293 Cells , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2B/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin/metabolism
16.
Mult Scler ; 20(14): 1851-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24852919

ABSTRACT

BACKGROUND: Natalizumab has shown its efficacy in reducing multiple sclerosis (MS) relapses and progression of disability; however, it has been associated with an increased risk of developing progressive multifocal leukoencephalopathy (PML). The differential expression of microRNA (miRNA), the small non-coding RNAs that regulate gene expression, in natalizumab-treated patients has been reported and miRNA have also been described as good candidates for disease biomarkers. OBJECTIVE: To characterize the effect of natalizumab therapy on the miRNA expression pattern and to search for miRNAs that can predict PML on an individual basis. METHODS: The expression of 754 microRNAs was measured in blood samples from 19 relapsing-remitting MS patients at three time points during natalizumab therapy, using TaqMan OpenArray panels. Two patients included in this study developed PML after more than 2 years of therapy. RESULTS: We found that the expression level of three miRNAs (let-7c, miR-125a-5p and miR-642) was affected after 6 months of therapy (t6). Furthermore, we observed a differential expression of another three miRNAs (miR-320, miR-320b and miR-629) between the PML and non-PML groups after 12 months of treatment (t12); and a positive correlation was found between therapy time and the expression of miR-320. CONCLUSIONS: Natalizumab modified the expression levels of three miRNAs after a 6-month treatment. We suggest miR-320, miR-320b and miR-629 as possible biomarkers for individual PML risk assessment.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Leukoencephalopathy, Progressive Multifocal/genetics , MicroRNAs/genetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adult , Case-Control Studies , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/chemically induced , Male , MicroRNAs/metabolism , Middle Aged , Natalizumab
17.
J Immunol ; 189(7): 3618-30, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942431

ABSTRACT

Virus-specific CD4(+) T cells play a central role in control of viral pathogens including JC polyoma virus (JCV) infection. JCV is a ubiquitous small DNA virus that leads to persistent infection of humans with no clinical consequences. However, under circumstances of immunocompromise, it is able to cause an opportunistic and often fatal infection of the brain called progressive multifocal leukoencephalopathy (PML). PML has emerged as a serious adverse event in multiple sclerosis patients treated with the anti-VLA-4 mAb natalizumab, which selectively inhibits cell migration across the blood-brain barrier and the gut's vascular endothelium thus compromising immune surveillance in the CNS and gut. In a multiple sclerosis patient who developed PML under natalizumab treatment and a vigorous immune response against JCV after Ab washout, we had the unique opportunity to characterize in detail JCV-specific CD4(+) T cell clones from the infected tissue during acute viral infection. The in-depth analysis of 14 brain-infiltrating, JCV-specific CD4(+) T cell clones demonstrated that these cells use an unexpectedly broad spectrum of different strategies to mount an efficient JCV-specific immune response including TCR bias, HLA cross-restriction that increases avidity and influences in vivo expansion, and a combination of Th1 and Th1-2 functional phenotypes. The level of combinatorial diversity in TCR- and HLA-peptide interactions used by brain-infiltrating, JCV-specific CD4(+) T cells has not, to our knowledge, been reported before in humans for other viral infections and confirms the exceptional plasticity that characterizes virus-specific immune responses.


Subject(s)
Brain/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Gene Rearrangement, T-Lymphocyte , HLA-D Antigens/metabolism , JC Virus/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Brain/metabolism , Brain/virology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Movement/genetics , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , HLA-D Antigens/genetics , Humans , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics
18.
Antiviral Res ; 222: 105817, 2024 02.
Article in English | MEDLINE | ID: mdl-38246207

ABSTRACT

JC polyomavirus (JCPyV) is a nonenveloped, double-stranded DNA virus that infects the majority of the population. Immunocompetent individuals harbor infection in their kidneys, while severe immunosuppression can result in JCPyV spread to the brain, causing the neurodegenerative disease progressive multifocal leukoencephalopathy (PML). Due to a lack of approved therapies to treat JCPyV and PML, the disease results in rapid deterioration, and is often fatal. In order to identify potential antiviral treatments for JCPyV, a high-throughput, large-scale drug screen was performed using the National Institutes of Health Clinical Collection (NCC). Drugs from the NCC were tested for inhibitory effects on JCPyV infection, and drugs from various classes that reduced JCPyV infection were identified, including receptor agonists and antagonists, calcium signaling modulators, and enzyme inhibitors. Given the role of calcium signaling in viral infection including Merkel cell polyomavirus and simian virus 40 polyomavirus (SV40), calcium signaling inhibitors were further explored for the capacity to impact JCPyV infection. Calcium and calmodulin inhibitors trifluoperazine (TFP), W-7, tetrandrine, and nifedipine reduced JCPyV infection, and TFP specifically reduced viral internalization. Additionally, TFP and W-7 reduced infection by BK polyomavirus, SV40, and SARS-CoV-2. These results highlight specific inhibitors, some FDA-approved, for the possible treatment and prevention of JCPyV and several other viruses, and further illuminate the calcium and calmodulin pathway as a potential target for antiviral drug development.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , Neurodegenerative Diseases , Polyomavirus Infections , Sulfonamides , Humans , Calcium , Calmodulin , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/genetics , JC Virus/genetics , Simian virus 40 , Antiviral Agents/pharmacology
19.
J Neurol Sci ; 461: 123046, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38761670

ABSTRACT

INTRODUCTION: Progressive multifocal leukoencephalopathy (PML) is a potentially life-threatening complication among Multiple Sclerosis (MS) patients under natalizumab treatment, with serum anti-JCV antibody titers being used for stratification risk. Given the critical role of interferon (IFN)/B-cell activating factor (BAFF) axis in humoral immune responses against viruses, we explored whether it is involved in the generation of serum anti-JCV antibodies among these patients. METHODS: 162 consecutive patients with relapsing-remitting MS under natalizumab treatment were included. Serum anti-JCV antibodies were measured at baseline, as well as 12 and 24 months after treatment initiation. Type I and II IFN-inducible genes and BAFF expression were quantitated in peripheral blood by qRT-PCR. Moreover, BAFF rs9514828, rs1041569, and rs9514827 gene variants were assessed by RFLP-PCR. RESULTS: While type I and II IFN inducible gene expression were not associated with anti-JCV serum titers, the latter were significantly correlated with BAFF gene expression. Of interest, the TTT haplotype of the studied BAFF variants was more frequently detected in male, but not female anti-JCV (+) MS patients compared to anti-JCV (-) counterparts at baseline, as well as at 12 months and 24 months of natalizumab treatment. Measures of clinical validity/utility for the BAFF TTT haplotype showed 88% specificity, 45%, positive predictive value, and sensitivity of 70% for the discrimination of anti-JCV (+) male MS patients after 24 months of treatment. CONCLUSIONS: Our study suggests an implication of the BAFF axis in the production of serum anti-JCV antibodies. Additionally, the BAFF TTT haplotype derived from the rs9514828, rs1041569, and rs9514827 variants may represent a novel risk factor for anti-JCV seropositivity and indirectly for PML development among male MS patients treated with natalizumab.


Subject(s)
B-Cell Activating Factor , Immunologic Factors , JC Virus , Leukoencephalopathy, Progressive Multifocal , Natalizumab , Humans , Natalizumab/therapeutic use , B-Cell Activating Factor/blood , B-Cell Activating Factor/genetics , Male , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/genetics , Adult , Female , Immunologic Factors/therapeutic use , JC Virus/immunology , JC Virus/genetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/immunology , Middle Aged , Antibodies, Viral/blood , Polymorphism, Single Nucleotide
20.
Nat Genet ; 3(3): 256-9, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8485581

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) has been recently reported as a cause of stroke. It is characterized, in the absence of hypertension, by recurrent subcortical ischaemic strokes, starting in early or midadulthood and leading in some patients to dementia. Magnetic resonance imaging and pathological examination show numerous small subcortical infarcts and a diffuse leukoencephalopathy underlaid by a non-arteriosclerotic, non-amyloid angiopathy. We performed genetic linkage analysis in two unrelated families and assigned the disease locus to chromosome 19q12. Multilocus analysis with the location scores method established the best estimate for the location of the affected gene within a 14 centimorgan interval bracketed by D19S221 and D19S222 loci.


Subject(s)
Cerebral Arterial Diseases/genetics , Cerebral Infarction/genetics , Chromosomes, Human, Pair 19 , Leukoencephalopathy, Progressive Multifocal/genetics , Adult , Brain/pathology , Cerebral Arterial Diseases/diagnosis , Cerebral Arterial Diseases/pathology , Cerebral Arteries/pathology , Cerebral Infarction/diagnosis , Cerebral Infarction/pathology , Chromosome Mapping , Female , Genes, Dominant , Genetic Linkage , Genetic Markers , Humans , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Pedigree , Polymerase Chain Reaction , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL