1
|
Cleaver J, Jeffery K, Klenerman P, Lim M, Handunnetthi L, Irani SR, Handel A. The immunobiology of herpes simplex virus encephalitis and post-viral autoimmunity. Brain 2024; 147:1130-1148. [PMID: 38092513 PMCID: PMC10994539 DOI: 10.1093/brain/awad419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 04/06/2024] Open
Abstract
Herpes simplex virus encephalitis (HSE) is the leading cause of non-epidemic encephalitis in the developed world and, despite antiviral therapy, mortality and morbidity is high. The emergence of post-HSE autoimmune encephalitis reveals a new immunological paradigm in autoantibody-mediated disease. A reductionist evaluation of the immunobiological mechanisms in HSE is crucial to dissect the origins of post-viral autoimmunity and supply rational approaches to the selection of immunotherapeutics. Herein, we review the latest evidence behind the phenotypic progression and underlying immunobiology of HSE including the cytokine/chemokine environment, the role of pathogen-recognition receptors, T- and B-cell immunity and relevant inborn errors of immunity. Second, we provide a contemporary review of published patients with post-HSE autoimmune encephalitis from a combined cohort of 110 patients. Third, we integrate novel mechanisms of autoimmunization in deep cervical lymph nodes to explore hypotheses around post-HSE autoimmune encephalitis and challenge these against mechanisms of molecular mimicry and others. Finally, we explore translational concepts where neuroglial surface autoantibodies have been observed with other neuroinfectious diseases and those that generate brain damage including traumatic brain injury, ischaemic stroke and neurodegenerative disease. Overall, the clinical and immunological landscape of HSE is an important and evolving field, from which precision immunotherapeutics could soon emerge.
Collapse
Affiliation(s)
- Jonathan Cleaver
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - Katie Jeffery
- Department of Microbiology, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, OX1 3SY, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Ming Lim
- Children’s Neurosciences, Evelina London Children’s Hospital at Guy’s and St Thomas’ NHS Foundation Trust, London, SE1 7EH, UK
- Department Women and Children’s Health, School of Life Course Sciences, King’s College London, London, WC2R 2LS, UK
| | - Lahiru Handunnetthi
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - Adam Handel
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK
| |
Collapse
|
2
|
Zhou L, Cheng A, Wang M, Wu Y, Yang Q, Tian B, Ou X, Sun D, Zhang S, Mao S, Zhao XX, Huang J, Gao Q, Zhu D, Jia R, Liu M, Chen S. Mechanism of herpesvirus protein kinase UL13 in immune escape and viral replication. Front Immunol 2022; 13:1088690. [PMID: 36531988 PMCID: PMC9749954 DOI: 10.3389/fimmu.2022.1088690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Upon infection, the herpes viruses create a cellular environment suitable for survival, but innate immunity plays a vital role in cellular resistance to viral infection. The UL13 protein of herpesviruses is conserved among all herpesviruses and is a serine/threonine protein kinase, which plays a vital role in escaping innate immunity and promoting viral replication. On the one hand, it can target various immune signaling pathways in vivo, such as the cGAS-STING pathway and the NF-κB pathway. On the other hand, it phosphorylates regulatory many cellular and viral proteins for promoting the lytic cycle. This paper reviews the research progress of the conserved herpesvirus protein kinase UL13 in immune escape and viral replication to provide a basis for elucidating the pathogenic mechanism of herpesviruses, as well as providing insights into the potential means of immune escape and viral replication of other herpesviruses that have not yet resolved the function of it.
Collapse
Affiliation(s)
- Lin Zhou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,*Correspondence: Mingshu Wang,
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bin Tian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xumin Ou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Di Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Sai Mao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xin-Xin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Juan Huang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qun Gao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Picard K, St-Pierre MK, Vecchiarelli HA, Bordeleau M, Tremblay MÈ. Neuroendocrine, neuroinflammatory and pathological outcomes of chronic stress: A story of microglial remodeling. Neurochem Int 2021; 145:104987. [PMID: 33587954 DOI: 10.1016/j.neuint.2021.104987] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident macrophage cells of the central nervous system (CNS), are involved in a myriad of processes required to maintain CNS homeostasis. These cells are dynamic and can adapt their phenotype and functions to the physiological needs of the organism. Microglia rapidly respond to changes occurring in their microenvironment, such as the ones taking place during stress. While stress can be beneficial for the organism to adapt to a situation, it can become highly detrimental when it turns chronic. Microglial response to prolonged stress may lead to an alteration of their beneficial physiological functions, becoming either maladaptive or pro-inflammatory. In this review, we aim to summarize the effects of chronic stress exerted on microglia through the neuroendocrine system and inflammation at adulthood. We also discuss how these effects of chronic stress could contribute to microglial involvement in neuropsychiatric and sleep disorders, as well as neurodegenerative diseases.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | | | - Maude Bordeleau
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Corbett RJ, Luttman AM, Wurtz KE, Siegford JM, Raney NE, Ford LM, Ernst CW. Weaning Induces Stress-Dependent DNA Methylation and Transcriptional Changes in Piglet PBMCs. Front Genet 2021; 12:633564. [PMID: 33613645 PMCID: PMC7893110 DOI: 10.3389/fgene.2021.633564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/14/2021] [Indexed: 11/19/2022] Open
Abstract
Changes to the epigenome, including those to DNA methylation, have been proposed as mechanisms by which stress can induce long-term physiological changes in livestock species. Pig weaning is associated with dietary and social stress, both of which elicit an immune response and changes to the hypothalamic–pituitary–adrenal (HPA) axis. While differential methylation following stress has been assessed in model organisms, it remains poorly understood how the pig methylome is altered by stressors in production settings. We quantified changes in CpG methylation and transcript abundance in piglet peripheral blood mononuclear cells (PBMCs) following weaning and also assessed differential patterns in pigs exhibiting high and low stress response as measured by cortisol concentration and lesion scores. Blood was collected from nine gilt piglets 24 h before and after weaning, and whole-genome bisulfite sequencing (WGBS) and RNA-sequencing were performed on six and nine animals, respectively, at both time points. We identified 2,674 differentially methylated regions (DMRs) that were enriched within promoters of genes associated with lymphocyte stimulation and transcriptional regulation. Stress groups displayed unique differential methylation and expression patterns associated with activation and suppression of T cell immunity in low and high stress animals, respectively. Differential methylation was strongly associated with differential expression; specifically, upregulated genes were enriched among hypomethylated genes. We observed post-weaning hypermethylation of the glucocorticoid receptor (NR3C1) promoter and a significant decrease in NR3C1 expression (n = 9, p = 6.1 × 10–3). Our results indicate that weaning-associated stress elicits genome-wide methylation changes associated with differential gene expression, reduced T cell activation, and an altered HPA axis response.
Collapse
Affiliation(s)
- Ryan J Corbett
- Genetics and Genome Sciences Graduate Program, Michigan State University, East Lansing, MI, United States
| | - Andrea M Luttman
- Genetics and Genome Sciences Graduate Program, Michigan State University, East Lansing, MI, United States
| | - Kaitlin E Wurtz
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Janice M Siegford
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Nancy E Raney
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Laura M Ford
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Catherine W Ernst
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
5
|
Wang Y, Jia J, Wang Y, Li F, Song X, Qin S, Wang Z, Kitazato K, Wang Y. Roles of HSV-1 infection-induced microglial immune responses in CNS diseases: friends or foes? Crit Rev Microbiol 2019; 45:581-594. [PMID: 31512533 DOI: 10.1080/1040841x.2019.1660615] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microglia, as brain-resident macrophages, are the first line of defense against brain invading pathogens. Further, their dysfunction has been recognized to be closely associated with mounting CNS diseases. Of note, chronic HSV-1 infection leads to the persistent activation of microglia, which elicit a comprehensive response by generating certain factors with neurotoxic and neuroprotective effects. CNS infection with HSV-1 results in herpes simplex encephalitis and herpes simplex keratitis. Microglial immune response plays a crucial role in the development of these diseases. Moreover, HSV-1 infection is strongly associated with several CNS diseases, especially Alzheimer's disease and schizophrenia. These CNS diseases can be effectively ameliorated by eliciting an appropriate immune response, such as inhibition of microglial proliferation and activation. Therefore, it is crucial to reassess the positive and negative roles of microglia in HSV-1 CNS infection for a more comprehensive and detailed understanding of the relationship between microglia and CNS diseases. Hence, the present review focuses on the dual roles of microglia in mediating HSV-1 CNS infection, as well as on the strategy of targeting microglia to ameliorate CNS diseases. Further research in this field can help comprehensively elucidate the dual role of the microglial immune response in HSV-1 CNS infection, providing a theoretical basis for identifying therapeutic targets against overactive microglia in CNS diseases and HSV-1 infection.
Collapse
Affiliation(s)
- Yiliang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Jiaoyan Jia
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Yun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Feng Li
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Xiaowei Song
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Shurong Qin
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhaoyang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| |
Collapse
|
6
|
To Go or Stay: The Development, Benefit, and Detriment of Tissue-Resident Memory CD8 T Cells during Central Nervous System Viral Infections. Viruses 2019; 11:v11090842. [PMID: 31514273 PMCID: PMC6784233 DOI: 10.3390/v11090842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022] Open
Abstract
CD8 T cells coordinate immune defenses against viral infections of the central nervous system (CNS). Virus-specific CD8 T cells infiltrate the CNS and differentiate into brain-resident memory CD8 T cells (CD8 bTRM). CD8 bTRM are characterized by a lack of recirculation and expression of phenotypes and transcriptomes distinct from other CD8 T cell memory subsets. CD8 bTRM have been shown to provide durable, autonomous protection against viral reinfection and the resurgence of latent viral infections. CD8 T cells have also been implicated in the development of neural damage following viral infection, which demonstrates that the infiltration of CD8 T cells into the brain can also be pathogenic. In this review, we will explore the residency and maintenance requirements for CD8 bTRM and discuss their roles in controlling viral infections of the brain.
Collapse
|
7
|
Pan H, Oliveira B, Saher G, Dere E, Tapken D, Mitjans M, Seidel J, Wesolowski J, Wakhloo D, Klein-Schmidt C, Ronnenberg A, Schwabe K, Trippe R, Mätz-Rensing K, Berghoff S, Al-Krinawe Y, Martens H, Begemann M, Stöcker W, Kaup FJ, Mischke R, Boretius S, Nave KA, Krauss JK, Hollmann M, Lühder F, Ehrenreich H. Uncoupling the widespread occurrence of anti-NMDAR1 autoantibodies from neuropsychiatric disease in a novel autoimmune model. Mol Psychiatry 2019; 24:1489-1501. [PMID: 29426955 PMCID: PMC6756099 DOI: 10.1038/s41380-017-0011-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 02/07/2023]
Abstract
Autoantibodies of the IgG class against N-methyl-D-aspartate-receptor subunit-NR1 (NMDAR1-AB) were considered pathognomonic for anti-NMDAR encephalitis. This view has been challenged by the age-dependent seroprevalence (up to >20%) of functional NMDAR1-AB of all immunoglobulin classes found in >5000 individuals, healthy or affected by different diseases. These findings question a merely encephalitogenic role of NMDAR1-AB. Here, we show that NMDAR1-AB belong to the normal autoimmune repertoire of dogs, cats, rats, mice, baboons, and rhesus macaques, and are functional in the NMDAR1 internalization assay based on human IPSC-derived cortical neurons. The age dependence of seroprevalence is lost in nonhuman primates in captivity and in human migrants, raising the intriguing possibility that chronic life stress may be related to NMDAR1-AB formation, predominantly of the IgA class. Active immunization of ApoE-/- and ApoE+/+ mice against four peptides of the extracellular NMDAR1 domain or ovalbumin (control) leads to high circulating levels of specific AB. After 4 weeks, the endogenously formed NMDAR1-AB (IgG) induce psychosis-like symptoms upon MK-801 challenge in ApoE-/- mice, characterized by an open blood-brain barrier, but not in their ApoE+/+ littermates, which are indistinguishable from ovalbumin controls. Importantly, NMDAR1-AB do not induce any sign of inflammation in the brain. Immunohistochemical staining for microglial activation markers and T lymphocytes in the hippocampus yields comparable results in ApoE-/- and ApoE+/+ mice, irrespective of immunization against NMDAR1 or ovalbumin. These data suggest that NMDAR1-AB of the IgG class shape behavioral phenotypes upon access to the brain but do not cause brain inflammation on their own.
Collapse
Affiliation(s)
- Hong Pan
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Bárbara Oliveira
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Gesine Saher
- 0000 0001 0668 6902grid.419522.9Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Ekrem Dere
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Daniel Tapken
- 0000 0004 0490 981Xgrid.5570.7Department of Biochemistry I—Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Marina Mitjans
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jan Seidel
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Janina Wesolowski
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Debia Wakhloo
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Christina Klein-Schmidt
- 0000 0004 0490 981Xgrid.5570.7Department of Biochemistry I—Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Anja Ronnenberg
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kerstin Schwabe
- 0000 0000 9529 9877grid.10423.34Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Ralf Trippe
- 0000 0004 0490 981Xgrid.5570.7Department of Biochemistry I—Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Kerstin Mätz-Rensing
- Department of Pathology, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Stefan Berghoff
- 0000 0001 0668 6902grid.419522.9Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Yazeed Al-Krinawe
- 0000 0000 9529 9877grid.10423.34Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | | | - Martin Begemann
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Winfried Stöcker
- Institute for Experimental Immunology, affiliated to Euroimmun, Lübeck, Germany
| | - Franz-Josef Kaup
- Department of Pathology, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Reinhard Mischke
- 0000 0001 0126 6191grid.412970.9Small Animal Clinic, University of Veterinary Medicine, Hannover, Germany
| | - Susann Boretius
- Functional Imaging Laboratory, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Klaus-Armin Nave
- 0000 0001 0668 6902grid.419522.9Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Joachim K. Krauss
- 0000 0000 9529 9877grid.10423.34Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Michael Hollmann
- 0000 0004 0490 981Xgrid.5570.7Department of Biochemistry I—Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Fred Lühder
- 0000 0001 0482 5331grid.411984.1Department of Neuroimmunology, Institute for Multiple Sclerosis Research and Hertie Foundation, University Medicine Göttingen, Göttingen, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
8
|
Labrousse VF, Leyrolle Q, Amadieu C, Aubert A, Sere A, Coutureau E, Grégoire S, Bretillon L, Pallet V, Gressens P, Joffre C, Nadjar A, Layé S. Dietary omega-3 deficiency exacerbates inflammation and reveals spatial memory deficits in mice exposed to lipopolysaccharide during gestation. Brain Behav Immun 2018; 73:427-440. [PMID: 29879442 DOI: 10.1016/j.bbi.2018.06.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/21/2018] [Accepted: 06/03/2018] [Indexed: 01/03/2023] Open
Abstract
Maternal immune activation (MIA) is a common environmental insult on the developing brain and represents a risk factor for neurodevelopmental disorders. Animal models of in utero inflammation further revealed a causal link between maternal inflammatory activation during pregnancy and behavioural impairment relevant to neurodevelopmental disorders in the offspring. Accumulating evidence point out that proinflammatory cytokines produced both in the maternal and fetal compartments are responsible for social, cognitive and emotional behavioral deficits in the offspring. Polyunsaturated fatty acids (PUFAs) are essential fatty acids with potent immunomodulatory activities. PUFAs and their bioactive derivatives can promote or inhibit many aspects of the immune and inflammatory response. PUFAs of the n-3 series ('n-3 PUFAs', also known as omega-3) exhibit anti-inflammatory/pro-resolution properties and promote immune functions, while PUFAs of the n-6 series ('n-6 PUFAs' or omega-6) favor pro-inflammatory responses. The present study aimed at providing insight into the effects of n-3 PUFAs on the consequences of MIA on brain development. We hypothesized that a reduction in n-3 PUFAs exacerbates both maternal and fetal inflammatory responses to MIA and later-life defects in memory in the offspring. Based on a lipopolysaccharide (LPS) model of MIA (LPS injection at embryonic day 17), we showed that n-3 PUFA deficiency 1) alters fatty acid composition of the fetal and adult offspring brain; 2) exacerbates maternal and fetal inflammatory processes with no significant alteration of microglia phenotype, and 3) induces spatial memory deficits in the adult offspring. We also showed a strong negative correlation between brain content in n-3 PUFA and cytokine production in MIA-exposed fetuses. Overall, our study is the first to address the deleterious effects of n-3 PUFA deficiency on brain lipid composition, inflammation and memory performances in MIA-exposed animals and indicates that it should be considered as a potent environmental risk factor for the apparition of neurodevelopmental disorders.
Collapse
Affiliation(s)
- V F Labrousse
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - Q Leyrolle
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France
| | - C Amadieu
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Aubert
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Sere
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - E Coutureau
- Centre National de la Recherche Scientifique, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Uité Mixte de Recherche 5287, 33076 Bordeaux, France; Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, 33076 Bordeaux, France
| | - S Grégoire
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - L Bretillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - V Pallet
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - P Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - C Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
9
|
Franklin TC, Wohleb ES, Zhang Y, Fogaça M, Hare B, Duman RS. Persistent Increase in Microglial RAGE Contributes to Chronic Stress-Induced Priming of Depressive-like Behavior. Biol Psychiatry 2018; 83:50-60. [PMID: 28882317 PMCID: PMC6369917 DOI: 10.1016/j.biopsych.2017.06.034] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic stress-induced inflammatory responses occur in part via danger-associated molecular pattern (DAMP) molecules, such as high mobility group box 1 protein (HMGB1), but the receptor(s) underlying DAMP signaling have not been identified. METHODS Microglia morphology and DAMP signaling in enriched rat hippocampal microglia were examined during the development and expression of chronic unpredictable stress (CUS)-induced behavioral deficits, including long-term, persistent changes after CUS. RESULTS The results show that CUS promotes significant morphological changes and causes robust upregulation of HMGB1 messenger RNA in enriched hippocampal microglia, an effect that persists for up to 6 weeks after CUS exposure. This coincides with robust and persistent upregulation of receptor for advanced glycation end products (RAGE) messenger RNA, but not toll-like receptor 4 in hippocampal microglia. CUS also increased surface expression of RAGE protein on hippocampal microglia as determined by flow cytometry and returned to basal levels 5 weeks after CUS. Importantly, exposure to short-term stress was sufficient to increase RAGE surface expression as well as anhedonic behavior, reflecting a primed state that results from a persistent increase in RAGE messenger RNA expression. Further evidence for DAMP signaling in behavioral responses is provided by evidence that HMGB1 infusion into the hippocampus was sufficient to cause anhedonic behavior and by evidence that RAGE knockout mice were resilient to stress-induced anhedonia. CONCLUSIONS Together, the results provide evidence of persistent microglial HMGB1-RAGE expression that increases vulnerability to depressive-like behaviors long after chronic stress exposure.
Collapse
|
10
|
Zhao Z, Ong LK, Johnson S, Nilsson M, Walker FR. Chronic stress induced disruption of the peri-infarct neurovascular unit following experimentally induced photothrombotic stroke. J Cereb Blood Flow Metab 2017; 37:3709-3724. [PMID: 28304184 PMCID: PMC5718325 DOI: 10.1177/0271678x17696100] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
How stress influences brain repair is an issue of considerable importance, as patients recovering from stroke are known to experience high and often unremitting levels of stress post-event. In the current study, we investigated how chronic stress modified the key cellular components of the neurovascular unit. Using an experimental model of focal cortical ischemia in male C57BL/6 mice, we examined how exposure to a persistently aversive environment, induced by the application of chronic restraint stress, altered the cortical remodeling post-stroke. We focused on systematically investigating changes in the key components of the neurovascular unit (i.e. neurons, microglia, astrocytes, and blood vessels) within the peri-infarct territories using both immunohistochemistry and Western blotting. The results from our study indicated that exposure to chronic stress exerted a significant suppressive effect on each of the key cellular components involved in neurovascular remodeling. Co-incident with these cellular changes, we observed that chronic stress was associated with an exacerbation of motor impairment 42 days post-event. Collectively, these results highlight the vulnerability of the peri-infarct neurovascular unit to the negative effects of chronic stress.
Collapse
Affiliation(s)
- Zidan Zhao
- 1 School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,2 Hunter Medical Research Institute, Newcastle, NSW, Australia.,3 NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Australia
| | - Lin Kooi Ong
- 1 School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,2 Hunter Medical Research Institute, Newcastle, NSW, Australia.,3 NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Australia
| | - Sarah Johnson
- 4 School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Nilsson
- 1 School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,2 Hunter Medical Research Institute, Newcastle, NSW, Australia.,3 NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Australia
| | - Frederick R Walker
- 1 School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,2 Hunter Medical Research Institute, Newcastle, NSW, Australia.,3 NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Australia
| |
Collapse
|
11
|
Zhang J, Liu H, Wei B. Immune response of T cells during herpes simplex virus type 1 (HSV-1) infection. J Zhejiang Univ Sci B 2017; 18:277-288. [PMID: 28378566 PMCID: PMC5394093 DOI: 10.1631/jzus.b1600460] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/07/2017] [Indexed: 12/14/2022]
Abstract
Herpes simplex virus type 1 (HSV-1), a neurotropic member of the alphaherpes virus family, is among the most prevalent and successful human pathogens. HSV-1 can cause serious diseases at every stage of life including fatal disseminated disease in newborns, cold sores, eye disease, and fatal encephalitis in adults. HSV-1 infection can trigger rapid immune responses, and efficient inhibition and clearance of HSV-1 infection rely on both the innate and adaptive immune responses of the host. Multiple strategies have been used to restrict host innate immune responses by HSV-1 to facilitate its infection in host cells. The adaptive immunity of the host plays an important role in inhibiting HSV-1 infections. The activation and regulation of T cells are the important aspects of the adaptive immunity. They play a crucial role in host-mediated immunity and are important for clearing HSV-1. In this review, we examine the findings on T cell immune responses during HSV-1 infection, which hold promise in the design of new vaccine candidates for HSV-1.
Collapse
|
12
|
Ramirez K, Sheridan JF. Antidepressant imipramine diminishes stress-induced inflammation in the periphery and central nervous system and related anxiety- and depressive- like behaviors. Brain Behav Immun 2016; 57:293-303. [PMID: 27223094 PMCID: PMC5010955 DOI: 10.1016/j.bbi.2016.05.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 12/30/2022] Open
Abstract
In order to relieve anxiety and depression accompanying stress, physicians resort to tricyclic antidepressants, such as imipramine. We had previously shown that imipramine reversed stress-induced social avoidance behavior, and down-regulated microglial activation 24days after stress cessation. To further characterize the effects of imipramine on stress induced neuroimmune dysregulation and associated changes in behavior, the aims of this study were to determine if imipramine 1) ameliorated stress-induced inflammation in the periphery and central nervous system, and 2) prevented stress related anxiety- and depressive-like behaviors. C57BL/6 mice were treated with imipramine (15mg/kg) in their drinking water, and exposed to repeated social defeat (RSD). Imipramine attenuated stress-induced corticosterone and IL-6 responses in plasma. Imipramine decreased the percentage of monocytes and granulocytes in the bone marrow and circulation. However, imipramine did not prevent splenomegaly, stress-related increased percentage of granulocytes in this organ, and the production of pro-inflammatory cytokines in the spleen, following RSD. Moreover, imipramine abrogated the accumulation of macrophages in the brain in mice exposed to RSD. Imipramine blocked neuroinflammatory signaling and prevented stress-related anxiety- and depressive-like behaviors. These data support the notion that pharmacomodulation of the monoaminergic system, besides exerting anxiolytic and antidepressant effects, may have therapeutic effects as a neuroimmunomodulator during stress.
Collapse
Affiliation(s)
- Karol Ramirez
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA; Faculty of Dentistry, University of Costa Rica, San Pedro, San José 11501-2060, Costa Rica; Neuroscience Research Center, University of Costa Rica, San Pedro, San José 11501-2060, Costa Rica.
| | - John F Sheridan
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
13
|
Ramirez K, Niraula A, Sheridan JF. GABAergic modulation with classical benzodiazepines prevent stress-induced neuro-immune dysregulation and behavioral alterations. Brain Behav Immun 2016; 51:154-168. [PMID: 26342944 PMCID: PMC4679551 DOI: 10.1016/j.bbi.2015.08.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Psychosocial stress is associated with altered immunity, anxiety, and depression. Repeated social defeat (RSD), a model of social stress, triggers egress of inflammatory myeloid progenitor cells (MPCs; CD11b(+)/Ly6C(hi)) that traffic to the brain, promoting anxiety-like behavior. In parallel, RSD enhances neuroinflammatory signaling and long-lasting social avoidant behavior. Lorazepam and clonazepam are routinely prescribed anxiolytics that act by enhancing GABAergic activity in the brain. Besides binding to the central benzodiazepine binding site (CBBS) in the central nervous system (CNS), lorazepam binds to the translocator protein (TSPO) with high affinity causing immunomodulation. Clonazepam targets the CBBS and has low affinity for the TSPO. Here the aims were to determine if lorazepam and clonazepam would: (1) prevent stress-induced peripheral and central inflammatory responses, and (2) block anxiety and social avoidance behavior in mice subjected to RSD. METHODS C57/BL6 mice were divided into experimental groups, and treated with either lorazepam (0.10mg/kg), clonazepam (0.25mg/kg) or vehicle (0.9% NaCl). Behavioral data and tissues were collected the morning after the last cycle of RSD. RESULTS Lorazepam and clonazepam were effective in attenuating mRNA expression of CRH in the hypothalamus and corticosterone in plasma in mice subjected to RSD. Both drugs blocked stress-induced levels of IL-6 in plasma. Lorazepam and clonazepam had different effects on stress-induced enhancement of myelopoiesis and inhibited trafficking of monocytes and granulocytes in circulation. Furthermore, lorazepam, but not clonazepam, inhibited splenomegaly and the production of pro-inflammatory cytokines in the spleen following RSD. Additionally, lorazepam and clonazepam, blocked stress-induced accumulation of macrophages (CD11b(+)/CD45(high)) in the CNS. In a similar manner, both lorazepam and clonazepam prevented neuroinflammatory signaling and reversed anxiety-like and depressive-like behavior in mice exposed to RSD. CONCLUSION These data support the notion that lorazepam and clonazepam, aside from exerting anxiolytic and antidepressant effects, may have therapeutic potential as neuroimmunomodulators during psychosocial stress. The reversal of RSD-induced behavioral outcomes may be due to the enhancement of GABAergic neurotransmission, or some other off-target effect. The peripheral actions of lorazepam, but not clonazepam, seem to be mediated by TSPO activation.
Collapse
Affiliation(s)
- Karol Ramirez
- Division of Biosciences, The Ohio State University, College of Dentistry, Columbus, OH 43210, USA; Facultad de Odontología, Universidad de Costa Rica, San Pedro, San José 11501-2060, Costa Rica.
| | - Anzela Niraula
- Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43212, USA.
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, College of Dentistry, Columbus, OH 43210, USA; Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
14
|
Ramirez K, Shea DT, McKim DB, B.F. R, Sheridan JF. Imipramine attenuates neuroinflammatory signaling and reverses stress-induced social avoidance. Brain Behav Immun 2015; 46:212-20. [PMID: 25701613 PMCID: PMC4414808 DOI: 10.1016/j.bbi.2015.01.016] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/29/2015] [Accepted: 01/31/2015] [Indexed: 12/18/2022] Open
Abstract
Psychosocial stress is associated with altered immunity, anxiety and depression. Previously we showed that repeated social defeat (RSD) promoted microglia activation and social avoidance behavior that persisted for 24days after cessation of RSD. The aim of the present study was to determine if imipramine (a tricyclic antidepressant) would reverse RSD-inducedsocial avoidance and ameliorate neuroinflammatory responses. To test this, C57BL/6 mice were divided into treatment groups. One group from RSD and controls received daily injections of imipramine for 24days, following 6 cycles of RSD. Two other groups were treated with saline. RSD mice spent significantly less time in the interaction zone when an aggressor was present in the cage. Administration of imipramine reversed social avoidance behavior, significantly increasing the interaction time, so that it was similar to that of control mice. Moreover, 24days of imipramine treatment in RSD mice significantly decreased stress-induced mRNA levels for IL-6 in brain microglia. Following ex vivo LPS stimulation, microglia from mice exposed to RSD, had higher mRNA expression of IL-6, TNF-α, and IL-1β, and this was reversed by imipramine treatment. In a second experiment, imipramine was added to drinking water confirming the reversal of social avoidant behavior and decrease in mRNA expression of IL-6 in microglia. These data suggest that the antidepressant imipramine may exert its effect, in part, by down-regulating microglial activation.
Collapse
Affiliation(s)
- Karol Ramirez
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA; Facultad de Odontología, Universidad de Costa Rica, San Pedro, San José 11501-2060, Costa Rica.
| | - Daniel T. Shea
- Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Daniel B. McKim
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA,Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Reader B.F.
- Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - John F. Sheridan
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA,Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA,Institute for Behavioral Medicine Research, The Ohio State University Medical Center, Columbus, OH 43210, USA,Corresponding author at: College of Dentistry, Division of Biosciences, PO BOX 182357, Columbus, OH 43218-2357, USA, Tel.: +1 614 688 4629, fax: +1 614 292 6087
| |
Collapse
|
15
|
Transgenic increase in n-3/n-6 fatty acid ratio protects against cognitive deficits induced by an immune challenge through decrease of neuroinflammation. Neuropsychopharmacology 2015; 40:525-36. [PMID: 25228141 PMCID: PMC4289942 DOI: 10.1038/npp.2014.196] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/17/2014] [Accepted: 07/25/2014] [Indexed: 12/19/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) display immunomodulatory properties in the brain, n-3 PUFAs being able to reduce inflammation whereas n-6 PUFAs are more pro-inflammatory. It has been extensively demonstrated that exposure to a peripheral immune challenge leads to the production and release of inflammatory mediators in the brain in association with cognitive deficits. The question arises whether n-3 PUFA supplementation could downregulate the brain inflammatory response and subsequent cognitive alterations. In this study, we used a genetically modified mouse line carrying the fat-1 gene from the roundworm Caenorhabditis elegans, encoding an n-3 PUFA desaturase that catalyzes conversion of n-6 into n-3 PUFA. Consequently, these mice display endogenously elevated n-3 PUFA tissue contents. Fat-1 mice or wild-type (WT) littermates were injected peripherally with lipopolysaccharide (LPS), a bacterial endotoxin, to induce an inflammatory episode. Our results showed that LPS altered differently the phenotype of microglia and the expression of cytokines and chemokines in Fat-1 and WT mice. In Fat-1 mice, pro-inflammatory factors synthesis was lowered compared with WT mice, whereas anti-inflammatory mechanisms were favored 24 h after LPS treatment. Moreover, LPS injection impaired spatial memory in WT mice, whereas interestingly, the Fat-1 mice showed normal cognitive performances. All together, these data suggest that the central n-3 PUFA increase observed in Fat-1 mice modulated the brain innate immune system activity, leading to the protection of animals against LPS-induced pro-inflammatory cytokine production and subsequent spatial memory alteration.
Collapse
|
16
|
Dey A, Hao S, Erion JR, Wosiski-Kuhn M, Stranahan AM. Glucocorticoid sensitization of microglia in a genetic mouse model of obesity and diabetes. J Neuroimmunol 2014; 269:20-27. [PMID: 24534266 DOI: 10.1016/j.jneuroim.2014.01.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/11/2013] [Accepted: 01/28/2014] [Indexed: 01/13/2023]
Abstract
db/db mice are a model of obesity and diabetes due to their lack of functional leptin receptors, which leads to insulin resistance, elevated corticosterone levels, and persistent inflammation. Because stress-induced elevations in glucocorticoids sensitize microglia to immune challenges, we hypothesized that corticosteroids might act similarly in the diabetic brain. To test this hypothesis, db/db and wildtype mice were treated with the glucocorticoid synthesis inhibitor metyrapone every day for 2weeks. This treatment revealed corticosterone-dependent increases in microglial number and accumulation of the pro-inflammatory cytokines interleukin 1beta and tumor necrosis factor alpha in the hippocampus of db/db mice. Analysis of microglial responses to lipopolysaccharide stimulation revealed that glucocorticoids lower the threshold for release of pro-inflammatory cytokines, underscoring the role of corticosteroids as a precipitating factor for neuroinflammation in obesity and diabetes.
Collapse
Affiliation(s)
- Aditi Dey
- Department of Physiology, Georgia Regents University, Augusta, GA, 30912, USA
| | - Shuai Hao
- Department of Physiology, Georgia Regents University, Augusta, GA, 30912, USA
| | - Joanna R Erion
- Department of Physiology, Georgia Regents University, Augusta, GA, 30912, USA
| | | | - Alexis M Stranahan
- Department of Physiology, Georgia Regents University, Augusta, GA, 30912, USA
| |
Collapse
|
17
|
Madore C, Joffre C, Delpech JC, De Smedt-Peyrusse V, Aubert A, Coste L, Layé S, Nadjar A. Early morphofunctional plasticity of microglia in response to acute lipopolysaccharide. Brain Behav Immun 2013; 34:151-8. [PMID: 23994463 DOI: 10.1016/j.bbi.2013.08.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 12/20/2022] Open
Abstract
Within the central nervous system (CNS) the traditional role of microglia has been in brain infection and disease, phagocytosing debris and secreting factors to modify disease progression. This led to the concept of "resting" versus "activated" microglia. However, this is misleading because multiple phenotypic and morphological stages of microglia can influence neuronal structure and function in any condition and recent evidence extends their role to healthy brain homeostasis. The present work was thus aimed at reappraising the concept of morphofunctional activity of microglia in a context of peripheral acute immune challenge, where microglial activity is known to be modified, using the new state-of-the-art techniques available. To do so, mice were injected peripherally with lipopolysaccharide, a potent inducer of cerebral inflammation, and we assessed early cytokines production, phenotype, motility and morphology of microglial cells. Our results showed that LPS induced a widespread inflammatory response both peripherally and centrally, as revealed by the quantification of cytokines levels. We also found an alteration of microglial motility that was confirmed by in vivo studies showing an overall reduction of microglial processes length in the hippocampus of LPS-treated animals. Finally, analysis of various surface receptors expression revealed that LPS did not significantly impact microglial phenotype 2h after the injection but rather induced an increase of CD11b(+)/CD45(high) cells. These latter may be at the vasculature, at the CNS vicinity, or may have invaded the CNS.
Collapse
Affiliation(s)
- C Madore
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France; Univ. Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Weber MD, Frank MG, Sobesky JL, Watkins LR, Maier SF. Blocking toll-like receptor 2 and 4 signaling during a stressor prevents stress-induced priming of neuroinflammatory responses to a subsequent immune challenge. Brain Behav Immun 2013; 32:112-21. [PMID: 23500798 PMCID: PMC3810175 DOI: 10.1016/j.bbi.2013.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 02/26/2013] [Accepted: 03/08/2013] [Indexed: 11/16/2022] Open
Abstract
Acute and chronic stressors sensitize or prime the neuroinflammatory response to a subsequent peripheral or central immunologic challenge. However, the neuroimmune process(es) by which stressors prime or sensitize subsequent neuroinflammatory responses remains unclear. Prior evidence suggested that toll-like receptors (TLRs) might be involved in the mediation of primed neuroinflammatory responses, but the role of TLRs during a stressor has never been directly tested. Here, a novel TLR2 and TLR4 antagonist, OxPAPC, was used to probe the contribution of TLRs in the stress sensitization phenomenon. OxPAPC has not previously been administered to the brain, and so its action in blocking TLR2 and TLR4 action in brain was first verified. Administration of OxPAPC into the CNS prior to stress prevented the stress-induced potentiation of hippocampal pro-inflammatory response to a subsequent peripheral LPS challenge occurring 24 h later. In addition, in vivo administration of OxPAPC prior to stress prevented the sensitized pro-inflammatory response from isolated microglia following administration of LPS ex vivo, further implicating microglia as a key neuroimmune substrate that mediates stress-induced sensitized neuroinflammation.
Collapse
Affiliation(s)
- Michael D. Weber
- Corresponding Author: Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0345, USA. Phone number: 614-937-2613. Fax number: 303-492-2967,
| | | | | | | | | |
Collapse
|
19
|
Márquez L, García-Bueno B, Madrigal JLM, Leza JC. Mangiferin decreases inflammation and oxidative damage in rat brain after stress. Eur J Nutr 2011; 51:729-39. [PMID: 21986672 DOI: 10.1007/s00394-011-0252-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/26/2011] [Indexed: 01/03/2023]
Abstract
PURPOSE Stress exposure elicits neuroinflammation and oxidative damage in brain, and stress-related neurological and neuropsychiatric diseases have been associated with cell damage and death. Mangiferin (MAG) is a polyphenolic compound abundant in the stem bark of Mangifera indica L. with antioxidant and anti-inflammatory properties in different experimental settings. In this study, the capacity of MAG to prevent neuroinflammation and brain oxidative damage induced by stress exposure was investigated. METHODS Young-adult male Wistar rats immobilized during 6 h were administered by oral gavage with increasing doses of MAG (15, 30, and 60 mg/Kg), respectively, 7 days before stress. RESULTS Prior treatment with MAG prevented all of the following stress-induced effects: (1) increase in glucocorticoids (GCs) and interleukin-1β (IL-1β) plasma levels, (2) loss of redox balance and reduction in catalase brain levels, (3) increase in pro-inflammatory mediators, such as tumor necrosis factor alpha TNF-α and its receptor TNF-R1, nuclear factor-kappa B (NF-κB) and synthesis enzymes, such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), (4) increase in lipid peroxidation. CONCLUSIONS These multifaceted protective effects suggest that MAG administration could be a new therapeutic strategy in neurological/neuropsychiatric pathologies in which hypothalamic/pituitary/adrenal (HPA) stress axis dysregulation, neuroinflammation, and oxidative damage take place in their pathophysiology.
Collapse
Affiliation(s)
- Lucía Márquez
- Department of Pharmacology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | | | | |
Collapse
|
20
|
Furr SR, Chauhan VS, Moerdyk-Schauwecker MJ, Marriott I. A role for DNA-dependent activator of interferon regulatory factor in the recognition of herpes simplex virus type 1 by glial cells. J Neuroinflammation 2011; 8:99. [PMID: 21838860 PMCID: PMC3168419 DOI: 10.1186/1742-2094-8-99] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 08/12/2011] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The rapid onset of potentially lethal neuroinflammation is a defining feature of viral encephalitis. Microglia and astrocytes are likely to play a significant role in viral encephalitis pathophysiology as they are ideally positioned to respond to invading central nervous system (CNS) pathogens by producing key inflammatory mediators. Recently, DNA-dependent activator of IFN regulatory factor (DAI) has been reported to function as an intracellular sensor for DNA viruses. To date, the expression and functional role of DAI in the inflammatory responses of resident CNS cells to neurotropic DNA viruses has not been reported. METHODS Expression of DAI and its downstream effector molecules was determined in C57BL/6-derived microglia and astrocytes, either at rest or following exposure to herpes simplex virus type 1 (HSV-1) and/or murine gammaherpesvirus-68 (MHV-68), by immunoblot analysis. In addition, such expression was studied in ex vivo microglia/macrophages and astrocytes from uninfected animals or mice infected with HSV-1. Inflammatory cytokine production by glial cultures following transfection with a DAI specific ligand (B-DNA), or following HSV-1 challenge in the absence or presence of siRNA directed against DAI, was assessed by specific capture ELISA. The production of soluble neurotoxic mediators by HSV-1 infected glia following DAI knockdown was assessed by analysis of the susceptibility of neuron-like cells to conditioned glial media. RESULTS We show that isolated microglia and astrocytes constitutively express DAI and its effector molecules, and show that such expression is upregulated following DNA virus challenge. We demonstrate that these resident CNS cells express DAI in situ, and show that its expression is similarly elevated in a murine model of HSV-1 encephalitis. Importantly, we show B-DNA transfection can elicit inflammatory cytokine production by isolated glial cells and DAI knockdown can significantly reduce microglial and astrocyte responses to HSV-1. Finally, we demonstrate that HSV-1 challenged microglia and astrocytes release neurotoxic mediators and show that such production is significantly attenuated following DAI knockdown. CONCLUSIONS The functional expression of DAI by microglia and astrocytes may represent an important innate immune mechanism underlying the rapid and potentially lethal inflammation associated with neurotropic DNA virus infection.
Collapse
MESH Headings
- Animals
- Astrocytes/cytology
- Astrocytes/metabolism
- Astrocytes/virology
- Cell Death
- Cells, Cultured
- DNA, B-Form/chemistry
- DNA, B-Form/metabolism
- Encephalitis, Viral/immunology
- Encephalitis, Viral/physiopathology
- Encephalitis, Viral/virology
- Female
- Herpesvirus 1, Human/immunology
- Humans
- Immunity, Innate/immunology
- Interferon Regulatory Factors/genetics
- Interferon Regulatory Factors/metabolism
- Interleukin-6/immunology
- Mice
- Mice, Inbred C57BL
- Microglia/cytology
- Microglia/metabolism
- Microglia/virology
- Neurons/pathology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- Samantha R Furr
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Vinita S Chauhan
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | | | - Ian Marriott
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
21
|
β-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J Neurosci 2011; 31:6277-88. [PMID: 21525267 DOI: 10.1523/jneurosci.0450-11.2011] [Citation(s) in RCA: 519] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Psychosocial stress is associated with altered immune function and development of psychological disorders including anxiety and depression. Here we show that repeated social defeat in mice increased c-Fos staining in brain regions associated with fear and threat appraisal and promoted anxiety-like behavior in a β-adrenergic receptor-dependent manner. Repeated social defeat also significantly increased the number of CD11b(+)/CD45(high)/Ly6C(high) macrophages that trafficked to the brain. In addition, several inflammatory markers were increased on the surface of microglia (CD14, CD86, and TLR4) and macrophages (CD14 and CD86) after social defeat. Repeated social defeat also increased the presence of deramified microglia in the medial amygdala, prefrontal cortex, and hippocampus. Moreover, mRNA analysis of microglia indicated that repeated social defeat increased levels of interleukin (IL)-1β and reduced levels of glucocorticoid responsive genes [glucocorticoid-induced leucine zipper (GILZ) and FK506 binding protein-51 (FKBP51)]. The stress-dependent changes in microglia and macrophages were prevented by propranolol, a β-adrenergic receptor antagonist. Microglia isolated from socially defeated mice and cultured ex vivo produced markedly higher levels of IL-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1 after stimulation with lipopolysaccharide compared with microglia from control mice. Last, repeated social defeat increased c-Fos activation in IL-1 receptor type-1-deficient mice, but did not promote anxiety-like behavior or microglia activation in the absence of functional IL-1 receptor type-1. These findings indicate that repeated social defeat-induced anxiety-like behavior and enhanced reactivity of microglia was dependent on activation of β-adrenergic and IL-1 receptors.
Collapse
|
22
|
Powell ND, Allen RG, Hufnagle AR, Sheridan JF, Bailey MT. Stressor-induced alterations of adaptive immunity to vaccination and viral pathogens. Immunol Allergy Clin North Am 2011; 31:69-79. [PMID: 21094924 PMCID: PMC3339561 DOI: 10.1016/j.iac.2010.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The stress response influences the immune system, and studies in laboratory animals indicate that the response to stress significantly reduces resistance to infectious challenge. Only a few studies, however, have determined the impact of the stress response on human susceptibility to infectious challenge due, in part, to the difficulties of using live, replicating pathogens in human research. As a result, many studies have assessed the immune response to vaccination as a surrogate for the immune response to an infectious challenge. Thus, much is known about how the stress response influences adaptive immunity, and memory responses, to vaccination. These studies have yielded data concerning the interactions of the nervous and immune systems and have provided important information for clinicians administering vaccines to susceptible populations. This review provides a brief overview of the immune response to commonly used vaccines and the impact that stress can have on vaccine-specific immunity.
Collapse
Affiliation(s)
- Nicole D Powell
- Division of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
23
|
Hunzeker JT, Elftman MD, Mellinger JC, Princiotta MF, Bonneau RH, Truckenmiller ME, Norbury CC. A marked reduction in priming of cytotoxic CD8+ T cells mediated by stress-induced glucocorticoids involves multiple deficiencies in cross-presentation by dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 186:183-94. [PMID: 21098225 DOI: 10.4049/jimmunol.1001737] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protracted psychological stress elevates circulating glucocorticoids, which can suppress CD8(+) T cell-mediated immunity, but the mechanisms are incompletely understood. Dendritic cells (DCs), required for initiating CTL responses, are vulnerable to stress/corticosterone, which can contribute to diminished CTL responses. Cross-priming of CD8(+) T cells by DCs is required for initiating CTL responses against many intracellular pathogens that do not infect DCs. We examined the effects of stress/corticosterone on MHC class I (MHC I) cross-presentation and priming and show that stress/corticosterone-exposed DCs have a reduced ability to cross-present OVA and activate MHC I-OVA(257-264)-specific T cells. Using a murine model of psychological stress and OVA-loaded β(2)-microglobulin knockout "donor" cells that cannot present Ag, DCs from stressed mice induced markedly less Ag-specific CTL proliferation in a glucocorticoid receptor-dependent manner, and endogenous in vivo T cell cytolytic activity generated by cross-presented Ag was greatly diminished. These deficits in cross-presentation/priming were not due to altered Ag donation, Ag uptake (phagocytosis, receptor-mediated endocytosis, or fluid-phase uptake), or costimulatory molecule expression by DCs. However, proteasome activity in corticosterone-treated DCs or splenic DCs from stressed mice was partially suppressed, which limits formation of antigenic peptide-MHC I complexes. In addition, the lymphoid tissue-resident CD11b(-)CD24(+)CD8α(+) DC subset, which carries out cross-presentation/priming, was preferentially depleted in stressed mice. At the same time, CD11b(-)CD24(+)CD8α(-) DC precursors were increased, suggesting a block in development of CD8α(+) DCs. Therefore, glucocorticoid-induced changes in both the cellular composition of the immune system and intracellular protein degradation contribute to impaired CTL priming in stressed mice.
Collapse
Affiliation(s)
- John T Hunzeker
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Elftman MD, Hunzeker JT, Mellinger JC, Bonneau RH, Norbury CC, Truckenmiller ME. Stress-induced glucocorticoids at the earliest stages of herpes simplex virus-1 infection suppress subsequent antiviral immunity, implicating impaired dendritic cell function. THE JOURNAL OF IMMUNOLOGY 2010; 184:1867-75. [PMID: 20089700 DOI: 10.4049/jimmunol.0902469] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The systemic elevation of psychological stress-induced glucocorticoids strongly suppresses CD8(+) T cell immune responses resulting in diminished antiviral immunity. However, the specific cellular targets of stress/glucocorticoids, the timing of exposure, the chronology of immunological events, and the underlying mechanisms of this impairment are incompletely understood. In this study, we address each of these questions in the context of a murine cutaneous HSV infection. We show that exposure to stress or corticosterone in only the earliest stages of an HSV-1 infection is sufficient to suppress, in a glucocorticoid receptor-dependent manner, the subsequent antiviral immune response after stress/corticosterone has been terminated. This suppression resulted in early onset and delayed resolution of herpetic lesions, reduced viral clearance at the site of infection and draining popliteal lymph nodes (PLNs), and impaired functions of HSV-specific CD8(+) T cells in PLNs, including granzyme B and IFN-gamma production and the ability to degranulate. In knockout mice lacking glucocorticoid receptors only in T cells, we show that these impaired CD8(+) T cell functions are not due to direct effects of stress/corticosterone on the T cells, but the ability of PLN-derived dendritic cells to prime HSV-1-specific CD8(+) T cells is functionally impaired. These findings highlight the susceptibility of critical early events in the generation of an antiviral immune response to neuroendocrine modulation and implicate dendritic cells as targets of stress/glucocorticoids in vivo. These findings also provide insight into the mechanisms by which the clinical use of glucocorticoids contributes to altered immune responses in patients with viral infections or tumors.
Collapse
Affiliation(s)
- Michael D Elftman
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
25
|
Kubera M, Maes M, Budziszewska B, Basta-Kaim A, Leśekiewicz M, Grygier B, Rogóż Z, Lasoń W. Inhibitory effects of amantadine on the production of pro-inflammatory cytokines by stimulated in vitro human blood. Pharmacol Rep 2009; 61:1105-12. [DOI: 10.1016/s1734-1140(09)70173-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 10/21/2009] [Indexed: 11/26/2022]
|
26
|
Galic MA, Riazi K, Henderson AK, Tsutsui S, Pittman QJ. Viral-like brain inflammation during development causes increased seizure susceptibility in adult rats. Neurobiol Dis 2009; 36:343-51. [PMID: 19660546 DOI: 10.1016/j.nbd.2009.07.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2009] [Revised: 07/26/2009] [Accepted: 07/29/2009] [Indexed: 11/26/2022] Open
Abstract
Viral infections of the CNS and their accompanying inflammation can cause long-term neurological effects, including increased risk for seizures. To examine the effects of CNS inflammation, we infused polyinosinic:polycytidylic acid, intracerebroventricularly to mimic a viral CNS infection in 14 day-old rats. This caused fever and an increase in the pro-inflammatory cytokine, interleukin (IL)-1beta in the brain. As young adults, these animals were more susceptible to lithium-pilocarpine and pentylenetetrazol-induced seizures and showed memory deficits in fear conditioning. Whereas there was no alteration in adult hippocampal cytokine levels, we found a marked increase in NMDA (NR2A and C) and AMPA (GluR1) glutamate receptor subunit mRNA expression. The increase in seizure susceptibility, glutamate receptor subunits, and hippocampal IL-1beta levels were suppressed by neonatal systemic minocycline. Thus, a novel model of viral CNS inflammation reveals pathophysiological relationships between brain cytokines, glutamate receptors, behaviour and seizures, which can be attenuated by anti-inflammatory agents like minocycline.
Collapse
Affiliation(s)
- M A Galic
- Epilepsy and Brain Circuits Program, Hotchkiss Brain Institute, Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
27
|
Sheridan PA, Beck MA. The dendritic and T cell responses to herpes simplex virus-1 are modulated by dietary vitamin E. Free Radic Biol Med 2009; 46:1581-8. [PMID: 19303435 PMCID: PMC2693096 DOI: 10.1016/j.freeradbiomed.2009.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 02/24/2009] [Accepted: 03/06/2009] [Indexed: 11/25/2022]
Abstract
Previous studies from our laboratory have shown that dietary alpha-tocopherol (vitamin E, or VE) is essential for regulating the cytokine and chemokine response in the brain to herpes simplex virus-1 (HSV-1) infection. The timing of T cell infiltration is critical to the resolution of central nervous system HSV-1 infections. Specifically, the appearance of "neuroprotective" CD8(+)IFN-gamma(+) T cells is crucial. During CNS infection, CD8(+) T cell priming and expansion in the draining lymph node, followed by recruitment and expansion, occurs in the spleen with subsequent accumulation in the brain. Weanling male BALB/cByJ mice were placed on VE-deficient (Def) or -adequate diets for 4 weeks followed by intranasal infection with HSV-1. VE-Def mice had fewer CD8(+)IFN-gamma(+) T cells trafficking to the brain despite increased CD8(+)IFN-gamma(+) T cells and activated dendritic cells in the periphery. VE-Def mice had increased T regulatory cells (Tregs) in the periphery and brain, and the increase in Tregs decreased CD8(+) T cell numbers in the brain. Our results demonstrate that adequate levels of VE are important for trafficking antigen-specific T cells to the brain, and dietary VE levels modulate T regulatory and dendritic cells in the periphery.
Collapse
Affiliation(s)
- Patricia A Sheridan
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
28
|
Steel CD, Hahto SM, Ciavarra RP. Peripheral dendritic cells are essential for both the innate and adaptive antiviral immune responses in the central nervous system. Virology 2009; 387:117-26. [PMID: 19264338 DOI: 10.1016/j.virol.2009.01.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Revised: 11/18/2008] [Accepted: 01/22/2009] [Indexed: 12/25/2022]
Abstract
Intranasal application of vesicular stomatitis virus (VSV) causes acute infection of the central nervous system (CNS). However, VSV encephalitis is not invariably fatal, suggesting that the CNS may contain a professional antigen-presenting cell (APC) capable of inducing or propagating a protective antiviral immune response. To examine this possibility, we first characterized the cellular elements that infiltrate the brain as well as the activation status of resident microglia in the brains of normal and transgenic mice acutely ablated of peripheral dendritic cells (DCs) in vivo. VSV encephalitis was characterized by a pronounced infiltrate of myeloid cells (CD45(high)CD11b(+)) and CD8(+) T cells containing a subset that was specific for the immunodominant VSV nuclear protein epitope. This T cell response correlated temporally with a rapid and sustained upregulation of MHC class I expression on microglia, whereas class II expression was markedly delayed. Ablation of peripheral DCs profoundly inhibited the inflammatory response as well as infiltration of virus-specific CD8(+) T cells. Unexpectedly, the VSV-induced interferon-gamma (IFN-gamma) response in the CNS remained intact in DC-deficient mice. Thus, both the inflammatory and certain components of the adaptive primary antiviral immune response in the CNS are dependent on peripheral DCs in vivo.
Collapse
Affiliation(s)
- Christina D Steel
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 W Olney Road, Norfolk, VA 23501, USA
| | | | | |
Collapse
|
29
|
Marques CP, Cheeran MCJ, Palmquist JM, Hu S, Urban SL, Lokensgard JR. Prolonged microglial cell activation and lymphocyte infiltration following experimental herpes encephalitis. THE JOURNAL OF IMMUNOLOGY 2009; 181:6417-26. [PMID: 18941232 DOI: 10.4049/jimmunol.181.9.6417] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Experimental murine herpes simplex virus (HSV)-1 brain infection stimulates microglial cell-driven proinflammatory chemokine production which precedes the presence of brain-infiltrating systemic immune cells. In the present study, we investigated the phenotypes and infiltration kinetics of leukocyte trafficking into HSV-infected murine brains. Using real-time bioluminescence imaging, the infiltration of luciferase-positive splenocytes, transferred via tail vein injection into the brains of HSV-infected animals, was followed over an 18-day time course. Flow cytometric analysis of brain-infiltrating leukocytes at 5, 8, 14, and 30 days postinfection (d.p.i.), was performed to assess their phenotype. A predominantly macrophage (CD45(high)CD11b(+)Ly6C(high)) and neutrophil (CD45(high)CD11b(+)Ly6G(+)) infiltration was seen early during infection, with elevated levels of TNF-alpha mRNA expression. By 14 d.p.i., the phenotypic profile shifted to a predominantly lymphocytic (CD45(high)CD3(+)) infiltrate. This lymphocyte infiltrate was detected until 30 d.p.i., when infectious virus could not be recovered, with CD8(+) and CD4(+) T cells present at a 3:1 ratio, respectively. This T lymphocyte infiltration paralleled increased IFN-gamma mRNA expression in the brain. Activation of resident microglia (CD45(int)CD11b(+)) was also detected until 30 d.p.i., as assessed by MHC class II expression. Activated microglial cells were further identified as the predominant source of IL-1beta. In addition, infected mice given primed immunocytes at 4 d.p.i. showed a significant increase in mortality. Taken together, these results demonstrate that intranasal infection results in early macrophage and neutrophil infiltration into the brain followed by prolonged microglial activation and T lymphocyte retention. Similar prolonged neuroimmune activation may contribute to the neuropathological sequelae observed in herpes encephalitis patients.
Collapse
Affiliation(s)
- Cristina P Marques
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
30
|
Ashcraft KA, Bonneau RH. Psychological stress exacerbates primary vaginal herpes simplex virus type 1 (HSV-1) infection by impairing both innate and adaptive immune responses. Brain Behav Immun 2008; 22:1231-40. [PMID: 18639627 PMCID: PMC3721735 DOI: 10.1016/j.bbi.2008.06.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/09/2008] [Accepted: 06/20/2008] [Indexed: 11/26/2022] Open
Abstract
Chronic psychological stress is generally immunosuppressive and contributes to an increase in herpes simplex virus (HSV) pathogenicity. We have previously shown that mice experiencing stress at the time of intranasal HSV infection have increased levels of infectious virus in their nasal cavity, as compared to control mice that were not subjected to stress. We have extended our studies to determine the effects of stress at another clinically-relevant mucosal site by examining the immune response to and pathogenesis of vaginal HSV infection. Mice experiencing psychological stress during vaginal HSV infection exhibited an increase in both vaginal viral titers and the pathology associated with this HSV infection. We demonstrate that these observations result from the failure of both the innate and HSV-specific adaptive immune responses. At 2 days post-infection, NK cell numbers were significantly decreased in mice experiencing restraint stress. Studies examining the adaptive immune response revealed a decrease in the number of HSV-specific CD8(+) T cells in not only the vaginal tissue itself but also the draining iliac lymph nodes (ILN). Furthermore, the number of functional cells, in terms of both their degranulation and interferon-gamma production, in the ILN of stressed mice was decreased as compared to non-stressed mice. We conclude that psychological stress, through its suppression of both innate and adaptive immune responses, may be an important factor in the ability to control vaginal HSV infection.
Collapse
Affiliation(s)
- Kathleen A. Ashcraft
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| | - Robert H. Bonneau
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA,Correspondence should be addressed to: Robert H. Bonneau, Ph.D., Department of Microbiology and Immunology (H107), The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, 500 University Drive, Hershey, Pennsylvania 17033, Telephone: 717-531-4078; Fax: 717-531-6522;
| |
Collapse
|
31
|
García-Bueno B, Caso JR, Leza JC. Stress as a neuroinflammatory condition in brain: Damaging and protective mechanisms. Neurosci Biobehav Rev 2008; 32:1136-51. [DOI: 10.1016/j.neubiorev.2008.04.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 04/02/2008] [Accepted: 04/02/2008] [Indexed: 01/07/2023]
|
32
|
Ashcraft KA, Hunzeker J, Bonneau RH. Psychological stress impairs the local CD8+ T cell response to mucosal HSV-1 infection and allows for increased pathogenicity via a glucocorticoid receptor-mediated mechanism. Psychoneuroendocrinology 2008; 33:951-63. [PMID: 18657369 PMCID: PMC3721759 DOI: 10.1016/j.psyneuen.2008.04.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 04/02/2008] [Accepted: 04/09/2008] [Indexed: 11/29/2022]
Abstract
Psychological stress and its associated increases in corticosterone are generally immunosuppressive and contribute to increased herpes simplex virus (HSV)-associated pathogenicity. However, the impact of stress on local control of the initial mucosal-based HSV infection has not been elucidated, nor have the ramifications of such failures of the immune response in terms of viral spread. To address these gaps in knowledge, the studies described herein sought to determine how psychological stress and associated increases in corticosterone may increase susceptibility to HSV encephalitis by allowing for increased viral titers at the site of initial infection. We have shown that in mice intranasally infected with HSV-1, a cell-mediated immune response occurs in the nasopharyngeal-associated lymphoid tissue (NALT), mediastinal lymph nodes (MLN), and superficial cervical lymph nodes (CLN). However, psychological stress induced by restraint decreased the number of lymphocytes in these tissues in HSV-infected mice. Surprisingly, the effects of this restraint stress on HSV-specific CTL function varied by immune tissue. Increased viral titers were found in the nasal cavity of stressed mice, an observation which correlated with an increased CD8+ cell response in the CLN. These findings led us to extend our studies to also determine the ramifications of decreased numbers of locally derived lymphocytes on viral titers following infection. Using an approach in which the NALT was surgically removed prior to infection, we confirmed that decreased numbers of NALT-derived lymphocytes at the time of infection allows for increased viral replication. We conclude that the increased viral titers observed in mice experiencing psychological stress are the consequence of a glucocorticoid-mediated reduction in the numbers of lymphocytes responsible for resolving the initial infection.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/physiology
- Cells, Cultured
- Corticosterone/physiology
- Down-Regulation/drug effects
- Down-Regulation/immunology
- Herpes Simplex/immunology
- Herpes Simplex/veterinary
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/pathogenicity
- Hormone Antagonists/pharmacology
- Immunity, Mucosal/immunology
- Immunity, Mucosal/physiology
- Lymphocyte Activation/immunology
- Lymphocytes/drug effects
- Lymphocytes/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mifepristone/pharmacology
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/physiology
- Stress, Psychological/immunology
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Kathleen A. Ashcraft
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| | - John Hunzeker
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| | - Robert H. Bonneau
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA
| |
Collapse
|
33
|
Bulakbasi N, Kocaoglu M. Central nervous system infections of herpesvirus family. Neuroimaging Clin N Am 2008; 18:53-84; viii. [PMID: 18319155 DOI: 10.1016/j.nic.2007.12.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Herpesviruses are one of the most common groups of pathogens causing central nervous system infections in humans. They mostly cause encephalitis, meningitis, or myelitis in immunocompetent and immunocompromised patients. Children, adults, and the elderly can all be affected. Although contrast-enhanced CT is more widely used for diagnosis, contrast-enhanced MR imaging combined with diffusion-weighted imaging is superior to CT in the detection of early changes and the real extent of the disease, and in assessing prognosis and monitoring response to antiviral treatment. More sophisticated techniques, such as MR spectroscopy and perfusion imaging, can aid in the differential diagnosis of herpesvirus infections from other tumoral, demyelinating, and ischemic processes.
Collapse
Affiliation(s)
- Nail Bulakbasi
- Department of Radiology, Gulhane Military Medical Academy and School of Medicine, Etlik, Ankara 06018, Turkey.
| | | |
Collapse
|
34
|
Zahwa H, Yorty JL, Bonneau RH. Elevated maternal corticosterone during lactation hinders the neonatal adaptive immune response to herpes simplex virus (HSV) infection. Brain Behav Immun 2008; 22:339-53. [PMID: 17950573 DOI: 10.1016/j.bbi.2007.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 08/15/2007] [Accepted: 08/16/2007] [Indexed: 11/29/2022] Open
Abstract
The neonate's immune system is relatively immature. For short-term protection against pathogens the neonate is reliant primarily on maternally derived antibodies delivered via the mother's milk. However, neonates soon acquire the ability to generate adaptive immune responses for long-term protection. Products of the nervous and endocrine systems that are elicited by psychological stress are known to modulate a variety of immune responses. Additionally, psychological stressors are well recognized for their ability to increase corticosterone levels. The studies described herein examined the effects of increases in maternally derived corticosterone on the neonatal cell-mediated immune response to, and pathogenicity of, herpes simplex virus (HSV) infection. Water containing corticosterone was made available to nursing mothers for a period of 6 consecutive days beginning on either the day of or 6 days post-delivery. At 12 days of age, neonates were infected with HSV-1 in the rear footpads. These neonates exhibited a decrease in the proliferative ability of splenic-derived cells due to the reduction of IL-2 production and IL-2 receptor alpha subunit (IL-2R alpha) expression by these cells. These neonates also exhibited a decrease in the number and function of popliteal lymph node-resident HSV-1 gB(498-505) peptide-specific CD8(+) T cells as measured by tetramer analysis, CTL lytic activity, expression of CD107a, cytokine production, and proliferation. Additionally, these HSV-infected neonates exhibited increased morbidity and mortality. Together, these studies indicate that exposure of neonates to maternally derived corticosterone via the milk hinders their ability to generate an adaptive cell-mediated immune response to a viral infection and illustrate the potential importance of maternal stress in neonatal resistance to infectious pathogens.
Collapse
Affiliation(s)
- Hassan Zahwa
- Cell and Molecular Biology Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | |
Collapse
|
35
|
Getts DR, Balcar VJ, Matsumoto I, Müller M, King NJC. Viruses and the immune system: their roles in seizure cascade development. J Neurochem 2008; 104:1167-76. [DOI: 10.1111/j.1471-4159.2007.05171.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Koelle D, Bergemann T. Doctor, Why Is My Herpes So Bad? The Search Continues. J Infect Dis 2008; 197:331-4. [DOI: 10.1086/525541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
37
|
Sheridan PA, Beck MA. The immune response to herpes simplex virus encephalitis in mice is modulated by dietary vitamin E. J Nutr 2008; 138:130-7. [PMID: 18156415 PMCID: PMC2430048 DOI: 10.1093/jn/138.1.130] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus encephalitis (HSE) is the most common fatal sporadic encephalitis in humans. HSE is primarily caused by herpes simplex virus (HSV)-1 infection of the brain. HSE results in increased levels of oxidative stress, including the production of reactive oxygen species, free radicals, and neuroinflammation. The most biologically active form of vitamin E (VE) is alpha-tocopherol (alpha-TOC). In cellular membranes, alpha-TOC prevents lipid peroxidation by scavenging free radicals and functioning as an antioxidant. Supplementation with VE has been shown to decrease immunosenescence, improve immune function, and may be neuroprotective. To determine how VE deficiency and VE supplementation would alter the pathogenesis of HSE, we placed weanling male BALB/cByJ mice on VE-deficient (VE-D), VE-adequate (VE-A), or 10x VE-supplemented diets for 4 wk, and then infected the mice intranasally with HSV-1. VE-D mice had more severe symptoms of encephalitis than VE-A mice, including weight loss, keratitis, hunched posture, and morbidity. VE-D mice had increased cytokine and chemokine expression in the brain and increased viral titers. In contrast, VE supplementation failed to decrease cytokine production and had no effect on viral titer. We demonstrated that adequate levels of VE are important in limiting HSE pathology and that 10x supplementation does not enhance protection.
Collapse
Affiliation(s)
- Patricia A Sheridan
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|