Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
J Neurol Sci ; 461: 123046, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38761670

RESUMEN

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.


Asunto(s)
Factor Activador de Células B , Factores Inmunológicos , Virus JC , Leucoencefalopatía Multifocal Progresiva , Natalizumab , Humanos , Natalizumab/uso terapéutico , Factor Activador de Células B/sangre , Factor Activador de Células B/genética , Masculino , Leucoencefalopatía Multifocal Progresiva/sangre , Leucoencefalopatía Multifocal Progresiva/genética , Adulto , Femenino , Factores Inmunológicos/uso terapéutico , Virus JC/inmunología , Virus JC/genética , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Persona de Mediana Edad , Anticuerpos Antivirales/sangre , Polimorfismo de Nucleótido Simple
2.
Antiviral Res ; 222: 105817, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38246207

RESUMEN

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.


Asunto(s)
Virus JC , Leucoencefalopatía Multifocal Progresiva , Enfermedades Neurodegenerativas , Infecciones por Polyomavirus , Sulfonamidas , Humanos , Calcio , Calmodulina , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Leucoencefalopatía Multifocal Progresiva/genética , Virus JC/genética , Virus 40 de los Simios , Antivirales/farmacología
3.
J Infect Dis ; 228(7): 829-833, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36988117

RESUMEN

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.


Asunto(s)
Virus JC , Leucoencefalopatía Multifocal Progresiva , MicroARNs , Humanos , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/patología , Virus JC/genética , MicroARNs/genética , Encéfalo/patología , Secuencia de Bases
4.
Expert Rev Mol Diagn ; 23(2): 143-157, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36786077

RESUMEN

INTRODUCTION: JC polyomavirus is the causative agent of progressive multifocal leukoencephalopathy (PML), a demyelinating disease resulting from the lytic infection of oligodendrocytes that may develop in immunosuppressed individuals: HIV1 infected or individuals under immunosuppressive therapies. Understanding the biology of JCPyV is necessary for a proper patient management, the development of diagnostic tests, and risk stratification. AREAS COVERED: The review covers different areas of expertise including the genomic characterization of JCPyV strains detected in different body compartments (urine, plasma, and cerebrospinal fluid) of PML patients, viral mutations, molecular diagnostics, viral miRNAs, and disease. EXPERT OPINION: The implementation of molecular biology techniques improved our understanding of JCPyV biology. Deep sequencing analysis of viral genomes revealed the presence of viral quasispecies in the cerebrospinal fluid of PML patients characterized by noncoding control region rearrangements and VP1 mutations. These neurotropic JCPyV variants present enhanced replication and an altered cell tropism that contribute to PML development. Monitoring these variants may be relevant for the identification of patients at risk of PML. Multiplex realtime PCR targeting both the LTAg and the archetype NCCR could be used to identify them. Failure to amplify NCCR should indicate the presence of a JCPyV prototype speeding up the diagnostic process.


Asunto(s)
Virus JC , Leucoencefalopatía Multifocal Progresiva , MicroARNs , Humanos , Biología , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/etiología , Leucoencefalopatía Multifocal Progresiva/genética , Mutación
5.
mBio ; 14(2): e0358322, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36786589

RESUMEN

JC polyomavirus (JCPyV) is a ubiquitous, double-stranded DNA virus that causes the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in immunocompromised patients. Current treatments for PML are limited to immune reconstitution, and no effective antivirals exist. In this report, we show that the oxindole GW-5074 (3-(3,5-dibromo-4-hydroxybenzylidene)-5-iodoindolin-2-one) reduces JCPyV infection in primary and immortalized cells. This compound potently inhibits virus spread, which suggests that it could control infection in PML patients. We demonstrate that GW-5074 inhibits endogenous ERK phosphorylation, and that JCPyV infection in GW-5074-treated cells cannot be rescued with ERK agonists, which indicates that the antiviral mechanism may involve its antagonistic effects on MAPK-ERK signaling. Importantly, GW-5074 exceeds thresholds of common pharmacological parameters that identify promising compounds for further development. This MAPK-ERK antagonist warrants further investigation as a potential treatment for PML. IMPORTANCE Human polyomaviruses, such as JCPyV and BKPyV, cause significant morbidity and mortality in immunocompromised or immunomodulated patients. There are no treatments for polyomavirus-induced diseases other than restoration of immune function. We discovered that the oxindole GW-5074 potently inhibits infection by both JCPyV and BKPyV. Further optimization of this compound could result in the development of antiviral therapies for polyomavirus-induced diseases.


Asunto(s)
Virus JC , Leucoencefalopatía Multifocal Progresiva , Infecciones por Polyomavirus , Poliomavirus , Humanos , Oxindoles/farmacología , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Leucoencefalopatía Multifocal Progresiva/genética , Virus JC/genética , Sistema de Señalización de MAP Quinasas , Antivirales
6.
J Neurovirol ; 28(4-6): 616-618, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35976539

RESUMEN

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.


Asunto(s)
Agammaglobulinemia , Inmunodeficiencia Variable Común , Virus JC , Leucoencefalopatía Multifocal Progresiva , Masculino , Humanos , Leucoencefalopatía Multifocal Progresiva/diagnóstico por imagen , Leucoencefalopatía Multifocal Progresiva/genética , Agammaglobulinemia/complicaciones , Agammaglobulinemia/genética , Virus JC/genética , Mutación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(1): 78-82, 2021 Jan 10.
Artículo en Chino | MEDLINE | ID: mdl-33423265

RESUMEN

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.


Asunto(s)
Ligando de CD40 , Síndrome de Inmunodeficiencia con Hiper-IgM Tipo 1 , Leucoencefalopatía Multifocal Progresiva , Adulto , Ligando de CD40/genética , Niño , Exones/genética , Femenino , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM Tipo 1/genética , Leucoencefalopatía Multifocal Progresiva/genética , Masculino , Mutación Missense , Reacción en Cadena de la Polimerasa
9.
Viruses ; 12(10)2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053912

RESUMEN

Polyomaviruses are ubiquitous human pathogens that cause lifelong, asymptomatic infections in healthy individuals. Although these viruses are restrained by an intact immune system, immunocompromised individuals are at risk for developing severe diseases driven by resurgent viral replication. In particular, loss of immune control over JC polyomavirus can lead to the development of the demyelinating brain disease progressive multifocal leukoencephalopathy (PML). Viral isolates from PML patients frequently carry point mutations in the major capsid protein, VP1, which mediates virion binding to cellular glycan receptors. Because polyomaviruses are non-enveloped, VP1 is also the target of the host's neutralizing antibody response. Thus, VP1 mutations could affect tropism and/or recognition by polyomavirus-specific antibodies. How these mutations predispose susceptible individuals to PML and other JCPyV-associated CNS diseases remains to be fully elucidated. Here, we review the current understanding of polyomavirus capsid mutations and their effects on viral tropism, immune evasion, and virulence.


Asunto(s)
Proteínas de la Cápside/genética , Evasión Inmune/genética , Virus JC/genética , Virus JC/inmunología , Leucoencefalopatía Multifocal Progresiva/inmunología , Animales , Cápside/inmunología , Proteínas de la Cápside/inmunología , Especificidad del Huésped/genética , Humanos , Evasión Inmune/inmunología , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/virología , Ratones , Mutación , Tropismo Viral/genética , Replicación Viral/genética
10.
PLoS Pathog ; 16(4): e1008523, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32324824

RESUMEN

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.


Asunto(s)
Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/genética , Adulto , Antígenos Virales de Tumores , Virus BK/genética , ADN Viral/genética , Femenino , Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Genoma , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Oligodendroglía/metabolismo , Poliomavirus/genética , Infecciones por Polyomavirus/genética , ARN Viral/genética , Transfección , Carga Viral , Replicación Viral
11.
Curr Opin Virol ; 40: 19-27, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32279025

RESUMEN

Progressive multifocal leukoencephalopathy (PML) is a frequently fatal brain infection caused by the JC polyomavirus (JCV). PML occurs in people with impaired cellular immunity, and the only effective treatment is restoration of immune function. Infection in immunocompromised hosts is often associated with immune exhaustion, which is mediated by inhibitory cell surface receptors known as immune checkpoints, leading to loss of T cell effector function. Blockade of immune checkpoints can reinvigorate host responses to fight infection. Recently, there have been several reports of checkpoint blockade to treat PML in patients in whom immune reconstitution is otherwise not possible, with some evidence for positive response. Larger studies are needed to better understand efficacy of checkpoint blockade in PML and factors that determine response.


Asunto(s)
Antivirales/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Proteínas de Punto de Control Inmunitario/inmunología , Virus JC/efectos de los fármacos , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Animales , Humanos , Proteínas de Punto de Control Inmunitario/genética , Virus JC/genética , Virus JC/inmunología , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/inmunología , Leucoencefalopatía Multifocal Progresiva/virología , Linfocitos T/inmunología
12.
J Cell Physiol ; 234(6): 8295-8315, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30390301

RESUMEN

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.


Asunto(s)
Genoma Viral/genética , MicroARNs/genética , Poliomavirus/genética , Proteínas Reguladoras y Accesorias Virales/genética , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/virología , ADN Viral/genética , Regulación Viral de la Expresión Génica/genética , Humanos , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/virología , Poliomavirus/patogenicidad , Replicación Viral/genética
13.
Front Immunol ; 9: 138, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29456537

RESUMEN

The increased use of newer potent immunomodulatory therapies for multiple sclerosis (MS), including natalizumab, fingolimod, and dimethyl fumarate, has expanded the patient population at risk for developing progressive multifocal leukoencephalopathy (PML). These MS therapies shift the profile of lymphocytes within the central nervous system (CNS) leading to increased anti-inflammatory subsets and decreased immunosurveillance. Similar to MS, PML is a demyelinating disease of the CNS, but it is caused by the JC virus. The manifestation of PML requires the presence of an active, genetically rearranged form of the JC virus within CNS glial cells, coupled with the loss of appropriate JC virus-specific immune responses. The reliability of metrics used to predict risk for PML could be improved if all three components, i.e., viral genetic strain, localization, and host immune function, were taken into account. Advances in our understanding of the critical lymphocyte subpopulation changes induced by these MS therapies and ability to detect viral mutation and reactivation will facilitate efforts to develop these metrics.


Asunto(s)
Inmunomodulación , Leucoencefalopatía Multifocal Progresiva/terapia , Esclerosis Múltiple/terapia , Animales , Humanos , Inmunosupresores/uso terapéutico , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/inmunología , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Riesgo
14.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29321332

RESUMEN

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.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Virus JC/metabolismo , Leucoencefalopatía Multifocal Progresiva/metabolismo , Sistema de Señalización de MAP Quinasas , Quinasas MAP Reguladas por Señal Extracelular/genética , Células HEK293 , Humanos , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/patología
15.
J Cell Physiol ; 233(5): 4137-4155, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29044559

RESUMEN

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.


Asunto(s)
Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/genética , Poliomavirus/genética , Trans-Empalme/genética , Encéfalo/virología , Codón de Terminación/genética , ADN Viral/clasificación , ADN Viral/genética , Exones/genética , Regulación Viral de la Expresión Génica , Genoma Viral/genética , Humanos , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/virología , Sistemas de Lectura Abierta , Poliomavirus/patogenicidad , Replicación Viral/genética
16.
Clin Immunol ; 179: 1-7, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28216420

RESUMEN

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.


Asunto(s)
Proteínas de Homeodominio/genética , Síndromes de Inmunodeficiencia/genética , Leucoencefalopatía Multifocal Progresiva/genética , Adulto , Linfocitos B/inmunología , Biopsia , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Proliferación Celular , Humanos , Inmunoglobulinas/sangre , Síndromes de Inmunodeficiencia/sangre , Síndromes de Inmunodeficiencia/diagnóstico por imagen , Síndromes de Inmunodeficiencia/inmunología , Leucoencefalopatía Multifocal Progresiva/sangre , Leucoencefalopatía Multifocal Progresiva/diagnóstico por imagen , Leucoencefalopatía Multifocal Progresiva/inmunología , Recuento de Linfocitos , Imagen por Resonancia Magnética , Masculino , Mutación , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Recombinación V(D)J
17.
J Neurovirol ; 22(6): 736-746, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27170332

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Factores Inmunológicos/uso terapéutico , Virus JC/inmunología , Leucoencefalopatía Multifocal Progresiva/inmunología , Esclerosis Múltiple/inmunología , Natalizumab/uso terapéutico , Seroconversión/efectos de los fármacos , Adulto , Anticuerpos Antivirales/biosíntesis , Endocitosis/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Integrinas/genética , Integrinas/inmunología , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/etiología , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/virología , Masculino , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/genética , Esclerosis Múltiple/virología , Pinocitosis/efectos de los fármacos , Estudios Prospectivos , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Factores de Riesgo , Seroconversión/genética , Transducción de Señal , Internalización del Virus/efectos de los fármacos
18.
Neurotherapeutics ; 13(3): 547-54, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27150390

RESUMEN

The study of neurological infections by viruses defines the field of neurovirology, which has emerged in the last 30 years and was founded upon the discovery of a number of viruses capable of infecting the human nervous system. Studies have focused on the molecular and biological basis of viral neurological diseases with the aim of revealing new therapeutic options. The first studies of neurovirological infections can be traced back to the discovery that some viruses have an affinity for the nervous system with research into rabies by Louis Pasteur and others in the 1880s. Today, the immense public health impact of neurovirological infections is illustrated by diseases such as neuroAIDS, progressive multifocal leukoencephalopathy, and viral encephalitis. Recent research has seen the development of powerful new techniques for gene editing that promise revolutionary opportunities for the development of novel therapeutic options. In particular, clustered regulatory interspaced short palindromic repeat-associated 9 system provides an effective, highly specific and versatile tool for targeting DNA viruses that are beginning to allow the development of such new approaches. In this short review, we discuss these recent developments, how they pertain to neurological infections, and future prospects.


Asunto(s)
Sistemas CRISPR-Cas , Enfermedades Virales del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/terapia , Edición Génica/métodos , Terapia Genética/métodos , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/terapia , Infecciones por VIH/genética , Infecciones por VIH/terapia , Herpesvirus Humano 1/genética , Humanos , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/terapia
19.
Brain ; 139(Pt 3): 782-94, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26912632

RESUMEN

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.


Asunto(s)
Leucoencefalopatía Multifocal Progresiva/diagnóstico , Leucoencefalopatía Multifocal Progresiva/genética , Imagen por Resonancia Magnética , Proteínas Mitocondriales/genética , Chaperonas Moleculares/genética , Mutación/genética , Adolescente , Secuencia de Aminoácidos , Niño , Preescolar , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética/métodos , Masculino , Datos de Secuencia Molecular , Saccharomyces cerevisiae
20.
Clin Infect Dis ; 62(8): 986-94, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26743090

RESUMEN

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.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Leucoencefalopatía Multifocal Progresiva/genética , Mutación , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/fisiología , Adulto , Encéfalo/diagnóstico por imagen , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/diagnóstico por imagen , Interferón gamma/farmacología , Virus JC/crecimiento & desarrollo , Leucoencefalopatía Multifocal Progresiva/complicaciones , Leucoencefalopatía Multifocal Progresiva/diagnóstico por imagen , Leucoencefalopatía Multifocal Progresiva/inmunología , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Activación Transcripcional , Carga Viral , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...