51
|
Nosocomial herpes simplex encephalitis: Does it really exist? J Infect Public Health 2017; 11:142. [PMID: 28209321 DOI: 10.1016/j.jiph.2016.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 11/25/2016] [Indexed: 12/26/2022] Open
|
52
|
Zimmermann J, Hafezi W, Dockhorn A, Lorentzen EU, Krauthausen M, Getts DR, Müller M, Kühn JE, King NJC. Enhanced viral clearance and reduced leukocyte infiltration in experimental herpes encephalitis after intranasal infection of CXCR3-deficient mice. J Neurovirol 2017; 23:394-403. [PMID: 28116674 DOI: 10.1007/s13365-016-0508-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 12/06/2016] [Accepted: 12/14/2016] [Indexed: 12/25/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) encephalitis (HSE) is the most common fatal sporadic encephalitis in developed countries. There is evidence from HSE animal models that not only direct virus-mediated damage caused but also the host's immune response contributes to the high mortality of the disease. Chemokines modulate and orchestrate this immune response. Previous experimental studies in HSE models identified the chemokine receptor CXCR3 and its ligands as molecules with a high impact on the course of HSE in mouse models. In this study, the role of the chemokine receptor CXCR3 was evaluated after intranasal infection with the encephalitogenic HSV-1 strain 17 syn+ using CXCR3-deficient mice (CXCR3-/-) and wild-type controls. We demonstrated a neurotropic viral spread into the CNS of after intranasal infection. Although viral load and histological distribution of infected neurons were independent from CXCR3 signaling early after infection, CXCR3-deficient mice cleared HSV-1 more efficiently 14 days after infection. Furthermore, CXCR3 deficiency led to a decreased weight loss in mice after HSV-1 infection. T cell infiltration and microglial activation was prominently reduced by inhibition of CXCR3 signaling. Quantitative PCR of proinflammatory cytokines and chemokines confirmed the reduced neuroinflammatory response in CXCR3-deficient mice during HSE. Our results demonstrate that the recruitment of peripheral immune cells into the CNS, induction of neuroinflammation, and consecutive weight loss during herpes encephalitis is modulated by CXCR3 signaling. Interruption of the CXCR3 pathway ameliorates the detrimental host immune response and in turn, leads paradoxically to an enhanced viral clearance after intranasal infection. Our data gives further insight into the role of CXCR3 during HSE after intranasal infection.
Collapse
Affiliation(s)
- J Zimmermann
- Department of Neurology, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - W Hafezi
- University Hospital Münster, Institute of Medical Microbiology-Clinical Virology, Münster, Germany
| | - A Dockhorn
- Department of Neurology, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - Eva U Lorentzen
- University Hospital Münster, Institute of Medical Microbiology-Clinical Virology, Münster, Germany
| | - M Krauthausen
- Department of Neurology, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - Daniel R Getts
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Cour Pharmaceutical Development Company, Elmhurst, IL, USA.,The Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - M Müller
- Department of Neurology, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany.
| | - Joachim E Kühn
- University Hospital Münster, Institute of Medical Microbiology-Clinical Virology, Münster, Germany
| | - Nicholas J C King
- The Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
53
|
Menasria R, Canivet C, Piret J, Gosselin J, Boivin G. Both Cerebral and Hematopoietic Deficiencies in CCR2 Result in Uncontrolled Herpes Simplex Virus Infection of the Central Nervous System in Mice. PLoS One 2016; 11:e0168034. [PMID: 27930721 PMCID: PMC5145225 DOI: 10.1371/journal.pone.0168034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022] Open
Abstract
CCR2 is a chemokine receptor expressed on the surface of blood leukocytes, particularly «Ly6Chi» inflammatory monocytes and microglia. Signaling through this receptor is thought to influence the immune activity of microglia as well as monocytes egress from the bone marrow (BM) and their trafficking into the central nervous system (CNS) in several neurological diseases. During experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE), CCR2 deficiency has been reported to exacerbate the outcome of the disease. However, the precise contribution of CCR2 expressed in cells of the CNS or peripheral monocytes in the protection against HSE remains unclear. To dissect the differential role of CCR2 during HSE, chimeric mice with receptor deficiency in the brain or blood cells were generated by transplanting wild-type (WT) C57BL/6 or CCR2-/- BM-derived cells in CCR2-/- (WT→CCR2-/-) and WT (CCR2-/-→WT) mice, respectively. Our results indicate that following intranasal infection with 1.2x106 plaque forming units of HSV-1, CCR2 deficiency in hematopoietic cells and, to a lesser extent, in CNS exacerbates the outcome of HSE. Mortality rates of CCR2-/- (71.4%) and CCR2-/-→WT (57.1%) mice were significantly higher than that of WT (15.3%; P<0.01 and P<0.05, respectively) but the difference did not reach statistical significance for WT→CCR2-/- animals (42.8%; P = 0.16). Both peripheral and CNS deficiencies in CCR2 resulted in increased infectious viral titers and wider dissemination of HSV antigens in the brain as well as an overproduction of inflammatory cytokines and chemokines including IL-1β, IL-6, CCL2, CCL3 and CCL5. Furthermore, CCR2 deficiency in the hematopoietic system altered monocytes egress from the BM and their recruitment to the CNS, which may contribute to the failure in HSV-1 containment. Collectively, these data suggest that CCR2 expressed on cells of CNS and especially on peripheral monocytes is important for the control of HSV-1 replication and inflammatory environment during experimental HSE.
Collapse
Affiliation(s)
- Rafik Menasria
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Coraline Canivet
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunity, CHU of Quebec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
54
|
Selmi C, Barin JG, Rose NR. Current trends in autoimmunity and the nervous system. J Autoimmun 2016; 75:20-29. [DOI: 10.1016/j.jaut.2016.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 08/06/2016] [Indexed: 01/17/2023]
|
55
|
Martínez-Martín N, Viejo-Borbolla A, Alcami A. Herpes simplex virus particles interact with chemokines and enhance cell migration. J Gen Virol 2016; 97:3007-3016. [DOI: 10.1099/jgv.0.000616] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Nadia Martínez-Martín
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid, Madrid, Spain
| | - Abel Viejo-Borbolla
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
56
|
Gurung HR, Carr MM, Carr DJJ. Cornea lymphatics drive the CD8 + T-cell response to herpes simplex virus-1. Immunol Cell Biol 2016; 95:87-98. [PMID: 27577867 PMCID: PMC5209249 DOI: 10.1038/icb.2016.80] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 01/19/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) infection of the cornea induces vascular endothelial growth factor (VEGF)-A-dependent lymphangiogenesis. However, the extent to which HSV-1-induced corneal lymphangiogenesis impacts the adaptive immune response has not been characterized. Here, we used floxed VEGF-A mice to study the importance of newly created corneal lymphatic vessels in the host adaptive immune response to infection. Whereas the mice infected with the parental virus (strain SC16) exhibited robust corneal lymphangiogenesis, mice that received the recombinant virus (SC16 ICP0-Cre) that expresses Cre recombinase under the control of infected cell protein 0 (ICP0), an HSV-1 immediate early gene, showed a significant reduction in lymphangiogenesis. There was no difference in virus recovered from the cornea of mice infected with SC16 vs SC16 ICP0-Cre. However, viral loads were significantly elevated in the trigeminal ganglia (TG) of mice with reduced corneal lymphangiogenesis. The increase in viral titer correlated with a significant loss of HSV-1-specific CD8+ T cells that traffic to the TG of mice infected with the recombinant virus. Intrastromal delivery of size exclusion dye (FITC-dextran) revealed a time-dependent defect in the ability of the lymphatic vessels in SC16 ICP0-Cre infected mice to transport soluble antigen from the cornea to the draining lymph nodes. We interpret these results to suggest that the newly created lymphatic vessels in the cornea driven by HSV-1 infection are critical in the delivery of soluble viral antigen to the draining lymph node and subsequent development of the CD8+ T cell response to HSV-1.
Collapse
Affiliation(s)
- Hem R Gurung
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Meghan M Carr
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Daniel J J Carr
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
57
|
Bradshaw MJ, Venkatesan A. Herpes Simplex Virus-1 Encephalitis in Adults: Pathophysiology, Diagnosis, and Management. Neurotherapeutics 2016; 13:493-508. [PMID: 27106239 PMCID: PMC4965403 DOI: 10.1007/s13311-016-0433-7] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Herpetic infections have plagued humanity for thousands of years, but only recently have advances in antiviral medications and supportive treatments equipped physicians to combat the most severe manifestations of disease. Prompt recognition and treatment can be life-saving in the care of patients with herpes simplex-1 virus encephalitis, the most commonly identified cause of sporadic encephalitis worldwide. Clinicians should be able to recognize the clinical signs and symptoms of the infection and familiarize themselves with a rational diagnostic approach and therapeutic modalities, as early recognition and treatment are key to improving outcomes. Clinicians should also be vigilant for the development of acute complications, including cerebral edema and status epilepticus, as well as chronic complications, including the development of autoimmune encephalitis associated with antibodies to the N-methyl-D-aspartate receptor and other neuronal cell surface and synaptic epitopes. Herein, we review the pathophysiology, differential diagnosis, and clinical and radiological features of herpes simplex virus-1 encephalitis in adults, including a discussion of the most common complications and their treatment. While great progress has been made in the treatment of this life-threatening infection, a majority of patients will not return to their previous neurologic baseline, indicating the need for further research efforts aimed at improving the long-term sequelae.
Collapse
Affiliation(s)
- Michael J Bradshaw
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arun Venkatesan
- Division of Neuroimmunology & Neuroinfectious Diseases, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
58
|
Zhou Y, Zeng YP, Zhou Q, Guan JX, Lu ZN. The effect of captopril on the expression of MMP-9 and the prognosis of neurological function in herpes simplex encephalitis mice. Neurol Res 2016; 38:733-9. [PMID: 27354147 DOI: 10.1080/01616412.2016.1202462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Early increased matrix metalloproteinase-9 (MMP-9) expression is involved in the evolution of herpes simplex encephalitis (HSE) by facilitating the development of cerebrovascular complications. However, the molecular mechanism underlying the detrimental effects of MMP-9 in HSE has not been elucidated. Recent research finds angiotensin II plays an important role in regulation of MMP-9 activity. The aim of this work was to identify the influence of angiotensin-converting enzyme inhibitor (ACEI) captopril on MMP-9 activation after herpes simplex virus 1 (HSV-1) infection. METHODS Animal models of HSE were established by intracerebral inoculation of HSV-1 into mice. Brain tissue ROS levels were measured by staining with dihydroethidium. MMP-9 protein expression was detected by immunofluorescence and brain water content was measured with dry-wet weight method. Neurological function score was quantified 5 d after HSV-1 infection. Microglial cells were treated with various concentrations of captopril. MMP-9 gelatinolytic activity in the supematant of the cell cultures was assessed by zymography. RT-PCR was used to detect the mRNA expressions of p47phox and MMP-9. RESULTS Immunofluorescence showed that expression of MMP-9 in brain tissue was mainly presented in OX-42 positive microglia. Quantification of gelatinolytic activity by densitometry showed that expression of MMP-9 in microglia was significantly increased after HSV-1 infection and inhibited by captopril treatment. NADPH oxidase subunit p47phox and MMP-9 mRNA expression were significantly increased 6 h after HSV-1 infection, and were seen reduced after captopril treatment in dose dependence. Captopril also downregulated ROS and MMP-9 protein expression following encephalitis in vivo, and attenuated brain edema, and improved neurological function. DISCUSSION This compelling evidence suggests that MMP-9 is a key pathogenic factor within HSE. ACEI captopril could reduce the expression of MMP-9 mediated by ROS, then relieve cerebral edema and improve neurological function, which may lay a foundation for further basic research and clinical application.
Collapse
Affiliation(s)
- Yu Zhou
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Yan-Ping Zeng
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Qin Zhou
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Jing-Xia Guan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zu-Neng Lu
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
59
|
Antiviral-guided fractionation and isolation of phenolic compounds from Limonium densiflorum hydroalcoholic extract. CR CHIM 2016. [DOI: 10.1016/j.crci.2016.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
60
|
Tsalenchuck Y, Steiner I, Panet A. Innate defense mechanisms against HSV-1 infection in the target tissues, skin and brain. J Neurovirol 2016; 22:641-649. [PMID: 27098517 DOI: 10.1007/s13365-016-0440-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 11/25/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) initiates productive infection in mucocutaneous tissues to cause cold sores and establishes latent infection in the trigeminal ganglia. Under certain circumstances, HSV-1 may cause encephalitis. Here, we compared host innate defenses against HSV-1 in the two clinically relevant tissues, skin and brain, using a unique ex vivo system of organ culture. Upon HSV-1 infection and spread, apoptosis induction was observed in the skin, but not in brain tissues. While the two tissues elicited interferon (IFN-β) response upon HSV1 infection, IFN induction was more robust in the skin compared to the brain. Moreover, antiviral response to exogenous IFNβ treatment was much stronger in the skin compared to brain tissues. This observation was not related to the availability of the IFN receptor on cells' surface. Taken together, our study demonstrates differential innate antiviral responses to HSV-1 infection that may be exploited in future development of selective and tissue-specific anti-viral treatments.
Collapse
Affiliation(s)
- Yael Tsalenchuck
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Israel Steiner
- Department of Neurology, Rabin Medical Center, Campus Beilinson, Petach Tikva, Israel
| | - Amos Panet
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
61
|
Michael BD, Griffiths MJ, Granerod J, Brown D, Davies NWS, Borrow R, Solomon T. Characteristic Cytokine and Chemokine Profiles in Encephalitis of Infectious, Immune-Mediated, and Unknown Aetiology. PLoS One 2016; 11:e0146288. [PMID: 26808276 PMCID: PMC4726626 DOI: 10.1371/journal.pone.0146288] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
Background Encephalitis is parenchymal brain inflammation due to infectious or immune-mediated processes. However, in 15–60% the cause remains unknown. This study aimed to determine if the cytokine/chemokine-mediated host response can distinguish infectious from immune-mediated cases, and whether this may give a clue to aetiology in those of unknown cause. Methods We measured 38 mediators in serum and cerebrospinal fluid (CSF) of patients from the Health Protection Agency Encephalitis Study. Of serum from 78 patients, 38 had infectious, 20 immune-mediated, and 20 unknown aetiology. Of CSF from 37 patients, 20 had infectious, nine immune-mediated and eight unknown aetiology. Results Heat-map analysis of CSF mediator interactions was different for infectious and immune-mediated cases, and that of the unknown aetiology group was similar to the infectious pattern. Higher myeloperoxidase (MPO) concentrations were found in infectious than immune-mediated cases, in serum and CSF (p = 0.01 and p = 0.006). Serum MPO was also higher in unknown than immune-mediated cases (p = 0.03). Multivariate analysis selected serum MPO; classifying 31 (91%) as infectious (p = 0.008) and 17 (85%) as unknown (p = 0.009) as opposed to immune-mediated. CSF data also selected MPO classifying 11 (85%) as infectious as opposed to immune-mediated (p = 0.036). CSF neutrophils were detected in eight (62%) infective and one (14%) immune-mediated cases (p = 0.004); CSF MPO correlated with neutrophils (p<0.0001). Conclusions Mediator profiles of infectious aetiology differed from immune-mediated encephalitis; and those of unknown cause were similar to infectious cases, raising the hypothesis of a possible undiagnosed infectious cause. Particularly, neutrophils and MPO merit further investigation.
Collapse
MESH Headings
- Adult
- Bacterial Infections/blood
- Bacterial Infections/cerebrospinal fluid
- Biomarkers
- Cell Adhesion Molecules/blood
- Cell Adhesion Molecules/cerebrospinal fluid
- Chemokines/cerebrospinal fluid
- Chemokines/classification
- Cytokines/blood
- Cytokines/cerebrospinal fluid
- Diagnosis, Differential
- Encephalitis/blood
- Encephalitis/cerebrospinal fluid
- Encephalitis/etiology
- Encephalitis/immunology
- Encephalitis, Viral/blood
- Encephalitis, Viral/cerebrospinal fluid
- Encephalitis, Viral/diagnosis
- England/epidemiology
- Female
- Humans
- Infectious Encephalitis/blood
- Infectious Encephalitis/cerebrospinal fluid
- Infectious Encephalitis/diagnosis
- Leukocyte Count
- Male
- Multicenter Studies as Topic
- Mycoses/blood
- Mycoses/cerebrospinal fluid
- Mycoses/diagnosis
- Paraneoplastic Syndromes, Nervous System/blood
- Paraneoplastic Syndromes, Nervous System/cerebrospinal fluid
- Paraneoplastic Syndromes, Nervous System/diagnosis
- Peroxidase/blood
- Peroxidase/cerebrospinal fluid
- Retrospective Studies
- Toxoplasmosis, Cerebral/blood
- Toxoplasmosis, Cerebral/cerebrospinal fluid
- Toxoplasmosis, Cerebral/diagnosis
Collapse
Affiliation(s)
- Benedict D. Michael
- The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| | - Michael J. Griffiths
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
- Alder Hey Children’s NHS Foundation Trust, Liverpool, United Kingdom
| | | | - David Brown
- Public Health England, London, United Kingdom
- Influenza and measles laboratory, IOC, Fiocruz, Rio de Janeiro, Brazil
| | | | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester, United Kingdom
| | - Tom Solomon
- The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
62
|
Michael BD, Griffiths MJ, Granerod J, Brown D, Keir G, Wnęk M, Cox DJ, Vidyasagar R, Borrow R, Parkes LM, Solomon T. The Interleukin-1 Balance During Encephalitis Is Associated With Clinical Severity, Blood-Brain Barrier Permeability, Neuroimaging Changes, and Disease Outcome. J Infect Dis 2015; 213:1651-60. [PMID: 26712949 DOI: 10.1093/infdis/jiv771] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/21/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Encephalitis is parenchymal brain inflammation, commonly due to herpes simplex virus (HSV). Key host inflammatory mediators and their relationship to blood-brain barrier (BBB) permeability, neuroimaging changes, and disease outcome are poorly understood. METHODS We measured levels of 38 mediators in serum (n = 78) and cerebrospinal fluid (n = 37) specimens from patients with encephalitis, including 17 with disease due to HSV infection. Outcome measures were Glasgow coma and outcome scores; CSF to serum albumin ratio, reflecting BBB permeability; and, in patients with HSV infection, magnetic resonance imaging-based temporal lobe volume. RESULTS Serum interleukin 1 receptor antagonist (IL-1RA) levels were elevated in patients with a good outcome (P= .004). Among patients infected with HSV, the ratio of CSF IL-1β to IL-1RA was associated with a worse outcome (P= .009); a ratio of ≥0.55 pg/mL had high specificity and sensitivity for a poor outcome (100% and 83%;P= .015). Temporal lobe volume had a negative correlation with serum IL-1RA level (P= .012) and a positive correlation with serum IL-1α level (P= .0003) and CSF IL-1β level (P= .007). A normal coma score was associated with an elevated interleukin 10 (IL-10) level in serum specimens from HSV-infected patients (P= .007) and CSF specimens from all patients (P= .016); the IL-10 level correlated inversely with BBB permeability (P= .005). CONCLUSIONS A proinflammatory cytokine response is associated with greater clinical severity, BBB permeability, and neuroimaging damage during encephalitis. IL-1 antagonists should be investigated as adjunctive treatment in encephalitis.
Collapse
Affiliation(s)
- Benedict Daniel Michael
- Institute of Infection and Global Health Health Protection Research Unit in Emerging and Zoonotic Infections, National Institute for Health Research, University of Liverpool Walton Centre National Health Service (NHS) Foundation Trust
| | - Michael J Griffiths
- Institute of Infection and Global Health Health Protection Research Unit in Emerging and Zoonotic Infections, National Institute for Health Research, University of Liverpool Alderhey Children's NHS Foundation Trust, Liverpool
| | | | - David Brown
- Public Health England, London Influenza and Measles Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Geoff Keir
- Walton Centre National Health Service (NHS) Foundation Trust
| | | | - Daniel J Cox
- Faculty of Medical and Human Sciences, Centre for Imaging Sciences, Institute of Population Health, University of Manchester
| | - Rishma Vidyasagar
- Faculty of Medical and Human Sciences, Centre for Imaging Sciences, Institute of Population Health, University of Manchester
| | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester, United Kingdom
| | - Laura M Parkes
- Faculty of Medical and Human Sciences, Centre for Imaging Sciences, Institute of Population Health, University of Manchester
| | - Tom Solomon
- Institute of Infection and Global Health Health Protection Research Unit in Emerging and Zoonotic Infections, National Institute for Health Research, University of Liverpool Walton Centre National Health Service (NHS) Foundation Trust
| |
Collapse
|
63
|
Infiltration Pattern of Blood Monocytes into the Central Nervous System during Experimental Herpes Simplex Virus Encephalitis. PLoS One 2015; 10:e0145773. [PMID: 26700486 PMCID: PMC4689369 DOI: 10.1371/journal.pone.0145773] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/08/2015] [Indexed: 12/14/2022] Open
Abstract
The kinetics and distribution of infiltrating blood monocytes into the central nervous system and their involvement in the cerebral immune response together with resident macrophages, namely microglia, were evaluated in experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE). To distinguish microglia from blood monocyte-derived macrophages, chimeras were generated by conditioning C57BL/6 recipient mice with chemotherapy regimen followed by transplantation of bone morrow-derived cells that expressed the green fluorescent protein. Mice were infected intranasally with a sub-lethal dose of HSV-1 (1.2x106 plaque forming units). Brains were harvested prior to and on days 4, 6, 8 and 10 post-infection for flow cytometry and immunohistochemistry analysis. The amounts of neutrophils (P<0.05) and «Ly6Chi» inflammatory monocytes (P<0.001) significantly increased in the CNS compared to non-infected controls on day 6 post-infection, which corresponded to more severe clinical signs of HSE. Levels decreased on day 8 for both leukocytes subpopulations (P<0.05 for inflammatory monocytes compared to non-infected controls) to reach baseline levels on day 10 following infection. The percentage of «Ly6Clow» patrolling monocytes significantly increased (P<0.01) at a later time point (day 8), which correlated with the resolution phase of HSE. Histological analysis demonstrated that blood leukocytes colonized mostly the olfactory bulb and the brainstem, which corresponded to regions where HSV-1 particles were detected. Furthermore, infiltrating cells from the monocytic lineage could differentiate into activated local tissue macrophages that express the microglia marker, ionized calcium-binding adaptor molecule 1. The lack of albumin detection in the brain parenchyma of infected mice showed that the infiltration of blood leukocytes was not necessarily related to a breakdown of the blood-brain barrier but could be the result of a functional recruitment. Thus, our findings suggest that blood monocyte-derived macrophages infiltrate the central nervous system and may contribute, with resident microglia, to the innate immune response seen during experimental HSE.
Collapse
|
64
|
Royer DJ, Zheng M, Conrady CD, Carr DJJ. Granulocytes in Ocular HSV-1 Infection: Opposing Roles of Mast Cells and Neutrophils. Invest Ophthalmol Vis Sci 2015; 56:3763-75. [PMID: 26066745 DOI: 10.1167/iovs.15-16900] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The contributions of mast cells (MCs) to immunologic defense against pathogens in the eye are unknown. We have characterized pericorneal MCs as tissue-resident innate sentinels and determined their impact on the immune response to herpes simplex virus type-1 (HSV-1), a common ocular pathogen. METHODS The impact of mast cells on the immune response to HSV-1 infection was investigated using MC-deficient Kit(W-sh) mice. Virus titers, inflammatory cytokine production, eicosanoid profiles, cellular immune responses, and ocular pathology were evaluated and compared with C57BL/6J mice during an acute corneal HSV-1 infection. RESULTS Corneas of Kit(W-sh) mice have higher viral titers, increased edema, and greater leukocyte infiltration following HSV-1 infection. Following infection, cytokine profiles were slightly elevated overall in Kit(W-sh) mice. Eicosanoid profiles were remarkably different only when comparing uninfected corneas from both groups. Neutrophils within infected corneas expressed HSV-1 antigen, lytic genes, and served as a disease-causing vector when adoptively transferred into immunocompromised animals. Myeloid-derived suppressor cells did not infiltrate into the cornea or suppress the expansion, recruitment, or cytokine production by CD8+ T cells following acute HSV-1 infection. CONCLUSIONS Collectively, these findings provide new insight into host defense in the cornea and the pathogenesis of HSV-1 infection by identifying previously unacknowledged MCs as protective innate sentinels for infection of the ocular surface and reinforcing that neutrophils are detrimental to corneal infection.
Collapse
Affiliation(s)
- Derek J Royer
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Min Zheng
- Department of Ophthalmology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Christopher D Conrady
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Daniel J J Carr
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States 2Department of Ophthalmology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
65
|
Desai DV, Kulkarni SS. Herpes Simplex Virus: The Interplay Between HSV, Host, and HIV-1. Viral Immunol 2015; 28:546-55. [PMID: 26331265 DOI: 10.1089/vim.2015.0012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus proteins interact with host (human) proteins and create an environment conducive for its replication. Genital ulceration due to herpes simplex virus type 2 (HSV-2) infections is an important clinical manifestation reported to increase the risk of human immunodeficiency virus type 1 (HIV-1) acquisition and replication in HIV-1/HSV-2 coinfection. Dampening the innate and adaptive immune responses of the skin-resident dendritic cells, HSV-2 not only helps itself, but creates a "yellow brick road" for one of the most dreaded viruses HIV, which is transmitted mainly through the sexual route. Although, data from clinical trials show that HSV-2 suppression reduces HIV-1 viral load, there are hardly any reports presenting conclusive evidence on the impact of HSV-2 coinfection on HIV-1 disease progression. Be that as it may, understanding the interplay between these three characters (HSV, host, and HIV-1) is imperative. This review endeavors to collate studies on the influence of HSV-derived proteins on the host response and HIV-1 replication. Studying such complex interactions may help in designing and developing common strategies for the two viruses to keep these "partners in crime" at bay.
Collapse
Affiliation(s)
- Dipen Vijay Desai
- Department of Virology, ICMR-National AIDS Research Institute , Pune, India
| | | |
Collapse
|
66
|
Chronic herpes simplex type-1 encephalitis with intractable epilepsy in an immunosuppressed patient. Infection 2015; 44:121-5. [PMID: 26187268 DOI: 10.1007/s15010-015-0822-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Chronic herpes simplex virus type-1 encephalitis (HSE-1) is uncommon. Past reports focused on its association with prior documented acute infection. Here, we describe a patient with increasingly intractable epilepsy from chronic HSE-1 reactivation without history of acute central nervous system infection. CASE PRESENTATION A 49-year-old liver transplant patient with 4-year history of epilepsy after initiation of cyclosporine developed increasingly frequent seizures over 3 months. Serial brain magnetic resonance imaging showed left temporoparietal cortical edema that gradually improved despite clinical decline. Herpes simplex virus type-1 (HSV-1) DNA was detected in cerebrospinal fluid by polymerase chain reaction. Cerebrospinal fluid HSV-1&2 IgM was negative. Seizures were controlled after acyclovir treatment, and the patient remained seizure free at 1-year follow-up. CONCLUSION Chronic HSE is a cause of intractable epilepsy, can occur without a recognized preceding acute phase, and the clinical course of infection may not directly correlate with neuroimaging changes.
Collapse
|
67
|
Liang Y, Yang K, Guo J, Wroblewska J, Fu YX, Peng H. Innate lymphotoxin receptor mediated signaling promotes HSV-1 associated neuroinflammation and viral replication. Sci Rep 2015; 5:10406. [PMID: 25993659 PMCID: PMC4438665 DOI: 10.1038/srep10406] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/10/2015] [Indexed: 12/13/2022] Open
Abstract
Host anti-viral innate immunity plays important roles in the defense against HSV-1 infection. In this study, we find an unexpected role for innate LT/LIGHT signaling in promoting HSV-1 replication and virus induced inflammation in immunocompromised mice. Using a model of footpad HSV-1 infection in Rag1–/– mice, we observed that blocking LT/LIGHT signaling with LTβR-Ig could significantly delay disease progression and extend the survival of infected mice. LTβR-Ig treatment reduced late proinflammatory cytokine release in the serum and nervous tissue, and inhibited chemokine expression and inflammatory cells infiltration in the dorsal root ganglia (DRG). Intriguingly, LTβR-Ig treatment restricted HSV-1 replication in the DRG but not the footpad. These findings demonstrate a critical role for LT/LIGHT signaling in modulating innate inflammation and promoting HSV-1 replication in the nervous system, and suggest a new target for treatment of virus-induced adverse immune response and control of severe HSV-1 infection.
Collapse
Affiliation(s)
- Yong Liang
- 1] Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaiting Yang
- 1] Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingya Guo
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Joanna Wroblewska
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Yang-Xin Fu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
68
|
Lafaille FG, Ciancanelli MJ, Studer L, Smith G, Notarangelo L, Casanova JL, Zhang SY. Deciphering Human Cell-Autonomous Anti-HSV-1 Immunity in the Central Nervous System. Front Immunol 2015; 6:208. [PMID: 26005444 PMCID: PMC4424875 DOI: 10.3389/fimmu.2015.00208] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/15/2015] [Indexed: 12/26/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a common virus that can rarely invade the human central nervous system (CNS), causing devastating encephalitis. The permissiveness to HSV-1 of the various relevant cell types of the CNS, neurons, astrocytes, oligodendrocytes, and microglia cells, as well as their response to viral infection, has been extensively studied in humans and other animals. Nevertheless, human CNS cell-based models of anti-HSV-1 immunity are of particular importance, as responses to any given neurotropic virus may differ between humans and other animals. Human CNS neuron cell lines as well as primary human CNS neurons, astrocytes, and microglia cells cultured/isolated from embryos or cadavers, have enabled the study of cell-autonomous anti-HSV-1 immunity in vitro. However, the paucity of biological samples and their lack of purity have hindered progress in the field, which furthermore suffers from the absence of testable primary human oligodendrocytes. Recently, the authors have established a human induced pluripotent stem cells (hiPSCs)-based model of anti-HSV-1 immunity in neurons, oligodendrocyte precursor cells, astrocytes, and neural stem cells, which has widened the scope of possible in vitro studies while permitting in-depth explorations. This mini-review summarizes the available data on human primary and iPSC-derived CNS cells for anti-HSV-1 immunity. The hiPSC-mediated study of anti-viral immunity in both healthy individuals and patients with viral encephalitis will be a powerful tool in dissecting the disease pathogenesis of CNS infections with HSV-1 and other neurotropic viruses.
Collapse
Affiliation(s)
- Fabien G Lafaille
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA
| | - Michael J Ciancanelli
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research , New York, NY , USA
| | - Gregory Smith
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine , Chicago, IL , USA
| | - Luigi Notarangelo
- Division of Immunology, Boston Children's Hospital and Harvard Medical School , Boston, MA , USA
| | - Jean-Laurent Casanova
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA ; Howard Hughes Medical Institute , New York, NY , USA ; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children , Paris , France ; Imagine Institute, Paris Descartes University , Paris , France ; Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children , Paris , France
| | - Shen-Ying Zhang
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA ; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children , Paris , France ; Imagine Institute, Paris Descartes University , Paris , France
| |
Collapse
|
69
|
Involvement of TLR2 and TLR9 in the anti-inflammatory effects of chlorogenic acid in HSV-1-infected microglia. Life Sci 2015; 127:12-8. [PMID: 25744394 DOI: 10.1016/j.lfs.2015.01.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/29/2015] [Accepted: 01/30/2015] [Indexed: 12/23/2022]
|
70
|
Royer DJ, Cohen A, Carr D. The Current State of Vaccine Development for Ocular HSV-1 Infection. EXPERT REVIEW OF OPHTHALMOLOGY 2015; 10:113-126. [PMID: 25983856 DOI: 10.1586/17469899.2015.1004315] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
HSV-1 continues to be the leading cause of infectious corneal blindness. Clinical trials for vaccines against genital HSV infection have been ongoing for more than three decades. Despite this, no approved vaccine exists, and no formal clinical trials have evaluated the impact of HSV vaccines on eye health. We review here the current state of development for an efficacious HSV-1 vaccine and call for involvement of ophthalmologists and vision researchers.
Collapse
Affiliation(s)
- D J Royer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center
| | - A Cohen
- Ophthalmology, University of Oklahoma Health Sciences Center
| | - Djj Carr
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center ; Ophthalmology, University of Oklahoma Health Sciences Center
| |
Collapse
|
71
|
Sun X, Shi L, Zhang H, Li R, Liang R, Liu Z. Effects of Toll-like receptor 3 on herpes simplex virus type-1-infected mouse neural stem cells. Can J Microbiol 2014; 61:201-8. [PMID: 25633775 DOI: 10.1139/cjm-2014-0540] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we aimed to investigate the effect of herpes simplex virus type-1 (HSV-1) infection on the phosphorylation of interferon regulatory factor 3 (IRF3) and the expression of interferon-β (IFN-β), as well as to clarify the functions of toll-like receptor 3 (TLR3) in mouse neural stem cells (NSCs) infected with HSV-1. In HSV-1-infected cultured NSCs, immunofluorescence, reverse transcription - polymerase chain reaction, Western blot, and ELISA were performed to reveal the expression patterns of TLR3, IRF3, and IFN-β. Then, lentivirus-mediated RNA interference (RNAi) was used to block the expression of TLR3, and its effect on host resistance to HSV-1 infection was investigated. Under uninfected conditions, NSCs expressed TLR3 and phosphorylated IRF3, but after infection, the expression level of TLR3 was upregulated and the phosphorylation level of IRF3 in the nucleus was significantly enhanced, while IFN-β was also expressed. After TLR3 expression was blocked by lentivirus-mediated RNAi, IRF3 phosphorylation and IFN-β expression were downregulated. Therefore, HSV-1 upregulated the expression of TLR3 in NSCs and promoted nuclear translocation after IRF3 was phosphorylated to induce IFN-β expression. TLR3 exhibited an anti-HSV-1 infection capacity via innate immune functions.
Collapse
Affiliation(s)
- Xiuning Sun
- Department of Parasitology, Weifang Medical University, Shandong 261053, People's Republic of China
| | | | | | | | | | | |
Collapse
|
72
|
Corilagin Protects Against HSV1 Encephalitis Through Inhibiting the TLR2 Signaling Pathways In Vivo and In Vitro. Mol Neurobiol 2014; 52:1547-1560. [DOI: 10.1007/s12035-014-8947-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022]
|
73
|
Libbey JE, Fujinami RS. Adaptive immune response to viral infections in the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2014. [PMID: 25015488 DOI: 10.1016/b978-0-444-0.00010-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Jane E Libbey
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
74
|
Libbey JE, Fujinami RS. Adaptive immune response to viral infections in the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2014; 123:225-47. [PMID: 25015488 DOI: 10.1016/b978-0-444-53488-0.00010-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jane E Libbey
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
75
|
Rajasekharan S, Rana J, Gulati S, Gupta V, Gupta S. Neuroinvasion by Chandipura virus. Acta Trop 2014; 135:122-6. [PMID: 24713200 DOI: 10.1016/j.actatropica.2014.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/14/2014] [Accepted: 03/26/2014] [Indexed: 01/13/2023]
Abstract
Chandipura virus (CHPV) is an arthropod borne rhabdovirus associated with acute encephalitis in children below the age of 15 years in the tropical states of India. Although the entry of the virus into the nervous system is among the crucial events in the pathogenesis of CHPV, the exact mechanism allowing CHPV to invade the central nervous system (CNS) is currently poorly understood. In the present review, based on the knowledge of host interactors previously predicted for CHPV, along with the support from experimental data available for other encephalitic viruses, the authors have speculated the various plausible modes by which CHPV could surpass the blood-brain barrier and invade the CNS to cause encephalitis whilst evading the host immune surveillance. Collectively, this review provides a conservative set of potential interactions that can be employed for future experimental validation with a view to better understand the neuropathogenesis of CHPV.
Collapse
Affiliation(s)
- Sreejith Rajasekharan
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Jyoti Rana
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Sahil Gulati
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India
| | - Vandana Gupta
- Department of Microbiology, Ram Lal Anand College, University of Delhi South Campus (UDSC), Benito Juarez Marg, New Delhi 110021, India
| | - Sanjay Gupta
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201 307, India.
| |
Collapse
|
76
|
Zolini GP, Lima GK, Lucinda N, Silva MA, Dias MF, Pessoa NL, Coura BP, Cartelle CT, Arantes RME, Kroon EG, Campos MA. Defense against HSV-1 in a murine model is mediated by iNOS and orchestrated by the activation of TLR2 and TLR9 in trigeminal ganglia. J Neuroinflammation 2014; 11:20. [PMID: 24479442 PMCID: PMC3922087 DOI: 10.1186/1742-2094-11-20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 01/14/2014] [Indexed: 12/24/2022] Open
Abstract
Background Herpes simplex 1 (HSV-1) causes various human clinical manifestations, ranging from simple cold sores to encephalitis. Innate immune cells recognize pathogens through Toll-like receptors (TLRs), thus initiating the immune response. Previously, we demonstrated that the immune response against HSV-1 is dependent on TLR2 and TLR9 expression and on IFN gamma production in the trigeminal ganglia (TG) of infected mice. In this work, we further investigated the cells, molecules, and mechanisms of HSV-1 infection control, especially those that are TLR-dependent. Methods C57BL/6 wild-type (WT), TLR2−/−, TLR9−/−, and TLR2/9−/− mice were intranasally infected with HSV-1. On the viral peak day, the TG and brains were collected from mice and TLR expression was measured in the TG and brain and inducible nitric oxide synthase (iNOS) expression was measured in the TG by real-time PCR. Immunofluorescence assays were performed in mice TG to detect iNOS production by F4/80+ cells. Intraperitoneal macrophages nitric oxide (NO) production was evaluated by the Griess assay. WT, CD8−/−, RAG−/−, and iNOS−/− mice were intranasally infected in a survival assay, and their cytokine expression was measured in the TG by real-time PCR. Results Infected WT mice exhibited significantly increased TLR expression, compared with their respective controls, in the TG but not in the brain. TLR-deficient mice had moderately increased TLR expression in the TG and brain in compare with the non-infected animals. iNOS expression in the WT infected mice TG was higher than in the other groups with increased production by macrophages in the WT infected mice, which did not occur in the TLR2/9−/− mice. Additionally, the intraperitoneal macrophages of the WT mice had a higher production of NO compared with those of the TLR-deficient mice. The CD8−/−, RAG−/−, and iNOS−/− mice had 100% mortality after the HSV-1 infection compared with 10% of the WT mice. Cytokines were overexpressed in the iNOS−/− infected mice, while the RAG−/− mice were nearly unresponsive to the virus. Conclusion TLRs efficiently orchestrate the innate immune cells, eliciting macrophage response (with NO production by the macrophages), thereby controlling the HSV-1 infection through the immune response in the TG of mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Marco Antônio Campos
- Laboratório de Imunopatologia, Imunologia de Doenças Virais, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Fiocruz, Av, Augusto de Lima 1715, Belo Horizonte, Minas Gerais 30190-002, Brazil.
| |
Collapse
|
77
|
The modulation of phosphatase expression impacts the proliferation efficiency of HSV-1 in infected astrocytes. PLoS One 2013; 8:e79648. [PMID: 24260274 PMCID: PMC3829861 DOI: 10.1371/journal.pone.0079648] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/02/2013] [Indexed: 01/06/2023] Open
Abstract
Herpes Simplex Virus 1 (HSV-1) is a major pathogen that causes human neurological diseases, including herpes simplex encephalitis (HSE). Previous studies have shown that astrocytes are involved in HSV-1 systemic pathogenesis in the central nervous system (CNS), although the mechanism remains unclear. In this study, a high-throughput RNAi library screening method was used to analyze the effect of host phosphatase gene regulation on HSV-1 replication using Macaca mulatta primary astrocytes in an in vitro culture system. The results showed that the downregulation of five phosphatase genes (PNKP, SNAP23, PTPRU, LOC714621 and PPM1M) significantly inhibited HSV-1 infection, suggesting that these phosphatases were needed in HSV-1 replication in rhesus astrocytes. Although statistically significant, the effect of downregulation of these phosphatases on HSV-1 replication in a human astrocytoma cell line appears to be more limited. Our results suggest that the phosphatase genes in astrocytes may regulate the immunological and pathological reactions caused by HSV-1 CNS infection through the regulation of HSV-1 replication or of multiple signal transduction pathways.
Collapse
|
78
|
Abstract
Viral encephalitis causes an altered level of consciousness, which may be associated with fever, seizures, focal deficits, CSF pleocytosis, and abnormal neuroimaging. Potential pathogens include HSV, VZV, enterovirus, and in some regions, arboviruses. Autoimmune (eg, anti-NMDA receptor) and paraneoplastic encephalitis are responsible for some cases where no pathogen is identified. Indications for ICU admission include coma, status epilepticus and respiratory failure. Timely initiation of anti-viral therapy is crucial while relevant molecular and serological test results are being performed. Supportive care should be directed at the prevention and treatment of cerebral edema and other physiological derangements which may contribute to secondary neurological injury.
Collapse
Affiliation(s)
- Andreas H Kramer
- Department of Critical Care Medicine and Clinical Neurosciences, Foothills Medical Center, McCaig Tower, 3134 Hospital Drive NW, Calgary, AB T2N 2T9, Canada.
| |
Collapse
|
79
|
Caignard G, Leiva-Torres GA, Leney-Greene M, Charbonneau B, Dumaine A, Fodil-Cornu N, Pyzik M, Cingolani P, Schwartzentruber J, Dupaul-Chicoine J, Guo H, Saleh M, Veillette A, Lathrop M, Blanchette M, Majewski J, Pearson A, Vidal SM. Genome-wide mouse mutagenesis reveals CD45-mediated T cell function as critical in protective immunity to HSV-1. PLoS Pathog 2013; 9:e1003637. [PMID: 24068938 PMCID: PMC3771889 DOI: 10.1371/journal.ppat.1003637] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/24/2013] [Indexed: 01/15/2023] Open
Abstract
Herpes simplex encephalitis (HSE) is a lethal neurological disease resulting from infection with Herpes Simplex Virus 1 (HSV-1). Loss-of-function mutations in the UNC93B1, TLR3, TRIF, TRAF3, and TBK1 genes have been associated with a human genetic predisposition to HSE, demonstrating the UNC93B-TLR3-type I IFN pathway as critical in protective immunity to HSV-1. However, the TLR3, UNC93B1, and TRIF mutations exhibit incomplete penetrance and represent only a minority of HSE cases, perhaps reflecting the effects of additional host genetic factors. In order to identify new host genes, proteins and signaling pathways involved in HSV-1 and HSE susceptibility, we have implemented the first genome-wide mutagenesis screen in an in vivo HSV-1 infectious model. One pedigree (named P43) segregated a susceptible trait with a fully penetrant phenotype. Genetic mapping and whole exome sequencing led to the identification of the causative nonsense mutation L3X in the Receptor-type tyrosine-protein phosphatase C gene (Ptprc(L3X)), which encodes for the tyrosine phosphatase CD45. Expression of MCP1, IL-6, MMP3, MMP8, and the ICP4 viral gene were significantly increased in the brain stems of infected Ptprc(L3X) mice accounting for hyper-inflammation and pathological damages caused by viral replication. Ptprc(L3X) mutation drastically affects the early stages of thymocytes development but also the final stage of B cell maturation. Transfer of total splenocytes from heterozygous littermates into Ptprc(L3X) mice resulted in a complete HSV-1 protective effect. Furthermore, T cells were the only cell population to fully restore resistance to HSV-1 in the mutants, an effect that required both the CD4⁺ and CD8⁺ T cells and could be attributed to function of CD4⁺ T helper 1 (Th1) cells in CD8⁺ T cell recruitment to the site of infection. Altogether, these results revealed the CD45-mediated T cell function as potentially critical for infection and viral spread to the brain, and also for subsequent HSE development.
Collapse
Affiliation(s)
- Grégory Caignard
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | | | - Michael Leney-Greene
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Benoit Charbonneau
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Anne Dumaine
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Nassima Fodil-Cornu
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Michal Pyzik
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Pablo Cingolani
- School of Computer Science and McGill Centre for Bioinformatics, McGill University, Montréal, Quebec, Canada
| | | | | | - Huaijian Guo
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Quebec, Canada
| | - Maya Saleh
- Departments of Biochemistry and Medicine, McGill University, Montréal, Quebec, Canada
| | - André Veillette
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Quebec, Canada
| | - Marc Lathrop
- McGill University and Genome Québec Innovation Centre, Montréal, Quebec, Canada
| | - Mathieu Blanchette
- School of Computer Science and McGill Centre for Bioinformatics, McGill University, Montréal, Quebec, Canada
| | - Jacek Majewski
- McGill University and Genome Québec Innovation Centre, Montréal, Quebec, Canada
| | - Angela Pearson
- INRS-Institut Armand-Frappier, Université du Québec, Laval, Quebec, Canada
| | - Silvia M. Vidal
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
- * E-mail:
| |
Collapse
|
80
|
Bryant-Hudson K, Conrady CD, Carr DJJ. Type I interferon and lymphangiogenesis in the HSV-1 infected cornea - are they beneficial to the host? Prog Retin Eye Res 2013; 36:281-91. [PMID: 23876483 DOI: 10.1016/j.preteyeres.2013.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 02/06/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) is a highly successful pathogen that can result in significant human morbidity. Within the cornea, it was thought the initial recognition of the pathogen was through Toll-like receptors expressed on/in resident cells that then elicit pro-inflammatory cytokine production, activation of anti-viral pathways, and recruitment of leukocytes. However, our lab has uncovered a novel, TLR-independent innate sensor that supersedes TLR induction of anti-viral pathways following HSV-1 infection. In addition, we have also found HSV-1 induces the genesis of lymphatic vessels into the cornea proper by a mechanism independent of TLRs and unique in the field of neovascularization. This review will focus on these two innate immune events during acute HSV-1 infection of the cornea.
Collapse
Affiliation(s)
- Katie Bryant-Hudson
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | |
Collapse
|
81
|
Prim N, Benito N, Montes G, Pomar V, Molet J, Rabella N. Human herpesvirus 1 meningoencephalitis after trigeminal neuralgia surgery. J Infect 2013; 67:79-81. [PMID: 23068448 DOI: 10.1016/j.jinf.2012.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/29/2012] [Accepted: 10/07/2012] [Indexed: 02/05/2023]
Abstract
We report a case of human herpesvirus 1 (HHV-1) meningoencephalitis in a patient who underwent trigeminal neuralgia surgery. Although this surgery has been reported to increase the risk of mucocutaneous HHV-1 recurrence, to our knowledge, an association between trigeminal surgery and HHV-1 encephalitis has not been previously described.
Collapse
Affiliation(s)
- Núria Prim
- Servei de Microbiologia, Hospital de la Santa Creu i Sant Pau, Sant Quintí 89, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
82
|
Advance in herpes simplex viruses for cancer therapy. SCIENCE CHINA-LIFE SCIENCES 2013; 56:298-305. [PMID: 23564184 DOI: 10.1007/s11427-013-4466-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 02/27/2013] [Indexed: 02/07/2023]
Abstract
Oncolytic virotherapy is an attractive approach that uses live viruses to selectively kill cancer cells. Oncolytic viruses can be genetically engineered to induce cell lyses through virus replication and cytotoxic protein expression. Herpes simplex virus (HSV) has become one of the most widely clinically used oncolytic agent. Various types of HSV have been studied in basic or clinical research. Combining oncolytic virotherapy with chemotherapy or radiotherapy generally produces synergic action with unclear molecular mechanisms. Arming HSV with therapeutic transgenes is a promising strategy and can be used to complement conventional therapies. As an efficient gene delivery system, HSV has been successfully used to deliver various immunomodulatory molecules. Arming HSV with therapeutic genes merits further investigation for potential clinical application.
Collapse
|
83
|
Ramakrishna C, Openshaw H, Cantin EM. The case for immunomodulatory approaches in treating HSV encephalitis. Future Virol 2013; 8:259-272. [PMID: 23956785 DOI: 10.2217/fvl.12.138] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HSV encephalitis (HSE) is the most prevalent sporadic viral encephalitis. Although safe and effective antiviral therapies and greatly improved noninvasive diagnostic procedures have significantly improved outcomes, mortality (~20%) and debilitating neurological sequelae in survivors remain unacceptably high. An encouraging new development is that the focus is now shifting away from the virus exclusively, to include consideration of the host immune response to infection in the pathology underlying development of HSE. In this article, the authors discuss results from recent studies in experimental mouse models, as well as clinical reports that demonstrate a role for exaggerated host inflammatory responses in the brain in the development of HSE that is motivating researchers and clinicians to consider new therapeutic approaches for treating HSE. The authors also discuss results from a few studies that have shown that immunomodulatory drugs can be highly protective against HSE, which supports a role for deleterious host inflammatory responses in HSE. The impressive outcomes of some immunomodulatory approaches in mouse models of HSE emphasize the urgent need for clinical trials to rigorously evaluate combination antiviral and immunomodulatory therapy in comparison with standard antiviral therapy for treatment of HSE, and support for such an initiative is gaining momentum.
Collapse
Affiliation(s)
- Chandran Ramakrishna
- Department of Virology, Beckman Research Institute of City of Hope; Duarte, CA 91010-3000, USA
| | | | | |
Collapse
|
84
|
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
|
85
|
Directional spread of alphaherpesviruses in the nervous system. Viruses 2013; 5:678-707. [PMID: 23435239 PMCID: PMC3640521 DOI: 10.3390/v5020678] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 02/04/2013] [Accepted: 02/05/2013] [Indexed: 12/30/2022] Open
Abstract
Alphaherpesviruses are pathogens that invade the nervous systems of their mammalian hosts. Directional spread of infection in the nervous system is a key component of the viral lifecycle and is critical for the onset of alphaherpesvirus-related diseases. Many alphaherpesvirus infections originate at peripheral sites, such as epithelial tissues, and then enter neurons of the peripheral nervous system (PNS), where lifelong latency is established. Following reactivation from latency and assembly of new viral particles, the infection typically spreads back out towards the periphery. These spread events result in the characteristic lesions (cold sores) commonly associated with herpes simplex virus (HSV) and herpes zoster (shingles) associated with varicella zoster virus (VZV). Occasionally, the infection spreads transsynaptically from the PNS into higher order neurons of the central nervous system (CNS). Spread of infection into the CNS, while rarer in natural hosts, often results in severe consequences, including death. In this review, we discuss the viral and cellular mechanisms that govern directional spread of infection in the nervous system. We focus on the molecular events that mediate long distance directional transport of viral particles in neurons during entry and egress.
Collapse
|
86
|
Conrady CD, Zheng M, van Rooijen N, Drevets DA, Royer D, Alleman A, Carr DJJ. Microglia and a functional type I IFN pathway are required to counter HSV-1-driven brain lateral ventricle enlargement and encephalitis. THE JOURNAL OF IMMUNOLOGY 2013; 190:2807-17. [PMID: 23382563 DOI: 10.4049/jimmunol.1203265] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
HSV-1 is the leading cause of sporadic viral encephalitis, with mortality rates approaching 30% despite treatment with the antiviral drug of choice, acyclovir. Permanent neurologic deficits are common in patients that survive, but the mechanism leading to this pathology is poorly understood, impeding clinical advancements in treatment to reduce CNS morbidity. Using magnetic resonance imaging and type I IFN receptor-deficient mouse chimeras, we demonstrate HSV-1 gains access to the murine brain stem and subsequently brain ependymal cells, leading to enlargement of the cerebral lateral ventricle and infection of the brain parenchyma. A similar enlargement in the lateral ventricles is found in a subpopulation of herpes simplex encephalitic patients. Associated with encephalitis is an increase in CXCL1 and CXCL10 levels in the cerebral spinal fluid, TNF-α expression in the ependymal region, and the influx of neutrophils of encephalitic mouse brains. Reduction in lateral ventricle enlargement using anti-secretory factor peptide 16 reduces mortality significantly in HSV-1-infected mice without any effect on expression of inflammatory mediators, infiltration of leukocytes, or changes in viral titer. Microglial cells but not infiltrating leukocytes or other resident glial cells or neurons are the principal source of resistance in the CNS during the first 5 d postinfection through a Toll/IL-1R domain-containing adapter inducing IFN-β-dependent, type I IFN pathway. Our results implicate lateral ventricle enlargement as a major cause of mortality in mice and speculate such an event transpires in a subpopulation of human HSV encephalitic patients.
Collapse
Affiliation(s)
- Christopher D Conrady
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Conrady CD, Zheng M, Mandal NA, van Rooijen N, Carr DJ. IFN-α-driven CCL2 production recruits inflammatory monocytes to infection site in mice. Mucosal Immunol 2013; 6:45-55. [PMID: 22692455 PMCID: PMC3449026 DOI: 10.1038/mi.2012.46] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) is the leading cause of corneal blindness in the developed world due to reactivation of infectious virus and the subsequent immune response. The innate response that facilitates viral control in the cornea is currently unknown. In the present study using a mouse chimera model, we found that a bone marrow component is crucial in inhibiting viral replication and identified inflammatory monocytes (F4/80(+) Gr1(+)) as the responsible cell. CCL2 was critical for recruiting inflammatory monocytes, and a loss of this chemokine in CCL2(-/-) mice resulted in a loss of viral containment and inflammatory monocyte recruitment. To confirm these results, clodronate depletion of inflammatory monocytes resulted in elevated viral titers. Furthermore, siRNA targeting the innate sensor p204/IFI-16 resulted in a loss of CCL2 production. In conclusion, CCL2 expression driven by IFI-16 recognition of HSV-1 facilitates the recruitment of inflammatory monocytes into the cornea proper to control viral replication.
Collapse
Affiliation(s)
- Christopher D. Conrady
- Department of Microbiology, Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Min Zheng
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nawajes A. Mandal
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Niko van Rooijen
- Department of Molecular Cell Biology and Immunology, VU Medical Center, Amsterdam, The Netherlands
| | - Daniel J.J. Carr
- Department of Microbiology, Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Corresponding author: Daniel J.J. Carr, Ph.D., Department of Ophthalmology, DMEI #415, OUHSC, 608 Stanton L. Young Blvd., Oklahoma City, OK. 73104 USA; telephone: 405-271-8784;
| |
Collapse
|
88
|
De Chiara G, Marcocci ME, Sgarbanti R, Civitelli L, Ripoli C, Piacentini R, Garaci E, Grassi C, Palamara AT. Infectious agents and neurodegeneration. Mol Neurobiol 2012; 46:614-38. [PMID: 22899188 PMCID: PMC3496540 DOI: 10.1007/s12035-012-8320-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/31/2012] [Indexed: 12/19/2022]
Abstract
A growing body of epidemiologic and experimental data point to chronic bacterial and viral infections as possible risk factors for neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis. Infections of the central nervous system, especially those characterized by a chronic progressive course, may produce multiple damage in infected and neighbouring cells. The activation of inflammatory processes and host immune responses cause chronic damage resulting in alterations of neuronal function and viability, but different pathogens can also directly trigger neurotoxic pathways. Indeed, viral and microbial agents have been reported to produce molecular hallmarks of neurodegeneration, such as the production and deposit of misfolded protein aggregates, oxidative stress, deficient autophagic processes, synaptopathies and neuronal death. These effects may act in synergy with other recognized risk factors, such as aging, concomitant metabolic diseases and the host’s specific genetic signature. This review will focus on the contribution given to neurodegeneration by herpes simplex type-1, human immunodeficiency and influenza viruses, and by Chlamydia pneumoniae.
Collapse
Affiliation(s)
- Giovanna De Chiara
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
The CNS, which consists of the brain and spinal cord, is continuously monitored by resident microglia and blood-borne immune cells such as macrophages, dendritic cells and T cells to detect for damaging agents that would disrupt homeostasis and optimal functioning of these vital organs. Further, the CNS must balance between vigilantly detecting for potentially harmful factors and resolving any immunological responses that in themselves can create damage if left unabated. We discuss the physiological roles of the immune sentinels that patrol the CNS, the molecular markers that underlie their surveillance duties, and the consequences of interrupting their functions following injury and infection by viruses such as JC virus, human immunodeficiency virus, herpes simplex virus and West Nile virus.
Collapse
|
90
|
Furr SR, Marriott I. Viral CNS infections: role of glial pattern recognition receptors in neuroinflammation. Front Microbiol 2012; 3:201. [PMID: 22723794 PMCID: PMC3379540 DOI: 10.3389/fmicb.2012.00201] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/15/2012] [Indexed: 12/13/2022] Open
Abstract
Viruses are the major causative agents of central nervous system (CNS) infection worldwide. RNA and DNA viruses trigger broad activation of glial cells including microglia and astrocytes, eliciting the release of an array of mediators that can promote innate and adaptive immune responses. Such responses can limit viral replication and dissemination leading to infection resolution. However, a defining feature of viral CNS infection is the rapid onset of severe neuroinflammation and overzealous glial responses are associated with significant neurological damage or even death. The mechanisms by which microglia and astrocytes perceive neurotropic RNA and DNA viruses are only now becoming apparent with the discovery of a variety of cell surface and cytosolic molecules that serve as sensors for viral components. In this review we discuss the role played by members of the Toll-like family of pattern recognition receptors (PRRs) in the inflammatory responses of glial cells to the principle causative agents of viral encephalitis. Importantly, we also describe the evidence for the involvement of a number of newly described intracellular PRRs, including retinoic acid-inducible gene I and DNA-dependent activator of IFN regulatory factors, that are thought to function as intracellular sensors of RNA and DNA viruses, respectively. Finally, we explore the possibility that cross-talk exists between these disparate viral sensors and their signaling pathways, and describe how glial cytosolic and cell surface/endosomal PRRs could act in a cooperative manner to promote the fulminant inflammation associated with acute neurotropic viral infection.
Collapse
Affiliation(s)
| | - Ian Marriott
- Department of Biology, University of North Carolina at Charlotte,Charlotte, NC, USA
| |
Collapse
|
91
|
Conrady CD, Zheng M, Stone DU, Carr DJJ. CD8+ T cells suppress viral replication in the cornea but contribute to VEGF-C-induced lymphatic vessel genesis. THE JOURNAL OF IMMUNOLOGY 2012; 189:425-32. [PMID: 22649204 DOI: 10.4049/jimmunol.1200063] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
HSV-1 is the leading cause of infectious corneal blindness in the industrialized world. CD4(+) T cells are thought to be the major leukocyte population mediating immunity to HSV-1 in the cornea as well as the likely source of immunopathology that reduces visual acuity. However, the role of CD8(+) T cells in immune surveillance of the cornea is unclear. Thus, we sought to evaluate the role of CD8(+) T cells in ocular immunity using transgenic mice in which >98% of CD8(+) T cells are specific for the immunodominant HSV-1 epitope (gBT-I.1). We found a significant reduction in virus, elevation in HSV-specific CD8(+) T cell influx, and more CD8(+) T cells expressing CXCR3 in the cornea of transgenic mice compared with those in the cornea of wild-type controls yet similar acute corneal pathology. However, by day 30 postinfection, wild-type mice had drastically more blood and lymphatic vessel projections into the cornea compared with gBT-I.1 mice, in which only lymphatic vessel growth in response to VEGF-C could be appreciated. Taken together, these results show that CD8(+) T cells are required to eliminate virus more efficiently from the cornea but play a minimal role in immunopathology as a source of VEGF-C.
Collapse
Affiliation(s)
- Christopher D Conrady
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
92
|
Sellner J, Trinka E. Seizures and epilepsy in herpes simplex virus encephalitis: current concepts and future directions of pathogenesis and management. J Neurol 2012; 259:2019-30. [DOI: 10.1007/s00415-012-6494-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 12/30/2022]
|
93
|
Zirger JM, Puntel M, Bergeron J, Wibowo M, Moridzadeh R, Bondale N, Barcia C, Kroeger KM, Liu C, Castro MG, Lowenstein PR. Immune-mediated loss of transgene expression from virally transduced brain cells is irreversible, mediated by IFNγ, perforin, and TNFα, and due to the elimination of transduced cells. Mol Ther 2012; 20:808-19. [PMID: 22233583 PMCID: PMC3321600 DOI: 10.1038/mt.2011.243] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 10/13/2011] [Indexed: 01/12/2023] Open
Abstract
The adaptive immune response to viral vectors reduces vector-mediated transgene expression from the brain. It is unknown, however, whether this loss is caused by functional downregulation of transgene expression or death of transduced cells. Herein, we demonstrate that during the elimination of transgene expression, the brain becomes infiltrated with CD4(+) and CD8(+) T cells and that these T cells are necessary for transgene elimination. Further, the loss of transgene-expressing brain cells fails to occur in the absence of IFNγ, perforin, and TNFα receptor. Two methods to induce severe immune suppression in immunized animals also fail to restitute transgene expression, demonstrating the irreversibility of this process. The need for cytotoxic molecules and the irreversibility of the reduction in transgene expression suggested to us that elimination of transduced cells is responsible for the loss of transgene expression. A new experimental paradigm that discriminates between downregulation of transgene expression and the elimination of transduced cells demonstrates that transduced cells are lost from the brain upon the induction of a specific antiviral immune response. We conclude that the anti-adenoviral immune response reduces transgene expression in the brain through loss of transduced cells.
Collapse
Affiliation(s)
- Jeffrey M Zirger
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mariana Puntel
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Josee Bergeron
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mia Wibowo
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rameen Moridzadeh
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Niyati Bondale
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Carlos Barcia
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kurt M Kroeger
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Deceased
| | - Chunyan Liu
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Maria G Castro
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Current address: Department of Neurosurgery, The University of Michigan, Medical School, Ann Arbor, Michigan, USA
- Current address: Department of Cell and Developmental Biology, The University of Michigan, Medical School, Ann Arbor, Michigan, USA
| | - Pedro R Lowenstein
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Current address: Department of Neurosurgery, The University of Michigan, Medical School, Ann Arbor, Michigan, USA
- Current address: Department of Cell and Developmental Biology, The University of Michigan, Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
94
|
Abstract
Toll-like receptors (TLRs) are innate sentinels required for clearance of bacterial and fungal infections of the cornea, but their role in viral immunity is currently unknown. We report that TLR signaling is expendable in herpes simplex virus (HSV)-1 containment as depicted by plaque assays of knockout mice (MyD88(-/-), Trif(-/-) and MyD88(-/-) Trif(-/-) double knockout) resembling wild-type controls. To identify the key sentinel in viral recognition of the cornea, in vivo knockdown of the DNA sensor IFI-16/p204 in the corneal epithelium was performed and resulted in a loss of IFN-regulatory factor-3 (IRF-3) nuclear translocation, interferon-α production, and viral containment. The sensor seems to have a similar function in other HSV clinically relevant sites such as the vaginal mucosa in which a loss of p204/IFI-16 results in significantly more HSV-2 shedding. Thus, we have identified an IRF-3-dependent, IRF-7- and TLR-independent innate sensor responsible for HSV containment at the site of acute infection.
Collapse
|
95
|
Puchhammer-Stöckl E, Aberle SW, Heinzl H. Association of age and gender with alphaherpesvirus infections of the central nervous system in the immunocompetent host. J Clin Virol 2012; 53:356-9. [PMID: 22265826 DOI: 10.1016/j.jcv.2011.12.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/22/2011] [Accepted: 12/08/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND The alphaherpesviruses Varicella-zoster virus (VZV) and human herpes simplex virus types 1 (HSV-1) and 2 (HSV-2) can cause severe infections of the central nervous system (CNS). OBJECTIVES To analyze whether age and gender of immunocompetent individuals are associated with the incidence of herpesvirus CNS diseases. STUDY DESIGN A total of 241 patients with virologically confirmed HSV-1, HSV-2 or VZV-infection of the CNS (excluding neonatal infection and varicella), diagnosed at the Department of Virology, Medical University Vienna, from 2001 to 2009 were analyzed retrospectively. The relative incidence of disease was evaluated statistically with respect to gender and age. RESULTS The relative incidence of VZV CNS disease increased with age (p<0.0001), and nonlinear age dependences were observed for HSV-1 (p=0.005) and HSV-2 disease (p=0.002). These effects were influenced significantly by the patient's gender in VZV (p=0.0003) and HSV-1 disease (p=0.008). Overall, 50.7% of VZV infections in males, but only 23.5% of those in females, occurred before age 45, and 28.9% of HSV-1 infections in males and 8.8% of those in females occurred before age 30. Women represented 71.9% of HSV-2 CNS infections (p=0.02). CONCLUSIONS The patient's gender is clearly associated with the incidence of CNS disease caused by VZV, HSV-1 and HSV-2, and its influence varies over one's lifetime.
Collapse
|
96
|
Kwiatkowski A, Gallois J, Bilbault N, Calais G, Mackowiak A, Hautecoeur P. Herpes encephalitis during natalizumab treatment in multiple sclerosis. Mult Scler 2011; 18:909-11. [DOI: 10.1177/1352458511428082] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this case report we describe the first non-fatal herpes simplex virus encephalitis (HSE) case with natalizumab for multiple sclerosis (MS). A 36-year-old woman, previously treated with immunomodulatory and immunosuppressive drugs for MS, developed acute encephalitis after 6 monthly natalizumab perfusions. Brain imaging demonstrated suggestive bi-temporal lesions. Herpes simplex virus type-1 DNA was detected in cerebrospinal fluid. The patient improved gradually after a 21-day course of intravenous acyclovir, but neuropsychiatric changes remained 5 months later. Our non-fatal case of HSE and other reported cases of herpes infections provide evidence of an increased risk with natalizumab and point to the need for clinicians to maintain awareness.
Collapse
Affiliation(s)
- A Kwiatkowski
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| | - J Gallois
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| | - N Bilbault
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| | - G Calais
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| | - A Mackowiak
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| | - P Hautecoeur
- PRES Lille Nord de France, Université Catholique de Lille, Groupe Hospitalier de l’Institut Catholique de Lille / Faculté Libre de Médecine, Clinique de Neurologie, Lille, France
| |
Collapse
|
97
|
Functional genomics reveals an essential and specific role for Stat1 in protection of the central nervous system following herpes simplex virus corneal infection. J Virol 2011; 85:12972-81. [PMID: 21994441 DOI: 10.1128/jvi.06032-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Innate immune deficiencies result in a spectrum of severe clinical outcomes following infection. In particular, there is a strong association between loss of the signal transducer and activator of transcription (Stat) pathway, breach of the blood-brain barrier (BBB), and virus-induced neuropathology. The gene signatures that characterize resistance, disease, and mortality in the virus-infected nervous system have not been defined. Herpes simplex virus type 1 (HSV-1) is commonly associated with encephalitis in humans, and humans and mice lacking Stat1 display increased susceptibility to HSV central nervous system (CNS) infections. In this study, two HSV-1 strains were used, KOS (wild type [WT]), and Δvhs, an avirulent recombinant lacking the virion host shutoff (vhs) function. In addition, two mouse strains were used: strain 129 (control) and a Stat1-deficient (Stat1(-/-)) strain. Using combinations of these virus and mouse strains, we established a model of infection resulting in three different outcomes: viral clearance without neurological disease (Δvhs infection of control mice), neurological disease followed by viral clearance (Δvhs infection of Stat1(-/-) mice and WT infection of control mice), or neurological disease followed by death (WT infection of Stat1(-/-) mice). Through the use of functional genomics on the infected brain stems, we determined gene signatures that were representative of the three infection outcomes. We demonstrated a pathological signature in the brain stem of Stat1-deficient mice characterized by upregulation of transcripts encoding chemokine receptors, inflammatory markers, neutrophil chemoattractants, leukocyte adhesion proteins, and matrix metalloproteases. Additionally, there was a greater than 100-fold increase in the inflammatory markers interleukin 1β (IL-1β) and IL-6. Consistent with this gene signature, we demonstrated profound CNS inflammation with a concomitant lethal breach of the BBB. Taken together, our results indicated an essential role for normal Stat1-dependent signaling in mediating a nonpathological immune response to viral CNS infection.
Collapse
|
98
|
Cohen M, Braun E, Tsalenchuck Y, Panet A, Steiner I. Restrictions that control herpes simplex virus type 1 infection in mouse brain ex vivo. J Gen Virol 2011; 92:2383-2393. [DOI: 10.1099/vir.0.031013-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Elucidating the cellular and molecular factors governing herpes simplex virus type 1 (HSV-1) neurotropism is a prerequisite for understanding HSV-1 encephalitis and for targeting HSV-1-derived vectors for gene transfer to the brain. Earlier we had described an ex vivo system of mouse brain slices and demonstrated a selective and unique infection pattern, mostly around the ventricles. Here, we examined tissue factors controlling HSV-1 infection of brain slices. We demonstrated that heparan sulphate, while an important factor, does not determine the infection pattern. Hyaluronic acid, but not collagen, appears to enhance HSV-1 brain infection. To investigate whether tissue distribution of viral receptors determines the infection pattern, we examined transcription of herpes virus entry mediator and nectin-1 receptor genes in infected and uninfected brain regions. Both the infected and the uninfected regions express the receptors. We also explored the influence of intra-cellular factors. HSV-1 does not preferentially infect proliferating cells in the brain slices, despite its predilection to the ventricular zones. To delineate the step at which the HSV-1 infection cascade is restricted, mRNA was isolated following tissue infection, and transcription of the immediate-early and late viral genes was evaluated. The results indicated that HSV-1 genes are not expressed in regions that do not express a viral reporter gene. Therefore, we conclude that tissue resistance to infection is associated with a block at or prior to the immediate-early mRNA synthesis. Taken together, using the ex vivo system of organotypic culture we describe here extra-cellular and intra-cellular restriction levels of HSV-1 brain infection.
Collapse
Affiliation(s)
- Meytal Cohen
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Efrat Braun
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yael Tsalenchuck
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amos Panet
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Israel Steiner
- Department of Neurology, Rabin Medical Center, Campus Beilinson, Petach Tikva, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
99
|
Krauthausen M, Ellis SL, Zimmermann J, Sarris M, Wakefield D, Heneka MT, Campbell IL, Müller M. Opposing roles for CXCR3 signaling in central nervous system versus ocular inflammation mediated by the astrocyte-targeted production of IL-12. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2346-59. [PMID: 21925471 DOI: 10.1016/j.ajpath.2011.07.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 06/17/2011] [Accepted: 07/18/2011] [Indexed: 12/26/2022]
Abstract
CXCR3 and its ligands are important for the trafficking of activated CD4(+) T(H)1 T cells, CD8(+) T cells, and natural killer cells during inflammation. Recent functional studies demonstrate a more diverse role of CXCR3 in inflammatory diseases of the central nervous system (CNS). We examined the impact of CXCR3 on a less complex interferon-γ-dependent, type 1 cell-mediated immune response in the CNS, induced in mice by the transgenic production of glial fibrillary acidic protein IL-12 (GF-IL12) by astrocytes and retinal Müller cells. GF-IL12 mice develop ataxia because of severe cerebellar inflammation but have little overt ocular disease. Surprisingly, CXCR3-deficient GF-IL12 mice (GF-IL12/CXCR3KO) have drastically reduced ataxia but developed cataracts, severe ocular inflammation, and eye atrophy. Most GF-IL12/CXCR3KO mice had minimal cerebellar inflammation but severe retinal disorganization, loss of photoreceptors, and lens destruction in the eye. The number of CD3(+), CD11b(+), and natural killer 1.1(+) cells were reduced in the CNS but highly increased in the eyes of GF-IL12/CXCR3KO compared with GF-IL12 mice. High levels of interferon-γ, IL-1, tumor necrosis factor α, CXCL9, CXCL10, and CCL5 were found in GF-IL12 cerebelli and GF-IL12/CXCR3KO eyes. Our findings demonstrate key but paradoxical functions for CXCR3 in IL-12-induced immune disease in the CNS, promoting inflammation in the brain yet restricting it in the eye. We conclude that the function of CXCR3 in cellular immune disease is driven by a common trigger and is controlled by tissue-specific factors.
Collapse
|
100
|
Abstract
PURPOSE OF REVIEW This review describes the pathogenesis, clinical presentation, course, and therapy of herpes simplex encephalitis (HSE), the most fatal viral encephalitis, in which prognosis is dependent on early diagnosis and effective therapy. RECENT FINDINGS Herpes simplex viruses types 1 and 2 (HSV-1 and HSV-2) are human neurotropic viruses that establish latent infection in dorsal-root ganglia for the entire life of the host. From this reservoir, they can reactivate to cause human morbidity and mortality. HSE is one of the most devastating disorders caused by these viruses. The biology of their ability to establish latency, maintain it for the entire life of the host, reactivate, and cause primary and recurrent disease is being studied in animal models and in humans. Of special interest is the question whether HSE is the result of primary infection or is it the outcome of reactivation. The present review covers the biological, medical, and neurological aspects of HSE, focusing among others on recent molecular findings of gene expression during latent infection of HSV-1. SUMMARY Despite accumulating knowledge, there are still several issues regarding both pathogenesis and therapy of HSV-1 that currently defy understanding.
Collapse
|