1
|
Nobach D, Müller J, Tappe D, Herden C. Update on immunopathology of bornavirus infections in humans and animals. Adv Virus Res 2020; 107:159-222. [PMID: 32711729 DOI: 10.1016/bs.aivir.2020.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Knowledge on bornaviruses has expanded tremendously during the last decade through detection of novel bornaviruses and endogenous bornavirus-like elements in many eukaryote genomes, as well as by confirmation of insectivores as reservoir species for classical Borna disease virus 1 (BoDV-1). The most intriguing finding was the demonstration of the zoonotic potential of lethal human bornavirus infections caused by a novel bornavirus of different squirrel species (variegated squirrel 1 bornavirus, VSBV-1) and by BoDV-1 known as the causative agent for the classical Borna disease in horses and sheep. Whereas a T cell-mediated immunopathology has already been confirmed as key disease mechanism for infection with BoDV-1 by experimental studies in rodents, the underlying pathomechanisms remain less clear for human bornavirus infections, infection with other bornaviruses or infection of reservoir species. Thus, an overview of current knowledge on the pathogenesis of bornavirus infections focusing on immunopathology is given.
Collapse
Affiliation(s)
- Daniel Nobach
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jana Müller
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany; Center for Brain, Mind and Behavior, Justus-Liebig-University Giessen, Giessen, Germany.
| |
Collapse
|
2
|
Zhang D, Thongda W, Li C, Zhao H, Beck BH, Mohammed H, Arias CR, Peatman E. More than just antibodies: Protective mechanisms of a mucosal vaccine against fish pathogen Flavobacterium columnare. FISH & SHELLFISH IMMUNOLOGY 2017; 71:160-170. [PMID: 28989091 DOI: 10.1016/j.fsi.2017.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/05/2017] [Accepted: 10/02/2017] [Indexed: 05/20/2023]
Abstract
A recently developed attenuated vaccine for Flavobacterium columnare has been demonstrated to provide superior protection for channel catfish, Ictalurus punctatus, against genetically diverse columnaris isolates. We were interested in examining the mechanisms of this protection by comparing transcriptional responses to F. columnare challenge in vaccinated and unvaccinated juvenile catfish. Accordingly, 58 day old fingerling catfish (28 days post-vaccination or unvaccinated control) were challenged with a highly virulent F. columnare isolate (BGSF-27) and gill tissues collected pre-challenge (0 h), and 1 h and 2 h post infection, time points previously demonstrated to be critical in early host-pathogen interactions. Following RNA-sequencing and transcriptome assembly, differential expression (DE) analysis within and between treatments revealed several patterns and pathways potentially underlying improved survival of vaccinated fish. Most striking was a pattern of dramatically higher basal expression of an array of neuropeptides (e.g. somatostatin), hormones, complement factors, and proteases at 0 h in vaccinated fish. Previous studies indicate these are likely the preformed mediators of neuroendocrine cells and/or eosinophilic granular (mast-like) cells within the fish gill. Following challenge, these elements fell to almost undetectable levels (>100-fold downregulated) by 1 h in vaccinated fish, suggesting their rapid release and/or cessation of synthesis following degranulation. Concomitantly, levels of pro-inflammatory cytokines (IL-1b, IL-8, IL-17) were induced in unvaccinated fish. In contrast, in vaccinated catfish, we observed widespread induction of genes needed for collagen deposition and tissue remodeling. Taken together, our results indicate an important component of vaccine protection in fish mucosal tissues may be the sensitization, proliferation and arming of resident secretory cells in the period between primary and secondary challenge.
Collapse
Affiliation(s)
- Dongdong Zhang
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wilawan Thongda
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Chao Li
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao 266109, China
| | - Honggang Zhao
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Benjamin H Beck
- United States Department of Agriculture, Agricultural Research Service, Aquatic Animal Health Research Unit, Auburn, AL 36832, USA
| | - Haitham Mohammed
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA; Department of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Covadonga R Arias
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Eric Peatman
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
3
|
Sarkar R, Verma SC. Egr-1 regulates RTA transcription through a cooperative involvement of transcriptional regulators. Oncotarget 2017; 8:91425-91444. [PMID: 29207655 PMCID: PMC5710935 DOI: 10.18632/oncotarget.20648] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 07/26/2017] [Indexed: 11/25/2022] Open
Abstract
Kaposi's sarcoma associated herpesvirus (KSHV) regulates the host cellular environment to establish life-long persistent infection by manipulating cellular signaling pathways, with approximately 1- 5% of cells undergoing lytic reactivation during the course of infection. Egr-1 (Early Growth Response Factor-1) is one such cellular transcription factor, which gets phosphorylated during the lytic phase of viral life cycle to perpetrate its function. This study demonstrates the mechanism of how Egr-1 mediates transcription of the immediate early gene, RTA (Replication and transcription activator), which is the lytic switch gene of KSHV. Egr-1 depleted KSHV infected cells exhibited reduced expression of RTA. Also, an increase in Egr-1 phosphorylation led to a higher virion production, which was suppressed in the presence of p38 and Raf inhibitors. Reporter assays showed that coexpression of Egr-1 and CBP (CREB-binding protein) enhances RTA promoter activity as compared to the expression of either Egr-1 or CBP alone. Binding of Egr-1 and CBP at RTA promoter was analyzed by chromatin immunoprecipitation assay (ChIP), which showed an enhanced accumulation during viral reactivation. Mutation in Egr-1 binding site of the RTA promoter eliminated Egr-1 response on promoter activation. Furthermore, de novo infection of THP-1 (monocytic) and HUVECs (endothelial) cells showed an upregulation of Egr-1 phosphorylation, whereas depletion of Egr-1 reduced the mRNA levels of RTA during primary infection. Together, these results demonstrate a cooperative role of Egr-1 and CBP in mediating RTA transcription, which significantly improves our understanding of the involvement of cellular factors controlling RTA transcription in KSHV pathogenesis.
Collapse
Affiliation(s)
- Roni Sarkar
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Subhash C Verma
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
4
|
Abstract
AbstractNatural bornavirus infections and their resulting diseases are largely restricted to horses and sheep in Central Europe. The disease also occurs naturally in cats, and can be induced experimentally in laboratory rodents and numerous other mammals. Borna disease virus-1 (BoDV-1), the cause of most cases of mammalian Borna disease, is a negative-stranded RNA virus that replicates within the nucleus of target cells. It causes severe, often lethal, encephalitis in susceptible species. Recent events, especially the discovery of numerous new species of bornaviruses in birds and a report of an acute, lethal bornaviral encephalitis in humans, apparently acquired from squirrels, have revived interest in this remarkable family of viruses. The clinical manifestations of the bornaviral diseases are highly variable. Thus, in addition to acute lethal encephalitis, they can cause persistent neurologic disease associated with diverse behavioral changes. They also cause a severe retinitis resulting in blindness. In this review, we discuss both the pathological lesions observed in mammalian bornaviral disease and the complex pathogenesis of the neurologic disease. Thus infected neurons may be destroyed by T-cell-mediated cytotoxicity. They may die as a result of excessive inflammatory cytokine release from microglia. They may also die as a result of a ‘glutaminergic storm’ due to a failure of infected astrocytes to regulate brain glutamate levels.
Collapse
|
5
|
Hilary Koprowski, MD: A Lifetime of Work. Monoclon Antib Immunodiagn Immunother 2014; 33:1-43. [DOI: 10.1089/mab.2014.kop.biblio] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
6
|
Chen Y, Garcia GE, Huang W, Constantini S. The involvement of secondary neuronal damage in the development of neuropsychiatric disorders following brain insults. Front Neurol 2014; 5:22. [PMID: 24653712 PMCID: PMC3949352 DOI: 10.3389/fneur.2014.00022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/20/2014] [Indexed: 12/12/2022] Open
Abstract
Neuropsychiatric disorders are one of the leading causes of disability worldwide and affect the health of billions of people. Previous publications have demonstrated that neuropsychiatric disorders can cause histomorphological damage in particular regions of the brain. By using a clinical symptom-comparing approach, 55 neuropsychiatric signs or symptoms related usually to 14 types of acute and chronic brain insults were identified and categorized in the present study. Forty percent of the 55 neuropsychiatric signs and symptoms have been found to be commonly shared by the 14 brain insults. A meta-analysis supports existence of the same neuropsychiatric signs or symptoms in all brain insults. The results suggest that neuronal damage might be occurring in the same or similar regions or structures of the brain. Neuronal cell death, neural loss, and axonal degeneration in some parts of the brain (the limbic system, basal ganglia system, brainstem, cerebellum, and cerebral cortex) might be the histomorphological basis that is responsible for the neuropsychiatric symptom clusters. These morphological alterations may be the result of secondary neuronal damage (a cascade of progressive neural injury and neuronal cell death that is triggered by the initial insult). Secondary neuronal damage causes neuronal cell death and neural injury in not only the initial injured site but also remote brain regions. It may be a major contributor to subsequent neuropsychiatric disorders following brain insults.
Collapse
Affiliation(s)
- Yun Chen
- BrightstarTech Inc. , Clarksburg, MD , USA
| | - Gregory E Garcia
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground , Aberdeen, MD , USA
| | - Wei Huang
- Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Shlomi Constantini
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
7
|
Kosmac K, Bantug GR, Pugel EP, Cekinovic D, Jonjic S, Britt WJ. Glucocorticoid treatment of MCMV infected newborn mice attenuates CNS inflammation and limits deficits in cerebellar development. PLoS Pathog 2013; 9:e1003200. [PMID: 23505367 PMCID: PMC3591306 DOI: 10.1371/journal.ppat.1003200] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 01/08/2013] [Indexed: 01/07/2023] Open
Abstract
Infection of the developing fetus with human cytomegalovirus (HCMV) is a major cause of central nervous system disease in infants and children; however, mechanism(s) of disease associated with this intrauterine infection remain poorly understood. Utilizing a mouse model of HCMV infection of the developing CNS, we have shown that peripheral inoculation of newborn mice with murine CMV (MCMV) results in CNS infection and developmental abnormalities that recapitulate key features of the human infection. In this model, animals exhibit decreased granule neuron precursor cell (GNPC) proliferation and altered morphogenesis of the cerebellar cortex. Deficits in cerebellar cortical development are symmetric and global even though infection of the CNS results in a non-necrotizing encephalitis characterized by widely scattered foci of virus-infected cells with mononuclear cell infiltrates. These findings suggested that inflammation induced by MCMV infection could underlie deficits in CNS development. We investigated the contribution of host inflammatory responses to abnormal cerebellar development by modulating inflammatory responses in infected mice with glucocorticoids. Treatment of infected animals with glucocorticoids decreased activation of CNS mononuclear cells and expression of inflammatory cytokines (TNF-α, IFN-β and IFNγ) in the CNS while minimally impacting CNS virus replication. Glucocorticoid treatment also limited morphogenic abnormalities and normalized the expression of developmentally regulated genes within the cerebellum. Importantly, GNPC proliferation deficits were normalized in MCMV infected mice following glucocorticoid treatment. Our findings argue that host inflammatory responses to MCMV infection contribute to deficits in CNS development in MCMV infected mice and suggest that similar mechanisms of disease could be responsible for the abnormal CNS development in human infants infected in-utero with HCMV.
Collapse
Affiliation(s)
- Kate Kosmac
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America.
| | | | | | | | | | | |
Collapse
|
8
|
Hoppes S, Gray PL, Payne S, Shivaprasad HL, Tizard I. The isolation, pathogenesis, diagnosis, transmission, and control of avian bornavirus and proventricular dilatation disease. Vet Clin North Am Exot Anim Pract 2010; 13:495-508. [PMID: 20682432 PMCID: PMC7110554 DOI: 10.1016/j.cvex.2010.05.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Proventricular dilatation disease (PDD) is a common infectious neurologic disease of birds comprising a dilatation of the proventriculus by ingested food as a result of defects in intestinal motility, which affects more than 50 species of psittacines, and is also known as Macaw wasting disease, neuropathic ganglioneuritis, or lymphoplasmacytic ganglioneuritis. Definitive diagnosis of PDD has been problematic due to the inconsistent distribution of lesions. Since its discovery, avian bornavirus (ABV) has been successfully cultured from the brains of psittacines diagnosed with PDD, providing a source of antigen for serologic assays and nucleic acid for molecular assays. This article provides evidence that ABV is the etiologic agent of PDD. Recent findings on the transmission, epidemiology, pathogenesis, diagnosis, and control of ABV infection and PDD are also reviewed.
Collapse
Affiliation(s)
- Sharman Hoppes
- Small Animal Clinical Sciences, Texas A&M University, 4474 TAMU, College Station, TX 77843, USA.
| | | | | | | | | |
Collapse
|
9
|
The immune response to rabies virus infection and vaccination. Vaccine 2010; 28:3896-901. [DOI: 10.1016/j.vaccine.2010.03.039] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/10/2010] [Accepted: 03/21/2010] [Indexed: 12/25/2022]
|
10
|
Ouyang N, Storts R, Tian Y, Wigle W, Villanueva I, Mirhosseini N, Payne S, Gray P, Tizard I. Histopathology and the detection of avian bornavirus in the nervous system of birds diagnosed with proventricular dilatation disease. Avian Pathol 2009; 38:393-401. [DOI: 10.1080/03079450903191036] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Nishino Y, Ooishi R, Kurokawa S, Fujino K, Murakami M, Madarame H, Hashimoto O, Sugiyama K, Funaba M. Gene expression of the TGF-β family in rat brain infected with Borna disease virus. Microbes Infect 2009; 11:737-43. [DOI: 10.1016/j.micinf.2009.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/02/2009] [Accepted: 04/07/2009] [Indexed: 11/17/2022]
|
12
|
Roy A, Hooper DC. Immune evasion by rabies viruses through the maintenance of blood-brain barrier integrity. J Neurovirol 2008; 14:401-11. [PMID: 19016377 DOI: 10.1080/13550280802235924] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The attenuated rabies virus (RV) strain Challenge Virus Standard (CVS)-F3 and a highly pathogenic strain associated with the silver-haired bats (SHBRV) can both be cleared from the central nervous system (CNS) tissues by appropriate antiviral immune mechanisms if the effectors are provided access across the blood-brain barrier (BBB). In the case of SHBRV infection, antiviral immunity develops normally in the periphery but fails to open the BBB, generally resulting in a lethal outcome. To determine whether or not an absence in the CNS targeted immune response is associated with the infection with other pathogenic RV strains, we have assessed the development of immunity, BBB permeability, and immune cell infiltration into the CNS tissues of mice infected with a variety of RV strains, including the dog variants responsible for the majority of human rabies cases. We demonstrate that the lethal outcomes of infection with a variety of known pathogenic RV strains are indeed associated with the inability to deliver immune effectors across the BBB. Survival from infection with certain of these viruses is improved in mice prone to CNS inflammation. The results suggest that competition between the activity of the immune effectors reaching CNS tissues and the inherent pathological attributes of the virus dictates the outcome and that intervention to deliver RV-specific immune effectors into CNS tissues may have general therapeutic value in rabies.
Collapse
Affiliation(s)
- Anirban Roy
- Center for Neurovirology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107-6799, USA
| | | |
Collapse
|
13
|
Chen SH, Yao HW, Chen IT, Shieh B, Li C, Chen SH. Suppression of transcription factor early growth response 1 reduces herpes simplex virus lethality in mice. J Clin Invest 2008; 118:3470-7. [PMID: 18769632 DOI: 10.1172/jci35114] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 07/16/2008] [Indexed: 02/06/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) infection is the most common cause of sporadic, fatal encephalitis, but current understanding of how the virus interacts with cellular factors to regulate disease progression is limited. Here, we show that HSV-1 infection induced the expression of the cellular transcription factor early growth response 1 (Egr-1) in a human neuronal cell line. Egr-1 increased viral replication by activating promoters of viral productive cycle genes through binding to its corresponding sequences in the viral promoters. Mouse studies confirmed that Egr-1 expression was enhanced in HSV-1-infected brains and that Egr-1 functions to promote viral replication in embryonic fibroblasts. Furthermore, Egr-1 deficiency or knockdown of Egr-1 by a DNA-based enzyme greatly reduced the mortality of HSV-1-infected mice by decreasing viral loads in tissues. This study provides what we believe is the first evidence that Egr-1 increases the mortality of HSV-1 encephalitis by enhancing viral replication. Moreover, blocking this cellular machinery exploited by the virus could prevent host mortality.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Republic of China
| | | | | | | | | | | |
Collapse
|
14
|
Pedras-Vasconcelos JA, Puig M, Sauder C, Wolbert C, Ovanesov M, Goucher D, Verthelyi D. Immunotherapy with CpG oligonucleotides and antibodies to TNF-alpha rescues neonatal mice from lethal arenavirus-induced meningoencephalitis. THE JOURNAL OF IMMUNOLOGY 2008; 180:8231-40. [PMID: 18523289 DOI: 10.4049/jimmunol.180.12.8231] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Viral encephalitides are life-threatening diseases in neonates partly due to the irreversible damage inflammation causes to the CNS. This study explored the role of proinflammatory cytokines in the balance between controlling viral replication and eliciting pathologic immune responses in nonlytic viral encephalitis. We show that neonatal mice challenged with arenavirus Tacaribe (TCRV) develop a meningoencephalitis characterized by high IFN-gamma and TNF-alpha levels and mild T cell infiltration. Neutralization of the TNF-alpha using mAb was associated with lower chemokine expression, reduced T cell infiltration, and lower levels of IFN-gamma, and TNF-alpha in the CNS and led to 100% survival. Moreover, treatment with Abs to TNF-alpha improved mobility and increased survival even after the mice developed bilateral hind limb paralysis. Of note, animals treated with anti-TNF-alpha Abs alone did not clear the virus despite generating Abs to TCRV. Direct activation of the innate immune response using CpG oligodeoxynucleotides in combination with anti-TNF-alpha Abs resulted in 100% survival and complete viral clearance. To our knowledge, this is the first demonstration of the use of innate immune modulators plus Abs to TNF-alpha as therapeutics for a lethal neurotropic viral infection.
Collapse
Affiliation(s)
- João A Pedras-Vasconcelos
- Laboratory of Immunology, Division of Therapeutic Proteins, Office of Biotechnology, Center for Drug Evaluation and Review, FDA, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Montgomery DL, Van Olphen A, Van Campen H, Hansen TR. The Fetal Brain in Bovine Viral Diarrhea Virus-infected Calves: Lesions, Distribution, and Cellular Heterogeneity of Viral Antigen at 190 Days Gestation. Vet Pathol 2008; 45:288-96. [DOI: 10.1354/vp.45-3-288] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies have shown that the brain is a target of persistent infection with bovine viral diarrhea virus (BVDV) and have demonstrated viral tropism for neurons as well as other endogenous cell types in diverse brain areas. Apart from foci of mild residual inflammation in some postnatal calves, consistent brain lesions, per se, have not been reported. No similar comprehensive studies of the brain have been reported in bovine fetuses. In the current study, 12 BVDV-seronegative heifers were inoculated intranasally with a 2-ml 4.4 log10 TCID50/ml dose of noncytopathic type 2 BVDV at 75 and 175 days of gestation to create persistently and transiently infected fetuses, respectively. In only persistently infected fetuses, encephaloclastic lesions resulting in pseudocysts were observed in the subependymal zone in the region of the median eminence and adjacent corona radiata as well as in the region of the external capsule associated with lenticulostriate arteries. Additionally, areas of rarefaction in white matter were observed at the tips of cerebrocortical gyri and in the external capsule. The distribution of viral antigen was examined by immunohistochemical labeling using the 15C5 anti-BVDV monoclonal antibody. Viral antigen was detected only in calves inoculated at 75 days of gestation, i.e., persistently infected. The pattern of BVDV immunolabeling revealed both similarities and differences compared with previous studies in postnatal calves, suggesting that viral infection in the brain is a dynamic and progressive rather than static process.
Collapse
Affiliation(s)
- D. L. Montgomery
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY
| | - A. Van Olphen
- Department of Global Health, College of Public Health, University of South Florida, Tampa, FL
| | - H. Van Campen
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO
| | - T. R. Hansen
- Department and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
16
|
Bourteele S, Oesterle K, Pleschka S, Unterstab G, Ehrhardt C, Wolff T, Ludwig S, Planz O. Constitutive activation of the transcription factor NF-kappaB results in impaired borna disease virus replication. J Virol 2005; 79:6043-51. [PMID: 15857990 PMCID: PMC1091684 DOI: 10.1128/jvi.79.10.6043-6051.2005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The inducible transcription factor NF-kappaB is commonly activated upon RNA virus infection and is a key player in the induction and regulation of the innate immune response. Borna disease virus (BDV) is a neurotropic negative-strand RNA virus, which replicates in the nucleus of the infected cell and causes a persistent infection that can lead to severe neurological disorders. To investigate the activation and function of NF-kappaB in BDV-infected cells, we stably transfected the highly susceptible neuronal guinea pig cell line CRL with a constitutively active (IKK EE) or dominant-negative (IKK KD) regulator of the IKK/NF-kappaB signaling pathway. While BDV titers were not affected in cells with impaired NF-kappaB signaling, the expression of an activated mutant of IkappaB kinase (IKK) resulted in a strong reduction in the intracellular viral titer in CRL cells. Electrophoretic mobility shift assays and luciferase reporter gene assays revealed that neither NF-kappaB nor interferon regulatory factors (IRFs) were activated upon acute BDV infection of wild-type or vector-transfected CRL cells. However, when IKK EE-transfected cells were used as target cells for BDV infection, DNA binding to an IRF3/7-responsive DNA element was detectable. Since IRF3/7 is a key player in the antiviral interferon response, our data indicate that enhanced NF-kappaB activity in the presence of BDV leads to the induction of antiviral pathways resulting in reduced virus titers. Consistent with this observation, the anti-BDV activity of NF-kappaB preferentially spread to areas of the brains of infected rats where activated NF-kappaB was not detectable.
Collapse
Affiliation(s)
- Soizic Bourteele
- Institut für Immunologie, Friedrich Loeffler Institut, Bundesforschungsinstitut für Tiergesundheit, Paul Ehrlich Str. 28, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Klein RS. Regulation of neuroinflammation: the role of CXCL10 in lymphocyte infiltration during autoimmune encephalomyelitis. J Cell Biochem 2005; 92:213-22. [PMID: 15108349 DOI: 10.1002/jcb.20052] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The movement of lymphocytes from the microvasculature into the central nervous system (CNS) parenchyma is an essential step in the pathogenesis of a variety of infectious and autoimmune neuroinflammatory diseases. The lymphocyte chemoattractant CXCL10 and its receptor, CXCR3, are expressed by the CNS and by CNS infiltrating lymphocytes, respectively, only in patients with ongoing CNS inflammation, suggesting an important role for these molecules in the pathogenic process. Numerous studies utilizing animal models and transgenic approaches have indeed supported a role for CXCL10 in the intraparenchymal trafficking of lymphocytes during acute CNS inflammation; however, other studies suggest that its expression is not required for the development of autoimmune forms of CNS inflammation and, in fact, that interference with CXCL10 signaling could lead to increased neuroinflammation. This review will consider the data from these studies and attempt to reconcile them through comparisons of both the neuroinflammatory models and the effects of CXCL10 in the CNS versus lymphoid tissues. Finally, it will define directions for future analyses of CXCL10 and CXCR3 in CNS inflammation so that their potential therapeutic utility can be more completely determined.
Collapse
Affiliation(s)
- Robyn S Klein
- Department Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63119, USA.
| |
Collapse
|
18
|
Bette M, Roehrenbeck A, Dietzschold B, Weihe E. Neuropeptide Y up-regulation in cerebrocortical neurons after Borna Disease Virus infection is unrelated to brain inflammation in rats. Neurosci Lett 2004; 366:197-200. [PMID: 15276246 DOI: 10.1016/j.neulet.2004.05.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Revised: 03/05/2004] [Accepted: 05/15/2004] [Indexed: 10/26/2022]
Abstract
Neuropeptides participate in the pathophysiology of cerebral inflammatory diseases. We analyzed the involvement of neuropeptide Y (NPY) in rat brain infected with Borna Disease Virus (BDV). NPY expressing cerebrocortical neurons were increased during the acute stage of BDV-induced encephalitis. The increase was resistant to immunosuppression by systemic dexamethasone, which greatly reduced inflammatory reactions in the brain. This indicates that the increase of cerebrocortical NPY expression is not causally related to inflammation. As cerebral NPY is known to be increased during experimental seizures and to have anticonvulsive actions, we propose that NPY up-regulated during BDV encephalitis limits seizures known to be associated with Borna Disease.
Collapse
Affiliation(s)
- Michael Bette
- Department of Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University, Robert-Koch Str. 8, D-35033 Marburg, Germany
| | | | | | | |
Collapse
|
19
|
Schmitz DN, Hofmann N, Tomov TL, Kovac AD, Neiss WF, Angelov DN. The correlation between severity of paraparesis and reduced density of resident antigen-presenting cells implicates an unknown role for the spinal perivascular macrophages in experimental autoimmune encephalomyelitis in rats. J Neuroimmunol 2003; 142:31-46. [PMID: 14512162 DOI: 10.1016/s0165-5728(03)00256-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To study alterations in the morphology of spinal perivascular macrophages (SPM) during experimental allergic encephalomyelitis (EAE), we labelled SPM by intracerebroventricular (i.c.v.) injection of horseradish peroxidase (HRP). As earlier electron microscopical analysis had shown severely damaged SPM, we suspected that each inflammatory process is accompanied by the death of SPM. To prove this hypothesis, we compared the numerical density of resident SPM (i.c.v. labelled in red by Fluoro-Ruby) with that of monocytes/macrophages recruited to the perivascular space (i.c.v. labelled in green by Fluoro-Emerald). At the peak of paraparesis, the density of resident SPM was reduced by 33%. Since this reduction contrasted sharply with earlier data indicating a massive increase in the density of SPM during EAE, we checked our findings after general or selective suppression of the immune response to myelin autoantigens with the drugs dexamethasone and copaxone, respectively. Dexamethasone treatment commenced after evident paraparesis accelerated recovery, but did not influence SPM density. Immunisation with copaxone completely prevented the occurrence of EAE (monitored by video-based motion analysis of tail motility); the subsequent histological analysis revealed no reduction in SPM density. Based on this inverse correlation between the severity of EAE and the density of resident macrophages, we conclude that SPM plays an important role in the pathogenesis of EAE.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/drug effects
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigen-Presenting Cells/pathology
- Cell Count
- Cell Movement/immunology
- Dexamethasone/administration & dosage
- Down-Regulation/drug effects
- Down-Regulation/immunology
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fluorescent Dyes/metabolism
- Injections, Intraperitoneal
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Paraparesis/blood
- Paraparesis/immunology
- Paraparesis/pathology
- Paraparesis/physiopathology
- Rats
- Rats, Inbred Lew
- Severity of Illness Index
- Spinal Cord/blood supply
- Spinal Cord/immunology
- Spinal Cord/pathology
- Tail/physiology
- Time Factors
Collapse
Affiliation(s)
- Dennis N Schmitz
- Institut I für Anatomie der Universität zu Köln, Joseph-Stelzmann-Strasse 9, D-50931 Cologne, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Watanabe M, Lee BJ, Yamashita M, Kamitani W, Kobayashi T, Tomonaga K, Ikuta K. Borna disease virus induces acute fatal neurological disorders in neonatal gerbils without virus- and immune-mediated cell destructions. Virology 2003; 310:245-53. [PMID: 12781712 DOI: 10.1016/s0042-6822(03)00158-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Borna disease virus (BDV) is a noncytolytic, neurotropic RNA virus that is known to cause neurological disturbances in various animal species. Our previous experiment demonstrated that neonate gerbils develop an acute fatal neurological disease following infection with BDV, Virology 282, 65-76). The study suggested that BDV directly causes functional damage of neuronal cells resulting in the lethal disorder in neonatal gerbils. To extend this finding, we examined whether BDV can induce neurological diseases in the absence of virus- and immune-mediated cell destruction, by using cyclosporine A (CsA)-treated neonatal gerbils. Although CsA completely suppressed specific antibody production and brain inflammation in the infected gerbil brains, the fatal neurological disorder was not inhibited by the treatment. Furthermore, we demonstrated that CsA treatment significantly decreased brain levels of cytokines, except interleukin (IL)-1 beta, in the infected gerbils. These results suggested that BDV replication, as well as brain cytokines, at least IL-1 beta, rapidly induces fatal disturbances in gerbil brain. We demonstrate here that BDV exhibits a unique neuropathogenesis in neonatal gerbil that may be pathologically and immunologically different from those in two other established rodent models, rats and mice. With this novel rodent model of virus infection it should be possible not only to examine acute neurological disturbances without severe neuroanatomical and immunopathological alterations but also to analyze molecular and cellular damage by virus replication in the central nervous system.
Collapse
Affiliation(s)
- Makiko Watanabe
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Freude S, Hausmann J, Hofer M, Pham-Mitchell N, Campbell IL, Staeheli P, Pagenstecher A. Borna disease virus accelerates inflammation and disease associated with transgenic expression of interleukin-12 in the central nervous system. J Virol 2002; 76:12223-32. [PMID: 12414961 PMCID: PMC136910 DOI: 10.1128/jvi.76.23.12223-12232.2002] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2002] [Accepted: 08/19/2002] [Indexed: 12/22/2022] Open
Abstract
Targeted expression of biologically active interleukin-12 (IL-12) in astrocytes of the central nervous system (CNS) results in spontaneous neuroimmunological disease of aged mice. Borna disease virus (BDV) can readily multiply in the mouse CNS but does not trigger disease in most strains. Here we show that a large percentage of IL-12 transgenic mice developed severe ataxia within 5 to 10 weeks after infection with BDV. By contrast, no disease developed in mock-infected IL-12 transgenic and wild-type mice until 4 months of age. Neurological symptoms were rare in infected wild-type animals, and if they occurred, these were milder and appeared later. Histological analyses showed that the cerebellum of infected IL-12 transgenic mice, which is the brain region with strongest transgene expression, contained large numbers of CD4(+) and CD8(+) T cells as well as lower numbers of B cells, whereas other parts of the CNS showed only mild infiltration by lymphocytes. The cerebellum of diseased mice further showed severe astrogliosis, calcifications and signs of neurodegeneration. BDV antigen and nucleic acids were present in lower amounts in the inflamed cerebellum of infected transgenic mice than in the noninflamed cerebellum of infected wild-type littermates, suggesting that IL-12 or IL-12-induced cytokines exhibited antiviral activity. We propose that BDV infection accelerates the frequency by which immune cells such as lymphocytes and NK cells enter the CNS and then respond to IL-12 present in the local milieu causing disease. Our results illustrate that infection of the CNS with a virus that is benign in certain hosts can be harmful in such normally disease-resistant hosts if the tissue is unfavorably preconditioned by proinflammatory cytokines.
Collapse
Affiliation(s)
- Susanna Freude
- Abteilung Neuropathologie, Pathologisches Institut, Universität Freiburg, D-79106 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Hofmann N, Lachnit N, Streppel M, Witter B, Neiss WF, Guntinas-Lichius O, Angelov DN. Increased expression of ICAM-1, VCAM-1, MCP-1, and MIP-1 alpha by spinal perivascular macrophages during experimental allergic encephalomyelitis in rats. BMC Immunol 2002; 3:11. [PMID: 12196270 PMCID: PMC126207 DOI: 10.1186/1471-2172-3-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2002] [Accepted: 08/26/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND T-cells extravasation and CNS parenchyma infiltration during autoimmune neurodegenerative disease can be evoked by local antigen presenting cells. Studying the chemoattracting potential of spinal perivascular macrophages (SPM) during experimental allergic encephalomyelitis (EAE), we observed numerous infiltrates of densely-packed mononuclear cells. Apart from the poor spatial and optical resolution, no differentiation between the resident SPM (mabs ED1+, ED2+) and the just recruited monocytes/macrophages (mab ED1+) was possible. RESULTS This is why we labeled SPM by injections of different fluoresecent dyes into the lateral cerebral ventricle before induction of active EAE. Within an additional experimental set EAE was induced by an intraperitoneal injection of T-cells specifically sensitized to myelin basic protein (MBP) and engineered to express the green fluorescent protein (GFP). In both experiments we observed a strong activation of SPM (mabs OX6+, SILK6+, CD40+, CD80+, CD86+) which was accompanied by a consistently increased expression of ICAM-1, VCAM-1, and the chemokines MCP-1 and MIP-1alpha. CONCLUSION These observations indicate that SPM play a role in promoting lymphocyte extravasation.
Collapse
Affiliation(s)
- Nils Hofmann
- Institut für Anatomie der Universität zu Köln, Germany
| | - Nina Lachnit
- Institut für Anatomie der Universität zu Köln, Germany
| | - Michael Streppel
- Klinik für Hals-, Nasen- und Ohrenheilkunde der Universität zu Köln, Germany
| | | | | | | | | |
Collapse
|
23
|
Janabi N. Selective inhibition of cyclooxygenase-2 expression by 15-deoxy-Delta(12,14)(12,14)-prostaglandin J(2) in activated human astrocytes, but not in human brain macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4747-55. [PMID: 11971025 DOI: 10.4049/jimmunol.168.9.4747] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Overexpression of the inducible cyclooxygenase (COX-2) and inducible NO synthase (iNOS) in activated brain macrophages (microglia) and astrocytes appears central to many neuroinflammatory conditions. 15-Deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) is a ligand for the peroxisome proliferator-activated receptor (PPAR)gamma. It has been proposed as an inhibitor of microglial activation, based on the study of iNOS down-regulation in rodent microglia. Because iNOS induction after cytokine activation remains controversial in human microglia, we examined the effect of 15d-PGJ(2) and other PPAR agonists on human microglia and astrocytes, using COX-2 induction as an index of activation. We found that PPAR alpha ligands (clofibrate and WY14643) enhanced IL-1 beta-induced COX-2 expression in human astrocytes and microglia, while inhibiting IL-1 beta plus IFN-gamma induction of iNOS in astrocytes. This is the first description of an inhibition of iNOS uncoupled from that of COX-2. 15d-PGJ(2) suppressed COX-2 induction in human astrocytes. It prevented NF-kappa B binding to the COX-2 promoter through a new pathway that is the repression of NF-kappa Bp50 induction by IL-1 beta. In contrast, 15d-PGJ(2) increased c-Jun and c-Fos DNA-binding activity in astrocytes, which may result in the activation of other inflammatory pathways. In human microglia, no effect of 15d-PGJ(2) on COX-2 and NF-kappa Bp65/p50 induction was observed. However, the entry of 15d-PGJ(2) occurred in microglia because STAT-1 and c-Jun expression was modulated. Our data suggest the existence of novel pathways mediated by 15d-PGJ(2) in human astrocytes. They also demonstrate that, unlike astrocytes and peripheral macrophages or rodent brain macrophages, human microglia are not subject to the anti-inflammatory effect of 15d-PGJ(2) in terms of COX-2 inhibition.
Collapse
Affiliation(s)
- Nazila Janabi
- Laboratory of Molecular Medicine and Neuroscience, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Bacher M, Weihe E, Dietzschold B, Meinhardt A, Vedder H, Gemsa D, Bette M. Borna disease virus-induced accumulation of macrophage migration inhibitory factor in rat brain astrocytes is associated with inhibition of macrophage infiltration. Glia 2002. [DOI: 10.1002/glia.10013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Hooper DC, Kean RB, Scott GS, Spitsin SV, Mikheeva T, Morimoto K, Bette M, Röhrenbeck AM, Dietzschold B, Weihe E. The central nervous system inflammatory response to neurotropic virus infection is peroxynitrite dependent. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3470-7. [PMID: 11544340 DOI: 10.4049/jimmunol.167.6.3470] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have recently demonstrated that increased blood-CNS barrier permeability and CNS inflammation in a conventional mouse model of experimental allergic encephalomyelitis are dependent upon the production of peroxynitrite (ONOO(-)), a product of the free radicals NO* and superoxide (O2*(-)). To determine whether this is a reflection of the physiological contribution of ONOO(-) to an immune response against a neurotropic pathogen, we have assessed the effects on adult rats acutely infected with Borna disease virus (BDV) of administration of uric acid (UA), an inhibitor of select chemical reactions associated with ONOO(-). The pathogenesis of acute Borna disease in immunocompetent adult rats results from the immune response to the neurotropic BDV, rather than the direct effects of BDV infection of neurons. An important stage in the BDV-specific neuroimmune response is the invasion of inflammatory cells into the CNS. UA treatment inhibited the onset of clinical disease, and prevented the elevated blood-brain barrier permeability as well as CNS inflammation seen in control-treated BDV-infected rats. The replication and spread of BDV in the CNS were unchanged by the administration of UA, and only minimal effects on the immune response to BDV Ags were observed. These results indicate that the CNS inflammatory response to neurotropic virus infection is likely to be dependent upon the activity of ONOO(-) or its products on the blood-brain barrier.
Collapse
Affiliation(s)
- D C Hooper
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Nakajima H, Kobayashi M, Pollard RB, Suzuki F. Monocyte chemoattractant protein‐1 enhances HSV‐induced encephalomyelitis by stimulating Th2 responses. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.3.374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Affiliation(s)
- Hideto Nakajima
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston
| | - Makiko Kobayashi
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston
| | - Richard B. Pollard
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston
| | - Fujio Suzuki
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston
| |
Collapse
|
27
|
Dörries R. The role of T-cell-mediated mechanisms in virus infections of the nervous system. Curr Top Microbiol Immunol 2001; 253:219-45. [PMID: 11417137 DOI: 10.1007/978-3-662-10356-2_11] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T lymphocytes play a decisive role in the course and clinical outcome of viral CNS infection. Summarizing the information presented in this review, the following sequence of events might occur during acute virus infection: After invasion of the host and a few initial rounds of replication, the virus reaches the CNS in most cases by hematogeneous spread. After passage through the BBB, CNS cells are infected and replication of virus in brain cells causes activation of the surrounding microglia population. Moreover, local production of IFN-alpha/beta induces expression of MHC antigens on CNS cells, and microglial cells start to phagocytose cellular debris, which accumulates as a result of virus-induced cytopathogenic effects. Upon phagocytosis, microglia becomes more activated; they up-regulate MHC molecules, acquire antigen presentation capabilities and secrete chemokines. This will initiate up-regulation of adhesion molecules on adjacent endothelial cells of the BBB. Transmigration of activated T lymphocytes through the BBB is followed by interaction with APC, presenting the appropriate peptides in the context of MHC antigens. It appears that CD8+ T lymphocytes are amongst the first mononuclear cells to arrive at the infected tissue. Without a doubt, their induction and attraction is deeply influenced by natural killer cells, which, after virus infection, secrete IFN-gamma, a cytokine that stimulates CD8+ T cells and diverts the immune response to a TH1-type CD4+ T cell-dominated response. Following the CD8+ T lymphocytes, tissue-penetrating, TH1 CD4+ T cells contact local APC. This results in a tremendous up-regulation of MHC molecules and secretion of more chemotactic and toxic substances. Consequently an increasing number of inflammatory cells, including macrophages/microglia and finally antibody-secreting plasma cells, are attracted to the site of virus infection. All trapped cells are mainly terminally differentiated cells that are going to enter apoptosis during or shortly after exerting their effector functions. The clinical consequences and the influence of the effector phase on the further course of the infection depends on the balance and fine-tuning of the contributing lymphoid cell populations. Generally, any delay in the recruitment of effector lymphocytes to the tissue or an unbalanced combination of lymphocyte subsets allows the virus to spread in the CNS, which in turn will cause severe immune-mediated tissue effects as well as disease. If either too late or partially deficient, the immune system response may contribute to a lethal outcome or cause autosensitization to brain-specific antigens by epitope spreading to the antigen-presenting system in peripheral lymphoid tissue. This could form the basis for subsequent booster reactions of autosensitized CD4+ T cells--a process that finally will end in an inflammatory autoimmune reaction, which in humans we call multiple sclerosis. In contrast, a rapid and specific local response in the brain tissue will result in efficient limitation of viral spread and thereby a subclinical immune system-mediated termination of the infection. After clearance of virus-infected cells, downsizing of the local response probably occurs via self-elimination of the contributing T cell populations and/or by so far unidentified signal pathways. However, much of this is highly speculative, and more data have to be collected to make decisive conclusions regarding this matter. Several strategies have been developed by viruses to escape T cell-mediated eradication, including interference with the MHC class I presentation pathway of the host cell or "hiding" in cells which lack MHC class I expression. This may result in life-long persistence of the virus in the brain, a state which probably is actively controlled by T lymphocytes. Under severe immunosuppression, however, reactivation of viral replication can occur, which is a lethal threat to the host.
Collapse
Affiliation(s)
- R Dörries
- Department of Virology, Institute of Medical Microbiology and Hygiene, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
28
|
Walther M, Popratiloff A, Lachnit N, Hofmann N, Streppel M, Guntinas-Lichius O, Neiss WF, Angelov DN. Exogenous antigen containing perivascular phagocytes induce a non-encephalitogenic extravasation of primed lymphocytes. J Neuroimmunol 2001; 117:30-42. [PMID: 11431002 DOI: 10.1016/s0165-5728(01)00302-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent evidence suggests that T-lymphocyte extravasation and CNS-parenchymal infiltration during autoimmune disease might be regulated by antigen-presenting (ED2(+)) cerebral/spinal perivascular phagocytes (CPP/SPP). Since the massive erythrocytic and leukocytic infiltrates in the CNS of rats with experimental allergic encephalomyelitis do not allow a precise differentiation between CPP/SPP and the invading cells in the Virchow-Robin space, we developed a new immune-response model whereby the extravasation of T-lymphocytes was not followed by other blood cells. Adult Lewis rats were sensitized to horseradish peroxidase (HRP). Subsequent intracerebroventricular (i.c.v.) injections of HRP and/or Fluoro-Emerald (FE) served to: (1) challenge the primed T-lymphocytes and (2) label the CPP/SPP for additional immunocytochemical analysis. We found that 24 h and 3 days after single, double, or triple antigen boosting T-lymphocytes (R73(+), W3/25(+), OX50(+)) entered the Virchow-Robin space but did not break through the astrocytic glia limitans. Instead they adhered to HRP-containing activated CPP/SPP (mabs OX-6(+), SILK6(+), CD40(+), CD80(+), CD86(+)). This selective contact was mediated neither by cell adhesion molecules (P-selectin, ICAM-1, VCAM-1), nor promoted by chemokine receptors (CCR1, CCR5) or chemokines (monocyte chemoattractant protein (MCP)-1, MIP-1alpha, MIP-1beta, RANTES). This non-inflammatory, but antigen-dependent lymphocyte extravasation provides optimal conditions to further study the CNS immune response.
Collapse
Affiliation(s)
- M Walther
- Institut I für Anatomie der Universität zu Köln, Joseph-Stelzmann-Strasse 9, D-50931, Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The biology of Borna disease virus (BDV) strongly supports the likelihood of human infection with BDV or a variant of BDV. Thus far, the evidence supporting BDV infection in humans has initiated much controversy among basic and clinical scientists; only time and additional research will support or refute the hypothesis of human BDV infection. Until an assay of acceptable specificity and sensitivity has been developed, validated, and used to document human BDV infection, scientists cannot reasonably begin to associate BDV infection with specific disease syndromes. Clinical studies seeking causal associations between BDV infection and specific diseases must ensure the proper identification of the BDV infection status of patients and control subjects by using a validated, highly sensitive, and highly specific assay (or series of assays). For clinical studies, a highly sensitive "screening" test followed by a highly specific confirmatory test will be of significant benefit. Although it is possible to formulate hypotheses about the clinical outcomes of human BDV infection based on animal model work, to date no human disease has been causally linked to human BDV infection. Scientists all over the world are actively pursuing these issues, and with continuing advances in clinical and basic BDV research, the answers cannot be far away.
Collapse
Affiliation(s)
- K M Carbone
- FDA/CBER, HFM 460, 8800 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Askovic S, Favara C, McAtee FJ, Portis JL. Increased expression of MIP-1 alpha and MIP-1 beta mRNAs in the brain correlates spatially and temporally with the spongiform neurodegeneration induced by a murine oncornavirus. J Virol 2001; 75:2665-74. [PMID: 11222690 PMCID: PMC115891 DOI: 10.1128/jvi.75.6.2665-2674.2001] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The chimeric murine oncornavirus FrCas(E) causes a rapidly progressive paralytic disease associated with spongiform neurodegeneration throughout the neuroaxis. Neurovirulence is determined by the sequence of the viral envelope gene and by the capacity of the virus to infect microglia. The neurocytopathic effect of this virus appears to be indirect, since the cells which degenerate are not infected. In the present study we have examined the possible role of inflammatory responses in this disease and have used as a control the virus F43. F43 is an highly neuroinvasive but avirulent virus which differs from FrCas(E) only in 3' pol and env sequences. Like FrCas(E), F43 infects large numbers of microglial cells, but it does not induce spongiform neurodegeneration. RNAase protection assays were used to detect differential expression of genes encoding a variety of cytokines, chemokines, and inflammatory cell-specific markers. Tumor necrosis factor alpha (TNF-alpha) and TNF-beta mRNAs were upregulated in advanced stages of disease but not early, even in regions with prominent spongiosis. Surprisingly there was no evidence for upregulation of the cytokines interleukin-1 alpha (IL-1 alpha), IL-1 beta, and IL-6 or of the microglial marker F4/80 at any stage of this disease. In contrast, increased levels of the beta-chemokines MIP-1 alpha and -beta were seen early in the disease and were concentrated in regions of the brain rich in spongiosis, and the magnitude of responses was similar to that observed in the brains of mice injected with the glutamatergic neurotoxin ibotenic acid. MIP-1alpha and MIP-1beta mRNAs were also upregulated in F43-inoculated mice, but the responses were three- to fivefold lower and occurred later in the course of infection than was observed in FrCas(E)-inoculated mice. These results suggest that the robust increase in expression of MIP-1 alpha and MIP-1 beta in the brain represents a correlate of neurovirulence in this disease, whereas the TNF responses are likely secondary events.
Collapse
Affiliation(s)
- S Askovic
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana 59840, USA
| | | | | | | |
Collapse
|
31
|
Sauder C, Hallensleben W, Pagenstecher A, Schneckenburger S, Biro L, Pertlik D, Hausmann J, Suter M, Staeheli P. Chemokine gene expression in astrocytes of Borna disease virus-infected rats and mice in the absence of inflammation. J Virol 2000; 74:9267-80. [PMID: 10982374 PMCID: PMC102126 DOI: 10.1128/jvi.74.19.9267-9280.2000] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Borna disease virus (BDV) causes CD8(+) T-cell-mediated meningoencephalitis in immunocompetent mice and rats, thus providing a valuable animal model for studying the mechanisms of virus-induced central nervous system (CNS) immunopathology. Chemokine-mediated leukocyte recruitment to the CNS is a crucial step in the development of neurological disease. We found increased mRNA levels of IP-10 and other chemokines in brains of adult rats following infection with BDV. The marked increase in chemokine gene expression at about day 8 postinfection seemed to immediately precede the inflammatory process. In brains of rats infected as newborns, in which inflammation was only mild and transient, sustained expression of IP-10 and RANTES genes was observed. In situ hybridization studies revealed that astrocytes were the major source of IP-10 mRNAs in brains of rats infected as newborns and as adults. In brains of infected mice lacking CD8(+) T cells (beta2m(0/0)), transcripts encoding IP-10 and RANTES were also observed. IP-10 transcripts were also present in a small number of scattered astrocytes of infected knockout mice lacking mature B and T cells as well as functional alpha/beta and gamma interferon receptors, indicating that BDV can induce chemokine synthesis in the absence of interferons and other B- or T-cell-derived cytokines. These data provide strong evidence that CNS-resident cells are involved in the early localized host immune response to infection with BDV and support the concept that chemokines are pivotal for the initiation of virus-induced CNS inflammation.
Collapse
Affiliation(s)
- C Sauder
- Abteilung Virologie, Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, D-79104 Freiburg
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nakamura Y, Watanabe M, Kamitani W, Taniyama H, Nakaya T, Nishimura Y, Tsujimoto H, Machida S, Ikuta K. High prevalence of Borna disease virus in domestic cats with neurological disorders in Japan. Vet Microbiol 1999; 70:153-69. [PMID: 10596800 DOI: 10.1016/s0378-1135(99)00135-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A total of 15 (T-1-T-15) domestic cats with neurological disorders in Tokyo area were examined for association with Borna disease virus (BDV). None had detectable antibodies to feline immunodeficiency virus (FIV), feline leukemia virus, feline infectious peritonitis virus and Toxoplasma gondii, and only cat T-8 had detectable antibody to FIV. Serological and molecular epidemiological studies revealed a significantly high prevalence of BDV infection in these cats: antibodies against BDV p24 and/or p40 proteins in 10/15 (66.7%) and p24 and/or p40 RNA in peripheral blood mononuclear cells in 8/15 (53.3%). Further, in situ hybridization and immunohistochemistry analyses of the autopsied brain samples derived from one of the cats (T-15) revealed BDV RNA predominantly in neuronal cells in restricted regions, such as olfactory bulb and medulla of cerebrum. Thus, BDV is present in Japanese domestic cats with neurological disorders at a high prevalence.
Collapse
Affiliation(s)
- Y Nakamura
- Section of Serology, Institute of Immunological Science, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lawrence MS, Foellmer HG, Elsworth JD, Kim JH, Leranth C, Kozlowski DA, Bothwell AL, Davidson BL, Bohn MC, Redmond DE. Inflammatory responses and their impact on beta-galactosidase transgene expression following adenovirus vector delivery to the primate caudate nucleus. Gene Ther 1999; 6:1368-79. [PMID: 10467361 DOI: 10.1038/sj.gt.3300958] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An E1, E3 deleted adenovirus vector, serotype 5, carrying the marker gene LacZ was bilaterally microinfused into the caudate nuclei of 10 St Kitts green monkeys. The location and number of cells expressing transgene and host immunologic response were evaluated at 1 week (n = 2) and 1 month (n = 8) following vector infusion. A large number of cells expressed beta-galactosidase in some monkeys, exceeding 600000 in one monkey, but no expression was seen in three of 10. All monkeys had positive adenoviral antibody titers before vector infusion, indicating the possibility of previous exposure to some adenovirus, but only one showed a significant increase in titer afterwards. Inflammatory cell markers revealed an inverse correlation between transgene expression and the extent of inflammatory response. Dexamethasone administered immediately before and for 8 days following vector delivery, however, had no effect on transgene expression. The demonstration of significant inflammatory responses in the brain of some individual primates, including demyelination, indicates the need for new generations of adenovirus vectors, or the successful suppression of inflammatory responses, before this vector is suitable for non-cytotoxic clinical applications in the CNS.
Collapse
Affiliation(s)
- M S Lawrence
- Yale University School of Medicine, Neural Transplantation and Repair Program, Department of Psychiatry, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Plata-Salamán CR, Ilyin SE, Gayle D, Romanovitch A, Carbone KM. Persistent Borna disease virus infection of neonatal rats causes brain regional changes of mRNAs for cytokines, cytokine receptor components and neuropeptides. Brain Res Bull 1999; 49:441-51. [PMID: 10483922 DOI: 10.1016/s0361-9230(99)00081-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Borna disease virus (BDV) replicates in brain cells. The neonatally infected rat with BDV exhibits developmental-neuromorphological abnormalities, neuronal cytolysis, and multiple behavioral and physiological alterations. Here, we report on the levels of interleukin-1beta (IL-1beta), IL-1 receptor antagonist (IL-1Ra), tumor necrosis factor-alpha (TNF-alpha), transforming growth factor-beta1 (TGF-beta1), IL-1 receptor type I (IL-1RI), IL-1 receptor accessory protein (IL-1R AcP) I and II, glycoprotein 130, and various neuropeptide mRNAs in the cerebellum, parieto-frontal cortex, hippocampus and hypothalamus of BDV-infected rats at 7 and 28 days postintracerebral BDV inoculation. The data show that cytokine and neuropeptide mRNA components are abnormal and differentially modulated in brain regions. IL-1beta, TNF-alpha and TGF-beta1 mRNA levels were up-regulated in all brain regions following BDV inoculation. The same cerebellar samples from BDV-infected animals exhibited the highest levels of IL-1beta, IL-1Ra, TNF-alpha, IL-1RI, and IL-1R AcP II mRNA expression. The profiles of IL-1beta, IL-1Ra, TNF-alpha, and TGF-beta1 mRNA induction in the cerebellar samples were highly intercorrelated, indicating an association among cytokine ligand mRNAs. Cytokine mRNA induction was differentially up-regulated among brain regions, except for TGF-beta1. Specificity of transcriptional changes in response to BDV infection is also suggested by the up-regulation of cytokine and neuropeptide Y mRNAs associated with down-regulation of pro-opiomelanocortin, and with no change of IL-1R AcPI, dynorphin and leptin receptor mRNAs in the same brain region samples. Other data also show a differential mRNA component modulation in distinct brain regions obtained from the same rats depending on the stage of BDV infection. The conclusion of these studies is that cytokines may play a role in the neuropathophysiology of neonatally BDV-infected rats.
Collapse
Affiliation(s)
- C R Plata-Salamán
- Division of Molecular Biology, School of Life and Health Sciences, University of Delaware, Newark, USA.
| | | | | | | | | |
Collapse
|
35
|
Sauder C, de la Torre JC. Cytokine expression in the rat central nervous system following perinatal Borna disease virus infection. J Neuroimmunol 1999; 96:29-45. [PMID: 10227422 DOI: 10.1016/s0165-5728(98)00272-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Borna disease virus (BDV) causes central nervous system (CNS) disease in several vertebrate species, which is frequently accompanied by behavioral abnormalities. In the adult rat, intracerebral (i.c.) BDV infection leads to immunomediated meningoencephalitis. In contrast, i.c. infection of neonates causes a persistent infection in the absence of overt signs of brain inflammation. These rats (designated PTI-NB) display distinct behavioral and neurodevelopmental abnormalities. However, the molecular mechanisms for these virally induced CNS disturbances are unknown. Cytokines play an important role in CNS function, both under normal physiological and pathological conditions. Astrocytes and microglia are the primary resident cells of the central nervous system with the capacity to produce cytokines. Strong reactive astrocytosis is observed in the PTI-NB rat brain. We have used a ribonuclease protection assay to investigate the mRNA expression levels of proinflammatory cytokines in different brain regions of PTI-NB and control rats. We show here evidence of a chronic upregulation of proinflammatory cytokines interleukin-6, tumor necrosis factor alpha, interleukins-1alpha, and -1beta in the hippocampus and cerebellum of the PTI-NB rat brain. These brain regions exhibited only a very mild and transient immune infiltration. In contrast, in addition to reactive astrocytes, a strong and sustained microgliosis was observed in the PTI-NB rat brains. Our data suggest that CNS resident cells, namely astrocytes and microglia, are the major source of cytokine expression in the PTI-NB rat brain. The possible implications of these findings are discussed.
Collapse
Affiliation(s)
- C Sauder
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
36
|
Nakamura Y, Nakaya T, Hagiwara K, Momiyama N, Kagawa Y, Taniyama H, Ishihara C, Sata T, Kurata T, Ikuta K. High susceptibility of Mongolian gerbil (Meriones unguiculatus) to Borna disease virus. Vaccine 1999; 17:480-9. [PMID: 10073727 DOI: 10.1016/s0264-410x(98)00222-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Borna disease virus (BDV) is a neurotropic enveloped virus with a nonsegmented, single-, negative-stranded RNA genome. This virus induced encephalitis in experimentally infected adult rats, but in newborn rats BDV established a persistent, tolerant infection with no apparent clinical signs. Here, we report evidence that newborn Mongolian gerbils (Meriones unguiculatus) are more susceptible to experimental intracranial inoculation of horse-derived BDV in persistently infected MDCK cells, compared with similar inoculation in newborn rats. All inoculated newborn gerbils, but not rats, died 30 days after infection. Reverse transcriptase-polymerase chain reaction amplified BDV-specific sequences in several regions including the brain. Histopathological analysis revealed apparent inflammatory reactions in the brains of inoculated gerbils but not rats, although similar levels of BDV RNA were detected in both gerbil and rat brains. BDV-specific antigen and RNA were identified predominantly in neurons in the brains by immunohistochemistry with antibodies to BDV and in situ hybridization with BDV-specific riboprobes, respectively. BDV in the gerbil brain was easily rescued by co-cultivation of the brain homogenate with human oligodendroglioma cells. Thus, gerbils seem to be a useful animal model for studying BDV-induced pathogenesis in the brain.
Collapse
Affiliation(s)
- Y Nakamura
- Section of Serology, Institute of Immunological Science, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Janabi N, Hau I, Tardieu M. Negative Feedback Between Prostaglandin and α- and β-Chemokine Synthesis in Human Microglial Cells and Astrocytes. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.3.1701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The understanding of immune surveillance and inflammation regulation in cerebral tissue is essential in the therapy of neuroimmunological disorders. We demonstrate here that primary human glial cells were able to produce α- and β-chemokines (IL-8 > growth related protein α (GROα) ≫ RANTES > microphage inflammatory protein (MIP)-1α and MIP-1β) in parallel to PGs (PGE2 and PGF2α) after proinflammatory cytokine stimulation: TNF-α + IL-1β induced all except RANTES, which was induced by TNF-α + IFN-γ. Purified cultures of astrocytes and microglia were also induced by the same combination of cytokines, to produce all these mediators except MIP-1α and MIP-1β, which were produced predominantly by astrocytes. The inhibition of PG production by indomethacin led to a 37–60% increase in RANTES, MIP-1α, and MIP-1β but not in GROα and IL-8 secretion. In contrast, inhibition of IL-8 and GRO activities using neutralizing Abs resulted in a specific 6-fold increase in PGE2 but not in PGF2α production by stimulated microglial cells and astrocytes, whereas Abs to β-chemokines had no effect. Thus, the production of PGs in human glial cells down-regulates their β-chemokine secretion, whereas α-chemokine production in these cells controls PG secretion level. These data suggest that under inflammatory conditions, the intraparenchymal production of PGs could control chemotactic gradient of β-chemokines for an appropriate effector cell recruitment or activation. Conversely, the elevated intracerebral α-chemokine levels could reduce PG secretion, preventing the exacerbation of inflammation and neurotoxicity.
Collapse
Affiliation(s)
- Nazila Janabi
- Laboratory of Virus, Neuron and Immunity, Unité de Formation et de Recherche, Kremlin Bicêtre, University of Paris-South, Paris, France
| | - Isabelle Hau
- Laboratory of Virus, Neuron and Immunity, Unité de Formation et de Recherche, Kremlin Bicêtre, University of Paris-South, Paris, France
| | - Marc Tardieu
- Laboratory of Virus, Neuron and Immunity, Unité de Formation et de Recherche, Kremlin Bicêtre, University of Paris-South, Paris, France
| |
Collapse
|
38
|
Röhrenbeck AM, Bette M, Hooper DC, Nyberg F, Eiden LE, Dietzschold B, Weihe E. Upregulation of COX-2 and CGRP expression in resident cells of the Borna disease virus-infected brain is dependent upon inflammation. Neurobiol Dis 1999; 6:15-34. [PMID: 10078970 DOI: 10.1006/nbdi.1998.0225] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Infection of immunocompetent adult rats with Borna disease virus (BDV) causes severe encephalitis and neural dysfunction. The expression of COX-2 and CGRP, genes previously shown to be implicated in CNS disease and peripheral inflammation, was dramatically upregulated in the cortical neurons of acutely BDV-infected rats. Neuronal COX-2 and CGRP upregulation was predominantly seen in brain areas where ED1-positive macrophages/microglia accumulated. In addition, COX-2 expression was strongly induced in brain endothelial cells and the number of COX-2 immunoreactive microglial cells was increased. In contrast, despite increased expression of viral antigens, neither COX-2 nor CGRP expression was altered in the CNS of BDV-infected rats treated with dexamethasone, or tolerant to BDV. Thus, increased CGRP and COX-2 expression in the BDV-infected brain is the result of the inflammatory response and likely to be involved in the pathogenesis of virus-induced encephalitis.
Collapse
Affiliation(s)
- A M Röhrenbeck
- Institute of Anatomy and Cell Biology, Philipps University Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Portis JL, Lynch WP. Dissecting the determinants of neuropathogenesis of the murine oncornaviruses. Virology 1998; 247:127-36. [PMID: 9705905 DOI: 10.1006/viro.1998.9240] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- J L Portis
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana 59840, USA.
| | | |
Collapse
|
40
|
Lane TE, Asensio VC, Yu N, Paoletti AD, Campbell IL, Buchmeier MJ. Dynamic Regulation of α- and β-Chemokine Expression in the Central Nervous System During Mouse Hepatitis Virus-Induced Demyelinating Disease. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.2.970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Infection of C57BL/6 mice with the V5A13.1 strain of mouse hepatitis virus (MHV-V5A13.1) results in an acute encephalomyelitis and chronic demyelinating disease with features similar to the human demyelinating disease multiple sclerosis. Chemokines are a family of proinflammatory cytokines associated with inflammatory pathology in various diseases. The kinetics and histologic localization of chemokine production in the central nervous system of MHV-infected mice were examined to identify chemokines that contribute to inflammation and demyelination. Transcripts for the chemokines cytokine-response gene-2 (CRG-2), regulated on activation, normal T cell expressed and secreted (RANTES), macrophage-chemoattractant protein-1 and protein-3 (MCP-1, MCP-3), macrophage-inflammatory protein-1β (MIP-1β), and MIP-2 were detected in the brains of MHV-infected mice at 3 days postinfection (p.i.), and these transcripts were increased markedly in brains and spinal cords at day 7 p.i., which coincides with the occurrence of acute viral encephalomyelitis. By day 35 p.i., RANTES, CRG-2, and MIP-1β were detected in brains and spinal cords of mice with chronic demyelination. CRG-2 mRNA expression colocalized with viral RNA and was associated with demyelinating lesions. Astrocytes were the predominant cell type expressing CRG-2 mRNA. These observations suggest a role for chemokines, notably CRG-2, in the initiation and maintenance of an inflammatory response following infection with MHV, which is important in contributing to demyelination.
Collapse
Affiliation(s)
- Thomas E. Lane
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Valérie C. Asensio
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Naichen Yu
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Alyssa D. Paoletti
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Iain L. Campbell
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Michael J. Buchmeier
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
41
|
Faraci FM, Heistad DD. Regulation of the cerebral circulation: role of endothelium and potassium channels. Physiol Rev 1998; 78:53-97. [PMID: 9457169 DOI: 10.1152/physrev.1998.78.1.53] [Citation(s) in RCA: 608] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Several new concepts have emerged in relation to mechanisms that contribute to regulation of the cerebral circulation. This review focuses on some physiological mechanisms of cerebral vasodilatation and alteration of these mechanisms by disease states. One mechanism involves release of vasoactive factors by the endothelium that affect underlying vascular muscle. These factors include endothelium-derived relaxing factor (nitric oxide), prostacyclin, and endothelium-derived hyperpolarizing factor(s). The normal vasodilator influence of endothelium is impaired by some disease states. Under pathophysiological conditions, endothelium may produce potent contracting factors such as endothelin. Another major mechanism of regulation of cerebral vascular tone relates to potassium channels. Activation of potassium channels appears to mediate relaxation of cerebral vessels to diverse stimuli including receptor-mediated agonists, intracellular second messenger, and hypoxia. Endothelial- and potassium channel-based mechanisms are related because several endothelium-derived factors produce relaxation by activation of potassium channels. The influence of potassium channels may be altered by disease states including chronic hypertension, subarachnoid hemorrhage, and diabetes.
Collapse
Affiliation(s)
- F M Faraci
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, USA
| | | |
Collapse
|
42
|
Dürrwald R, Ludwig H. Borna disease virus (BDV), a (zoonotic?) worldwide pathogen. A review of the history of the disease and the virus infection with comprehensive bibliography. ZENTRALBLATT FUR VETERINARMEDIZIN. REIHE B. JOURNAL OF VETERINARY MEDICINE. SERIES B 1997; 44:147-84. [PMID: 9197210 DOI: 10.1111/j.1439-0450.1997.tb00962.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A comprehensive history of Borna disease virus (BDV) and this infection, including the complete bibliography, is presented. Over the last 200 years, descriptions of this 'head disease' of horses ('Kopfkrankheit der Pferde') have been given. Considerable losses in the horse population (< 0.8%) led to intensive clinical and (neuro-)pathological investigations of this meningitis cerebrospinalis which occurs with faint behavioural changes, occasionally followed by severe neurological symptomatology and death. The broad experimental host range reflects infections in nature which include horses, sheep, cattle, cats, dogs, rodents, ostriches, and some zoo animals. BDV infections are associated with phylogentically old brain areas, and the retina. Occasionally, expression in the autonomic nervous system occurs, besides its neurotropism BDV can spread to peripheral organs, especially to epithelial tissues and peripheral blood mononuclear cells. Infections of humans that can be monitored by antibodies, antigens or nucleic acids in blood samples are prominent features of future interest. BDV, the prototype of the family Bornaviridae is an enveloped spherical virus carrying an 8.9 kb single-stranded, non-segmented RNA with negative polarity which replicates in the nucleus. These features together with its considerable genetic stability make this non-cytopathogenic virus an evolutionary 'old pathogen' in nature.
Collapse
Affiliation(s)
- R Dürrwald
- Institut für Virologie, Freie Universität Berlin, Germany
| | | |
Collapse
|
43
|
Gonzalez-Dunia D, Sauder C, de la Torre JC. Borna disease virus and the brain. Brain Res Bull 1997; 44:647-64. [PMID: 9421127 PMCID: PMC7126547 DOI: 10.1016/s0361-9230(97)00276-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/1997] [Revised: 06/30/1997] [Accepted: 07/07/1997] [Indexed: 02/05/2023]
Abstract
Viruses with the ability to establish persistent infection in the central nervous system (CNS) can induce progressive neurologic disorders associated with diverse pathological manifestations. Clinical, epidemiological, and virological evidence supports the hypothesis that viruses contribute to human mental diseases whose etiology remains elusive. Therefore, the investigation of the mechanisms whereby viruses persist in the CNS and disturb normal brain function represents an area of research relevant to clinical and basic neurosciences. Borna disease virus (BDV) causes CNS disease in several vertebrate species characterized by behavioral abnormalities. Based on its unique features, BDV represents the prototype of a new virus family. BDV provides an important model for the investigation of the mechanisms and consequences of viral persistence in the CNS. The BDV paradigm is amenable to study virus-cell interactions in the CNS that can lead to neurodevelopmental abnormalities, immune-mediated damage, as well as alterations in cell differentiated functions that affect brain homeostasis. Moreover, seroepidemiological data and recent molecular studies indicate that BDV is associated with certain neuropsychiatric diseases. The potential role of BDV and of other yet to be uncovered BDV-related viruses in human mental health provides additional impetus for the investigation of this novel neurotropic infectious agent.
Collapse
Affiliation(s)
- D Gonzalez-Dunia
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|