Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 200
1.
Proc Natl Acad Sci U S A ; 119(30): e2122236119, 2022 07 26.
Article En | MEDLINE | ID: mdl-35858406

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) readily infects a variety of cell types impacting the function of vital organ systems, with particularly severe impact on respiratory function. Neurological symptoms, which range in severity, accompany as many as one-third of COVID-19 cases, indicating a potential vulnerability of neural cell types. To assess whether human cortical cells can be directly infected by SARS-CoV-2, we utilized stem-cell-derived cortical organoids as well as primary human cortical tissue, both from developmental and adult stages. We find significant and predominant infection in cortical astrocytes in both primary tissue and organoid cultures, with minimal infection of other cortical populations. Infected and bystander astrocytes have a corresponding increase in inflammatory gene expression, reactivity characteristics, increased cytokine and growth factor signaling, and cellular stress. Although human cortical cells, particularly astrocytes, have no observable ACE2 expression, we find high levels of coronavirus coreceptors in infected astrocytes, including CD147 and DPP4. Decreasing coreceptor abundance and activity reduces overall infection rate, and increasing expression is sufficient to promote infection. Thus, we find tropism of SARS-CoV-2 for human astrocytes resulting in inflammatory gliosis-type injury that is dependent on coronavirus coreceptors.


Astrocytes , Cerebral Cortex , SARS-CoV-2 , Viral Tropism , Angiotensin-Converting Enzyme 2/metabolism , Astrocytes/enzymology , Astrocytes/virology , Cerebral Cortex/virology , Humans , Organoids/virology , Primary Cell Culture , SARS-CoV-2/physiology
2.
J Virol ; 96(14): e0012622, 2022 07 27.
Article En | MEDLINE | ID: mdl-35862705

Human cytomegalovirus (HCMV) is a prevalent betaherpesvirus that is asymptomatic in healthy individuals but can cause serious disease in immunocompromised patients. HCMV is also the leading cause of virus-mediated birth defects. Many of these defects manifest within the central nervous system and include microcephaly, sensorineural hearing loss, and cognitive developmental delays. Nitric oxide is a critical effector molecule produced as a component of the innate immune response during infection. Congenitally infected fetal brains show regions of brain damage, including necrotic foci with infiltrating macrophages and microglia, cell types that produce nitric oxide during infection. Using a 3-dimensional cortical organoid model, we demonstrate that nitric oxide inhibits HCMV spread and simultaneously disrupts neural rosette structures, resulting in tissue disorganization. Nitric oxide also attenuates HCMV replication in 2-dimensional cultures of neural progenitor cells (NPCs), a prominent cell type in cortical organoids that differentiate into neurons and glial cells. The multipotency factor SOX2 was decreased during nitric oxide exposure, suggesting that early neural differentiation is affected. Nitric oxide also reduced maximal mitochondrial respiration in both uninfected and infected NPCs. We determined that this reduction likely influences neural differentiation, as neurons (Tuj1+ GFAP- Nestin-) and glial populations (Tuj1- GFAP+ Nestin-) were reduced following differentiation. Our studies indicate a prominent, immunopathogenic role of nitric oxide in promoting developmental defects within the brain despite its antiviral activity during congenital HCMV infection. IMPORTANCE Human cytomegalovirus (HCMV) is the leading cause of virus-mediated congenital birth defects. Congenitally infected infants can have a variety of symptoms manifesting within the central nervous system. The use of 3-dimensional (3-D) cortical organoids to model infection of the fetal brain has advanced the current understanding of development and allowed broader investigation of the mechanisms behind disease. However, the impact of the innate immune molecule nitric oxide during HCMV infection has not been explored in neural cells or cortical 3-D models. Here, we investigated the effect of nitric oxide on cortical development during HCMV infection. We demonstrate that nitric oxide plays an antiviral role during infection yet results in disorganized cortical tissue. Nitric oxide contributes to differentiation defects of neuron and glial cells from neural progenitor cells despite inhibiting viral replication. Our results indicate that immunopathogenic consequences of nitric oxide during congenital infection promote developmental defects that undermine its antiviral activity.


Cell Differentiation , Cytomegalovirus Infections , Neural Stem Cells , Nitric Oxide , Antiviral Agents , Cerebral Cortex/virology , Cytomegalovirus/physiology , Cytomegalovirus Infections/pathology , Humans , Nestin , Neural Stem Cells/virology , Nitric Oxide/pharmacology , Organoids/virology
3.
Front Immunol ; 12: 753683, 2021.
Article En | MEDLINE | ID: mdl-34899705

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Cytopathogenic Effect, Viral , Disease Models, Animal , Encephalitis, Viral/metabolism , Parechovirus/pathogenicity , Picornaviridae Infections/metabolism , Animals , Animals, Newborn , Apoptosis , Autophagy , Cell Line, Tumor , Cerebral Cortex/virology , Chlorocebus aethiops , Cytokines/biosynthesis , Cytokines/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Glioblastoma/pathology , Hippocampus/virology , Humans , Inflammation , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon Type I/pharmacology , Interferons/biosynthesis , Interferons/genetics , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Parechovirus/drug effects , Parechovirus/physiology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Pyroptosis , Vero Cells , Virus Replication/drug effects , Interferon Lambda
4.
PLoS One ; 16(12): e0261208, 2021.
Article En | MEDLINE | ID: mdl-34890434

PURPOSE: Changes in cerebral cortical regions occur in HIV-infected patients, even in those with mild neurocognitive disorders. Working memory / attention is one of the most affected cognitive domain in these patients, worsening their quality of life. Our objective was to assess whether cortical thickness differs between HIV-infected patients with and without working memory deficit. METHODS: Forty-one adult HIV-infected patients with and without working memory deficit were imaged on a 1.5 T scanner. Working memory deficit was classified by composite Z scores for performance on the Digits and Letter-Number Sequencing subtests of the Wechsler Adult Intelligence Scale (third edition; WAIS-III). Cortical thickness was determined using FreeSurfer software. Differences in mean cortical thickness between groups, corrected for multiple comparisons using Monte-Carlo simulation, were examined using the query design estimate contrast tool of the FreeSurfer software. RESULTS: Greater cortical thickness in left pars opercularis of the inferior frontal gyrus, and rostral and caudal portions of the left middle frontal gyrus (cluster 1; p = .004), and left superior frontal gyrus (cluster 2; p = .004) was observed in HIV-infected patients with working memory deficit compared with those without such deficit. Negative correlations were found between WAIS-III-based Z scores and cortical thickness in the two clusters (cluster 1: ρ = -0.59; cluster 2: ρ = -0.47). CONCLUSION: HIV-infected patients with working memory deficit have regions of greater thickness in the left frontal cortices compared with those without such deficit, which may reflect increased synaptic contacts and/or an inflammatory response related to the damage caused by HIV infection.


Cerebral Cortex/pathology , Cerebral Cortex/virology , HIV Infections/pathology , Memory Disorders/virology , Memory, Short-Term/physiology , Adult , Aged , Brazil/epidemiology , Female , HIV/isolation & purification , HIV Infections/epidemiology , HIV Infections/psychology , HIV Infections/virology , Humans , Male , Memory Disorders/epidemiology , Memory Disorders/pathology , Memory Disorders/psychology , Middle Aged , Neuropsychological Tests
5.
Clin Neurol Neurosurg ; 210: 106956, 2021 11.
Article En | MEDLINE | ID: mdl-34583276

Influenza virus-associated encephalopathy/encephalitis is a rare entity in adults that can lead to severe neurological sequelae and even death. The clinical presentation can be quite diverse. This absence of a typical presentation along with the difficulty detecting the virus in the cerebrospinal fluid represents a diagnostic challenge. We present the case of a 79-year-old male with sudden onset of decreased consciousness and signs of right hemisphere damage. The presence of influenza A (H3N2) virus in respiratory sample along with compatible findings in cranial magnetic resonance led to the diagnosis. The patient died without responding to treatment with antivirals and immunomodulators and the anatomopathological study did not detect infectious agent. Early diagnostic suspicion is essential to establish adequate treatment and improve the prognosis.


Cerebral Cortex/diagnostic imaging , Encephalitis, Viral/diagnostic imaging , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/diagnostic imaging , Aged , Cerebral Cortex/virology , Humans , Magnetic Resonance Imaging , Male
6.
Cells ; 10(9)2021 08 31.
Article En | MEDLINE | ID: mdl-34571912

COVID-19 presents with a wide range of clinical neurological manifestations. It has been recognized that SARS-CoV-2 infection affects both the central and peripheral nervous system, leading to smell and taste disturbances; acute ischemic and hemorrhagic cerebrovascular disease; encephalopathies and seizures; and causes most surviving patients to have long lasting neurological symptoms. Despite this, typical neuropathological features associated with the infection have still not been identified. Studies of post-mortem examinations of the cerebral cortex are obtained with difficulty due to laboratory safety concerns. In addition, they represent cases with different neurological symptoms, age or comorbidities, thus a larger number of brain autoptic data from multiple institutions would be crucial. Histopathological findings described here are aimed to increase the current knowledge on neuropathology of COVID-19 patients. We report post-mortem neuropathological findings of ten COVID-19 patients. A wide range of neuropathological lesions were seen. The cerebral cortex of all patients showed vascular changes, hyperemia of the meninges and perivascular inflammation in the cerebral parenchyma with hypoxic neuronal injury. Perivascular lymphocytic inflammation of predominantly CD8-positive T cells mixed with CD68-positive macrophages, targeting the disrupted vascular wall in the cerebral cortex, cerebellum and pons were seen. Our findings support recent reports highlighting a role of microvascular injury in COVID-19 neurological manifestations.


COVID-19/pathology , Cerebral Cortex/pathology , Aged , Aged, 80 and over , Autopsy , Brain/pathology , Brain/virology , Brain Diseases/pathology , Brain Diseases/virology , CD8-Positive T-Lymphocytes/pathology , Cerebral Cortex/virology , Female , Humans , Inflammation , Macrophages/pathology , Male , Microvessels/pathology , Microvessels/virology , Middle Aged , Nervous System Diseases/pathology , Nervous System Diseases/virology , SARS-CoV-2/pathogenicity
7.
J Immunol Res ; 2021: 5317662, 2021.
Article En | MEDLINE | ID: mdl-34327243

Zika virus (ZIKV) has attracted the wide global attention due to its causal link to microcephaly. In this study, two amino acid (aa) mutation (E143K and R3394K) were identified at the fourth generation (named ZKC2P4) during the serial passage of ZIKV-Asian lineage ZKC2/2016 strain in the newborn mouse brain, while another seven aa deletions in envelope (E) protein were detected in ZKC2P6. ZKC2P6 is a novel nonglycosylated E protein Asian ZIKV we first identified and provides the first direct supporting evidence that glycosylation motif could be lost during the passage in neonatal mice. To study the impact of E protein glycosylation ablation, we compared the pathogenicity of ZKC2P6 with that of ZKC2P4. The results showed that the loss of E protein glycosylation accelerated the disease progression, as evidenced by an earlier weight loss and death, a thinner cerebral cortex, and more serious tissue lesions and inflammation/necrosis. Furthermore, ZKC2P6 exhibited a greater ability to replicate and caused severer cell apoptosis than that of ZKC2P4. Therefore, the ablation of E glycosylation generally enhances the neurovirulence of ZIKV and cell apoptosis in newborn mice.


Cerebral Cortex/pathology , Viral Envelope Proteins/metabolism , Zika Virus Infection/immunology , Zika Virus/pathogenicity , Animals , Animals, Newborn , Apoptosis , Cell Line, Tumor , Cerebral Cortex/immunology , Cerebral Cortex/virology , Chlorocebus aethiops , Disease Models, Animal , Disease Progression , Glycosylation , Humans , Mice , Sequence Deletion , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virus Replication/immunology , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/metabolism , Zika Virus Infection/pathology , Zika Virus Infection/virology
8.
J Exp Med ; 218(3)2021 03 01.
Article En | MEDLINE | ID: mdl-33433624

Although COVID-19 is considered to be primarily a respiratory disease, SARS-CoV-2 affects multiple organ systems including the central nervous system (CNS). Yet, there is no consensus on the consequences of CNS infections. Here, we used three independent approaches to probe the capacity of SARS-CoV-2 to infect the brain. First, using human brain organoids, we observed clear evidence of infection with accompanying metabolic changes in infected and neighboring neurons. However, no evidence for type I interferon responses was detected. We demonstrate that neuronal infection can be prevented by blocking ACE2 with antibodies or by administering cerebrospinal fluid from a COVID-19 patient. Second, using mice overexpressing human ACE2, we demonstrate SARS-CoV-2 neuroinvasion in vivo. Finally, in autopsies from patients who died of COVID-19, we detect SARS-CoV-2 in cortical neurons and note pathological features associated with infection with minimal immune cell infiltrates. These results provide evidence for the neuroinvasive capacity of SARS-CoV-2 and an unexpected consequence of direct infection of neurons by SARS-CoV-2.


Angiotensin-Converting Enzyme 2 , Antibodies, Blocking/chemistry , COVID-19 , Cerebral Cortex , Neurons , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , COVID-19/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/virology , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Neurons/metabolism , Neurons/pathology , Neurons/virology , Organoids/metabolism , Organoids/pathology , Organoids/virology
9.
J Clin Invest ; 131(1)2021 01 04.
Article En | MEDLINE | ID: mdl-33393505

Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/ß induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-ß protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-ß secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-ß immunity.


Cerebral Cortex/immunology , Fibroblasts/immunology , Herpesvirus 1, Human/immunology , Interferon-beta/immunology , Neurons/immunology , Toll-Like Receptor 3/immunology , Vesiculovirus/immunology , Animals , Cell Line , Cerebral Cortex/pathology , Cerebral Cortex/virology , Fibroblasts/pathology , Fibroblasts/virology , Humans , Interferon-beta/genetics , Mice , Mice, Knockout , Neurons/pathology , Neurons/virology , Toll-Like Receptor 3/genetics
10.
J Neurovirol ; 26(6): 846-862, 2020 12.
Article En | MEDLINE | ID: mdl-32910432

Previous studies showed that persons living with HIV (PLWH) demonstrate higher brain prefrontal cortex neuroinflammation and immunoproteasome expression compared to HIV-negative individuals; these associate positively with HIV levels. Lower expression of the antioxidant enzyme heme oxygenase 1 (HO-1) was observed in PLWH with HIV-associated neurocognitive impairment (HIV-NCI) compared to neurocognitively normal PLWH. We hypothesized that similar expression patterns occur throughout cortical, subcortical, and brainstem regions in PLWH, and that neuroinflammation and immunoproteasome expression associate with lower expression of neuronal markers. We analyzed autopsied brains (15 regions) from 9 PLWH without HIV-NCI and 7 matched HIV-negative individuals. Using Western blot and RT-qPCR, we quantified synaptic, inflammatory, immunoproteasome, endothelial, and antioxidant biomarkers, including HO-1 and its isoform heme oxygenase 2 (HO-2). In these PLWH without HIV-NCI, we observed higher expression of neuroinflammatory, endothelial, and immunoproteasome markers in multiple cortical and subcortical regions compared to HIV-negative individuals, suggesting a global brain inflammatory response to HIV. Several regions, including posterior cingulate cortex, globus pallidus, and cerebellum, showed a distinct pattern of higher type I interferon (IFN)-stimulated gene and immunoproteasome expression. PLWH without HIV-NCI also had (i) stable or higher HO-1 expression and positive associations between (ii) HO-1 and HIV levels (CSF, plasma) and (iii) HO-1 expression and neuroinflammation, in multiple cortical, subcortical, and brainstem regions. We observed no differences in synaptic marker expression, suggesting little, if any, associated neuronal injury. We speculate that this may reflect a neuroprotective effect of a concurrent HO-1 antioxidant response despite global neuroinflammation, which will require further investigation.


Cerebral Cortex/metabolism , Cognitive Dysfunction/genetics , HIV Infections/genetics , HIV-1/pathogenicity , Heme Oxygenase-1/genetics , Aged , Amygdala/metabolism , Amygdala/virology , Autopsy , Biomarkers/metabolism , Brain Stem/metabolism , Brain Stem/virology , Case-Control Studies , Caudate Nucleus/metabolism , Caudate Nucleus/virology , Cerebral Cortex/virology , Cognitive Dysfunction/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/virology , Female , Gene Expression Regulation , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/virology , Heme Oxygenase-1/metabolism , Humans , Inflammation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
11.
J Neurovirol ; 26(5): 769-778, 2020 10.
Article En | MEDLINE | ID: mdl-32839948

The blood-brain barrier (BBB) is a major obstacle for the treatment of central nervous system (CNS) disorders. Significant progress has been made in developing adeno-associated virus (AAV) variants with increased ability to cross the BBB in mice. However, these variants are not efficacious in non-human primates. Herein, we employed various bioinformatic techniques to identify lymphocyte antigen-6E (LY6E) as a candidate for mediating transport of AAV across the human BBB based on the previously determined mechanism of transport in mice. Our results provide insight into future discovery and optimization of AAV variants for CNS gene delivery in humans.


Antigens, Ly/metabolism , Antigens, Surface/metabolism , Blood-Brain Barrier/metabolism , Dependovirus/metabolism , Genetic Vectors/metabolism , Membrane Proteins/metabolism , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Antigens, Ly/chemistry , Antigens, Ly/genetics , Antigens, Surface/chemistry , Antigens, Surface/genetics , Biological Transport , Blood-Brain Barrier/virology , Capillary Permeability , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/virology , Computational Biology/methods , Dependovirus/chemistry , Dependovirus/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/virology , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/chemistry , Humans , Macaca mulatta , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Sequence Alignment , Sequence Homology, Amino Acid
12.
J Neurovirol ; 26(5): 790-792, 2020 10.
Article En | MEDLINE | ID: mdl-32671810

Enteroviruses are one of the most important causes of viral encephalitis in the neonatal period. However, the non-specificity of the symptoms presented renders its diagnosis challenging. Intracranial MRI has been reported to be a very useful imaging modality that can detect the characteristic white matter lesions around the periventricular regions. In this study, we report a case of a patient with neonatal encephalitis who presented with normal white blood cell counts in the initial cerebrospinal fluid analysis. A lumbar puncture retap identified pleocytosis, and polymerase chain reaction assays detected enterovirus 71 in the blood and stool samples. Furthermore, MRI revealed atypical disseminated cortical and subcortical white matter lesions on diffusion weighted images, and neuroradiological re-evaluation showed necrotic changes 2 weeks later. This unique case expands our knowledge of the spectrum of neurological disorders due to enterovirus 71 infection in neonatal period.


Encephalitis, Viral/diagnostic imaging , Enterovirus A, Human/pathogenicity , Enterovirus Infections/diagnostic imaging , White Matter/diagnostic imaging , Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/virology , Diffusion Magnetic Resonance Imaging/methods , Encephalitis, Viral/drug therapy , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Enterovirus A, Human/drug effects , Enterovirus A, Human/genetics , Enterovirus A, Human/growth & development , Enterovirus Infections/drug therapy , Enterovirus Infections/pathology , Enterovirus Infections/virology , Humans , Infant, Newborn , Male , Neuroimaging/methods , Spinal Puncture/methods , White Matter/pathology , White Matter/virology
13.
J Neuroinflammation ; 16(1): 248, 2019 Dec 02.
Article En | MEDLINE | ID: mdl-31791351

BACKGROUND: Herpes simplex virus-1 (HSV-1) infections of the central nervous system (CNS) can result in HSV-1 encephalitis (HSE) which is characterized by severe brain damage and long-term disabilities. Different cell types including neurons and astrocytes become infected in the course of an HSE which leads to an activation of glial cells. Activated glial cells change their neurotrophic factor profile and modulate inflammation and repair. The superfamily of fibroblast growth factors (FGFs) is one of the largest family of neurotrophic factors comprising 22 ligands. FGFs induce pro-survival signaling in neurons and an anti-inflammatory answer in glial cells thereby providing a coordinated tissue response which favors repair over inflammation. Here, we hypothesize that FGF expression is altered in HSV-1-infected CNS cells. METHOD: We employed primary murine cortical cultures comprising a mixed cell population of astrocytes, neurons, microglia, and oligodendrocytes. Astrocyte reactivity was morphometrically monitored by an automated image analysis algorithm as well as by analyses of A1/A2 marker expression. Altered FGF expression was detected by quantitative real-time PCR and its paracrine FGF activity. In addition, HSV-1 mutants were employed to characterize viral factors important for FGF responses of infected host cells. RESULTS: Astrocytes in HSV-1-infected cortical cultures were transiently activated and became hypertrophic and expressed both A1- and A2-markers. Consistently, a number of FGFs were transiently upregulated inducing paracrine neurotrophic signaling in neighboring cells. Most prominently, FGF-4, FGF-8, FGF-9, and FGF-15 became upregulated in a switch-on like mechanism. This effect was specific for CNS cells and for a fully functional HSV-1. Moreover, the viral protein ICP0 critically mediated the FGF switch-on mechanism. CONCLUSIONS: HSV-1 uses the viral protein ICP0 for the induction of FGF-expression in CNS cells. Thus, we propose that HSV-1 triggers FGF activity in the CNS for a modulation of tissue response upon infection.


Cerebral Cortex/metabolism , Fibroblast Growth Factors/metabolism , Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Paracrine Communication/physiology , Viral Proteins/metabolism , Animals , Animals, Newborn , Cerebral Cortex/virology , Chlorocebus aethiops , Coculture Techniques , Cricetinae , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Vero Cells
14.
Front Immunol ; 10: 814, 2019.
Article En | MEDLINE | ID: mdl-31110499

Purpose: The aim of this study was to investigate how human immunodeficiency virus (HIV) affects brain development in adolescents, what are susceptible brain regions, and how these brain structural changes correlate with cognitive abilities. Methods: We used structural magnetic resonance imaging to examine gray matter volume and cortical thickness in 16 HIV-infected children (mean age = 13.63 years) and 25 HIV-exposed uninfected children (mean age = 13.32 years), 12 of them were subjected to a 1-year repetitive magnetic resonance scan of the brain. Five neurocognitive tests were performed on each subject to assess cognitive performance in different areas. Results: Cross-sectional studies showed that the gray matter volume of HIV-infected children was widely reduced (mainly in the bilateral frontal, temporal, and insular regions, and cerebellum). The changes in cortical thickness were mainly due to thinning of the right temporal lobe and thickening of the left occipital lobe. Longitudinal studies showed that the gray matter volume reduction of HIV-infected children after 1 year mainly occurs in the advanced functional area of the right prefrontal, parietal lobe and the motor area, cortical thinning of brain regions were sensorimotor cortex and the limbic system. The gray matter volume of the bilateral cerebellum was positively correlated with the performance of the Wisconsin Card Sorting Test, while the cortical thickness of the right dorsolateral prefrontal cortex was negatively correlated with this test. Conclusion: This study found that HIV-infected pubertal children showed a delayed cortical maturation with atrophy. This abnormal pattern of cortical development may be the structural basis for cognitive impairment in HIV-infected children.


Cerebral Cortex/anatomy & histology , Gray Matter/anatomy & histology , HIV Infections/physiopathology , Adolescent , Cerebral Cortex/virology , Child , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/virology , Cross-Sectional Studies , Female , Gray Matter/virology , Humans , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Organ Size/physiology
15.
Glia ; 67(9): 1719-1729, 2019 09.
Article En | MEDLINE | ID: mdl-31124192

The HIV-1 protein Tat is continually released by HIV-infected cells despite effective combination antiretroviral therapies (cART). Tat promotes neurotoxicity through enhanced expression of proinflammatory molecules from resident and infiltrating immune cells. These molecules include matrix metalloproteinases (MMPs), which are pathologically elevated in HIV, and are known to drive central nervous system (CNS) injury in varied disease settings. A subset of MMPs can activate G-protein coupled protease-activated receptor 1 (PAR-1), a receptor that is highly expressed on astrocytes. Although PAR-1 expression is increased in HIV-associated neurocognitive disorder (HAND), its role in HAND pathogenesis remains understudied. Herein, we explored Tat's ability to induce expression of the PAR-1 agonists MMP-3 and MMP-13. We also investigated MMP/PAR-1-mediated release of CCL2, a chemokine that drives CNS entry of HIV infected monocytes and remains a significant correlate of cognitive dysfunction in the era of cART. Tat exposure significantly increased the expression of MMP-3 and MMP-13. These PAR-1 agonists both stimulated the release of astrocytic CCL2, and both genetic knock-out and pharmacological inhibition of PAR-1 reduced CCL2 release. Moreover, in HIV-infected post-mortem brain tissue, within-sample analyses revealed a correlation between levels of PAR-1-activating MMPs, PAR-1, and CCL2. Collectively, these findings identify MMP/PAR-1 signaling to be involved in the release of CCL2, which may underlie Tat-induced neuroinflammation.


Astrocytes/metabolism , Astrocytes/virology , Chemokine CCL2/metabolism , Matrix Metalloproteinases/metabolism , Protein Serine-Threonine Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Adult , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/virology , Female , HIV-1 , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Protein Serine-Threonine Kinases/genetics , Signal Transduction
16.
J Neurovirol ; 25(4): 605-607, 2019 08.
Article En | MEDLINE | ID: mdl-31140129

Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease caused by JC virus reactivation. Its occurrence is very rare after solid organ transplantation, especially liver transplantation. We report a patient who received liver transplantation due to liver failure resulting from autoimmune hepatitis and advanced PML presenting with aphasia. A 41-year-old female with a history of liver transplantation who received a usual immunosuppression regimen was admitted with a stroke attack resulting in right hemiplegia 2 months after liver transplantation. Surprisingly, she gradually developed dysarthria and left central facial paresis. A brain MRI showed an abnormal multifocal area with a high T2/flair signal in the deep subcortical white matter of the left hemisphere as well as the splenium of the corpus callosum. PCR evaluation of CSF for JCV was positive while other PCR results were negative. A liver transplant recipient receiving immunosuppressive treatment for a long time could develop PML due to JCV reactivation. Only eight cases of JCV infection were reported after liver transplantation by the time of reporting this case. Unfortunately, there is no definite treatment for PML.


Hepatitis, Autoimmune/immunology , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/immunology , Liver Transplantation , Adult , Aphasia/diagnostic imaging , Aphasia/physiopathology , Aphasia/virology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/virology , Corpus Callosum/diagnostic imaging , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/virology , Dysarthria/diagnostic imaging , Dysarthria/physiopathology , Dysarthria/virology , Female , Hemiplegia/diagnostic imaging , Hemiplegia/physiopathology , Hemiplegia/virology , Hepatitis, Autoimmune/pathology , Hepatitis, Autoimmune/surgery , Hepatitis, Autoimmune/virology , Humans , Immunosuppressive Agents/administration & dosage , JC Virus/immunology , JC Virus/isolation & purification , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/surgery , Liver/drug effects , Liver/immunology , Liver/pathology , Liver/surgery , Magnetic Resonance Imaging , Stroke/diagnostic imaging , Stroke/physiopathology , Stroke/virology , Virus Activation/immunology
17.
Emerg Microbes Infect ; 8(1): 307-326, 2019.
Article En | MEDLINE | ID: mdl-30866785

The harmful effects of ZIKA virus (ZIKV) infection are reflected by severe neurological manifestations such as microcephaly in neonates and other complications associated with Guillain-Barré syndrome in adults. The transmission dynamics of ZIKV in or between neurons, or within the developing brains of the foetuses are not fully understood. Using primary cultures of murine cortical neurons, we show that ZIKV uses exosomes as mediators of viral transmission between neurons. Cryo-electron microscopy showed heterogeneous population of neuronal exosomes with a size range of 30-200 nm. Increased production of exosomes from neuronal cells was noted upon ZIKV infection. Neuronal exosomes contained both ZIKV viral RNA and protein(s) that were highly infectious to naïve cells. RNaseA and neutralizing antibodies treatment studies suggest the presence of viral RNA/proteins inside exosomes. Exosomes derived from time- and dose-dependent incubations showed increasing viral loads suggesting higher packaging and delivery of ZIKV RNA and proteins. Furthermore, we noted that ZIKV induced both activity and gene expression of neutral Sphingomyelinase (nSMase)-2/SMPD3, an important molecule that regulates production and release of exosomes. Silencing of SMPD3 in neurons resulted in reduced viral burden and transmission through exosomes. Treatment with SMPD3 specific inhibitor GW4869, significantly reduced ZIKV loads in both cortical neurons and in exosomes derived from these neuronal cells. Taken together, our results suggest that ZIKV modulates SMPD3 activity in cortical neurons for its infection and transmission through exosomes perhaps leading to severe neuronal death that may result in neurological manifestations such as microcephaly in the developing embryonic brains.


Cerebral Cortex/virology , Exosomes/virology , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Zika Virus/physiology , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cryoelectron Microscopy , Exosomes/genetics , Exosomes/metabolism , Gene Expression Regulation, Enzymologic , Mice , Neurons/cytology , Neurons/virology , Time Factors , Viral Load/drug effects , Virus Assembly/drug effects
18.
PLoS Pathog ; 15(1): e1007507, 2019 01.
Article En | MEDLINE | ID: mdl-30657788

Zika virus (ZIKV) infection during pregnancy in humans is associated with an increased incidence of congenital anomalies including microcephaly as well as fetal death and miscarriage and collectively has been referred to as Congenital Zika Syndrome (CZS). Animal models for ZIKV infection in pregnancy have been developed including mice and non-human primates (NHPs). In macaques, fetal CZS outcomes from maternal ZIKV infection range from none to significant. In the present study we develop the olive baboon (Papio anubis), as a model for vertical transfer of ZIKV during pregnancy. Four mid-gestation, timed-pregnant baboons were inoculated with the French Polynesian ZIKV isolate (104 ffu). This study specifically focused on the acute phase of vertical transfer. Dams were terminated at 7 days post infection (dpi; n = 1), 14 dpi (n = 2) and 21 dpi (n = 1). All dams exhibited mild to moderate rash and conjunctivitis. Viremia peaked at 5-7 dpi with only one of three dams remaining mildly viremic at 14 dpi. An anti-ZIKV IgM response was observed by 14 dpi in all three dams studied to this stage, and two dams developed a neutralizing IgG response by either 14 dpi or 21 dpi, the latter included transfer of the IgG to the fetus (cord blood). A systemic inflammatory response (increased IL2, IL6, IL7, IL15, IL16) was observed in three of four dams. Vertical transfer of ZIKV to the placenta was observed in three pregnancies (n = 2 at 14 dpi and n = 1 at 21 dpi) and ZIKV was detected in fetal tissues in two pregnancies: one associated with fetal death at ~14 dpi, and the other in a viable fetus at 21 dpi. ZIKV RNA was detected in the fetal cerebral cortex and other tissues of both of these fetuses. In the fetus studied at 21 dpi with vertical transfer of virus to the CNS, the frontal cerebral cortex exhibited notable defects in radial glia, radial glial fibers, disorganized migration of immature neurons to the cortical layers, and signs of pathology in immature oligodendrocytes. In addition, indices of pronounced neuroinflammation were observed including astrogliosis, increased microglia and IL6 expression. Of interest, in one fetus examined at 14 dpi without detection of ZIKV RNA in brain and other fetal tissues, increased neuroinflammation (IL6 and microglia) was observed in the cortex. Although the placenta of the 14 dpi dam with fetal death showed considerable pathology, only minor pathology was noted in the other three placentas. ZIKV was detected immunohistochemically in two placentas (14 dpi) and one placenta at 21 dpi but not at 7 dpi. This is the first study to examine the early events of vertical transfer of ZIKV in a NHP infected at mid-gestation. The baboon thus represents an additional NHP as a model for ZIKV induced brain pathologies to contrast and compare to humans as well as other NHPs.


Cerebral Cortex/pathology , Zika Virus Infection/pathology , Zika Virus/pathogenicity , Animals , Brain/pathology , Cerebral Cortex/injuries , Cerebral Cortex/virology , Disease Models, Animal , Female , Fetal Death , Fetal Diseases/pathology , Fetus/virology , Infectious Disease Transmission, Vertical , Microcephaly , Papio anubis/microbiology , Papio anubis/virology , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/virology , Viremia , Zika Virus/genetics , Zika Virus Infection/virology
19.
J Neurovirol ; 25(2): 254-262, 2019 04.
Article En | MEDLINE | ID: mdl-30617850

Rapid maturation of major white matter pathways occurs in the first 2 years of life, indicating a critical neuronal developmental period. The impact of initiating antiretroviral therapy (ART) in children perinatally infected with HIV-1, after the age of 2 years on neurocognitive functioning and white matter development in adolescence has not been studied. Forty-six adolescents who initiated ART during the first 2 years of life (< 2 years) and 79 adolescents who initiated ART after 2 years of age (> 2 years), with perinatally acquired HIV were enrolled in the Cape Town Adolescent Antiretroviral Cohort. Adolescents completed a comprehensive neurocognitive battery testing a number of cognitive domains. Diffusion tensor imaging (DTI) was done to determine fractional anisotropy (FA), mean diffusivity (MD), axial diffusion (AD), and radial diffusion (RD) in a region of interest analysis. Neurocognitive performance was similar between adolescents who initiated ART < 2 years or > 2 years. There was a trend towards attention (p = .07) and working memory (p = .05) being poorer in the group who initiated ART > 2 years. FA was lower in the > 2-year group in the superior corona radiata (p = .03), and the external capsule (p = .04). MD was higher in the > 2-year group in the cerebral peduncle (p = .02), the superior corona radiata (p = .01), and the superior fronto-occipital fasciculus (p = .03). RD was higher in the > 2-year group in the superior corona radiata (p = .02), the cerebral peduncle (p = .01), and the superior fronto-occipital fasciculus (p = .01). However, the higher AD in the > 2-year group in the superior corona radiata was not in the expected direction (p = .01). Initiation of ART after the neuronal development period of the second postnatal year is associated with white matter alterations on neuroimaging.


Anti-HIV Agents/therapeutic use , Brain Stem/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Cognitive Dysfunction/drug therapy , HIV Infections/drug therapy , White Matter/diagnostic imaging , Antiretroviral Therapy, Highly Active , Attention/drug effects , Attention/physiology , Brain Stem/physiopathology , Brain Stem/virology , Cerebral Cortex/physiopathology , Cerebral Cortex/virology , Child , Child, Preschool , Cognition/drug effects , Cognition/physiology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/virology , Diffusion Tensor Imaging , Female , HIV Infections/diagnostic imaging , HIV Infections/physiopathology , HIV Infections/virology , Humans , Infant , Male , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Neuroimaging , Neuropsychological Tests , Prospective Studies , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , South Africa , Time Factors , White Matter/physiopathology , White Matter/virology
20.
J Neurovirol ; 25(1): 9-21, 2019 02.
Article En | MEDLINE | ID: mdl-30298203

It is yet unclear if people infected with human immunodeficiency virus (HIV+) on stable, combined antiretroviral therapies (cARTs) decline with age at the same or greater rate than healthy people. In this study, we examined independent and interactive effects of HIV, age, and HIV-related clinical parameters on neuropsychological functioning and brain regional volume in a sizable group of Polish HIV+ men receiving cART. We also estimated the impact of nadir CD4 cell count, CD4 cell count during participation in the study, duration of HIV infection, or duration of cART along with age. Ninety-one HIV+ and 95 control (HIV-) volunteers ages 23-75 completed a battery of neuropsychological tests, and 54 HIV+ and 62 HIV- of these volunteers participated in a brain imaging assessment. Regional brain volume in the cortical and subcortical regions was measured using voxel-based morphometry. We have found that HIV and older age were independently related to lower attention, working memory, nonverbal fluency, and visuomotor dexterity. Older age but not HIV was associated with less volume in several cortical and subcortical brain regions. In the oldest HIV+ participants, age had a moderating effect on the relationship between the duration of cART and visuomotor performance, such as that older age decreased speed of visuomotor performance along with every year on cART. Such results may reflect the efficacy of cART in preventing HIV-associated brain damage. They also highlight the importance of monitoring neuropsychological functioning and brain structure in HIV+ patients. This is particularly important in older patients with long adherence to cART.


Anti-HIV Agents/therapeutic use , Cerebral Cortex/physiopathology , HIV Infections/physiopathology , Adult , Age Factors , Aged , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Cerebral Cortex/virology , HIV Infections/diagnostic imaging , HIV Infections/drug therapy , HIV Infections/virology , Humans , Magnetic Resonance Imaging , Male , Memory, Short-Term/drug effects , Middle Aged , Neuroimaging , Neuropsychological Tests , Organ Size/drug effects
...