1
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
2
|
Rashidi AS, Tran DN, Peelen CR, van Gent M, Ouwendijk WJD, Verjans GMGM. Herpes simplex virus infection induces necroptosis of neurons and astrocytes in human fetal organotypic brain slice cultures. J Neuroinflammation 2024; 21:38. [PMID: 38302975 PMCID: PMC10832279 DOI: 10.1186/s12974-024-03027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Herpes simplex virus (HSV) encephalitis (HSE) is a serious and potentially life-threatening disease, affecting both adults and newborns. Progress in understanding the virus and host factors involved in neonatal HSE has been hampered by the limitations of current brain models that do not fully recapitulate the tissue structure and cell composition of the developing human brain in health and disease. Here, we developed a human fetal organotypic brain slice culture (hfOBSC) model and determined its value in mimicking the HSE neuropathology in vitro. METHODS Cell viability and tissues integrity were determined by lactate dehydrogenase release in supernatant and immunohistological (IHC) analyses. Brain slices were infected with green fluorescent protein (GFP-) expressing HSV-1 and HSV-2. Virus replication and spread were determined by confocal microscopy, PCR and virus culture. Expression of pro-inflammatory cytokines and chemokines were detected by PCR. Cell tropism and HSV-induced neuropathology were determined by IHC analysis. Finally, the in situ data of HSV-infected hfOBSC were compared to the neuropathology detected in human HSE brain sections. RESULTS Slicing and serum-free culture conditions were optimized to maintain the viability and tissue architecture of ex vivo human fetal brain slices for at least 14 days at 37 °C in a CO2 incubator. The hfOBSC supported productive HSV-1 and HSV-2 infection, involving predominantly infection of neurons and astrocytes, leading to expression of pro-inflammatory cytokines and chemokines. Both viruses induced programmed cell death-especially necroptosis-in infected brain slices at later time points after infection. The virus spread, cell tropism and role of programmed cell death in HSV-induced cell death resembled the neuropathology of HSE. CONCLUSIONS We developed a novel human brain culture model in which the viability of the major brain-resident cells-including neurons, microglia, astrocytes and oligodendrocytes-and the tissue architecture is maintained for at least 2 weeks in vitro under serum-free culture conditions. The close resemblance of cell tropism, spread and neurovirulence of HSV-1 and HSV-2 in the hfOBSC model with the neuropathological features of human HSE cases underscores its potential to detail the pathophysiology of other neurotropic viruses and as preclinical model to test novel therapeutic interventions.
Collapse
Affiliation(s)
- Ahmad S Rashidi
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Diana N Tran
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Caithlin R Peelen
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Michiel van Gent
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Werner J D Ouwendijk
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Georges M G M Verjans
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Shen S, Wei R, Gao Y, Yang X, Zhang G, Yan B, Xiao Z, Li J. Cortical atrophy in early-stage patients with anti-NMDA receptor encephalitis: a machine-learning MRI study with various feature extraction. Cereb Cortex 2024; 34:bhad499. [PMID: 38185983 DOI: 10.1093/cercor/bhad499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Conventional brain magnetic resonance imaging (MRI) of anti-N-methyl-D-aspartate-receptor encephalitis (NMDARE) is non-specific, thus showing little differential diagnostic value, especially for MRI-negative patients. To characterize patterns of structural alterations and facilitate the diagnosis of MRI-negative NMDARE patients, we build two support vector machine models (NMDARE versus healthy controls [HC] model and NMDARE versus viral encephalitis [VE] model) based on radiomics features extracted from brain MRI. A total of 109 MRI-negative NMDARE patients in the acute phase, 108 HCs and 84 acute MRI-negative VE cases were included for training. Another 29 NMDARE patients, 28 HCs and 26 VE cases were included for validation. Eighty features discriminated NMDARE patients from HCs, with area under the receiver operating characteristic curve (AUC) of 0.963 in validation set. NMDARE patients presented with significantly lower thickness, area, and volume and higher mean curvature than HCs. Potential atrophy predominately presented in the frontal lobe (cumulative weight = 4.3725, contribution rate of 29.86%), and temporal lobe (cumulative weight = 2.573, contribution rate of 17.57%). The NMDARE versus VE model achieved certain diagnostic power, with AUC of 0.879 in validation set. Our research shows potential atrophy across the entire cerebral cortex in acute NMDARE patients, and MRI machine learning model has a potential to facilitate the diagnosis MRI-negative NMDARE.
Collapse
Affiliation(s)
- Sisi Shen
- Department of Neurology, West China Hospital of Sichuan University, 37 GuoXue Alley, Chengdu 610041, China
| | - Ran Wei
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Yu Gao
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Xinyuan Yang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Guoning Zhang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Bo Yan
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Zhuoling Xiao
- School of Information and Communication Engineering, University of Electronic Science and Technology of China No.2006, Xiyuan Ave, West Hi-Tech Zone, 611731| Chengdu, Sichuan, P.R. China
| | - Jinmei Li
- Department of Neurology, West China Hospital of Sichuan University, 37 GuoXue Alley, Chengdu 610041, China
| |
Collapse
|
4
|
Hodzic E, Hasbun R, Granillo A, Tröscher AR, Wagner H, von Oertzen TJ, Wagner JN. Steroids for the treatment of viral encephalitis: a systematic literature review and meta-analysis. J Neurol 2023:10.1007/s00415-023-11715-0. [PMID: 37060361 PMCID: PMC10105360 DOI: 10.1007/s00415-023-11715-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Specific antiviral treatment is only available for a small subset of viral encephalitis (VE). Adjunctive steroids are used, but there is scant evidence evaluating its utility. We present a systematic review and meta-analysis on the outcome of steroid use in VE. METHODS We conducted a systematic literature review and reported it according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standards. Two observational studies from unpublished or partially published data were added. For the meta-analysis, we employed the metaphor package of the statistical software R-4.3.1. RESULTS We screened 378 studies and included 50. 155 patients were added from the Houston and Linz cohorts. Individual data were available for 281 persons, 120 (43%) of whom received steroids. The most common pathogens were herpes simplex virus 1, West Nile virus, and measles. Study designs and patient outcomes were heterogeneous. Only three of the trials report an advantage of steroid therapy. Steroid-induced side effects were scarce. Ten cohorts were included into the meta-analysis. For the pooled data, the null hypothesis could not be rejected (p = 0.245) using a random effects model, i.e., a benefit of steroid treatment on survival in VE could not be shown. CONCLUSIONS Steroids as potent anti-inflammatory agents may act through a reduction of secondary inflammation-mediated damage. Our data do not support the use of steroids in VE. However, multiple shortcomings apply. Standardized controlled trials are needed to investigate optimal dosing and timing of steroid administration and to explore potential subgroups that could benefit.
Collapse
Affiliation(s)
- Emira Hodzic
- Institute of Applied Statistics, Johannes Kepler University, Linz, Austria
| | - Rodrigo Hasbun
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School, UT Health, Houston, TX, USA
| | - Alejandro Granillo
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School, UT Health, Houston, TX, USA
| | - Anna R Tröscher
- Department of Neurology, Evangelisches Klinikum Gelsenkirchen, Teaching Hospital University Duisburg-Essen, Munckelstrasse 27, 45879, Gelsenkirchen, Germany
| | - Helga Wagner
- Institute of Applied Statistics, Johannes Kepler University, Linz, Austria
| | - Tim J von Oertzen
- Department of Neurology, Evangelisches Klinikum Gelsenkirchen, Teaching Hospital University Duisburg-Essen, Munckelstrasse 27, 45879, Gelsenkirchen, Germany
- Johannes Kepler University, Linz, Austria
| | - Judith N Wagner
- Department of Neurology, Evangelisches Klinikum Gelsenkirchen, Teaching Hospital University Duisburg-Essen, Munckelstrasse 27, 45879, Gelsenkirchen, Germany.
- Johannes Kepler University, Linz, Austria.
| |
Collapse
|
5
|
Fontes-Dantas FL, Fernandes GG, Gutman EG, De Lima EV, Antonio LS, Hammerle MB, Mota-Araujo HP, Colodeti LC, Araújo SM, Froz GM, da Silva TN, Duarte LA, Salvio AL, Pires KL, Leon LA, Vasconcelos CCF, Romão L, Savio LEB, Silva JL, da Costa R, Clarke JR, Da Poian AT, Alves-Leon SV, Passos GF, Figueiredo CP. SARS-CoV-2 Spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep 2023; 42:112189. [PMID: 36857178 PMCID: PMC9935273 DOI: 10.1016/j.celrep.2023.112189] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Cognitive dysfunction is often reported in patients with post-coronavirus disease 2019 (COVID-19) syndrome, but its underlying mechanisms are not completely understood. Evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein or its fragments are released from cells during infection, reaching different tissues, including the CNS, irrespective of the presence of the viral RNA. Here, we demonstrate that brain infusion of Spike protein in mice has a late impact on cognitive function, recapitulating post-COVID-19 syndrome. We also show that neuroinflammation and hippocampal microgliosis mediate Spike-induced memory dysfunction via complement-dependent engulfment of synapses. Genetic or pharmacological blockage of Toll-like receptor 4 (TLR4) signaling protects animals against synapse elimination and memory dysfunction induced by Spike brain infusion. Accordingly, in a cohort of 86 patients who recovered from mild COVID-19, the genotype GG TLR4-2604G>A (rs10759931) is associated with poor cognitive outcome. These results identify TLR4 as a key target to investigate the long-term cognitive dysfunction after COVID-19 infection in humans and rodents.
Collapse
Affiliation(s)
- Fabricia L. Fontes-Dantas
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Gabriel G. Fernandes
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elisa G. Gutman
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emanuelle V. De Lima
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia S. Antonio
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana B. Hammerle
- Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hannah P. Mota-Araujo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lilian C. Colodeti
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Suzana M.B. Araújo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabrielle M. Froz
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Talita N. da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Larissa A. Duarte
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andreza L. Salvio
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karina L. Pires
- Neurology Department, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, RJ, Brazil
| | - Luciane A.A. Leon
- Laboratório de Desenvolvimento Tecnológico em Virologia, IOC/FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Luciana Romão
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luiz Eduardo B. Savio
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jerson L. Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Robson da Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R. Clarke
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andrea T. Da Poian
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Soniza V. Alves-Leon
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Division of Neurology, Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Giselle F. Passos
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Claudia P. Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| |
Collapse
|
6
|
Constant O, Maarifi G, Blanchet FP, Van de Perre P, Simonin Y, Salinas S. Role of Dendritic Cells in Viral Brain Infections. Front Immunol 2022; 13:862053. [PMID: 35529884 PMCID: PMC9072653 DOI: 10.3389/fimmu.2022.862053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
To gain access to the brain, a so-called immune-privileged organ due to its physical separation from the blood stream, pathogens and particularly viruses have been selected throughout evolution for their use of specific mechanisms. They can enter the central nervous system through direct infection of nerves or cerebral barriers or through cell-mediated transport. Indeed, peripheral lymphoid and myeloid immune cells can interact with the blood-brain and the blood-cerebrospinal fluid barriers and allow viral brain access using the "Trojan horse" mechanism. Among immune cells, at the frontier between innate and adaptive immune responses, dendritic cells (DCs) can be pathogen carriers, regulate or exacerbate antiviral responses and neuroinflammation, and therefore be involved in viral transmission and spread. In this review, we highlight an important contribution of DCs in the development and the consequences of viral brain infections.
Collapse
Affiliation(s)
- Orianne Constant
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Ghizlane Maarifi
- Institut de Recherche en Infectiologie de Montpellier, Centre national de la recherche scientifique (CNRS), Université de Montpellier, Montpellier, France
| | - Fabien P. Blanchet
- Institut de Recherche en Infectiologie de Montpellier, Centre national de la recherche scientifique (CNRS), Université de Montpellier, Montpellier, France
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Yannick Simonin
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Sara Salinas
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| |
Collapse
|
7
|
Yan J, Han VX, Heng B, Guillemin GJ, Bandodkar S, Dale RC. Development of a translational inflammation panel for the quantification of cerebrospinal fluid Pterin, Tryptophan-Kynurenine and Nitric oxide pathway metabolites. EBioMedicine 2022; 77:103917. [PMID: 35279631 PMCID: PMC8914118 DOI: 10.1016/j.ebiom.2022.103917] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 02/08/2023] Open
Abstract
Background Neuroinflammatory diseases such as encephalitis, meningitis, multiple sclerosis and other neurological diseases with inflammatory components, have demonstrated a need for diagnostic biomarkers to define treatable and reversible neuroinflammation. The development and clinical validation of a targeted translational inflammation panel using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) could provide early diagnosis, rapid treatment and insights into neuroinflammatory mechanisms. Methods An inflammation panel of 13 metabolites (neopterin, tryptophan, kynurenine, kynurenic acid, 3-hydroxykynurenine, xanthurenic acid, anthranilic acid, 3-hydroxyanthranilic acid, quinolinic acid, picolinic acid, arginine, citrulline and methylhistamine) was measured based on a simple precipitation and filtration method using minimal CSF volume. The chromatographic separation was achieved using the Acquity UPLC BEH C18 column in combination with a gradient elution within a 12-min time frame. Acute encephalitis (n=10; myelin oligodendrocyte glycoprotein encephalitis n=3, anti-N-methyl-D-aspartate encephalitis n=2, acute disseminated encephalomyelitis n=2, herpes simplex encephalitis n=1, enteroviral encephalitis n=1) and frequency-matched non-inflammatory neurological disease controls (n=10) were examined. Findings The method exhibited good sensitivity as the limits of quantification ranged between 0.75 and 3.00 ng mL−1, good linearity (r2 > 0.99), acceptable matrix effects (<± 19.4%) and high recoveries (89.8-109.1 %). There were no interferences observed from common endogenous CSF metabolites, no carryover and concordance with well-established clinical methods. The accuracy and precision for all analytes were within tolerances, at <± 15 mean relative error and < 15 % coefficient of variation respectively. All analytes in matrix-matched pooled human CSF calibrators and human CSF extracts were stable for 24 h after extraction and two freeze-thaw cycles. Interpretation The method was successfully applied to a pilot study investigating acute brain inflammation case-control groups. Statistical discrimination between encephalitis (n=10) and control groups (n=10) was achieved using orthogonal partial least squares discriminant analysis and heatmap cluster analysis. Statistical analysis of the measured metabolites identified significant alterations of seven metabolites in the tryptophan-kynurenine pathway (tryptophan, kynurenine, kynurenic acid, 3-hydroxykynurenine, anthranilic acid, 3-hydroxyanthranilic acid, quinolinic acid), arginine and neopterin in presence of acute neuroinflammation. Furthermore, elevated ratios of CSF kynurenine/tryptophan ratio, quinolinic acid/kynurenic acid and anthranilic acid/3-hydroxyanthranilic acid provided strong discriminative power for neuroinflammatory conditions. Studies of large groups of neurological diseases are required to explore the sensitivity and specificity of the inflammation panel. Funding Financial support for the study was granted by Dale NHMRC Investigator grant APP1193648, Petre Foundation, Cerebral Palsy Alliance and Department of Biochemistry at the Children's Hospital at Westmead.
Collapse
Affiliation(s)
- Jingya Yan
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia
| | - Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Benjamin Heng
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Sushil Bandodkar
- Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| |
Collapse
|
8
|
Caldwell M, Boruah AP, Thakur KT. Acute neurologic emerging flaviviruses. Ther Adv Infect Dis 2022; 9:20499361221102664. [PMID: 35719177 PMCID: PMC9198421 DOI: 10.1177/20499361221102664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/30/2022] [Indexed: 11/24/2022] Open
Abstract
The COVID-19 pandemic has shed light on the challenges we face as a global society in preventing and containing emerging and re-emerging pathogens. Multiple intersecting factors, including environmental changes, host immunological factors, and pathogen dynamics, are intimately connected to the emergence and re-emergence of communicable diseases. There is a large and expanding list of communicable diseases that can cause neurological damage, either through direct or indirect routes. Novel pathogens of neurotropic potential have been identified through advanced diagnostic techniques, including metagenomic next-generation sequencing, but there are also known pathogens which have expanded their geographic distribution to infect non-immune individuals. Factors including population growth, climate change, the increase in animal and human interface, and an increase in international travel and trade are contributing to the expansion of emerging and re-emerging pathogens. Challenges exist around antimicrobial misuse giving rise to antimicrobial-resistant infectious neurotropic organisms and increased susceptibility to infection related to the expanded use of immunomodulatory treatments. In this article, we will review key concepts around emerging and re-emerging pathogens and discuss factors associated with neurotropism and neuroinvasion. We highlight several neurotropic pathogens of interest, including West Nile virus (WNV), Zika Virus, Japanese Encephalitis Virus (JEV), and Tick-Borne Encephalitis Virus (TBEV). We emphasize neuroinfectious diseases which impact the central nervous system (CNS) and focus on flaviviruses, a group of vector-borne pathogens that have expanded globally in recent years and have proven capable of widespread outbreak.
Collapse
Affiliation(s)
- Marissa Caldwell
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Abhilasha P. Boruah
- Department of Neurology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital (CUIMC/NYP), New York, NY, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kiran T. Thakur
- Division of Critical Care and Hospitalist Neurology, Department of Neurology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital (CUIMC/NYP), 177 Fort Washington Avenue, Milstein Hospital, 8GS-300, New York, NY 10032, USA
| |
Collapse
|
9
|
Siciliano V, Rosà T, Del Vecchio P, D'Angelillo A, Brigida M, Longhitano Y, Zanza C, Santoro MC, Candelli M, Franceschi F, Piccioni A. Viral Encephalitis in Adults: A Narrative Review. Rev Recent Clin Trials 2022; 17:259-267. [PMID: 34792015 DOI: 10.2174/1574887116666211118141117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/14/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023]
Abstract
Viral infections of the central nervous system cause frequent hospitalization. The pathogenesis of viral encephalitis involves both the direct action of invading pathogens and the damage generated by the inflammatory reaction they trigger. The type of signs and symptoms presented by the patient depends on the severity and location of the ongoing inflammatory process. Most of the viral encephalitides are characterized by an acute development, fever, variable alterations in consciousness (confusion, lethargy, even coma), seizures (focal and generalized) and focal neurologic signs. The specific diagnosis of encephalitis is usually based on lumbar puncture. Cerebrospinal fluid examination should be performed in all patients unless absolutely contraindicated. Also, electroencephalogram and neuroimaging play a prominent role in diagnosis. Airway protection, ventilatory support, the management of raised intracranial pressure and correction of electrolyte disorders must be immediately considered in a patient with altered mental status. The only therapy strictly recommended is acyclovir in HSV encephalitis. The use of adjunctive glucocorticoids has poor-quality evidence in HSV, EBV, or VZV encephalitis. The role of antiviral therapy in other types of viral encephalitis is not well defined.
Collapse
Affiliation(s)
- Valentina Siciliano
- Department of Laboratory and Infectious Diseases, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Tommaso Rosà
- Department of Anesthesiology and Intensive Care Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Pierluigi Del Vecchio
- Department of Laboratory and Infectious Diseases, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Anna D'Angelillo
- Department of Laboratory and Infectious Diseases, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Mattia Brigida
- Department of Systems Medicine, Unit of Gastroenterology, Tor Vergata University, Rome, Italy
| | - Yaroslava Longhitano
- Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Department of Anesthesia and Critical Care, Alessandria, Italy
| | - Christian Zanza
- Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Department of Anesthesia and Critical Care, Alessandria, Italy
- Foundation of "Nuovo Ospedale Alba-Bra" and Department of Anesthesia, Critical Care and Emergency Medicine, Pietro and Michele Ferrero Hospital Verduno, Italy
- Department of Emergency Medicine, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Michele Cosimo Santoro
- Department of Emergency Medicine, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Marcello Candelli
- Department of Emergency Medicine, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| | - Andrea Piccioni
- Department of Emergency Medicine, University Polyclinic Foundation A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
10
|
Mesquita LP, Costa RC, Mesquita LLR, Lara MDCCSH, Villalobos EMC, Mori CMC, Mori E, Howerth EW, Maiorka PC. Pathogenesis of Equid Alphaherpesvirus 1 Infection in the Central Nervous System of Mice. Vet Pathol 2021; 58:1075-1085. [PMID: 34128432 DOI: 10.1177/03009858211020670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Equid alphaherpesvirus 1 (EHV-1) causes myeloencephalopathy in horses and occasionally in non-equid species. Although mouse models have been developed to understand EHV-1 pathogenesis, few EHV-1 strains have been identified as highly neurovirulent to mice. The aim of this study was to evaluate the pathogenesis of 2 neurovirulent EHV-1 strains in mice, and to characterize the inflammatory cells and expression of chemokines and the apoptosis marker caspase-3 in the brain of infected mice. C57BL/6J mice were inoculated intranasally with EHV-1 strains A4/72 or A9/92 and evaluated on 1, 2, and 3 days post inoculation (DPI). EHV-1-infected mice showed severe neurological signs at 3 DPI. Ultrastructural analysis revealed numerous viral nucleocapsids and fewer enveloped virions within degenerated and necrotic neurons and in the surrounding neuropil. Histologically, at 3 DPI, there was severe diffuse neuronal degeneration and liquefactive necrosis, prominent microgliosis, and perivascular cuffing composed of CD3+ cells (T cells) and Iba-1+ cells (macrophages), mainly in the olfactory bulb and ventral portions of the brain. In these areas, moderate numbers of neuroglial cells expressed CCL5 and CCL2 chemokines. Numerous neurons, including those in less affected areas, were immunolabeled for cleaved caspase-3. In conclusion, neurovirulent EHV-1 strains induced a fulminant necrotizing lymphohistiocytic meningoencephalitis in mice, with microgliosis and expression of chemokines and caspase-3. This model will be useful for understanding the mechanisms underlying the extensive neuropathology induced by these viral infections.
Collapse
Affiliation(s)
- Leonardo P Mesquita
- 28133University of Sao Paulo, Sao Paulo, Brazil
- 1355University of Georgia, Athens, GA, USA
| | | | | | | | | | | | - Enio Mori
- 27058Pasteur Institute, Sao Paulo, Brazil
| | | | | |
Collapse
|
11
|
Mesquita LP, Costa RC, Zanatto DA, Bruhn FRP, Mesquita LLR, Lara MCCSH, Villalobos EMC, Massoco CO, Mori CMC, Mori E, Maiorka PC. Equine herpesvirus 1 elicits a strong pro-inflammatory response in the brain of mice. J Gen Virol 2021; 102. [PMID: 33528354 DOI: 10.1099/jgv.0.001556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Equine herpesvirus type 1 (EHV-1) is an emerging pathogen that causes encephalomyelitis in horses and non-equid species. Several aspects of the immune response in the central nervous system (CNS), mainly regarding the role of inflammatory mediators during EHV-1 encephalitis, remain unknown. Moreover, understanding the mechanisms underlying extensive neuropathology induced by viruses would be helpful to establish therapeutic strategies. Therefore, we aimed to evaluate some aspects of the innate immune response during highly neurovirulent EHV-1 infection. C57BL/6 mice infected intranasally with A4/72 and A9/92 EHV-1 strains developed a fulminant neurological disease at 3 days post-inoculation with high viral titres in the brain. These mice developed severe encephalitis with infiltration of monocytes and CD8+ T cells to the brain. The inflammatory infiltrate followed the detection of the chemokines CCL2, CCL3, CCL4, CCL5, CXCL2, CXCL9 and CXCL-10 in the brain. Notably, the levels of CCL3, CCL4, CCL5 and CXCL9 were higher in A4/72-infected mice, which presented higher numbers of inflammatory cells within the CNS. Pro-inflammatory cytokines, such as interleukins (ILs) IL-1α, IL-1β, IL-6, IL-12β, and tumour necrosis factor (TNF), were also detected in the CNS, and Toll-like receptor (TLR) TLR2, TLR3 and TLR9 genes were also upregulated within the brain of EHV-1-infected mice. However, no expression of interferon-γ (IFN-γ) and IL-12α, which are important for controlling the replication of other herpesviruses, was detected in EHV-1-infected mice. The results show that the activated innate immune mechanisms could not prevent EHV-1 replication within the CNS, but most likely contributed to the extensive neuropathology. The mouse model of viral encephalitis proposed here will also be useful to study the mechanisms underlying extensive neuropathology.
Collapse
Affiliation(s)
- Leonardo P Mesquita
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Rafael C Costa
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Dennis A Zanatto
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Fábio R P Bruhn
- College of Veterinary Medicine, Federal University of Pelotas, Campus Universitário, Capão do Leão, Rio Grande do Sul, RS, 96160-000, Brazil
| | - Laís L R Mesquita
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - M C C S H Lara
- Biological Institute, Av. Conselheiro Rodrigues Alves, 1252, São Paulo, SP, 04014-002, Brazil
| | - E M C Villalobos
- Biological Institute, Av. Conselheiro Rodrigues Alves, 1252, São Paulo, SP, 04014-002, Brazil
| | - Cristina O Massoco
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Claudia M C Mori
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Enio Mori
- Pasteur Institute, Av. Paulista, 393, São Paulo, SP, 01311-000, Brazil
| | - Paulo C Maiorka
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| |
Collapse
|
12
|
Toscano ECDB, Sousa LFDC, Lima GK, Mesquita LA, Vilela MC, Rodrigues DH, Ferreira RN, Soriani FM, Campos MA, Kroon EG, Teixeira MM, de Miranda AS, Rachid MA, Teixeira AL. Neuroinflammation is associated with reduced SOCS2 and SOCS3 expression during intracranial HSV-1 infection. Neurosci Lett 2020; 736:135295. [PMID: 32800922 DOI: 10.1016/j.neulet.2020.135295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/27/2020] [Accepted: 08/04/2020] [Indexed: 12/22/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) is the main etiological agent of acute and sporadic encephalitis. Proteins of the suppressor of cytokine signaling (SOCS) family have shown to regulate the inflammation during HSV-1 infection in the brain. However, the effects of SOCS2 and SOCS3 in viral encephalitis remain unclear. The aim of the current study is to investigate the potential association between SOCS2, SOCS3, cytokines, and hippocampal damage, especially neuronal apoptosis, during acute intracranial HSV-1 infection in mice. Male C57BL/6 mice were infected by intracranial route with 102 plaque-forming units (PFU) inoculum of purified HSV-1. At three days post-infection (3 d.p.i.), mice were euthanized and their hippocampi were collected for histopathological analysis, immunohistochemical reaction against active caspase-3 and quantification of SOCS2, SOCS3 and cytokines (tumoral necrosis factor (TNF), interleukin (IL) 1β, IL-6, IL-10; interferon (IFN) -α, IFN-β, IFN-γ) mRNA expression. Infected mice exhibited neuronal loss and hemorrhagic focus in Cornu Ammonis (CA) region. The apoptotic index was higher in infected mice compared to controls. HSV-1 infection was associated with increased hippocampal expression of TNF, IL1-β, IL-6 and IFNα/IFNβ and decreased expression of IL-10, IFN-γ, SOCS2 and SOCS3. Our results suggest that down regulation of SOCS2 and SOCS3 contributes to a pro-inflammatory environment associated with hippocampal damage and neuronal apoptosis during acute HSV-1 infection in mice.
Collapse
Affiliation(s)
| | | | - Graciela Kunrath Lima
- Departamento De Morfologia, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil
| | - Leonardo Antunes Mesquita
- Departamento De Microbiologia, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil
| | | | | | - Rodrigo Novaes Ferreira
- Departamento De Morfologia, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil
| | | | | | - Erna Geessien Kroon
- Departamento De Microbiologia, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Departamento De Bioquímica e Imunologia, Instituto De Ciências Biológicas, Universidade Federal De Juiz De Fora, Brazil
| | - Aline Silva de Miranda
- Departamento De Morfologia, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil
| | - Milene Alvarenga Rachid
- Departamento De Patologia Geral, Instituto De Ciências Biológicas, Universidade Federal De Minas Gerais, Brazil.
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, School of Medicine, University of Texas Health Science Center at Houston, TX, United States
| |
Collapse
|
13
|
The diverse roles of RIP kinases in host-pathogen interactions. Semin Cell Dev Biol 2020; 109:125-143. [PMID: 32859501 PMCID: PMC7448748 DOI: 10.1016/j.semcdb.2020.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/09/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
Receptor Interacting Protein Kinases (RIPKs) are cellular signaling molecules that are critical for homeostatic signaling in both communicable and non-communicable disease processes. In particular, RIPK1, RIPK2, RIPK3 and RIPK7 have emerged as key mediators of intracellular signal transduction including inflammation, autophagy and programmed cell death, and are thus essential for the early control of many diverse pathogenic organisms. In this review, we discuss the role of each RIPK in host responses to bacterial and viral pathogens, with a focus on studies that have used pathogen infection models rather than artificial stimulation with purified pathogen associated molecular patterns. We also discuss the intricate mechanisms of host evasion by pathogens that specifically target RIPKs for inactivation, and finally, we will touch on the controversial issue of drug development for kinase inhibitors to treat chronic inflammatory and neurological disorders, and the implications this may have on the outcome of pathogen infections.
Collapse
|
14
|
Potter H, Boyd TD, Clarke P, Pelak VS, Tyler KL. Recruiting the innate immune system with GM-CSF to fight viral diseases, including West Nile Virus encephalitis and COVID-19. F1000Res 2020; 9:345. [PMID: 32704352 PMCID: PMC7359749 DOI: 10.12688/f1000research.23729.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 01/08/2023] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic grows throughout the world, it is imperative that all approaches to ameliorating its effects be investigated, including repurposing drugs that show promise in other diseases. We have been investigating an approach to multiple disorders that involves recruiting the innate immune system to aid the body's healing and regenerative mechanism(s). In the case of West Nile Virus encephalitis and potentially COVID-19, the proposed intervention to stimulate the innate immune system may give the adaptive immune response the necessary time to develop, finish clearing the virus, and provide future immunity. Furthermore, we have found that GM-CSF-induced recruitment of the innate immune system is also able to reverse brain pathology, neuroinflammation and cognitive deficits in mouse models of Alzheimer's disease and Down syndrome, as well as improving cognition in normal aging and in human patients with cognitive deficits due to chemotherapy, both of which exhibit neuroinflammation. Others have shown that GM-CSF is an effective treatment for both bacterial and viral pneumonias, and their associated inflammation, in animals and that it has successfully treated pneumonia-associated Acute Respiratory Distress Syndrome in humans. These and other data strongly suggest that GM-CSF may be an effective treatment for many viral infections, including COVID-19.
Collapse
Affiliation(s)
- Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Victoria S. Pelak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
15
|
Tuttolomondo A, Di Raimondo D, Vasto S, Casuccio A, Colomba C, Norrito RL, Di Bona D, Arnao V, Siciliano L, Cascio A, Pinto A. Protective and causative killer Ig-like receptor (KIR) and metalloproteinase genetic patterns associated with Herpes simplex virus 1 (HSV-1) encephalitis occurrence. J Neuroimmunol 2020; 344:577241. [PMID: 32334204 DOI: 10.1016/j.jneuroim.2020.577241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND The cerebral innate immune system has a critical role in control processes of viral replication in the brain after primary infactivo and immunologic disregulation and inflammation has been reported as a primary determinant of pathogenesis and prognosis of subsequent HSV-1 related encephalitis (HSE). Interaction linking LTR3-activated DCs is also represented by the killer Ig-like receptor (KIR) + pathways on NK cells. Only a few studies analyzed the role of of MMP-9 activity regulating genetic polymorphism on clinical outcome of viral infections. Susceptibility to symptomatic encephalitis depends on SNC viral invasion and BBB disruption. We hypothesize that certain KIR genes and MMP allele may help to characterize a risk profile of developing an acute encephalitis due to HSV 1. AIM OF THE STUDY Analyze the frequency of KIR genes and the C(-1562)T MMP-9 allels in subjects with HSV-1 encephalitis and to analyze their interaction with regard of the risk of occurrence of a symptomatic encephalitis. MATERIALS AND METHODS Between November 2014 and January 2019, all consecutive patients with symptomatic acute encephalitis were recruited from three wards (Internal Medicine, Neurology, and Infectious Diseases) of "P. Giaccone" University Hospital, Palermo. RESULTS Patients with acute viral encephalitis in comparison to controls showed a higher frequency AA KIR haplotype, HLA-C2 and of HLA-A-Bw4 alleles. With regard of HLA allele frequency patients with acute viral encephalitis In comparison to controls also showed a higher frequency of HLA-C2 and of HLA-A-Bw4 alleles. With regard of MMP-9 alleles, subjects with acute viral encephalitis were more likely to have the TT genotype. The multiple logistic regression analysis considering variables predictive of the occurrence of acute viral encephalitis showed the detrimental effect of AA KIR, HLAC1, HLA-A-BW4 and HLA-B-BW4T and of TT aplotype of MMP-9 genotype. CONCLUSIONS Our study shows that in immunocompetent adult subjects there is an association between some KIR genes, MMP-9 alleles and HLA-ligand alleles and susceptibility to develop a symptomatic acute viral encephalitis. Definition of the genetic and immunological background of acute viral encephalitis can play a key role to determine personalized medicine.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy.
| | - Domenico Di Raimondo
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | - Sonya Vasto
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Italy
| | - Alessandra Casuccio
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | - Claudia Colomba
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | - Rosario Luca Norrito
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | - Danilo Di Bona
- School and Chair of Allergology, Dipartimento delle Emergenze e Trapianti d'Organo, University of Bari, Bari, Italy
| | | | - Luisa Siciliano
- Psychology, Neuroscience and Human Sciences, Department of Brain and Behavioural Sciences of the University of Pavia (Italy), Department of Humanities and Life Sciences of the University School for Advanced Studies (IUSS), Pavia, Italy
| | - Antonio Cascio
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | - Antonio Pinto
- Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Italy
| | | |
Collapse
|
16
|
Imaizumi T, Satake U, Miyashita R, Kawaguchi S, Matsumiya T, Seya K, Ding J, Tanaka H. Interferon-induced transmembrane protein 1 and Myxovirus resistance protein 1 are induced by polyinosinic-polycytidylic acid in cultured hCMEC/D3 human cerebral microvascular endothelial cells. J Neuroimmunol 2019; 337:577047. [PMID: 31520792 DOI: 10.1016/j.jneuroim.2019.577047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
The molecular mechanisms of antiviral innate immune reactions in brain microvascular endothelial cells remain unclear. Interferon (IFN)-induced transmembrane protein 1 (IFITM1) and Myxovirus resistance protein 1 (MX1), the members of IFN-stimulated genes, are known as antiviral molecules. IFITM1 inhibits virus entry into host cell cytoplasm, whereas MX1 antagonizes virus replication. Here we observed that IFITM1 and MX1, and a proinflammatory cytokine IL-6 expression was induced by polyinosinic-polycytidylic acid (poly IC) in hCMEC/D3 human brain microvascular endothelial cells. Poly IC-induced IFITM1 and MX1 expression were decreased by NF-κB inhibitor SN50, IFN regulatory factor 3 inhibitor MRT67307 and human type I IFN neutralizing antibody mixture. These findings suggest that IFITM1 and MX1 may help protect the brain from viruses.
Collapse
Affiliation(s)
- Tadaatsu Imaizumi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | - Urara Satake
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Ruri Miyashita
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Shogo Kawaguchi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Tomoh Matsumiya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Kazuhiko Seya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Jiangli Ding
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Hiroshi Tanaka
- Department of School Health Science, Hirosaki University Faculty of Education, Hirosaki 036-8560, Japan
| |
Collapse
|
17
|
Nutma E, Willison H, Martino G, Amor S. Neuroimmunology - the past, present and future. Clin Exp Immunol 2019; 197:278-293. [PMID: 30768789 PMCID: PMC6693969 DOI: 10.1111/cei.13279] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Neuroimmunology as a separate discipline has its roots in the fields of neurology, neuroscience and immunology. Early studies of the brain by Golgi and Cajal, the detailed clinical and neuropathology studies of Charcot and Thompson's seminal paper on graft acceptance in the central nervous system, kindled a now rapidly expanding research area, with the aim of understanding pathological mechanisms of inflammatory components of neurological disorders. While neuroimmunologists originally focused on classical neuroinflammatory disorders, such as multiple sclerosis and infections, there is strong evidence to suggest that the immune response contributes to genetic white matter disorders, epilepsy, neurodegenerative diseases, neuropsychiatric disorders, peripheral nervous system and neuro-oncological conditions, as well as ageing. Technological advances have greatly aided our knowledge of how the immune system influences the nervous system during development and ageing, and how such responses contribute to disease as well as regeneration and repair. Here, we highlight historical aspects and milestones in the field of neuroimmunology and discuss the paradigm shifts that have helped provide novel insights into disease mechanisms. We propose future perspectives including molecular biological studies and experimental models that may have the potential to push many areas of neuroimmunology. Such an understanding of neuroimmunology will open up new avenues for therapeutic approaches to manipulate neuroinflammation.
Collapse
Affiliation(s)
- E. Nutma
- Department of PathologyAmsterdam UMC, VUmc siteAmsterdamthe Netherlands
| | - H. Willison
- University of Glasgow, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesGlasgowUK
| | - G. Martino
- Neuroimmunology Unit, Division of NeuroscienceInstitute of Experimental Neurology (INSpe), San Raffaele Scientific Institute and Vita‐Salute San Raffaele UniversityMilanItaly
| | - S. Amor
- Department of PathologyAmsterdam UMC, VUmc siteAmsterdamthe Netherlands
- Centre for Neuroscience and TraumaThe Blizard Institute, Barts and The London School of Medicine and DentistryLondonUK
| |
Collapse
|
18
|
Mancuso R, Sicurella M, Agostini S, Marconi P, Clerici M. Herpes simplex virus type 1 and Alzheimer's disease: link and potential impact on treatment. Expert Rev Anti Infect Ther 2019; 17:715-731. [PMID: 31414935 DOI: 10.1080/14787210.2019.1656064] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Alzheimer's disease (AD), the most common form of dementia worldwide, is a multifactorial disease with a still unknown etiology. Herpes simplex virus 1 (HSV-1) has long been suspected to be one of the factors involved in the pathogenesis of the disease. Areas covered: We review the literature focusing on viral characteristics of HSV-1, the mechanisms this virus uses to infect neural cells, its interaction with the host immune system and genetic background and summarizes results and research that support the hypothesis of an association between AD and HSV-1. The possible usefulness of virus-directed pharmaceutical approaches as potential treatments for AD will be discussed as well. Expert opinion: We highlight crucial aspects that must be addressed to clarify the possible role of HSV-1 in the pathogenesis of the disease, and to allow the design of new therapeutical approaches for AD.
Collapse
Affiliation(s)
| | | | | | - Peggy Marconi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara , Ferrara , Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi , Milan , Italy.,Department of Pathophysiology and Transplantation, University of Milan , Milan , Italy
| |
Collapse
|
19
|
Qin Q, Li Y. Herpesviral infections and antimicrobial protection for Alzheimer's disease: Implications for prevention and treatment. J Med Virol 2019; 91:1368-1377. [PMID: 30997676 DOI: 10.1002/jmv.25481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/04/2019] [Indexed: 02/05/2023]
Abstract
Accumulating evidence suggests that infections by herpesviruses might be closely linked to Alzheimer's disease (AD). Pathological hallmarks of AD brains include senile plaques induced by amyloid β peptide (Aβ) in the extracellular space and intracellular neurofibrillary tangles (NFTs) consisting of phosphorylated tau protein. The prevailing hypothesis for the mechanism of AD is amyloid cascade reaction. Recent studies revealed that infections by herpesviruses induce the similar pathological hallmarks of AD, including Aβ production, phosphorylation of tau (P-tau), oxidative stress, neuroinflammation, etc. Aβ peptide is regarded as one of the antimicrobial peptides, which inhibits HSV-1 replication. In the elderly, reactivation of herpesviruses might act as an initiator for amyloid cascade reaction in vulnerable individuals, triggering the neurofibrillary formation of phosphorylated tau and inducing oxidative stress and neuroinflammation, which can further contribute to the accumulation of Aβ and P-tau by impairing mitochondria and autophagosome. Epidemiological studies have shown AD susceptibility genes, such as APOE-ε4 allele, are highly linked to infections by herpesviruses. Interestingly, anti-herpesviral therapy significantly reduced the risk of AD in a large population study. Given that herpesviruses are arguably the most prevalent opportunistic pathogens and often reactivate in the elderly, it is reasonable to argue reactivation of herpesviruses might be major culprits for initiating AD in individuals carrying AD susceptibility genes. In this review, we summarize epidemiological and molecular evidence that support for a hypothesis of herpesviral infections and antimicrobial protection in the development of AD, and discuss the implications for future prevention and treatment of the disease.
Collapse
Affiliation(s)
- Qingsong Qin
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yun Li
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
20
|
Sassa N, Hirono K, Shiratori T, Kawaguchi S, Matsumiya T, Yoshida H, Seya K, Tanaka H, Imaizumi T. Induction of C‐C motif chemokine ligand 2 through Toll‐like receptor 3 signaling in human cerebral microvascular endothelial cell/D3 cells: possible regulation by nuclear factor‐κB. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/cen3.12514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Naoko Sassa
- Department of Vascular Biology Institute of Brain Science Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Koji Hirono
- Department of Pediatrics Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Toshihiro Shiratori
- Department of Respiratory Medicine Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Shogo Kawaguchi
- Department of Gastroenterology and Hematology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Tomoh Matsumiya
- Department of Vascular Biology Institute of Brain Science Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Hidemi Yoshida
- Department of Vascular Biology Institute of Brain Science Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Kazuhiko Seya
- Department of Vascular Biology Institute of Brain Science Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Hiroshi Tanaka
- Department of Pediatrics Hirosaki University Graduate School of Medicine Hirosaki Japan
- Department of School Health Science Hirosaki University Faculty of Education Hirosaki Japan
| | - Tadaatsu Imaizumi
- Department of Vascular Biology Institute of Brain Science Hirosaki University Graduate School of Medicine Hirosaki Japan
| |
Collapse
|
21
|
The STING agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) stimulates an antiviral state and protects mice against herpes simplex virus-induced neurological disease. Virology 2019; 529:23-28. [PMID: 30648635 DOI: 10.1016/j.virol.2019.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 12/15/2022]
Abstract
Herpes simplex virus (HSV)- 1 is the most common cause of sporadic viral encephalitis and accounts for 5-10% of cases worldwide. A key factor in host control of viral infection is the initiation of the interferon (IFN) response, mediated in part by the stimulator of interferon genes (STING) pathway. In these studies, we examined the ability of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a STING agonist, to protect against HSV-1 infection. DMXAA reduced viral replication through increased production of type I IFN in vitro. Furthermore, administration of DMXAA to HSV-1 infected mice resulted in a reduction of viral burden in the peripheral and central nervous systems. This reduced viral burden also correlated with increased survival of DMXAA-treated infected mice. These results therefore demonstrate the potential of STING agonists for immunotherapy against HSV-1.
Collapse
|
22
|
Yoshida H, Imaizumi T, Matsumiya T, Seya K, Kawaguchi S, Tanaka H. Gnetin C suppresses double-stranded RNA-induced C-C motif chemokine ligand 2 (CCL2) and CCL5 production by inhibiting Toll-like receptor 3 signaling pathway. Biomed Res 2018; 39:231-240. [PMID: 30333430 DOI: 10.2220/biomedres.39.231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The innate immune system is a prerequisite for biophylactic ability, but its dysregulation can cause inflammatory and autoimmune diseases. To determine a safe method of controlling inflammatory reactions in the brain, we examined the effects of gnetin C, a natural resveratrol dimer, on C-C motif chemokine ligand 2 (CCL2) and CCL5 (pro-inflammatory chemokines) production observed after treatment with polyinosinic-polycytidylic acid [poly IC; a synthetic analog of dsRNA as a Toll-like receptor 3 (TRL3) ligand, 30 μg/mL] in cultured human astrocytoma U373MG and neuroblastoma SH-SY5Y cells. The addition of gnetin C (10 μM) to the media moderately reduced the CCL2 production and markedly suppressed CCL5 production in both cells. In the TLR3-interferon (IFN)-β-phosphorylated-STAT1 (signal transducer and activator of transcription protein 1)RIG-I (retinoic acid-inducible gene-I) pathway that mediates CCL2 and CCL5 production, gnetin C first inhibits IFN-β expression in SH-SY5Y cells and primarily inhibits STAT1 phosphorylation in U373MG cells. In any case, gnetin C attenuated the dsRNA-activated TLR3 signaling resulting in CCL2 and CCL5 production, thus, may be useful for controlling TLR3-mediated inflammation in the brain.
Collapse
Affiliation(s)
- Hidemi Yoshida
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine
| | - Tomoh Matsumiya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine
| | - Kazuhiko Seya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine
| | - Shogo Kawaguchi
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine
| | - Hiroshi Tanaka
- Department of Pediatrics, Hirosaki University Graduate School of Medicine.,Department of School Health Science, Faculty of Education
| |
Collapse
|
23
|
Walters AW, Kujawa MR, Albe JR, Reed DS, Klimstra WB, Hartman AL. Vascular permeability in the brain is a late pathogenic event during Rift Valley fever virus encephalitis in rats. Virology 2018; 526:173-179. [PMID: 30396029 DOI: 10.1016/j.virol.2018.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/08/2018] [Accepted: 10/19/2018] [Indexed: 01/19/2023]
Abstract
Rift Valley fever virus (RVFV) is a zoonotic disease of livestock that causes several clinical outcomes in people including febrile disease, hemorrhagic fever, and/or encephalitis. After aerosol infection with RVFV, Lewis rats develop lethal encephalitic disease, and we use this as a model for studying disease mechanisms of RVFV infection in the brain. Permeability of the brain vasculature in relation to virus invasion and replication is not known. Here, we found that vascular permeability in the brain occurred late in the course of infection and corresponded temporally to expression of matrix metalloproteinase-9 (MMP-9). Virus replication was ongoing within the central nervous system for several days prior to detectable vascular leakage. Based on this study, vascular permeability was not required for entry of RVFV into the brain of rats. Prevention of vascular leakage late in infection may be an important component for prevention of lethal neurological disease in the rat model.
Collapse
Affiliation(s)
- Aaron W Walters
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Michael R Kujawa
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Joseph R Albe
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Douglas S Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William B Klimstra
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Brown B, Fidell A, Ingolia G, Murad E, Beckham JD. Defining diagnostic approaches and outcomes in patients with inflammatory CSF: A retrospective cohort study. Clin Neurol Neurosurg 2018; 172:105-111. [PMID: 29986194 DOI: 10.1016/j.clineuro.2018.06.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/07/2018] [Accepted: 06/30/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Define the etiologies, clinical features, time to diagnosis, and outcomes of patients that present with cerebral spinal fluid (CSF) pleocytosis. PATIENTS AND METHODS This is retrospective cohort study of patients with CSF pleocytosis, defined as WBC count >5 cells/mm3 in the CSF, from July 2015 to June 2016 at a large tertiary care hospital. The proportion of patients within specific diagnostic categories were analyzed for differences in diagnostic testing and outcomes. RESULTS 53% of patients had CSF pleocytosis due to an unknown etiology. The leading known cause of neuroinflammation was CNS infection (n = 43/244, 18%), followed by malignancy (n = 28/244, 11%). Mean time to diagnosis was 125 days in patients with autoimmune neuroinflammation and was 16 days in patients with an infection or malignancy. CSF parameters and peripheral white blood cell counts did not distinguish between categories of disease. The presence of CSF oligoclonal bands or a positive biopsy result most commonly supported a diagnosis of an autoimmune disease or malignancy, respectively. Neuroimaging changes were present in only 44% of infections but were found in 80-90% of other categories of neuroinflammation. Patients presenting with a severe neurologic deficit had 22.29 higher odds of a severe deficit at the last neurologic assessment, and mortality was highest (29%) in patients with malignancy-associated neuroinflammation. CONCLUSIONS This study to defines general diagnostic categories of neuroinflammatory disease in patients and provides new insight on the value of specific diagnostic testing, time to diagnosis, and outcomes in these patient populations.
Collapse
Affiliation(s)
- Bethany Brown
- Clinical Science Graduate Program, University of Colorado Graduate School, Aurora, CO, United States
| | - Andrea Fidell
- Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO, United States
| | - Gregory Ingolia
- Department of Medicine, University of Chicago School of Medicine, Chicago, IL, United States
| | - Eias Murad
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
| | - J David Beckham
- Clinical Science Graduate Program, University of Colorado Graduate School, Aurora, CO, United States; Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States; Veterans Administration, Eastern Colorado Health System, Denver, CO, United States; Department of Neurology, Division of NeuroImmunology and Neurological Infections, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
25
|
Chattopadhyay D, Mukhopadhyay A, Ojha D, Sadhukhan P, Dutta S. Immuno-metabolic changes in herpes virus infection. Cytokine 2018; 112:52-62. [PMID: 29960669 DOI: 10.1016/j.cyto.2018.06.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/18/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022]
Abstract
Recent evidences indicate that change in cellular metabolic pathways can alter immune response and function of the host; emphasizing the role of metabolome in health and diseases. Human Herpes simplex virus type-1 (HSV-1) and type-2 (HSV-2) causes diseases from asymptomatic to highly prevalent oral and genital herpes, recurrent blisters or neurological complications. Immune responses against HSV are complex with delicate interplay between innate signaling pathways and adaptive immune responses. The innate response involves the induction of protective IFN-1; while Natural Killer (NK) cells and plasmacytoid Dendritic Cells (pDC) confer in vivo adaptive anti-HSV response along with humoral and cellular components in controlling infection and latency. Metabolic changes lead to up-/down-regulation of several cytokines and chemokines like IFN-γ, IL-2, IL-4, IL-10 and MIP1β in HSV infection and recurrences. Recently, the viral protein ICP0 has been identified as an attenuator of TLR signaling, that inhibit innate responses to HSV. This review will summarize the role of metabolome in innate and adaptive effectors in infection, pathogenesis and immune control of HSV, highlighting the delicate interplay between the metabolic changes and immunity.
Collapse
Affiliation(s)
- Debprasad Chattopadhyay
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; ICMR-Virus Unit, Infectious Diseases and Beliaghata General Hospital, 57 Dr Suresh Chandra Banerjee Road, Beliaghata, Kolkata, West Bengal 700010, India.
| | - Aparna Mukhopadhyay
- Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata 700073, India
| | - Durbadal Ojha
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
| | - Provash Sadhukhan
- ICMR-Virus Unit, Infectious Diseases and Beliaghata General Hospital, 57 Dr Suresh Chandra Banerjee Road, Beliaghata, Kolkata, West Bengal 700010, India
| | - Shanta Dutta
- ICMR-National Institute of Cholera & Enteric Diseases, P- C.I.T. Scheme XM, 33 CIT Road, Beliaghata, Kolkata, West Bengal 700010, India
| |
Collapse
|
26
|
Harris SA, Harris EA. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer's Disease. Front Aging Neurosci 2018; 10:48. [PMID: 29559905 PMCID: PMC5845560 DOI: 10.3389/fnagi.2018.00048] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
This review focuses on research in the areas of epidemiology, neuropathology, molecular biology and genetics that implicates herpes simplex virus type 1 (HSV-1) as a causative agent in the pathogenesis of sporadic Alzheimer’s disease (AD). Molecular mechanisms whereby HSV-1 induces AD-related pathophysiology and pathology, including neuronal production and accumulation of amyloid beta (Aβ), hyperphosphorylation of tau proteins, dysregulation of calcium homeostasis, and impaired autophagy, are discussed. HSV-1 causes additional AD pathologies through mechanisms that promote neuroinflammation, oxidative stress, mitochondrial damage, synaptic dysfunction, and neuronal apoptosis. The AD susceptibility genes apolipoprotein E (APOE), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1) and clusterin (CLU) are involved in the HSV lifecycle. Polymorphisms in these genes may affect brain susceptibility to HSV-1 infection. APOE, for example, influences susceptibility to certain viral infections, HSV-1 viral load in the brain, and the innate immune response. The AD susceptibility gene cholesterol 25-hydroxylase (CH25H) is upregulated in the AD brain and is involved in the antiviral immune response. HSV-1 interacts with additional genes to affect cognition-related pathways and key enzymes involved in Aβ production, Aβ clearance, and hyperphosphorylation of tau proteins. Aβ itself functions as an antimicrobial peptide (AMP) against various pathogens including HSV-1. Evidence is presented supporting the hypothesis that Aβ is produced as an AMP in response to HSV-1 and other brain infections, leading to Aβ deposition and plaque formation in AD. Epidemiologic studies associating HSV-1 infection with AD and cognitive impairment are discussed. Studies are reviewed supporting subclinical chronic reactivation of latent HSV-1 in the brain as significant in the pathogenesis of AD. Finally, the rationale for and importance of clinical trials treating HSV-1-infected MCI and AD patients with antiviral medication is discussed.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, United States
| | - Elizabeth A Harris
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
27
|
Kakooza-Mwesige A, Mohammed AH, Kristensson K, Juliano SL, Lutwama JJ. Emerging Viral Infections in Sub-Saharan Africa and the Developing Nervous System: A Mini Review. Front Neurol 2018. [PMID: 29527187 PMCID: PMC5829034 DOI: 10.3389/fneur.2018.00082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The global public health concern is heightened over the increasing number of emerging viruses, i.e., newly discovered or previously known that have expanded into new geographical zones. These viruses challenge the health-care systems in sub-Saharan Africa (SSA) countries from which several of them have originated and been transmitted by insects worldwide. Some of these viruses are neuroinvasive, but have been relatively neglected by neuroscientists. They may provide experiments by nature to give a time window for exposure to a new virus within sizeable, previously non-infected human populations, which, for instance, enables studies on potential long-term or late-onset effects on the developing nervous system. Here, we briefly summarize studies on the developing brain by West Nile, Zika, and Chikungunya viruses, which are mosquito-borne and have spread worldwide out of SSA. They can all be neuroinvasive, but their effects vary from malformations caused by prenatal infections to cognitive disturbances following perinatal or later infections. We also highlight Ebola virus, which can leave surviving children with psychiatric disturbances and cause persistent infections in the non-human primate brain. Greater awareness within the neuroscience community is needed to emphasize the menace evoked by these emerging viruses to the developing brain. In particular, frontline neuroscience research should include neuropediatric follow-up studies in the field on long-term or late-onset cognitive and behavior disturbances or neuropsychiatric disorders. Studies on pathogenetic mechanisms for viral-induced perturbations of brain maturation should be extended to the vulnerable periods when neurocircuit formations are at peaks during infancy and early childhood.
Collapse
Affiliation(s)
- Angelina Kakooza-Mwesige
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences and Mulago Hospital, Kampala, Uganda
| | | | | | - Sharon L Juliano
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Julius J Lutwama
- Arbovirology Laboratory, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
28
|
Howe CL, LaFrance-Corey RG, Goddery EN, Johnson RK, Mirchia K. Neuronal CCL2 expression drives inflammatory monocyte infiltration into the brain during acute virus infection. J Neuroinflammation 2017; 14:238. [PMID: 29202854 PMCID: PMC5715496 DOI: 10.1186/s12974-017-1015-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022] Open
Abstract
Background Viral encephalitis is a dangerous compromise between the need to robustly clear pathogen from the brain and the need to protect neurons from bystander injury. Theiler’s murine encephalomyelitis virus (TMEV) infection of C57Bl/6 mice is a model of viral encephalitis in which the compromise results in hippocampal damage and permanent neurological sequelae. We previously identified brain-infiltrating inflammatory monocytes as the primary driver of this hippocampal pathology, but the mechanisms involved in recruiting these cells to the brain were unclear. Methods Chemokine expression levels in the hippocampus were assessed by microarray, ELISA, RT-PCR, and immunofluorescence. Monocyte infiltration during acute TMEV infection was measured by flow cytometry. CCL2 levels were manipulated by immunodepletion and by specific removal from neurons in mice generated by crossing a line expressing the Cre recombinase behind the synapsin promoter to animals with floxed CCL2. Results Inoculation of the brain with TMEV induced hippocampal production of the proinflammatory chemokine CCL2 that peaked at 6 h postinfection, whereas inoculation with UV-inactivated TMEV did not elicit this response. Immunofluorescence revealed that hippocampal neurons expressed high levels of CCL2 at this timepoint. Genetic deletion of CCR2 and systemic immunodepletion of CCL2 abrogated or blunted the infiltration of inflammatory monocytes into the brain during acute infection. Specific genetic deletion of CCL2 from neurons reduced serum and hippocampal CCL2 levels and inhibited inflammatory monocyte infiltration into the brain. Conclusions We conclude that intracranial inoculation with infectious TMEV rapidly induces the expression of CCL2 in neurons, and this cellular source is necessary for CCR2-dependent infiltration of inflammatory monocytes into the brain during the most acute stage of encephalitis. These findings highlight a unique role for neuronal production of chemokines in the initiation of leukocytic infiltration into the infected central nervous system.
Collapse
Affiliation(s)
- Charles L Howe
- Translational Neuroimmunology Lab, Mayo Clinic, Rochester, USA. .,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, USA. .,Department of Neurology, Mayo Clinic, Rochester, USA. .,Department of Neuroscience, Mayo Clinic, Rochester, USA. .,Department of Immunology, Mayo Clinic, Rochester, USA. .,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, USA. .,Mayo Clinic, Guggenheim 1542C, 200 First St SW, Rochester, MN, 55905, USA.
| | - Reghann G LaFrance-Corey
- Translational Neuroimmunology Lab, Mayo Clinic, Rochester, USA.,Department of Neurology, Mayo Clinic, Rochester, USA
| | - Emma N Goddery
- Translational Neuroimmunology Lab, Mayo Clinic, Rochester, USA.,Department of Immunology, Mayo Clinic, Rochester, USA.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, USA
| | - Renee K Johnson
- Translational Neuroimmunology Lab, Mayo Clinic, Rochester, USA.,Department of Neurology, Mayo Clinic, Rochester, USA
| | - Kanish Mirchia
- Translational Neuroimmunology Lab, Mayo Clinic, Rochester, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, USA.,Department of Neurology, Mayo Clinic, Rochester, USA
| |
Collapse
|
29
|
Abstract
Activation of the receptor interacting serine/threonine kinase (RIPK) 3 mediates an inflammatory type of cell death called necroptosis; in addition, RIPK3 has necroptosis-independent roles in inflammation, although these are not well defined. In a recent study published in Cell, Daniels and colleagues demonstrate that RIPK3 controls West Nile virus infection by promoting neuroinflammation in the central nervous system without affecting neuronal death.
Collapse
|