Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Virol ; 96(4): e0196921, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34935438

ABSTRACT

Unlike SARS-CoV-1 and MERS-CoV, infection with SARS-CoV-2, the viral pathogen responsible for COVID-19, is often associated with neurologic symptoms that range from mild to severe, yet increasing evidence argues the virus does not exhibit extensive neuroinvasive properties. We demonstrate SARS-CoV-2 can infect and replicate in human iPSC-derived neurons and that infection shows limited antiviral and inflammatory responses but increased activation of EIF2 signaling following infection as determined by RNA sequencing. Intranasal infection of K18 human ACE2 transgenic mice (K18-hACE2) with SARS-CoV-2 resulted in lung pathology associated with viral replication and immune cell infiltration. In addition, ∼50% of infected mice exhibited CNS infection characterized by wide-spread viral replication in neurons accompanied by increased expression of chemokine (Cxcl9, Cxcl10, Ccl2, Ccl5 and Ccl19) and cytokine (Ifn-λ and Tnf-α) transcripts associated with microgliosis and a neuroinflammatory response consisting primarily of monocytes/macrophages. Microglia depletion via administration of colony-stimulating factor 1 receptor inhibitor, PLX5622, in SARS-CoV-2 infected mice did not affect survival or viral replication but did result in dampened expression of proinflammatory cytokine/chemokine transcripts and a reduction in monocyte/macrophage infiltration. These results argue that microglia are dispensable in terms of controlling SARS-CoV-2 replication in in the K18-hACE2 model but do contribute to an inflammatory response through expression of pro-inflammatory genes. Collectively, these findings contribute to previous work demonstrating the ability of SARS-CoV-2 to infect neurons as well as emphasizing the potential use of the K18-hACE2 model to study immunological and neuropathological aspects related to SARS-CoV-2-induced neurologic disease. IMPORTANCE Understanding the immunological mechanisms contributing to both host defense and disease following viral infection of the CNS is of critical importance given the increasing number of viruses that are capable of infecting and replicating within the nervous system. With this in mind, the present study was undertaken to evaluate the role of microglia in aiding in host defense following experimental infection of the central nervous system (CNS) of K18-hACE2 with SARS-CoV-2, the causative agent of COVID-19. Neurologic symptoms that range in severity are common in COVID-19 patients and understanding immune responses that contribute to restricting neurologic disease can provide important insight into better understanding consequences associated with SARS-CoV-2 infection of the CNS.


Subject(s)
Angiotensin-Converting Enzyme 2/immunology , COVID-19/immunology , Central Nervous System Viral Diseases/immunology , Microglia/immunology , SARS-CoV-2/physiology , Virus Replication/immunology , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Central Nervous System/immunology , Central Nervous System/virology , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/virology , Chemokines/genetics , Chemokines/immunology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Microglia/virology , Neurons/immunology , Neurons/virology , Virus Replication/genetics
2.
mBio ; 12(6): e0271221, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34781742

ABSTRACT

Poliomyelitis-like illness is a common clinical manifestation of neurotropic viral infections. Functional loss and death of motor neurons often lead to reduced muscle tone and paralysis, causing persistent motor sequelae among disease survivors. Despite several reports demonstrating the molecular basis of encephalopathy, the pathogenesis behind virus-induced flaccid paralysis remained largely unknown. The present study for the first time aims to elucidate the mechanism responsible for limb paralysis by studying clinical isolates of Japanese encephalitis virus (JEV) and Chandipura virus (CHPV) responsible for causing acute flaccid paralysis (AFP) in vast regions of Southeast Asia and the Indian subcontinent. An experimental model for studying virus-induced AFP was generated by intraperitoneal injection of 10-day-old BALB/c mice. Progressive decline in motor performance of infected animals was observed, with paralysis being correlated with death of motor neurons (MNs). Furthermore, we demonstrated that upon infection, MNs undergo an extrinsic apoptotic pathway in a RIG-I-dependent fashion via transcription factors pIRF-3 and pIRF-7. Both gene-silencing experiments using specific RIG-I-short interfering RNA and in vivo morpholino abrogated cellular apoptosis, validating the important role of pattern recognition receptor (PRR) RIG-I in MN death. Hence, from our experimental observations, we hypothesize that host innate response plays a significant role in deterioration of motor functioning upon neurotropic virus infections. IMPORTANCE Neurotropic viral infections are an increasingly common cause of immediate or delayed neuropsychiatric sequelae, cognitive impairment, and movement disorders or, in severe cases, death. Given the highest reported disability-adjusted life years and mortality rate worldwide, a better understanding of molecular mechanisms for underlying clinical manifestations like AFP will help in development of more effective tools for therapeutic solutions.


Subject(s)
Central Nervous System Viral Diseases/metabolism , Central Nervous System Viral Diseases/physiopathology , DEAD Box Protein 58/metabolism , Encephalitis Virus, Japanese/physiology , Motor Neurons/cytology , Myelitis/metabolism , Myelitis/physiopathology , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/physiopathology , Vesiculovirus/physiology , Animals , Cell Death , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/virology , DEAD Box Protein 58/genetics , Encephalitis Virus, Japanese/genetics , Female , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Male , Mice , Motor Activity , Motor Neurons/metabolism , Motor Neurons/virology , Myelitis/genetics , Myelitis/virology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/virology , Vesiculovirus/genetics
3.
Int J Mol Sci ; 22(17)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34502105

ABSTRACT

The human brain and central nervous system (CNS) harbor a select sub-group of potentially pathogenic microRNAs (miRNAs), including a well-characterized NF-kB-sensitive Homo sapiens microRNA hsa-miRNA-146a-5p (miRNA-146a). miRNA-146a is significantly over-expressed in progressive and often lethal viral- and prion-mediated and related neurological syndromes associated with progressive inflammatory neurodegeneration. These include ~18 different viral-induced encephalopathies for which data are available, at least ~10 known prion diseases (PrD) of animals and humans, Alzheimer's disease (AD) and other sporadic and progressive age-related neurological disorders. Despite the apparent lack of nucleic acids in prions, both DNA- and RNA-containing viruses along with prions significantly induce miRNA-146a in the infected host, but whether this represents part of the host's adaptive immunity, innate-immune response or a mechanism to enable the invading prion or virus a successful infection is not well understood. Current findings suggest an early and highly interactive role for miRNA-146a: (i) as a major small noncoding RNA (sncRNA) regulator of innate-immune responses and inflammatory signaling in cells of the human brain and CNS; (ii) as a critical component of the complement system and immune-related neurological dysfunction; (iii) as an inducible sncRNA of the brain and CNS that lies at a critical intersection of several important neurobiological adaptive immune response processes with highly interactive associations involving complement factor H (CFH), Toll-like receptor pathways, the innate-immunity, cytokine production, apoptosis and neural cell decline; and (iv) as a potential biomarker for viral infection, TSE and AD and other neurological diseases in both animals and humans. In this report, we review the recent data supporting the idea that miRNA-146a may represent a novel and unique sncRNA-based biomarker for inflammatory neurodegeneration in multiple species. This paper further reviews the current state of knowledge regarding the nature and mechanism of miRNA-146a in viral and prion infection of the human brain and CNS with reference to AD wherever possible.


Subject(s)
Brain/pathology , Central Nervous System Viral Diseases/immunology , Gene Expression Regulation/immunology , MicroRNAs/metabolism , Prion Diseases/immunology , Apoptosis/genetics , Apoptosis/immunology , Biomarkers/analysis , Biomarkers/metabolism , Brain/immunology , Brain/virology , Central Nervous System Viral Diseases/diagnosis , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/virology , Complement Factor H/metabolism , Cytokines/metabolism , Humans , MicroRNAs/analysis , MicroRNAs/genetics , NF-kappa B/metabolism , Prion Diseases/diagnosis , Prion Diseases/genetics , Prion Diseases/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/metabolism
4.
Nat Med ; 25(11): 1748-1752, 2019 11.
Article in English | MEDLINE | ID: mdl-31636453

ABSTRACT

Since 2012, the United States of America has experienced a biennial spike in pediatric acute flaccid myelitis (AFM)1-6. Epidemiologic evidence suggests non-polio enteroviruses (EVs) are a potential etiology, yet EV RNA is rarely detected in cerebrospinal fluid (CSF)2. CSF from children with AFM (n = 42) and other pediatric neurologic disease controls (n = 58) were investigated for intrathecal antiviral antibodies, using a phage display library expressing 481,966 overlapping peptides derived from all known vertebrate and arboviruses (VirScan). Metagenomic next-generation sequencing (mNGS) of AFM CSF RNA (n = 20 cases) was also performed, both unbiased sequencing and with targeted enrichment for EVs. Using VirScan, the viral family significantly enriched by the CSF of AFM cases relative to controls was Picornaviridae, with the most enriched Picornaviridae peptides belonging to the genus Enterovirus (n = 29/42 cases versus 4/58 controls). EV VP1 ELISA confirmed this finding (n = 22/26 cases versus 7/50 controls). mNGS did not detect additional EV RNA. Despite rare detection of EV RNA, pan-viral serology frequently identified high levels of CSF EV-specific antibodies in AFM compared with controls, providing further evidence for a causal role of non-polio EVs in AFM.


Subject(s)
Central Nervous System Viral Diseases/genetics , Enterovirus Infections/genetics , Enterovirus/genetics , Myelitis/genetics , Neuromuscular Diseases/genetics , Seroepidemiologic Studies , Antibodies, Viral/cerebrospinal fluid , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Central Nervous System Viral Diseases/cerebrospinal fluid , Central Nervous System Viral Diseases/epidemiology , Central Nervous System Viral Diseases/virology , Child, Preschool , Enterovirus/pathogenicity , Enterovirus Infections/cerebrospinal fluid , Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Female , Humans , Infant , Male , Myelitis/cerebrospinal fluid , Myelitis/epidemiology , Myelitis/virology , Neuromuscular Diseases/cerebrospinal fluid , Neuromuscular Diseases/epidemiology , Neuromuscular Diseases/virology , United States
5.
Genes (Basel) ; 10(5)2019 04 30.
Article in English | MEDLINE | ID: mdl-31052348

ABSTRACT

Identifying the causative pathogen in central nervous system (CNS) infections is crucial for patient management and prognosis. Many viruses can cause CNS infections, yet screening for each individually is costly and time-consuming. Most metagenomic assays can theoretically detect all pathogens, but often fail to detect viruses because of their small genome and low viral load. Viral metagenomics overcomes this by enrichment of the viral genomic content in a sample. VIDISCA-NGS is one of the available workflows for viral metagenomics, which requires only a small input volume and allows multiplexing of multiple samples per run. The performance of VIDISCA-NGS was tested on 45 cerebrospinal fluid (CSF) samples from patients with suspected CNS infections in which a virus was identified and quantified by polymerase chain reaction. Eighteen were positive for an RNA virus, and 34 for a herpesvirus. VIDISCA-NGS detected all RNA viruses with a viral load >2 × 104 RNA copies/mL (n = 6) and 8 of 12 of the remaining low load samples. Only one herpesvirus was identified by VIDISCA-NGS, however, when withholding a DNase treatment, 11 of 18 samples with a herpesvirus load >104 DNA copies/mL were detected. Our results indicate that VIDISCA-NGS has the capacity to detect low load RNA viruses in CSF. Herpesvirus DNA in clinical samples is probably non-encapsidated and therefore difficult to detect by VIDISCA-NGS.


Subject(s)
Central Nervous System Viral Diseases/cerebrospinal fluid , Genome, Viral/genetics , RNA, Viral/cerebrospinal fluid , Viruses/isolation & purification , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/virology , High-Throughput Nucleotide Sequencing , Humans , Metagenomics , Viral Load/genetics , Viruses/genetics , Viruses/pathogenicity
6.
J Neuroimmunol ; 308: 25-29, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28187911

ABSTRACT

Central nervous system consequences of viral infections are rare, but when they do occur, they are often serious and clinically challenging to manage. Our awareness of the perils of neuroinvasion by viruses is growing: the recently appreciated impact of Ebola and Zika virus infections on CNS integrity, decreases in vaccination coverage for potentially neurotropic viruses such as measles, and increased neurovirulence of some influenza strains collectively highlight the need for a better understanding of the viral-neural interaction. Defining these interactions and how they result in neuropathogenesis is paramount for the development of better clinical strategies, especially given the limited treatment options that are available due to the unique physiology of the brain that limits migration of blood-borne molecules into the CNS parenchyma. In this perspective, we discuss some unique aspects of neuronal viral infections and immune-mediated control that impact the pathogenic outcomes of these infections. Further, we draw attention to an often overlooked aspect of neuropathogenesis research: that lack of overt disease, which is often equated with survival post-infection, likely only scratches the surface of the myriad ways by which neurotropic infections can impair CNS function.


Subject(s)
Central Nervous System Viral Diseases/mortality , Central Nervous System/pathology , Kaplan-Meier Estimate , Animals , Central Nervous System/virology , Central Nervous System Viral Diseases/genetics , Disease Models, Animal , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/genetics , Mice , Mice, Transgenic , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics
7.
Neurotherapeutics ; 13(3): 547-54, 2016 07.
Article in English | MEDLINE | ID: mdl-27150390

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/therapy , Gene Editing/methods , Genetic Therapy/methods , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/therapy , HIV Infections/genetics , HIV Infections/therapy , Herpesvirus 1, Human/genetics , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Leukoencephalopathy, Progressive Multifocal/therapy
8.
Brain Pathol ; 25(5): 600-4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26276023

ABSTRACT

The range of viruses implicated in central nervous system disease continues to grow with globalization of travel and trade, emergence and reemergence of zoonoses and investments in discovery science. Diagnosis of viral central nervous system infections is challenging in that brain tissue, where the pathogen concentration is likely to be highest, is not readily obtained and sensitive methods for molecular and serological detection of infection are not available in most clinical microbiology laboratories. Here we review these challenges and discuss how they may be addressed using advances in molecular, proteomic and immunological methods.


Subject(s)
Brain/virology , Central Nervous System Viral Diseases/diagnosis , Animals , Central Nervous System Viral Diseases/genetics , Humans , Polymerase Chain Reaction , Proteomics , Serologic Tests
9.
Curr Opin Immunol ; 36: 47-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26163762

ABSTRACT

The innate immune system mediates protection against neurotropic viruses that replicate in the central nervous system (CNS). Virus infection within specific cells of the CNS triggers activation of several families of pattern recognition receptors including Toll-like receptors, retinoic acid-inducible gene I like receptors, nucleotide-binding oligomerization domain-like receptors, and cytosolic DNA sensors. In this review, we highlight recent advances in our understanding of how cell-intrinsic host defenses within the CNS modulate infection of different DNA and RNA viruses.


Subject(s)
Central Nervous System Viral Diseases/immunology , Central Nervous System Viral Diseases/virology , Host-Pathogen Interactions , Immunity, Innate , Immunomodulation , Virus Diseases/immunology , Virus Diseases/virology , Animals , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , MicroRNAs/genetics , Receptors, Immunologic/metabolism , Signal Transduction , Transcription Factors/metabolism , Virus Diseases/genetics , Virus Diseases/metabolism
10.
J Interferon Cytokine Res ; 35(5): 373-84, 2015 May.
Article in English | MEDLINE | ID: mdl-25536401

ABSTRACT

Human immunodeficiency virus-1 (HIV-1)-infected monocytes/macrophages and microglia release increased levels of proinflammatory cytokines and chemokines, including ELR+ (containing glutamic acid-leucine-arginine motif) chemokines. To investigate the role of HIV-1 infection on chemokine regulation, monocyte-derived macrophages (MDMs) from normal donors were infected with HIV-1 and the expression of chemokines and their downstream biological functions were evaluated. Among the tested chemokines, CXCL5 was upregulated significantly both at the mRNA and protein level in the HIV-1-infected MDMs compared with mock-infected cultures. Upregulation of CXCL5 in the HIV-1-infected MDMs is, in part, regulated by increased interleukin-1ß (IL-1ß) production and phosphorylation of ERK1/2. Functional analyses indicate that HIV-1-induced overexpression of CXCL5 has enhanced the ability to attract neutrophils, as observed by chemotaxis assay. However, exposure of NT2, SH-SY5Y cells, and primary neurons to HIV-1-infected MDM supernatants resulted in cell death that was not rescued by anti-CXCL5 antibody suggesting that CXCL5 does not have direct effect on neuronal death. Together, these results suggest that the increased level of CXCL5 in tissue compartments, including the central nervous system of HIV-1-infected individuals might alter the inflammatory response through the infiltration of neutrophils into tissue compartment, thus causing secondary effects on resident cells.


Subject(s)
Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/virology , Chemokine CXCL5/genetics , Gene Expression Regulation , HIV-1/physiology , Macrophages/metabolism , Macrophages/virology , Transcription, Genetic , Cell Line , Cell Survival/genetics , Central Nervous System Viral Diseases/immunology , Central Nervous System Viral Diseases/metabolism , Chemokine CXCL5/metabolism , Chemokines/genetics , Chemokines/metabolism , Gene Expression Profiling , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , MAP Kinase Signaling System , Macrophages/immunology , Neurons/metabolism , Neutrophil Infiltration/immunology , Virus Replication
11.
Comp Immunol Microbiol Infect Dis ; 34(6): 503-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22005334

ABSTRACT

The present study was focused on the modulation of innate immune response genes in CNS of mouse in response to rabies virus (RABV) infection. The global gene expression changes in brains of RABV- or mock-infected mice were investigated using DNA microarray analysis and quantitative real-time PCR. Then functional enrichment of the differentially expressed mRNAs was performed. Microarray analysis showed that 390 genes in brain were significantly (P<0.01) regulated in response to RABV infection, with obviously up-regulated genes like interferon (IFN) stimulated genes (ISGs), IFN inducible transcription factors, cytokines and complement, etc. The significant pathways of differentially expressed genes are mainly involved in JAK-STAT signaling pathway, antigen processing and presentation, ubiquitin mediated proteolysis and complement cascades. The results suggest that the modulated genes in infected CNS were possibly involved in pathogenesis of rabies. Conversely, they may have protective effects.


Subject(s)
Brain/immunology , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/immunology , Immunity, Innate/genetics , Rabies virus/isolation & purification , Rabies/genetics , Rabies/immunology , Animals , Brain/metabolism , Brain/pathology , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Inbred BALB C , Signal Transduction , Viral Load
12.
AIDS ; 25(17): 2057-67, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-21857495

ABSTRACT

OBJECTIVE: Plasma microRNAs (miRNAs) are modulated during disease and are emerging biomarkers; they have not been characterized in HIV infection. Using our macaque/simian immunodeficiency virus (SIV) model of HIV, we sought to identify a plasma miRNA profile of acute lentiviral infection, evaluate its relationship with known cellular and viral determinants of lentivirus-associated central nervous system (CNS) disease, and explore the potential of miRNAs to predict CNS disease. DESIGN: Plasma samples were obtained before inoculation and 10 days after inoculation from SIV-infected macaques. METHODS: Plasma miRNA expression profiles were determined by TaqMan low-density array for six individuals. miRNA expression was compared with levels of cytokines, virus, and plasma platelet count. miRNA results were confirmed by single miRNA-specific assays for 10 macaques. Nineteen individuals were used to validate a disease prediction test. RESULTS: A 45-miRNA signature of acute infection (differential expression with P < 0.05 after multiple comparison correction) classified plasma as infected or not. Several differentially expressed miRNAs correlated with CNS disease-associated cytokines interleukin-6 and CCL2 and included predicted and/or validated regulators of the corresponding mRNAs. miRNAs tracked with viral load and platelet count were also predictors of CNS disease. At least six miRNAs were significantly differentially expressed in individuals with severe versus no CNS disease; in an unweighted expression test, they predicted CNS disease. CONCLUSION: Acute-phase differential expression of plasma miRNAs predicts CNS disease and suggests that CNS damage or predisposition to disease progression begins in the earliest phase of infection. Plasma miRNAs should be investigated further as leading indicators of HIV diseases as early as acute infection.


Subject(s)
Central Nervous System Viral Diseases/blood , Chemokine CCL2/blood , Interleukin-6/blood , MicroRNAs/blood , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/blood , Animals , Biomarkers/blood , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/physiopathology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Macaca , Polymerase Chain Reaction , Predictive Value of Tests , RNA, Messenger/blood , Sequence Analysis, RNA , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/physiopathology , Virus Replication
13.
Curr Neurol Neurosci Rep ; 10(2): 147-54, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20425240

ABSTRACT

Viral infections of the pediatric central nervous system (CNS) encompass a broad spectrum of both perinatally and postnatally acquired diseases with potentially devastating effects on the developing brain. In children, viral infections have been associated with chronic encephalopathy, encephalitis, demyelinating disease, tumors, and epilepsy. Older diagnostic techniques of biopsy, viral culture, electron microscopy, gel-based polymerase chain reaction (PCR), and viral titer quantification are being replaced with more rapid, sensitive, and specific real-time and microarray-based PCR technologies. Advances in neuroimaging technologies have provided for earlier recognition of CNS injury without elucidation of specific viral etiology. Although the mainstay therapy of many pediatric neurovirologic diseases, aside from HIV, includes intravenous acyclovir, much work is being done to develop novel antiviral immunotherapies aimed at both treating and preventing pediatric CNS viral disease.


Subject(s)
Central Nervous System Viral Diseases , Developmental Disabilities/virology , Pediatrics , Central Nervous System Viral Diseases/diagnosis , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/therapy , Humans , Magnetic Resonance Imaging/methods
15.
Curr Opin Pharmacol ; 8(4): 472-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18562249

ABSTRACT

Viral infections of the central nervous system (CNS) necessitate rapid, yet tightly controlled responses to contain viral spread while limiting tissue damage. All CNS resident cell types are equipped with pattern recognition receptors (PRRs) to respond to viruses. The resulting activation of IFN-alpha/beta, pro-inflammatory cytokines and chemokines is dependent on the virus replication strategy, tropism and PRR distribution. Although IFN-alpha/beta induced antiviral mediators are essential to restrict initial viral spread, adaptive immunity promoted by chemokines, cytokines and metalloproteinases is equally crucial in lowering viral burden. Recognition of viral antigen presented by MHC molecules is crucial for T cell retention and function. Non-lytic clearance mechanisms mediated by IFN-gamma and antibodies prevail in providing protection. Targeted intervention can be achieved by PRR stimulation, chemokine-receptor blockade and immune modulation of T cell function. However, owing to the extensive positive and negative feedback signaling cascades linking innate and adaptive immune responses, enhanced anti-viral functions will have to be counterbalanced to avoid pathology.


Subject(s)
Central Nervous System Viral Diseases/immunology , Immunity, Cellular/physiology , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Central Nervous System Viral Diseases/drug therapy , Central Nervous System Viral Diseases/genetics , Chemokines/physiology , Humans , Immunity, Cellular/genetics , Immunologic Factors/genetics , Immunologic Factors/physiology , Inflammation Mediators/physiology , Interferons/immunology , Interferons/physiology , Receptors, Chemokine/physiology , Receptors, Pattern Recognition/physiology
16.
J Neuroimmune Pharmacol ; 2(1): 20-5, 2007 Mar.
Article in English | MEDLINE | ID: mdl-18040822

ABSTRACT

On May 15 and 16, 2006, the National Institute of Mental Health sponsored a workshop designed to facilitate discussion and collaboration between basic and clinical investigators in neuroAIDS. Day 2 of the meeting featured a series of talks by clinical investigators. Three of these (those by J. McArthur, P. Tebas, and C. Flexner) are described in detail here. The remaining four talks (those by R. Ellis, R. Price, S. Letendre, and R. Robertson) are briefly introduced here, and full-length papers may be found elsewhere in this issue of the Journal.


Subject(s)
Central Nervous System Viral Diseases/drug therapy , Clinical Trials as Topic/methods , Clinical Trials as Topic/trends , HIV Infections/drug therapy , Animals , Antiretroviral Therapy, Highly Active/methods , Antiretroviral Therapy, Highly Active/trends , Central Nervous System Viral Diseases/epidemiology , Central Nervous System Viral Diseases/genetics , Disease Management , HIV Infections/epidemiology , HIV Infections/genetics , Humans
17.
J Neurosurg ; 107(1): 136-44, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17639883

ABSTRACT

OBJECT: The purpose of this study was to evaluate the gene transfer capability and tolerability of plasmid DNA/polyethylenimine (PEI) complexes in comparison with adenovirus and naked plasmid DNA in the canine brain. METHODS: Plasmid or adenoviral vectors encoding firefly luciferase were injected directly into the cerebral parenchyma of five adult dogs at varying doses and volumes. Serial physical and neurological examinations, as well as blood and cerebrospinal fluid (CSF) analyses, were conducted before and after the surgery for 3 days. Three days after gene delivery, a luciferase activity assay and immunofluorescence analysis were used to test the brain tissue for gene expression. RESULTS: Injection into the brain parenchyma resulted in gene transfer throughout the cerebrum with every vector tested. Luciferase expression was highest when adenovirus vectors were used. Injection of plasmid DNA/PEI complexes and naked DNA resulted in similar levels of luciferase expression, which were on average 0.5 to 1.5% of the expression achieved with adenovirus vectors. Immunofluorescent microscopy analysis revealed that plasmid DNA/PEI complexes transduced mainly neurons, whereas adenovirus transduced mainly astrocytes. No significant acute side effects or neurological complications were observed in any of the dogs. Mononuclear cell counts significantly increased in the CSF after adenovirus injection and modestly increased after injection of plasmid DNA/PEI complexes, suggesting that a mild, acute inflammatory response occurred in the central nervous system (CNS). CONCLUSIONS: Compared with rodent models that are limited by very small brains, the dog is an excellent preclinical model in which to assess the distribution and safety of emerging gene transfer technologies. In this study, short-term gene transfer was evaluated as a prelude to long-term expression and safety studies. The authors conclude that the viral and nonviral vectors tested were well tolerated and effective at mediating gene transfer throughout a large portion of the canine brain. The nonviral plasmid vectors were less effective than adenovirus, yet they still achieved appreciable gene expression levels. Due to reduced gene transfer efficiency relative to viral vectors, nonviral vectors may be most useful when the expressed protein is secreted or exerts a bystander effect. Nonviral vectors offer an alternative means to genetically modify cells within the CNS of large mammals.


Subject(s)
Adenoviruses, Canine/genetics , Gene Transfer Techniques/instrumentation , Genetic Therapy/instrumentation , Plasmids/genetics , Animals , Astrocytes/cytology , Astrocytes/virology , Blood Chemical Analysis , Brain/cytology , Brain/enzymology , Brain/virology , Brain Neoplasms/therapy , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/pathology , Central Nervous System Viral Diseases/virology , Dogs , Feasibility Studies , Genetic Vectors/genetics , Glioma/therapy , Inflammation/pathology , Inflammation/virology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Neurons/virology , Plasmids/physiology , Polyethyleneimine/therapeutic use , Transduction, Genetic/methods , Vaccines, DNA/genetics
18.
Neuromolecular Med ; 8(4): 547-65, 2006.
Article in English | MEDLINE | ID: mdl-17028375

ABSTRACT

Lissencephaly 1 (LIS1) was the first gene implicated in the pathogenesis of type-1 lissencephaly. More than a decade of research by multiple laboratories has revealed that LIS1 is a key node protein, which participates in several pathways, including association with the molecular motor cytoplasmic dynein, the reelin signaling pathway, and the platelet-activating factor pathway. Mutations in LIS1-interacting proteins, either in human, or in mouse models has suggested that LIS1 might play a role in the pathogenesis of numerous diseases such as male sterility, schizophrenia, neuronal degeneration, and viral infections.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Microtubule-Associated Proteins/genetics , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/metabolism , Classical Lissencephalies and Subcortical Band Heterotopias/metabolism , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mice , Microtubule-Associated Proteins/metabolism , Mutation , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Protein Binding , Reelin Protein , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction
19.
Biochem Biophys Res Commun ; 350(2): 444-9, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-17010311

ABSTRACT

The alpha and beta isoforms of rat Ca(2+)/calmodulin kinase II inhibitor (CaMKIINalpha/beta) expressed in brain or brain and testis, respectively, are potent inhibitors of Ca(2+)/calmodulin kinase II (CaMKII) in vitro. However, the regulation or function of CaMKIINalpha/beta in the central nervous system (CNS) is not known. In this study, we demonstrate that mouse CaMKIINalpha gene encodes a 2.9kb brain-specific transcript whose expression is downregulated in mouse brain during Japanese encephalitis virus (JEV) and rabies virus infection. The downregulation is specific for CaMKIINalpha but not CaMKIINbeta mRNA. In addition to these changes in CaMKIINalpha mRNA, distinct changes are also observed in the phosphorylation as well as subcellular localization of CaMKIIalpha leading to an increase in cytosolic CaMKII activity in JEV-infected mouse brain. The differential regulation of CaMKIIalpha and CaMKIINalpha during JEV infection suggests a possible role for these proteins in viral infection and/or virus-induced neuropathogenesis in the CNS.


Subject(s)
Brain/metabolism , Carrier Proteins/genetics , Central Nervous System Viral Diseases/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/metabolism , Central Nervous System Viral Diseases/enzymology , Down-Regulation , Encephalitis, Japanese/enzymology , Encephalitis, Japanese/genetics , Encephalitis, Japanese/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Rabies/genetics , Rabies/metabolism
20.
J Neurol Neurosurg Psychiatry ; 77(8): 938-42, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16844949

ABSTRACT

OBJECTIVE: To study cerebrospinal fluid (CSF) and serum samples from 34 consecutive patients suspected of having varicella-zoster virus (VZV) infection of the central nervous system (CNS). POPULATION AND METHODS: The patients were divided into three groups. The first group consisted of 27 patients with a rash in one to three dermatomes and clinical suspicion of meningitis and radiculitis; among them, three subgroups were distinguished according to the affected dermatome: ophthalmicus (n = 9), oticus (n = 11) and cervico-thoraco-lumbar zoster (n = 7). Four cases of zoster sine herpete (ZSH) were included in the second group: these patients presented with either radiculitis (n = 2) or meningoencephalitis (n = 2), without cutaneous eruption. The third group consisted of three patients with a generalised rash and encephalitis. A polymerase chain reaction (PCR) for VZV DNA and antigen-driven immunoblots for oligoclonal anti-VZV antibodies were carried out on all CSF samples. RESULTS: PCR of the CSF was positive in 44% of the patients from the first group, mainly within the first 7 days after eruption. In addition, intrathecal synthesis of anti-VZV antibodies was detected in 37% of patients, always after an interval of 7 days (p<0.0001). Among the four patients with ZSH, a positive VZV PCR was detected in three patients and CSF-specific oligoclonal anti-VZV antibodies in two. PCR was also positive in the CSF of two of the three patients with generalised rash and encephalitis; local production of anti-VZV antibodies was seen in a second CSF sample in one patient, and was also present in the third patient. CONCLUSION: Amplification of VZV DNA by PCR in the CSF and antigen-driven immunoblots have important diagnostic value in suspected VZV infection, although their presence depends on the timing of the CSF sampling. VZV is thought to be a causative agent in unexplained cases of meningitis associated with radiculitis or focal CNS symptoms, even in the absence of skin manifestations. In such patients, rapid diagnosis by this combined approach permits early antiviral treatment.


Subject(s)
Central Nervous System Viral Diseases/genetics , Central Nervous System Viral Diseases/immunology , Herpes Zoster/genetics , Herpes Zoster/immunology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, Viral/analysis , Central Nervous System Viral Diseases/cerebrospinal fluid , Central Nervous System Viral Diseases/diagnosis , Cerebrospinal Fluid/virology , DNA, Viral/analysis , Female , Herpes Zoster/cerebrospinal fluid , Herpes Zoster/diagnosis , Humans , Immunoblotting , Male , Middle Aged , Oligoclonal Bands , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...