1
|
Lopez-Atalaya JP, Bhojwani-Cabrera AM. Type I interferon signalling and interferon-responsive microglia in health and disease. FEBS J 2025. [PMID: 40299722 DOI: 10.1111/febs.70126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
Recent evidence suggests that type I interferon (IFN-I) signalling extends beyond its canonical roles in antiviral defence and immunomodulation. Over the past decade, dysregulated IFN-I signalling has been linked to genetic disorders and neurodegenerative diseases, where it may contribute to neurological impairments. Microglia have emerged as key mediators of IFN-I responses in the central nervous system. A distinct transcriptional state responsive to interferons has recently been identified in microglia. The activation of the IFN-I pathway in these cells is now recognised as pivotal in both development and neurodegeneration. This review is divided into two main sections: the first examines the broader role of IFN-I signalling in the central nervous system, particularly its contribution to neurological dysfunction; the second focuses on the specific state of interferon-responsive microglia, exploring its mechanisms and relevance in neurodegenerative conditions. Finally, we discuss how these areas intersect and their implications for both healthy and diseased states.
Collapse
Affiliation(s)
- Jose P Lopez-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| | - Aysha M Bhojwani-Cabrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
2
|
Periche-Tomas E, Cattaneo A, Cattane N, Bone C, Tibble J, Bullmore ET, Pariante C, Harrison NA. Acute effects of interferon-alpha on cellular anabolic and catabolic processes are associated with the development of fatigue during Interferon-alpha-based therapy for Hepatitis-C: A preliminary study. Brain Behav Immun 2025; 123:717-724. [PMID: 39414178 DOI: 10.1016/j.bbi.2024.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
INTRODUCTION Interferon-alpha (IFN-α) is a key mediator of antiviral immune responses used to treat Hepatitis-C virus (HCV) infection. Though clinically effective, IFN-α frequently induces functionally impairing mood and motivation symptoms, particularly fatigue. Unlike mood impairment, which typically emerges after weeks of treatment, fatigue tends to emerge and evolve rapidly, typically within hours of the first IFN-α injection. Despite being a major source of functional impairment during IFN-α and other immune-based therapies, the biological mechanisms underlying fatigue remain poorly understood. Here, we aimed to identify acute immune-response signatures to IFN-α that could predict the later development of fatigue. METHODS In this exploratory study, we analyzed whole blood transcriptomics in a longitudinal sample of 27 HCV patients initiating IFN-α and Ribavirin therapy. Blood samples were obtained at baseline and 4½ hours after the first IFN-α dose and transcriptomic data was obtained using Affymetrix Human Gene 1.1 ST Array Strips. Gene expression data visualization and quality control were assessed using Partek Genomics Suite V6.6 and protein-protein interaction networks using STRING and Ingenuity Pathway Analysis (IPA). A Fatigue Visual Analogue Scale (fVAS) was utilized to record fatigue symptoms at baseline, 4½ hours and 4 weeks after initiation of treatment. RESULTS IFN-α was associated with an upregulation of 526 transcripts and a downregulation of 228 genes, indicating a rapid transcriptomic response in whole blood within 4½ hours of injection. 93 genes were significantly positively correlated with changes in fatigue, with gene expression changes measured from baseline to 4.5 h and increases in fatigue assessed from baseline to week 4 on the fVAS. We identified a novel network of predominantly cytosolic ribosomal units and ubiquitin proteins implicated in modulating mTOR signaling that was associated with the development of fatigue 4 weeks after initiation of IFN-α treatment (p = 0.0078). CONCLUSION Our findings suggest that acute activation of this anabolic/catabolic network by IFN-α may predispose to the experience of fatigue similar to evidence found in cancer-related fatigue. Further investigation is warranted to confirm the exploratory nature of these observations.
Collapse
Affiliation(s)
- Eva Periche-Tomas
- CUBRIC, Cardiff University Brain Research Imaging Centre, Cardiff, UK.
| | - Annamaria Cattaneo
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Claudia Bone
- CUBRIC, Cardiff University Brain Research Imaging Centre, Cardiff, UK
| | - Jeremy Tibble
- Department of Gastroenterology, Brighton & Sussex University Hospitals, Brighton, UK
| | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge Clinical School, Cambridge, UK
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Neil A Harrison
- CUBRIC, Cardiff University Brain Research Imaging Centre, Cardiff, UK; Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
3
|
Cuddy SR, Cliffe AR. The Intersection of Innate Immune Pathways with the Latent Herpes Simplex Virus Genome. J Virol 2023; 97:e0135222. [PMID: 37129520 PMCID: PMC10231182 DOI: 10.1128/jvi.01352-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023] Open
Abstract
Innate immune responses can impact different stages of viral life cycles. Herpes simplex virus latent infection of neurons and subsequent reactivation provide a unique context for immune responses to intersect with different stages of infection. Here, we discuss recent findings linking neuronal innate immune pathways with the modulation of latent infection, acting at the time of reactivation and during initial neuronal infection to have a long-term impact on the ability of the virus to reactivate.
Collapse
Affiliation(s)
- Sean R. Cuddy
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Anna R. Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Viengkhou B, Hofer MJ. Breaking down the cellular responses to type I interferon neurotoxicity in the brain. Front Immunol 2023; 14:1110593. [PMID: 36817430 PMCID: PMC9936317 DOI: 10.3389/fimmu.2023.1110593] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Since their original discovery, type I interferons (IFN-Is) have been closely associated with antiviral immune responses. However, their biological functions go far beyond this role, with balanced IFN-I activity being critical to maintain cellular and tissue homeostasis. Recent findings have uncovered a darker side of IFN-Is whereby chronically elevated levels induce devastating neuroinflammatory and neurodegenerative pathologies. The underlying causes of these 'interferonopathies' are diverse and include monogenetic syndromes, autoimmune disorders, as well as chronic infections. The prominent involvement of the CNS in these disorders indicates a particular susceptibility of brain cells to IFN-I toxicity. Here we will discuss the current knowledge of how IFN-Is mediate neurotoxicity in the brain by analyzing the cell-type specific responses to IFN-Is in the CNS, and secondly, by exploring the spectrum of neurological disorders arising from increased IFN-Is. Understanding the nature of IFN-I neurotoxicity is a crucial and fundamental step towards development of new therapeutic strategies for interferonopathies.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
5
|
Lang R, Li H, Luo X, Liu C, Zhang Y, Guo S, Xu J, Bao C, Dong W, Yu Y. Expression and mechanisms of interferon-stimulated genes in viral infection of the central nervous system (CNS) and neurological diseases. Front Immunol 2022; 13:1008072. [PMID: 36325336 PMCID: PMC9618809 DOI: 10.3389/fimmu.2022.1008072] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/28/2022] [Indexed: 09/16/2023] Open
Abstract
Interferons (IFNs) bind to cell surface receptors and activate the expression of interferon-stimulated genes (ISGs) through intracellular signaling cascades. ISGs and their expression products have various biological functions, such as antiviral and immunomodulatory effects, and are essential effector molecules for IFN function. ISGs limit the invasion and replication of the virus in a cell-specific and region-specific manner in the central nervous system (CNS). In addition to participating in natural immunity against viral infections, studies have shown that ISGs are essential in the pathogenesis of CNS disorders such as neuroinflammation and neurodegenerative diseases. The aim of this review is to present a macroscopic overview of the characteristics of ISGs that restrict viral neural invasion and the expression of the ISGs underlying viral infection of CNS cells. Furthermore, we elucidate the characteristics of ISGs expression in neurological inflammation, neuropsychiatric disorders such as depression as well as neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Finally, we summarize several ISGs (ISG15, IFIT2, IFITM3) that have been studied more in recent years for their antiviral infection in the CNS and their research progress in neurological diseases.
Collapse
Affiliation(s)
- Rui Lang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, DeYang, China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShunYu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological diseases and brain function laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Beier KT. The Serendipity of Viral Trans-Neuronal Specificity: More Than Meets the Eye. Front Cell Neurosci 2021; 15:720807. [PMID: 34671244 PMCID: PMC8521040 DOI: 10.3389/fncel.2021.720807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Trans-neuronal viruses are frequently used as neuroanatomical tools for mapping neuronal circuits. Specifically, recombinant one-step rabies viruses (RABV) have been instrumental in the widespread application of viral circuit mapping, as these viruses have enabled labs to map the direct inputs onto defined cell populations. Within the neuroscience community, it is widely believed that RABV spreads directly between neurons via synaptic connections, a hypothesis based principally on two observations. First, the virus labels neurons in a pattern consistent with known anatomical connectivity. Second, few glial cells appear to be infected following RABV injections, despite the fact that glial cells are abundant in the brain. However, there is no direct evidence that RABV can actually be transmitted through synaptic connections. Here we review the immunosubversive mechanisms that are critical to RABV’s success for infiltration of the central nervous system (CNS). These include interfering with and ultimately killing migratory T cells while maintaining levels of interferon (IFN) signaling in the brain parenchyma. Finally, we critically evaluate studies that support or are against synaptically-restricted RABV transmission and the implications of viral-host immune responses for RABV transmission in the brain.
Collapse
Affiliation(s)
- Kevin Thomas Beier
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
7
|
Viengkhou B, White MY, Cordwell SJ, Campbell IL, Hofer MJ. A novel phosphoproteomic landscape evoked in response to type I interferon in the brain and in glial cells. J Neuroinflammation 2021; 18:237. [PMID: 34656141 PMCID: PMC8520650 DOI: 10.1186/s12974-021-02277-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/16/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Type I interferons (IFN-I) are key responders to central nervous system infection and injury and are also increased in common neurodegenerative diseases. Their effects are primarily mediated via transcriptional regulation of several hundred interferon-regulated genes. In addition, IFN-I activate several kinases including members of the MAPK and PI3K families. Yet, how changes to the global protein phosphoproteome contribute to the cellular response to IFN-I is unknown. METHODS The cerebral phosphoproteome of mice with brain-targeted chronic production of the IFN-I, IFN-α, was obtained. Changes in phosphorylation were analyzed by ontology and pathway analysis and kinase enrichment predictions. These were verified by phenotypic analysis, immunohistochemistry and immunoblots. In addition, primary murine microglia and astrocytes, the brain's primary IFN-I-responding cells, were acutely treated with IFN-α and the global phosphoproteome was similarly analyzed. RESULTS We identified widespread protein phosphorylation as a novel mechanism by which IFN-I mediate their effects. In our mouse model for IFN-I-induced neurodegeneration, protein phosphorylation, rather than the proteome, aligned with the clinical hallmarks and pathological outcome, including impaired development, motor dysfunction and seizures. In vitro experiments revealed extensive and rapid IFN-I-induced protein phosphorylation in microglia and astrocytes. Response to acute IFN-I stimulation was independent of gene expression and mediated by a small number of kinase families. The changes in the phosphoproteome affected a diverse range of cellular processes and functional analysis suggested that this response induced an immediate reactive state and prepared cells for subsequent transcriptional responses. CONCLUSIONS Our studies reveal a hitherto unappreciated role for changes in the protein phosphorylation landscape in cellular responses to IFN-I and thus provide insights for novel diagnostic and therapeutic strategies for neurological diseases caused by IFN-I.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Melanie Y White
- School of Life and Environmental Sciences, School of Medical Sciences, Charles Perkins Centre and Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Stuart J Cordwell
- School of Life and Environmental Sciences, School of Medical Sciences, Charles Perkins Centre and Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
8
|
Rodriguez S, Sahin A, Schrank BR, Al-Lawati H, Costantino I, Benz E, Fard D, Albers AD, Cao L, Gomez AC, Evans K, Ratti E, Cudkowicz M, Frosch MP, Talkowski M, Sorger PK, Hyman BT, Albers MW. Genome-encoded cytoplasmic double-stranded RNAs, found in C9ORF72 ALS-FTD brain, propagate neuronal loss. Sci Transl Med 2021; 13:eaaz4699. [PMID: 34233951 PMCID: PMC8779652 DOI: 10.1126/scitranslmed.aaz4699] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 06/29/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
Triggers of innate immune signaling in the CNS of patients with amyotrophic lateral sclerosis and frontotemporal degeneration (ALS/FTD) remain elusive. We report the presence of cytoplasmic double-stranded RNA (cdsRNA), an established trigger of innate immunity, in ALS-FTD brains carrying C9ORF72 intronic hexanucleotide expansions that included genomically encoded expansions of the G4C2 repeat sequences. The presence of cdsRNA in human brains was coincident with cytoplasmic TAR DNA binding protein 43 (TDP-43) inclusions, a pathologic hallmark of ALS/FTD. Introducing cdsRNA into cultured human neural cells induced type I interferon (IFN-I) signaling and death that was rescued by FDA-approved JAK inhibitors. In mice, genomically encoded dsRNAs expressed exclusively in a neuronal class induced IFN-I and death in connected neurons non-cell-autonomously. Our findings establish that genomically encoded cdsRNAs trigger sterile, viral-mimetic IFN-I induction and propagated death within neural circuits and may drive neuroinflammation and neurodegeneration in patients with ALS/FTD.
Collapse
Affiliation(s)
- Steven Rodriguez
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA 02115, USA
| | - Asli Sahin
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Benjamin R Schrank
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Hawra Al-Lawati
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Isabel Costantino
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Eric Benz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Darian Fard
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Alefiya D Albers
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
- Department of Psychology, Endicott College, Beverly, MA 01915, USA
| | - Luxiang Cao
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Alexis C Gomez
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Kyle Evans
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Ratti
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Michael Talkowski
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA 02115, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Wu W, Wu G, Cao D. Acteoside Presents Protective Effects on Cerebral Ischemia/reperfusion Injury Through Targeting CCL2, CXCL10, and ICAM1. Cell Biochem Biophys 2021; 79:301-310. [PMID: 33439460 DOI: 10.1007/s12013-020-00965-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2020] [Indexed: 11/26/2022]
Abstract
The objective of this study is to investigate the roles of acteoside (ACT) in cells with oxygen-glucose deprivation and reoxygenation (OGD/R)-induced injury and the underlying mechanisms. The differentially expressed genes (DEGs) in rats with middle cerebral artery occlusion were identified using GSE61616 data set. Kyoto Encyclopedia of Genes and Genomes pathway enrichment with the DEGs and the prediction of ACT's targets were conducted using The Comparative Toxicogenomics Database. The OGD/R model was established with bEnd.3 cells. Following that, bEnd.3 cells were treated by distinct concentrations of ACT and IL-10. The proliferation and apoptosis of cells were analyzed by cell counting kit-8 and flow cytometry assays, respectively. Western blot was used to check involved proteins. Herein, we identified CCL2, CXCL10, and ICAM1 as the targets of ACT, which were upregulated in tissues of MACO rats and cells with OGD/R-induced injury. ACT promoted the proliferation but reduce the apoptosis of cells with OGD/R-induced injury. Moreover, these effects of ACT were enhanced by IL-10. After being treated with ACT, IL-10, or ACT together with IL-10, the levels of CCL2, CXCL10, and ICAM1 were all decreased, whereas p-Stat3 was raised in cells with OGD/R-induced injury, while Stat3 expression presented no significant difference among groups. ACT protected cells against OGD/R-induced injury through regulating the IL-10/Stat3 signaling, indicating that ACT might be an effective therapy drug to lower cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Weijiang Wu
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, PR China
| | - Gang Wu
- Department of Neurology, Binzhou People's Hospital, Binzhou, Shandong Province, PR China
| | - Deyan Cao
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, PR China.
| |
Collapse
|
10
|
Huang W, Cho KY, Meng D, Walker WA. The impact of indole-3-lactic acid on immature intestinal innate immunity and development: a transcriptomic analysis. Sci Rep 2021; 11:8088. [PMID: 33850185 PMCID: PMC8044159 DOI: 10.1038/s41598-021-87353-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/23/2021] [Indexed: 02/02/2023] Open
Abstract
An excessive intestinal inflammatory response may have a role in the pathogenesis of necrotizing enterocolitis (NEC) in very preterm infants. Indole-3-lactic acid (ILA) of breastmilk tryptophan was identified as the anti-inflammatory metabolite involved in probiotic conditioned media from Bifidobacteria longum subsp infantis. This study aimed to explore the molecular endocytic pathways involved in the protective ILA effect against inflammation. H4 cells, Caco-2 cells, C57BL/6 pup and adult mice were used to compare the anti-inflammatory mechanisms between immature and mature enterocytes in vitro and in vivo. The results show that ILA has pleiotropic protective effects on immature enterocytes including anti-inflammatory, anti-viral, and developmental regulatory potentials in a region-dependent and an age-dependent manner. Quantitative transcriptomic analysis revealed a new mechanistic model in which STAT1 pathways play an important role in IL-1β-induced inflammation and ILA has a regulatory effect on STAT1 pathways. These studies were validated by real-time RT-qPCR and STAT1 inhibitor experiments. Different protective reactions of ILA between immature and mature enterocytes indicated that ILA's effects are developmentally regulated. These findings may be helpful in preventing NEC for premature infants.
Collapse
Affiliation(s)
- Wuyang Huang
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing, People's Republic of China
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Harvard Medical School, 16th Street Building (114-3503), Charlestown, MA, 02129, USA
| | - Ky Young Cho
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Harvard Medical School, 16th Street Building (114-3503), Charlestown, MA, 02129, USA
| | - Di Meng
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Harvard Medical School, 16th Street Building (114-3503), Charlestown, MA, 02129, USA
| | - W Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Harvard Medical School, 16th Street Building (114-3503), Charlestown, MA, 02129, USA.
| |
Collapse
|
11
|
Davies KA, Cooper E, Voon V, Tibble J, Cercignani M, Harrison NA. Interferon and anti-TNF therapies differentially modulate amygdala reactivity which predicts associated bidirectional changes in depressive symptoms. Mol Psychiatry 2021; 26:5150-5160. [PMID: 32457424 PMCID: PMC8589643 DOI: 10.1038/s41380-020-0790-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
A third of patients receiving Interferon-α (IFN-α) treatment for Hepatitis-C develop major depressive disorder (MDD). Conversely, anti-Tumor Necrosis Factor (TNF) therapies improve depression providing key empirical support for the "inflammatory theory" of depression. Heightened amygdala reactivity (particularly to negatively valanced stimuli) is a consistent finding within MDD; can predict treatment efficacy and reverses following successful treatment. However, whether IFN-α and anti-TNF enhance/attenuate depressive symptoms through modulation of amygdala emotional reactivity is unknown. Utilizing a prospective study design, we recruited 30 patients (mean 48.0 ± 10.5 years, 21 male) initiating IFN-α treatment for Hepatitis-C and 30 (mean 50.4 ± 15.7 years, 10 male) anti-TNF therapy for inflammatory arthritis. All completed an emotional face-processing task during fMRI and blood sampling before and after their first IFN-α (4-h) or anti-TNF (24-h) injection and follow-up psychiatric assessments for 3 months of treatment. IFN-α significantly increased depression symptoms (Hamilton Depression Rating Scale HAM-D) at 4 weeks (p < 0.001) but not 4-h after first dose (p > 0.1). Conversely, anti-TNF significantly improved depressive symptoms (Hospital Anxiety and Depression Rating Scale HADS) at both 24-h (P = 0.015) and 12 weeks (p = 0.018). In support of our a-priori hypothesis, both IFN-α and anti-TNF significantly modulated amygdala reactivity with IFN-α acutely enhancing right amygdala responses to sad (compared with neutral) faces (p = 0.032) and anti-TNF conversely decreasing right amygdala reactivity (across emotional valence) (p = 0.033). Furthermore, these changes predicted IFN-induced increases in HAM-D 4 weeks later (R2 = 0.17, p = 0.022) and anti-TNF-associated decreases in HADS at 24-h (R2 = 0.23, p = 0.01) suggesting that actions of systemic inflammation on amygdala emotional reactivity play a mechanistic role in inflammation-associated depressive symptoms.
Collapse
Affiliation(s)
- Kevin A. Davies
- grid.12082.390000 0004 1936 7590Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex Campus, Brighton, BN1 9RY UK ,grid.511096.aDepartment of Rheumatology, Brighton & Sussex University Hospitals, Brighton, UK
| | - Ella Cooper
- grid.12082.390000 0004 1936 7590Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex Campus, Brighton, BN1 9RY UK
| | - Valerie Voon
- grid.5335.00000000121885934Department of Psychiatry, University of Cambridge, Cambridge, CB2 0QQ UK
| | - Jeremy Tibble
- grid.511096.aDepartment of Hepatology, Brighton & Sussex University Hospitals, Brighton, UK
| | - Mara Cercignani
- grid.12082.390000 0004 1936 7590Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex Campus, Brighton, BN1 9RY UK
| | - Neil A. Harrison
- grid.12082.390000 0004 1936 7590Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex Campus, Brighton, BN1 9RY UK ,grid.5600.30000 0001 0807 5670Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, CF24 4HQ UK
| |
Collapse
|
12
|
Roy ER, Wang B, Wan YW, Chiu G, Cole A, Yin Z, Propson NE, Xu Y, Jankowsky JL, Liu Z, Lee VMY, Trojanowski JQ, Ginsberg SD, Butovsky O, Zheng H, Cao W. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J Clin Invest 2020; 130:1912-1930. [PMID: 31917687 PMCID: PMC7108898 DOI: 10.1172/jci133737] [Citation(s) in RCA: 337] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/03/2020] [Indexed: 12/18/2022] Open
Abstract
Type I interferon (IFN) is a key cytokine that curbs viral infection and cell malignancy. Previously, we demonstrated a potent IFN immunogenicity of nucleic acid-containing (NA-containing) amyloid fibrils in the periphery. Here, we investigated whether IFN is associated with β-amyloidosis inside the brain and contributes to neuropathology. An IFN-stimulated gene (ISG) signature was detected in the brains of multiple murine Alzheimer disease (AD) models, a phenomenon also observed in WT mouse brain challenged with generic NA-containing amyloid fibrils. In vitro, microglia innately responded to NA-containing amyloid fibrils. In AD models, activated ISG-expressing microglia exclusively surrounded NA+ amyloid β plaques, which accumulated in an age-dependent manner. Brain administration of rIFN-β resulted in microglial activation and complement C3-dependent synapse elimination in vivo. Conversely, selective IFN receptor blockade effectively diminished the ongoing microgliosis and synapse loss in AD models. Moreover, we detected activated ISG-expressing microglia enveloping NA-containing neuritic plaques in postmortem brains of patients with AD. Gene expression interrogation revealed that IFN pathway was grossly upregulated in clinical AD and significantly correlated with disease severity and complement activation. Therefore, IFN constitutes a pivotal element within the neuroinflammatory network of AD and critically contributes to neuropathogenic processes.
Collapse
Affiliation(s)
- Ethan R. Roy
- Huffington Center on Aging
- Translational Biology & Molecular Medicine Program, and
| | | | - Ying-wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - Zhuoran Yin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas E. Propson
- Huffington Center on Aging
- Molecular and Cellular Biology Program, Department of Molecular and Cellular Biology
| | - Yin Xu
- Huffington Center on Aging
| | | | - Zhandong Liu
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - Virginia M.-Y. Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, USA
- Departments of Psychiatry, Neuroscience & Physiology and the NYU Neuroscience Institute, New York University Langone Medical Center, New York, New York, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Zheng
- Huffington Center on Aging
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Wei Cao
- Huffington Center on Aging
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
13
|
Li HL, Huang Y, Zhou YL, Teng RH, Zhou SZ, Lin JP, Yang Y, Zhu SM, Xu H, Yao YX. C-X-C Motif Chemokine 10 Contributes to the Development of Neuropathic Pain by Increasing the Permeability of the Blood-Spinal Cord Barrier. Front Immunol 2020; 11:477. [PMID: 32265928 PMCID: PMC7098954 DOI: 10.3389/fimmu.2020.00477] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/02/2020] [Indexed: 01/10/2023] Open
Abstract
Neuropathic pain is among the most debilitating forms of chronic pain. Studies have suggested that chronic pain pathogenesis involves neuroimmune interactions and blood-spinal cord barrier (BSCB) disruption. However, the underlying mechanisms are poorly understood. We modeled neuropathic pain in rats by inducing chronic constriction injury (CCI) of the sciatic nerve and analyzed the effects on C-X-C motif chemokine 10 (CXCL10)/CXCR3 activation, BSCB permeability, and immune cell migration from the circulation into the spinal cord. We detected CXCR3 expression in spinal neurons and observed that CCI induced CXCL10/CXCR3 activation, BSCB disruption, and mechanical hyperalgesia. CCI-induced BSCB disruption enabled circulating T cells to migrate into the spinal parenchyma. Intrathecal administration of an anti-CXCL10 antibody not only attenuated CCI-induced hyperalgesia, but also reduced BSCB permeability, suggesting that CXCL10 acts as a key regulator of BSCB integrity. Moreover, T cell migration may play a critical role in the neuroimmune interactions involved in the pathogenesis of CCI-induced neuropathic pain. Our results highlight CXCL10 as a new potential drug target for the treatment of nerve injury-induced neuropathic pain.
Collapse
Affiliation(s)
- Hao-Ling Li
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan Huang
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Anesthesia, The Central Hospital of Lishui City, Lishui, China
| | - Ya-Lan Zhou
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Run-Hua Teng
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-Zhuan Zhou
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jia-Piao Lin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Yang
- Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng-Mei Zhu
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Xu
- Department of Anesthesia, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Xing Yao
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Immunopathology in the brain of mice following vertical transmission of Coxsackievirus B4. Microb Pathog 2020; 140:103965. [PMID: 31904449 DOI: 10.1016/j.micpath.2020.103965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022]
Abstract
Coxsackie B viruses (CV-B) are associated with several central nervous system (CNS) disorders. These viruses are predominantly transmitted by fecal-oral route but vertical transmission can also occur. This work attempted to study the immune response ensuing vertical transmission of CV-B to the brain, and its eventual implementation in the brain pathogenesis. To this end, pregnant Swiss albino mice were inoculated with CV-B4 E2 or with sterile medium for control animals. At different ages after birth, brains were collected and analyzed for virus infection, histopathological changes and immune response. Infectious particles were detected in offspring's brain which demonstrates vertical transmission of the virus. This infection is persistent since the long lasting detection of viral RNA in offspring's brain. Some pathological signs including meningitis, edema and accumulation of inflammatory cells within and surrounding the inflammatory areas were observed. Immunoflorescence staining unveiled the presence of T lymphocytes and microgliosis in the sites of lesion for a long period after birth. Multiplex cytokines measurement upon supernatants of in vitro mixed brain cells and extracted mononuclear cells from offspring's brain has demonstrated an elevated secretion of the pro-inflammatory cytokines TNFα, IL-6 and IFNα and the chemokines RANTES and MCP-1. Hence, vertical transmission of CV-B4 and its persistence within offspring's brain can lead to pathological features linked to increased and sustained immune response.
Collapse
|
15
|
West PK, Viengkhou B, Campbell IL, Hofer MJ. Microglia responses to interleukin-6 and type I interferons in neuroinflammatory disease. Glia 2019; 67:1821-1841. [PMID: 31033014 DOI: 10.1002/glia.23634] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2025]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They are a heterogenous, exquisitely responsive, and highly plastic cell population, which enables them to perform diverse roles. They sense and respond to the local production of many different signals, including an assorted range of cytokines. Microglia respond strongly to interleukin-6 (IL-6) and members of the type I interferon (IFN-I) family, IFN-alpha (IFN-α), and IFN-beta (IFN-β). Although these cytokines are essential in maintaining homeostasis and for activating and regulating immune responses, their chronic production has been linked to the development of distinct human neurological diseases, termed "cerebral cytokinopathies." IL-6 and IFN-α have been identified as key mediators in the pathogenesis of neuroinflammatory disorders including neuromyelitis optica and Aicardi-Goutières syndrome, respectively, whereas IFN-β has an emerging role as a causal factor in age-associated cognitive decline. One of the key features that unites these diseases is the presence of highly reactive microglia. The current understanding is that microglia contribute to the development of cerebral cytokinopathies and represent an important therapeutic target. However, it remains to be resolved whether microglia have beneficial or detrimental effects. Here we review and discuss what is currently known about the microglial response to IL-6 and IFN-I, based on both animal models and clinical studies. Foundational knowledge regarding the microglial response to IL-6 and IFN-I is now being used to devise therapeutic strategies to ameliorate neuroinflammation and promote repair: either through targeting microglia, or by targeting the reduction of CNS levels or downstream biological pathways of IL-6 or IFN-I.
Collapse
Affiliation(s)
- Phillip K West
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Barney Viengkhou
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Zeng J, Meng X, Zhou P, Yin Z, Xie Q, Zou H, Shen N, Ye Z, Tang Y. Interferon-α exacerbates neuropsychiatric phenotypes in lupus-prone mice. Arthritis Res Ther 2019; 21:205. [PMID: 31481114 PMCID: PMC6724270 DOI: 10.1186/s13075-019-1985-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022] Open
Abstract
Background Neuropsychiatric systemic lupus erythaematosus (NP-SLE) is one of the major manifestations of lupus. However, the mechanisms involved in NP-SLE are still largely unknown. The abnormal activation of the type I IFN signalling pathway is involved in SLE pathogenesis and is linked to NP-SLE, but the effect of IFN-α on NP-SLE encephalopathy has not been systematically studied. Methods An intravenous injection of Adv-IFN-α (10 mice, 10 × 109 vp) was administered to the IFN-α-treated group, and Adv-ctrl (10 mice, 10 × 109 vp) (ViGene Biosciences, China) was administered to the control group. Gene expression was determined by real-time quantitative polymerase chain reaction (RT-qPCR). Enzyme-linked immunosorbent assay (ELISA) was used to detect antibodies in the serum, and urinary protein levels were measured with a BCA Protein Assay kit. Haematoxylin-eosin (H&E) and periodic acid-Schiff (PAS)-light green staining were used for kidney histology. The elevated plus-maze test, novelty-suppressed feeding assay, open-field test, tail suspension test, social dominance tube test, three-chamber social interaction test, step-down passive avoidance test and novelty Y-maze task were used to assess behaviour. Results In this study, we performed a series of behavioural tests to assess the neuropsychiatric phenotypes of IFN-α-treated NZB/NZW F1 mice and found that these mice developed a series of mental disorders such as anxiety-like phenotypes, depression-like phenotypes, deficits in sociability and cognitive impairments, which mimic the neuropsychiatric manifestations of NP-SLE, with a consistent onset and progression. Conclusions Our research verified that IFN-α plays a critical role in NP-SLE and provides a comprehensive NP-SLE mouse model for dissecting the mechanisms of NP-SLE and developing novel therapies for intervention. Electronic supplementary material The online version of this article (10.1186/s13075-019-1985-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Zeng
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai, China
| | - Xinyu Meng
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai, China
| | - Ping Zhou
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, 22 Nong Lin Road, Shenzhen, China
| | - Qinglian Xie
- Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, China
| | - Hong Zou
- Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai, China. .,Shenzhen Futian Hospital for Rheumatic Diseases, 22 Nong Lin Road, Shenzhen, China. .,Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, China. .,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, 2200 Lane 25 Xietu Road, Shanghai, China. .,Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China. .,Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, USA.
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, 22 Nong Lin Road, Shenzhen, China.
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shan Dong Road (c), Shanghai, China. .,Shenzhen Futian Hospital for Rheumatic Diseases, 22 Nong Lin Road, Shenzhen, China.
| |
Collapse
|
17
|
Li W, Viengkhou B, Denyer G, West PK, Campbell IL, Hofer MJ. Microglia have a more extensive and divergent response to interferon-α compared with astrocytes. Glia 2018; 66:2058-2078. [PMID: 30051922 DOI: 10.1002/glia.23460] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022]
Abstract
Type I interferons (IFN-I) are crucial for effective antimicrobial defense in the central nervous system (CNS) but also can cause severe neurological disease (termed cerebral interferonopathy) as exemplified by Aicardi-Goutières Syndrome. In the CNS, microglia and astrocytes have essential roles in host responses to infection and injury, with both cell types responding to IFN-I. While the IFN-I signaling pathways are the same in astrocytes and microglia, the extent to which the IFN-I responses of these cells differ, if at all, is unknown. Here we determined the global transcriptional responses of astrocytes and microglia to the IFN-I, IFN-α. We found that under basal conditions, each cell type has a unique gene expression pattern reflective of its developmental origin and biological function. Following stimulation with IFN-α, astrocytes and microglia also displayed a common core response that was characterized by the increased expression of genes required for pathogen detection and elimination. Compared with astrocytes, microglia had a more extensive and diverse response to IFN-α with significantly more genes with expression upregulated (282 vs. 141) and downregulated (81 vs. 3). Further validation was documented for selected IFN-I-regulated genes in a murine model of cerebral interferonopathy. In all, the findings highlight not only overlapping but importantly divergent responses to IFN-I by astrocytes versus microglia. This suggests specialized roles for these cells in host defense and in the development of cerebral interferonopathy.
Collapse
Affiliation(s)
- Wen Li
- The University of Sydney, School of Molecular Bioscience, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Charles Perkins Centre, and the Bosch Institute, Sydney, Australia
| | - Barney Viengkhou
- The University of Sydney, School of Molecular Bioscience, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Charles Perkins Centre, and the Bosch Institute, Sydney, Australia.,The University of Sydney, School of Life and Environmental Sciences, Sydney, Australia
| | - Gareth Denyer
- The University of Sydney, School of Life and Environmental Sciences, Sydney, Australia
| | - Phillip K West
- The University of Sydney, School of Molecular Bioscience, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Charles Perkins Centre, and the Bosch Institute, Sydney, Australia.,The University of Sydney, School of Life and Environmental Sciences, Sydney, Australia
| | - Iain L Campbell
- The University of Sydney, School of Molecular Bioscience, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Charles Perkins Centre, and the Bosch Institute, Sydney, Australia
| | - Markus J Hofer
- The University of Sydney, School of Molecular Bioscience, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Charles Perkins Centre, and the Bosch Institute, Sydney, Australia.,The University of Sydney, School of Life and Environmental Sciences, Sydney, Australia
| |
Collapse
|
18
|
Savitz J, Harrison NA. Interoception and Inflammation in Psychiatric Disorders. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 3:514-524. [PMID: 29884282 PMCID: PMC5995132 DOI: 10.1016/j.bpsc.2017.12.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/28/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
Abstract
Despite a historical focus on neurally mediated interoceptive signaling mechanisms, humoral (and even cellular) signals also play an important role in communicating bodily physiological state to the brain. These signaling pathways can perturb neuronal structure, chemistry, and function, leading to discrete changes in behavior. They are also increasingly implicated in the pathophysiology of psychiatric disorders. The importance of these humoral signaling pathways is perhaps most powerfully illustrated in the context of infection and inflammation. Here we provide an overview of how interaction of immune activation of neural and humoral interoceptive mechanisms mediates discrete changes in brain and behavior and highlight how activation of these pathways at specific points in neural development may predispose to psychiatric disorder. As our mechanistic understanding of these interoceptive pathways continues to emerge, it is revealing novel therapeutic targets, potentially heralding an exciting new era of immunotherapies in psychiatry.
Collapse
Affiliation(s)
- Jonathan Savitz
- Laureate Institute for Brain Research, the University of Tulsa, Tulsa, Oklahoma; Oxley College of Health Sciences, the University of Tulsa, Tulsa, Oklahoma
| | - Neil A Harrison
- Clinical Imaging Sciences Centre, Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom; Sackler Centre for Consciousness Science, University of Sussex, Brighton, United Kingdom; Sussex Partnership NHS Foundation Trust, Brighton, United Kingdom.
| |
Collapse
|
19
|
Dowell NG, Bouyagoub S, Tibble J, Voon V, Cercignani M, Harrison NA. Interferon-alpha-Induced Changes in NODDI Predispose to the Development of Fatigue. Neuroscience 2017; 403:111-117. [PMID: 29292074 PMCID: PMC6458994 DOI: 10.1016/j.neuroscience.2017.12.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
NODDI, was used to probe subtle changes to tissue microstructure associated with IFN-α. We found a strong correlation between changes in neurite density index with acute and long-term fatigue following IFN-α. This observation confirms that the striatum is a key brain area targeted by IFN with implications for impaired motivation.
Interferon-alpha (IFN-α) is an important mediator of antiviral immune responses. It is also used clinically in the treatment of hepatitis-C infection. Though effective, IFN-α-based therapies can often impair mood, motivation and cognition, which when severe can appear indistinguishable from major depression. In susceptible patients, fatigue and motivational impairment emerge early and have been linked to changes in basal ganglia (striatal) metabolism, neurochemistry and microstructural integrity. Here we use neurite orientation dispersion and density imaging (NODDI) modeling of multi-shell diffusion MRI to investigate whether changes in orientation-dispersion index (ODI) or neurite density index (NDI) can predict the later emergence of IFN-α-induced fatigue. Eighteen patients initiating IFN-α-based treatment for hepatitis-C underwent diffusion MRI and blood sampling at baseline and 4 h after their first IFN-α injection. They were then followed up with regular psychological assessments for 12 weeks of treatment. IFN-α injection stimulated an acute inflammatory cytokine response and evoked acute fatigue that peaked between 4 and 12 weeks of treatment. Within the brain, IFN-α induced an acute increase in NDI in patients that experienced a simultaneous increase in IFN-α-induced fatigue but not in patients that did not. Acute changes in striatal microstructure additionally predicted the continued development of fatigue but not mood symptoms 4 and 8 weeks later into treatment. Our findings highlight the value of NODDI as a potential in vivo biomarker of the central effects of peripheral inflammation. We highlight the exquisite sensitivity of the striatum to IFN-α and further implicate striatal perturbation in IFN-α-induced fatigue.
Collapse
Affiliation(s)
- N G Dowell
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RR, UK.
| | - S Bouyagoub
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RR, UK
| | - J Tibble
- Department of Gastroenterology, Brighton & Sussex University Hospitals, Brighton, UK
| | - V Voon
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0QQ, UK
| | - M Cercignani
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RR, UK; Neuroimaging Laboratory, Santa Lucia Foundation, Rome, Italy
| | - N A Harrison
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RR, UK; Sackler Centre for Consciousness Science, University of Sussex, Falmer BN1 9RR, UK; Sussex Partnership NHS Foundation Trust, Brighton, UK
| |
Collapse
|
20
|
Borsini A, Cattaneo A, Malpighi C, Thuret S, Harrison NA, MRC ImmunoPsychiatry Consortium, Zunszain PA, Pariante CM. Interferon-Alpha Reduces Human Hippocampal Neurogenesis and Increases Apoptosis via Activation of Distinct STAT1-Dependent Mechanisms. Int J Neuropsychopharmacol 2017; 21:187-200. [PMID: 29040650 PMCID: PMC5793815 DOI: 10.1093/ijnp/pyx083] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In humans, interferon-α treatment for chronic viral hepatitis is a well-recognized clinical model for inflammation-induced depression, but the molecular mechanisms underlying these effects are not clear. Following peripheral administration in rodents, interferon-α induces signal transducer and activator of transcription-1 (STAT1) within the hippocampus and disrupts hippocampal neurogenesis. METHODS We used the human hippocampal progenitor cell line HPC0A07/03C to evaluate the effects of 2 concentrations of interferon-α, similar to those observed in human serum during its therapeutic use (500 pg/mL and 5000 pg/mL), on neurogenesis and apoptosis. RESULTS Both concentrations of interferon-α decreased hippocampal neurogenesis, with the high concentration also increasing apoptosis. Moreover, interferon-α increased the expression of interferon-stimulated gene 15 (ISG15), ubiquitin-specific peptidase 18 (USP18), and interleukin-6 (IL-6) via activation of STAT1. Like interferon-α, co-treatment with a combination of ISG15, USP18, and IL-6 was able to reduce neurogenesis and enhance apoptosis via further downstream activation of STAT1. Further experiments showed that ISG15 and USP18 mediated the interferon-α-induced reduction in neurogenesis (potentially through upregulation of the ISGylation-related proteins UBA7, UBE2L6, and HERC5), while IL-6 mediated the interferon-α-induced increase in apoptosis (potentially through downregulation of aquaporin 4). Using transcriptomic analyses, we showed that interferon-α regulated pathways involved in oxidative stress and immune response (e.g., Nuclear Factor (erythroid-derived 2)-like 2 [Nrf2] and interferon regulatory factor [IRF] signaling pathway), neuronal formation (e.g., CAMP response element-binding protein [CREB] signaling), and cell death regulation (e.g., tumor protein(p)53 signaling). CONCLUSIONS We identify novel molecular mechanisms mediating the effects of interferon-α on the human hippocampus potentially involved in inflammation-induced neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Alessandra Borsini
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, United Kingdom,Correspondence: Alessandra Borsini, PhD, Stress, Psychiatry and Immunology Lab and Perinatal Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King’s College London, Cutcombe Road, London, SE5 9RT ()
| | - Annamaria Cattaneo
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| | - Chiara Malpighi
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| | - Sandrine Thuret
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, United Kingdom
| | - Neil A Harrison
- University of Sussex, Department of Neuroscience, Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Patricia A Zunszain
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom
| | - Carmine M Pariante
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| |
Collapse
|
21
|
Type-I interferon pathway in neuroinflammation and neurodegeneration: focus on Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:797-807. [PMID: 28676934 DOI: 10.1007/s00702-017-1745-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/09/2017] [Indexed: 12/18/2022]
Abstract
Past research in Alzheimer's disease (AD) has largely been driven by the amyloid hypothesis; the accompanying neuroinflammation seen in AD has been assumed to be consequential and not disease modifying or causative. However, recent data from both clinical and preclinical studies have established that the immune-driven neuroinflammation contributes to AD pathology. Key evidence for the involvement of neuroinflammation in AD includes enhanced microglial and astroglial activation in the brains of AD patients, increased pro-inflammatory cytokine burden in AD brains, and epidemiological evidence that chronic non-steroidal anti-inflammatory drug use prior to disease onset leads to a lower incidence of AD. Identifying critical mediators controlling this neuroinflammation will prove beneficial in developing anti-inflammatory therapies for the treatment of AD. The type-I interferons (IFNs) are pleiotropic cytokines that control pro-inflammatory cytokine secretion and are master regulators of the innate immune response that impact on disorders of the central nervous system. This review provides evidence that the type-I IFNs play a critical role in the exacerbation of neuroinflammation and actively contribute to the progression of AD.
Collapse
|
22
|
Eising E, Shyti R, 't Hoen PAC, Vijfhuizen LS, Huisman SMH, Broos LAM, Mahfouz A, Reinders MJT, Ferrari MD, Tolner EA, de Vries B, van den Maagdenberg AMJM. Cortical Spreading Depression Causes Unique Dysregulation of Inflammatory Pathways in a Transgenic Mouse Model of Migraine. Mol Neurobiol 2017; 54:2986-2996. [PMID: 27032388 PMCID: PMC5390001 DOI: 10.1007/s12035-015-9681-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/23/2015] [Indexed: 01/03/2023]
Abstract
Familial hemiplegic migraine type 1 (FHM1) is a rare monogenic subtype of migraine with aura caused by mutations in CACNA1A that encodes the α1A subunit of voltage-gated CaV2.1 calcium channels. Transgenic knock-in mice that carry the human FHM1 R192Q missense mutation ('FHM1 R192Q mice') exhibit an increased susceptibility to cortical spreading depression (CSD), the mechanism underlying migraine aura. Here, we analysed gene expression profiles from isolated cortical tissue of FHM1 R192Q mice 24 h after experimentally induced CSD in order to identify molecular pathways affected by CSD. Gene expression profiles were generated using deep serial analysis of gene expression sequencing. Our data reveal a signature of inflammatory signalling upon CSD in the cortex of both mutant and wild-type mice. However, only in the brains of FHM1 R192Q mice specific genes are up-regulated in response to CSD that are implicated in interferon-related inflammatory signalling. Our findings show that CSD modulates inflammatory processes in both wild-type and mutant brains, but that an additional unique inflammatory signature becomes expressed after CSD in a relevant mouse model of migraine.
Collapse
Affiliation(s)
- Else Eising
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Reinald Shyti
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisanne S Vijfhuizen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd M H Huisman
- Department of Intelligent Systems, Faculty of EEMCS, Delft University of Technology, Delft, Netherlands
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Ludo A M Broos
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ahmed Mahfouz
- Department of Intelligent Systems, Faculty of EEMCS, Delft University of Technology, Delft, Netherlands
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Marcel J T Reinders
- Department of Intelligent Systems, Faculty of EEMCS, Delft University of Technology, Delft, Netherlands
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Else A Tolner
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Boukje de Vries
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
23
|
Li W, Hofer MJ, Songkhunawej P, Jung SR, Hancock D, Denyer G, Campbell IL. Type I interferon-regulated gene expression and signaling in murine mixed glial cells lacking signal transducers and activators of transcription 1 or 2 or interferon regulatory factor 9. J Biol Chem 2017; 292:5845-5859. [PMID: 28213522 DOI: 10.1074/jbc.m116.756510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/23/2017] [Indexed: 01/12/2023] Open
Abstract
Type I interferons (IFN-I) are critical in antimicrobial and antitumor defense. Although IFN-I signal via the interferon-stimulated gene factor 3 (ISGF3) complex consisting of STAT1, STAT2, and IRF9, IFN-I can mediate significant biological effects via ISGF3-independent pathways. For example, the absence of STAT1, STAT2, or IRF9 exacerbates neurological disease in transgenic mice with CNS production of IFN-I. Here we determined the role of IFN-I-driven, ISGF3-independent signaling in regulating global gene expression in STAT1-, STAT2-, or IRF9-deficient murine mixed glial cell cultures (MGCs). Compared with WT, the expression of IFN-α-stimulated genes (ISGs) was reduced in number and magnitude in MGCs that lacked STAT1, STAT2, or IRF9. There were significantly fewer ISGs in the absence of STAT1 or STAT2 versus in the absence of IRF9. The majority of ISGs regulated in the STAT1-, STAT2-, or IRF9-deficient MGCs individually were shared with WT. However, only a minor number of ISGs were common to WT and STAT1-, STAT2-, and IRF9-deficient MGCs. Whereas signal pathway activation in response to IFN-α was rapid and transient in WT MGCs, this was delayed and prolonged and correlated with increased numbers of ISGs expressed at 12 h versus 4 h of IFN-α exposure in all three IFN-I signaling-deficient MGCs. In conclusion, 1) IFN-I can mediate ISG expression in MGCs via ISGF3-independent signaling pathways but with reduced efficiency, with delayed and prolonged kinetics, and is more dependent on STAT1 and STAT2 than IRF9; and 2) signaling pathways not involving STAT1, STAT2, or IRF9 play a minor role only in mediating ISG expression in MGCs.
Collapse
Affiliation(s)
- Wen Li
- From the School of Molecular Bioscience
| | - Markus J Hofer
- From the School of Molecular Bioscience.,Bosch Institute, and.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | | | | | | | - Iain L Campbell
- From the School of Molecular Bioscience, .,Bosch Institute, and.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
24
|
Transcriptomics in Interferon-α-Treated Patients Identifies Inflammation-, Neuroplasticity- and Oxidative Stress-Related Signatures as Predictors and Correlates of Depression. Neuropsychopharmacology 2016; 41:2502-11. [PMID: 27067128 PMCID: PMC4983179 DOI: 10.1038/npp.2016.50] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/07/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022]
Abstract
Owing to the unique opportunity to assess individuals before and after they develop depression within a short timeframe, interferon-α (IFN-α) treatment for chronic hepatitis C virus (HCV) infection is an ideal model to identify molecular mechanisms relevant to major depression, especially in the context of enhanced inflammation. Fifty-eight patients were assessed prospectively, at baseline and monthly over 24 weeks of IFN-α treatment. New-onset cases of depression were determined using the Mini International Neuropsychiatric Interview (MINI). Whole-blood transcriptomic analyses were conducted to investigate the following: (1) baseline gene expression differences associated with future development of IFN-α-induced depression, before IFN-α, and (2) longitudinal gene expression changes from baseline to weeks 4 or 24 of IFN-α treatment, separately in those who did and did not develop depression. Transcriptomics data were analyzed using Partek Genomics Suite (1.4-fold, FDR adjusted p⩽0.05) and Ingenuity Pathway Analysis Software. Twenty patients (34%) developed IFN-α-induced depression. At baseline, 73 genes were differentially expressed in patients who later developed depression compared with those who did not. After 4 weeks of IFN-α treatment, 592 genes were modulated in the whole sample, representing primarily IFN-α-responsive genes. Substantially more genes were modulated only in patients who developed depression (n=506, compared with n=70 in patients who did not), with enrichment in inflammation-, neuroplasticity- and oxidative stress-related pathways. A similar picture was observed at week 24. Our data indicate that patients who develop IFN-α-induced depression have an increased biological sensitivity to IFN-α, as shown by larger gene expression changes, and specific signatures both as predictors and as correlates.
Collapse
|
25
|
Olianas MC, Dedoni S, Onali P. Protection from interferon-β-induced neuronal apoptosis through stimulation of muscarinic acetylcholine receptors coupled to ERK1/2 activation. Br J Pharmacol 2016; 173:2910-28. [PMID: 27474091 DOI: 10.1111/bph.13570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/30/2016] [Accepted: 07/20/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Although clinically useful for their immunomodulatory, antiproliferative and antiviral properties, type I interferons (IFNs) are involved in the pathogenesis of several neurodegenerative/neuroinflammatory diseases. In the present study, we investigated the ability of cholinergic stimulation to protect from IFN-β-induced neuronal apoptosis. EXPERIMENTAL APPROACH The effects of the ACh receptor agonist carbachol (CCh) on IFN-β-induced apoptosis of human SH-SY5Y neuroblastoma cells were examined by using western blots, immunofluorescence and cytofluorimetry. The involvement of muscarinic acetylcholine receptors (mAChRs) was assessed by using selective antagonists and siRNA transfection. Pharmacological inhibitors and overexpression of ERK2 and an ERK2 constitutively active form (ERK2-CA) were employed to study ERK1/2 signalling. The effects of oxotremorine-M (Oxo-M) on IFN-β-induced apoptosis of mouse hippocampal neurons were examined by measuring cleaved caspase 3 expression. KEY RESULTS In SH-SY5Y cells, CCh inhibited IFN-β-induced mitochondrial cytochrome c release, activation of caspases 9, 7 and 3, PARP cleavage and DNA fragmentation. The anti-apoptotic effect of CCh was mediated by M3 receptors, blocked by Gq/11 antagonist YM254890 and PKC inhibitor Go 6983, impaired by inhibition of ERK1/2 pathway, potentiated by overexpression of ERK2 and mimicked by ERK2-CA. Blockade of JNK activation enhanced the CCh anti-apoptotic response. IFN-β inhibited JNK activation and up-regulated CCh-induced ERK1/2 signalling. In hippocampal neurons, Oxo-M reduced IFN-β-induced apoptosis; this effect was antagonized by blockade of M1 /M3 receptors and ERK1/2. CONCLUSIONS AND IMPLICATIONS Stimulation of mAChRs counteracted IFN-β-induced neuronal apoptosis through the activation of ERK1/2 signalling. The data indicate that activation of ERK1/2-coupled mAChRs may be an effective strategy for preventing IFNs neurotoxicity.
Collapse
Affiliation(s)
- Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
26
|
Interferon-induced sterile alpha motif and histidine/aspartic acid domain-containing protein 1 expression in astrocytes and microglia is mediated by microRNA-181a. AIDS 2016; 30:2053-64. [PMID: 27219130 DOI: 10.1097/qad.0000000000001166] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1), a newly discovered HIV-1 host restriction factor, has been found to be induced by interferons and to be regulated by microRNA-181a (miR-181a). However, the mechanism of interferons-induced SAMHD1 expression is unclear. DESIGN We hypothesized that interferons induce SAMHD1 expression through Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathways, which is mediated by miR-181a. METHODS We examined the effect of IFN-α and IFN-γ on SAMHD1 mRNA and protein expression, as well as the levels of phosphorylated SAMHD1 and miR-181a in astrocytes and microglia. To determine whether interferons-induced SAMHD1 expression was mediated by miR-181a, we overexpressed or inhibited miR-181a in these cells and exposed them to interferons. We also detected the effect of SAMHD1 and miR-181a on HIV-1 infection in astrocytes and microglia. RESULTS Both IFN-α and IFN-γ increased SAMHD1 mRNA and protein expression, and reduced miR-181a levels, particularly in microglia. Phosphorylated SAMHD1was not induced by interferons. Overexpression of miR-181a counteracted induction of SAMHD1 expression by interferons, and inhibition of miR-181a mimicked interferons treatment. Inhibition of JAK-STAT signaling pathways resulted in increased miR-181a levels and decreased SAMHD1 mRNA expression. Knock-down of SAMHD1 or overexpression of miR-181a enhanced HIV-1 infection, whereas inhibition of miR-181a reduced HIV-1 infection. However, inhibition of HIV-1 infection induced by IFN-α was not significantly affected by miR-181a and SAMHD1. CONCLUSION MiR-181a is an important mediator for interferons-induced SAMHD1 expression in astrocytes and microglia, but not for inhibition of HIV-1 infection induced by IFN-α.
Collapse
|
27
|
Dowell NG, Cooper EA, Tibble J, Voon V, Critchley HD, Cercignani M, Harrison NA. Acute Changes in Striatal Microstructure Predict the Development of Interferon-Alpha Induced Fatigue. Biol Psychiatry 2016; 79:320-8. [PMID: 26169252 PMCID: PMC4725575 DOI: 10.1016/j.biopsych.2015.05.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/21/2015] [Accepted: 05/24/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND Interferon-alpha (IFN-α) is a key mediator of antiviral immune responses used clinically for hepatitis C treatment. Though effective, IFN-α induces marked behavioral changes that, when severe, can appear indistinguishable from major depression. Curiously, fatigue and motivational impairment evolve rapidly, suggesting acute engagement of immune-brain communicatory pathways, yet mood impairments typically emerge later, after weeks of treatment. Whether this reflects prolonged modulation of motivational processes underpinning fatigue or separate neurobiological mechanisms is currently unclear. METHODS Here, we used quantitative magnetization transfer (qMT) imaging, an advanced microstructural neuroimaging technique sensitive to effects of inflammation, in a prospective study design to measure acute brain changes to IFN-α and relate these to later development of discrete behavioral changes. Twenty-three patients initiating IFN-α treatment for hepatitis C underwent qMT imaging and blood sampling at baseline and 4 hours after their first IFN-α injection. Comprehensive behavioral and psychological assessments were completed at both scanning sessions and at treatment weeks 4, 8, 12, and 24. RESULTS IFN-α injection stimulated an acute inflammatory cytokine response and evoked fatigue that peaked between 4 and 12 weeks, preceding mood change by 4 weeks. In the brain, IFN-α induced an acute change in striatal microstructure that additionally predicted development of fatigue but not mood symptoms. CONCLUSIONS Our findings highlight qMT as an in vivo biomarker of central effects of peripheral inflammation. We demonstrate exquisite sensitivity of the striatum to IFN-α, implicate striatal perturbation in IFN-α-induced fatigue, and dissociate this from mechanisms underlying IFN-α-induced mood symptoms, providing empirical support for distinct neural substrates mediating actions on motivation and mood.
Collapse
Affiliation(s)
| | - Ella A Cooper
- Brighton and Sussex Medical School, University of Sussex; Department of Gastroenterology, Brighton & Sussex University Hospitals, Brighton
| | - Jeremy Tibble
- Department of Gastroenterology, Brighton & Sussex University Hospitals, Brighton
| | - Valerie Voon
- Department of Psychiatry, University of Cambridge; Cambridge and Peterborough National Health Service Foundation Trust, Cambridge
| | - Hugo D Critchley
- Brighton and Sussex Medical School, University of Sussex; Sackler Centre for Consciousness Science, University of Sussex, Falmer; Sussex Partnership National Health Service Trust, Brighton, United Kingdom
| | - Mara Cercignani
- Brighton and Sussex Medical School, University of Sussex; Neuroimaging Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Neil A Harrison
- Brighton and Sussex Medical School, University of Sussex; Sackler Centre for Consciousness Science, University of Sussex, Falmer; Neuroimaging Laboratory, Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
28
|
Roberts WK, Blachère NE, Frank MO, Dousmanis A, Ransohoff RM, Darnell RB. A destructive feedback loop mediated by CXCL10 in central nervous system inflammatory disease. Ann Neurol 2015. [PMID: 26224283 PMCID: PMC4583819 DOI: 10.1002/ana.24494] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective Paraneoplastic neurologic disorders (PND) are autoimmune diseases associated with cancer and ectopic expression of a neuronal antigen in a peripheral tumor. Patients with PND harbor high‐titer antibodies and T cells in their serum and cerebrospinal fluid (CSF) that are specific to the tumor antigen, and treatment with the immunosuppressant FK506 (tacrolimus) decreases CSF white blood cell counts. The objective of this study was to determine the effect of FK506 on CSF chemokine levels in PND patients. Methods CSF samples before and after FK506 treatment were tested by multiplex assay for the presence of 27 cytokines. Follow‐up in vitro experiments aimed to determine whether T cells secrete CXCL10 in response to cognate antigen. Results Here we report that PND patients harbor high levels of the chemokine CXCL10 in their CSF. CXCL10 is a cytokine that recruits CXCR3+ cells such as activated T cells, and we found that FK506 treatment specifically decreased CSF CXCL10 from among 27 cytokines tested. In vitro, CXCL10 was only produced during antigen‐specific cognate interactions between T cells and antigen‐presenting cells (APCs) when interferon‐γ (IFNγ) receptors were present on the T cell. Interpretation These results support a model in which antigen‐specific T cell stimulation by PND APCs triggers IFNγ, followed by CXCL10 production and further lymphocyte recruitment, suggesting that treatments targeting T cells or CXCL10 in the central nervous system (CNS) may interrupt a destructive positive feedback loop present in CNS inflammation. Ann Neurol 2015;78:619–629
Collapse
Affiliation(s)
- Wendy K Roberts
- Laboratory of Molecular Neuro-oncology, Rockefeller University, New York, NY
| | - Nathalie E Blachère
- Laboratory of Molecular Neuro-oncology, Rockefeller University, New York, NY.,Howard Hughes Medical Institute, Rockefeller University, New York, NY
| | - Mayu O Frank
- Laboratory of Molecular Neuro-oncology, Rockefeller University, New York, NY
| | | | | | - Robert B Darnell
- Laboratory of Molecular Neuro-oncology, Rockefeller University, New York, NY.,Howard Hughes Medical Institute, Rockefeller University, New York, NY.,New York Genome Center, New York, NY
| |
Collapse
|
29
|
Rosato PC, Leib DA. Neurons versus herpes simplex virus: the innate immune interactions that contribute to a host-pathogen standoff. Future Virol 2015; 10:699-714. [PMID: 26213562 PMCID: PMC4508759 DOI: 10.2217/fvl.15.45] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Herpes simplex virus (HSV) is a prevalent neurotropic virus, which establishes lifelong latent infections in the neurons of sensory ganglia. Despite our long-standing knowledge that HSV predominately infects sensory neurons during its life cycle, little is known about the neuronal antiviral response to HSV infection. Recent studies show that while sensory neurons have impaired intrinsic immunity to HSV infection, paracrine IFN signaling can potentiate a potent antiviral response. Additionally, antiviral autophagy plays an important role in neuronal control of HSV infection. Here we review the literature of antiviral signaling and autophagy in neurons, the mechanisms by which HSV can counteract these responses, and postulate how these two pathways may synergize to mediate neuronal control of HSV infection and yet result in lifelong persistence of the virus.
Collapse
Affiliation(s)
- Pamela C Rosato
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - David A Leib
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
30
|
Nallar SC, Kalvakolanu DV. Interferons, signal transduction pathways, and the central nervous system. J Interferon Cytokine Res 2015; 34:559-76. [PMID: 25084173 DOI: 10.1089/jir.2014.0021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The interferon (IFN) family of cytokines participates in the development of innate and acquired immune defenses against various pathogens and pathogenic stimuli. Discovered originally as a proteinaceous substance secreted from virus-infected cells that afforded immunity to neighboring cells from virus infection, these cytokines are now implicated in various human pathologies, including control of tumor development, cell differentiation, and autoimmunity. It is now believed that the IFN system (IFN genes and the genes induced by them, and the factors that regulate these processes) is a generalized alarm of cellular stress, including DNA damage. IFNs exert both beneficial and deleterious effects on the central nervous system (CNS). Our knowledge of the IFN-regulated processes in the CNS is far from being clear. In this article, we reviewed the current understanding of IFN signal transduction pathways and gene products that might have potential relevance to diseases of the CNS.
Collapse
Affiliation(s)
- Shreeram C Nallar
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine , Baltimore, Maryland
| | | |
Collapse
|
31
|
Wollmann G, Paglino JC, Maloney PR, Ahmadi SA, van den Pol AN. Attenuation of vesicular stomatitis virus infection of brain using antiviral drugs and an adeno-associated virus-interferon vector. Virology 2014; 475:1-14. [PMID: 25462341 DOI: 10.1016/j.virol.2014.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/04/2014] [Accepted: 10/20/2014] [Indexed: 12/21/2022]
Abstract
Vesicular stomatitis virus (VSV) shows promise as a vaccine-vector and oncolytic virus. However, reports of neurotoxicity of VSV remain a concern. We compared 12 antiviral compounds to control infection of VSV-CT9-M51 and VSV-rp30 using murine and human brain cultures, and in vivo mouse models. Inhibition of replication, cytotoxicity and infectivity was strongest with ribavirin and IFN-α and to some extent with mycophenolic acid, chloroquine, and adenine 9-β-d-arabinofuranoside. To generate continuous IFN exposure, we made an adeno-associated virus vector expressing murine IFN; AAV-mIFN-β protected mouse brain cells from VSV, as did a combination of IFN, ribavirin and chloroquine. Intracranial AAV-mIFN-β protected the brain against VSV-CT9-M51. In SCID mice bearing human glioblastoma, AAV-mIFN-β moderately enhanced survival. VSV-CT9-M51 doubled median survival when administered after AAV-mIFN-β; some surviving mice showed complete tumor destruction. Together, these data suggest that AAV-IFN or IFN with ribavirin and chloroquine provide an optimal anti-virus combination against VSV in the brain.
Collapse
Affiliation(s)
- Guido Wollmann
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Justin C Paglino
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Patrick R Maloney
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Sebastian A Ahmadi
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Anthony N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
32
|
Comet NR, Aguiló JI, Rathoré MG, Catalán E, Garaude J, Uzé G, Naval J, Pardo J, Villalba M, Anel A. IFNα signaling through PKC-θ is essential for antitumor NK cell function. Oncoimmunology 2014; 3:e948705. [PMID: 25960930 DOI: 10.4161/21624011.2014.948705] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 06/06/2014] [Indexed: 11/19/2022] Open
Abstract
We have previously shown that the development of a major histocompatibility complex class I (MHC-I)-deficient tumor was favored in protein kinase C-θ knockout (PKC-θ-/-) mice compared to that occurring in wild-type mice. This phenomenon was associated with scarce recruitment of natural killer (NK) cells to the tumor site, as well as impaired NK cell activation and reduced cytotoxicity ex vivo. Poly-inosinic:cytidylic acid (poly I:C) treatment activated PKC-θ in NK cells depending on the presence of a soluble factor produced by a different splenocyte subset. In the present work, we sought to analyze whether interleukin-15 (IL-15) and/or interferon-α (IFNα) mediate PKC-θ-dependent antitumor NK cell function. We found that IL-15 improves NK cell viability, granzyme B expression, degranulation capacity and interferon-γ (IFNγ) secretion independently of PKC-θ. In contrast, we found that IFNα improves the degranulation capability of NK cells against target cancer cells in a PKC-θ-dependent fashion both ex vivo and in vivo. Furthermore, IFNα induces PKC-θ auto-phosphorylation in NK cells, in a signal transduction pathway involving both phosphatidylinositol-3-kinase (PI3K) and phospholipase-C (PLC) activation. PKC-θ dependence was further implicated in IFNα-induced transcriptional upregulation of chemokine (C-X-C motif) ligand 10 (CXCL10), a signal transducer and activator of transcription-1 (STAT-1)-dependent target of IFNα. The absence of PKC-θ did not affect IFNα-induced STAT-1 Tyr701 phosphorylation but affected the increase in STAT-1 phosphorylation on Ser727, attenuating CXCL10 secretion. This connection between IFNα and PKC-θ in NK cells may be exploited in NK cell-based tumor immunotherapy.
Collapse
Key Words
- CDK8, cyclin-dependent kinase 8
- CXCL10
- CXCL10, (C-X-C motif) ligand 10/CXCL10
- FCS, fetal calf serum
- IFN-α, IL-15
- IFNA1
- IFNα, interferon-α
- IFNγ, interferon-γ, IFNG
- IL-15, interleukin-15/IL15
- MACS, magnetic cell separation
- MEF, murine embryonic fibroblast
- MHC-I, major histocompability complex class I/MHC-I
- NK cells
- NK, natural killer
- PI3K, phosphatidylinositol-3-kinase
- PKC-θ
- PKC-θ, protein kinase C-θ, PRKCQ
- PLC, phospholipase-C
- Poly I:C, poly-inosinic:cytidilic acid
- RT-PCR, real-time polymerase chain reaction
- STAT-1, signal transducer and activator of transcription-1/STAT1.
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Natalia R Comet
- Apoptosis, Immunity & Cancer Group; Department of Biochemistry and Molecular and Cell Biology ; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Juan Ignacio Aguiló
- Apoptosis, Immunity & Cancer Group; Department of Biochemistry and Molecular and Cell Biology ; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Moeez G Rathoré
- INSERM U1040; Université de Montpellier 1; UFR Médecine ; Montpellier, France
| | - Elena Catalán
- Apoptosis, Immunity & Cancer Group; Department of Biochemistry and Molecular and Cell Biology ; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Johan Garaude
- INSERM U1040; Université de Montpellier 1; UFR Médecine ; Montpellier, France
| | - Gilles Uzé
- CNRS UMR 5235; Université de Montpellier II; Place Eugene Bataillon ; Montpellier, France
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group; Department of Biochemistry and Molecular and Cell Biology ; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Julián Pardo
- Immune Effector Cells Group; IIS Aragón; Biomedical Research Center of Aragón (CIBA); Nanoscience Institute of Aragon (INA); Zaragoza, Spain ; Aragón I+D Foundation (ARAID) ; Zaragoza, Spain
| | - Martín Villalba
- INSERM U1040; Université de Montpellier 1; UFR Médecine ; Montpellier, France ; Institut de Recherche en Biothérapie (IRB); CHU Montpellier ; Montpellier, France
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group; Department of Biochemistry and Molecular and Cell Biology ; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| |
Collapse
|
33
|
Taylor JM, Minter MR, Newman AG, Zhang M, Adlard PA, Crack PJ. Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer's disease. Neurobiol Aging 2014; 35:1012-23. [DOI: 10.1016/j.neurobiolaging.2013.10.089] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 10/02/2013] [Accepted: 10/25/2013] [Indexed: 11/26/2022]
|
34
|
Characterization of a murine neuron-enriched model of primary trigeminal ganglia cultures to study the interferon-β antiviral effect against Herpes Simplex Virus type 1. INFECTIO 2014. [DOI: 10.1016/j.infect.2014.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
35
|
Minter MR, Zhang M, Ates RC, Taylor JM, Crack PJ. Type-1 interferons contribute to oxygen glucose deprivation induced neuro-inflammation in BE(2)M17 human neuroblastoma cells. J Neuroinflammation 2014; 11:43. [PMID: 24602263 PMCID: PMC3995960 DOI: 10.1186/1742-2094-11-43] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/21/2014] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Hypoxic-ischaemic injuries such as stroke and traumatic brain injury exhibit features of a distinct neuro-inflammatory response in the hours and days post-injury. Microglial activation, elevated pro-inflammatory cytokines and macrophage infiltration contribute to core tissue damage and contribute to secondary injury within a region termed the penumbra. Type-1 interferons (IFNs) are a super-family of pleiotropic cytokines that regulate pro-inflammatory gene transcription via the classical Jak/Stat pathway; however their role in hypoxia-ischaemia and central nervous system neuro-inflammation remains unknown. Using an in vitro approach, this study investigated the role of type-1 IFN signalling in an inflammatory setting induced by oxygen glucose deprivation (OGD). METHODS Human BE(2)M17 neuroblastoma cells or cells expressing a type-1 interferon-α receptor 1 (IFNAR1) shRNA or negative control shRNA knockdown construct were subjected to 4.5 h OGD and a time-course reperfusion period (0 to 24 h). Q-PCR was used to evaluate IFNα, IFNβ, IL-1β, IL-6 and TNF-α cytokine expression levels. Phosphorylation of signal transducers and activators of transcription (STAT)-1, STAT-3 and cleavage of caspase-3 was detected by western blot analysis. Post-OGD cellular viability was measured using a MTT assay. RESULTS Elevated IFNα and IFNβ expression was detected during reperfusion post-OGD in parental M17 cells. This correlated with enhanced phosphorylation of STAT-1, a downstream type-1 IFN signalling mediator. Significantly, ablation of type-1 IFN signalling, through IFNAR1 knockdown, reduced IFNα, IFNβ, IL-6 and TNF-α expression in response to OGD. In addition, MTT assay confirmed the IFNAR1 knockdown cells were protected against OGD compared to negative control cells with reduced pro-apoptotic cleaved caspase-3 levels. CONCLUSIONS This study confirms a role for type-1 IFN signalling in the neuro-inflammatory response following OGD in vitro and suggests its modulation through therapeutic blockade of IFNAR1 may be beneficial in reducing hypoxia-induced neuro-inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Peter John Crack
- Department of Pharmacology, University of Melbourne, 8th floor, Medical building, Grattan St, Parkville 3010, VIC, Australia.
| |
Collapse
|
36
|
Inefficient type I interferon-mediated antiviral protection of primary mouse neurons is associated with the lack of apolipoprotein l9 expression. J Virol 2014; 88:3874-84. [PMID: 24453359 DOI: 10.1128/jvi.03018-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED We examined the antiviral response promoted by type I interferons (IFN) in primary mouse neurons. IFN treatment of neuron cultures strongly upregulated the transcription of IFN-stimulated genes but conferred a surprisingly low resistance to infection by neurotropic viruses such as Theiler's murine encephalomyelitis virus (TMEV) or vesicular stomatitis virus (VSV). Response of primary mouse neurons to IFN treatment was heterogeneous, as many neurons failed to express the typical IFN response marker Mx1 after IFN treatment. This heterogeneous response of primary neurons correlated with a low level of basal expression of IFN-stimulated genes, such as Stat1, that are involved in signal transduction of the IFN response. In addition, transcriptomic analysis identified 15 IFN-responsive genes whose expression was low in IFN-treated primary neurons compared to that of primary fibroblasts derived from the same mice (Dhx58, Gvin1, Sp100, Ifi203 isoforms 1 and 2, Irgm2, Lgals3bp, Ifi205, Apol9b, Ifi204, Ifi202b, Tor3a, Slfn2, Ifi35, Lgals9). Among these genes, the gene coding for apolipoprotein L9b (Apol9b) displayed antiviral activity against Theiler's virus when overexpressed in L929 cells or in primary neurons. Accordingly, knocking down Apol9b expression in L929 cells increased viral replication. Therefore, we identified a new antiviral protein induced by interferon, ApoL9b, whose lack of expression in primary neurons likely contributes to the high sensitivity of these cells to viral infection. IMPORTANCE The type I interferon (IFN) response is an innate immune defense mechanism that is critical to contain viral infection in the host until an adaptive immune response can be mounted. Neurons are a paradigm for postmitotic, highly differentiated cells. Our data show that primary mouse neurons that are exposed to type I interferon remain surprisingly susceptible to viral infection. On one hand, the low level of basal expression of some factors in neurons might prevent a rapid response of these cells. On the other hand, some genes that are typically activated by type I interferon in other cell types are expressed at much lower levels in neurons. Among these genes is the gene encoding apolipoprotein L9, a protein that proved to have antiviral activity against the neurotropic Theiler's murine encephalomyelitis virus. Our data suggest important functional differences in the IFN response mounted by specific cell populations.
Collapse
|
37
|
Hoyo-Becerra C, Huebener A, Trippler M, Lutterbeck M, Liu ZJ, Truebner K, Bajanowski T, Gerken G, Hermann DM, Schlaak JF. Concomitant interferon alpha stimulation and TLR3 activation induces neuronal expression of depression-related genes that are elevated in the brain of suicidal persons. PLoS One 2013; 8:e83149. [PMID: 24391741 PMCID: PMC3877033 DOI: 10.1371/journal.pone.0083149] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/30/2013] [Indexed: 02/07/2023] Open
Abstract
We have previously identified 15 genes that are associated with the development of severe depressive side effects during the standard therapy with interferon alpha and ribavirin in the peripheral blood of hepatitis C virus infected patients. An enhanced expression of these genes was also found in the blood of psychiatric patients suffering severe depressive episode. Herein, we demonstrate that the same depression-related interferon-inducible genes (DRIIs) are also upregulated in post-mortem brains of suicidal individuals. Using cultured mouse hippocampal and prefrontal neurons we show that costimulation with murine IFN (mIFN) and the TLR3 agonist poly(I:C) promotes the expression of the described DRIIs, at the same time inducing pro-inflammatory cytokine expression through Stat1 and Stat3 activation, promoting neuronal apoptosis. Consequently, the upregulation of selective DRIIs, production of inflammatory cytokines and inhibition of neuronal plasticity may be involved in the pathogenesis of IFN-associated depression.
Collapse
Affiliation(s)
- Carolina Hoyo-Becerra
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Anastasia Huebener
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Martin Trippler
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Melanie Lutterbeck
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Zijian J. Liu
- Department of Anatomy, Tongji Medical College of Huazhong, University of Science and Technology, Wuhan City, P.R.China
| | - Kurt Truebner
- Institute for Forensic Medicine, University Hospital of Essen, Essen, Germany
| | - Thomas Bajanowski
- Institute for Forensic Medicine, University Hospital of Essen, Essen, Germany
| | - Guido Gerken
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Hospital of Essen, Essen, Germany
| | - Joerg F. Schlaak
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
- * E-mail:
| |
Collapse
|
38
|
Hofer MJ, Campbell IL. Type I interferon in neurological disease-the devil from within. Cytokine Growth Factor Rev 2013; 24:257-67. [PMID: 23548179 DOI: 10.1016/j.cytogfr.2013.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/05/2013] [Indexed: 12/31/2022]
Abstract
The members of the type I interferon (IFN-I) family of cytokines are pleiotropic factors that have seminal roles in host defence, acting as antimicrobial and antitumor mediators as well as potent immunomodulatory factors that bridge the innate and adaptive immune responses. Despite these beneficial actions there is mounting evidence that link inappropriate or chronic production of IFN-I in the CNS to the development of a number of severe neuroinflammatory disorders. The most persuasive example is the genetically determined inflammatory encephalopathy, Aicardi-Goutières syndrome (AGS) in which patients have chronically elevated IFN-α production in the CNS. The presentation of AGS can often mimic congenital viral infection, however, molecular genetic studies have identified mutations in six genes that can cause AGS, most likely via dysregulated nucleic acid metabolism and activation of the innate immune response leading to increased intrathecal production of IFN-α. The role of IFN-α as a pathogenic factor in AGS and other neurological disorders has gained considerable support from experimental studies. In particular, a transgenic mouse model with CNS-restricted production of IFN-α replicates many of the cardinal neuropathologic features of AGS and reveal IFN-I to be the "devil from within", mediating molecular and cellular damage within the CNS. Thus, targeting IFN-I may be an effective strategy for the treatment of AGS as well as some other autoimmune and infectious neurological "interferonopathies".
Collapse
Affiliation(s)
- Markus J Hofer
- School of Molecular Bioscience and the Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| | | |
Collapse
|
39
|
Matsui T, Motoki Y, Yoshida Y. Hypothermia reduces toll-like receptor 3-activated microglial interferon-β and nitric oxide production. Mediators Inflamm 2013; 2013:436263. [PMID: 23589665 PMCID: PMC3621171 DOI: 10.1155/2013/436263] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 11/17/2022] Open
Abstract
Therapeutic hypothermia protects neurons after injury to the central nervous system (CNS). Microglia express toll-like receptors (TLRs) that play significant roles in the pathogenesis of sterile CNS injury. To elucidate the possible mechanisms involved in the neuroprotective effect of therapeutic hypothermia, we examined the effects of hypothermic culture on TLR3-activated microglial release of interferon (IFN)- β and nitric oxide (NO), which are known to be associated with neuronal cell death. When rat or mouse microglia were cultured under conditions of hypothermia (33°C) and normothermia (37°C) with a TLR3 agonist, polyinosinic-polycytidylic acid, the production of IFN- β and NO in TLR3-activated microglia at 48 h was decreased by hypothermia compared with that by normothermia. In addition, exposure to recombinant IFN- β and sodium nitroprusside, an NO donor, caused death of rat neuronal pheochromocytoma PC12 cells in a concentration-dependent manner after 24 h. Taken together, these results suggest that the attenuation of microglial production of IFN- β and NO by therapeutic hypothermia leads to the inhibition of neuronal cell death.
Collapse
Affiliation(s)
- Tomohiro Matsui
- Department of Laboratory Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yukari Motoki
- Department of Laboratory Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yusuke Yoshida
- ACEL, Inc., SIC1 1201, 5-4-21 Nishihashimoto, Midori-ku, Sagamihara, Kanagawa 252-0131, Japan
| |
Collapse
|
40
|
Li W, Hofer MJ, Noçon AL, Manders P, Campbell IL. Interferon regulatory factor 7 (IRF7) is required for the optimal initial control but not subsequent clearance of lymphocytic choriomeningitis virus infection in mice. Virology 2013; 439:152-62. [PMID: 23490048 DOI: 10.1016/j.virol.2013.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 10/29/2012] [Accepted: 02/19/2013] [Indexed: 01/02/2023]
Abstract
The role of IRF7 in the host response to lymphocytic choriomeningitis virus (LCMV) Armstrong 53b infection of mice was investigated. Intracranial infection of IRF7 KO mice was associated with delayed onset of LCM, increased survival and significantly reduced expression of the Ifng gene in the brain but not in the periphery. IRF7 KO mice showed impaired control of LCMV replication and delayed clearance of LCMV. Similar numbers of activated anti-LCMV-GP(33-41) CD8+ T cells were present in the brain and spleens of infected WT and IRF7 KO mice. While plasma IFN-β was increased to similar levels, IFN-α was markedly reduced in IRF7 KO compared with WT mice. Compared with IFN-β, IFN-α was a less potent inhibitor of LCMV infection in vitro. In conclusion, IRF7 (1) is required for the early innate control of LCMV infection, likely through the regulation of the appropriate type I IFN response, and (2) is not required for the antiviral CD8+ T cell-dependent clearance of LCMV from infected tissues.
Collapse
Affiliation(s)
- Wen Li
- School of Molecular Bioscience and the Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | | | | | | | | |
Collapse
|
41
|
Cho H, Proll SC, Szretter KJ, Katze MG, Gale M, Diamond MS. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat Med 2013; 19:458-64. [PMID: 23455712 PMCID: PMC3618596 DOI: 10.1038/nm.3108] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/28/2013] [Indexed: 12/30/2022]
Abstract
Different types of neurons are differentially susceptible to West Nile virus (WNV) infection. Michael Diamond and colleagues now show that cerebellar granule cell neurons (GCN) have a higher basal level of expression of type I interferon–inducible genes than cortical neurons, making GCN more resistant to infection by a variety of positive-stranded RNA viruses, including WNV. Although susceptibility of neurons in the brain to microbial infection is a major determinant of clinical outcome, little is known about the molecular factors governing this vulnerability. Here we show that two types of neurons from distinct brain regions showed differential permissivity to replication of several positive-stranded RNA viruses. Granule cell neurons of the cerebellum and cortical neurons from the cerebral cortex have unique innate immune programs that confer differential susceptibility to viral infection ex vivo and in vivo. By transducing cortical neurons with genes that were expressed more highly in granule cell neurons, we identified three interferon-stimulated genes (ISGs; Ifi27, Irg1 and Rsad2 (also known as Viperin)) that mediated the antiviral effects against different neurotropic viruses. Moreover, we found that the epigenetic state and microRNA (miRNA)-mediated regulation of ISGs correlates with enhanced antiviral response in granule cell neurons. Thus, neurons from evolutionarily distinct brain regions have unique innate immune signatures, which probably contribute to their relative permissiveness to infection.
Collapse
Affiliation(s)
- Hyelim Cho
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | |
Collapse
|
42
|
Rauch I, Müller M, Decker T. The regulation of inflammation by interferons and their STATs. JAKSTAT 2013; 2:e23820. [PMID: 24058799 PMCID: PMC3670275 DOI: 10.4161/jkst.23820] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 12/30/2022] Open
Abstract
Interferons (IFN) are subdivided into type I IFN (IFN-I, here synonymous with IFN-α/β), type II (IFN-γ) and type III IFN (IFN-III/IFN-λ) that reprogram nuclear gene expression through STATs 1 and 2 by forming STAT1 dimers (mainly IFN-γ) or the ISGF3 complex, a STAT1-STAT2-IRF9 heterotrimer (IFN-I and IFN-III). Dominant IFN activities in the immune system are to protect cells from viral replication and to activate macrophages for enhanced effector function. However, the impact of IFN and their STATs on the immune system stretches far beyond these activities and includes the control of inflammation. The goal of this review is to give an overview of the different facets of the inflammatory process that show regulatory input by IFN/STAT.
Collapse
Affiliation(s)
- Isabella Rauch
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics and Biomodels Austria; University of Veterinary Medicine Vienna; Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| |
Collapse
|
43
|
Lukasz B, O'Sullivan NC, Loscher JS, Pickering M, Regan CM, Murphy KJ. Peripubertal viral-like challenge and social isolation mediate overlapping but distinct effects on behaviour and brain interferon regulatory factor 7 expression in the adult Wistar rat. Brain Behav Immun 2013; 27:71-9. [PMID: 23036922 DOI: 10.1016/j.bbi.2012.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 09/12/2012] [Accepted: 09/21/2012] [Indexed: 12/14/2022] Open
Abstract
A range of adverse, early life environmental influences such as viral infection and social deprivation are thought to increase risk of psychiatric illness later in life. Here, we used peripheral administration of the viral infection mimic polyriboinosinic-polyribocytidylic acid (polyI:C) to compare the consequences of peripubertal infection and isolation rearing. Isolation rearing induced deficits in sensorimotor gating and recognition memory while no changes in social interaction or spatial learning were observed. PolyI:C injection during the peripubertal period markedly increased expression of interferon-stimulated genes (Ifit2, Prkr, Mx2 and Irf7) in the hippocampal dentate gyrus demonstrating that peripheral administration of the viral mimic in the adolescent animal does have direct effects in the brain. Peripubertal infection mimicry induced a similar but later emerging behavioural deficit in prepulse inhibition implying the existence of a peripubertal window of opportunity for viral-mediated cytokine increases to impact brain development and function. PolyI:C treatment also impaired novel object recognition but did not alter spatial reference memory or social interaction. Combining the polyI:C challenge with social isolation did not exacerbate the behavioural deficits seen with isolation rearing alone. Using Irf7 as a marker, peripubertal viral infection mimicry, isolation rearing and a combination of both were all seen to produce a long-lasting molecular imprint on the interferon-associated signalling pathway in the principal neuron population of the hippocampal dentate gyrus. The data suggest that the sensitivity of brain structure and function to disruption by viral infection extends into the peripubertal period. Moreover, augmented interferon signalling in hippocampus may represent a common molecular imprint of environmental insults associated with neuropsychiatric illnesses like schizophrenia.
Collapse
Affiliation(s)
- Bartlomiej Lukasz
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
44
|
Podolsky MA, Solomos AC, Durso LC, Evans SM, Rall GF, Rose RW. Extended JAK activation and delayed STAT1 dephosphorylation contribute to the distinct signaling profile of CNS neurons exposed to interferon-gamma. J Neuroimmunol 2012; 251:33-8. [PMID: 22769061 DOI: 10.1016/j.jneuroim.2012.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 06/06/2012] [Accepted: 06/11/2012] [Indexed: 01/19/2023]
Abstract
Although interferon-gamma (IFN-γ) plays a critical role in the noncytolytic elimination of many neurotropic viral infections, the signaling response to this cytokine has not been extensively characterized in primary CNS neurons. We previously demonstrated that the IFN-γ response at the signaling and gene expression levels is temporally extended in primary mouse hippocampal neurons, as compared to the transient response of primary mouse embryonic fibroblasts (MEF). We hypothesize that the protracted kinetics of STAT1 phosphorylation in IFN-γ-treated neurons are due to extended receptor activation and/or delayed STAT1 dephosphorylation in the nucleus. Here, we show that in response to IFN-γ, the Janus kinases (JAK1/JAK2) associated with the neuronal IFN-γ receptor complex remain active for an extended period as compared to MEF. Experimental inactivation of JAK1/JAK2 in neurons after IFN-γ treatment did not reverse the extended STAT1 phosphorylation phenotype. These results suggest that the extended kinetics of neuronal IFN-γ signaling are a product of distinct negative feedback mechanisms operating at both the receptor and within the nucleus.
Collapse
Affiliation(s)
- Michael A Podolsky
- Department of Biology, Arcadia University, 450 South Easton Road, Glenside, PA 19038, USA
| | | | | | | | | | | |
Collapse
|
45
|
Dedoni S, Olianas MC, Ingianni A, Onali P. Type I interferons impair BDNF-induced cell signaling and neurotrophic activity in differentiated human SH-SY5Y neuroblastoma cells and mouse primary cortical neurons. J Neurochem 2012; 122:58-71. [PMID: 22533963 DOI: 10.1111/j.1471-4159.2012.07766.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Type I interferons (IFNs) have been shown to act on neurons and to cause neuronal damage through mechanisms not completely defined. Here, we investigated the effects of type I IFNs on brain-derived neurotrophic factor (BDNF)-induced TrkB receptor signaling and neurotrophic activity. In retinoic acid-treated human SH-SY5Y neuroblastoma cells and mouse primary cortical neurons, long-term exposure to IFNs curtailed BDNF-induced activation of phosphatidylinositol 3-kinase, phospholipase Cγ and extracellular-regulated kinases 1 and 2 signaling. Moreover, IFN-β inhibited BDNF-induced cell survival, neurite outgrowth, and expression of neuronal markers, such as neurofilament proteins, growth-associated protein-43 and glutamate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor subunit GluR1. The IFN inhibitory effects were associated with down-regulation of TrkB and inhibition of TrkB autophosphorylation. In SH-SY5Y cells, blockade of either Janus kinase with pyridone 6 or signal transducer and activator of transcription (STAT) 1 with siRNA transfection attenuated IFN-β-induced TrkB down-regulation. Quantitative real time RT-PCR indicated that IFN-β significantly reduced TrkB mRNA levels. Moreover, blockade of protein kinase R counteracted IFN-β-induced inhibition of TrkB expression and signaling. These data indicate that in neuronal cells IFNs negatively regulate BDNF signaling and neurotrophic activity through inhibition of TrkB activation and Janus kinase/Signal transducer and activator of transcription-dependent down-regulation of TrkB.
Collapse
Affiliation(s)
- Simona Dedoni
- Department of Neuroscience, University of Cagliari, Cagliari, Italy
| | | | | | | |
Collapse
|
46
|
Mathieu C, Guillaume V, Sabine A, Ong KC, Wong KT, Legras-Lachuer C, Horvat B. Lethal Nipah virus infection induces rapid overexpression of CXCL10. PLoS One 2012; 7:e32157. [PMID: 22393386 PMCID: PMC3290546 DOI: 10.1371/journal.pone.0032157] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 01/19/2012] [Indexed: 12/15/2022] Open
Abstract
Nipah virus (NiV) is a recently emerged zoonotic Paramyxovirus that causes regular outbreaks in East Asia with mortality rate exceeding 75%. Major cellular targets of NiV infection are endothelial cells and neurons. To better understand virus-host interaction, we analyzed the transcriptome profile of NiV infection in primary human umbilical vein endothelial cells. We further assessed some of the obtained results by in vitro and in vivo methods in a hamster model and in brain samples from NiV-infected patients. We found that NiV infection strongly induces genes involved in interferon response in endothelial cells. Among the top ten upregulated genes, we identified the chemokine CXCL10 (interferon-induced protein 10, IP-10), an important chemoattractant involved in the generation of inflammatory immune response and neurotoxicity. In NiV-infected hamsters, which develop pathology similar to what is seen in humans, expression of CXCL10 mRNA was induced in different organs with kinetics that followed NiV replication. Finally, we showed intense staining for CXCL10 in the brain of patients who succumbed to lethal NiV infection during the outbreak in Malaysia, confirming induction of this chemokine in fatal human infections. This study sheds new light on NiV pathogenesis, indicating the role of CXCL10 during the course of infection and suggests that this chemokine may serve as a potential new marker for lethal NiV encephalitis.
Collapse
Affiliation(s)
- Cyrille Mathieu
- Inserm U758, Human Virology, Ecole Normale Supérieure de Lyon, IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon 1, Lyon, France
| | - Vanessa Guillaume
- Inserm U758, Human Virology, Ecole Normale Supérieure de Lyon, IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon 1, Lyon, France
| | - Amélie Sabine
- Inserm U758, Human Virology, Ecole Normale Supérieure de Lyon, IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon 1, Lyon, France
| | | | | | | | - Branka Horvat
- Inserm U758, Human Virology, Ecole Normale Supérieure de Lyon, IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon 1, Lyon, France
- * E-mail:
| |
Collapse
|
47
|
Fritz-French C, Tyor W. Interferon-α (IFNα) neurotoxicity. Cytokine Growth Factor Rev 2012; 23:7-14. [DOI: 10.1016/j.cytogfr.2012.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 01/09/2012] [Indexed: 01/20/2023]
|
48
|
Functional genomics reveals an essential and specific role for Stat1 in protection of the central nervous system following herpes simplex virus corneal infection. J Virol 2011; 85:12972-81. [PMID: 21994441 DOI: 10.1128/jvi.06032-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Innate immune deficiencies result in a spectrum of severe clinical outcomes following infection. In particular, there is a strong association between loss of the signal transducer and activator of transcription (Stat) pathway, breach of the blood-brain barrier (BBB), and virus-induced neuropathology. The gene signatures that characterize resistance, disease, and mortality in the virus-infected nervous system have not been defined. Herpes simplex virus type 1 (HSV-1) is commonly associated with encephalitis in humans, and humans and mice lacking Stat1 display increased susceptibility to HSV central nervous system (CNS) infections. In this study, two HSV-1 strains were used, KOS (wild type [WT]), and Δvhs, an avirulent recombinant lacking the virion host shutoff (vhs) function. In addition, two mouse strains were used: strain 129 (control) and a Stat1-deficient (Stat1(-/-)) strain. Using combinations of these virus and mouse strains, we established a model of infection resulting in three different outcomes: viral clearance without neurological disease (Δvhs infection of control mice), neurological disease followed by viral clearance (Δvhs infection of Stat1(-/-) mice and WT infection of control mice), or neurological disease followed by death (WT infection of Stat1(-/-) mice). Through the use of functional genomics on the infected brain stems, we determined gene signatures that were representative of the three infection outcomes. We demonstrated a pathological signature in the brain stem of Stat1-deficient mice characterized by upregulation of transcripts encoding chemokine receptors, inflammatory markers, neutrophil chemoattractants, leukocyte adhesion proteins, and matrix metalloproteases. Additionally, there was a greater than 100-fold increase in the inflammatory markers interleukin 1β (IL-1β) and IL-6. Consistent with this gene signature, we demonstrated profound CNS inflammation with a concomitant lethal breach of the BBB. Taken together, our results indicated an essential role for normal Stat1-dependent signaling in mediating a nonpathological immune response to viral CNS infection.
Collapse
|
49
|
Fil D, Borysiewicz E, Konat GW. A broad upregulation of cerebral chemokine genes by peripherally-generated inflammatory mediators. Metab Brain Dis 2011; 26:49-59. [PMID: 21258854 DOI: 10.1007/s11011-010-9231-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 12/21/2010] [Indexed: 12/22/2022]
Abstract
Previously, we have shown that peripheral challenge of mice with double stranded RNA (dsRNA), a viral mimic, evokes global upregulation of cerebral inflammatory genes and, particularly, genes encoding chemokines. Because chemokine networks are potent modulators of brain function, the present study was undertaken to comprehensively characterize the cerebral response of chemokine ligand and receptor genes to peripheral immune system stimulation. Briefly, C57BL/6 mice were intraperitoneally injected with 12 mg/kg of polyinosinic-polycytidylic acid (PIC) and the expression of 39 mouse chemokine ligand and 20 receptor genes was monitored in the cerebellum by real time quantitative RT-PCR within 24 h. Almost half of the ligand genes featured either transient or sustained upregulation from several- to several thousand-fold. Five CXC type genes, i.e., Cxcl9, Cxcl11, Cxcl10, Cxcl2 and Cxcl1, were the most robustly upregulated, and were followed by six CC type genes, i.e., Ccl2, Ccl7, Ccl5, Ccl12, Ccl4 and Ccl11. Seven genes showed moderate upregulation, whereas the remaining genes were unresponsive. Six receptor genes, i.e., Cxcr2, Ccr7, Cxcr5, Ccr6, Ccr1 and Ccr5, featured a several-fold upregulation. Similar chemokine gene response was observed in the forebrain and brainstem. This upregulation of chemokine genes could be induced in naïve mice by transfer of blood plasma from PIC-challenged mice. Employing oligodeoxynucleotide-labeled PIC we further showed that intraperitoneally injected PIC was not transferred to the blood. In conclusion, peripheral PIC challenge elicits a broad upregulation of cerebral chemokine genes, and this upregulation is mediated by blood-borne agents.
Collapse
Affiliation(s)
- Daniel Fil
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, 4052 HSN, P.O. Box 9128, Morgantown, WV 26506-9128, USA
| | | | | |
Collapse
|
50
|
Dedoni S, Olianas MC, Onali P. Interferon-β induces apoptosis in human SH-SY5Y neuroblastoma cells through activation of JAK-STAT signaling and down-regulation of PI3K/Akt pathway. J Neurochem 2010; 115:1421-33. [PMID: 21044071 DOI: 10.1111/j.1471-4159.2010.07046.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Type I interferons (IFNs) are known to cause neuropsychiatric side effects, which have been proposed to be mediated by either peripheral actions or activation of glial cells. In the present study, we have investigated whether these cytokines could act directly on neuronal cells and regulate signaling pathways involved in cell death. In human SH-SY5Y neuroblastoma cells, type I IFNs rapidly stimulated tyrosine phosphorylation of Janus kinase and signal transducer and activator of transcription (STAT) through type I IFN receptor. Prolonged exposure to IFN-β induced apoptotic cell death accompanied by cytochrome C release, cleavage of caspases 9, 7, 3 and poly-(ADP ribose) polymerase and DNA fragmentation. Janus kinase inhibition reduced IFN-β-stimulated TyK2 and STAT1 phosphorylation, STAT1 transcriptional activity, induction of double-stranded RNA-activated protein kinase (PKR) and caspase cleavage. PKR induction was associated with enhanced PKR activity and chemical inhibition of PKR reduced IFN-stimulated caspase activation. Moreover, long-term IFN-β treatment led to down-regulation of phosphatidylinositol 3-kinase/Akt signaling and IFN-β-induced apoptosis was attenuated in cells expressing constitutively active Akt. Similarly, in mouse primary neurons IFN-β induced STAT phosphorylation, caspase 3 cleavage and inhibition of Akt signaling. Thus, type I IFNs can directly impair neuronal survival by regulating multiple signaling molecules promoting the intrinsic apoptotic pathway. This effect may contribute to the cytokine neurotoxicity.
Collapse
Affiliation(s)
- Simona Dedoni
- Department of Neuroscience, Section of Biochemical Pharmacology, University of Cagliari, Cagliari, Italy
| | | | | |
Collapse
|