Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Nat Commun ; 15(1): 3969, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730242

RESUMEN

Encephalitis is a rare and potentially fatal manifestation of herpes simplex type 1 infection. Following genome-wide genetic analyses, we identified a previously uncharacterized and very rare heterozygous variant in the E3 ubiquitin ligase WWP2, in a 14-month-old girl with herpes simplex encephalitis. The p.R841H variant (NM_007014.4:c.2522G > A) impaired TLR3 mediated signaling in inducible pluripotent stem cells-derived neural precursor cells and neurons; cells bearing this mutation were also more susceptible to HSV-1 infection compared to control cells. The p.R841H variant increased TRIF ubiquitination in vitro. Antiviral immunity was rescued following the correction of p.R841H by CRISPR-Cas9 technology. Moreover, the introduction of p.R841H in wild type cells reduced such immunity, suggesting that this mutation is linked to the observed phenotypes.


Asunto(s)
Encefalitis por Herpes Simple , Herpesvirus Humano 1 , Mutación , Ubiquitina-Proteína Ligasas , Humanos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Femenino , Encefalitis por Herpes Simple/genética , Lactante , Herpesvirus Humano 1/genética , Células Madre Pluripotentes Inducidas/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Ubiquitinación , Neuronas/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/virología , Sistemas CRISPR-Cas
2.
Brain ; 146(10): 4306-4319, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37453099

RESUMEN

Patients with herpes simplex virus (HSV) encephalitis (HSE) often develop neuronal autoantibody-associated encephalitis (AE) post-infection. Risk factors of AE are unknown. We tested the hypotheses that predisposition for AE post-HSE may be involved, including genetic variants at specific loci, human leucocyte (HLA) haplotypes, or the blood innate immune response against HSV, including type I interferon (IFN) immunity. Patients of all ages with HSE diagnosed between 1 January 2014 and 31 December 2021 were included in one of two cohorts depending on whether the recruitment was at HSE onset (Spanish Cohort A) or by the time of new neurological manifestations (international Cohort B). Patients were assessed for the type of neurological syndromes; HLA haplotypes; blood type I-IFN signature [RNA quantification of 6 or 28 IFN-response genes (IRG)] and toll-like receptor (TLR3)-type I IFN-related gene mutations. Overall, 190 patients (52% male) were recruited, 93 in Cohort A and 97 in Cohort B. Thirty-nine (42%) patients from Cohort A developed neuronal autoantibodies, and 21 (54%) of them developed AE. Three syndromes (choreoathetosis, anti-NMDAR-like encephalitis and behavioural-psychiatric) showed a high (≥95% cases) association with neuronal autoantibodies. Patients who developed AE post-HSE were less likely to carry the allele HLA-A*02 (4/21, 19%) than those who did not develop AE (42/65, 65%, P = 0.0003) or the Spanish general population (2005/4335, 46%, P = 0.0145). Blood IFN signatures using 6 or 28 IRG were positive in 19/21 (91%) and 18/21 (86%) patients at HSE onset, and rapidly decreased during follow-up. At Day 21 after HSE onset, patients who later developed AE had higher median IFN signature compared with those who did not develop AE [median Zs-6-IRG 1.4 (0.6; 2.0) versus 0.2 (-0.4; 0.8), P = 0.03]. However, a very high median Zs-6-IRG (>4) or persistently increased IFN signature associated with uncontrolled viral infection. Whole exome sequencing showed that the percentage of TLR3-IFN-related mutations in patients who developed AE was not different from those who did not develop AE [3/37 (8%) versus 2/57 (4%), P = 0.379]. Multivariate logistic regression showed that a moderate increase of the blood IFN signature at Day 21 (median Zs-6-IRG >1.5 but <4) was the most important predictor of AE post-HSE [odds ratio 34.8, interquartile ratio (1.7-691.9)]. Altogether, these findings show that most AE post-HSE manifest with three distinct syndromes, and HLA-A*02, but not TLR3-IFN-related mutations, confer protection from developing AE. In addition to neuronal autoantibodies, the blood IFN signature in the context of HSE may be potentially useful for the diagnosis and monitoring of HSE complications.


Asunto(s)
Encefalitis por Herpes Simple , Interferón Tipo I , Enfermedades del Sistema Nervioso , Humanos , Masculino , Femenino , Encefalitis por Herpes Simple/complicaciones , Encefalitis por Herpes Simple/genética , Receptor Toll-Like 3/genética , Autoanticuerpos , Antígenos HLA-A
3.
Immunology ; 170(1): 83-104, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37278103

RESUMEN

Autosomal recessive (AR) and dominant (AD) deficiencies of TLR3 and TRIF are believed to be crucial genetic causes of herpes simplex encephalitis (HSE), which is a fatal disease causing focal or global cerebral dysfunction following infection with herpes simplex virus type 1 (HSV-1). However, few studies have been conducted on the immunopathological networks of HSE in the context of TLR3 and TRIF defects at the cellular and molecular levels. In this work, we deciphered the crosstalk between type I IFN (IFN-I)-producing epithelial layer and IL-15-producing dendritic cells (DC) to activate NK cells for the protective role of TLR3/TRIF pathway in HSE progression after vaginal HSV-1 infection. TLR3- and TRIF-ablated mice showed enhanced susceptibility to HSE progression, along with high HSV-1 burden in vaginal tract, lymphoid tissues and CNS. The increased HSV-1 burden in TLR3- and TRIF-ablated mice did not correlate with increased infiltration of Ly-6C+ monocytes, but it was closely associated with impaired NK cell activation in vaginal tract. Furthermore, using delicate ex vivo experiments and bone marrow transplantation, TRIF deficiency in tissue-resident cells, such as epithelial cells in vaginal tract, was found to cause impaired NK cell activation by means of low IFN-I production, whereas IFN-I receptor in DC was required for NK cell activation via IL-15 production in response to IFN-I produced from epithelial layer. These results provide new information about IFN-I- and IL-15-mediated crosstalk between epithelial cells and DC at the primary infection site, which suppresses HSE progression in a TLR3- and TRIF-dependent manner.


Asunto(s)
Encefalitis por Herpes Simple , Herpesvirus Humano 1 , Femenino , Animales , Ratones , Encefalitis por Herpes Simple/genética , Receptor Toll-Like 3/genética , Interleucina-15/genética , Células Dendríticas , Proteínas Adaptadoras del Transporte Vesicular/genética
4.
J Clin Invest ; 133(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37097753

RESUMEN

Neonatal herpes simplex virus (HSV) infection is a devastating disease with substantial morbidity and mortality. The genetic basis of susceptibility to HSV in neonates remains undefined. We evaluated a male infant with neonatal skin/eye/mouth (SEM) HSV-1 disease, who had complete recovery after acyclovir but developed HSV-1 encephalitis at 1 year of age. An immune workup showed an anergic PBMC cytokine response to TLR3 stimulation but no other TLRs. Exome sequencing identified rare missense variants in IFN-regulatory factor 7 (IRF7) and UNC-93 homolog B1 (UNC93B1). PBMC single-cell RNA-Seq done during childhood revealed decreased expression of several innate immune genes and a repressed TLR3 pathway signature at baseline in several immune cell populations, including CD14 monocytes. Functional studies in fibroblasts and human leukemia monocytic THP1 cells showed that both variants individually suppressed TLR3-driven IRF3 transcriptional activity and the type I IFN response in vitro. Furthermore, fibroblasts expressing the IRF7 and UNC93B1 variants had higher intracellular viral titers with blunting of the type I IFN response upon HSV-1 challenge. This study reports an infant with recurrent HSV-1 disease complicated by encephalitis associated with deleterious variants in the IRF7 and UNC93B1 genes. Our results suggest that TLR3 pathway mutations may predispose neonates to recurrent, severe HSV.


Asunto(s)
Encefalitis por Herpes Simple , Herpes Simple , Herpesvirus Humano 1 , Interferón Tipo I , Humanos , Lactante , Recién Nacido , Masculino , Encefalitis por Herpes Simple/genética , Herpes Simple/genética , Leucocitos Mononucleares/metabolismo , Proteínas de Transporte de Membrana , Receptor Toll-Like 3/genética
5.
Sci Immunol ; 8(82): eade2860, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37083451

RESUMEN

Inborn errors of TLR3-dependent type I IFN immunity in cortical neurons underlie forebrain herpes simplex virus-1 (HSV-1) encephalitis (HSE) due to uncontrolled viral growth and subsequent cell death. We report an otherwise healthy patient with HSE who was compound heterozygous for nonsense (R422*) and frameshift (P493fs9*) RIPK3 variants. Receptor-interacting protein kinase 3 (RIPK3) is a ubiquitous cytoplasmic kinase regulating cell death outcomes, including apoptosis and necroptosis. In vitro, the R422* and P493fs9* RIPK3 proteins impaired cellular apoptosis and necroptosis upon TLR3, TLR4, or TNFR1 stimulation and ZBP1/DAI-mediated necroptotic cell death after HSV-1 infection. The patient's fibroblasts displayed no detectable RIPK3 expression. After TNFR1 or TLR3 stimulation, the patient's cells did not undergo apoptosis or necroptosis. After HSV-1 infection, the cells supported excessive viral growth despite normal induction of antiviral IFN-ß and IFN-stimulated genes (ISGs). This phenotype was, nevertheless, rescued by application of exogenous type I IFN. The patient's human pluripotent stem cell (hPSC)-derived cortical neurons displayed impaired cell death and enhanced viral growth after HSV-1 infection, as did isogenic RIPK3-knockout hPSC-derived cortical neurons. Inherited RIPK3 deficiency therefore confers a predisposition to HSE by impairing the cell death-dependent control of HSV-1 in cortical neurons but not their production of or response to type I IFNs.


Asunto(s)
Encefalitis por Herpes Simple , Herpes Simple , Herpesvirus Humano 1 , Humanos , Muerte Celular , Encefalitis por Herpes Simple/genética , Herpesvirus Humano 1/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores Tipo I de Factores de Necrosis Tumoral , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
6.
Sci Immunol ; 7(77): eabq4531, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36399538

RESUMEN

Herpes simplex virus 1 (HSV-1) infects several billion people worldwide and can cause life-threatening herpes simplex encephalitis (HSE) in some patients. Monogenic defects in components of the type I interferon system have been identified in patients with HSE, emphasizing the role of inborn errors of immunity underlying HSE pathogenesis. Here, we identify compound heterozygous loss-of-function mutations in the gene GTF3A encoding for transcription factor IIIA (TFIIIA), a component of the RNA polymerase III complex, in a patient with common variable immunodeficiency and HSE. Patient fibroblasts and GTF3A gene-edited cells displayed impaired HSV-1-induced innate immune responses and enhanced HSV-1 replication. Chromatin immunoprecipitation sequencing analysis identified the 5S ribosomal RNA pseudogene 141 (RNA5SP141), an endogenous ligand of the RNA sensor RIG-I, as a transcriptional target of TFIIIA. GTF3A mutant cells exhibited diminished RNA5SP141 expression and abrogated RIG-I activation upon HSV-1 infection. Our work unveils a crucial role for TFIIIA in transcriptional regulation of a cellular RIG-I agonist and shows that GTF3A genetic defects lead to impaired cell-intrinsic anti-HSV-1 responses and can predispose to HSE.


Asunto(s)
Encefalitis por Herpes Simple , Herpesvirus Humano 1 , Humanos , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/patología , Seudogenes , ARN , Ligandos , Factor de Transcripción TFIIIA/genética , Herpesvirus Humano 1/genética , Mutación
7.
Ugeskr Laeger ; 184(16)2022 04 18.
Artículo en Danés | MEDLINE | ID: mdl-35485776

RESUMEN

Herpes simplex encephalitis is a devastating neurological disorder with a poor prognosis. For years, it remained elusive why a fraction of otherwise healthy individuals presented with the condition; this lack of insight has hampered understanding of disease pathogenesis and the development of novel effective therapies. However, recent studies have shown that the lack of viral containment can be caused by functionally related monogenic inborn errors of immunity at least in a subset of patients. This knowledge renders prophylactic measures and design of targeted therapies possible, as argued in this review.


Asunto(s)
Encefalitis por Herpes Simple , Encefalitis por Herpes Simple/genética , Humanos
8.
J Clin Immunol ; 42(3): 606-617, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35040013

RESUMEN

PURPOSE: Enterovirus A71 (EV71) causes a broad spectrum of childhood diseases, ranging from asymptomatic infection or self-limited hand-foot-and-mouth disease (HFMD) to life-threatening encephalitis. The molecular mechanisms underlying these different clinical presentations remain unknown. We hypothesized that EV71 encephalitis in children might reflect an intrinsic host single-gene defect of antiviral immunity. We searched for mutations in the toll-like receptor 3 (TLR3) gene. Such mutations have already been identified in children with herpes simplex virus encephalitis (HSE). METHODS: We sequenced TLR3 and assessed the impact of the mutations identified. We tested dermal fibroblasts from a patient with EV71 encephalitis and a TLR3 mutation and other patients with known genetic defects of TLR3 or related genes, assessing the response of these cells to TLR3 agonist poly(I:C) stimulation and EV71 infection. RESULTS: Three children with EV71 encephalitis were heterozygous for rare mutations-TLR3 W769X, E211K, and R867Q-all of which were shown to affect TLR3 function. Furthermore, fibroblasts from the patient heterozygous for the W769X mutation displayed an impaired, but not abolished, response to poly(I:C). We found that TLR3-deficient and TLR3-heterozygous W769X fibroblasts were highly susceptible to EV71 infection. CONCLUSIONS: Autosomal dominant TLR3 deficiency may underlie severe EV71 infection with encephalitis. Human TLR3 immunity is essential to protect the central nervous system against HSV-1 and EV71. Children with severe EV71 infections, such as encephalitis in particular, should be tested for inborn errors of TLR3 immunity.


Asunto(s)
Encefalitis por Herpes Simple , Encefalitis Viral , Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Células Cultivadas , Niño , Encefalitis por Herpes Simple/diagnóstico , Encefalitis por Herpes Simple/genética , Encefalitis Viral/diagnóstico , Encefalitis Viral/genética , Enterovirus Humano A/genética , Infecciones por Enterovirus/diagnóstico , Infecciones por Enterovirus/genética , Humanos , Poli I-C , Receptor Toll-Like 3/genética
9.
J Int Med Res ; 49(10): 3000605211049645, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34644193

RESUMEN

OBJECTIVES: Herpes simplex encephalitis (HSE) is one of the most common causes of severe viral encephalitis. The characteristic manifestations of HSE include cerebrospinal fluid with mild cytopenia, dominated by lymphocytes, elevated protein, and normal blood glucose values (3.9-6.1 mmol/L). Although it is not difficult to diagnose classical HSE, diagnosing clinically atypical cases is more difficult. METHODS: We reviewed the results of next-generation sequencing (NGS) of CSF in a series of patients diagnosed with atypical HSE. RESULTS: Four patients lacking classical clinical manifestations of HSE, including no fever, headache, or other typical neurological deficit symptoms, 1-2 × 106 cells/L CSF leucocyte count, and no typical imaging features, were diagnosed with atypical HSE by NGS of CSF. The NGS reads corresponding to herpes simplex virus type 1 ranged from 2 to 13,174. CONCLUSIONS: Mild HSE may not present with classic frontotemporal lobe syndrome and fever may not be an inevitable symptom in patients with immunosuppression. However, the possibility of HSE should be considered in patients with atypical intracranial infection, and these patients should be tested by NGS.


Asunto(s)
Encefalitis por Herpes Simple , Encefalitis por Herpes Simple/diagnóstico , Encefalitis por Herpes Simple/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Simplexvirus/genética
10.
Am J Hum Genet ; 108(6): 1012-1025, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34015270

RESUMEN

The human genetic dissection of clinical phenotypes is complicated by genetic heterogeneity. Gene burden approaches that detect genetic signals in case-control studies are underpowered in genetically heterogeneous cohorts. We therefore developed a genome-wide computational method, network-based heterogeneity clustering (NHC), to detect physiological homogeneity in the midst of genetic heterogeneity. Simulation studies showed our method to be capable of systematically converging genes in biological proximity on the background biological interaction network, and capturing gene clusters harboring presumably deleterious variants, in an efficient and unbiased manner. We applied NHC to whole-exome sequencing data from a cohort of 122 individuals with herpes simplex encephalitis (HSE), including 13 individuals with previously published monogenic inborn errors of TLR3-dependent IFN-α/ß immunity. The top gene cluster identified by our approach successfully detected and prioritized all causal variants of five TLR3 pathway genes in the 13 previously reported individuals. This approach also suggested candidate variants of three reported genes and four candidate genes from the same pathway in another ten previously unstudied individuals. TLR3 responsiveness was impaired in dermal fibroblasts from four of the five individuals tested, suggesting that the variants detected were causal for HSE. NHC is, therefore, an effective and unbiased approach for unraveling genetic heterogeneity by detecting physiological homogeneity.


Asunto(s)
Biología Computacional/métodos , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/patología , Fibroblastos/inmunología , Redes Reguladoras de Genes , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Estudios de Casos y Controles , Encefalitis por Herpes Simple/inmunología , Fibroblastos/metabolismo , Humanos , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 3/metabolismo , Secuenciación del Exoma
11.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32960813

RESUMEN

Inborn errors of TLR3-dependent IFN-α/ß- and IFN-λ-mediated immunity in the CNS can underlie herpes simplex virus 1 (HSV-1) encephalitis (HSE). The respective contributions of IFN-α/ß and IFN-λ are unknown. We report a child homozygous for a genomic deletion of the entire coding sequence and part of the 3'-UTR of the last exon of IFNAR1, who died of HSE at the age of 2 years. An older cousin died following vaccination against measles, mumps, and rubella at 12 months of age, and another 17-year-old cousin homozygous for the same variant has had other, less severe, viral illnesses. The encoded IFNAR1 protein is expressed on the cell surface but is truncated and cannot interact with the tyrosine kinase TYK2. The patient's fibroblasts and EBV-B cells did not respond to IFN-α2b or IFN-ß, in terms of STAT1, STAT2, and STAT3 phosphorylation or the genome-wide induction of IFN-stimulated genes. The patient's fibroblasts were susceptible to viruses, including HSV-1, even in the presence of exogenous IFN-α2b or IFN-ß. HSE is therefore a consequence of inherited complete IFNAR1 deficiency. This viral disease occurred in natural conditions, unlike those previously reported in other patients with IFNAR1 or IFNAR2 deficiency. This experiment of nature indicates that IFN-α/ß are essential for anti-HSV-1 immunity in the CNS.


Asunto(s)
Encefalitis por Herpes Simple , Herpesvirus Humano 1/metabolismo , Receptor de Interferón alfa y beta/deficiencia , Adolescente , Preescolar , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/metabolismo , Encefalitis por Herpes Simple/patología , Células HEK293 , Herpesvirus Humano 1/genética , Humanos , Interferones/genética , Interferones/metabolismo , Masculino , Receptor de Interferón alfa y beta/metabolismo
13.
Nitric Oxide ; 108: 8-11, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347982

RESUMEN

In this study, a hypothesis that genetic variations in neuronal nitric oxide synthase (nNOS) could influence the susceptibility and outcome of herpes simplex encephalitis was investigated. Polymorphic loci of nNOS gene, G84A and C276T were genotyped in 132 HSE cases (Age 8.2 ± 1.3yr) and 143 in healthy individuals (Age-9.2 ± 1.6yr) of the same ethnic background from Odisha. A significantly increased risk for HSVE was associated with the AG genotype (OR = 1.73, 95%CI = 1.03-2.9, P = 0.03) and AA genotype (OR = 2.96, 95%CI = 1.04-8.4, P = 0.04) of nNOS 84G →A locus. In case of nNOS 276C→T variation, HSVE risk was linked to CT genotype (OR = 1.79, 95%CI = 1.07-3.0, P = 0.03) and TT genotype (OR = 3.6, 95%CI = 1.2-10.8, P = 0.02). Patients with poor outcome either had homo or heterozygous genotype for both SNPs, but separate genotype analysis could not show significance. But combined genotype analysis of both SNPs confirmed that GG + CC was a risk factor for development of poor outcome. (OR = 6.3, CI-1.9-20.7, P = 0.0033). Haplotype analysis of both SNP did show that "at" haplotype was significantly higher and associated with HSVE cases (OR = 2.322,CI: 1.43-3.77, P = 0.00070). The result observed in this study suggested that variation at these loci of nNOS may have decreased its expression and caused low production of NO, which have resulted in risk of HSVE but provided good outcome in these patients.


Asunto(s)
Encefalitis por Herpes Simple/genética , Óxido Nítrico Sintasa de Tipo I/genética , Polimorfismo de Nucleótido Simple , Pueblo Asiatico , Niño , Encefalitis por Herpes Simple/diagnóstico , Encefalitis por Herpes Simple/mortalidad , Predisposición Genética a la Enfermedad , Humanos , India , Óxido Nítrico/metabolismo , Pronóstico , Factores de Riesgo
14.
Hum Genet ; 139(6-7): 911-918, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32040615

RESUMEN

Herpes simplex virus 1 (HSV-1) encephalitis (HSE) is the most common sporadic viral encephalitis in Western countries. Over the last 15 years, human genetic and immunological studies have provided proof-of-principle that childhood HSE can result from inborn errors of central nervous system (CNS)-specific, cell-intrinsic immunity to HSV-1. HSE-causing mutations of eight genes disrupt known (TLR3-dependent IFN-α/ß immunity) and novel (dependent on DBR1 or snoRNA31) antiviral mechanisms. Monogenic inborn errors confer susceptibility to forebrain (TLR3-IFN or snoRNA31) or brainstem (DBR1) HSE. Most of these disorders display incomplete clinical penetrance, with the possible exception of DBR1 deficiency. They account for a small, but non-negligible proportion of cases (about 7%). These findings pave the way for the gradual definition of the genetic and immunological architecture of childhood HSE, with both biological and clinical implications.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Encefalitis por Herpes Simple/genética , Predisposición Genética a la Enfermedad , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno/genética , Inmunidad Celular/inmunología , Enfermedades del Sistema Nervioso Central/epidemiología , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/virología , Niño , Encefalitis por Herpes Simple/epidemiología , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/virología , Herpes Simple/complicaciones , Herpes Simple/virología , Herpesvirus Humano 1/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Humanos , Mutación
15.
J Infect Dis ; 221(8): 1295-1303, 2020 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-31268141

RESUMEN

BACKGROUND: Herpes zoster ophthalmicus occurs primarily in elderly or immunocompromised individuals after reactivation of varicella zoster virus (VZV). Recurrences of zoster ophthalmicus are uncommon because the reactivation efficiently boosts anti-VZV immunity. A 28-year-old female presented to our clinic with a history of multiple recurrences of zoster ophthalmicus. METHODS: Whole-exome sequencing (WES), analyses of VZV T-cell immunity, and pathogen recognition receptor function in primary antigen-presenting cells (APCs) and fibroblasts were performed. RESULTS: Normal VZV-specific T-cell immunity and antibody response were detected. Whole-exome sequencing identified a heterozygous nonsynonymous variant (c.2324C > T) in the Toll-like receptor 3 (TLR3) gene resulting in formation of a premature stop-codon. This alteration could potentially undermine TLR3 signaling in a dominant-negative fashion. Therefore, we investigated TLR3 signaling responses in APCs and fibroblasts from the patient. The APCs responded efficiently to stimulation with TLR3 ligands, whereas the responses from the fibroblasts were compromised. CONCLUSIONS: We report a novel TLR3 variant associated with recurrent zoster ophthalmicus. Toll-like receptor 3 responses that were unaffected in APCs but diminished in fibroblasts are in line with previous reports linking TLR3 deficiency with herpes simplex virus encephalitis. Mechanisms involving compromised viral sensing in infected cells may thus be central to the described immunodeficiency.


Asunto(s)
Herpes Zóster Oftálmico/virología , Herpesvirus Humano 3/patogenicidad , Mutación/genética , Receptor Toll-Like 3/genética , Adulto , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/virología , Femenino , Fibroblastos/virología , Herpes Zóster/genética , Herpes Zóster/virología , Herpes Zóster Oftálmico/genética , Humanos , Huésped Inmunocomprometido/genética
16.
Nat Med ; 25(12): 1873-1884, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31806906

RESUMEN

Herpes simplex virus-1 (HSV-1) encephalitis (HSE) is typically sporadic. Inborn errors of TLR3- and DBR1-mediated central nervous system cell-intrinsic immunity can account for forebrain and brainstem HSE, respectively. We report five unrelated patients with forebrain HSE, each heterozygous for one of four rare variants of SNORA31, encoding a small nucleolar RNA of the H/ACA class that are predicted to direct the isomerization of uridine residues to pseudouridine in small nuclear RNA and ribosomal RNA. We show that CRISPR/Cas9-introduced bi- and monoallelic SNORA31 deletions render human pluripotent stem cell (hPSC)-derived cortical neurons susceptible to HSV-1. Accordingly, SNORA31-mutated patient hPSC-derived cortical neurons are susceptible to HSV-1, like those from TLR3- or STAT1-deficient patients. Exogenous interferon (IFN)-ß renders SNORA31- and TLR3- but not STAT1-mutated neurons resistant to HSV-1. Finally, transcriptome analysis of SNORA31-mutated neurons revealed normal responses to TLR3 and IFN-α/ß stimulation but abnormal responses to HSV-1. Human SNORA31 thus controls central nervous system neuron-intrinsic immunity to HSV-1 by a distinctive mechanism.


Asunto(s)
Encefalitis por Herpes Simple/genética , Herpesvirus Humano 1/genética , Neuronas/inmunología , ARN Nucleolar Pequeño/genética , Adulto , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Preescolar , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/patología , Encefalitis por Herpes Simple/virología , Femenino , Predisposición Genética a la Enfermedad , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Inmunidad/genética , Lactante , Masculino , Metagenoma/genética , Metagenoma/inmunología , Persona de Mediana Edad , Neuronas/virología , ARN Nucleolar Pequeño/inmunología
17.
PLoS Pathog ; 15(12): e1008168, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31869396

RESUMEN

We report here two cases of Herpes simplex virus encephalitis (HSE) in adult patients with very rare, previously uncharacterized, non synonymous heterozygous G634R and R203W substitution in mannan-binding lectin serine protease 2 (MASP2), a gene encoding a key protease of the lectin pathway of the complement system. None of the 2 patients had variants in genes involved in the TLR3-interferon signaling pathway. Both MASP2 variants induced functional defects in vitro, including a reduced (R203W) or abolished (G634R) protein secretion, a lost capability to cleave MASP-2 precursor into its active form (G634R) and an in vivo reduced antiviral activity (G634R). In a murine model of HSE, animals deficient in mannose binding lectins (MBL, the main pattern recognition molecule associated with MASP-2) had a decreased survival rate and an increased brain burden of HSV-1 compared to WT C57BL/6J mice. Altogether, these data suggest that MASP-2 deficiency can increase susceptibility to adult HSE.


Asunto(s)
Encefalitis por Herpes Simple/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/deficiencia , Adulto , Animales , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/inmunología , Humanos , Inmunidad Innata/genética , Lectinas/genética , Lectinas/metabolismo , Masculino , Lectina de Unión a Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
J Neurovirol ; 25(4): 525-539, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31144288

RESUMEN

Herpes simplex virus 1 (HSV-1) is a predominant cause of herpes simplex encephalitis (HSE), leading to a high mortality rate and severe neurological sequelae worldwide. HSE is typically accompanied by the blood-brain barrier (BBB) disruption, but the underlying mechanisms are unclear. To explore the disruption mechanisms of the BBB, quantitative analysis of the cellular proteome was carried out to investigate the proteomic changes that occur after infection. In this study, bEnd.3 cells were infected with HSV-1, followed by liquid chromatography-tandem mass spectrometry. A total of 6761 proteins were identified in three independent mass spectrometry analyses. Compared to the uninfected cells, 386 and 293 differentially expressed proteins were markedly upregulated or downregulated, respectively. Bioinformatic analysis showed that the activator protein-1 factor, including Fos, Jun, and ATF family proteins and cell adhesion molecules were significantly changed. Further validation of the changes observed for these proteins was carried out by western blotting and quantitative real-time PCR. Transendothelial electrical resistance (TEER) studies were performed to explore the effects of ATF3, Fra1, or JunB overexpression on the function of bEnd.3 cells. Characterization of the differential expression of these proteins in bEnd.3 cells will facilitate further exploration of BBB disruption upon HSV-1 infection.


Asunto(s)
Factor de Transcripción Activador 3/genética , Encefalitis por Herpes Simple/genética , Células Endoteliales/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Proto-Oncogénicas c-fos/genética , Factores de Transcripción/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/virología , Encéfalo/metabolismo , Encéfalo/virología , Línea Celular , Encefalitis por Herpes Simple/metabolismo , Encefalitis por Herpes Simple/virología , Células Endoteliales/virología , Regulación de la Expresión Génica , Ontología de Genes , Herpesvirus Humano 1/patogenicidad , Interacciones Huésped-Patógeno/genética , Humanos , Ratones , Modelos Biológicos , Anotación de Secuencia Molecular , Proteoma/clasificación , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Replicación Viral
19.
Genes Immun ; 20(1): 10-22, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217828

RESUMEN

We selected two sets of naturally occurring human missense allelic variants within innate immune genes. The first set represented eleven non-synonymous variants in six different genes involved in interferon (IFN) induction, present in a cohort of patients suffering from herpes simplex encephalitis (HSE) and the second set represented sixteen allelic variants of the IFNLR1 gene. We recreated the variants in vitro and tested their effect on protein function in a HEK293T cell based assay. We then used an array of 14 available bioinformatics tools to predict the effect of these variants upon protein function. To our surprise two of the most commonly used tools, CADD and SIFT, produced a high rate of false positives, whereas SNPs&GO exhibited the lowest rate of false positives in our test. As the problem in our test in general was false positive variants, inclusion of mutation significance cutoff (MSC) did not improve accuracy.


Asunto(s)
Biología Computacional/normas , Encefalitis por Herpes Simple/genética , Pruebas Genéticas/normas , Estudio de Asociación del Genoma Completo/normas , Programas Informáticos/normas , Niño , Reacciones Falso Positivas , Femenino , Células HEK293 , Humanos , Masculino , Mutación Missense , Polimorfismo de Nucleótido Simple , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de Interferón
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA