1
|
Lisi F, Zelikin AN, Chandrawati R. Nitric Oxide to Fight Viral Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003895. [PMID: 33850691 PMCID: PMC7995026 DOI: 10.1002/advs.202003895] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Indexed: 05/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) that has quickly and deeply affected the world, with over 60 million confirmed cases. There has been a great effort worldwide to contain the virus and to search for an effective treatment for patients who become critically ill with COVID-19. A promising therapeutic compound currently undergoing clinical trials for COVID-19 is nitric oxide (NO), which is a free radical that has been previously reported to inhibit the replication of several DNA and RNA viruses, including coronaviruses. Although NO has potent antiviral activity, it has a complex role in the immunological host responses to viral infections, i.e., it can be essential for pathogen control or detrimental for the host, depending on its concentration and the type of virus. In this Essay, the antiviral role of NO against SARS-CoV, SARS-CoV-2, and other human viruses is highlighted, current development of NO-based therapies used in the clinic is summarized, existing challenges are discussed and possible further developments of NO to fight viral infections are suggested.
Collapse
Affiliation(s)
- Fabio Lisi
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN)The University of New South Wales (UNSW Sydney)SydneyNSW2052Australia
| | - Alexander N. Zelikin
- Department of Chemistry and iNANO Interdisciplinary Nanoscience CenterAarhus UniversityAarhus8000Denmark
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN)The University of New South Wales (UNSW Sydney)SydneyNSW2052Australia
| |
Collapse
|
2
|
Abstract
Herpes simplex virus 1 (HSV-1) can be responsible for life-threatening HSV encephalitis (HSE). The mortality rate of patients with HSE who do not receive antiviral treatment is 70%, with most survivors suffering from permanent neurological sequelae. The use of intravenous acyclovir together with improved diagnostic technologies such as PCR and magnetic resonance imaging has resulted in a reduction in the mortality rate to close to 20%. However, 70% of surviving patients still do not recover complete neurological functions. Thus, there is an urgent need to develop more effective treatments for a better clinical outcome. It is well recognized that cerebral damage resulting from HSE is caused by viral replication together with an overzealous inflammatory response. Both of these processes constitute potential targets for the development of innovative therapies against HSE. In this review, we discuss recent progress in therapy that may be used to ameliorate the outcome of patients with HSE, with a particular emphasis on immunomodulatory agents. Ideally, the administration of adjunctive immunomodulatory drugs should be initiated during the rise of the inflammatory response, and its duration should be limited in time to reduce undesired effects. This critical time frame should be optimized by the identification of reliable biomarkers of inflammation.
Collapse
|
3
|
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: 5.4] [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
|
4
|
Mechanisms of Blood-Brain Barrier Disruption in Herpes Simplex Encephalitis. J Neuroimmune Pharmacol 2018; 14:157-172. [PMID: 30456443 DOI: 10.1007/s11481-018-9821-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022]
Abstract
Herpes simplex encephalitis (HSE) is often caused by infection with herpes simplex virus 1 (HSV-1), a neurotropic double-stranded DNA virus. HSE infection always impacts the temporal and frontal lobes or limbic system, leading to edema, hemorrhage, and necrotic changes in the brain parenchyma. Additionally, patients often exhibit severe complications following antiviral treatment, including dementia and epilepsy. HSE is further associated with disruptions to the blood-brain barrier (BBB), which consists of microvascular endothelial cells, tight junctions, astrocytes, pericytes, and basement membranes. Following an HSV-1 infection, changes in BBB integrity and permeability can result in increased movement of viruses, immune cells, and/or cytokines into the brain parenchyma. This leads to an enhanced inflammatory response in the central nervous system and further damage to the brain. Thus, it is important to protect the BBB from pathogens to reduce brain damage from HSE. Here, we discuss HSE and the normal structure and function of the BBB. We also discuss growing evidence indicating an association between BBB breakdown and the pathogenesis of HSE, as well as future research directions and potential new therapeutic targets. Graphical Abstract During herpes simplex encephalitis, the functions and structures of each composition of BBB have been altered by different factors, thus the permeability and integrity of BBB have been broken. The review aim to explore the potential mechanisms and factors in the process, probe the next research targets and new therapeutic targets.
Collapse
|
5
|
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: 16.5] [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
|
6
|
Gu N, Tian Y, Di Z, Han C, Lei H, Zhang G. Shuanghuanglian injection downregulates nuclear factor-kappa B expression in mice with viral encephalitis. Neural Regen Res 2012; 7:2592-9. [PMID: 25368635 PMCID: PMC4200726 DOI: 10.3969/j.issn.1673-5374.2012.33.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 10/10/2012] [Indexed: 11/18/2022] Open
Abstract
A mouse model of viral encephalitis was induced by intracranial injection of a Coxsackie virus B3 suspension. Quantitative real-time reverse transcription-PCR and western blot assay were applied to detect mRNA and protein expression of intelectin-2 and nuclear factor-kappa B in the viral encephalitis and control groups. Nuclear factor-kappa B and intelectin-2 mRNA and protein expression were significantly increased in mice with viral encephalitis. After intraperitoneal injection of Shuanghuanglian at a dose of 1.5 mg/kg for 5 successive days, intelectin-2 and nuclear factor-kappa B protein and mRNA expression were significantly decreased. To elucidate the relationship between intelectin-2 and nuclear factor-kappa B, mice with viral encephalitis were administered an intracerebral injection of 107 pfu recombinant lentivirus expressing intelectin shRNA. Both protein and mRNA levels of intelectin and nuclear factor-kappa B in brain tissue of mice were significantly decreased. Experimental findings suggest that Shuanghuanglian injection may downregulate nuclear factor-kappa B production via suppression of intelectin production, thus inhibiting inflammation associated with viral encephalitis.
Collapse
Affiliation(s)
- Naibing Gu
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| | - Ye Tian
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| | - Zhengli Di
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| | - Caiping Han
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| | - Hui Lei
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| | - Gejuan Zhang
- Department of Neurology, Xi’an Center Hospital, Xi’an 710003, Shaanxi Province, China
| |
Collapse
|
7
|
Brown WD, Bearer EL, Donahue JE. Chronic active herpes simplex type 2 encephalitis in an asymptomatic immunocompetent child. J Child Neurol 2010; 25:901-8. [PMID: 20179002 PMCID: PMC3376078 DOI: 10.1177/0883073809353449] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A unique form of chronic, active, granulomatous herpes simplex type 2 encephalitis is described in an asymptomatic, immunocompetent 8-year-old girl who acquired the virus as a neonate. The extensive, bilateral cerebral parenchymal involvement was discovered incidentally. Diagnosis was confirmed by a combination of serial neuroimaging, brain biopsy, and quantitative polymerase chain reaction targeted to DNA sequences in the glycoprotein G gene, allowing differentiation between herpes simplex virus types 1 and 2. The clinical course over a 5-year period, treatment with intermittent intravenous steroids, and daily valacyclovir, diagnostic imaging, and laboratory studies are reviewed in detail. This form of herpes simplex virus type 2 encephalitis hasn't been described previously and is significant because of its prolonged indolent course, absence of neurological findings or suggestive history, and benign behavior in this child, who is now 14 years old. The authors believe this entity can be unsuspected and underdiagnosed in the general pediatric population, especially in those with a prior maternal history of herpes simplex virus type 2 infection.
Collapse
Affiliation(s)
- William D. Brown
- Warren Alpert Medical School at Brown University, Providence, RI, Department of Pediatrics and Department of Neurology, Hasbro Children’s Hospital/Rhode Island Hospital, Providence, RI
| | - Elaine L. Bearer
- Warren Alpert Medical School at Brown University, Providence, RI, Department of Pathology, University of New Mexico Medical School, Albuquerque, NM
| | - John E. Donahue
- Warren Alpert Medical School at Brown University, Providence, RI, Department of Pathology (Neuropathology) and Clinical Neurosciences (Neurology), Rhode Island Hospital, Providence, RI
| |
Collapse
|
8
|
Kuhlmann I, Minihane AM, Huebbe P, Nebel A, Rimbach G. Apolipoprotein E genotype and hepatitis C, HIV and herpes simplex disease risk: a literature review. Lipids Health Dis 2010; 9:8. [PMID: 20109174 PMCID: PMC2830997 DOI: 10.1186/1476-511x-9-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 01/28/2010] [Indexed: 01/22/2023] Open
Abstract
Apolipoprotein E is a polymorphic and multifunctional protein with numerous roles in lipoprotein metabolism. The three common isoforms apoE2, apoE3 and apoE4 show isoform-specific functional properties including different susceptibilities to diseases. ApoE4 is an accepted risk factor for Alzheimer's disease and cardiovascular disorders. Recently, associations between apoE4 and infectious diseases have been demonstrated. This review summarises how apoE4 may be involved in the infection incidence and associated pathologies of specific infectious diseases, namely hepatitis C, human immunodeficiency virus disease and herpes simplex.ApoE4 seems to be protective against chronic hepatitis C virus infection and retards fibrosis progression. In contrast apoE4 enhances the fusion rate of human immunodeficiency virus with target cell membranes, resulting in accelerated cell entry and faster disease progression. Its association with human immunodeficiency virus-associated dementia remains controversial. Regarding herpes simplex virus infection, apoE4 intensifies virus latency and is associated with increased oxidative damage of the central nervous system, and there is some evidence that herpes simplex virus infection in combination with the apoE4 genotype may be associated with an increased risk of Alzheimer's disease. In addition to reviewing available data from human trials, evidence derived from a variety of cell culture and animal models are considered in this review in order to provide mechanistic insights into observed association between apoE4 genotype and viral disease infection and pathology.
Collapse
Affiliation(s)
- Inga Kuhlmann
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Hermann-Rodewald-Strasse 6, 24098 Kiel, Germany
| | | | | | | | | |
Collapse
|
9
|
Kavouras JH, Prandovszky E, Valyi-Nagy K, Kovacs SK, Tiwari V, Kovacs M, Shukla D, Valyi-Nagy T. Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation by-products in mouse P19N neural cell cultures. J Neurovirol 2008; 13:416-25. [PMID: 17994426 DOI: 10.1080/13550280701460573] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
To determine whether herpes simplex virus type 1 (HSV-1) infection causes oxidative stress and lipid peroxidation in cultured neural cells, mouse P19 embryonal carcinoma cells were differentiated into cells with neural phenotypes (P19N cells) by retinoic acid and were then infected with HSV-1. Cellular levels of reactive oxygen species (ROS) and the release of lipid peroxidation by-products into the tissue culture medium were then measured by the generation of fluorescent markers hydroxyphenyl fluorescein and a stable chromophore produced by lipid peroxidation products, malondialdehyde (MDA) and hydroxyalkenals (4-HAEs; predominantly 4-hydroxy-2-nonenal [HNE]), respectively. HSV-1 infection increased ROS levels in neural cells as early as 1 h post infection (p.i.) and ROS levels remained elevated at 24 h p.i. This viral effect required viral entry and replication as heat- and ultraviolet light-inactivated HSV-1 were ineffective. HSV-1 infection also was associated with increased levels of MDA/HAE in the culture medium at 2 and 4 h p.i., but MDA/HAE levels were not different from those detected in mock infected control cultures at 1, 6, and 24 h p.i. HSV-1 replication in P19N cells was inhibited by the antioxidant compound ebselen and high concentrations of HNE added to the cultures, but was increased by low concentrations of HNE. These findings indicate that HSV-1 infection of neural cells causes oxidative stress that is required for efficient viral replication. Furthermore, these observations raise the possibility that soluble, bioactive lipid peroxidation by-products generated in infected neural cells may be important regulators of HSV-1 pathogenesis in the nervous system.
Collapse
Affiliation(s)
- Jerry H Kavouras
- Department of Pathology, University of Illinois at Chicago, College of Medicine, 840 South Wood Street, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Sellner J, Simon F, Meyding-Lamade U, Leib SL. Herpes-simplex virus encephalitis is characterized by an early MMP-9 increase and collagen type IV degradation. Brain Res 2006; 1125:155-62. [PMID: 17109833 DOI: 10.1016/j.brainres.2006.09.093] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 09/26/2006] [Accepted: 09/27/2006] [Indexed: 11/22/2022]
Abstract
Cerebrovascular complications including cerebral edema, raised intracranial pressure and hemorrhage contribute to the high mortality and morbidity of herpes-simplex virus encephalitis (HSE). We examined changes of collagen type IV, the major constituent of the neurovascular matrix, together with expression and localization of matrix-degrading enzymes during the development of acute HSE. In an experimental model of focal HSE, we found that early, symptomatic HSE (3 days after infection) and acute, fully developed HSE (7 days after infection) are associated with significantly raised levels of matrix-metalloproteinase-9 (MMP-9) (both P<0.05). In situ zymography of brain sections revealed that the increase of MMP-9 was restricted to the cerebral vasculature in early HSE and further expanded towards the perivascular space and adjacent tissue in acute HSE. Around the cerebral vasculature, we observed that MMP-9 activity was insufficiently counterbalanced by its endogenous tissue inhibitor of MMP (TIMP) TIMP-1, resulting in loss of collagen type IV. Our findings suggest that MMP-9 is involved in the evolution of HSE by causing damage to the cerebral vasculature. The degradation of the neurovascular matrix in HSE facilitates the development of cerebrovascular complications and may represent a target for novel adjuvant treatment strategies.
Collapse
Affiliation(s)
- Johann Sellner
- Department of Neurology, University Hospital Bern, Inselspital, Freiburgstrasse, CH-3010 Bern, Switzerland
| | | | | | | |
Collapse
|
11
|
Marques CP, Hu S, Sheng W, Lokensgard JR. Microglial cells initiate vigorous yet non-protective immune responses during HSV-1 brain infection. Virus Res 2006; 121:1-10. [PMID: 16621100 DOI: 10.1016/j.virusres.2006.03.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 03/14/2006] [Accepted: 03/14/2006] [Indexed: 11/25/2022]
Abstract
Central nervous system (CNS) infection with herpes simplex virus (HSV)-1 triggers neuroinflammatory responses leading to peripheral immune cell infiltration into the brain. Previous in vitro studies from our laboratory, using primary human brain cells, implicated microglia as the cellular source of infection-induced chemokines, such as CXC ligand 10 (CXCL10) and CC ligand 2 (CCL2). Here, we evaluated the role of microglial cells in HSV-induced neuroimmune responses using an in vivo murine model of herpes encephalitis. Data obtained during this study demonstrated robust levels of CXCL10, CCL2 and CXCL9 detectable in the brains of infected BALB/c mice between 5 and 8 days post-infection (p.i.). Microglial cells were identified as a source of this HSV-induced chemokine production. Additional experiments established that induction of these immune mediators preceded the presence of CD3, CD4, CD8, and CD45 mRNA in the brain, and immunohistochemical analysis confirmed the presence of infiltrating CD3(+) cells. Further analysis suggested that microglia-derived chemokines drive peripheral immune cell chemotaxis, as antibodies to CXCL10 and CCL2 blocked the migration of murine splenocytes toward HSV-infected microglia by approximately 59.3+/-4.1% and 17.5+/-1.4%, respectively. Taken together, these results demonstrate that a vigorous microglia-driven cascade of pro-inflammatory immune responses is not sufficient to protect susceptible mice from HSV-1 brain infection.
Collapse
Affiliation(s)
- Cristina P Marques
- Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, USA
| | | | | | | |
Collapse
|
12
|
Sellner J, Lenhard T, Haas J, Einsiedel RV, Meyding-Lamadé U. Differential mRNA expression of neurotrophic factors GDNF, BDNF, and NT-3 in experimental herpes simplex virus encephalitis. ACTA ACUST UNITED AC 2005; 137:267-71. [PMID: 15950786 DOI: 10.1016/j.molbrainres.2005.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2004] [Revised: 02/26/2005] [Accepted: 03/13/2005] [Indexed: 01/26/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) mRNA levels were studied in the course of murine herpes simplex virus encephalitis. Induction of GNDF and NT-3 (both P < 0.05) was found during acute encephalitis. Despite absence of clinical impairment, both neurotrophic factors were overexpressed 2 months (NT-3) and 6 months (GDNF) following infection (both P < 0.05). Neurotrophic factors play an important role in neuronal survival and recovery after acute injury to the central nervous system (CNS) and may represent an additional therapeutic target for treatment of viral encephalitis.
Collapse
Affiliation(s)
- Johann Sellner
- Department of Neurology, Ruprecht-Karls-University Heidelberg, D-69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
13
|
Sellner J, Dvorak F, Zhou Y, Haas J, Kehm R, Wildemann B, Meyding-Lamadè U. Acute and long-term alteration of chemokine mRNA expression after anti-viral and anti-inflammatory treatment in herpes simplex virus encephalitis. Neurosci Lett 2005; 374:197-202. [PMID: 15663962 DOI: 10.1016/j.neulet.2004.10.054] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Revised: 10/12/2004] [Accepted: 10/21/2004] [Indexed: 11/24/2022]
Abstract
Mortality and morbidity rates remain high among patients with herpes simplex virus encephalitis (HSVE). Chemokine-mediated recruitment and activation of leukocytes to focal areas of viral CNS infection are crucial steps in antiviral response and clearance. However, the inflammatory reaction and cellular antiviral response may enhance collateral damage to neurons and account for chronic progressive brain damage. We identified a specific mRNA expression of the interferon-gamma-inducible chemokines (CXCL9, CXCL10 and CXCL11), and RANTES (CCL5) in the acute course and long-term of experimental HSVE. This pattern was substantially altered by anti-viral and anti-inflammatory treatment. Our findings indicate a pivotal role of these chemokines in the immunopathogenesis of HSVE.
Collapse
Affiliation(s)
- Johann Sellner
- Department of Neurology, Im Neuenheimer Feld 400, Ruprecht-Karls-University Heidelberg, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
Rock RB, Gekker G, Hu S, Sheng WS, Cheeran M, Lokensgard JR, Peterson PK. Role of microglia in central nervous system infections. Clin Microbiol Rev 2004; 17:942-64, table of contents. [PMID: 15489356 PMCID: PMC523558 DOI: 10.1128/cmr.17.4.942-964.2004] [Citation(s) in RCA: 500] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The nature of microglia fascinated many prominent researchers in the 19th and early 20th centuries, and in a classic treatise in 1932, Pio del Rio-Hortega formulated a number of concepts regarding the function of these resident macrophages of the brain parenchyma that remain relevant to this day. However, a renaissance of interest in microglia occurred toward the end of the 20th century, fueled by the recognition of their role in neuropathogenesis of infectious agents, such as human immunodeficiency virus type 1, and by what appears to be their participation in other neurodegenerative and neuroinflammatory disorders. During the same period, insights into the physiological and pathological properties of microglia were gained from in vivo and in vitro studies of neurotropic viruses, bacteria, fungi, parasites, and prions, which are reviewed in this article. New concepts that have emerged from these studies include the importance of cytokines and chemokines produced by activated microglia in neurodegenerative and neuroprotective processes and the elegant but astonishingly complex interactions between microglia, astrocytes, lymphocytes, and neurons that underlie these processes. It is proposed that an enhanced understanding of microglia will yield improved therapies of central nervous system infections, since such therapies are, by and large, sorely needed.
Collapse
Affiliation(s)
- R Bryan Rock
- Neuroimmunology Laboratory, Minneapolis Medical Research Foundation, and University of Minnesota Medical School, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Dvorak F, Martinez-Torres F, Sellner J, Haas J, Schellinger PD, Schwaninger M, Meyding-Lamadé UK. Experimental herpes simplex virus encephalitis: a long-term study of interleukin-6 expression in mouse brain tissue. Neurosci Lett 2004; 367:289-92. [PMID: 15337251 DOI: 10.1016/j.neulet.2004.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Revised: 06/02/2004] [Accepted: 06/03/2004] [Indexed: 11/20/2022]
Abstract
This study aimed to investigate the expression of interleukin-6 (IL-6) in acute and chronic herpes simplex virus encephalitis. In the brain of 15 SJL mice infected with herpes simplex virus type 1, strain F, and 14 control animals we performed a sequential quantitative analysis of expression of IL-6 mRNA with reverse transcription real-time polymerase chain reaction. The viral burden peaked in the acute disease, and then returned to a low baseline value. At day 7 following infection, IL-6 expression was significantly (2.05-fold) increased as compared with the baseline expression in uninfected animals. Twenty-one days after infection the mRNA expression still was significantly (1.78-fold) upregulated. No significant differences of IL-6 mRNA expression between infected and control mice were found after 2 and 6 months. We observed a 2.5-fold increase of IL-6 mRNA expression in control mice with increasing age of animals. We have additionally studied the clinical evolution of HSVE in IL-6 deficient mice. In experimental herpes simplex virus encephalitis IL-6, as a potent mediator of neuronal injury, is upregulated in the acute but not in the chronic disease. IL-6 deficient mice presented early and severe clinical signs of HSVE as compared to the wild-type C57/bl6 mice.
Collapse
Affiliation(s)
- Florian Dvorak
- Department of Neurology, University of Heidelberg, INF 400, 69120, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
Mori I, Goshima F, Koshizuka T, Imai Y, Kohsaka S, Koide N, Sugiyama T, Yoshida T, Yokochi T, Kimura Y, Nishiyama Y. Iba1-expressing microglia respond to herpes simplex virus infection in the mouse trigeminal ganglion. ACTA ACUST UNITED AC 2004; 120:52-6. [PMID: 14667577 DOI: 10.1016/j.molbrainres.2003.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microglial response in the trigeminal ganglion of mice corneally inoculated with herpes simplex virus (HSV) was investigated. Virus-infected neurons of the trigeminal ganglion did not exhibit apoptotic signal, while those of the trigeminal sensory brainstem nucleus did. Cells expressing ionized calcium binding adapter molecule 1 (Iba1), a specific marker of microglia/macrophages, increased in number in the virally infected region of the trigeminal ganglion, with morphological transformation to an activated phenotype, frequently detected as perineural satellites. Further microglial transformation to macropahges was not evident. Iba1-immunopositive perineural satellites also appeared in the vicinity of virally infected region. Such activated microglia expressed basic fibroblast growth factor (bFGF) molecules. The reverse transcription-polymerase chain reaction (RT-PCR) detected upregulated synthesis of mRNA for bFGF in the trigeminal ganglion. In contrast, in the trigeminal sensory brainstem nucleus, a small number of bFGF-producing cells appeared only in the vicinity of virally infected area. Collectively, Iba1-bearing microglia exist in the mouse trigeminal ganglion and respond to herpes simplex virus infection, most likely conferring neuroprotective functions upon the trigeminal ganglion.
Collapse
Affiliation(s)
- Isamu Mori
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mori I, Goshima F, Koshizuka T, Koide N, Sugiyama T, Yoshida T, Yokochi T, Nishiyama Y, Kimura Y. Differential activation of the c-Jun N-terminal kinase/stress-activated protein kinase and p38 mitogen-activated protein kinase signal transduction pathways in the mouse brain upon infection with neurovirulent influenza A virus. J Gen Virol 2003; 84:2401-2408. [PMID: 12917461 DOI: 10.1099/vir.0.19188-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The temporal and spatial distribution of active c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) in the brain was investigated in an experimental virus-mouse system in which neurovirulent influenza A virus caused lethal acute encephalitis. Following stereotaxic microinjection into the olfactory bulb, virus-infected neurons appeared in several midbrain structures, including the ventral tegmental area, amygdala and the pyramidal layer of the hippocampus. Infected neurons exhibited apoptosis on day 5, as demonstrated by in situ detection of DNA fragmentation and active caspase-3. The stress-responsive JNK signal transduction pathway was activated in virus-infected neurons. Activation of p38 MAPK was widespread and occurred in astrocytes on day 7 after infection. Active p38 MAPK in astrocytes showed no association with apoptosis but appeared to be involved in regulation of TNF-alpha production. These results indicate that these two stress-activated protein kinases may play distinct roles during the course of lethal acute influenza virus encephalitis.
Collapse
Affiliation(s)
- Isamu Mori
- Department of Microbiology, Fukui Medical University School of Medicine, 23-3 Matsuoka-cho, Yoshida-gun, Fukui 910-1193, Japan
- Laboratory of Virology, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Aichi, Japan
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Aichi, Japan
| | - Fumi Goshima
- Laboratory of Virology, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Aichi, Japan
| | - Tetsuro Koshizuka
- Laboratory of Virology, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Aichi, Japan
| | - Naoki Koide
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Aichi, Japan
| | - Tsuyoshi Sugiyama
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Aichi, Japan
| | - Tomoaki Yoshida
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Aichi, Japan
| | - Takashi Yokochi
- Department of Microbiology and Immunology, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Aichi, Japan
| | - Yukihiro Nishiyama
- Laboratory of Virology, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Aichi, Japan
| | - Yoshinobu Kimura
- Department of Microbiology, Fukui Medical University School of Medicine, 23-3 Matsuoka-cho, Yoshida-gun, Fukui 910-1193, Japan
| |
Collapse
|
18
|
Meyding-Lamadé UK, Oberlinner C, Rau PR, Seyfer S, Heiland S, Sellner J, Wildemann BT, Lamadé WR. Experimental herpes simplex virus encephalitis: a combination therapy of acyclovir and glucocorticoids reduces long-term magnetic resonance imaging abnormalities. J Neurovirol 2003; 9:118-25. [PMID: 12587075 DOI: 10.1080/13550280390173373] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2002] [Revised: 07/31/2002] [Accepted: 08/06/2002] [Indexed: 10/20/2022]
Abstract
Despite early antiviral treatment, herpes simples virus encephalitis (HSVE) still remains a life-threatening sporadic disease with high mortality and morbidity. In patients and in experimental disease, chronic progressive magnetic resonance imaging (MRI) abnormalities have been found even after antiviral therapy. Secondary autoimmune-mediated and not directly virus-mediated mechanisms might play a key role for the outcome of disease. This study aimed to evaluate a possible beneficial effect of a therapy of acyclovir and corticosteroids versus acyclovir only. In a mouse model of HSVE (intranasal inoculation with 10(5) pfu [plaque-forming units] of HSV-1 strain F), a long-term MRI study was realized. Cranial MRI was performed serially at days 2, 7, 14, 21, 60, and 180 in different therapy groups: 1, saline; 2, acyclovir; 3, acyclovir, subsequently methylprednisolone; 4, sham-infected with saline. Brain viral load peaked at day 7 to decline thereafter to a low baseline value. Viral load in group 1 was significantly higher than in animals with antiviral therapy. In group 4, no viral DNA was detectable. Viral load did not differ significantly between acyclovir and acyclovir/corticosteroid-treated groups, suggesting that the use of corticosteroids in addition to acyclovir does not increase viral burden. MRI findings in untreated and acyclovir-treated animals revealed chronic progressive changes. In contrast, there was a significant reduction of the severity of long-term MRI abnormalities in acyclovir/corticosteroid-treated animals. With respect to abnormal MRI findings, this study demonstrates a clear beneficial effect of an acyclovir and corticosteroid therapy without influencing brain viral load.
Collapse
|
19
|
Lokensgard JR, Cheeran MCJ, Hu S, Gekker G, Peterson PK. Glial cell responses to herpesvirus infections: role in defense and immunopathogenesis. J Infect Dis 2002; 186 Suppl 2:S171-9. [PMID: 12424694 DOI: 10.1086/344272] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Glial cells can respond to herpesvirus infections through the production of cytokines and chemokines. Although specific interactions between resident glia and lymphocytes that infiltrate the infected brain remain to be defined, the presence of T cell chemotactic signals in microglial cell supernatants following infection with cytomegalovirus or herpes simplex virus has led to the concept that chemokines initiate a cascade of neuroimmune responses that result in defense of the brain against herpesviruses. While chemokines may play a defensive role by attracting T cells into the brain, aberrant accumulation of lymphocytes may also induce brain damage. Host defense mechanisms must balance control of herpesvirus spread with associated undesirable immunopathologic effects. A growing body of evidence suggests that through complex networks of chemokines and cytokines produced in response to herpesvirus infection, glial cells orchestrate a cascade of events that result in successful defense of or damage to the brain.
Collapse
Affiliation(s)
- James R Lokensgard
- Neuroimmunology Laboratory, Minneapolis Medical Research Foundation, 914 S. 8th Street, Bldg. D-3, Minneapolis, MN 55404, USA.
| | | | | | | | | |
Collapse
|
20
|
Lim YJ, Chang SE, Choi JH, Sung KJ, Bahk JH, Do SH, Lee DS. Expression of inducible nitric oxide synthase in skin lesions of acute herpes zoster. J Dermatol Sci 2002; 29:201-5. [PMID: 12234710 DOI: 10.1016/s0923-1811(02)00035-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Histopathologically, the skin lesions of acute herpes zoster (AHZ) are characterized by epidermal necrotic vesicles with inflammation. Nitric oxide (NO) is generated from L-arginine by nitric oxide synthase (NOS), and immune inflammation involves the activation of NOS in both effector cells and target cells. NO can cause apoptosis and necrosis of target cells such as keratinocytes. We proposed that a large burst of NO in AHZ may cause the epidermal necrosis. Skin biopsies were taken from 13 patients with AHZ. The expression of inducible-type NOS (iNOS) was examined by immunoperoxidase staining and reverse transcription-polymerase chain reaction (RT-PCR). In the skin specimen of AHZ, moderate-to-strong staining for iNOS was observed in inflammatory cells and necrotic keratinocytes, while weak staining was observed in non-necrotic peripheral keratinocytes. RT-PCR using skin specimen of AHZ corroborated the immunoperoxidase findings, yielding bright bands for iNOS. Normal control skin showed minimal or negative expression both by immunoperoxidase stains and RT-PCR. Increased expression of iNOS is consistent with the hypothesis that high level of NO induced by iNOS may be associated with the epidermal necrosis with inflammation seen in the skin lesions of AHZ.
Collapse
Affiliation(s)
- Young-Jin Lim
- Department of Anesthesiology and Clinical Research Institute, Seoul National University Hospital, 28 Yongon-dong, Chongno-gu, 110-744 Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
21
|
Milatovic D, Zhang Y, Olson SJ, Montine KS, Roberts LJ, Morrow JD, Montine TJ, Dermody TS, Valyi-Nagy T. Herpes simplex virus type 1 encephalitis is associated with elevated levels of F2-isoprostanes and F4-neuroprostanes. J Neurovirol 2002; 8:295-305. [PMID: 12161814 DOI: 10.1080/13550280290100743] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To better understand the pathogenesis of herpes simplex virus type 1 (HSV-1) infections of the nervous system, concentrations of F(4)-neuroprostanes (F(4)-NP) and F(2)-isoprostanes (F(2)-IP) in the murine brain were determined following intracerebral inoculation of HSV-1 or normal saline. F(4)-NP are highly selective, quantitative markers of neuronal oxidative damage, while F(2)-IP are markers of oxidative damage to brain tissue not limited to a certain cell type. In contrast to saline-treated control animals, HSV-1-infected animals developed encephalitic symptoms associated with severe inflammation, widespread HSV-1 protein expression, and significantly elevated F(4)-NP and F(2)-IP levels in the brain. Survivors of acute HSV-1 infection showed no encephalitic symptoms 2 to 3 weeks following virus inoculation. Brain tissue derived from mice euthanized 2 month after virus inoculation demonstrated expression of HSV-1 latency-associated transcripts without detectable HSV-1 protein expression. However, brain tissue from these animals showed focal chronic inflammation, moderately elevated F(2)-IP levels, and normal levels of F(4)-NP. These observations provide novel biochemical evidence that oxidant tissue injury is a mechanism underlying neuronal damage during acute HSV-1 encephalitis and suggest that oxidative damage to tissue may continue in the mammalian brain until at least several weeks after recovery from the symptomatic phase of HSV-1 infection.
Collapse
Affiliation(s)
- Dejan Milatovic
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Meyding-Lamadé U, Seyfer S, Haas J, Dvorak F, Kehm R, Lamadé W, Hacke W, Wildemann B. Experimental herpes simplex virus encephalitis: inhibition of the expression of inducible nitric oxide synthase in mouse brain tissue. Neurosci Lett 2002; 318:21-4. [PMID: 11786216 DOI: 10.1016/s0304-3940(01)02469-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In the brain tissue of 36 mice infected with herpes simplex virus type 1, strain F, we determined the expression of inducible nitric oxide synthase (iNOS) with semiquantitative reverse transcription polymerase chain reaction. The viral burden was quantitated by polymerase chain reaction. Nitric oxide, induced by iNOS, may contribute to neuronal cell damage following virus infection. As the experimental therapeutic strategy in herpes simplex virus encephalitis (HSVE), we used: N-nitro-L-arginin (L-NA), a selective inhibitor of iNOS; and combination therapies of either methylprednisolone/acyclovir or L-NA/acyclovir. The viral burden peaked in acute disease, and then returned to a low baseline value, except in untreated controls. The expression of iNOS mRNA was suppressed by L-NA and by acyclovir/corticosteroids. INOS inhibition may provide an additional therapeutic strategy targeted specifically to suppress iNOS expression as a potential secondary mechanism of tissue damage in acute and chronic HSVE.
Collapse
|
23
|
Chen BP, Lane TE. Lack of nitric oxide synthase type 2 (NOS2) results in reduced neuronal apoptosis and mortality following mouse hepatitis virus infection of the central nervous system. J Neurovirol 2002; 8:58-63. [PMID: 11847593 PMCID: PMC7094997 DOI: 10.1080/135502802317247820] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The role of nitric oxide synthase type-2 (NOS2)-derived nitric oxide (NO) in the pathogenesis of mouse hepatitis virus (MHV)-induced central nervous system disease was examined. Infection of NOS2 knockout ((-/-)) and NOS2(+/+) mice with MHV resulted in similar kinetics of viral clearance from the brain and comparable levels of demyelination. MHV-infected NOS2(-/-) mice displayed a marked decrease in mortality as compared to infected NOS2(+/+) mice that correlated with a significant decrease (P < or = 0.001) in the number of apoptotic cells (determined by TUNEL staining) present in the brain. Confocal microscopy revealed that the majority of cells (>70%) undergoing apoptosis were neurons. These studies indicate that NOS2-generated NO contributes to apoptosis of neurons but not demyelination following MHV infection.
Collapse
Affiliation(s)
- Benjamin P. Chen
- Dept. of Molecular Biology and Biochemistry, University of California-Irvine, 3205 Biological Sciences II, 92697 Irvine, CA USA
| | - Thomas E. Lane
- Dept. of Molecular Biology and Biochemistry, University of California-Irvine, 3205 Biological Sciences II, 92697 Irvine, CA USA
- Reeve-Irvine Research Center, University of California-Irvine, Irvine, California USA
| |
Collapse
|
24
|
Affiliation(s)
- J R Anderson
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK.
| |
Collapse
|
25
|
Valyi-Nagy T, Olson SJ, Valyi-Nagy K, Montine TJ, Dermody TS. Herpes simplex virus type 1 latency in the murine nervous system is associated with oxidative damage to neurons. Virology 2000; 278:309-21. [PMID: 11118355 DOI: 10.1006/viro.2000.0678] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pathological consequences of herpes simplex virus type 1 (HSV-1) latency in the nervous system are not well understood. To determine whether acute and latent HSV-1 infections of the nervous system are associated with oxidative damage, mice were inoculated with HSV-1 by the corneal route, and the extent of viral infection and oxidative damage in trigeminal ganglia and brain was assessed at 7, 90, and 220 days after inoculation. Abundant HSV-1 protein expression in the nervous system was observed in neurons and non-neuronal cells at 7 days after inoculation, consistent with viral replication and spread through the trigeminal and olfactory systems. Acute HSV-1 infection was associated with focal, neuronal and non-neuronal 4-hydroxy-2-nonenal- and 8-hydroxyguanosine-specific immunoreactivity, indicating oxidative damage. Rare HSV-1 antigen-positive cells were observed at 90 and 220 days after inoculation; however, widespread HSV-1 latency-associated transcript expression was detected, consistent with latent HSV-1 infection in the nervous system. HSV-1 latency was detected predominantly in the trigeminal ganglia, brainstem, olfactory bulbs, and temporal cortex. Latent HSV-1 infection was associated with focal chronic inflammation and consistently detectable evidence of oxidative damage involving primarily neurons. These results indicate that both acute and latent HSV-1 infections in the murine nervous system are associated with oxidative damage.
Collapse
Affiliation(s)
- T Valyi-Nagy
- Departments of Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
26
|
Hu WH, Qiang WA, Li F, Liu N, Wang GQ, Wang HY, Wan XS, Liao WH, Liu JS, Jen MF. Constitutive and inducible nitric oxide synthases after dynorphin-induced spinal cord injury. J Chem Neuroanat 2000; 17:183-97. [PMID: 10697245 DOI: 10.1016/s0891-0618(99)00039-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
It has recently been demonstrated that selective inhibition of both neuronal constitutive and inducible nitric oxide synthases (ncNOS and iNOS) is neuroprotective in a model of dynorphin (Dyn) A(1-17)-induced spinal cord injury. In the present study, various methods including the conversion of 3H-L-arginine to 3H-citrulline, immunohistochemistry and in situ hybridization are employed to determine the temporal profiles of the enzymatic activities, immunoreactivities, and mRNA expression for both ncNOS and iNOS after intrathecal injection of a neurotoxic dose (20 nmol) of Dyn A(1-17). The expression of ncNOS immunoreactivity and mRNA increased as early as 30 min after injection and persisted for 1-4 h. At 24-48 h, the number of ncNOS positive cells remained elevated while most neurons died. The cNOS enzymatic activity in the ventral spinal cord also significantly increased at 30 min 48 h, but no significant changes in the dorsal spinal cord were observed. However, iNOS mRNA expression increased later at 2 h, iNOS immunoreactivity and enzymatic activity increased later at 4 h and persisted for 24-48 h after injection of 20 nmol Dyn A(1-17). These results indicate that both ncNOS and iNOS are associated with Dyn-induced spinal cord injury, with ncNOS predominantly involved at an early stage and iNOS at a later stage.
Collapse
Affiliation(s)
- W H Hu
- Department of Spinal Cord Injury, Research Institute of Surgery and Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Noack H, Possel H, Rethfeldt C, Keilhoff G, Wolf G. Peroxynitrite mediated damage and lowered superoxide tolerance in primary cortical glial cultures after induction of the inducible isoform of NOS. Glia 1999; 28:13-24. [PMID: 10498818 DOI: 10.1002/(sici)1098-1136(199910)28:1<13::aid-glia2>3.0.co;2-l] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The effect of the induction of i-NOS in primary glial cultures was studied with respect to the protein levels of reactive oxygen species (ROS) scavenging enzymes and the cytotoxicity of nitric oxide (.NO) formation at different levels of artificially generated superoxide. Stimulation of the cultures by bacterial lipopolysaccharides and gamma-interferon resulted in an induction of i-NOS exclusively in microglial cells. Among the ROS scavenging enzymes superoxide dismutase (Cu/Zn- and Mn-isoform), glutathione peroxidase and catalase only mitochondrial Mn-SOD was found to be upregulated in the course of i-NOS induction (Western blots). Although .NO formation did not affect cell viability at physiological levels of superoxide over a time period of 4 days, it caused an oxidative load particularly in microglial cells as observed by monitoring the oxidation of dichloro-dihydrofluorescein, an indicator for the formation of peroxynitrite and ROS. Elevated levels of superoxide, generated either intracellularly by paraquat or extracellularly via xanthine oxidase and hypoxanthine, resulted dose-dependently in a larger decline of cell viability in the .NO forming cultures compared to controls (release of lactate dehydrogenase, citrate synthase, stainability by propidium iodide, and tetramethylrhodamine). NOS-inhibitors reduced the degree of cell damage to that seen for control cultures, indicating an ONOO--/.NO mediated mechanism of cell damage. Our data support the concept that i-NOS catalyzed .NO-formation leads to an ONOO--mediated increased oxidative load. At physiological levels of superoxide and within a wide range of higher superoxide levels this nitrosative stress is well balanced in cultured glial cells by protective mechanisms.
Collapse
Affiliation(s)
- H Noack
- Institut für Medizinische Neurobiologie, Otto-von-Guericke-Universität, Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
28
|
Meyding-Lamadé U, Lamadé W, Kehm R, Oberlinner C, Fäth A, Wildemann B, Haas J, Hacke W. Herpes simplex virus encephalitis: chronic progressive cerebral MRI changes despite good clinical recovery and low viral load - an experimental mouse study. Eur J Neurol 1999; 6:531-8. [PMID: 10457385 DOI: 10.1046/j.1468-1331.1999.650531.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cranial magnetic resonance imaging (MRI) is a sensitive diagnostic tool for the in vivo detection of morphological abnormalities in herpes simplex virus encephalitis (HSVE). We performed a long-term MRI study in a mouse model of HSVE. Cranial MRI findings were compared with the viral load within brain tissue, the presence of HSV DNA in the cerebrospinal fluid (CSF), a daily clinical assessment and post-mortem neurohistopathological studies. A 1.5 T cranial MRI scanner with standard spin-echo sequences was used. Viral load within the brain and the presence of HSV DNA in cerebrospinal fluid were determined by a polymerase chain reaction assay. Clinically, animals were severely affected within the first 2 weeks and recovered thereafter. Focal histopathological and MRI abnormalities involved predominantly limbic structures, a pattern that mimics human disease. Severity and extent of abnormalities had increased at 6 months despite clinical improvement. HSV DNA was present in CSF during the acute disease only. Brain viral load peaked at day 10 and declined thereafter. MRI as an in vivo monitoring approach may reveal chronic progressive changes in HSVE, despite clinical recovery and low viral load in the brain. Secondary, not directly virus-mediated, mechanisms of tissue damage may contribute to tissue damage of HSVE.
Collapse
Affiliation(s)
- U Meyding-Lamadé
- Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
New Thoughts on Pathogenesis and Diagnosis of Encephalitis. Curr Infect Dis Rep 1999; 1:178-186. [PMID: 11095786 DOI: 10.1007/s11908-996-0027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Knowledge regarding the pathogenesis of viral encephalitis, defined as inflammation and destruction of the central nervous system (CNS) from viral infection and the resulting immune response, has improved with advances in molecular biology techniques and recent advances in immunology and neuroscience research. An increasingly complex understanding has developed with regard to viral CNS infection. In addition to advances in viral genetics exploring increased viral spread and neurovirulence, improved understanding from research on neurochemistry, neurodevelopment, and cytokine expression in the CNS has led to new hypotheses regarding the mechanism of CNS damage during viral CNS infection. This review explores three advances in the understanding of viral encephalitis in the past few years: 1) the relationship between viral load and extent of viral CNS disease, 2) chemokines and their role in the CNS inflammatory response as well as in the pathogenesis of encephalitis, and 3) secondary damage from the release of neurotoxins during encephalitis. By examining this research, the reviewers intend to introduce novel therapeutic modalities that are developing for the management of patients with viral encephalitis beyond the timely use of antiviral therapy.
Collapse
|
30
|
Haas J, Meyding-Lamadé U, Fäth A, Stingele K, Storch-Hagenlocher B, Wildemann B. Acyclovir treatment of experimentally induced herpes simplex virus encephalitis: monitoring the changes in immunologic NO synthase expression and viral load within brain tissue of SJL mice. Neurosci Lett 1999; 264:129-32. [PMID: 10320031 DOI: 10.1016/s0304-3940(99)00191-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The effect of acyclovir treatment on viral burden and the expression of immunologic nitric oxide synthase (iNOS) within brains of 42 HSV-1 F infected mice was studied by using a titration PCR assay for HSV-1 DNA and a semiquantitative RT-PCR for iNOS mRNA. iNOS mediated NO-production may possibly be involved in secondary mechanisms of brain injury following virus infection, which may account for treatment failures in human herpes simplex virus encephalitis (HSVE). Following infection, a parallel increase of iNOS mRNA and HSV-1F-DNA occurred with peaks after 7 days that were both significantly lower under acyclovir treatment. Six months post infection viral load had declined, but iNOS mRNA expression in both treated and untreated mice was still enhanced as compared with mock infected controls. This suggests that acyclovir decreases iNOS expression via inhibition of viral replication shortly after infection but fails to influence elevated iNOS within the brain late in the course of experimental HSVE.
Collapse
Affiliation(s)
- J Haas
- Department of Neurology, University of Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
Meyding-Lamadé U, Lamadé W, Kehm R, Knopf KW, Hess T, Gosztonyi G, Degen O, Hacke W. Herpes simplex virus encephalitis: cranial magnetic resonance imaging and neuropathology in a mouse model. Neurosci Lett 1998; 248:13-6. [PMID: 9665652 DOI: 10.1016/s0304-3940(98)00319-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We performed a long-term magnetic resonance imaging (MRI) study in a mouse model of herpes simplex virus encephalitis. Mice were infected with herpes simplex virus type 1 (HSV-1) strain F. A 1.5-T cranial MRI scanner with standard spin-echo sequences was used. Neuropathological studies included immunohistochemistry. The presence of HSV DNA in brain tissue was determined with a polymerase chain reaction assay. Clinical assessment was performed daily: within the first 2 weeks the animals were severely affected and recovered thereafter. MRI and histopathological abnormalities corresponded well. HSV DNA was detectable initially and at 6 months. Extent and severity of structural abnormalities increased at 6 months. MRI offers a new in vivo approach for the detection of structural changes in the disease course of experimental herpes simplex virus encephalitis.
Collapse
Affiliation(s)
- U Meyding-Lamadé
- Department of Neurology, INF 400 Kopfklinik, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|