Open access peer-reviewed chapter

Neurotropic Virus-Induced Meningoencephalomyelitis

Written By

Fareeha Saadi, Debanjana Chakravarty, Grishma Kasle and Jayasri Das Sarma

Submitted: 06 January 2022 Reviewed: 14 January 2022 Published: 25 February 2022

DOI: 10.5772/intechopen.102674

From the Edited Volume

RNA Viruses Infection

Edited by Yogendra Shah

Chapter metrics overview

243 Chapter Downloads

View Full Metrics

Abstract

Meningoencephalomyelitis emanates under the umbrella relating inflammatory changes of the Central Nervous System (CNS). Meningitis denotes inflammation in the meningeal layers, encephalitis is an acute diffuse inflammation of the brain, and inflammation in the spinal cord is denoted as myelitis. These can be interrelated or independent of each other depending on the etiology. The entire mechanism of meningoencephalomyelitis is governed by an acute innate inflammatory branch followed by a chronic progressive, adaptive branch of immunity with clinical signs like hyperthermia, weight loss, hypoxia, leukocytosis. This book chapter will focus on viral-induced meningitis, encephalitis, and myelitis. Thirty years of experience working with a murine-β-coronavirus (m-CoV); Mouse hepatitis virus (MHV)-A59 induced experimental model system provided us a thorough understanding of neuroglial cell-mediated acute neuroinflammation, denoted by the accumulation of leukocyte-common-antigen (LCA) positive or CD45+ leukocytes in perivascular infiltrates referred to as perivascular cuff formation and microglial nodules in the brain parenchyma, which mimics specific pathology of human neurological disease multiple sclerosis (MS). Additionally, in this chapter, we summarized the role of CNS resident microglial activation and its interaction with peripheral migratory T cells in mounting neuropathogenesis and host immunity in different families of neurotrophic encephalomyelitis viruses that cause CNS inflammation.

Keywords

  • meningioencephalomyelitis
  • demyelination
  • axonal loss
  • murine-β-coronavirus (m-CoV)
  • neuroinflammation
  • multiple sclerosis (MS)

1. Introduction

Encephalitis is a pathological entity that refers to the inflammation of the encephalon or the brain parenchyma due to infection, autoimmunity, and brain injury. It is a rare medical condition with clinically serious consequences ranging from headaches, fever, seizures, permanent disability, and brain damage. Encephalitis majorly affects infants and the elderly above the age of 65, whereas, the incidence is transitional in the youth and is of significant public health importance because of the associated morbidity and mortality [1]. It results in varied clinical symptoms, such as mild fever and headaches to severe cognitive impairment accompanied by loss of physical vigor and unconsciousness or even life-threatening symptoms that can result in permanent brain damage [2, 3]. It is believed that the severe inflammation associated with encephalitis causes swelling in the brain, which in turn gives rise to headaches, stiff neck, mental confusion, and even seizures [1, 4]. Though cases are recognized in all populations and ages, pediatric populations, young adults, and especially males have a higher propensity to encephalitis [5, 6, 7].

Infection by a virus is the most common and important cause of encephalitis [8]. Virus infections can also cause aseptic meningitis and myelitis [9, 10]. Research on viral encephalitis has gained much momentum with the recognition of encephalitis in human immunodeficiency virus (HIV) infection of the CNS and the emerging viruses such as West Nile virus (WNV), Nipah virus, and severe acute respiratory syndrome viruses (SARS-CoV and SARS-CoV-2) [11]. Members of several virus families like flaviviruses, paramyxoviruses, alphaviruses, bunyaviruses, orthomyxoviruses, arenaviruses, enteroviruses, rhabdoviruses, and astroviruses are also known to cause encephalitis [12].

Viruses may directly enter into the CNS or replicate away from the CNS at first and gain entry to the CNS through various routes [13, 14]. Local factors, like pH, mucosal immune responses, or the integrity of skin and mucosal barriers, govern the entry of the virus into the CNS and resultant encephalitis. Virus entry and replication activate the CNS resident immune cells, which, together with peripheral leukocytes, induce host immune response and promote encephalitis and neuroinflammation, resulting in multiple changes to the CNS physiology [15]. Histopathologically, characteristic microglial nodule formation, i.e., the accumulation of activated microglia in the brain parenchyma and the perivascular cuff formation, is observed in encephalitic brains [16, 17, 18]. Once the virus clears, the acute behavioral symptoms are resolved; however, long-term psychiatric, neurocognitive, and degenerative issues persist due to the ongoing immune responses in the CNS even after pathogen clearance [18, 19].

Interestingly, people infected with neurotropic viruses may not always develop encephalitis, indicating that host cell factors may play a critical role in regulating the outcome of the disease process. While mounting shreds of evidence are available to understand these host factors, limited information is available to understand the genomic control of the pathogenic properties and host factors that mediate a balance between neurovirulence and neuroprotection. Host cell response pathways like UPR and ER stress and oxidative stress may alter due to robust viral replication and intracellular assembly, causing imbalance between reactive oxygen species and antioxidants. These are governed by a battery of cellular mediators like DJ-1, Nrf-2, catalase, HMOX, MMPs, NADPH oxidase, cytokines, chemokines, and secondary messengers. These mediators are either CNS resident proteins or may be produced by the CNS resident neuro-glial cells like microglia, astrocytes, endothelial cells, and peripherally derived leukocytes that enter the CNS upon inflammation and breaching of the blood-brain barrier [13]. The nexus of these immune-inflammatory mediators with endogenous host proteins are well studied for ages in mounting host immunity. The question lies in whether host immunity plays a protective or pathogenic role.

Moreover, understanding the inflammatory mechanisms of meningioencephalomyelitis is a challenge in human patients due to the unavailability of the high throughput data from non-invasive techniques like MRI, fMRI, CT, and detailed invasive histopathological data from punched-biopsy/autopsy tissues. Thus, a thorough understanding of the cellular factors ranging from genomic control of pathogenic properties to viral host factors and immunomodulatory effects requires cause-effect relationship experimental animal models/systems that can provide detailed insight into the disease process instrumental in the diagnosis and designing therapeutics. Though the use of mouse models to understand human disease has its limitations, critical pathological features of encephalitis can be efficiently reproduced in viral-induced experimental models. This book chapter will majorly summarize studies on viral encephalitis and its consecutive neuroinflammatory demyelination and axonal loss.

Advertisement

2. Routes of infection emanating to neurotropic virus infection

The term “neurotropic” refers to affinity towards the nervous system and displays the properties of neuroinvasion (entry into the CNS), and has direct neuroglial tropism. The viral entry to the neuroglial cells may be via receptor-mediated endocytosis and its fusion with the cell cytoplasm. It may also enter via direct endocytosis irrespective of engaging a receptor. Mounting shreds of evidence reported that neurotropic virus enters the brain parenchyma from the olfactory epithelium or retinal ganglionic cells via retrograded axonal transport. Upon entry to the brain parenchyma, infectious virus particles may also follow anterograde axonal transport via the optic nerve to reach retinal ganglionic cells and also can spread to different anatomic regions of the brain like the hippocampus, cortex, anterior commissure, basal forebrain, amygdala, brain stem as well as down the spinal cord. The neurotropic viruses can also travel through the lung-brain axis and cause inflammation in the brain stem region, the respiratory center. Neurotropic viruses can also access brain parenchyma via the gut-brain axis through the vagus nerve.

Advertisement

3. Neurotropic virus infection in mice is employed as an experimental model system to understand the underlying mechanisms of encephalitis, poliomyelitis, neuroinflammation, and demyelination concurrent with axonal loss

Neuroinflammation is responsible for initiating direct neuroglial dystrophy, which in turn can activate CNS resident microglia to release immune modulators. Microglia, the major resident immune cells in the central nervous system (CNS), are considered as the key cellular mediators of neuroinflammatory processes. Microglial research has become a central focus in cellular neuroimmunology and neuroinflammation in the past few years. Chronic/remitting neurological disease such as multiple sclerosis (MS) has long been considered an inflammatory autoimmune disease with the infiltration of peripheral myelin-specific T cells into the CNS. With the rapid advancement in the field of microglia and astrocytic neurobiology, the term neuroinflammation progressively started to denote chronic CNS cell-specific inflammation in MS. The direct glial responses in MS are different from conventional peripheral immune responses. This book chapter attempts to summarize current findings of neuroinflammatory responses within the CNS by direct infection of neural cells by mouse hepatitis virus (MHV) and the mechanisms by which glial cell responses ultimately contribute to the meningioencephalomyelitis and demyelination concurrent with axonal loss (Figure 1). Microglia can be persistently infected by MHV. Microglial activation and phagocytosis are recognized to be critically important in the pathogenesis of demyelination. Emerging evidence for the pathogenic role of microglia and the activation of inflammatory pathways in these cells in MHV infection supports the concept that microglia-induced neuroinflammation is an amplifier of virus-induced neuropathology. Conventional understanding was that the peripheral immune cells are the major players for mounting CNS inflammatory responses. But the current studies revealed that if microglial activation and its immune modulators can check the infection, then the peripheral immune system need not be involved in mounting host immunity, and meningioencephalomyelitis may not shadow. In most cases, CNS resident microglial activation sets the stage for innate immune inflammation that results in the proinflammatory milieu of cytokines and chemokines, known as cytokine storm, which, while trying to combat pathogens, also causes damage to the CNS tissues. Amelioration of the proinflammatory condition requires anti-inflammatory cytokines where the peripheral immune system plays a major role. A series of recent studies on neurotropic murine β-coronavirus demonstrated acute-innate neuroinflammation mediated by CNS resident microglial interplay with peripheral leucocytes comprising monocytes and neutrophils NKT cells, CD4+ and CD8+ T cells to eradicate the pathogen and protect host tissue against aberrant tissue damage (Figure 2). In the below-mentioned sections of this book chapter, we are discussing in detail MHV infection as a prototype of β-coronavirus infection and its pathogenesis to understand the underpinning mechanism of meningioencephalomyelitis, demyelination and axonal loss.

Figure 1.

Disease kinetics and pathological manifestations of murine β-CoV, MHV-A59 intracranial inoculation in C57BL/6 mice, a model to understand viral induced meningioencephalomyelitis and demyelination concurrent with axonal loss. (A) Shows the timeline of infection namely acute stage and its corresponding neuropathologies: meningitis denoting inflammation in the meningeal layers, encephalitis,an acute diffuse inflammation of the brain, and inflammation in the spinal cord denoted as myelitis and chronic progressive demyelination characterized by myelin loss concurrent with axonal loss. (B) Kinetics of MHV-A59 replication and viral clearance represented by viral RNA and infectious viral particles respectively and occurrence of demyelination [20].

Figure 2.

Temporal kinetics of CNS resident glial cell activation associated with peripheral cell migration in response to RSA59 infection in the mouse CNS is key to cause meningioencephalomyeltits. Intracranial inoculation of RSA59 directly infects CNS resident neuroglial cells that in-turn activates CNS resident immune-glial cells like astrocytes and microglia. Activated CNS resident cells secrete a large number of inflammatory mediators like pro-inflammatory cytokines and chemokines. Microglial activation and its pro-inflammatory mileu in the inflamed CNS make a chemoattractant gradient to help the migration of peripheral leukocytes in the CNS. A differential infiltration of total myeloid (neutrophils, macrophages/monocytes, and microglia) and lymphoid (CD4, CD8, and NKT) cell populations observed at different time post infection is critical for orchestration of the clearance of the viral particle mounting host immunity by balancing the pro-inflammatory condition with the anti-inflammatory condition and restoring the CNS homeostasis.

3.1 MHV

Mouse hepatitis virus (MHV) is a β-CoV of the family Coronaviridae. It poses no threat to humans but shows similarities with other human viruses of the same family, such as SARS-CoV, MERS-CoV, and SARS-CoV-2 though they are evolutionarily distinct. MHV can infect the CNS and cause white matter lesions, which makes it an excellent viral model of neuroinflammatory demyelinating disease. Depending on the inoculation route and the strain of MHV-CoV, different outcomes are expected [21].

For example, a highly neurovirulent strain of MHV, JHM, J2.2-V-1, upon intracranial inoculation, induces a monophasic disease course, characterized by inflammatory cell infiltrates in the CNS with subsequent demyelination and clinical symptoms of hind limb weakness, ataxia, and paralysis [22, 23]. No auto-reactive T cells have ever been found in the CNS of J2.2-V-1-infected mice, and the disease is the resultant of virus-specific T cells, which indicated that virus alone can cause myelin destruction. Earlier it was believed that demyelination in JHM infection may be solely due to the lytic oligodendrocyte infection [24], but with the application of immune-deficient animal models, it became clear that immune-mediated mechanisms may be more important [25].

MHV-A59, a hepatotropic and neurotropic MHV strain, caused demyelination in C57BL/6 mice even in the absence of B and T cells [20]. The disease upon MHV-A59 intracranial administration also follows a biphasic course, where encephalomyelitis is characteristic of an acute phase peaking during days 5/6 post infection (p.i.) and chronic stage where demyelination and axonal degeneration peak on day 30 p.i. [26, 27, 28, 29]. Thus, it can be said that different, but related MHV strains may induce demyelination via distinct mechanisms. MHV-A59 induced neuroinflammation and neuroimmune modulation mediated neuroglial dystrophy is triggered by the activation of cellular censors like Toll-like receptor (TLRs)/Rig-I-like receptor (RLRs)/synthase for the second messenger cyclic GMP-AMP and the cyclic GMP-AMP receptor stimulator of interferon genes (cGAS-STING) which can further activate the interferon regulatory factors (IRFs), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and downstream type I interferon (IFN) genes. Acute-innate neuroinflammation is rather dependent on the CNS resident immune cell activation in association with peripheral derived myeloid cells, which in turn involve lymphoid cells in ameliorating proinflammatory condition and bring the anti-inflammatory condition in order to restore the homeostasis of the CNS compartment [30].

Studies are focused on understanding mechanisms from cellular sensing to the disease outcome comprising the MHV-A59 induced neuroinflammation encompassing encephalitis and microglial nodule formation and its progressive myelin pathology concurrent with axonal pathology. We have used and compared spike gene recombinant strains of MHV, a demyelinating strain (DM) RSA59, and non-demyelinating strain (NDM) RSMHV2 to understand the genomic control of encephalitic properties. A plethora of studies from the eminent scientists in this field, along with ours, have contributed to understanding the pathogenesis of MHV infection. Strains of MHV can cause direct CNS cell infection or access the CNS via retrograde axonal transport, but irrespective of the route, they cause encephalitis [31, 32, 33].

3.1.1 Comparative studies between spike gene recombinants murine coronaviruses RSA59 and RSMHV2 to understand the genomic control of meningioencephalomyelitic properties

Using targeted RNA recombination, two isogenic spike protein recombinant strains of MHV, RSA59, and RSMHV2 (background is from demyelinating strain MHV-A59) were generated. For RSA59, the spike was taken from the parental demyelinating strain MHV-A59, and RSMHV2 had the spike from the parental non-demyelinating MHV-2 strain. Enhanced green fluorescent protein (EGFP) was also inserted in the recombinants by replacing the nonessential gene 4a and part of 4b in the MHV-A59 genome by heterologous targeted recombination [34].

Comparative studies between the two recombinants revealed similar pathology to their parental strains for RSA59 and RSMHV2, respectively. Intracerebral (IC) inoculation of RSA59 in 4-week-old C57BL/6 mice caused acute hepatitis, neuroinflammation comprising of meningitis, encephalitis, myelitis, and chronic demyelination and axonal loss, characterized by lymphocytic infiltrates and microglial nodules with focal neuronophagia associated lepto-meningitis [29, 35]. IC inoculation of RSMHV2 caused acute stage hepatitis, meningitis, and encephalitis but no myelitis or chronic demyelination. The encephalitis was indeed more robust compared to RSA59. MHV-2 does not induce encephalitis; it cannot even enter the brain parenchyma and restricts to the meninges inducing meningitis alone [34].

Both RSA59 and RSMHV2 showed similar infection in the brain where they successfully infected and replicated in meninges, the site of inoculation (near the lateral geniculate nucleus), ventral striatum/basal forebrain, hippocampus, and brainstem, and infect the neurons, astrocytes, microglia, and oligodendrocytes in the brain. However, they show different tropism in the spinal cord. DM infects the grey matter neurons and takes an axonal route to be released at the nerve end, whereas in the white matter, it preferentially infects oligodendrocytes [36]. By day 7 p.i. most of the viruses have traversed to the spinal cord white matter. Though NDM can also infect the neurons in the grey matter, they fail to infect the white matter oligodendrocytes [35, 36] due to their inefficiency to translocate through neurites and fusion at the nerve end [37]. The difference in the disease outcome can be attributed to their differential spinal cord tropism and persistence. Thus, it can be inferred that the combined action of spike mediated axon transport of DM strain to evade the heightened immune response and ability to infect the white matter oligodendrocytes and persist in the white matter could be the key to inducing demyelination and axonal loss during the chronic phase of neuroinflammation. DM strain-induced axonal loss and myelin loss are associated with profuse accumulation of macrophages filled with myelin debris within the demyelinating plaques which is not observed in NDM strain infection. High resolution TEM microscopy revealed that microglia/macrophages are indeed responsible for direct myelin stripping, leading to demyelination [29, 35, 38].

It is important to note that neuroinflammation and encephalitis in MHV infection is accompanied with pronounced activation of CNS resident immune cells, microglia, and astrocytes. Upon activation, they take their characteristic activated phenotype and start expressing microglia/macrophage-specific protein Iba1 (ionized calcium-binding adaptor molecule 1), which promotes ruffling and phagocytosis [20, 38]. Detailed Affymetrix microarray analysis revealed that both RSA59 and RSMHV2 initiate innate immune responses during the acute phase with the expression of chemokines like CXCL10, CXCL9, CCL5, and CCL12 and other CD molecules that represent activation of microglia/macrophages [39]. Results also showed the induction of antiviral host response represented by the expression of perforins and IFN gamma signaling genes. Together, the acute stage innate-immune responses and encephalitis were comparable in both RSA59 and RSMHV2 infection [39].

The inflammatory responses gradually declined in RSMHV2 infection following virus clearance, but RSA59 chronic infection showed persistent microglia in the demyelinating plaques and the production of microglia-associated inflammatory mediators. Studies have shown that IFN responses can promote phagolysosomes maturation and autophagy in the persistently activated microglia/macrophages, which can promote myelin sheath engulfment leading to demyelination. This evidence demonstrated that RSA59 induced demyelination could occur through innate immune neuroinflammation denoted by meningioencephalomyelitis triggered during the acute infection stage. Although innate immune responses contribute partially towards controlling of initial virus spread, virus-specific T cell effector functions are essential to eliminate the infectious virus load during most acute infections. Control of m-CoV spread requires the functioning of both CD4+ and CD8+ T cells [40]. CD8+ T cells are the primary effectors but require support from CD4+ T cells. A recent study in CD4−/− mice showed impaired RSA59 clearance, despite the presence of functional CD8+ T cells, demonstrating the importance of CD4+ T cells for the efficient functioning of CD8+ T responses [41].

The distinct cause-effect relationship-driven studies demonstrate that RSA59 infection can be instrumental towards understanding a direct CNS resident immune cell-mediated as well as antibody-mediated encephalomyelitis and demyelination pathologies.

Both DM and NDM strains show a reduction in the expression of genes responsible for innate immune response, and this reduction is more pronounced in the NDM strain-infected mice. In contrast, the genes involved in adaptive immune cell response are upregulated only in DM strain, specifically during the chronic stage of spinal cord infection. A significant upregulation of genes involved in T helper cell signaling pathways, B-cell development, and communication between innate and adaptive immune cells as well as of the expression of IgG genes are observed in the DM strain infection leading to chronic pathology but not in NDM strain [42].

While MHV infection in mouse is a prototype to understand the cellular and molecular consequences of encephalitis and demyelination, Theiler’s murine encephalomyelitis virus (TMEV) in SJL mice also serves as another excellent experimental model of MS because of its histopathological and immunological similarities with MS.

3.2 Theiler’s murine encephalomyelitis virus (TMEV) induced encephalitis

TMEV is a non-enveloped, single-stranded positive-sense RNA virus belonging to the family Picornaviridae and also used as a model to understand the immune-mediated mechanism of demyelination [43, 44].

Upon intracranial (i.c.) infection in SJL mice induces characteristic polioencephalomyelitis in the CNS. In a biphasic CNS disease, first, during the acute phase immune cells including the CD4+ T cells and CD8+ T cells infiltrate the CNS in response to profuse virus replication, and inflammation [45]. They exert rather protective effects by helping clear the virus from the grey matter and result in immune-mediated encephalomyelitis [46]. During the chronic phase, TMEV persistently infects glial cells and macrophages in the white matter, further, there is an infiltration of leukocytes, including macrophages, TMEV-specific T cells and antibodies. The immune effectors (CD4+ and CD8+ T cells) that exerted protective functions during the acute phase exert detrimental effects in the chronic phase by participating in epitope spreading to myelin antigens resulting in severe immune pathology in the white matter of the spinal cords, which causes demyelination [47]. White matter lesions harbor monocytes/macrophages and a few B cells in addition to the T lymphocytes [48]. Initially, the CD4+ T cells recognized the abundant myelin protein PLP, but later, the CD4+ T cells subsets start to recognize subdominant myelin protein epitopes and led to autoreactivity [47]. Additionally, the CD8+ T cells have also been suggested to function as autoreactive cytotoxic cells or regulatory cells in TMEV infection [49]. TMEV induced CNS pathology is immunologically mediated like in MS, wherein MHC plays an important role, and substantial similarities exist in neuropathology, including axonal damage and remyelination [43]. Also, like MS, T-cell apoptosis is less in TMEV induced disease [50].

Cytokines are known to play important functions in the induction and regulation of immune responses against the neurotropic virus. Similarly, the cytokine production by the CNS resident cells astrocytes and microglia, as well as the peripheral immune cells such as T cells and macrophages heavily, influence the encephalitic response induced by TMEV infection [51, 52]. Studies have shown that a critical balance between the inflammatory cytokines governing the propagation of antiviral response to clear the virus during early infection and controlling of immune pathology and establish homeostasis during the late stage. That being said, no particular cytokine pattern is yet established and successfully associated with resistance or susceptibility to TMEV-induced encephalitis and demyelination by the different strains of the virus. A study compared the cytokine response between TMEV DA-infected susceptible (SJL) and resistant (B6) mice. Results showed a high expression of proinflammatory cytokines (IFN-γ, TNF-α, IL-1, IL-2, and IL-6) and low levels of anti-inflammatory cytokines (IL-4, IL-5, and IL-10) in the brains of both SJL and B6 mice during the early acute phase decreasing thereafter [51]. However, only in the SJL mice after a peak of the inflammatory response during day 8–12 p.i. in the brain with a minimum recorded during days 20–25 p.i., the second wave of inflammatory cytokine production was observed later in the spinal cord, which could explain the inflammatory demyelination only in the SJL mice. TGF-β, an important anti-inflammatory cytokine, has shown a significantly higher upregulation in SJL mice compared to B6 [51]. TGF-β can specifically inhibit the cytotoxic T lymphocyte (CTL) response [53] which is noted to be significantly impaired in the SJL mice resulting in reduced TMEV clearance and persistence, leading to virus-mediated encephalitis and demyelination.

Advertisement

4. Overview of microglial activation in encephalomyelitis: amplifier of virus-induced neuropathology

In the context of viral encephalitis that is characterized by an inflammatory response with meningeal, perivascular, and parenchymal infiltrates of peripheral leukocytes, studies have revealed that microglial activation acts as a double-edged sword [16]. On the one hand they promote multiple antiviral functions; microglia sense the ATP released by virus-infected neurons through the purinergic receptor P2Y12 and quickly migrate towards the infected neurons to exert their phagocytic activity [54]. They directly exert their antiviral effect by producing type 1 interferon (IFN-1), inducing IFN-stimulated gene (ISG) to activate corresponding signaling pathways [55]. Additionally, microglia induce autophagy and secrete cytokines to clear the virus from the tissue [56, 57, 58]. On the other hand, their persistent activation leads to tissue damage due to autophagy and apoptotic pathway activation, presynaptic membrane damage in the hippocampus mediated by the complement system activation, which further results in long-term memory impairment and cognitive dysfunction in patients with viral encephalitis [59, 60, 61, 62].

The most common and important example of virus-induced chronic brain infection is the HIV [63]. HIV-induced encephalitis is typified by the accumulation of activated microglia in nodules-like phenotype throughout the parenchyma [64, 65]. HIV enters the CNS via the myelomonocytic cells such as monocytes, perivascular cells, and microglia [66]. HIV particularly targets and disables microglia in the CNS and T cells in the periphery, the key players in neuroinflammation [64, 67]. In fact, the persistence of HIV in microglia indicates that the virus uses the cells as the reservoir [68]. Even though reports suggest that microglia may perform protective functions early on during HIV infections, their functions are considerably compromised. Most studies suggest that active infection of microglia results in their secretion of a variety of neurotoxins, increasing neural apoptosis and neuronal autophagy [16]. Astrogliosis is another characteristic pathology following microglial activation that, together with microgliosis, ultimately leads to myelin paleness and neuronal loss. Patients with advanced AIDS are likely to develop severe encephalitis upon human cytomegalovirus (HCMV) infection [69, 70]. HCMV infection is characterized by microglial nodular encephalitis consisting of microglia, astrocytes, and giant cells, and ventriculoencephalitis and is the main cause of dementia in AIDS patients [71].

Microglia are susceptible to congenital Zika virus (ZIKV) infection [61]. Histopathological analysis showed that ZIKV infects and activates microglia in the perivascular regions causing localized neuroinflammation [61]. Further, the virus is disseminated throughout the parenchyma, which is later associated with neuron damage, especially in the cortical regions [72]. Pronounced neuronal injury results in microcephaly noted in many cases of congenital ZIKV infection [72].

E3 ubiquitin ligase pellino (pelia) expressed by microglia promotes the replication of West nile virus (WNV) in microglia and neurons [73]. It also induces NF-κB and/or p38-MAPK signaling in the microglia that causes an upregulation of inflammatory cytokines and chemokines, leading to peripheral leukocytes infiltration [73, 74]. The robust neuroinflammation may lead to lethal WNV encephalitis.

It is interesting to note that many viruses from diverse families of viruses have been studied in microglia depletion models. Results from most studies showed increased viral replication upon microglial depletion [16, 75, 76, 77]. Additionally, microglial depletion was also associated with overt neurological symptoms and/or death along with high viral burden, which indicated their importance in survival in encephalitis [77]. Though confirmatory results on how these protective functions are exerted by microglia are lacking, some studies show a dependence of T cell responses on microglia activation.

Studies on several mouse models of viral encephalitis have shown that viral clearance depends on efficient T cell responses, including WNV, MHV, and TMEV [78, 79, 80, 81]. TMEV model shows strain-specific differences in disease phenotype and viral clearance, which was associated with underlying differences in CD8+ T cell responses subject to Treg suppression [82, 83]. Further investigation revealed that microglia depletion did not impact CD8+ T cell recruitment but resulted in increased infiltration of Tregs, which caused clinical severity in C57BL/6 mice which is normally not susceptible to TMEV induced disease [84]. In the MHV-induced neuroinflammation model, both CD8+ and CD4+ T cells are implicated in viral clearance, but CD4+ T cells have also been reported to contribute to pathogenesis [80, 85, 86]. A study showed that microglial depletion in mice infected with JHMV strain of MHV, rJ2.2 significantly reduced the infiltration of CD4+ T cells and Tregs in the CNS along with a significant reduction in IFN-γ expression by CD4+ T cells, but there was no impact on the CD8+ T cell population [77]. Thus, showing the importance of microglia in especially orchestrating virus-specific CD4+ T cell response.

In addition to these studies using the JHMV strain, MHV-A59 or its isogenic recombinant strain RSA59 have also elucidated a critical communication between microglia and CD4+ T cell response. Using a CD4−/− mice model very recent study demonstrated for the first time that the mice are highly susceptible to RSA59 induced chronic demyelination with axonal loss [80]. Though the overall inflammation was not affected during the early time-points (day 5–6) i.e., the acute neuroinflammation phase, the CD11b + microglial activation was significantly impaired. The entire inflammatory response was skewed towards an M2 type which was also reflected in the persistence of characteristic amoeboid shaped phagocytic microglia in the CNS of the mice during the chronic phase (day 30 p.i.). Encephalitis, which usually resolves after the acute phase in RSA59 infection, persisted for as long as day 30 p.i. The brain stems of CD4−/− mice were populated with CD11b + microglia surrounding bulbar vacuolated pathology, which signified axonal death and damage [80]. Additionally, CD4+ T cell deficiency resulted in severe grey matter inflammation in the form of poliomyelitis in the spinal cords as well as the dorsal root ganglion (Figure 3). Together these results showed a critical interdependence of microglia and CD4+ T cells in RSA59 infection. Typically, M2 microglial activation fails to resolve during the chronic infection, rendering mice more susceptible to demyelination and axonal bulbar vacuolation [80].

Figure 3.

CD4 deficiency causes poliomyelitis and the dorsal root ganglionic inflammation at acute phase and abnormal bulbar vacuolation at chronic phase of RSA59 infection. Serial sections of spinal cord, dorsal root ganglion and brain from CD4+/+ and CD4−/− mouse were immunostained with anti-CD45, LFB and/or H&E. CD45 immunostining showed heightened poliomyelitis/inflammation of gray matter and inflammation of the dorsal root ganglia in the CD4−/− mice compared to CD4+/+ mice at the acute phase of infection. Spinal cord sections form these mice when further analyzed at chronic stage for demyelination by LFB showed increased myelin loss in CD4−/− mice compared to CD4+/+ mice, CD45 + inflammatory cells were observed in demyelinating lesions of both wildtype and CD4 deficient mice but were elevated in case of CD4−/− mice. Sagittal sections of the brain at chronic stage, stained with H&E showed large number of vacuoles in the brain stem region denoting abnormal bulbar vacuolation which was populated with and Cd11b + micgrogila/monocyte macrophages. Adapted from Chakravarty et al [41].

A very recent study on neurotropic coronavirus MHV-RSA59 infection in Ifit2−/− mice revealed that Ifit2 protects mice from uncontrolled replication and spread throughout the brain parenchyma as well as the spinal cord. Ifit2 deficiency showed pronounced morbidity and mortality in RSA59 infected mice. Furthermore, microglial activation in the CNS was impaired in infected Ifit2−/− mice compared to WT infected mice, and as a consequence, peripheral lymphocyte specifically NK1.1 T cells and CD4+ T cells migration to the CNS was restricted in the Ifit2−/− mice possibly contributing to the lack of viral clearance. Impaired microglial activation and reduced migration of inflammatory cells in the CNS may be associated with less encephalitis and devoid of mounting host immunity. These deficiencies were associated with a lower level of microglial expression of CX3CR1, the cognate receptor of the CX3CL1 (fractalkine) chemokine, which plays a critical role in both microglial activation and leukocyte recruitment. These findings highlighted a pivotal role of interferon stimulating genes and its tetratricopeptide protein as host cell factors in the induction of encephalitis and uncovered a new potential role of an interferon-induced protein in immune protection (Figure 4) [30].

Figure 4.

Ifit2−/− mice upon a murine β-CoV RSA59 infection show increased viral spread and decreased microglia/macrophage activation. About 4–5-week-old Ifit2−/− mice upon RSA59 infection showed a robust viral replication and antigen spread throughout the brain parenchyma compared to the wildtype mice infection. Infectious viral load was significantly higher in the Ifit2−/− mice when assessed by plaque assay (A). The cryosections form Ifit2−/− and wildtype mice brain sections showed similar overall distribution of RSA59 but the total EGFP expression (viral antigen) was more in Ifit2−/− mice (A). At day 5 p.i. H&E staining for the sagittal sections of the whole brain form WT (B) and Ifit2−/− mice (I) showed much milder meningitis (J) and encephalitis characterized by perivascular cuffing (K) and microglial nodule (L) formation in the Ifit2−/− compared to WT (C–E) mice. Similarly activated microglia/macrophage were much less in the Ifit2−/− mice (M–O) compared to the WT (F–H) mice as seen in Iba1 immunostaining. (P) Brain section of Ifit2−/− mice showed heightened viral infection as evident by the profuse viral N protein immunostaining. Ifit2−/− mice however showed a comparatively decreased Iba1 immunostaining indicative of impaired activation of microglia/macrophages compared to the wildtype mice. Adapted form Das Sarma et al [30].

Taking the above-mentioned experimental evidence of the role of CD4+ T cells and monocyte/macrophage activation in viral-induced neuroinflammation, further studies were geared to explore the interactome between the CD4+ T cell expressed CD40 Ligand and CD40 expressed on microglia. CD40-CD40L dyad is an important immune dyad that controls both CD4+ T cell and microglia functions [87, 88]. Our studies in CD40L−/− mice showed that the absence of CD40L renders mice highly susceptible to RSA59 infection due to reduced microglia/macrophage activation during the acute phase of infection required to eliminate the virus (Figure 5) [89]. Effector CD4+ T recruitment to the CNS is significantly dampened, and due to the impaired CD40-CD40L signaling in CD40L−/− mice, their priming is reduced substantially in the draining lymph nodes [89]. Effector CD4+ T cell population was reduced as well as the antiviral response was diminished, and phagocytic microglia persisted in the CNS at a substantial amount in the CD40L−/− mice. As a result, CD40L−/− mice exhibited greater demyelination, axonal loss, and persistent poliomyelitis at the chronic phase of infection [89]. Together, these studies highlight that migration of peripheral T cells and their interaction with microglia via CD40-CD40L is essential to eliminate the virus and provide long-term neuroprotection.

Figure 5.

RSA59 infection in CD40L deficient mice showed impaired microglia/macrophage activation during acute phase of neuroinflammation but causes profuse chronic demyelination concurrent with diminished PLP staining. (a and b) CD40L−/− and WT mice at day 5 upon RSA59 infection showed acute encephalitis and myelitis.Iba1 immunostaining in these brain and spinal cord sections showed that the CD40L−/− mice showed reduced Iba1+ cells compared to WT mice. (c) At day 30 p.i., spinal cord of CD40L−/− mice showed more intense demyelination and reduced PLP staining compared to wildtype mice. Adapted from Saadi et al [89].

Independent of the effect on T cells, IFN produced by microglia acts on other cells that exert indirect antiviral effects [90]. For example, microglia induce antiviral functions in neurons via STING signaling and stimulate IFN-1 production in astrocytes by the TLR3 pathway [91]. Studies have shown that the Type 1 IFNAR signaling in astrocytes helps to protect the blood-brain barrier against virus infection and immunopathology [92]. Depletion of IFNAR signaling in astrocytes resulted in increased inflammatory cytokines and chemokines production, which caused blood-brain barrier inflammation during neurotropic viral infection [92].

Additionally, microglia also mediate viral clearance by autophagy [59]. Viral infections induce NFkB-dependent inflammatory effectors that produce antiviral molecules, including those promoting autophagy. ZIKV infection in Drosophila induces a stimulator of interferon genes (dSTING) in the brain, which promotes autophagy and helps protect the brain [93]. In mammals, autophagy has been shown to restrict HSV-1 infection [94]. Autophagy by microglia helps to clear the virus without causing cell death which protects mature neurons. Microglial phagocytosis is another mechanism that helps protect the neurons from severe damage. Microglia and neurons express C3aR that recognizes C3 cleavage products. In response to C3 and its cleavage products, microglia surround the neurons and phagocytose the presynaptic ends of the neurons [95]. This prevents the trans-synaptic spread of the virus and keeps neurons from firing abnormal signals that may result in cognitive impairment and physical disabilities.

Additionally, detrimental effects of microglia are also reported in many studies on viral encephalitis. Microglia are reported to remain persistently activated in several viral infections [80, 89]. Their activation is further associated with the production of TNF-α [96, 97], which can activate the astrocytic TNFR-1 pathway [98]. This signaling accentuates their crosstalk with neurons leading to modification of the excitatory synapses, which emerges in cognitive disabilities. TNF-α secreted by microglia can directly affect synaptic transmission and plasticity [62]. ZIKV infection has been associated with neurological damage among infants. Studies have found that ZIKV majorly infects the fetal microglia and activates them [61]. This induces an intense pro-inflammatory response by the secretion of mediators like IL-6, TNF-α, IL-1β, and MCP-1 [61]. Also, in HIV encephalitis, many microglia genes undergo significant changes, including immune activation and function, kinases, phosphatases, and pro-/anti-apoptotic and neurotrophic factors, which indicates that microglia functions are compromised and skewed towards pro-inflammation [99].

Thus, it can be ascertained that, microglia are not only important to maintain homeostasis in the CNS but are also critical for responding to injury, infection, and neurodegeneration. Often microglia act quickly in response to injury but with varied stimuli received, their activation profile can differ and may result in harmful or beneficial effects. It is true that viral encephalitis has caused high morbidity, which is a grave concern, but it is also imperative that research on microglia and viral encephalitis can provide new and efficient targets for treatment. Considering the unique response of microglia with different viral infections and at different stages of encephalitis, it is needless to say that the current research on microglia and viral encephalitis remains is at a nascent state. Depending on the type of encephalitis, careful fine-tuning of microglial activation has the potential to improve the therapeutic effect of encephalitis, its prognosis and also reduce the sequelae of encephalitis.

Advertisement

5. Encephalitis caused by several other neurotropic virus families

Many viruses from numerous virus families in different geographical areas can induce immediate or delayed neuropathological manifestations in humans and animals [18, 100]. Infection by neurotropic viruses and their resultant immune response has the potential to irreversibly disrupt the complex structural and functional dynamics of the central nervous system, frequently leaving the patient with a poor or fatal prognosis. The incidences of virus-induced CNS disorder are significantly higher than the damage caused by other pathogens.

Members of several virus families are known to be neurotropic, e.g., herpes family viruses, flaviviruses, paramyxoviruses, alphaviruses, bunyaviruses, orthomyxoviruses, arenaviruses, enteroviruses, rhabdoviruses, coronaviruses, and picornaviruses. Specifically, some of the viruses from these families are viruses like SARS-CoV, MERS-CoV, herpes simplex virus, poliovirus, West Nile virus (WNV), Chikungunya virus (CHIKV), Zika virus (ZIKV), Japanese encephalitis virus (JEV), La Crosse encephalitis (LACV), Epstein-Barr virus (EBV), measles, and mumps viruses, among many others [100, 101]. These viruses have been frequently associated with significant encephalitis, as well as meningitis and myelitis in the CNS. The clinical disease outcome of the CNS virus infection depends on several factors, like the host immune status, viral genomic constitution, and other environmental factors [100]. There is also evidence suggesting coronaviruses such as α-CoVs (NL63 and 229E) and the β-CoVs (OC43, HKU1) being positive-sense single-stranded, enveloped RNA viruses, can induce numerous neurological manifestations along with systemic inflammations in humans. A very recent outbreak of human β coronavirus SARS-CoV-2 is primarily known for its ARDS and paramount evidence suggest that it may enter the brain via olfactory route or can enter through the lung brain axis or gut-brain axis and is also known to cause meningitis, encephalitis, and demyelination [102, 103, 104].

The etiology of CNS infections induced by viruses can also depend on and vary across variable geographical locations. For example, reports have shown that herpes simplex viruses are the most common pathogens observed among both children and adults in the United States (US), Australia, and Italy. In contrast, in Southeast Asian countries like southern Vietnam, the Japanese encephalitis virus has been shown to be one of the most frequent inducers of viral encephalitis, especially among children. Enteroviruses have been commonly isolated to be involved in causing encephalitis in several parts of India. At the same time, HSVs have been observed to be more prevalent in the eastern parts of India, both among adults as well as children. Furthermore, the virulence of the viruses also varies geographically [105]. Therefore, understanding the etio-biology and epidemiology of neurotropic viruses is paramount in designing the targeted intervention.

Therefore, based on epidemiological prevalence and episodic occurrence evidence as well as the employment of these viruses as experimental model systems for human disorders, this book chapter will also briefly discuss some of the other viruses.

Flaviviruses are the enveloped, single-stranded, positive-sense RNA viruses that consist of the world’s most clinically critical viruses like the following species: Japanese encephalitis virus (JEV), tick-borne encephalitis virus (TBEV), and Powassan encephalitis virus (POWV), as well as other mosquito-borne viruses, like Dengue virus (DENV), yellow fever virus (YFV), West Nile virus (WNV), St. Louis encephalitis virus (SLEV), and Zika virus (ZIKV) [106, 107]. JEV is highly prevalent in the Southeast Asian countries as well as the Indian subcontinent, affecting infants and children, and can also be transmitted to the fetus during pregnancy [108, 109]. The incidences of Tick-borne encephalitis (transmitted to humans by the bites of ticks) are progressively expanding in European and Asian countries with severe neurological complications [110]. WNV infection is endemic in temperate and tropical regions throughout the world, triggering yearly outbreaks of encephalitis [111]. St. Louis encephalitis virus (SLEV) is found predominantly in North, Central, and South America and accounts for nearly 35–60% of meningitis in all symptomatic cases in children [112, 113]. Zika virus is also an emerging pathogen with substantial clinical impact significantly on the CNS, as reported, in the form of severe congenital malformations (microcephaly) and neurological complications, mainly Guillain-Barré syndrome (GBS) [114]. It has shown explosive outbreaks in African, South, and Central American countries [112, 115].

Alphaviruses like Eastern equine encephalitis virus (EEEV), Western equine encephalitis virus (WEEV), and Venezuelan equine encephalitis virus (VEEV), as well as the Mayaro virus (MAYV), Una virus (UNAV), and Chikungunya virus (CHIKV) are the small, enveloped viruses with a single-stranded, positive-sense RNA [100, 113]. The most critical neurotropic alphavirus is VEEV, which induced many outbreaks in South, Central, and North America [116]. CHIKV has also caused severe neurological complications in humans at all age groups, especially in infants, in Europe, Asia, and Africa [117, 118]. EEEV can also induce encephalitis in humans in about 50–75% of the cases [8, 119].

Herpes family viruses which are double-stranded DNA viruses, have been commonly associated with severe encephalitis and meningitis in the CNS and have been distributed globally. The members of herpes family viruses that are shown to be neurotropic include HSV types 1 and 2, varicella-zoster, Epstein-Barr virus, and cytomegalovirus [120, 121]. Both children and immunocompromised individuals are most vulnerable to herpes simplex meningoencephalitis [120]. Another critical property to herpes family viruses, especially varicella-zoster virus, is reactivation [122]. Primary infection with VZV during childhood induces chickenpox, but the virus becomes latent in the spinal and cranial ganglia. However, deteriorating cellular immunity with senescence or immunocompromised conditions may lead to virus reactivation that promotes zoster (shingles) [123, 124].

Paramyxoviruses that induce neurological diseases are from genera Rubulavirus (consisting of the mumps virus), which is neurotropic [125]; Morbillivirus genera (consisting of measles virus) [126] and Henipavirus with the emerging Nipah virus (NiV) being one of the neurotropic variants [127]. These are single-stranded, nonsegmented RNA viruses [128]. Measles virus-induced encephalitis is one of the leading causes of morbidity and mortality in the developing world [129]. Nipah virus is one of the emerging viruses that present with numerous cases of acute encephalitis in humans [127, 130].

Lymphocytic choriomeningitis virus (LCMV) belonging to the family Arenaviridae is an enveloped, single-stranded RNA virus. Although its primary host is mice, it is also present in other rodents and has the ability to infect humans, especially laboratory workers, pet owners, and individuals living in impoverished conditions. It is predominant in Europe, Asia, American continents, and Africa [131, 132, 133].

Picornaviruses are single-stranded, non-enveloped RNA viruses encompassing enteroviruses (echoviruses, coxsackieviruses) and parechoviruses (PeVs) pathogenic against humans. Infants and children are highly susceptible to human pathogenic picornaviruses that induce aseptic meningitis and meningoencephalitis [134, 135]. It is highly predominant in the UK, Ireland, the US and some Southeast Asian countries [105, 136, 137].

Advertisement

6. Chronic viral encephalitis and neurodegeneration

The research on viral encephalitis is a rather dynamic and large field. However, its relationship with neurodegeneration is less explored. As research on viral encephalitis and neurodegenerative disease is progressing on diverse fronts many similarities are being identified between the classical neurodegenerative pathways and viral-induced neurodegeneration [138, 139]. It is well established that many neurotropic viruses result in neuronal dysfunction which can have devastating life threatening consequences for the host [138, 140, 141]. Virus can either directly infect the neurons and kill the cells directly by replication and lysis or by apoptosis as observed in poliomyelitis [139, 142]. Additionally viral-induced encephalitis can damage the neurons in an immune mechanism as all neurotropic virus infections irrespective of the route of entry can trigger both innate and adaptive immune responses [139, 143, 144]. Supporting this concept, a number of viruses have been associated with neurodegeneration outcomes [141]. For example, In addition to causing encephalitis, CMV infection can result in transverse myelitis [145], HIV is associated with severe dementia [146] and Echo virus can cause neuro-muscular disorder [147]. Additionally, yearlong infection with JEV in humans can cause postencephalitic parkinsonism (PEP) which shows symptoms similar to sporadic Parkinson’s disease (PD) [148]. Evidence also suggests that H5N1 influenza virus induces many PD-like symptoms [149, 150]. Specifically, the virus first infects the peripheral nervous system (PNS) and later gains entry into the CNS where it causes degeneration of susceptible dopamine (DA) neurons in midbrain regions similar to PD patients [149, 151]. Another influenza virus strain H3N2 causes many neurodegenerative symptoms like amyotrophy, MS flares and relapsing delirium [152, 153]. However, the direct role of viruses in neurodegeneration is less understood. Most likely, several viruses have developed means to evade the immune response and are present in subclinical levels [154, 155, 156]. The local inflammatory response in the CNS in response to persistent virus infections results in chronic encephalomyelitis even without overt cell death which may lead to neuronal damage resulting in degeneration.

HSV-1 is one of the most common virus infections that can remain dormant in the neurons for life-long [157]. It is highly neurotropic and periodic reactivation is observed to establish productive infection of the neurons [140]. It is one of the largely associated viruses with Alzheimer’s disease (AD) [158]. The virus presence is detected in the AD brains, in fact the presence of HSV-1 DNA on APOE gene carriers is a risk factor for AD [159]. Studies also showed HSV-1 DNA and amyloid β to be present in close proximity in AD plaques [160, 161]. Mechanistically, HSV-1 infection can promote neurotoxic Aβ accumulation, tau phosphorylation and cleavage as observed in vitro [161, 162, 163]. In addition to the direct interaction which the virus exploits to travel to the cell surface it also interferes with post-transcriptional regulation by up regulating microRNA-146a, which is another marker for AD [164].

Parkinson’s disease (PD) the second most common neurodegenerative disorder has been linked to Influenza viruses [140, 165]. Highly neurotropic and pathogenic H5N1 virus can enter the CNS, induce encephalitis associated with microglial activation, loss of dopaminergic neurons and accumulation of α-synuclein aggregates in infected regions resembling PD symptoms and pathology [149, 166]. It is well documented that with the 1918 epidemic of H1N1 has greatly increased the incidence of PD [166]. Both H1N1 and H5N1 are found in the substantia nigra region which is also majorly affected in PD patients [167]. In fact, post-mortem brain sections from PD patients show the presence of influenza A virus [168]. No direct mechanism is yet established but is majorly thought to be a contribution of the neuroinflammation process activated by the virus in the CNS [151]. Moreover, HIV infection of the CNS is associated with amyotrophic lateral sclerosis (ALS), which is a fatal neurodegenerative disease with characteristic degeneration of the spinal cord and cortical neurons [169, 170]. Several other neurological symptoms are associated with HIV infections; the most common is HIV-associated dementia which shows certain complications presented with MS [171, 172]. HIV-associated dementia (HAD) also has many similarities with AD and PD including the target anatomical region hippocampus and substantia nigra [173]. The similarities of HAD with neurodegenerative disorders is reported at genomic, proteomic as well as transcription levels [140].

Modern times have seen a drastic increase in the average life expectancy which has come with its own limitation i.e., the incidence of ageing disorders. As discussed, virus infection can significantly act as co factors of neurodegeneration if not the causative agents. Also, the mechanism of viral and non-viral neurodegeneration is not very different [138, 140]. Both show an involvement of immune system by means of neuroinflammation and direct or indirect damage of neurons. Viruses can significantly modulate the structure and function of cytoskeletal proteins that are instrumental in neuronal dysfunction associated with neurodegeneration [174]. Thus, neurotropic virus infection for encephalitis and neuroinflammation can serve as excellent models for understanding neurodegeneration. Moreover, a better understanding of targeting the immune system, which has profound implications, especially in viral-induced neurodegeneration and deciphering the critical overlaps and distinctions between classical neurodegeneration and viral-induced neurodegeneration, can lead to developing new and efficient therapeutic strategies.

Advertisement

7. Conclusion

Encephalitis was considered a rare syndrome, but the incidence of encephalitis is likely to be elevated than previously estimated. This underrated approach towards understanding encephalitis prevails because it is challenging to diagnose, manage and study. Encephalitis is a condition with multiple etiologies and pathogeneses, ranging from direct infectious to immune-mediated; however, each of these specific mechanisms is diverse and often incompletely understood. Accurate diagnosis of encephalitis cases is made complicated because of the difficulties involved in distinguishing between encephalitic and non-encephalitis mimics, autoimmune vs. infectious encephalitis, and the limitation of standard clinical case/laboratory definitions. Ancillary testing, and clinical correlations, along with a clinical follow-up, are important to establish more specific diagnoses. Determining the etiology is the key first step to improving patient outcomes, and it needs advanced neuropathologic and clinical algorithms. Furthermore, the most recent antibody-associated forms of encephalitis also pose a challenge due to their significantly varying clinical manifestations. Understanding the antigenic specificity of intrathecal IgGs found in the CSF may help to identify clues to the cause of infection or inflammation in several cases of encephalitis.

Also, it is critical to study host-immune responses and other host factors to design novel therapeutic interventions, given the paradoxical role of the immune system in encephalitis. The increased urbanization, travel, and climate change are some of the factors, which contribute to the evolution and spread of new pathogens. Infectious diseases are emerging profoundly. Neuroinfectious diseases might occur as occurrences in small, localized regions or may rapidly spread over large geographical areas as pandemics, just like SARS-CoV2. Understanding emerging viruses need better experimental animal models, which will help to comprehend the cause-effect relationship between the virus and its associated neuropathogenesis. Strong consideration should be given to trials of combination therapy that include treatment strategies with both anti-inflammatory and anti-pathogen drugs.

Advertisement

Acknowledgments

Neurotopic m-CoV research in an experimental animal model has provided a timely and appropriate platform to take a step toward writing this comprehensive book chapter on encephalitis. This work has been a collaborative effort for two decades between the Indian Institute of Science Education and Research Kolkata (IISER-K), India; Indian Institute of Sciences (IISc), India; University of Pennsylvania, USA; Thomas Jefferson University, USA and University of Colorado Denver, USA. The joint endeavor has been supported and nurtured by the Indo-U.S. Science and Technology Forum (IUSSTF) on several occasions. m-CoV research was generously funded by the Department of Biotechnology (DBT), India, Council for Scientific and Industrial Research (CSIR), India; National Multiple Sclerosis Society, USA; M.E. Groff Surgical Medical Research and Education Charitable Trust; and Lindback Foundation Career Enhancement Award, USA. The authors would also like to express their gratefulness to Dr. Dhriti Chatterjee, Dr. Kaushiki Biswas, Dr. Abhinoy Kishore, Dr. Subhajit Das Sarma, Dr. Afaq Hussain, Mr. Soumya Kundu, Mr. Abhishek Bose, Mr. Sourodip Sengupta, Ms. Mithila Kamble, Ms. Vaishali Mulchandani, Mr. Saurav, Mr. A. Safiriyu Abass, Dr. Mahua Maulik, and Ms. Soma Nag who have contributed immensely to the generation of a large volume of work on m-CoV research over the years. Authors would like to thank Dr. Kenneth S. Shindler, Dr. Lawrence C. Kenyon, and Dr. Randall J. Cohrs for their long-standing collaboration on m-CoV research. The success of this work has been primarily dependent on the collaborative efforts of the national and international m-COVID team and many researchers whose names are listed in the references.

Advertisement

Ethical statement

Most of the m-CoV work discussed in this chapter either adopted from our published work or from other studies adhered to the experimental procedures and animal care and use in accordance with good animal ethics approved by the Institutional Animal Care and use Committee.

References

  1. 1. Roos KL. Encephalitis. In: Handbook of Clinical Neurology. Vol. 121. Elsevier; 2014. pp. 1377-1381
  2. 2. Granerod J, Ambrose HE, Davies NW, Clewley JP, Walsh AL, Morgan D, et al. Causes of encephalitis and differences in their clinical presentations in England: A multicentre, population-based prospective study. The Lancet Infectious Diseases. 2010;10(12):835-844
  3. 3. Kennedy PGE. Viral encephalitis: Causes, differential diagnosis, and management. Journal of Neurology, Neurosurgery, and Psychiatry. 2004;75(Suppl. 1):i10-ii5
  4. 4. Mace SE. Central nervous system infections as a cause of an altered mental status? What is the pathogen growing in your central nervous system? Emergency Medicine Clinics of North America. 2010;28(3):535-570
  5. 5. Solomon T, Michael BD, Smith PE, Sanderson F, Davies NW, Hart IJ, et al. Management of suspected viral encephalitis in adults--Association of British Neurologists and British Infection Association National Guidelines. The Journal of Infection. 2012;64(4):347-373
  6. 6. Kneen R, Michael BD, Menson E, Mehta B, Easton A, Hemingway C, et al. Management of suspected viral encephalitis in children—Association of British Neurologists and British Paediatric Allergy, Immunology and Infection Group national guidelines. The Journal of Infection. 2012;64(5):449-477
  7. 7. Tunkel AR, Glaser CA, Bloch KC, Sejvar JJ, Marra CM, Roos KL, et al. The management of encephalitis: Clinical practice guidelines by the Infectious Diseases Society of America. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2008;47(3):303-327
  8. 8. Whitley RJ. Viral encephalitis. New England Journal of Medicine. 1990;323(4):242-250
  9. 9. Irani DN. Aseptic meningitis and viral myelitis. Neurologic Clinics. 2008;26(3):635-655
  10. 10. Rotbart HA. Viral meningitis. Seminars in Neurology. 2000;20(03):277-292
  11. 11. Kennedy PGE, Quan P-L, Lipkin WI. Viral encephalitis of unknown cause: Current perspective and recent advances. Viruses. 2017;9(6):138
  12. 12. Venkatesan A, Murphy OC. Viral encephalitis. Neurologic Clinics. 2018;36(4):705-724
  13. 13. Koyuncu OO, Hogue IB, Enquist LW. Virus infections in the nervous system. Cell Host & Microbe. 2013;13(4):379-393
  14. 14. Ayala-Nunez NV, Gaudin R. A viral journey to the brain: Current considerations and future developments. PLoS Pathogens. 2020;16(5):e1008434
  15. 15. Rozenberg F. Acute viral encephalitis. In: Handbook of Clinical Neurology. Vol. 112. Elsevier; 2013. pp. 1171-1181
  16. 16. Chen Z, Zhong D, Li G. The role of microglia in viral encephalitis: A review. Journal of Neuroinflammation. 2019;16(1):76
  17. 17. Tröscher AR, Wimmer I, Quemada-Garrido L, Köck U, Gessl D, Verberk SGS, et al. Microglial nodules provide the environment for pathogenic T cells in human encephalitis. Acta Neuropathologica. 2019;137(4):619-635
  18. 18. Chhatbar C, Prinz M. The roles of microglia in viral encephalitis: From sensome to therapeutic targeting. Cellular & Molecular Immunology. 2021;18(2):250-258
  19. 19. Griffin DE, Metcalf T. Clearance of virus infection from the CNS. Current Opinion in Virology. 2011;1(3):216-221
  20. 20. Das SJ. A mechanism of virus-induced demyelination. Interdisciplinary Perspectives on Infectious Diseases. 2010;2010:109239
  21. 21. Saadi F, Pal D, Sarma JD. Spike Glycoprotein Is Central to Coronavirus Pathogenesis-Parallel Between m-CoV and SARS-CoV-2. Annals of Neurosciences. 2021. doi: 10.1177/09727531211023755
  22. 22. Lane TE, Buchmeier MJ. Murine coronavirus infection: A paradigm for virus-induced demyelinating disease. Trends in Microbiology. 1997;5(1):9-14
  23. 23. Stohlman SA, Bergmann CC, Perlman S. In: Ahmed R, Chen I, editors. Persistent Viral Infections. New York: Wiley & Sons; 1998. pp. 537-557
  24. 24. Buchmeier MJ, Lewicki HA, Talbot PJ, Knobler RL. Murine hepatitis virus-4 (strain JHM)-induced neurologic disease is modulated in vivo by monoclonal antibody. Virology. 1984;132(2):261-270
  25. 25. Houtman JJ, Fleming JO. Dissociation of demyelination and viral clearance in congenitally immunodeficient mice infected with murine coronavirus JHM. Journal of Neurovirology. 1996;2(2):101-110
  26. 26. Lavi E, Gilden DH, Highkin MK, Weiss SR. Persistence of mouse hepatitis virus A59 RNA in a slow virus demyelinating infection in mice as detected by in situ hybridization. Journal of Virology. 1984;51(2):563-566
  27. 27. Lavi E, Gilden DH, Wroblewska Z, Rorke LB, Weiss SR. Experimental demyelination produced by the A59 strain of mouse hepatitis virus. Neurology. 1984;34(5):597-603
  28. 28. Das Sarma J, Fu L, Hingley ST, Lavi E. Mouse hepatitis virus type-2 infection in mice: An experimental model system of acute meningitis and hepatitis. Experimental and Molecular Pathology. 2001;71(1):1-12
  29. 29. Shindler KS, Kenyon LC, Dutt M, Hingley ST, Sarma JD. Experimental optic neuritis induced by a demyelinating strain of mouse hepatitis virus. Journal of Virology. 2008;82(17):8882-8886
  30. 30. Das Sarma J, Burrows A, Rayman P, Hwang MH, Kundu S, Sharma N, et al. Ifit2 deficiency restricts microglial activation and leukocyte migration following murine coronavirus (m-CoV) CNS infection. PLoS Pathogens. 2020;16(11):e1009034
  31. 31. Perlman S, Evans G, Afifi A. Effect of olfactory bulb ablation on spread of a neurotropic coronavirus into the mouse brain. The Journal of Experimental Medicine. 1990;172(4):1127-1132. DOI: 10.1084/jem.172.4.1127
  32. 32. Lavi E, Gilden DH, Highkin MK, Weiss SR. The organ tropism of mouse hepatitis virus A59 in mice is dependent on dose and route of inoculation. Laboratory Animal Science. 1986;36(2):130-135
  33. 33. Rout SS, Singh M, Shindler KS, Das SJ. One proline deletion in the fusion peptide of neurotropic mouse hepatitis virus (MHV) restricts retrograde axonal transport and neurodegeneration. The Journal of Biological Chemistry. 2020;295(20):6926-6935. DOI: 10.1074/jbc.RA119.011918
  34. 34. Das Sarma J, Scheen E, Seo SH, Koval M, Weiss SR. Enhanced green fluorescent protein expression may be used to monitor murine coronavirus spread in vitro and in the mouse central nervous system. Journal of Neurovirology. 2002;8(5):381-391
  35. 35. Das Sarma J, Iacono K, Gard L, Marek R, Kenyon LC, Koval M, et al. Demyelinating and nondemyelinating strains of mouse hepatitis virus differ in their neural cell tropism. Journal of Virology. 2008;82(11):5519
  36. 36. Kenyon LC, Biswas K, Shindler KS, et al. Gliopathy of Demyelinating and Non-Demyelinating Strains of Mouse Hepatitis Virus. Frontiers in Cellular Neuroscience. 2015;9:488. doi: 10.3389/fncel.2015.00488
  37. 37. Biswas K, Das SJ. Effect of microtubule disruption on neuronal spread and replication of demyelinating and nondemyelinating strains of mouse hepatitis virus in vitro. Journal of Virology. 2014;88(5):3043
  38. 38. Das Sarma J, Kenyon LC, Hingley ST, Shindler KS. Mechanisms of primary axonal damage in a viral model of multiple sclerosis. The Journal of Neuroscience. 2009;29(33):10272
  39. 39. Chatterjee D, Addya S, Khan RS, Kenyon LC, Choe A, Cohrs RJ, et al. Mouse hepatitis virus infection upregulates genes involved in innate immune responses. PLoS One. 2014;9(10):e111351
  40. 40. Bergmann CC, Lane TE, Stohlman SA. Coronavirus infection of the central nervous system: Host-virus stand-off. Nature Reviews Microbiology. 2006;4(2):121-132
  41. 41. Chakravarty D, Saadi F, Kundu S, Bose A, Khan R, Dine K, et al. CD4 deficiency causes poliomyelitis and axonal blebbing in murine coronavirus-induced neuroinflammation. Journal of virology. 2020;94(14):e00548-20. DOI: 10.1128/JVI.00548-20
  42. 42. Biswas K, Chatterjee D, Addya S, Khan RS, Kenyon LC, Choe A, et al. Demyelinating strain of mouse hepatitis virus infection bridging innate and adaptive immune response in the induction of demyelination. Clinical Immunology. 2016;170:9-19
  43. 43. Oleszak EL, Chang JR, Friedman H, Katsetos CD, Platsoucas CD. Theiler's virus infection: A model for multiple sclerosis. Clinical Microbiology Reviews. 2004;17(1):174-207
  44. 44. Theiler M. Spontaneous encephalomyelitis of mice, a new virus disease. The Journal of Experimental Medicine. 1937;65(5):705-719
  45. 45. Oleszak EL, Kuzmak J, Good RA, Platsoucas CD. Immunology of Theiler's murine encephalomyelitis virus infection. Immunologic Research. 1995;14(1):13-33
  46. 46. Murray PD, Pavelko KD, Leibowitz J, Lin X, Rodriguez M. CD4(+) and CD8(+) T cells make discrete contributions to demyelination and neurologic disease in a viral model of multiple sclerosis. Journal of Virology. 1998;72(9):7320-7329
  47. 47. Tsunoda I, Fujinami RS. TMEV and neuroantigens: Myelin genes and proteins, molecular mimicry, epitope spreading, and autoantibody-mediated remyelination. In: Lavi E, Constantinescu CS, editors. Experimental Models of Multiple Sclerosis. Boston, MA: Springer US; 2005. pp. 593-616
  48. 48. Drescher KM, Pease LR, Rodriguez M. Antiviral immune responses modulate the nature of central nervous system (CNS) disease in a murine model of multiple sclerosis. Immunological Reviews. 1997;159:177-193
  49. 49. Tsunoda I, Kuang LQ , Fujinami RS. Induction of autoreactive CD8+ cytotoxic T cells during Theiler's murine encephalomyelitis virus infection: Implications for autoimmunity. Journal of Virology. 2002;76(24):12834-12844
  50. 50. Oleszak EL, Hoffman BE, Chang JR, Zaczynska E, Gaughan J, Katsetos CD, et al. Apoptosis of infiltrating T cells in the central nervous system of mice infected with Theiler's murine encephalomyelitis virus. Virology. 2003;315(1):110-123
  51. 51. Chang JR, Zaczynska E, Katsetos CD, Platsoucas CD, Oleszak EL. Differential expression of TGF-beta, IL-2, and other cytokines in the CNS of Theiler's murine encephalomyelitis virus-infected susceptible and resistant strains of mice. Virology. 2000;278(2):346-360
  52. 52. Sato S, Reiner SL, Jensen MA, Roos RP. Central nervous system cytokine mRNA expression following Theiler's murine encephalomyelitis virus infection. Journal of Neuroimmunology. 1997;76(1-2):213-223
  53. 53. Fontana A, Frei K, Bodmer S, Hofer E, Schreier MH, Palladino MA Jr, et al. Transforming growth factor-beta inhibits the generation of cytotoxic T cells in virus-infected mice. Journal of Immunology (Baltimore, Md: 1950). 1989;143(10):3230-3234
  54. 54. Fekete R, Cserép C, Lénárt N, Tóth K, Orsolits B, Martinecz B, et al. Microglia control the spread of neurotropic virus infection via P2Y12 signalling and recruit monocytes through P2Y12-independent mechanisms. Acta Neuropathologica. 2018;136(3):461-482
  55. 55. Drokhlyansky E, Göz Aytürk D, Soh TK, Chrenek R, O’Loughlin E, Madore C, et al. The brain parenchyma has a type I interferon response that can limit virus spread. Proceedings of the National Academy of Sciences. 2017;114(1):E95
  56. 56. Liu Y, Gordesky-Gold B, Leney-Greene M, Weinbren NL, Tudor M, Cherry S. Inflammation-induced, STING-dependent autophagy restricts Zika virus infection in the Drosophila brain. Cell Host & Microbe. 2018;24(1):57-68.e3
  57. 57. Moy RH, Gold B, Molleston JM, Schad V, Yanger K, Salzano MV, et al. Antiviral autophagy restricts Rift Valley fever virus infection and is conserved from flies to mammals. Immunity. 2014;40(1):51-65
  58. 58. Moretti J, Roy S, Bozec D, Martinez J, Chapman JR, Ueberheide B, et al. STING senses microbial viability to orchestrate stress-mediated autophagy of the endoplasmic reticulum. Cell. 2017;171(4):809-23.e13
  59. 59. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469(7330):323-335
  60. 60. Alirezaei M, Kiosses WB, Fox HS. Decreased neuronal autophagy in HIV dementia: A mechanism of indirect neurotoxicity. Autophagy. 2008;4(7):963-966
  61. 61. Lum FM, Low DK, Fan Y, Tan JJ, Lee B, Chan JK, et al. Zika virus infects human fetal brain microglia and induces inflammation. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2017;64(7):914-920
  62. 62. Santello M, Bezzi P, Volterra A. TNFα controls glutamatergic gliotransmission in the hippocampal dentate gyrus. Neuron. 2011;69(5):988-1001
  63. 63. Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, et al. HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment. Nature Reviews Neurology. 2016;12(4):234-248
  64. 64. Persidsky Y, Gendelman HE. Mononuclear phagocyte immunity and the neuropathogenesis of HIV-1 infection. Journal of Leukocyte Biology. 2003;74(5):691-701
  65. 65. Gras G, Kaul M. Molecular mechanisms of neuroinvasion by monocytes-macrophages in HIV-1 infection. Retrovirology. 2010;7(1):30
  66. 66. Burdo TH, Lackner A, Williams KC. Monocyte/macrophages and their role in HIV neuropathogenesis. Immunological Reviews. 2013;254(1):102-113
  67. 67. Williams KC, Hickey WF. Central nervous system damage, monocytes and macrophages, and neurological disorders in AIDS. Annual Review of Neuroscience. 2002;25:537-562
  68. 68. Wallet C, De Rovere M, Van Assche J, Daouad F, De Wit S, Gautier V, et al. Microglial cells: The main HIV-1 reservoir in the brain. Frontiers in Cellular and Infection Microbiology. 2019;9(362). DOI: 10.3389/fcimb.2019.00362
  69. 69. Micallef S, Galea R. CMV encephalitis in an immune-competent patient. BMJ Case Reports. 2018:bcr2018224740. DOI: 10.1136/bcr-2018-224740
  70. 70. Arribas JR, Storch GA, Clifford DB, Tselis AC. Cytomegalovirus encephalitis. Annals of Internal Medicine. 1996;125(7):577-587
  71. 71. Grassi MP, Clerici F, Perin C, D'Arminio Monforte A, Vago L, Borella M, et al. Microglial nodular encephalitis and ventriculoencephalitis due to cytomegalovirus infection in patients with AIDS: Two distinct clinical patterns. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 1998;27(3):504-508
  72. 72. Mlakar J, Korva M, Tul N, Popović M, Poljšak-Prijatelj M, Mraz J, et al. Zika virus associated with microcephaly. The New England Journal of Medicine. 2016;374(10):951-958
  73. 73. Luo H, Winkelmann ER, Zhu S, Ru W, Mays E, Silvas JA, et al. Peli1 facilitates virus replication and promotes neuroinflammation during West Nile virus infection. The Journal of Clinical Investigation. 2018;128(11):4980-4991
  74. 74. Town T, Jeng D, Alexopoulou L, Tan J, Flavell RA. Microglia recognize double-stranded RNA via TLR3. Journal of immunology (Baltimore, Md: 1950). 2006;176(6):3804-3812
  75. 75. Seitz S, Clarke P, Tyler KL. Pharmacologic depletion of microglia increases viral load in the brain and enhances mortality in murine models of flavivirus-induced encephalitis. Journal of Virology. 2018;92(16):e00525-18. DOI: 10.1128/JVI.00525-18
  76. 76. Chhatbar C, Detje CN, Grabski E, Borst K, Spanier J, Ghita L, et al. Type I interferon receptor signaling of neurons and astrocytes regulates microglia activation during viral encephalitis. Cell Reports. 2018;25(1):118-29.e4
  77. 77. Wheeler DL, Sariol A, Meyerholz DK, Perlman S. Microglia are required for protection against lethal coronavirus encephalitis in mice. The Journal of Clinical Investigation. 2018;128(3):931-943
  78. 78. Mendez-Fernandez YV, Johnson AJ, Rodriguez M, Pease LR. Clearance of Theiler's virus infection depends on the ability to generate a CD8+ T cell response against a single immunodominant viral peptide. European Journal of Immunology. 2003;33(9):2501-2510
  79. 79. Sitati EM, Diamond MS. CD4+ T-cell responses are required for clearance of West Nile virus from the central nervous system. Journal of Virology. 2006;80(24):12060-12069
  80. 80. Chakravarty D, Saadi F, Kundu S, Bose A, Khan R, Dine K, et al. CD4 deficiency causes poliomyelitis and axonal blebbing in murine coronavirus-induced neuroinflammation. Journal of Virology. 2020;94(14):e00548-20. DOI: 10.1128/JVI.00548-20
  81. 81. Stohlman Stephen A, Hinton David R, Parra B, Atkinson R, Bergmann CC. CD4 T cells contribute to virus control and pathology following central nervous system infection with neurotropic mouse hepatitis virus. Journal of Virology. 2008;82(5):2130-2139
  82. 82. Ciurkiewicz M, Herder V, Beineke A. Beneficial and detrimental effects of regulatory T cells in neurotropic virus infections. International Journal of Molecular Sciences. 2020;21(5):1705. doi: 10.3390/ijms21051705
  83. 83. Omura S, Kawai E, Sato F, Martinez NE, Minagar A, Al-Kofahi M, et al. Theiler's virus-mediated immunopathology in the CNS and heart: Roles of organ-specific cytokine and lymphatic responses. Frontiers in Immunology. 2018;9:2870. DOI: 10.3389/fimmu.2018.02870
  84. 84. Ciurkiewicz M, Herder V, Khan MA, Uhde AK, Teich R, Floess S, et al. Cytotoxic CD8(+) T cell ablation enhances the capacity of regulatory T cells to delay viral elimination in Theiler's murine encephalomyelitis. Brain Pathology (Zurich, Switzerland). 2018;28(3):349-368
  85. 85. Wu GF, Dandekar AA, Pewe L, Perlman S. CD4 and CD8 T cells have redundant but not identical roles in virus-induced demyelination. Journal of Immunology (Baltimore, Md: 1950). 2000;165(4):2278-2286
  86. 86. Chua MM, MacNamara KC, San Mateo L, Shen H, Weiss SR. Effects of an epitope-specific CD8+ T-cell response on murine coronavirus central nervous system disease: Protection from virus replication and antigen spread and selection of epitope escape mutants. Journal of Virology. 2004;78(3):1150-1159
  87. 87. Ponomarev ED, Shriver LP, Dittel BN. CD40 expression by microglial cells is required for their completion of a two-step activation process during central nervous system autoimmune inflammation. Journal of Immunology (Baltimore, Md: 1950). 2006;176(3):1402-1410
  88. 88. Chen K, Huang J, Gong W, Zhang L, Yu P, Wang JM. CD40/CD40L dyad in the inflammatory and immune responses in the central nervous system. Cellular & Molecular Immunology. 2006;3(3):163-169
  89. 89. Saadi F, Chakravarty D, Kumar S, Kamble M, Saha B, Shindler KS, et al. CD40L protects against mouse hepatitis virus-induced neuroinflammatory demyelination. PLoS Pathogens. 2021;17(12):e1010059
  90. 90. Perry AK, Chen G, Zheng D, Tang H, Cheng G. The host type I interferon response to viral and bacterial infections. Cell Research. 2005;15(6):407-422
  91. 91. Reinert LS, Lopušná K, Winther H, Sun C, Thomsen MK, Nandakumar R, et al. Sensing of HSV-1 by the cGAS-STING pathway in microglia orchestrates antiviral defence in the CNS. Nature Communications. 2016;7:13348
  92. 92. Daniels BP, Holman DW, Cruz-Orengo L, Jujjavarapu H, Durrant DM, Klein RS. Viral pathogen-associated molecular patterns regulate blood-brain barrier integrity via competing innate cytokine signals. MBio. 2014;5(5):e01476-e01414
  93. 93. Morris O, Liu X, Domingues C, Runchel C, Chai A, Basith S, et al. Signal integration by the iκb protein pickle shapes Drosophila innate host defense. Cell Host & Microbe. 2016;20(3):283-295
  94. 94. Joubert PE, Werneke SW, de la Calle C, Guivel-Benhassine F, Giodini A, Peduto L, et al. Chikungunya virus-induced autophagy delays caspase-dependent cell death. The Journal of Experimental Medicine. 2012;209(5):1029-1047
  95. 95. Vasek MJ, Garber C, Dorsey D, Durrant DM, Bollman B, Soung A, et al. A complement-microglial axis drives synapse loss during virus-induced memory impairment. Nature. 2016;534(7608):538-543
  96. 96. Lokensgard JR, Hu S, Sheng W, van Oijen M, Cox D, Cheeran MC, et al. Robust expression of TNF-alpha, IL-1beta, RANTES, and IP-10 by human microglial cells during nonproductive infection with herpes simplex virus. Journal of Neurovirology. 2001;7(3):208-219
  97. 97. Hanisch UK. Microglia as a source and target of cytokines. Glia. 2002;40(2):140-155
  98. 98. Habbas S, Santello M, Becker D, Stubbe H, Zappia G, Liaudet N, et al. Neuroinflammatory TNFα impairs memory via astrocyte signaling. Cell. 2015;163(7):1730-1741
  99. 99. Ginsberg SD, Alldred MJ, Gunnam SM, Schiroli C, Lee SH, Morgello S, et al. Expression profiling suggests microglial impairment in human immunodeficiency virus neuropathogenesis. Annals of Neurology. 2018;83(2):406-417
  100. 100. Ludlow M, Kortekaas J, Herden C, Hoffmann B, Tappe D, Trebst C, et al. Neurotropic virus infections as the cause of immediate and delayed neuropathology. Acta Neuropathologica. 2016;131(2):159-184
  101. 101. Hatanpaa KJ, Kim JH. Neuropathology of viral infections. In: Handbook of Clinical Neurology. Vol. 123. Elsevier; 2014. pp. 193-214
  102. 102. Mao L, Wang M, Chen S, He Q , Chang J, Hong C, et al. Neurological manifestations of hospitalized patients with COVID-19 in Wuhan, China: A retrospective case series study. medRxiv. 2020;77(6):683-690. DOI: 10.1001/jamaneurol.2020.1127
  103. 103. Koralnik IJ, Tyler KL. COVID-19: A global threat to the nervous system. Annals of Neurology. 2020;88(1):1-11
  104. 104. Chakravarty D, Das SJ. Murine-beta-coronavirus-induced neuropathogenesis sheds light on CNS pathobiology of SARS-CoV2. Journal of Neurovirology. 2021;27(2):197-216
  105. 105. Abdullahi AM, Sarmast ST, Singh R. Molecular biology and epidemiology of neurotropic viruses. Cureus. 2020;12(8):e9674
  106. 106. Salimi H, Cain MD, Klein RS. Encephalitic arboviruses: Emergence, clinical presentation, and neuropathogenesis. Neurotherapeutics. 2016;13(3):514-534
  107. 107. Solomon T. Flavivirus encephalitis. The New England Journal of Medicine. 2004;351(4):370-378. DOI: 10.1056/NEJMra030476
  108. 108. Zhang Y, Wang Z, Chen H, Chen Z, Tian Y. Antioxidants: Potential antiviral agents for Japanese encephalitis virus infection. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases. 2014;24:30-36
  109. 109. Upadhyay RK. Epidemiology, disease transmission and pathogenesis caused by JE virus: Its prevention and control. American Journal of Infectious Diseases and Microbiology. 2015;3(1):38-64
  110. 110. Bogovic P, Strle F. Tick-borne encephalitis: A review of epidemiology, clinical characteristics, and management. World Journal of Clinical Cases. 2015;3(5):430-441
  111. 111. Hernández-Triana LM, Jeffries CL, Mansfield KL, Carnell G, Fooks AR, Johnson N. Emergence of West Nile virus lineage 2 in Europe: A review on the introduction and spread of a mosquito-borne disease. Frontiers in Public Health. 2014;2:271. DOI: 10.3389/fpubh.2014.00271
  112. 112. Higuera A, Ramírez JD. Molecular epidemiology of dengue, yellow fever, Zika and Chikungunya arboviruses: An update. Acta Tropica. 2019;190:99-111
  113. 113. Go YY, Balasuriya UBR, Lee C-k. Zoonotic encephalitides caused by arboviruses: Transmission and epidemiology of alphaviruses and flaviviruses. Clinical and Experimental Vaccine Research. 2014;3(1):58-77
  114. 114. Beckham JD, Pastula DM, Massey A, Tyler KL. Zika virus as an emerging global pathogen: Neurological complications of Zika virus. JAMA Neurology. 2016;73(7):875-879
  115. 115. Thomas SJ, Endy TP, Rothman AL, Barrett AD. 155—Flaviviruses (dengue, yellow fever, Japanese encephalitis, West Nile encephalitis, St. Louis encephalitis, tick-borne encephalitis, Kyasanur Forest disease, Alkhurma hemorrhagic fever, Zika). In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: W.B. Saunders; 2015. pp. 1881-903.e6
  116. 116. Guzmán-Terán C, Calderón-Rangel A, Rodriguez-Morales A, Mattar S. Venezuelan equine encephalitis virus: The problem is not over for tropical America. Annals of Clinical Microbiology and Antimicrobials. 2020;19(1):19
  117. 117. Mehta R, Gerardin P, de Brito CAA, Soares CN, Ferreira MLB, Solomon T. The neurological complications of chikungunya virus: A systematic review. Reviews in Medical Virology. 2018;28(3):e1978
  118. 118. Gérardin P, Couderc T, Bintner M, Tournebize P, Renouil M, Lémant J, et al. Chikungunya virus–associated encephalitis. A cohort study on La Réunion Island, 2005-2009. 2016;86(1):94-102
  119. 119. Carrera JP, Cucunubá ZM, Neira K, Lambert B, Pittí Y, Liscano J, et al. Endemic and epidemic human alphavirus infections in Eastern Panama: An analysis of population-based cross-sectional surveys. The American Journal of Tropical Medicine and Hygiene. 2020;103(6):2429-2437
  120. 120. Meyding-Lamadé U, Strank C. Herpesvirus infections of the central nervous system in immunocompromised patients. Therapeutic Advances in Neurological Disorders. 2012;5(5):279-296
  121. 121. Gilden DH, Mahalingam R, Cohrs RJ, Tyler KL. Herpesvirus infections of the nervous system. Nature Clinical Practice Neurology. 2007;3(2):82-94
  122. 122. Duarte LF, Farías MA, Álvarez DM, Bueno SM, Riedel CA, González PA. Herpes simplex virus type 1 infection of the central nervous system: Insights into proposed interrelationships with neurodegenerative disorders. Frontiers in Cellular Neuroscience. 2019;13:46. DOI: 10.3389/fncel.2019.00046
  123. 123. Gershon AA, Breuer J, Cohen JI, Cohrs RJ, Gershon MD, Gilden D, et al. Varicella zoster virus infection. Nature Reviews. Disease Primers. 2015;1:15016
  124. 124. Kennedy PGE, Gershon AA. Clinical features of varicella-zoster virus infection. Viruses. 2018;10(11):609
  125. 125. Rubin S, Eckhaus M, Rennick LJ, Bamford CG, Duprex WP. Molecular biology, pathogenesis and pathology of mumps virus. The Journal of Pathology. 2015;235(2):242-252. DOI: 10.1002/path.4445
  126. 126. Fisher DL, Defres S, Solomon T. Measles-induced encephalitis. QJM: An International Journal of Medicine. 2015;108(3):177-182
  127. 127. Hossain MJ, Gurley ES, Montgomery JM, Bell M, Carroll DS, Hsu VP, et al. Clinical presentation of nipah virus infection in Bangladesh. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2008;46(7):977-984
  128. 128. Vries RD, Jong A, Verburgh RJ, Sauerhering L, Nierop GP, Binnendijk RS, et al. Human paramyxovirus infections induce T cells that cross-react with zoonotic henipaviruses. mBio. 2020;11(4):e00972-20. DOI: 10.1128/mBio.00972-20
  129. 129. Ferren M, Horvat B, Mathieu C. Measles encephalitis: Towards new therapeutics. Viruses. 2019;11(11):1017
  130. 130. Singh RK, Dhama K, Chakraborty S, Tiwari R, Natesan S, Khandia R, et al. Nipah virus: Epidemiology, pathology, immunobiology and advances in diagnosis, vaccine designing and control strategies—a comprehensive review. The Veterinary Quarterly. 2019;39(1):26-55
  131. 131. Wilson MR, Peters CJ. Diseases of the central nervous system caused by lymphocytic choriomeningitis virus and other arenaviruses. In: Handbook of Clinical Neurology. Vol. 123. Elsevier; 2014. pp. 671-681
  132. 132. Bonthius DJ. Lymphocytic choriomeningitis virus: An underrecognized cause of neurologic disease in the fetus, child, and adult. Seminars in Pediatric Neurology. 2012;19(3):89-95
  133. 133. Dräger S, Marx A-F, Pigny F, Cherpillod P, Eisermann P, Sendi P, et al. Lymphocytic choriomeningitis virus meningitis after needlestick injury: A case report. Antimicrobial Resistance & Infection Control. 2019;8(1):77
  134. 134. de Crom SCM, Rossen JWA, van Furth AM, Obihara CC. Enterovirus and parechovirus infection in children: A brief overview. European Journal of Pediatrics. 2016;175(8):1023-1029
  135. 135. Baggen J, Thibaut HJ, Strating JRPM, van Kuppeveld FJM. The life cycle of non-polio enteroviruses and how to target it. Nature Reviews Microbiology. 2018;16(6):368-381
  136. 136. Kadambari S, Braccio S, Ribeiro S, Allen DJ, Pebody R, Brown D, et al. Enterovirus and parechovirus meningitis in infants younger than 90 days old in the UK and Republic of Ireland: A British paediatric surveillance unit study. Archives of Disease in Childhood 2019;104(6):552-557. DOI: 10.1136/archdischild-2018-315643
  137. 137. Dumaidi K, Al-Jawabreh A, Samarah F, Zraiqi A, Yaseen D. Genetic diversity of the enteroviruses detected from cerebrospinal fluid (CSF) samples of patients with suspected aseptic meningitis in northern West Bank, Palestine in 2017. PLoS One. 2018;13(12):e0202243
  138. 138. Deleidi M, Isacson O. Viral and inflammatory triggers of neurodegenerative diseases. Science Translational Medicine. 2012;4(121):121ps3
  139. 139. van den Pol AN. Viral infections in the developing and mature brain. Trends in Neurosciences. 2006;29(7):398-406
  140. 140. Zhou L, Miranda-Saksena M, Saksena NK. Viruses and neurodegeneration. Virology Journal. 2013;10(1):172
  141. 141. Berth SH, Leopold PL, Morfini GN. Virus-induced neuronal dysfunction and degeneration. Frontiers in Bioscience (Landmark Edition). 2009;14:5239-5259
  142. 142. Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nature Medicine. 2004;10(12):1366-1373
  143. 143. Boulanger LM. Immune proteins in brain development and synaptic plasticity. Neuron. 2009;64(1):93-109
  144. 144. Préhaud C, Mégret F, Lafage M, Lafon M. Virus infection switches TLR-3-positive human neurons to become strong producers of beta interferon. Journal of Virology. 2005;79(20):12893-12904
  145. 145. Fux CA, Pfister S, Nohl F, Zimmerli S. Cytomegalovirus-associated acute transverse myelitis in immunocompetent adults. Clinical Microbiology and Infection. 2003;9(12):1187-1190
  146. 146. Brew BJ, González-Scarano F. HIV-associated dementia: An inconvenient truth. Neurology. 2007;68(5):324-325
  147. 147. Horta-Barbosa L, Fuccillo DA, Sever JL. Chronic viral infections of the central nervous system. Journal of the American Medical Association. 1971;218(8):1185-1188
  148. 148. Shoji H, Watanabe M, Itoh S, Kuwahara H, Hattori F. Japanese encephalitis and parkinsonism. Journal of Neurology. 1993;240(1):59-60
  149. 149. Jang H, Boltz D, Sturm-Ramirez K, Shepherd KR, Jiao Y, Webster R, et al. Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(33):14063-14068
  150. 150. Jang H, Boltz D, McClaren J, Pani AK, Smeyne M, Korff A, et al. Inflammatory effects of highly pathogenic H5N1 influenza virus infection in the CNS of mice. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience. 2012;32(5):1545-1559
  151. 151. Rohn TT, Catlin LW. Immunolocalization of influenza A virus and markers of inflammation in the human Parkinson's disease brain. PLoS One. 2011;6(5):e20495
  152. 152. Sulkava R, Rissanen A, Pyhälä R. Post-influenzal encephalitis during the influenza A outbreak in 1979/1980. Journal of Neurology, Neurosurgery, and Psychiatry. 1981;44(2):161-163
  153. 153. Neurological complications of influenza. British Medical Journal. 1970;1(5691):248-249
  154. 154. Nath A, Johnson TP. Mechanisms of viral persistence in the brain and therapeutic approaches. The FEBS Journal. 2021. Online ahead of Print. DOI: 10.1111/febs.15871
  155. 155. Lafon M. Immune evasion, a critical strategy for rabies virus. Developments in Biologicals. 2008;131:413-419
  156. 156. Xie X, Zeng J. Neuroimmune evasion of Zika virus to facilitate viral pathogenesis. Frontiers in Cellular and Infection Microbiology. 2021;11:662447. DOI: 10.3389/fcimb.2021.662447
  157. 157. Whitley RJ. Herpesviruses. 4th ed. Galveston (TX): University of Texas Medical Branch at Galveston; 1996
  158. 158. Letenneur L, Pérès K, Fleury H, Garrigue I, Barberger-Gateau P, Helmer C, et al. Seropositivity to herpes simplex virus antibodies and risk of Alzheimer's disease: A population-based cohort study. PLoS One. 2008;3(11):e3637
  159. 159. Itzhaki RF, Wozniak MA. Herpes simplex virus type 1 in Alzheimer's disease: The enemy within. Journal of Alzheimer's Disease: JAD. 2008;13(4):393-405
  160. 160. Denaro FJ, Staub P, Colmer J, Freed DM. Coexistence of Alzheimer disease neuropathology with herpes simplex encephalitis. Cellular and Molecular Biology (Noisy-le-Grand, France). 2003;49(8):1233-1240
  161. 161. Wozniak MA, Mee AP, Itzhaki RF. Herpes simplex virus type 1 DNA is located within Alzheimer's disease amyloid plaques. The Journal of Pathology. 2009;217(1):131-138
  162. 162. Santana S, Recuero M, Bullido MJ, Valdivieso F, Aldudo J. Herpes simplex virus type I induces the accumulation of intracellular β-amyloid in autophagic compartments and the inhibition of the non-amyloidogenic pathway in human neuroblastoma cells. Neurobiology of Aging. 2012;33(2):430.e19-430.e33
  163. 163. Piacentini R, Civitelli L, Ripoli C, Marcocci ME, De Chiara G, Garaci E, et al. HSV-1 promotes Ca2+−mediated APP phosphorylation and Aβ accumulation in rat cortical neurons. Neurobiology of Aging. 2011;32(12):2323.e13-2323.e26
  164. 164. Hill JM, Zhao Y, Clement C, Neumann DM, Lukiw WJ. HSV-1 infection of human brain cells induces miRNA-146a and Alzheimer-type inflammatory signaling. Neuroreport. 2009;20(16):1500-1505
  165. 165. Obeso JA, Rodríguez-Oroz MC, Benitez-Temino B, Blesa FJ, Guridi J, Marin C, et al. Functional organization of the basal ganglia: Therapeutic implications for Parkinson's disease. Movement Disorders: Official Journal of the Movement Disorder Society. 2008;23(Suppl. 3):S548-S559
  166. 166. Hoffman LA, Vilensky JA. Encephalitis lethargica: 100 years after the epidemic. Brain. 2017;140(8):2246-2251
  167. 167. Takahashi M, Yamada T, Nakajima S, Nakajima K, Yamamoto T, Okada H. The substantia nigra is a major target for neurovirulent influenza A virus. The Journal of Experimental Medicine. 1995;181(6):2161-2169
  168. 168. Gamboa ET, Wolf A, Yahr MD, Harter DH, Duffy PE, Barden H, et al. Influenza virus antigen in postencephalitic parkinsonism brain. Detection by immunofluorescence. Archives of Neurology. 1974;31(4):228-232
  169. 169. Verma A, Berger JR. ALS syndrome in patients with HIV-1 infection. Journal of the Neurological Sciences. 2006;240(1-2):59-64
  170. 170. MacGowan DJ, Scelsa SN, Waldron M. An ALS-like syndrome with new HIV infection and complete response to antiretroviral therapy. Neurology. 2001;57(6):1094-1097
  171. 171. Zhou L, Diefenbach E, Crossett B, Tran SL, Ng T, Rizos H, et al. First evidence of overlaps between HIV-associated dementia (HAD) and non-viral neurodegenerative diseases: Proteomic analysis of the frontal cortex from HIV+ patients with and without dementia. Molecular Neurodegeneration. 2010;5:27
  172. 172. Maruszak H, Brew BJ, Giovannoni G, Gold J. Could antiretroviral drugs be effective in multiple sclerosis? A case report. European Journal of Neurology. 2011;18(9):e110-e111
  173. 173. Brew BJ, Crowe SM, Landay A, Cysique LA, Guillemin G. Neurodegeneration and ageing in the HAART era. Journal of Neuroimmune Pharmacology : The Official Journal of the Society on NeuroImmune Pharmacology. 2009;4(2):163-174
  174. 174. Zambrano Á, Solis L, Salvadores N, Cortés M, Lerchundi R, Otth C. Neuronal cytoskeletal dynamic modification and neurodegeneration induced by infection with herpes simplex virus type 1. Journal of Alzheimer's Disease. 2008;14:259-269

Written By

Fareeha Saadi, Debanjana Chakravarty, Grishma Kasle and Jayasri Das Sarma

Submitted: 06 January 2022 Reviewed: 14 January 2022 Published: 25 February 2022