51
|
Saiz JC, Martín-Acebes MA, Blázquez AB, Escribano-Romero E, Poderoso T, Jiménez de Oya N. Pathogenicity and virulence of West Nile virus revisited eight decades after its first isolation. Virulence 2021; 12:1145-1173. [PMID: 33843445 PMCID: PMC8043182 DOI: 10.1080/21505594.2021.1908740] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
West Nile virus (WNV) is a flavivirus which transmission cycle is maintained between mosquitoes and birds, although it occasionally causes sporadic outbreaks in horses and humans that can result in serious diseases and even death. Since its first isolation in Africa in 1937, WNV had been considered a neglected pathogen until its recent spread throughout Europe and the colonization of America, regions where it continues to cause outbreaks with severe neurological consequences in humans and horses. Although our knowledge about the characteristics and consequences of the virus has increased enormously lately, many questions remain to be resolved. Here, we thoroughly update our knowledge of different aspects of the WNV life cycle: virology and molecular classification, host cell interactions, transmission dynamics, host range, epidemiology and surveillance, immune response, clinical presentations, pathogenesis, diagnosis, prophylaxis (antivirals and vaccines), and prevention, and we highlight those aspects that are still unknown and that undoubtedly require further investigation.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Miguel A Martín-Acebes
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Ana B Blázquez
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Estela Escribano-Romero
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Teresa Poderoso
- Molecular Virology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nereida Jiménez de Oya
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| |
Collapse
|
52
|
Tembusu Virus entering the central nervous system caused nonsuppurative encephalitis without disrupting the blood-brain barrier. J Virol 2021; 95:JVI.02191-20. [PMID: 33472933 PMCID: PMC8092698 DOI: 10.1128/jvi.02191-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tembusu Virus (TMUV) is an emerging and re-emerging zoonotic pathogen that adversely affects poultry industry in recent years. TMUV disease is characterized by nonsuppurative encephalitis in ducklings. The duckling infection model was established to study the mechanism of TMUV crossing the blood-brain barrier (BBB) into the central nervous system (CNS). Here, we showed that no obvious clinical symptoms and enhancement of BBB permeability occurred at the early stage of infection (3∼5 dpi). While simultaneously virus particles were observed by transmission electron microscopy in the brain, inducing the accumulation of inflammatory cytokines. Neurological symptoms and disruption of BBB appeared at the intermediate stage of infection (7∼9 dpi). It was confirmed that TMUV could survive and propagate in brain microvascular endothelial cells (BMECs), but did not affect the permeability of BBB in vivo and in vitro at an early date. In conclusion, TMUV enters the CNS then causes encephalitis, and finally destruct the BBB, which may be due to the direct effect of TMUV on BMECs and the subsequent response of "inflammatory storm".IMPORTANCE The TMUV disease has caused huge losses to the poultry industry in Asia, which is potentially harmful to public health. Neurological symptoms and their sequelae are the main characters of this disease. However, the mechanism of how this virus enters the brain and causes encephalitis is unclear. In this study, we confirmed that the virus entered the CNS and then massively destroyed BBB and the BBB damage was closely associated with the subsequent outbreak of inflammation. TMUV may enter the CNS through the transcellular and "Trojan horse" pathways. These findings can fill the knowledge gap in the pathogenesis of TMUV-infected poultry and be benefit for the treatment of TMUV disease. What's more, TMUV is a representative to study the infection of avian flavivirus. Therefore, our studies have significances both for understanding of the full scope of mechanisms of TMUV and other flavivirus infection, and conceivably, for therapeutics.
Collapse
|
53
|
Chhatbar C, Prinz M. The roles of microglia in viral encephalitis: from sensome to therapeutic targeting. Cell Mol Immunol 2021; 18:250-258. [PMID: 33437050 PMCID: PMC7802409 DOI: 10.1038/s41423-020-00620-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 01/31/2023] Open
Abstract
Viral encephalitis is a devastating disease with high mortality, and survivors often suffer from severe neurological complications. Microglia are innate immune cells of the central nervous system (CNS) parenchyma whose turnover is reliant on local proliferation. Microglia express a diverse range of proteins, which allows them to continuously sense the environment and quickly react to changes. Under inflammatory conditions such as CNS viral infection, microglia promote innate and adaptive immune responses to protect the host. However, during viral infection, a dysregulated microglia-T-cell interplay may result in altered phagocytosis of neuronal synapses by microglia that causes neurocognitive impairment. In this review, we summarize the current knowledge on the role of microglia in viral encephalitis, propose questions to be answered in the future and suggest possible therapeutic targets.
Collapse
Affiliation(s)
- Chintan Chhatbar
- grid.5963.9Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- grid.5963.9Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany ,grid.5963.9Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany ,grid.5963.9Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
54
|
Cortes‐Selva D, Fairfax K. Schistosome and intestinal helminth modulation of macrophage immunometabolism. Immunology 2021; 162:123-134. [PMID: 32614982 PMCID: PMC7808165 DOI: 10.1111/imm.13231] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are fundamental to sustain physiological equilibrium and to regulate the pathogenesis of parasitic and metabolic processes. The functional heterogeneity and immune responses of macrophages are shaped by cellular metabolism in response to the host's intrinsic factors, environmental cues and other stimuli during disease. Parasite infections induce a complex cascade of cytokines and metabolites that profoundly remodel the metabolic status of macrophages. In particular, helminths polarize macrophages to an M2 state and induce a metabolic shift towards reliance on oxidative phosphorylation, lipid oxidation and amino acid metabolism. Accumulating data indicate that helminth-induced activation and metabolic reprogramming of macrophages underlie improvement in overall whole-body metabolism, denoted by improved insulin sensitivity, body mass in response to high-fat diet and atherogenic index in mammals. This review aims to highlight the metabolic changes that occur in human and murine-derived macrophages in response to helminth infections and helminth products, with particular interest in schistosomiasis and soil-transmitted helminths.
Collapse
Affiliation(s)
- Diana Cortes‐Selva
- Division of Microbiology and ImmunologyDepartment of PathologyUniversity of UtahSalt Lake CityUTUSA
- Janssen BiotherapeuticsJanssen R&DSpring HousePAUSA
| | - Keke Fairfax
- Division of Microbiology and ImmunologyDepartment of PathologyUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
55
|
Mai CL, Tan Z, Xu YN, Zhang JJ, Huang ZH, Wang D, Zhang H, Gui WS, Zhang J, Lin ZJ, Meng YT, Wei X, Jie YT, Grace PM, Wu LJ, Zhou LJ, Liu XG. CXCL12-mediated monocyte transmigration into brain perivascular space leads to neuroinflammation and memory deficit in neuropathic pain. Theranostics 2021; 11:1059-1078. [PMID: 33391521 PMCID: PMC7738876 DOI: 10.7150/thno.44364] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging clinical and experimental evidence demonstrates that neuroinflammation plays an important role in cognitive impairment associated with neuropathic pain. However, how peripheral nerve challenge induces remote inflammation in the brain remains largely unknown. Methods: The circulating leukocytes and plasma C-X-C motif chemokine 12 (CXCL12) and brain perivascular macrophages (PVMs) were analyzed by flow cytometry, Western blotting, ELISA, and immunostaining in spared nerve injury (SNI) mice. The memory function was evaluated with a novel object recognition test (NORT) in mice and with Montreal Cognitive Assessment (MoCA) in chronic pain patients. Results: The classical monocytes and CXCL12 in the blood, PVMs in the perivascular space, and gliosis in the brain, particularly in the hippocampus, were persistently increased following SNI in mice. Using the transgenic CCR2RFP/+ and CX3CR1GFP/+ mice, we discovered that at least some of the PVMs were recruited from circulating monocytes. The SNI-induced increase in hippocampal PVMs, gliosis, and memory decline were substantially prevented by either depleting circulating monocytes via intravenous injection of clodronate liposomes or blockade of CXCL12-CXCR4 signaling. On the contrary, intravenous injection of CXCL12 at a pathological concentration in naïve mice mimicked SNI effects. Significantly, we found that circulating monocytes and plasma CXCL12 were elevated in chronic pain patients, and both of them were closely correlated with memory decline. Conclusion: CXCL12-mediated monocyte recruitment into the perivascular space is critical for neuroinflammation and the resultant cognitive impairment in neuropathic pain.
Collapse
Affiliation(s)
- Chun-Lin Mai
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ya-Nan Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing-Jun Zhang
- Department of Anesthesiology and Pain Clinic, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhen-Hua Huang
- Division of Emergency Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Zhang
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Wen-Shan Gui
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Zhang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Tong Meng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Wei
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Tao Jie
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Peter M. Grace
- Department of Critical Care & Respiratory Care Research (PMG), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
| | - Xian-Guo Liu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
| |
Collapse
|
56
|
Therapeutic role of inflammasome inhibitors in neurodegenerative disorders. Brain Behav Immun 2021; 91:771-783. [PMID: 33157255 DOI: 10.1016/j.bbi.2020.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation, characterized by the activation of glial cells, is a hallmark in several neurological and neurodegenerative disorders. Inadequate inflammation cannot eliminate the infection of pathogens, while excessive or hyper-reactive inflammation can cause chronic or systemic inflammatory diseases affecting the central nervous system (CNS). In response to a brain injury or pathogen invasion, the pathogen recognition receptors (PRRs) expressed on glial cells are activated via binding to cellular damage-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs). This subsequently leads to the activation of NOD (nucleotide-binding oligomerization domain)-like receptor proteins (NLRs). In neurodegenerative diseases such as HIV-1-associated neurocognitive disorders (HAND), Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS), chronic inflammation is a critical contributing factor for disease manifestation including pathogenesis. Emerging evidence points to the involvement of "inflammasomes", especially the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing (NLRP) complex in the development of these diseases. The activated NLRP3 results in the proteolytic activation of caspase-1 that facilitates the cleavage of pro-IL-1β and the secretion of IL-1β and IL-18 proinflammatory cytokines. Accordingly, these and other seminal findings have led to the development of NLRP-targeting small-molecule therapeutics as possible treatment options for neurodegenerative disorders. In this review, we will discuss the new advances and evidence-based literature concerning the role of inflammasomes in neurodegenerative diseases, its role in the neurological repercussions of CNS chronic infection, and the examples of preclinical or clinically tested NLRP inhibitors as potential strategies for the treatment of chronic neurological diseases.
Collapse
|
57
|
Xing Z, Zuo Z, Hu D, Zheng X, Wang X, Yuan L, Zhou L, Qi F, Yao Z. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-β accumulation and improves cognition in APP/PS1 mice. Brain Behav Immun 2021; 91:128-141. [PMID: 32956831 DOI: 10.1016/j.bbi.2020.09.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Immune dysfunction is implicated in Alzheimer's disease (AD), whereas systemic immune modulation may be neuroprotective. Our previous results have indicated immune challenge with Bacillus Calmette-Guerin attenuates AD pathology in animal models by boosting the systemic immune system. Similarly, independent studies have shown that boosting systemic immune system, by blocking PD-1 checkpoint pathway, modifies AD. Here we hypothesized that influenza vaccine would potentiate function of moderate dose anti-PD-1 and therefore combining them might allow reducing the dose of PD-1 antibody needed to modify the disease. We found that moderate-dose PD-1 in combination with influenza vaccine effectively attenuated cognitive deficit and prevented amyloid-β pathology build-up in APP/PS1 mice in a mechanism dependent on recruitment of peripheral monocyte-derived macrophages into the brain. Eliminating peripheral macrophages abrogated the beneficial effect. Moreover, by comparing CD11b+ compartments in the mouse parenchyma, we observed an elevated subset of Ly6C+ microglia-like cells, which are reportedly derived from peripheral monocytes. In addition, myeloid-derived suppressor cells are strongly elevated in the transgenic model used and normalized by combination treatment, indicating restoration of brain immune homeostasis. Overall, our results suggest that revitalizing brain immunity by combining IV with moderate-dose PD-1 inhibition may represent a therapeutic immunotherapy for AD.
Collapse
Affiliation(s)
- Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, PR China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lifang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lihua Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Anatomy, Sun Yat-sen University, School of Medicine, Guangzhou 510089, PR China.
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
58
|
Blaszkiewicz M, Wood E, Koizar S, Willows J, Anderson R, Tseng YH, Godwin J, Townsend KL. The involvement of neuroimmune cells in adipose innervation. Mol Med 2020; 26:126. [PMID: 33297933 PMCID: PMC7727151 DOI: 10.1186/s10020-020-00254-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Innervation of adipose tissue is essential for the proper function of this critical metabolic organ. Numerous surgical and chemical denervation studies have demonstrated how maintenance of brain-adipose communication through both sympathetic efferent and sensory afferent nerves helps regulate adipocyte size, cell number, lipolysis, and 'browning' of white adipose tissue. Neurotrophic factors are growth factors that promote neuron survival, regeneration, and plasticity, including neurite outgrowth and synapse formation. Peripheral immune cells have been shown to be a source of neurotrophic factors in humans and mice. Although a number of immune cells reside in the adipose stromal vascular fraction (SVF), it has remained unclear what roles they play in adipose innervation. We previously demonstrated that adipose SVF secretes brain derived neurotrophic factor (BDNF). METHODS We now show that deletion of this neurotrophic factor from the myeloid lineage of immune cells led to a 'genetic denervation' of inguinal subcutaneous white adipose tissue (scWAT), thereby causing decreased energy expenditure, increased adipose mass, and a blunted UCP1 response to cold stimulation. RESULTS We and others have previously shown that noradrenergic stimulation via cold exposure increases adipose innervation in the inguinal depot. Here we have identified a subset of myeloid cells that home to scWAT upon cold exposure and are Ly6C+ CCR2+ Cx3CR1+ monocytes/macrophages that express noradrenergic receptors and BDNF. This subset of myeloid lineage cells also clearly interacted with peripheral nerves in the scWAT and were therefore considered neuroimmune cells. CONCLUSIONS We propose that these myeloid lineage, cold induced neuroimmune cells (CINCs) are key players in maintaining adipose innervation as well as promoting adipose nerve remodeling under noradrenergic stimulation, such as cold exposure.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- School of Biology and Ecology, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Elizabeth Wood
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Sigi Koizar
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Jake Willows
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Ryan Anderson
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - James Godwin
- Jackson Laboratory, Bar Harbor, ME, USA
- MDI Biological Laboratory, Bar Harbor, ME, USA
| | - Kristy L Townsend
- School of Biology and Ecology, University of Maine, Orono, ME, USA.
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA.
- The Ohio State University, 1014 Biomedical Research Tower, 460 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
59
|
Spiteri AG, Wishart CL, King NJC. Immovable Object Meets Unstoppable Force? Dialogue Between Resident and Peripheral Myeloid Cells in the Inflamed Brain. Front Immunol 2020; 11:600822. [PMID: 33363542 PMCID: PMC7752943 DOI: 10.3389/fimmu.2020.600822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammation of the brain parenchyma is characteristic of neurodegenerative, autoimmune, and neuroinflammatory diseases. During this process, microglia, which populate the embryonic brain and become a permanent sentinel myeloid population, are inexorably joined by peripherally derived monocytes, recruited by the central nervous system. These cells can quickly adopt a morphology and immunophenotype similar to microglia. Both microglia and monocytes have been implicated in inducing, enhancing, and/or maintaining immune-mediated pathology and thus disease progression in a number of neuropathologies. For many years, experimental and analytical systems have failed to differentiate resident microglia from peripherally derived myeloid cells accurately. This has impeded our understanding of their precise functions in, and contributions to, these diseases, and hampered the development of novel treatments that could target specific cell subsets. Over the past decade, microglia have been investigated more intensively in the context of neuroimmunological research, fostering the development of more precise experimental systems. In light of our rapidly growing understanding of these cells, we discuss the differential origins of microglia and peripherally derived myeloid cells in the inflamed brain, with an analysis of the problems resolving these cell types phenotypically and morphologically, and highlight recent developments enabling more precise identification.
Collapse
Affiliation(s)
- Alanna G. Spiteri
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Claire L. Wishart
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas J. C. King
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Nano Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
60
|
Honarpisheh P, Lee J, Banerjee A, Blasco-Conesa MP, Honarpisheh P, d'Aigle J, Mamun AA, Ritzel RM, Chauhan A, Ganesh BP, McCullough LD. Potential caveats of putative microglia-specific markers for assessment of age-related cerebrovascular neuroinflammation. J Neuroinflammation 2020; 17:366. [PMID: 33261619 PMCID: PMC7709276 DOI: 10.1186/s12974-020-02019-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/29/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The ability to distinguish resident microglia from infiltrating myeloid cells by flow cytometry-based surface phenotyping is an important technique for examining age-related neuroinflammation. The most commonly used surface markers for the identification of microglia include CD45 (low-intermediate expression), CD11b, Tmem119, and P2RY12. METHODS In this study, we examined changes in expression levels of these putative microglia markers in in vivo animal models of stroke, cerebral amyloid angiopathy (CAA), and aging as well as in an ex vivo LPS-induced inflammation model. RESULTS We demonstrate that Tmem119 and P2RY12 expression is evident within both CD45int and CD45high myeloid populations in models of stroke, CAA, and aging. Interestingly, LPS stimulation of FACS-sorted adult microglia suggested that these brain-resident myeloid cells can upregulate CD45 and downregulate Tmem119 and P2RY12, making them indistinguishable from peripherally derived myeloid populations. Importantly, our findings show that these changes in the molecular signatures of microglia can occur without a contribution from the other brain-resident or peripherally sourced immune cells. CONCLUSION We recommend future studies approach microglia identification by flow cytometry with caution, particularly in the absence of the use of a combination of markers validated for the specific neuroinflammation model of interest. The subpopulation of resident microglia residing within the "infiltrating myeloid" population, albeit small, may be functionally important in maintaining immune vigilance in the brain thus should not be overlooked in neuroimmunological studies.
Collapse
Affiliation(s)
- Pedram Honarpisheh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.,UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Juneyoung Lee
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Anik Banerjee
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.,UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Maria P Blasco-Conesa
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Parisa Honarpisheh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - John d'Aigle
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Abdullah A Mamun
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Rodney M Ritzel
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
61
|
Ma RY, Zhang H, Li XF, Zhang CB, Selli C, Tagliavini G, Lam AD, Prost S, Sims AH, Hu HY, Ying T, Wang Z, Ye Z, Pollard JW, Qian BZ. Monocyte-derived macrophages promote breast cancer bone metastasis outgrowth. J Exp Med 2020; 217:e20191820. [PMID: 32780802 PMCID: PMC7596825 DOI: 10.1084/jem.20191820] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/08/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Bone metastasis is the major cause of death in breast cancer. The lack of effective treatment suggests that disease mechanisms are still largely unknown. As a key component of the tumor microenvironment, macrophages promote tumor progression and metastasis. In this study, we found that macrophages are abundant in human and mouse breast cancer bone metastases. Macrophage ablation significantly inhibited bone metastasis growth. Lineage tracking experiments indicated that these macrophages largely derive from Ly6C+CCR2+ inflammatory monocytes. Ablation of the chemokine receptor, CCR2, significantly inhibited bone metastasis outgrowth and prolonged survival. Immunophenotyping identified that bone metastasis-associated macrophages express high levels of CD204 and IL4R. Furthermore, monocyte/macrophage-restricted IL4R ablation significantly inhibited bone metastasis growth, and IL4R null mutant monocytes failed to promote bone metastasis outgrowth. Together, this study identified a subset of monocyte-derived macrophages that promote breast cancer bone metastasis in an IL4R-dependent manner. This suggests that IL4R and macrophage inhibition can have potential therapeutic benefit against breast cancer bone disease.
Collapse
Affiliation(s)
- Ruo-Yu Ma
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Hui Zhang
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY
| | - Xue-Feng Li
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cheng-Bin Zhang
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cigdem Selli
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Giulia Tagliavini
- Medical Research Council Centre for Inflammation Research, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Alyson D. Lam
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Sandrine Prost
- Medical Research Council Centre for Inflammation Research, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Andrew H. Sims
- Edinburgh Cancer Research UK Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Hai-Yan Hu
- Shanghai Jiao Tong University Affiliated Sixth People`s Hospital, Shanghai, China
| | - Tianlei Ying
- Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhan Wang
- Department of Orthopaedics, Centre for Orthopaedic Research, Orthopaedics Research Institute of Zhejiang University, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopaedics, Centre for Orthopaedic Research, Orthopaedics Research Institute of Zhejiang University, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Jeffrey W. Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY
| | - Bin-Zhi Qian
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research UK Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Guangdong Provincial Education Department Key Laboratory of Nano-immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
62
|
Mid-life microbiota crises: middle age is associated with pervasive neuroimmune alterations that are reversed by targeting the gut microbiome. Mol Psychiatry 2020; 25:2567-2583. [PMID: 31092898 DOI: 10.1038/s41380-019-0425-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/23/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022]
Abstract
Male middle age is a transitional period where many physiological and psychological changes occur leading to cognitive and behavioural alterations, and a deterioration of brain function. However, the mechanisms underpinning such changes are unclear. The gut microbiome has been implicated as a key mediator in the communication between the gut and the brain, and in the regulation of brain homeostasis, including brain immune cell function. Thus, we tested whether targeting the gut microbiome by prebiotic supplementation may alter microglia activation and brain function in ageing. Male young adult (8 weeks) and middle-aged (10 months) C57BL/6 mice received diet enriched with a prebiotic (10% oligofructose-enriched inulin) or control chow for 14 weeks. Prebiotic supplementation differentially altered the gut microbiota profile in young and middle-aged mice with changes correlating with faecal metabolites. Functionally, this translated into a reversal of stress-induced immune priming in middle-aged mice. In addition, a reduction in ageing-induced infiltration of Ly-6Chi monocytes into the brain coupled with a reversal in ageing-related increases in a subset of activated microglia (Ly-6C+) was observed. Taken together, these data highlight a potential pathway by which targeting the gut microbiome with prebiotics can modulate the peripheral immune response and alter neuroinflammation in middle age. Our data highlight a novel strategy for the amelioration of age-related neuroinflammatory pathologies and brain function.
Collapse
|
63
|
Xia X, Wang W, Yin K, Wang S. Interferon regulatory factor 8 governs myeloid cell development. Cytokine Growth Factor Rev 2020; 55:48-57. [DOI: 10.1016/j.cytogfr.2020.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
|
64
|
Singh S, Singh G, Tiwari S, Kumar A. CCR2 Inhibition Reduces Neurotoxic Microglia Activation Phenotype After Japanese Encephalitis Viral Infection. Front Cell Neurosci 2020; 14:230. [PMID: 32903799 PMCID: PMC7439097 DOI: 10.3389/fncel.2020.00230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022] Open
Abstract
Controlling the proinflammatory response of microglia by targeting chemokines (C-C motif) receptor 2 (CCR2) could be an important therapeutic approach for Japanese encephalitis virus (JEV) infection. Here, through JEV infection to BV2 microglia and young BALB/c mice, we investigated that CCR2 is highly upregulated after JEV infection and plays a key role in determining microglia activation phenotype and associated with neurotoxic proinflammatory mediators of TNF-α and IFNγ. In addition, we found JEV infection to BV2 microglia causes an increase in microglial proliferation and cell body area at day 1 and day 3. Using the agonist molecule of CCR2 inhibition; RS102895, significantly reduces microglia reactive phenotype and nitric oxide production. Further, to define the role of CCR2 in functional responses of microglia and their activation phenotype, we performed in vitro cell scratch functional assay and ImageJ analysis. When compared with control, microglia cells showed a significant increase in elongated or rod-like activated phenotype in JEV-infected cells at 24 h post-infection and CCR2 inhibition significantly reduced the elongated activation phenotype induced by JEV infection, suggesting that CCR2 acts as a critical regulator for microglia activation phenotype after JEV infection. We found that JEV-infected mice treated with RS102895 had less microglia activation and reduced mRNA expression of CCR2 and proinflammatory mediators such as IFN-γ in cortical tissue. Collectively, our data indicate that CCR2 drives reactive phenotype of microglia and its inhibition reduces microglia activation and neurotoxic proinflammatory mediators after JEV infection.
Collapse
Affiliation(s)
- Swati Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| |
Collapse
|
65
|
Trzeciak A, Pietropaoli AP, Kim M. Biomarkers and Associated Immune Mechanisms for Early Detection and Therapeutic Management of Sepsis. Immune Netw 2020; 20:e23. [PMID: 32655971 PMCID: PMC7327151 DOI: 10.4110/in.2020.20.e23] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Sepsis is conceptually defined as life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Although there has been significant advancement in recent decades in defining and understanding sepsis pathology, clinical management of sepsis is challenging due to difficulties in diagnosis, a lack of reliable prognostic biomarkers, and treatment options that are largely limited to antibiotic therapy and fundamental supportive measures. The lack of reliable diagnostic and prognostic tests makes it difficult to triage patients who are in need of more urgent care. Furthermore, while the acute inpatient treatment of sepsis warrants ongoing attention and investigation, efforts must also be directed toward longer term survival and outcomes. Sepsis survivors experience incomplete recovery, with long-term health impairments that may require both cognitive and physical treatment and rehabilitation. This review summarizes recent advances in sepsis prognosis research and discusses progress made in elucidating the underlying causes of prolonged health deficits experienced by patients surviving the early phases of sepsis.
Collapse
Affiliation(s)
- Alissa Trzeciak
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony P Pietropaoli
- Pulmonary and Critical Care Medicine Division, University of Rochester, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
66
|
Meghraoui-Kheddar A, Barthelemy S, Boissonnas A, Combadière C. Revising CX3CR1 Expression on Murine Classical and Non-classical Monocytes. Front Immunol 2020; 11:1117. [PMID: 32582197 PMCID: PMC7283740 DOI: 10.3389/fimmu.2020.01117] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/07/2020] [Indexed: 12/25/2022] Open
Abstract
In mice, monocytes (Mo) are conventionally described as CX3CR1low classical Mo (CMo) and CX3CR1high non-classical Mo (NCMo) based on the expression of EGFP in Cx3cr1+/EGFP mice and by analogy with human CX3CR1 expression. Although this terminology is widely used, it may not reflect the expression of CX3CR1 on Mo subsets. Using an unsupervised multiparametric analysis of blood Mo in steady state and after sterile peritonitis, we observed that CX3CR1 expression did not discriminate the CMo from the NCMo subsets. Our results highlight that despite being a reliable reporter to discriminate Mo subpopulations, EGFP level in Cx3cr1+/EGFP mice does not reflect CX3CR1 expression measured by a fluorescently-labeled CX3CL1 chemokine and a CX3CR1 specific antibody. In conclusion, authors should be cautious not to identify murine classical and non-classical Mo as CX3CR1low and CX3CR1high but rather use alternative markers such as the combination of Ly6C and CD43.
Collapse
Affiliation(s)
- Aïda Meghraoui-Kheddar
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Sandrine Barthelemy
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Christophe Combadière
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| |
Collapse
|
67
|
Mutso M, St John JA, Ling ZL, Burt FJ, Poo YS, Liu X, Žusinaite E, Grau GE, Hueston L, Merits A, King NJ, Ekberg JA, Mahalingam S. Basic insights into Zika virus infection of neuroglial and brain endothelial cells. J Gen Virol 2020; 101:622-634. [PMID: 32375993 PMCID: PMC7414445 DOI: 10.1099/jgv.0.001416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/24/2020] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) has recently emerged as an important human pathogen due to the strong evidence that it causes disease of the central nervous system, particularly microcephaly and Guillain-Barré syndrome. The pathogenesis of disease, including mechanisms of neuroinvasion, may include both invasion via the blood-brain barrier and via peripheral (including cranial) nerves. Cellular responses to infection are also poorly understood. This study characterizes the in vitro infection of laboratory-adapted ZIKV African MR766 and two Asian strains of (1) brain endothelial cells (hCMEC/D3 cell line) and (2) olfactory ensheathing cells (OECs) (the neuroglia populating cranial nerve I and the olfactory bulb; both human and mouse OEC lines) in comparison to kidney epithelial cells (Vero cells, in which ZIKV infection is well characterized). Readouts included infection kinetics, intracellular virus localization, viral persistence and cytokine responses. Although not as high as in Vero cells, viral titres exceeded 104 plaque-forming units (p.f.u.) ml-1 in the endothelial/neuroglial cell types, except hOECs. Despite these substantial titres, a relatively small proportion of neuroglial cells were primarily infected. Immunolabelling of infected cells revealed localization of the ZIKV envelope and NS3 proteins in the cytoplasm; NS3 staining overlapped with that of dsRNA replication intermediate and the endoplasmic reticulum (ER). Infected OECs and endothelial cells produced high levels of pro-inflammatory chemokines. Nevertheless, ZIKV was also able to establish persistent infection in hOEC and hCMEC/D3 cells. Taken together, these results provide basic insights into ZIKV infection of endothelial and neuroglial cells and will form the basis for further study of ZIKV disease mechanisms.
Collapse
Affiliation(s)
- Margit Mutso
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - James A. St John
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan 4111, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Zheng Lung Ling
- Discipline of Pathology, Bosch Institute, Marie Bashir Institute for Infectious diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Felicity J. Burt
- National Health Laboratory Services, University of the Free State, Bloemfontein, South Africa
| | - Yee Suan Poo
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Xiang Liu
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Eva Žusinaite
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Georges E. Grau
- Vascular Immunology Unit, Discipline of Pathology, Sydney Medical School, University of Sydney, New South Wales 2050, Australia
| | - Linda Hueston
- Arbovirus Emerging Disease Unit, CIDMLS-ICPMR, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Nicholas J.C. King
- Discipline of Pathology, Bosch Institute, Marie Bashir Institute for Infectious diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Jenny A.K. Ekberg
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Suresh Mahalingam
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| |
Collapse
|
68
|
Amarilla AA, Santos-Junior NN, Figueiredo ML, Luiz JPM, Fumagalli MJ, Colón DF, Lippi V, Alfonso HL, Lima-Junior DS, Trabuco AC, Spinieli RL, Desidera AC, Leite-Panissi CRA, Lauretti F, Mendoza SES, Silva CLA, Rego EM, Galvao-Lima LJ, Bassi GS, Penharvel Martíns SLB, Manrique WG, Alves-Filho JC, Cunha FQ, Peng NYG, Modhiran N, Setoh YX, Khromykh AA, Figueiredo LTM, Aquino VH. CCR2 Plays a Protective Role in Rocio Virus-Induced Encephalitis by Promoting Macrophage Infiltration Into the Brain. J Infect Dis 2020; 219:2015-2025. [PMID: 30715407 PMCID: PMC7107438 DOI: 10.1093/infdis/jiz029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 12/15/2022] Open
Abstract
Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975–1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.
Collapse
Affiliation(s)
- Alberto A Amarilla
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil.,Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | | | - Mario Luis Figueiredo
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | | | - David F Colón
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Veronica Lippi
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Helda Liz Alfonso
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Djalma S Lima-Junior
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, Ribeirao Preto, SP, Brazil
| | - Amanda C Trabuco
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Richard L Spinieli
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Amanda C Desidera
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Christie R A Leite-Panissi
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Silvia Elena Sánchez Mendoza
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | | | - Eduardo Magalhaes Rego
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | - Leonardo J Galvao-Lima
- Department of Immunology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Gabriel S Bassi
- Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sandra L B Penharvel Martíns
- Department of Surgery and Anatomy, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Wilson Gomez Manrique
- Animal Health Laboratory, Veterinary Medicine Course, Federal University of Rondonia - UNIR, Rolim de Moura, RO, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Nias Y G Peng
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Naphak Modhiran
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Yin Xiang Setoh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Alexander A Khromykh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Luiz T M Figueiredo
- Virology Research Center, Ribeirao Preto, SP, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Victor H Aquino
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
69
|
Sellier Y, Marliot F, Bessières B, Stirnemann J, Encha-Razavi F, Guilleminot T, Haicheur N, Pages F, Ville Y, Leruez-Ville M. Adaptive and Innate Immune Cells in Fetal Human Cytomegalovirus-Infected Brains. Microorganisms 2020; 8:microorganisms8020176. [PMID: 31991822 PMCID: PMC7074756 DOI: 10.3390/microorganisms8020176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
Background: The understanding of the pathogenesis of cytomegalovirus (CMV)-induced fetal brain lesions is limited. We aimed to quantify adaptive and innate immune cells and CMV-infected cells in fetal brains with various degrees of brain damage. Methods: In total, 26 archived embedded fetal brains were studied, of which 21 were CMV-infected and classified in severely affected (n = 13) and moderately affected (n = 8), and 5 were uninfected controls. The respective magnitude of infected cells, immune cells (CD8+, B cells, plasma cells, NK cells, and macrophages), and expression of immune checkpoint receptors (PD-1/PD-L1 and LAG-3) were measured by immunochemistry and quantified by quantitative imaging analysis. Results: Quantities of CD8+, plasma cells, NK cells, macrophages, and HCMV+ cells and expression of PD-1/PD-L1 and LAG-3 were significantly higher in severely affected than in moderately affected brains (all p values < 0.05). A strong link between higher number of stained cells for HCMV/CD8 and PD-1 and severity of brain lesions was found by component analysis. Conclusions: The higher expression of CD8, PD-1, and LAG-3 in severely affected brains could reflect immune exhaustion of cerebral T cells. These exhausted T cells could be ineffective in controlling viral multiplication itself, leading to more severe brain lesions. The study of the functionality of brain leucocytes ex vivo is needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Yann Sellier
- Service de Gynécologie-Obstétrique, Hôpital Universitaire Necker-Enfants-malades, AP-HP, 149 rue de Sèvres, 75015 Paris, France; (Y.S.); (J.S.)
- EHU 7328 PACT, 75015 Paris, France;
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
| | - Florence Marliot
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- INSERM U872, plateforme d’Immuno-monitoring, service d’Immunologie Biologique, Hôpital Européen Georges-Pompidou, AP-HP, 75015 Paris, France;
| | - Bettina Bessières
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- Service d’histologie-Embryologie-Cytogénétique Hôpital Universitaire Necker-Enfants-malades, AP-HP, 75015 Paris, France
- Institut Imagine, Université Paris Descartes (INSERM U) 1163, 75015 Paris, France
| | - Julien Stirnemann
- Service de Gynécologie-Obstétrique, Hôpital Universitaire Necker-Enfants-malades, AP-HP, 149 rue de Sèvres, 75015 Paris, France; (Y.S.); (J.S.)
- EHU 7328 PACT, 75015 Paris, France;
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
| | - Ferechte Encha-Razavi
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- Service d’histologie-Embryologie-Cytogénétique Hôpital Universitaire Necker-Enfants-malades, AP-HP, 75015 Paris, France
| | - Tiffany Guilleminot
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- Service d’histologie-Embryologie-Cytogénétique Hôpital Universitaire Necker-Enfants-malades, AP-HP, 75015 Paris, France
- Laboratoire de Virologie, Hôpital Universitaire Necker-Enfants-malades, AP-HP, Centre National de Référence, laboratoire associé Cytomégalovirus, 75015 Paris, France
| | - Nacilla Haicheur
- INSERM U872, plateforme d’Immuno-monitoring, service d’Immunologie Biologique, Hôpital Européen Georges-Pompidou, AP-HP, 75015 Paris, France;
| | - Franck Pages
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- INSERM U872, plateforme d’Immuno-monitoring, service d’Immunologie Biologique, Hôpital Européen Georges-Pompidou, AP-HP, 75015 Paris, France;
| | - Yves Ville
- Service de Gynécologie-Obstétrique, Hôpital Universitaire Necker-Enfants-malades, AP-HP, 149 rue de Sèvres, 75015 Paris, France; (Y.S.); (J.S.)
- EHU 7328 PACT, 75015 Paris, France;
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- Correspondence: ; Tel.: +33-1-44-49-63-32
| | - Marianne Leruez-Ville
- EHU 7328 PACT, 75015 Paris, France;
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France; (F.M.); (B.B.); (F.E.-R.); (T.G.); (F.P.)
- Laboratoire de Virologie, Hôpital Universitaire Necker-Enfants-malades, AP-HP, Centre National de Référence, laboratoire associé Cytomégalovirus, 75015 Paris, France
| |
Collapse
|
70
|
Chhatbar C, Detje CN, Grabski E, Borst K, Spanier J, Ghita L, Elliott DA, Jordão MJC, Mueller N, Sutton J, Prajeeth CK, Gudi V, Klein MA, Prinz M, Bradke F, Stangel M, Kalinke U. Type I Interferon Receptor Signaling of Neurons and Astrocytes Regulates Microglia Activation during Viral Encephalitis. Cell Rep 2020; 25:118-129.e4. [PMID: 30282022 PMCID: PMC7103936 DOI: 10.1016/j.celrep.2018.09.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/06/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
In sterile neuroinflammation, a pathological role is proposed for microglia, whereas in viral encephalitis, their function is not entirely clear. Many viruses exploit the odorant system and enter the CNS via the olfactory bulb (OB). Upon intranasal vesicular stomatitis virus instillation, we show an accumulation of activated microglia and monocytes in the OB. Depletion of microglia during encephalitis results in enhanced virus spread and increased lethality. Activation, proliferation, and accumulation of microglia are regulated by type I IFN receptor signaling of neurons and astrocytes, but not of microglia. Morphological analysis of myeloid cells shows that type I IFN receptor signaling of neurons has a stronger impact on the activation of myeloid cells than of astrocytes. Thus, in the infected CNS, the cross talk among neurons, astrocytes, and microglia is critical for full microglia activation and protection from lethal encephalitis.
Collapse
Affiliation(s)
- Chintan Chhatbar
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Claudia N Detje
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Elena Grabski
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - David A Elliott
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Disease Research (DZNE), Bonn, Germany
| | - Marta Joana Costa Jordão
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Nora Mueller
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - James Sutton
- Novartis Institutes for Biomedical Research, Emeryville, CA, USA
| | - Chittappen K Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Michael A Klein
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Disease Research (DZNE), Bonn, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
71
|
Sasai M, Yamamoto M. Innate, adaptive, and cell-autonomous immunity against Toxoplasma gondii infection. Exp Mol Med 2019; 51:1-10. [PMID: 31827072 PMCID: PMC6906438 DOI: 10.1038/s12276-019-0353-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
Hosts have been fighting pathogens throughout the evolution of all infectious diseases. Toxoplasma gondii is one of the most common infectious agents in humans but causes only opportunistic infection in healthy individuals. Similar to antimicrobial immunity against other organisms, the immune response against T. gondii activates innate immunity and in turn induces acquired immune responses. After activation of acquired immunity, host immune cells robustly produce the proinflammatory cytokine interferon-γ (IFN-γ), which activates a set of IFN-γ-inducible proteins, including GTPases. IFN-inducible GTPases are essential for cell-autonomous immunity and are specialized for effective clearance and growth inhibition of T. gondii by accumulating in parasitophorous vacuole membranes. Recent studies suggest that the cell-autonomous immune response plays a protective role in host defense against not only T. gondii but also various intracellular bacteria. Moreover, the negative regulatory mechanisms of such strong immune responses are also important for host survival after infection. In this review, we will discuss in detail recent advances in the understanding of host defenses against T. gondii and the roles played by cell-autonomous immune responses. Researchers are extensively studying immune responses to the single-celled parasite Toxoplasma gondii, which infects around one-third of humans, often harmlessly, but can cause life-threatening toxoplasmosis infections in patients with weakened immune systems. Masahiro Yamamoto and Miwa Sasai at Osaka University in Japan review recent advances in understanding the interactions between the immune system and the parasite. They consider non-specific ‘innate’ immune responses and also the ‘acquired’ responses that target specific parts of the parasite, referred to as antigens. Methods that selectively switch off genes in mice are revealing details presumed to also be relevant for humans. Significant molecules, molecular signaling pathways and immune-regulating processes are being identified. Recent studies suggest cell-autonomous immunity, the ability of host cells to defend themselves against attack, plays a significant role in fighting Toxoplasma gondii infection.
Collapse
Affiliation(s)
- Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan. .,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
72
|
Flavivirus infection—A review of immunopathogenesis, immunological response, and immunodiagnosis. Virus Res 2019; 274:197770. [DOI: 10.1016/j.virusres.2019.197770] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022]
|
73
|
Imaging the dynamic recruitment of monocytes to the blood-brain barrier and specific brain regions during Toxoplasma gondii infection. Proc Natl Acad Sci U S A 2019; 116:24796-24807. [PMID: 31727842 DOI: 10.1073/pnas.1915778116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain infection by the parasite Toxoplasma gondii in mice is thought to generate vulnerability to predation by mechanisms that remain elusive. Monocytes play a key role in host defense and inflammation and are critical for controlling T. gondii However, the dynamic and regional relationship between brain-infiltrating monocytes and parasites is unknown. We report the mobilization of inflammatory (CCR2+Ly6Chi) and patrolling (CX3CR1+Ly6Clo) monocytes into the blood and brain during T. gondii infection of C57BL/6J and CCR2RFP/+CX3CR1GFP/+ mice. Longitudinal analysis of mice using 2-photon intravital imaging of the brain through cranial windows revealed that CCR2-RFP monocytes were recruited to the blood-brain barrier (BBB) within 2 wk of T. gondii infection, exhibited distinct rolling and crawling behavior, and accumulated within the vessel lumen before entering the parenchyma. Optical clearing of intact T. gondii-infected brains using iDISCO+ and light-sheet microscopy enabled global 3D detection of monocytes. Clusters of T. gondii and individual monocytes across the brain were identified using an automated cell segmentation pipeline, and monocytes were found to be significantly correlated with sites of T. gondii clusters. Computational alignment of brains to the Allen annotated reference atlas [E. S. Lein et al., Nature 445:168-176 (2007)] indicated a consistent pattern of monocyte infiltration during T. gondii infection to the olfactory tubercle, in contrast to LPS treatment of mice, which resulted in a diffuse distribution of monocytes across multiple brain regions. These data provide insights into the dynamics of monocyte recruitment to the BBB and the highly regionalized localization of monocytes in the brain during T. gondii CNS infection.
Collapse
|
74
|
Peng BH, Wang T. West Nile Virus Induced Cell Death in the Central Nervous System. Pathogens 2019; 8:pathogens8040215. [PMID: 31683807 PMCID: PMC6963722 DOI: 10.3390/pathogens8040215] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
West Nile virus (WNV), a mosquito-borne, single-stranded flavivirus, has caused annual outbreaks of viral encephalitis in the United States since 1999. The virus induces acute infection with a clinical spectrum ranging from a mild flu-like febrile symptom to more severe neuroinvasive conditions, including meningitis, encephalitis, acute flaccid paralysis, and death. Some WNV convalescent patients also developed long-term neurological sequelae. Neither the treatment of WNV infection nor an approved vaccine is currently available for humans. Neuronal death in the central nervous system (CNS) is a hallmark of WNV-induced meningitis and encephalitis. However, the underlying mechanisms of WNV-induced neuronal damage are not well understood. In this review, we discuss current findings from studies of WNV infection in vitro in the CNS resident cells and the in vivo animal models, and provide insights into WNV-induced neuropathogenesis.
Collapse
Affiliation(s)
- Bi-Hung Peng
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
75
|
Trzeciak A, Lerman YV, Kim TH, Kim MR, Mai N, Halterman MW, Kim M. Long-Term Microgliosis Driven by Acute Systemic Inflammation. THE JOURNAL OF IMMUNOLOGY 2019; 203:2979-2989. [PMID: 31619539 DOI: 10.4049/jimmunol.1900317] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/19/2019] [Indexed: 12/20/2022]
Abstract
Severe sepsis, a systemic inflammatory response to infection, is an increasing cause of morbidity in intensive care units. During sepsis, the vasculature is profoundly altered, leading to release of microbial virulence factors and proinflammatory mediators to surrounding tissue, causing severe systemic inflammatory responses and hypoxic injury of multiple organs. To date, multiple studies have explored pathologic conditions in many vital organs, including lungs, liver, and kidneys. Although data suggest that sepsis is emerging as a key driver of chronic brain dysfunction, the immunological consequence of severe inflammatory responses in the brain remain poorly understood. In this study, we used C57BL/6 sepsis mouse models to establish a disease phenotype in which septic mice with various degrees of severity recover. In the early phases of sepsis, monocytes infiltrate the brain with significantly elevated proinflammatory cytokine levels. In recovered animals, monocytes return to vehicle levels, but the number of brain-resident microglia is significantly increased in the cortex, the majority of which remain activated. The increase in microglia number is mainly due to self-proliferation, which is completely abolished in CCR2 knockout mice. Collectively our data suggest that early monocyte infiltration causes permanent changes to microglia during sepsis, which may ultimately dictate the outcome of future infections and neuropathological diseases.
Collapse
Affiliation(s)
- Alissa Trzeciak
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642
| | - Yelena V Lerman
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642
| | - Tae-Hyoun Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642
| | - Ma Rie Kim
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627
| | - Nguyen Mai
- Department of Neurology, University of Rochester, Rochester, NY 14642; and
| | - Marc W Halterman
- Department of Neurology, University of Rochester, Rochester, NY 14642; and.,Center for Neurotherapeutics Discovery, University of Rochester, Rochester, NY 14642
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642;
| |
Collapse
|
76
|
van de Wouw M, Boehme M, Dinan TG, Cryan JF. Monocyte mobilisation, microbiota & mental illness. Brain Behav Immun 2019; 81:74-91. [PMID: 31330299 DOI: 10.1016/j.bbi.2019.07.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal microbiome has emerged as a key player in regulating brain and behaviour. This has led to the strategy of targeting the gut microbiota to ameliorate disorders of the central nervous system. Understanding the underlying signalling pathways in which the microbiota impacts these disorders is crucial for the development of future therapeutics for improving CNS functionality. One of the major pathways through which the microbiota influences the brain is the immune system, where there is an increasing appreciation for the role of monocyte trafficking in regulating brain homeostasis. In this review, we will shed light on the role of monocyte trafficking as a relay of microbiota signals in conditions where the central nervous system is in disorder, such as stress, peripheral inflammation, ageing, traumatic brain injury, stroke, multiple sclerosis, Alzheimer's disease and Parkinson's disease. We also cover how the gastrointestinal microbiota is implicated in these mental illnesses. In addition, we aim to discuss how the monocyte system can be modulated by the gut microbiota to mitigate disorders of the central nervous system, which will lead to novel microbiota-targeted strategies.
Collapse
Affiliation(s)
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
77
|
Ayala-Nunez NV, Follain G, Delalande F, Hirschler A, Partiot E, Hale GL, Bollweg BC, Roels J, Chazal M, Bakoa F, Carocci M, Bourdoulous S, Faklaris O, Zaki SR, Eckly A, Uring-Lambert B, Doussau F, Cianferani S, Carapito C, Jacobs FMJ, Jouvenet N, Goetz JG, Gaudin R. Zika virus enhances monocyte adhesion and transmigration favoring viral dissemination to neural cells. Nat Commun 2019; 10:4430. [PMID: 31562326 PMCID: PMC6764950 DOI: 10.1038/s41467-019-12408-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) invades and persists in the central nervous system (CNS), causing severe neurological diseases. However the virus journey, from the bloodstream to tissues through a mature endothelium, remains unclear. Here, we show that ZIKV-infected monocytes represent suitable carriers for viral dissemination to the CNS using human primary monocytes, cerebral organoids derived from embryonic stem cells, organotypic mouse cerebellar slices, a xenotypic human-zebrafish model, and human fetus brain samples. We find that ZIKV-exposed monocytes exhibit higher expression of adhesion molecules, and higher abilities to attach onto the vessel wall and transmigrate across endothelia. This phenotype is associated to enhanced monocyte-mediated ZIKV dissemination to neural cells. Together, our data show that ZIKV manipulates the monocyte adhesive properties and enhances monocyte transmigration and viral dissemination to neural cells. Monocyte transmigration may represent an important mechanism required for viral tissue invasion and persistence that could be specifically targeted for therapeutic intervention. Zika virus (ZIKV) can infect the central nervous system, but it is not clear how it reaches the brain. Here, Ayala-Nunez et al. show in ex vivo and in vivo models that ZIKV can hitch a ride in monocytes in a Trojan Horse manner to cross the endothelium and disseminate the virus.
Collapse
Affiliation(s)
- Nilda Vanesa Ayala-Nunez
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France.,Université de Strasbourg, INSERM, 67000, Strasbourg, France
| | | | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Emma Partiot
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France
| | - Gillian L Hale
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Brigid C Bollweg
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Judith Roels
- University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Maxime Chazal
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | - Florian Bakoa
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | - Margot Carocci
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S1255, FMTS, 67000, Strasbourg, France
| | - Sandrine Bourdoulous
- INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Orestis Faklaris
- MRI Core facility, Biocampus, CNRS UMS 3426, 34293, Montpellier, France
| | - Sherif R Zaki
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S1255, FMTS, 67000, Strasbourg, France
| | - Béatrice Uring-Lambert
- Hôpitaux universitaires de Strasbourg, laboratoire central d'immunologie, 67000, Strasbourg, France
| | - Frédéric Doussau
- Institut des Neurosciences Cellulaires et Intégratives, CNRS, Université de Strasbourg, 67000, Strasbourg, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Frank M J Jacobs
- University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Nolwenn Jouvenet
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | | | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France. .,Université de Strasbourg, INSERM, 67000, Strasbourg, France.
| |
Collapse
|
78
|
Hayashida E, Ling ZL, Ashhurst TM, Viengkhou B, Jung SR, Songkhunawej P, West PK, King NJC, Hofer MJ. Zika virus encephalitis in immunocompetent mice is dominated by innate immune cells and does not require T or B cells. J Neuroinflammation 2019; 16:177. [PMID: 31511023 PMCID: PMC6740023 DOI: 10.1186/s12974-019-1566-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022] Open
Abstract
Background Until the end of the twentieth century, Zika virus (ZIKV) was thought to cause a mostly mild, self-limiting disease in humans. However, as the geographic distribution of ZIKV has shifted, so too has its pathogenicity. Modern-day ZIKV infection is now known to cause encephalitis, acute disseminated encephalomyelitis, and Guillain-Barré syndrome in otherwise healthy adults. Nevertheless, the underlying pathogenetic mechanisms responsible for this shift in virulence remain unclear. Methods Here, we investigated the contribution of the innate versus the adaptive immune response using a new mouse model involving intracranial infection of adult immunocompetent mice with a moderately low dose of ZIKV MR766. To determine the contribution of type I interferons (IFN-Is) and adaptive immune cells, we also studied mice deficient for the IFN-I receptor 1 (Ifnar1−/−) and recombination-activating gene 1 (Rag1−/−). Results We show that intracranial infection with ZIKV resulted in lethal encephalitis. In wild-type mice, ZIKV remained restricted predominantly to the central nervous system (CNS) and infected neurons, whereas astrocytes and microglia were spared. Histological and molecular analysis revealed prominent activation of resident microglia and infiltrating monocytes that were accompanied by an expression of pro-inflammatory cytokines. The disease was independent of T and B cells. Importantly, unlike peripheral infection, IFN-Is modulated but did not protect from infection and lethal disease. Lack of IFN-I signaling resulted in spread of the virus, generalized inflammatory changes, and accelerated disease onset. Conclusions Using intracranial infection of immunocompetent wild-type mice with ZIKV, we demonstrate that in contrast to the peripheral immune system, the CNS is susceptible to infection and responds to ZIKV by initiating an antiviral immune response. This response is dominated by resident microglia and infiltrating monocytes and macrophages but does not require T or B cells. Unlike in the periphery, IFN-Is in the CNS cannot prevent the establishment of infection. Our findings show that ZIKV encephalitis in mice is dependent on the innate immune response, and adaptive immune cells play at most a minor role in disease pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12974-019-1566-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emina Hayashida
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia
| | - Zheng Lung Ling
- Discipline of Pathology, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Bosch Institute, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Thomas M Ashhurst
- Discipline of Pathology, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Bosch Institute, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Sydney Cytometry Facility, The University of Sydney and the Centenary Institute, Sydney, Australia
| | - Barney Viengkhou
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia
| | - So Ri Jung
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia
| | - Pattama Songkhunawej
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia
| | - Phillip K West
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia
| | - Nicholas J C King
- Discipline of Pathology, the Marie Bashir Institute for Infectious Diseases and Biosecurity, the Bosch Institute, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Sydney Cytometry Facility, The University of Sydney and the Centenary Institute, Sydney, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, Australia. .,School of Life and Environmental Sciences, The University of Sydney, Molecular Bioscience Bldg., Maze Crescent G08, Sydney, NSW, 2006, Australia.
| |
Collapse
|
79
|
RIG-I-like receptors direct inflammatory macrophage polarization against West Nile virus infection. Nat Commun 2019; 10:3649. [PMID: 31409781 PMCID: PMC6692387 DOI: 10.1038/s41467-019-11250-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/17/2019] [Indexed: 01/20/2023] Open
Abstract
RIG-I-Like Receptors (RLRs) RIG-I, MDA5, and LGP2, are vital pathogen recognition receptors in the defense against RNA viruses. West Nile Virus (WNV) infections continue to grow in the US. Here, we use a systems biology approach to define the contributions of each RLR in the innate immune response to WNV. Genome-wide RNAseq and bioinformatics analyses of macrophages from mice lacking either RLR reveal that the RLRs drive distinct immune gene activation and response polarization to mediate an M1/inflammatory signature while suppressing the M2/wound healing phenotype. While LGP2 functions to modulate inflammatory signaling, RIG-I and MDA5 together are essential for M1 macrophage polarization in vivo and the control of WNV infection through potential downstream control of ATF4 and SMAD4 to regulate target gene expression for cell polarization. These analyses reveal the RLR-driven signature of macrophage polarization, innate immune protection, and immune programming against WNV infection.
Collapse
|
80
|
Park J, Zhang Y, Saito E, Gurczynski SJ, Moore BB, Cummings BJ, Anderson AJ, Shea LD. Intravascular innate immune cells reprogrammed via intravenous nanoparticles to promote functional recovery after spinal cord injury. Proc Natl Acad Sci U S A 2019; 116:14947-14954. [PMID: 31285339 PMCID: PMC6660718 DOI: 10.1073/pnas.1820276116] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Traumatic primary spinal cord injury (SCI) results in paralysis below the level of injury and is associated with infiltration of hematogenous innate immune cells into the injured cord. Methylprednisolone has been applied to reduce inflammation following SCI, yet was discontinued due to an unfavorable risk-benefit ratio associated with off-target effects. In this study, i.v. administered poly(lactide-coglycolide) nanoparticles were internalized by circulating monocytes and neutrophils, reprogramming these cells based on their physicochemical properties and not by an active pharmaceutical ingredient, to exhibit altered biodistribution, gene expression, and function. Approximately 80% of nanoparticle-positive immune cells were observed within the injury, and, additionally, the overall accumulation of innate immune cells at the injury was reduced 4-fold, coinciding with down-regulated expression of proinflammatory factors and increased expression of antiinflammatory and proregenerative genes. Furthermore, nanoparticle administration induced macrophage polarization toward proregenerative phenotypes at the injury and markedly reduced both fibrotic and gliotic scarring 3-fold. Moreover, nanoparticle administration with the implanted multichannel bridge led to increased numbers of regenerating axons, increased myelination with about 40% of axons myelinated, and an enhanced locomotor function (score of 6 versus 3 for control group). These data demonstrate that nanoparticles provide a platform that limits acute inflammation and tissue destruction, at a favorable risk-benefit ratio, leading to a proregenerative microenvironment that supports regeneration and functional recovery. These particles may have applications to trauma and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Yining Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Steve J Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48105
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48105
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105
| |
Collapse
|
81
|
Coulon PG, Dhanushkodi N, Prakash S, Srivastava R, Roy S, Alomari NI, Nguyen AM, Warsi WR, Ye C, Carlos-Cruz EA, Mai UT, Cruel AC, Ekmekciyan KM, Pearlman E, BenMohamed L. NLRP3, NLRP12, and IFI16 Inflammasomes Induction and Caspase-1 Activation Triggered by Virulent HSV-1 Strains Are Associated With Severe Corneal Inflammatory Herpetic Disease. Front Immunol 2019; 10:1631. [PMID: 31367214 PMCID: PMC6644090 DOI: 10.3389/fimmu.2019.01631] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
The crosstalk between the host's inflammasome system and the invading virulent/less-virulent viruses determines the outcome of the ensuing inflammatory response. An appropriate activation of inflammasomes triggers antiviral inflammatory responses that clear the virus and heal the inflamed tissue. However, an aberrant activation of inflammasomes can result in a harmful and overwhelming inflammation that could damage the infected tissue. The underlying host's immune mechanisms and the viral virulent factors that impact severe clinical inflammatory disease remain to be fully elucidated. In this study, we used herpes simplex virus type 1 (HSV-1), the causative agent of corneal inflammatory herpetic disease, as a model pathogen to determine: (i) Whether and how the virulence of a virus affects the type and the activation level of the inflammasomes; and (ii) How triggering specific inflammasomes translates into protective or damaging inflammatory response. We showed that, in contrast to the less-virulent HSV-1 strains (RE, F, KOS, and KOS63), corneal infection of B6 mice with the virulent HSV-1 strains (McKrae, 17 or KOS79): (i) Induced simultaneous expression of the NLRP3, NLRP12, and IFI16 inflammasomes; (ii) Increased production of the biologically active Caspase-1 and pro-inflammatory cytokines IL-1β and IL-18; (iii) Heightened recruitment into the inflamed cornea of CD45highLy6C+Ly6G-F4/80+CD11b+CD11c- inflammatory monocytes and CD45highCD11b+F4/80-Ly6GhiLy6Cmed neutrophils; and (iv) This intensified inflammatory response was associated with a severe corneal herpetic disease, irrespective of the level of virus replication in the cornea. Similarly, in vitro infection of human corneal epithelial cells and human monocytic THP-1 cells with the virulent HSV-1 strains triggered a synchronized early expression of NLRP3, NLRP12 and IFI16, 2 h post-infection, associated with formation of single and dense specks of the adapter molecule ASC in HSV(+) cells, but not in the neighboring bystander HSV(-) cells. This was associated with increased cleavages of Caspase-1, IL-1β, and IL-18. These findings suggest a previously unappreciated role of viral virulence in a synchronized early induction of the NLRP3, NLRP12, and IFI16 inflammasomes that lead to a damaging inflammatory response. A potential role of common virus virulent factors that stimulate this harmful inflammatory corneal disease is currently under investigation.
Collapse
Affiliation(s)
- Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Nisha Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Soumyabrata Roy
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Nuha I. Alomari
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Angela M. Nguyen
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Wasay R. Warsi
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Caitlin Ye
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Edgar A. Carlos-Cruz
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Uyen T. Mai
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Audrey C. Cruel
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Keysi M. Ekmekciyan
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - Eric Pearlman
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
- School of Medicine, Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
- School of Medicine, Institute for Immunology, University of California, Irvine, Irvine, CA, United States
- Department of Molecular Biology and Biochemistry, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
82
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
83
|
Choi JY, Kim JH, Hossain FMA, Uyangaa E, Park SO, Kim B, Kim K, Eo SK. Indispensable Role of CX 3CR1 + Dendritic Cells in Regulation of Virus-Induced Neuroinflammation Through Rapid Development of Antiviral Immunity in Peripheral Lymphoid Tissues. Front Immunol 2019; 10:1467. [PMID: 31316515 PMCID: PMC6610490 DOI: 10.3389/fimmu.2019.01467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
A coordinated host immune response mediated via chemokine network plays a crucial role in boosting defense mechanisms against pathogenic infections. The speed of Ag presentation and delivery by CD11c+ dendritic cells (DCs) to cognate T cells in lymphoid tissues may decide the pathological severity of the infection. Here, we investigated the role of CX3CR1 in the neuroinflammation induced by infection with Japanese encephalitis virus (JEV), a neurotrophic virus. Interestingly, CX3CR1 deficiency strongly enhanced susceptibility to JEV only after peripheral inoculation via footpad. By contrast, both CX3CR1+/+ and CX3CR1-/- mice showed comparable susceptibility to JEV following inoculation via intranasal and intraperitoneal routes. CX3CR1-/- mice exhibited lethal neuroinflammation after JEV inoculation via footpad route, showing high mortality, morbidity, pro-inflammatory cytokine expression, and uncontrolled CNS-infiltration of peripheral leukocytes including Ly-6Chi monocytes and Ly-6Ghi granulocytes. Furthermore, the absence of CX3CR1+CD11c+ DCs appeared to enhance susceptibility of CX3CR1-/- mice to JE after peripheral JEV inoculation. CX3CR1 ablation impaired the migration of CX3CR1+CD11c+ DCs from JEV-inoculated sites to draining lymph nodes (dLNs), resulting in decreased NK cell activation and JEV-specific CD4+/CD8+ T-cell responses. However, CX3CR1-competent mice showed rapid temporal expression of viral Ags in dLNs. Subsequently, JEV was rapidly cleared, with concomitant generation of antiviral NK cell activation and T-cell responses mediated by rapid migration of JEV Ag+CX3CR1+CD11c+ DCs. Using biallelic functional CX3CR1 expression system, the functional expression of CX3CR1 on CD11chi DCs appeared to be essentially required for inducing rapid and effective responses of NK cell activation and Ag-specific CD4+ T cells in dLNs. Strikingly, adoptive transfer of CX3CR1+CD11c+ DCs was found to completely restore the resistance of CX3CR1-/- recipients to JEV, as corroborated by the rapid delivery of JEV Ags in dLNs and attenuation of neuroinflammation in the CNS. Collectively, these results indicate that CX3CR1+CD11c+ DCs play an important role in generating rapid and effective responses of antiviral NK cell activation and Ag-specific T cells after peripheral inoculation with the virus, thereby resulting in conferring resistance to viral infection by reducing the peripheral viral burden.
Collapse
Affiliation(s)
- Jin Young Choi
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Jin Hyoung Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Ferdaus Mohd Altaf Hossain
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea.,Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Erdenebelig Uyangaa
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Seong Ok Park
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Bumseok Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan-si, South Korea
| | - Seong Kug Eo
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| |
Collapse
|
84
|
PD1 +CCR2 +CD8 + T Cells Infiltrate the Central Nervous System during Acute Japanese Encephalitis Virus Infection. Virol Sin 2019; 34:538-548. [PMID: 31215000 DOI: 10.1007/s12250-019-00134-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/08/2019] [Indexed: 12/20/2022] Open
Abstract
Japanese encephalitis (JE) is a viral encephalitis disease caused by Japanese encephalitis virus (JEV) infection. Uncontrolled inflammatory responses in the central nervous system (CNS) are a hallmark of severe JE. Although the CCR2-CCL2 axis is important for monocytes trafficking during JEV infection, little is known about its role in CNS trafficking of CD8+ T cells. Here, we characterized a mouse model of JEV infection, induced via intravenous injection (i.v.) and delineated the chemokines and infiltrating peripheral immune cells in the brains of infected mice. The CNS expression of chemokines, Ccl2, Ccl3, and Ccl5, and their receptors, Ccr2 or Ccr5, was significantly up-regulated after JEV infection and was associated with the degree of JE pathogenesis. Moreover, JEV infection resulted in the migration of a large number of CD8+ T cells into the CNS. In the brains of JEV-infected mice, infiltrating CD8+ T cells expressed CCR2 and CCR5 and were found to comprise mainly effector T cells (CD44+CD62L-). JEV infection dramatically enhanced the expression of programmed death 1 (PD-1) on infiltrating CD8+ T cells in the brain, as compared to that on peripheral CD8+ T cells in the spleen. This effect was more pronounced on infiltrating CCR2+CD8+ T cells than on CCR2-CD8+ T cells. In conclusion, we identified a new subset of CD8+ T cells (PD1+CCR2+CD8+ T cells) present in the CNS of mice during acute JEV infection. These CD8+ T cells might play a role in JE pathogenesis.
Collapse
|
85
|
Sasai M, Pradipta A, Yamamoto M. Host immune responses to Toxoplasma gondii. Int Immunol 2019; 30:113-119. [PMID: 29408976 DOI: 10.1093/intimm/dxy004] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/19/2018] [Indexed: 12/24/2022] Open
Abstract
Toxoplasma gondii can infect homoeothermic animals including humans and cause lethal toxoplasmosis in immunocompromised individuals. When hosts are infected with T. gondii, the cells induce immune responses against T. gondii. The pathogen infection is recognized by immune sensors that directly detect T. gondii structural components, leading to production of pro-inflammatory cytokines and chemokines. Antigen-presenting cells such as macrophages and dendritic cells strongly activate T cells and induce development of Th1 cells and antigen-specific killer CD8 T cells. These T cells and Group 1 innate lymphoid cells are main producers of IFN-γ, which robustly stimulates cell-autonomous immunity in cells infected with T. gondii. IFN-γ-inducible effectors such as IFN-inducible GTPases, inducible nitric oxide synthase and indoleamine-2,3-dioxygenase differentially play important roles in suppression of T. gondii growth and its direct killing in anti-T. gondii cell-autonomous immune responses. In this review, we will describe our current knowledge of innate, adaptive and IFN-γ-mediated cell-autonomous immunity against T. gondii infection.
Collapse
Affiliation(s)
- Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Ariel Pradipta
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
86
|
Niewold P, Cohen A, van Vreden C, Getts DR, Grau GE, King NJC. Experimental severe malaria is resolved by targeting newly-identified monocyte subsets using immune-modifying particles combined with artesunate. Commun Biol 2018; 1:227. [PMID: 30564748 PMCID: PMC6292940 DOI: 10.1038/s42003-018-0216-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/31/2018] [Indexed: 12/29/2022] Open
Abstract
Current treatment of severe malaria and associated cerebral malaria (CM) and respiratory distress syndromes are directed primarily at the parasite. Targeting the parasite has only partial efficacy in advanced infection, as neurological damage and respiratory distress are due to accumulation of host blood cells in the brain microvasculature and lung interstitium. Here, computational analysis identifies Ly6Clo monocytes as a major component of the immune infiltrate in both organs in a preclinical mouse model. Specifically targeting Ly6Clo monocyte precursors, identified by adoptive transfer, with immune-modifying particles (IMP) prevents experimental CM (ECM) in 50% of Plasmodium berghei ANKA-infected mice in early treatment protocols. Furthermore, treatment at onset of clinical ECM with 2 doses of a novel combination of IMP and anti-malarial drug artesunate results in 88% survival. This combination confers protection against ECM and mortality in late stage severe experimental malaria and provides a viable advance on current treatment regimens.
Collapse
Affiliation(s)
- Paula Niewold
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Amy Cohen
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Caryn van Vreden
- 3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| | - Daniel R Getts
- 4Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA.,TcR2, Therapeutics, 100 Binney Street, Suite 710, Cambridge, MA 02142 USA
| | - Georges E Grau
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Nicholas J C King
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia.,3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| |
Collapse
|
87
|
Recasens M, Shrivastava K, Almolda B, González B, Castellano B. Astrocyte-targeted IL-10 production decreases proliferation and induces a downregulation of activated microglia/macrophages after PPT. Glia 2018; 67:741-758. [PMID: 30548340 DOI: 10.1002/glia.23573] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 01/03/2023]
Abstract
When central nervous system (CNS) homeostasis is altered, microglial cells become rapidly activated, proliferate and release a broad range of molecules. Among the plethora of molecules involved in the regulation of microglial activation, cytokines are considered crucial. Although production of interleukin-10 (IL-10) has been demonstrated after different types of CNS injuries and associated with protective functions, the specific role played by IL-10 modulating microglial cells remains unclear. Hence, the objective of this study was to evaluate the effects of transgenic astrocyte IL-10 production on microglial activation associated with axonal anterograde degeneration. To address it, the hippocampal area subjected to perforant pathway transection (PPT) was analyzed by immunohistochemistry (IHC), flow cytometry and protein microarray in transgenic (GFAP-IL10Tg) mice and their corresponding wild types (WT) littermates. Our results demonstrated increased microglial/macrophages density in nonlesioned and PPT-lesioned GFAP-IL10Tg animals when compared with nonlesioned and lesioned WT, respectively. This increase was not due to proliferation, as GFAP-IL10Tg mice showed a reduced proliferation of microglial cells, but was related to an increased population of CD11b+/CD45high monocyte/macrophages. Despite this higher number, the microglia/macrophage population in transgenic animals displayed a downregulated phenotype characterized by lower MHCII, ICOSL, and CD11c. Moreover, a sustained T-cell infiltration was found in transgenic animals. We strongly suggest these modifications must be associated with indirect effects derived from the influence of IL-10 on astrocytes and/or neurons, which express IL-10R. We finally suggested that TGF-β produced by astrocytes, along with IL-2 and CXCL10 might be crucial molecules mediating the effects of transgenic IL-10.
Collapse
Affiliation(s)
- Mireia Recasens
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kalpana Shrivastava
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Beatriz Almolda
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta González
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Bernardo Castellano
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
88
|
Schultz KLW, Troisi EM, Baxter VK, Glowinski R, Griffin DE. Interferon regulatory factors 3 and 7 have distinct roles in the pathogenesis of alphavirus encephalomyelitis. J Gen Virol 2018; 100:46-62. [PMID: 30451651 DOI: 10.1099/jgv.0.001174] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7-/- and IRF3/7-/- mice died within 3-4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3-/- and IRF7-/- mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNβ and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3-/- mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7-/- mice died 5-8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3-/- mice in the induction of higher CNS levels of IFNβ, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7-/- mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.
Collapse
Affiliation(s)
- Kimberly L W Schultz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,†Present address: Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Elizabeth M Troisi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Victoria K Baxter
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,‡Present address: University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rebecca Glowinski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,§Present address: Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
89
|
Hu S, Liu Q, Zang S, Zhang Z, Wang J, Cai X, He X. Microglia Are Derived from Peripheral Blood Mononuclear Cells After Pseudorabies Infection in Mice. Viral Immunol 2018; 31:596-604. [PMID: 30339053 DOI: 10.1089/vim.2018.0064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pseudorabies virus (PRV) can spread along the peripheral nerves near the site of infection in the animals, and gradually migrates into the central nervous system, where it leads to the development of brain lesions. The aim of this study was to investigate the dynamics of microglia after PRV inoculation. A mouse model inoculated with PRV was established to study the interactions between PRV and microglia, microglial recruitment, and polarization effects. The mice were subcutaneously inoculated with different doses of PRV-Bartha K61 vaccine strain. The obtained results showed that mouse mortality rates increased with the applied doses of virus, and brain lesions, located in the brain tail and brain stem, were observed in each investigated group. Inflammatory cells were shown to infiltrate through the vasculature into perivascular cuff, and the number of microglia was increased as well. Mouse group treated with a medium infection dose demonstrated a high survival rate while developing serious brain lesions, and therefore, this dose was selected for further experiments. Immunohistochemistry, flow cytometry, and confocal laser scanning microscopy were used to analyze PRV-microglia interactions. After PRV inoculation, proliferating cell nuclear antigen (Pcna) and Iba1 double-positive cells were observed in the brain lesions, together with the activated microglia, suggesting that PRV can induce microglial proliferation and activation. Furthermore, 5-bromo-deoxy-uridine (BrdU) labeling demonstrated that microglial cells did not proliferate in situ and the proliferating cells originated from peripheral blood monocytes, mainly from the inflammatory monocytes (Ly6Chigh). In addition, microglia polarized into both M1 and M2 phenotypes by PRV infection. The results obtained in this study may help understand the development of pseudorabies infection and help improve the treatment, by recruiting and enhancing immune response.
Collapse
Affiliation(s)
- Shouping Hu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Qiang Liu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Sufang Zang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhuo Zhang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jingfei Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuehui Cai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xijun He
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
90
|
Lesteberg KE, Beckham JD. Immunology of West Nile Virus Infection and the Role of Alpha-Synuclein as a Viral Restriction Factor. Viral Immunol 2018; 32:38-47. [PMID: 30222521 DOI: 10.1089/vim.2018.0075] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
West Nile virus (WNV) is a single-stranded RNA flavivirus and is a major cause of viral encephalitis worldwide. Experimental models of WNV infection in mice are commonly used to define acute neuroinflammatory responses in the brain. Alpha-synuclein (Asyn) is a protein of primarily neuronal origin and is a major cause of Parkinson's disease (PD), a disorder characterized by loss of dopaminergic neurons. Both WNV and PD pathologies are largely mediated by inflammation of the central nervous system (neuroinflammation) and have overlapping inflammatory pathways. In this review, we highlight the roles of the immune system in both diseases while comparing and contrasting both protective and pathogenic roles of immune cells and their effector proteins. Additionally, we review the current literature showing that Asyn is an important mediator of the immune response with diverging roles in PD (pathogenic) and WNV disease (neuroprotective).
Collapse
Affiliation(s)
- Kelsey E Lesteberg
- 1 Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine , Aurora, Colorado
| | - John David Beckham
- 1 Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine , Aurora, Colorado.,2 Division of Neuroimmunology and Neurological Infections, Department of Neurology, University of Colorado School of Medicine , Aurora, Colorado.,3 Veterans Administration, Eastern Colorado Health System , Denver, Colorado
| |
Collapse
|
91
|
Chemokine receptors CCR2 and CX3CR1 regulate viral encephalitis-induced hippocampal damage but not seizures. Proc Natl Acad Sci U S A 2018; 115:E8929-E8938. [PMID: 30181265 PMCID: PMC6156634 DOI: 10.1073/pnas.1806754115] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Viral encephalitis is a major risk factor for the development of seizures, epilepsy, and hippocampal damage with associated cognitive impairment, markedly reducing quality of life in survivors. The mechanisms underlying seizures and hippocampal neurodegeneration developing during and after viral encephalitis are only incompletely understood, hampering the development of preventive treatments. Recent findings suggest that brain invasion of blood-born monocytes may be critically involved in both seizures and brain damage in response to encephalitis, whereas the relative role of microglia, the brain's resident immune cells, in these processes is not clear. CCR2 and CX3CR1 are two chemokine receptors that regulate the responses of myeloid cells, such as monocytes and microglia, during inflammation. We used Ccr2-KO and Cx3cr1-KO mice to understand the role of these receptors in viral encephalitis-associated seizures and neurodegeneration, using the Theiler's virus model of encephalitis in C57BL/6 mice. Our results show that CCR2 as well as CX3CR1 plays a key role in the accumulation of myeloid cells in the CNS and activation of hippocampal myeloid cells upon infection. Furthermore, by using Cx3cr1-creER+/-tdTomatoSt/Wt reporter mice, we show that, with regard to CD45 and CD11b expression, some microglia become indistinguishable from monocytes during CNS infection. Interestingly, the lack of CCR2 or CX3CR1 receptors was associated with almost complete prevention of hippocampal damage but did not prevent seizure development after viral CNS infection. These data are compatible with the hypothesis that CNS inflammatory mechanism(s) other than the infiltrating myeloid cells trigger the development of seizures during viral encephalitis.
Collapse
|
92
|
Pharmacologic Depletion of Microglia Increases Viral Load in the Brain and Enhances Mortality in Murine Models of Flavivirus-Induced Encephalitis. J Virol 2018; 92:JVI.00525-18. [PMID: 29899084 DOI: 10.1128/jvi.00525-18] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses account for most arthropod-borne cases of human encephalitis in the world. However, the exact mechanisms of injury to the central nervous system (CNS) during flavivirus infections remain poorly understood. Microglia are the resident immune cells of the CNS and are important for multiple functions, including control of viral pathogenesis. Utilizing a pharmacologic method of microglia depletion (PLX5622 [Plexxikon Inc.], an inhibitor of colony-stimulating factor 1 receptor), we sought to determine the role of microglia in flaviviral pathogenesis. Depletion of microglia resulted in increased mortality and viral titer in the brain following infection with either West Nile virus (WNV) or Japanese encephalitis virus (JEV). Interestingly, microglial depletion did not prevent virus-induced increases in the expression of relevant cytokines and chemokines at the mRNA level. In fact, the expression of several proinflammatory genes was increased in virus-infected, microglia-depleted mice compared to virus-infected, untreated controls. In contrast, and as expected, expression of the macrophage marker triggering receptor expressed on myeloid cells 2 (TREM2) was decreased in virus-infected, PLX5622-treated mice compared to virus-infected controls.IMPORTANCE As CNS invasion by flaviviruses is a rare but life-threatening event, it is critical to understand how brain-resident immune cells elicit protection or injury during disease progression. Microglia have been shown to be important in viral clearance but may also contribute to CNS injury as part of the neuroinflammatory process. By utilizing a microglial depletion model, we can begin to parse out the exact roles of microglia during flaviviral pathogenesis with hopes of understanding specific mechanisms as potential targets for therapeutics.
Collapse
|
93
|
Pellegatta S, Savoldo B, Di Ianni N, Corbetta C, Chen Y, Patané M, Sun C, Pollo B, Ferrone S, DiMeco F, Finocchiaro G, Dotti G. Constitutive and TNFα-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: Implications for CAR-T cell therapy. Sci Transl Med 2018; 10:eaao2731. [PMID: 29491184 PMCID: PMC8713441 DOI: 10.1126/scitranslmed.aao2731] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/31/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022]
Abstract
The heterogeneous expression of tumor-associated antigens limits the efficacy of chimeric antigen receptor (CAR)-redirected T cells (CAR-Ts) for the treatment of glioblastoma (GBM). We have found that chondroitin sulfate proteoglycan 4 (CSPG4) is highly expressed in 67% of the GBM specimens with limited heterogeneity. CSPG4 is also expressed on primary GBM-derived cells, grown in vitro as neurospheres (GBM-NS), which recapitulate the histopathology and molecular characteristics of primary GBM. CSPG4.CAR-Ts efficiently controlled the growth of GBM-NS in vitro and in vivo upon intracranial tumor inoculation. Moreover, CSPG4.CAR-Ts were also effective against GBM-NS with moderate to low expression of CSPG4. This effect was mediated by the in vivo up-regulation of CSPG4 on tumor cells, induced by tumor necrosis factor-α (TNFα) released by the microglia surrounding the tumor. Overall, the constitutive and TNFα-inducible expression of CSPG4 in GBM may greatly reduce the risk of tumor cell escape observed when targeted antigens are heterogeneously expressed on tumor cells.
Collapse
Affiliation(s)
- Serena Pellegatta
- Unit of Molecular Neuro-Oncology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan 20133, Italy
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Natalia Di Ianni
- Unit of Molecular Neuro-Oncology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Cristina Corbetta
- Unit of Molecular Neuro-Oncology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Yuhui Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Monica Patané
- Unit of Neuropathology, Fondazione IRCCS Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Chuang Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Bianca Pollo
- Unit of Neuropathology, Fondazione IRCCS Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Francesco DiMeco
- Department of Neuro-Surgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
94
|
Bryan MA, Giordano D, Draves KE, Green R, Gale M, Clark EA. Splenic macrophages are required for protective innate immunity against West Nile virus. PLoS One 2018; 13:e0191690. [PMID: 29408905 PMCID: PMC5800658 DOI: 10.1371/journal.pone.0191690] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/09/2018] [Indexed: 01/25/2023] Open
Abstract
Although the spleen is a major site for West Nile virus (WNV) replication and spread, relatively little is known about which innate cells in the spleen replicate WNV, control viral dissemination, and/or prime innate and adaptive immune responses. Here we tested if splenic macrophages (MΦs) were necessary for control of WNV infection. We selectively depleted splenic MΦs, but not draining lymph node MΦs, by injecting mice intravenously with clodronate liposomes several days prior to infecting them with WNV. Mice missing splenic MΦs succumbed to WNV infection after an increased and accelerated spread of virus to the spleen and the brain. WNV-specific Ab and CTL responses were normal in splenic MΦ-depleted mice; however, numbers of NK cells and CD4 and CD8 T cells were significantly increased in the brains of infected mice. Splenic MΦ deficiency led to increased WNV in other splenic innate immune cells including CD11b- DCs, newly formed MΦs and monocytes. Unlike other splenic myeloid subsets, splenic MΦs express high levels of mRNAs encoding the complement protein C1q, the apoptotic cell clearance protein Mertk, the IL-18 cytokine and the FcγR1 receptor. Splenic MΦ-deficient mice may be highly susceptible to WNV infection in part to a deficiency in C1q, Mertk, IL-18 or Caspase 12 expression.
Collapse
Affiliation(s)
- Marianne A. Bryan
- Department of Immunology, University of Washington, Seattle, WA, United States of America
| | - Daniela Giordano
- Department of Immunology, University of Washington, Seattle, WA, United States of America
- The Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, United States of America
| | - Kevin E. Draves
- Department of Immunology, University of Washington, Seattle, WA, United States of America
- The Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, United States of America
| | - Richard Green
- Department of Immunology, University of Washington, Seattle, WA, United States of America
- The Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, United States of America
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA, United States of America
- The Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, United States of America
| | - Edward A. Clark
- Department of Immunology, University of Washington, Seattle, WA, United States of America
- The Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
95
|
Peralta Ramos JM, Bussi C, Gaviglio EA, Arroyo DS, Baez NS, Rodriguez-Galan MC, Iribarren P. Type I IFNs Are Required to Promote Central Nervous System Immune Surveillance through the Recruitment of Inflammatory Monocytes upon Systemic Inflammation. Front Immunol 2017; 8:1666. [PMID: 29255461 PMCID: PMC5722985 DOI: 10.3389/fimmu.2017.01666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/14/2017] [Indexed: 01/09/2023] Open
Abstract
Brain-resident microglia and peripheral migratory leukocytes play essential roles in shaping the immune response in the central nervous system. These cells activate and migrate in response to chemokines produced during active immune responses and may contribute to the progression of neuroinflammation. Herein, we addressed the participation of type I–II interferons in the response displayed by microglia and inflammatory monocytes to comprehend the contribution of these cytokines in the establishment and development of a neuroinflammatory process. Following systemic lipopolysaccharide (LPS) challenge, we found glial reactivity and an active recruitment of CD45hi leukocytes close to CD31+ vascular endothelial cells in circumventricular organs. Isolated CD11b+ CD45hi Ly6Chi Ly6G−-primed inflammatory monocytes were able to induce T cell proliferation, unlike CD11b+ CD45lo microglia. Moreover, ex vivo re-stimulation with LPS exhibited an enhancement of T cell proliferative response promoted by inflammatory monocytes. These myeloid cells also proved to be recruited in a type I interferon-dependent fashion as opposed to neutrophils, unveiling a role of these cytokines in their trafficking. Together, our results compares the phenotypic and functional features between tissue-resident vs peripheral recruited cells in an inflamed microenvironment, identifying inflammatory monocytes as key sentinels in a LPS-induced murine model of neuroinflammation.
Collapse
Affiliation(s)
- Javier María Peralta Ramos
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudio Bussi
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Emilia Andrea Gaviglio
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniela Soledad Arroyo
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia Soledad Baez
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Cecilia Rodriguez-Galan
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo Iribarren
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
96
|
Giordano D, Draves KE, Young LB, Roe K, Bryan MA, Dresch C, Richner JM, Diamond MS, Gale M, Clark EA. Protection of mice deficient in mature B cells from West Nile virus infection by passive and active immunization. PLoS Pathog 2017; 13:e1006743. [PMID: 29176765 PMCID: PMC5720816 DOI: 10.1371/journal.ppat.1006743] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 12/07/2017] [Accepted: 11/10/2017] [Indexed: 01/02/2023] Open
Abstract
B cell activating factor receptor (BAFFR)-/- mice have a profound reduction in mature B cells, but unlike μMT mice, they have normal numbers of newly formed, immature B cells. Using a West Nile virus (WNV) challenge model that requires antibodies (Abs) for protection, we found that unlike wild-type (WT) mice, BAFFR-/- mice were highly susceptible to WNV and succumbed to infection within 8 to 12 days after subcutaneous virus challenge. Although mature B cells were required to protect against lethal infection, infected BAFFR-/- mice had reduced WNV E-specific IgG responses and neutralizing Abs. Passive transfer of immune sera from previously infected WT mice rescued BAFFR-/- and fully B cell-deficient μMT mice, but unlike μMT mice that died around 30 days post-infection, BAFFR-/- mice survived, developed WNV-specific IgG Abs and overcame a second WNV challenge. Remarkably, protective immunity could be induced in mature B cell-deficient mice. Administration of a WNV E-anti-CD180 conjugate vaccine 30 days prior to WNV infection induced Ab responses that protected against lethal infection in BAFFR-/- mice but not in μMT mice. Thus, the immature B cells present in BAFFR-/- and not μMT mice contribute to protective antiviral immunity. A CD180-based vaccine may promote immunity in immunocompromised individuals.
Collapse
Affiliation(s)
- Daniela Giordano
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Kevin E. Draves
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Lucy B. Young
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Kelsey Roe
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Marianne A. Bryan
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Christiane Dresch
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Justin M. Richner
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Michael S. Diamond
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
- The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, United States of America
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, United States of America
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, United States of America
| | - Edward A. Clark
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
97
|
Liu S, Pan S, Tan J, Zhao W, Liu F. Oxytocin inhibits ox-LDL-induced adhesion of monocytic THP-1 cells to human brain microvascular endothelial cells. Toxicol Appl Pharmacol 2017; 337:104-110. [PMID: 29104011 DOI: 10.1016/j.taap.2017.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 12/28/2022]
Abstract
The attachment of monocytes to human brain microvascular endothelial cells (HBMVEs) caused by oxidized low-density lipoprotein (ox-LDL) is associated with an early event and the pathological progression of cerebrovascular diseases. Oxytocin (OT) is a human peptide hormone that is traditionally used as a medication to facilitate childbirth. However, little information is available regarding the physiological function of OT in brain endothelial dysfunction. In the present study, our results indicate that the oxytocin receptor (OTR) was expressed in human brain microvascular endothelial cells (HBMVEs) and was upregulated in response to ox-LDL in a concentration-dependent manner. Notably, OT significantly suppressed ox-LDL-induced attachment of THP-1 monocytes to HBMVEs. Furthermore, we found that OT reduced the expression of adhesion molecules, such as VCAM-1 and E-selectin. Interestingly, it was shown that OT could restore ox-LDL-induced reduction of KLF4 in HBMVEs. Importantly, knockdown of KLF4 abolished the inhibitory effects of OT on ox-LDL-induced expressions of VCAM-1 and E-selectin as well as the adhesion of human monocytic THP-1 cells to endothelial HBMVEs. Mechanistically, we found that the stimulatory effects of OT on KLF4 expression are mediated by the MEK5/MEF2A pathway.
Collapse
Affiliation(s)
- Shuyan Liu
- Department of neurology, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu 212300, China
| | - Shengying Pan
- Department of neurology, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu 212300, China
| | - Jing Tan
- Department of neurology, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu 212300, China
| | - Weina Zhao
- Department of neurology, Hongqi Hospital Affiliated to Mudanjiang Medical university, Mudanjiang, Heilongjiang 157000, China
| | - Fengguo Liu
- Department of neurology, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu 212300, China; Department of neurology, Hongqi Hospital Affiliated to Mudanjiang Medical university, Mudanjiang, Heilongjiang 157000, China.
| |
Collapse
|
98
|
Michlmayr D, Andrade P, Gonzalez K, Balmaseda A, Harris E. CD14 +CD16 + monocytes are the main target of Zika virus infection in peripheral blood mononuclear cells in a paediatric study in Nicaragua. Nat Microbiol 2017; 2:1462-1470. [PMID: 28970482 PMCID: PMC5997390 DOI: 10.1038/s41564-017-0035-0] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/01/2017] [Indexed: 01/13/2023]
Abstract
The recent Zika pandemic in the Americas is linked to congenital birth defects and Guillain-Barré syndrome. White blood cells (WBCs) play an important role in host immune responses early in arboviral infection. Infected WBCs can also function as 'Trojan horses' and carry viruses into immune-sheltered spaces, including the placenta, testes and brain. Therefore, defining which WBCs are permissive to Zika virus (ZIKV) is critical. Here, we analyse ZIKV infectivity of peripheral blood mononuclear cells (PBMCs) in vitro and from Nicaraguan Zika patients and show CD14+CD16+ monocytes are the main target of infection, with ZIKV replication detected in some dendritic cells. The frequency of CD14+ monocytes was significantly decreased, while the CD14+CD16+ monocyte population was significantly expanded during ZIKV infection compared to uninfected controls. Viral RNA was detected in PBMCs from all patients, but in serum from only a subset, suggesting PBMCs may be a reservoir for ZIKV. In Zika patients, the frequency of infected cells was lower but the percentage of infected CD14+CD16+ monocytes was significantly higher compared to dengue cases. The gene expression profile in monocytes isolated from ZIKV- and dengue virus-infected patients was comparable, except for significant differences in interferon-γ, CXCL12, XCL1, interleukin-6 and interleukin-10 levels. Thus, our study provides a detailed picture of the innate immune profile of ZIKV infection and highlights the important role of monocytes, and CD14+CD16+ monocytes in particular.
Collapse
Affiliation(s)
- Daniela Michlmayr
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, 94720-3370, CA, USA
| | - Paulina Andrade
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, 94720-3370, CA, USA
- Universidad de San Francisco de Quito, Quito, 170157, Ecuador
| | - Karla Gonzalez
- Sustainable Sciences Institute, Managua, 14007, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, 16064, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, 14007, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, 16064, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, 94720-3370, CA, USA.
| |
Collapse
|
99
|
Minocycline Has Anti-inflammatory Effects and Reduces Cytotoxicity in an Ex Vivo Spinal Cord Slice Culture Model of West Nile Virus Infection. J Virol 2017; 91:JVI.00569-17. [PMID: 28878079 DOI: 10.1128/jvi.00569-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/27/2017] [Indexed: 12/13/2022] Open
Abstract
West Nile virus (WNV) is a neurotropic flavivirus that can cause significant neurological disease. Mouse models of WNV infection demonstrate that a proinflammatory environment is induced within the central nervous system (CNS) after WNV infection, leading to entry of activated peripheral immune cells. We utilized ex vivo spinal cord slice cultures (SCSC) to demonstrate that anti-inflammatory mechanisms may also play a role in WNV-induced pathology and/or recovery. Microglia are a type of macrophage that function as resident CNS immune cells. Similar to mouse models, infection of SCSC with WNV induces the upregulation of proinflammatory genes and proteins that are associated with microglial activation, including the microglial activation marker Iba1 and CC motif chemokines CCL2, CCL3, and CCL5. This suggests that microglia assume a proinflammatory phenotype in response to WNV infection similar to the proinflammatory (M1) activation that can be displayed by other macrophages. We now show that the WNV-induced expression of these and other proinflammatory genes was significantly decreased in the presence of minocycline, which has antineuroinflammatory properties, including the ability to inhibit proinflammatory microglial responses. Minocycline also caused a significant increase in the expression of anti-inflammatory genes associated with alternative anti-inflammatory (M2) macrophage activation, including interleukin 4 (IL-4), IL-13, and FIZZ1. Minocycline-dependent alterations to M1/M2 gene expression were associated with a significant increase in survival of neurons, microglia, and astrocytes in WNV-infected slices and markedly decreased levels of inducible nitric oxide synthase (iNOS). These results demonstrate that an anti-inflammatory environment induced by minocycline reduces viral cytotoxicity during WNV infection in ex vivo CNS tissue.IMPORTANCE West Nile virus (WNV) causes substantial morbidity and mortality, with no specific therapeutic treatments available. Antiviral inflammatory responses are a crucial component of WNV pathology, and understanding how they are regulated is important for tailoring effective treatments. Proinflammatory responses during WNV infection have been extensively studied, but anti-inflammatory responses (and their potential protective and reparative capabilities) following WNV infection have not been investigated. Minocycline induced the expression of genes associated with the anti-inflammatory (M2) activation of CNS macrophages (microglia) in WNV-infected SCSC while inhibiting the expression of genes associated with proinflammatory (M1) macrophage activation and was protective for multiple CNS cell types, indicating its potential use as a therapeutic reagent. This ex vivo culture system can uniquely address the ability of CNS parenchymal cells (neurons, astrocytes, and microglia) to respond to minocycline and to modulate the inflammatory environment and cytotoxicity in response to WNV infection without peripheral immune cell involvement.
Collapse
|
100
|
Duyen HTL, Cerny D, Trung DT, Pang J, Velumani S, Toh YX, Qui PT, Hao NV, Simmons C, Haniffa M, Wills B, Fink K. Skin dendritic cell and T cell activation associated with dengue shock syndrome. Sci Rep 2017; 7:14224. [PMID: 29079750 PMCID: PMC5660158 DOI: 10.1038/s41598-017-14640-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/12/2017] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of severe dengue remains unclear, particularly the mechanisms underlying the plasma leakage that results in hypovolaemic shock in a small proportion of individuals. Maximal leakage occurs several days after peak viraemia implicating immunological pathways. Skin is a highly vascular organ and also an important site of immune reactions with a high density of dendritic cells (DCs), macrophages and T cells. We obtained skin biopsies and contemporaneous blood samples from patients within 24 hours of onset of dengue shock syndrome (DSS), and from healthy controls. We analyzed cell subsets by flow cytometry, and soluble mediators and antibodies by ELISA; the percentage of migratory CD1a+ dermal DCs was significantly decreased in the DSS patients, and skin CD8+ T cells were activated, but there was no accumulation of dengue-specific antibodies. Inflammatory monocytic cells were not observed infiltrating the skin of DSS cases on whole-mount histology, although CD14dim cells disappeared from blood.
Collapse
Affiliation(s)
- Huynh Thi Le Duyen
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Daniela Cerny
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dinh The Trung
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Jassia Pang
- Biological Resource Centre (BRC), Singapore, Singapore
| | - Sumathy Velumani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Ying Xiu Toh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Phan Tu Qui
- Hospital for Tropical Diseases, 764 Vo Van Kiet, Ho Chi Minh City, Vietnam
| | - Nguyen Van Hao
- Hospital for Tropical Diseases, 764 Vo Van Kiet, Ho Chi Minh City, Vietnam.,University of Medicine and Pharmacy of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Cameron Simmons
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam.,Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Muzlifah Haniffa
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bridget Wills
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam. .,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom.
| | - Katja Fink
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|