A role for viral infections in Parkinson's etiology - Neuronal Signaling

2 downloads 0 Views 675KB Size Report
Mar 19, 2018 - -Causes rash, white spots, cough, red eyes .... Anderson L, Vilensky J, Duvoisin R. Neuropathology of acute phase encephalitis lethargica: a review of cases from ..... Zhang J, Perry G, Smith MA, Robertson D, Olson SJ, Graham DG, et al. .... Prüss H, Finke C, Höltje M, Hofmann J, Klingbeil C, Probst C, et al.
Neuronal Signaling: this is an Accepted Manuscript, not the final Version of Record. You are encouraged to use the Version of Record that, when published, will replace this version. The most up-to-date version is available at http://dx.doi.org/10.1042/NS20170166. Please cite using the DOI 10.1042/NS20170166

A role for viral infections in Parkinson’s etiology – more than just “shaky” evidence?

Laura K Olsen, Eilís Dowd & Declan P McKernan*.

* Corresponding author

Address: Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland H91 TK33. Phone: +353 91 493826. ACCEPTED MANUSCRIPT

Fax: +353 91 495586. Email: [email protected].

1 Use of open access articles is permitted based on the terms of the specific Creative Commons Licence under which the article is published. Archiving of non-open access articles is permitted in accordance with the Archiving Policy of Portland Press ( http://www.portlandpresspublishing.com/content/open-access-policy#Archiving).

Abstract Despite over 200 years since its first description by James Parkinson, the cause(s) of most cases of Parkinson’s disease (PD) have yet to be elucidated. The disparity between the current understanding of PD symptomology and pathology has led to numerous symptomatic therapies, but no strategy for prevention or disease cure. An association between certain viral infections and neurodegenerative diseases has been recognized, but largely ignored or dismissed as controversial, for decades. Recent epidemiological studies have renewed scientific interest in investigating microbial interactions with the CNS. This review examines past and current clinical findings and overviews the potential molecular implications of viruses in PD pathology. Keywords: Parkinson’s Disease, virus, neurodegeneration, pathogen, neuroinflammation, synapse, autophagy

2

Abbreviation List AD = Alzheimer's Disease $03$ Į-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid BBB = Blood-brain Barrier cAMP = Cyclic Adenosine Monophosphate CD4+ = Cluster of Differentiation 4 CD8+ = Cluster of Differentiation 8 CNS = Central Nervous System CSF = Cerebrospinal Fluid DA = Dopamine dsDNA = Double-stranded Deoxyribonucleic Acid dsRNA = Double-stranded Ribonucleic Acid EL = Encephalitis Lethargica HCV = Hepatitis C Virus HIV = Human Immunodeficiency Virus HSE = Herpes Simplex Encephalitis HSV = Herpes Simplex Virus

3

IFN = Interferon IFNAR1 = Interferon-Įȕ5HFHSWRU IL-6 = Interleukin-6 iNOS = Inducible Nitric Oxide Synthase iPD = Idiopathic Parkinson's Disease IRF = Interferon Regulatory Factor ISG = Interferon-stimulated Genes LAT = Latency Associated Transcript LC3-II = Microtubule-associated Protein 1A/1B-light Chain 3 NF-ț% 1XFOHDU)DFWRU.DSSD-light-chain-enhancer of Activated B Cells NMDAR = N-methyl-D-aspartate Receptor NP = Nucleoprotein NS1 = Non-structural Protein 1 OS = Oxidative Stress PAMP = Pathogen-associated Molecular Patterns PD = Parkinson's Disease PDZ = PSD-95/Discs-large/ZO-1

4

PEP = Post-encephalitic Parkinsonism PKR = Protein Kinase R PRR = Pathogen Recognition Receptor PSD-95 = Post-synaptic Density Protein-95 ROS = Reactive Oxygen Species SN = Substantia Nigra SNAP25 = Synaptosomal-associated Protein 25 ssRNA = Single-stranded Ribonucleic Acid TG = Trigeminal Ganglia TLR = Toll-like Receptor TNF-Į 7XPRU1HFURVLV)DFWRU-alpha VTA = Ventral Tegmental Area

5

Introduction Parkinson’s disease (PD), the most common neurodegenerative motor disorder, is generally characterized by the selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SN) of the midbrain, resulting in decreased dopamine (DA) transmission throughout the nigrostriatal pathway (1). PD symptoms include resting tremors, unstable posture, bradykinesia, rigidity, and non-motor symptoms (such as dysphagia, olfactory impairment, sleep disturbances, dementia, and constipation) (2-4). This progressive neurodegenerative disease can be familial (associated with early onset) or sporadic (5-8). Hallmark pathological features of both familial and sporadic 3'LQFOXGHXQFRQWUROOHGSURWHLQDJJUHJDWLRQ SULPDULO\Į-synuclein fibrils forming

Lewy

bodies),

oxidative

stress

(OS),

mitochondrial

dysfunction,

chronic

neuroinflammation (including microglia activation and astrogliosis), and autophagy disruption (5, 9-11). Despite over two centuries of investigation, the cause(s) of most cases of PD are still unknown. Epidemiological studies suggest there is a gene-environment interaction involved in the development of sporadic/idiopathic PD (iPD). Previous studies suggest a correlation between iPD development and exposure to pesticides or heavy metals, traumatic brain injury, and viral/bacterial infection (12-14). Although multiple reviews describe the association between pesticides and PD (most notably rotenone and paraquat) (15, 16), there are few reviews detailing all the epidemiological, post-mortem, and pre-clinical evidence surrounding the association between viral infections and PD. As urgently expressed by both scientists and clinicians in a recent article by Itzhaki et al. (17), previous studies identifying microbial associations with Alzheimer’s disease (AD) (the most common neurodegenerative disorder) have largely been ignored or dismissed as controversial despite the current lack of progress for understanding or

6

curing this disease. Similarly, the same lack of progress and dismissal of previous epidemiological findings regarding viral infections can be said of PD. The purpose of this review is to revisit historical conclusions, provide a comprehensive update on recent clinical findings, and overview the potential molecular and cellular implications of neurotropic viruses in PD pathology.

Viruses as a Risk Factor for PD: Sifting Through Historical and Clinical Evidence The first suggestion of a relationship between viral infections and PD was the 1920s-1930s influenza epidemic, which was associated with Encephalitis Lethargica (EL) (18). Although EL patients exhibited drastic irregularities in disease progression and displayed ‘symptomatological polymorphisms,’ EL has been described as a type of ‘sleeping sickness’ which can include headache, nausea, fever, uncontrollable sleepiness, catatonia and sometimes coma (19). The EL epidemic coincided with an equally significant influenza pandemic (the Spanish Influenza), leading many clinicians and other prominent scientists from that time to believe there was a causal relationship (or at least an epidemiological association) between these conditions (20). Multiple studies investigating the preserved brain samples of EL patients from the epidemic years (1918-1930) found no evidence of the 1918 influenza virus in these tissues (21-23). Also, 1918 influenza derived sequences revealed mutations in two surface protein-encoding genes that suggest this viral strain was incapable of replicating outside of the respiratory system (24, 25). Reports from more current cases of EL suggest that EL may be an auto-antibody disorder (18, 26-32).

7

Since the EL epidemic, numerous cases of Post-encephalitic Parkinsonism (PEP) after certain viral infections (H5N1, coxsackie virus, Japanese encephalitis B., St. Louis viral encephalopathy, and HIV) have been reported, but these cases of parkinsonism often do not exhibit the same cellular or molecular pathologies as seen in PD and are suggested to be ‘phenocopies’ of PD (33-39). Although these acute cases of viral infection did not present with classical PD, these findings (along with the believed EL association with PD) led to multiple clinical studies in the late 1970s and early 1980s investigating the relationship between viral infections and PD. Studies examining the relationship between viruses and PD are detailed in Table 1. A study by Elizan et al. (40) found a significant relationship between viruses (HSV, measles, and influenza A) and iPD, but these findings may be confounded since their control group included Amyotrophic Lateral Sclerosis and AD patients (conditions which have since been suggested to be associated with certain viral infections themselves). Another set of studies found an increased incidence of PD among those chronically infected with hepatitis C virus (HCV), but these studies are confounded by the fact that some HCV patients received interferon (IFN) treatments; with a follow-up investigation finding a much stronger relationship between IFN treated groups and PD (249 PD incidents/100,000 person-years) than non-IFN treated groups (30 PD incidents/ 100,000 person-years) (41-43). Multiple studies by Marttila et al., using a variety of antibody detection techniques (complement fixation, radioimmunoassay, indirect immunofluorescent assay, microindirect hemagglutination), found a significant increase in HSV antibody titres and mean HSV titre in iPD patient serum (44-46). A study using the microindirect hemagglutination test was able to differentiate between HSV-I and HSV-II; finding that increases in antibody titres and mean titre among iPD patients was specific to HSV-I only (not HSV-II) (46). Other studies have

8

questioned iPD patients for their history of HSV, measles, and influenza A infection with conflicting results (see Table 1). Although a significant association was found between severity and frequency of influenza A infection and PD incidence (with no association for HSV), the conclusions based on these studies are limited in that they rely on accurate patient memory and interpretation of their condition (patients cannot be expected to correctly diagnose their previous exposure or infection with viruses) (47, 48). A more recent study that examined PD patient serum found a more frequent incidence of HSV-I infections among iPD patients, further supporting the findings of the Marttila studies (49). Based on the aforementioned clinical evidence, HSV-I and strains of influenza A will be reviewed for their neurovirulence and association with PD-like pathology in the CNS. Molecular and cellular events associated with HSV-I/influenza A infection will be discussed for their potential implications in PD.

CNS Viral Entry: HSV-I and Influenza A Since the primary disease pathology characteristics of PD exist in the CNS, it may be relevant (but possibly not crucial) to study the neurovirulence of viruses associated with PD. HSV-I and influenza A have very different life cycles, resulting in different strategies for survival/ replication within the host. Influenza A is generally a transient infection, only lasting a few weeks inside the host (50). On the other hand, an acute infection of HSV-I (presented as epithelial blistering of the mouth or genitalia) is followed by viral latency, which is generally established in the trigeminal ganglia (TG) (51). Although dormant, HSV-I is a chronic infection that maintains latency in sensory ganglia that innervate the brainstem and cerebellum of the CNS (52, 53). Primarily residing in the respiratory system during acute infection, influenza A can

9

enter the CNS through the olfactory nerve (54). Found to be axonally transported via cytoskeleton intermediate filaments, influenza A can follow olfactory neuron projections through the cribiform plate in the nasal cavity into the olfactory bulbs and olfactory tracts of the CNS (55, 56). Previously, viral entry of either HSV-I or influenza A into the CNS was considered fatal (or nearly fatal) via the rare condition of herpes simplex encephalitis (HSE) or acute encephalitis, respectively (33, 57, 58). More recent findings now suggest that viral entry into the CNS does not necessarily result in a drastic, usually fatal, immune response. Although many studies have not found the existence of HSV-I DNA or antigens in post-mortem PD patient brains (59, 60), multiple studies have found HSV-I DNA in the brains of normal aged humans and AD patients (59, 61-64). The presence of HSV-I DNA was associated with increased age and the characteristic amyloid-beta plaques found in AD (63, 64). Determining the neurovirulence and brain cell localization of influenza A in the CNS of PD patients is far more difficult since it is a transient infection. Although partially determined by the route of infection, pre-clinical mouse models of neurovirulent strains of influenza A have found this infection to successfully enter the CNS and localize in the SN, thalamus, hippocampus, locus coeruleus, ganglia (trigeminal, vagal, spinal and sympathetic trunk ganglia), olfactory bulbs, and thoracic spinal cord around day 10 post-infection (34, 65-67). Influenza A antigens were also found to preferentially exist in catecholaminergic neurons, meninges, and ependymal areas (68). Despite influenza A entry into the CNS in these mouse models, viral replication and maintenance in the CNS did not extend past two weeks, and was generally non-existent in the CNS by day 21 post-infection. More relevant to PD pathology, the neurotropic H5N1 influenza virus was found to induce long lasting

10

PLFURJOLD DFWLYDWLRQ DQG Į-synuclein phosphorylation and aggregation in the mouse SN postinfection (34, 69). An increased incidence of HSV-I DNA in the CNS and increased sensitivity to respiratory infections among the elderly (70, 71) is worth noting since the greatest risk factor for PD is old age (72). With age, the blood-brain barrier (BBB) becomes more permeable, resulting in more fluid entry of peripheral proteins into the CNS (including neurotoxic peripheral proinflammatory mediators) (73). The immune system is also compromised in the elderly, with increases in pro-inflammatory cytokines and decreases in lymphocytes (74). Disruptions to the BBB and normal immune processes among the aged population could also result in increased entry of HSV-I and influenza A into the CNS during infection/HSV-I reactivation. Since HSV-I and some strains of influenza A have demonstrated their ability to infect the CNS (especially among the elderly) without immediately fatal consequences, the effects of these viral infections in host neurons in the CNS will be reviewed, with a focus on PD-related pathology.

Viral Infection in the CNS: Inflammation, Synaptic Dysfunction, and Autophagy Disruption Upon viral infection, the host immune system usually becomes activated and attempts to remove or destroy the invading pathogen via inflammatory mediators, autophagy degradation, or sometimes controlled cell death of infected cells (75). Although viral pathogens have evolved multiple ways of evading the host immune response (and so host clearance of viral pathogens), host immune circumvention is dependent on virus strain, evasion strategy, and host cell type. Of relevance to PD, the viral evasion of the host immune response may be modulated by BBB

11

integrity, CNS immune cell sensitivity, and duration/severity of infection. The next few sections review viral modulation of the host immune/autophagy response due to HSV-I or influenza A infection. Viral mediated inflammation, synaptic dysfunction, and autophagy disruption in the CNS will be discussed.

Neuroinflammation The human immune system is divided into the adaptive (memory based, specific response) and innate (genetically conserved, non-specific response) immune systems (76). Macrophages are able to attack pathogens due to pattern-recognition receptors (PRRs) that have evolved to recognize pathogen-associated molecular patterns (PAMPs) (77, 78). Meanwhile, the adaptive immune response uses lymphocytes (B-cells and T-cells) to ‘remember’ and attack the pathogen more efficiently (79, 80). This immune response sometimes includes cytotoxic lymphocytes, which sacrifice and destroy infected host cells. Of importance to HSV-I, CD8+ T-cells have been found to have HSV-I epitopes and block reactivation (81, 82). Although involved in hindering HSV-I reactivation, there are suggestions that these T-cells lead to chronic inflammation. Residual lymphocytes were found to recognize HSV-I during latency in the TG, resulting in cytokine release, T-cell exhaustion, and eventual allowing of viral reactivation (83-85). The H5N1 influenza A strain was also found to induce excessive peripheral T-cell activation (86). There is evidence of T-cell population modulation in PD as well. T-cell population increases/ decreases and impairment in PD depends on T-cell type, and more recently T-FHOOVKDYHEHHQIRXQGWRUHFRJQL]HĮ-synuclein epitopes (87, 88). Interestingly, recent studies identified homologous cross-reactivity between HSV-, DQG Į-

12

synuclein, suggesting that HSV-I may induce an auto-immune response (89). Indeed, autoantibodies against a HSV-I peptide were cross-UHDFWLYHZLWKDQĮ-synuclein epitope (89). While lymphocytes are involved in the adaptive immune response, the innate immune system also initiates an immediate response due to PAMPs. A key set of PRRs regulating the innate immune system are the Toll-like receptors (TLRs). TLRs are glycoprotein transmembrane receptors that recognize PAMPs (such as lipopolysaccharides, dsDNA/RNA, ssRNA) (90). Of significance to HSV-I and influenza A, TLR3 is known to recognize viral dsRNA that is present during the viral life cycle within infected host cells (91, 92). TLR3 activation leads to proinflammatory cytokine and type I IFN-Įȕ production, and regulation of DNA expression through nuclear factor kappa-light-chain-enhancer of activated B (NF-Ƹ% and interferon regulatory factor (IRF) activation (93). Neuroinflammation in PD patients has previously been investigated to characterize potential bio-markers. Genetic mutations in PD-related genes (lrrk2 and parkin) have been found to regulate the immune system response (94-98). Also, single nucleotide polymorphisms in the Major Histocompatibility Complex-II (an antigen-presenting component of specific adaptive immune cells) locus were associated with an increased incidence of PD (99-102). Post-mortem studies have found increased levels of pro-inflammatory cytokines (103). They also found increased levels of IFNs and p65 subunits of NF-ț% (104). CSF and peripheral levels of cytokines are also elevated in PD (104, 105). Although the role of these cytokines/IFNs in PD is unknown, animal models have demonstrated that increases in pro-inflammatory mediators results in dopaminergic neurodegeneration (106, 107). Examination of the TLR profile in animal models found increases in TLR3/4 expression in the striatum in response to OS and the pesticide rotenone, possibly leaving these cell populations/brain regions more sensitive to an infection

13

(108). Interestingly, certain viral infections have found ways to circumvent IFN-stimulated pathways, possibly allowing them to enter and remain dormant in the CNS. Although the host immune system is well evolved to combat viral infections through type 1 IFNs and IFN-stimulated genes (ISGs), HSV-I and influenza A have also evolved ways to evade this host immune response. HSV-I proteins inhibit NF-ț%and IRF3 (a TLR3 downstream regulator of IFNs) activation (109-111). The influenza A non-structural protein 1 (NS1) prevents the host innate immune response and cellular apoptosis of infected cells by suppressing IFN activation through multiple routes (112). Also, NS1 regulates IFN-Įȕ UHFHSWRU VXEXQLW  (IFNAR1) surface expression (113). Due to suppression of the innate immune system, influenza A infection of neurons only leads to increases in tumor necrosis factor-Į TNF-Į) release, not interleukin 6 (IL-6) or IFNs (114). Despite multiple HSV-I proteins working to dampen IFN signaling, there still has been clear evidence of IFN signaling and regulation of viral replication in HSV-I infected cells. This is not surprising since HSV-I inhibition or activation of IRF3 appears to be cell type dependent (111). Sensory neurons, where HSV-I latency is generally maintained, are innately unable to mount a large IFN response (115). This may be why sensory neurons are ideal for HSV-I to maintain latency, but even so, some level of IFN signaling may be required for HSV-I reactivation. Latency-associated transcripts (LATs) have not been found to produce an inflammatory cytokine/IFN response themselves, but instead may require some cytokines/IFNs to initiate reactivation (116, 117). One study suggested that IFNs regulate LAT expression in a way that benefits HSV-I infection; by promoting neuron cell survival throughout latency, HSV-I is provided an opportunity for reactivation and viral spread (117). Interestingly, neuronal IFN-ȕ VXSSUHVVLRQZDVDVVRFLDWHGZLWKĮ-synuclein accumulation and PD-like neurodegeneration (118).

14

Although HSV-I and influenza A viruses have evolved ways to circumvent neuronal innate immune sensing of infection, other CNS cells can still sense and defend against pathogens (see Table 2). Glial cells in the CNS mainly function as regulators of the cellular environment to promote healthy neuronal cell function. Astrocytes (the most abundant cell type in the CNS) support neuron homeostasis by regulating synaptic activity, assisting in BBB formation, and interacting with immune cells. They regulate neurotransmission and metabolism by controlling extracellular potassium levels, uptake of neurotransmitters (such as glutamate), and storing glycogens/exporting lactate (119). Microglia cells act as resident immune cells in the CNS, with the capability of sensing, engulfing, and degrading invading pathogens (120). The activation of microglia can have neuroprotective or neurotoxic effects depending on their microenvironment. When activated, some microglia release reactive oxygen species (ROS), inducible nitric oxide synthase (iNOS), and cytokines (121). These oxidative species and cytokines interact with dopaminergic neurons to regulate cell fate during stress (121). Astrocytes and microglia participate in the defence against viral spread throughout the CNS (122). Although HSV-I may find a safe haven in sensory neurons, replication in these neurons for reactivation may alert neighboring astrocytes. These cells recognize extracellular dsRNA since they can express cell surface TLR3 (123). Indeed, previous studies have found astrocytes to be reactive to a synthetic mimetic of dsRNA, albeit with conflicting conclusions (123-128). One study found synthetic dsRNA to produce an anti-inflammatory response in astrocytes (124), while others found a pro-inflammatory response (125, 128). The reasons for these differences may be due to astrocyte source (fetal or adult). Overall, synthetic dsRNA treatment in human astrocytes in vitro was found to cause increases in IFNs, IL-6, and a downregulation in connexin 43 (a crucial protein for intercellular gap junctions between

15

astrocytes and maintaining BBB integrity) (125, 128, 129). Interestingly, a rat study also found synthetic dsRNA to attenuate astrocytic L-glutamate uptake by inhibiting EAAT1/GLAST transporter gene transcription (130). Studies examining HSV-I infection in the mouse CNS found increased inflammation and ROS (131, 132). These studies suggest viral infection and replication in neurons near astrocytes could cause an inflammatory response and disrupt healthy astrocyte function, possibly leading to neuronal signaling dysfunction and cell death. Of relevance to HSV-I and glia activity, a study describing a mouse model of HSE found lytic genes (ICP0 and ICP27) to sustain their expression long into ‘latency’ within the brain ependymal after HSE recovery (133). This HSV-I gene expression profile differs from its life in the TG and was associated with a loss of effector T-cell function and an increase in microglia in the region. Although most humans infected with HSV-I never experience an episode of HSE during their lifetime, this study not only further demonstrates that not all cell types respond in the same way to HSV-I, but that HSV-I can infect regions of the CNS without lethal consequences.

Synaptic Dysfunction Previous models of PD have suggested that synaptic dysfunction (such as alterations to long term potentiation/depression, changes in synaptic proteins, and NMDA receptor (NMDAR) subunit composition) in nigrostriatal and corticostriatal pathways could be responsible for the physical manifestations of DA loss in the SN (134-136). Post-mortem studies have found decreases in glutamatergic synapses and AMPA GluR1 in the striatal regions of PD patients (137, 138). Also, human induced pluripotent stem cell derived neurons from familial PD patients demonstrated reduced synaptic connectivity and hindered neurite outgrowth (139). Although more research

16

needs to be conducted to better understand synaptic dysfunction in PD, HSV-I and influenza A associated modifications to normal synaptic function are worth review (see Table 2). Influenza A infection has been found to disrupt synaptic activity through modulation of host gene expression and interaction with synaptic related proteins. Pandemic and seasonal influenza A infections were examined for their modulation of genes in the CNS (140). Pandemic influenza strains were associated with downregulation of ‘neuron projection,’ ‘synapse assembly,’ and ‘calcium channel activity’ related genes (140). Genes that were downregulated compared WRWKHVHDVRQDOIOXVWUDLQLQFOXGHG*O\FRSURWHLQ0$3URWRFDGKHULQĮ-subfamily C2, and CAMP regulated phosphoprotein. Influenza A NS1 and nucleoprotein (NP) were also found to modify the host synapse. The influenza A NP was found to localize within dendritic spine-like structures of hippocampal neurons, resulting in reduced spontaneous excitatory synaptic frequency and decreased amplitude of excitatory post-synaptic currents (141). Also in hippocampal neurons, the PDZ motif of the C-terminus of H5N1 influenza A NS1 (not H1N1 influenza A NS1) was found to bind to post-synaptic density protein-95 (PSD-95) (142). NS1 binding to PSD-95 was suggested to prevent normal post-synaptic processes. Although there is no evidence of direct inhibition of synaptic proteins, HSV-I infection is associated with changes in the host synapse. HSE patients are often found to have NMDAR antibodies, with a reduction in NMDAR and synapsin protein in murine hippocampal neurons after treatment with HSE patient serum (143). In an animal model, HSV-I infection of murine cortical neurons resulted in reduction of synapsin-1 and synaptophysin proteins, and disrupted synaptic transmission (144). Although synaptic dysfunction in PD may be a result of other features of 3' SDWKRORJ\ VXFK DV Į-synuclein aggregation or DA striatal denervation), it is worth noting viral induced changes in synaptic function. HSV-I or influenza A infection may

17

exacerbate already stressed synaptic connections or weaken synaptic activity before other PD pathological features have fully manifested.

Autophagy The autophagy process is fundamental for cellular homeostasis. Briefly, unwanted components (misfolded proteins, foreign structures, dysfunctional proteins, etc.) are engulfed in doublemembraned vesicles (autophagosomes) for digestion and substrate recycling (145). Autophagy pathways have been suggested to be very important for amounting an antiviral defence in nonreplicating cells (146). Epithelial cells infected with HSV-I can produce pro-inflammatory cytokines and undergo cell death to prevent viral spread without permanent tissue damage because they can be replaced afterwards. Non-replicating cells, such as neurons, may be more reliant on autophagy processes to limit viral replication and viral spread without undergoing cell death (147). Similar to IFN signaling evasion, HSV-I and influenza A proteins have evolved ways to disrupt autophagy events to prevent clearance of viral components from host cells during latency and replication. Previous studies have identified ICP34.5 as a crucial HSV-I protein for inhibiting autophagic degradation of virion structures (148, 149). Multiple HSV-I and influenza A proteins are able to disrupt autophagy. HSV-I ICP34.5 is able to indirectly inhibit protein kinase RNAactivated (PKR), while HSV-I US11 directly binds to PKR to prevent activation (150-152). Influenza A NS1 and NP also inhibit PKR (153). This PKR activation inhibition prevents PKRmediated autophagy activation (149, 153). Also, HSV-I ICP34.5 has been found to bind directly to Beclin-1 (154). Beclin-1 binds with other autophagy components to promote the formation of

18

autophagosomes (155). The amino acid region 68-87 of ICP34.5 binds to Beclin-1, leaving the section that functions to inhibit PKR signaling to remain open (154). HSV-I ICP34.5 inhibition of autophagy through Beclin-1 binding also prevents antigen presentation and CD4+ T-cell response (156). Further investigation into the consequences of ICP34.5-mediated autophagy disruption needs to be done to understand the effects on host neuron homeostasis beyond increased neurovirulence of HSV-I. Viral mediated autophagy disruption or suppression could lead to a decrease in clearance of misfolded/aggregated proteins. Studies examining post-mortem AD brains found HSV-I DNA to be associated with amyloid-beta plaques (63, 64, 157). Although a majority of HSV-I DNA positive CNS neurons were found to have amyloid-beta plaques, there was no correlation between amyloid-beta plaque containing neurons and presence of HSV-I DNA (63). These findings suggest that HSV-I infection may cause an increase in amyloid protein aggregation. Further investigation should be conducted to determine if there may also be an association between HSV-, '1$ SRVLWLYH QHXURQV DQG Į-synuclein aggregation. A study by Santana et al. found HSV-I infection in neuronal cell cultures to cause an increase in amyloid-beta aggregation accumulation, along with an increase in LC3-II (158). This study further supports the theory that HSV-I may contribute to amyloid protein aggregation, but does not refute the possibility that this is related to HSV-I mediated autophagy disruption since ICP34.5 disrupts autophagy through Beclin-1 binding, not LC3-II. Also, the lack of correlation between amyloid-beta plaque containing neurons and HSV-I DNA positivity may be due to cross-seeding. HSV-I may cause autophagy disruption in innervating neurons (possibly latent sensory neurons),

leading to

amyloid fibril accumulations that may have the capability of spreading to other non-HSV-I DNA positive neurons. These protein aggregates may then be transported to innervating neurons or be

19

exocytosed for extracellular cross-seeding. In general, autophagy disruption has been previously found to cause an increase in neurodegeneration, pre-synaptic Į-synuclein accumulation, neuronal inclusions, and dopaminergic axon and dendritic degeneration (159, 160).

Conclusion The purpose of this review was to demonstrate the need to revisit the association between viral infections and PD. It is clear that parallels can be drawn between viral-induced changes in the CNS (ranging from chronic inflammation to synaptic dysfunction) and PD pathology (Figure 1). Further investigation of viral infections (specifically HSV-I and influenza A) should be conducted to determine if intervention can suppress the long-term consequences in the CNS and possibly mitigate the association between viral infections and incidence of PD.

20

Viral Infection

Relation to PD

Study

Herpes Simplex Virus

Ĺ,QFLGHQFH +69-I) Ĺ,QFLGHQFH Ĺ,QFLGHQFH +69-I) No Association Ĺ,QFLGHQFH Ĺ,QFLGHQFH No Association Ļ,QFLGHQFH No Association No Association Ļ,QFLGHQFH No Association No Association No Association

(44-46, 161) (48) (49) (47) (48) (47) (44) (47) (48) (161) (47) (46) (49) (162)

Ĺ,QFLGHQFH

(48)

-Enveloped, linear dsDNA genome (152kb- 154 kb) -Causes blistering of mouth/genitals, latency in neurons

Influenzavirus A -Enveloped, linear ssRNA(-) genome (13.5kb) -Causes fever, cough, runny nose, malaise

Measles Virus -Enveloped, linear ssRNA(-) genome (15-16kb) -Causes rash, white spots, cough, red eyes

Cytomegalovirus -Enveloped, linear dsDNA genome (~200kb) -May cause mononucleosis, pnuemonia

Coronavirus -Enveloped, linear ssRNA(+) genome (27- 32kb) -Causes upper/lower respiratory infection, sometimes gastroenteritis

Mumps -Enveloped, linear ssRNA(-) genome (15kb) -Causes parotid gland swelling, malaise

Table 1. Viral Infection Associations with Parkinson’s Disease.

21

THEME Inflammation Glial Cells

T-cells

Cytokines

CONDITION PD

HSV-I

Influenza A

Activated microglia and astrogliosis in PD midbrain (163,

ĹPLFURJOLDLQHSV-I infected ependymal

H1N1 ĹDVWURJOLRVLVDQGDFWLYDWHG

164) T-cell modulation and UHFRJQLWLRQRIĮsynuclein epitopes in PD (87, 166)

Ĺ&6)DQG peripheral cytokines in PD (104, 105)

microglia in SN and VTA (165)

(133) Exhausted T-cells in HSV-I infected brain stem and TG express HSV-I epitope (81, 83, 133)

H5N1 causes excessive peripheral T-cell activation (86)

T-cell associated F\WRNLQHVĹLQ+69 infected TG (82, 84,

+1ĹDVWURF\WHQHXURQDO cytokines (167, 168)

85)

Autophagy

Disruption

PD

HSV-I

Influenza A

Autophagic and lysosomal defects in PD neurons (159,

PKR inhibition disrupts autophagy/ autophagosome formation (149, 154,

PKR inhibition and autophagosome/lysosome fusion blocked (153, 170)

169)

155)

Synapse Proteins

PD

HSV-I

Influenza A

Redistribution of synaptic proteins in PD models (171,

Ļ synapsin-1 and synaptophysin in murine cortical neurons (143, 144) Reduced NMDAR and synaptic activity in HSE patients (143,

H5N1 inhibits PSD-95; SNAP25 differentially expressed in neonatal infection

172)

(142, 173)

Ļsynaptic ĻQHXURQDOH[FLWDWRU\synaptic connectivity and activity and amplitude (141) Activity glutamatergic 174) synapse loss in PD models (138, 139) Table 2. Viral Induced Molecular/Cellular Changes Related to PD Pathology. As detailed above, multiple parallels can be drawn between PD pathology and the potential molecular and cellular consequences of HSV-I/influenza A infection.

22

Figure 1. HSV-I and Influenza A Viral Infections May Lead to Parkinson’s Disease-like Pathology. HSV-I and influenza A viral infections have the potential to cause molecular and cellular changes that can alter healthy neuron function within the CNS. Viral transcripts/proteins due to HSV-I/influenza A infection may cause inflammation, autophagy disruption, and synapse dysfunction, possibly contributing to a Parkinson’s Disease-like pathology.

23

Bibliography 1. Tanner CM, Goldman SM. Epidemiology of Parkinson's disease. Neurologic clinics. 1996;14(2):317-35. 2. Hoehn MM, Yahr MD. Parkinsonism onset, progression, and mortality. Neurology. 1967;17(5):427-. 3. Martinez-Martin P, Chaudhuri KR, Rojo-Abuin JM, Rodriguez-Blazquez C, Alvarez-Sanchez M, Arakaki T, et al. Assessing the non-motor symptoms of Parkinson's disease: MDS-UPDRS and NMS Scale. European journal of neurology. 2015;22(1):37-43. 4. Pfeiffer RF. Non-motor symptoms in Parkinson's disease. Parkinsonism & related disorders. 2016;22:S119-S22. 5. Braak H, Del Tredici K, Rüb U, De Vos RA, Steur ENJ, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of aging. 2003;24(2):197-211. 6. Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold Spring Harbor perspectives in medicine. 2012;2(1):a008888. 7. 3RO\PHURSRXORV0+/DYHGDQ&/HUR\(,GH6('HKHMLD$'XWUD$HWDO0XWDWLRQLQWKHĮsynuclein gene identified in families with Parkinson's disease. science. 1997;276(5321):2045-7. 8. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, (.RIĮ-synuclein causes parkinson and Lewy body dementia. Annals of neurology. 2004;55(2):16473. 9. Chung KK, Zhang Y, Lim KL, Tanaka Y, Huang H, Gao J, et al. Parkin ubiTXLWLQDWHVWKHĮsynuclein–interacting protein, synphilin-1: implications for Lewy-body formation in Parkinson disease. Nature medicine. 2001;7(10):1144-50. 10. Lynch-Day MA, Mao K, Wang K, Zhao M, Klionsky DJ. The Role of Autophagy in Parkinson’s Disease. Cold Spring Harbor perspectives in medicine. 2012;2(4):a009357. 11. Taylor JM, Main BS, Crack PJ. Neuroinflammation and oxidative stress: Co-conspirators in the pathology of Parkinson’s disease. Neurochemistry International. 2013;62(5):803-19. 12. Lai B, Marion S, Teschke K, Tsui J. Occupational and environmental risk factors for Parkinson's disease. Parkinsonism & related disorders. 2002;8(5):297-309. 13. Liou H, Tsai M, Chen C, Jeng J, Chang Y, Chen S, et al. Environmental risk factors and Parkinson's disease A case-control study in Taiwan. Neurology. 1997;48(6):1583-8. 14. Seidler A, Hellenbrand W, Robra B-P, Vieregge P, Nischan P, Joerg J, et al. Possible environmental, occupational, and other etiologic factors for Parkinson's disease A case-control study in Germany. Neurology. 1996;46(5):1275-. 15. Breckenridge CB, Berry C, Chang ET, Sielken Jr RL, Mandel JS. Association between Parkinson’s disease and cigarette smoking, rural living, well-water consumption, farming and pesticide use: systematic review and meta-analysis. PloS one. 2016;11(4):e0151841. 16. Van Maele-Fabry G, Hoet P, Vilain F, Lison D. Occupational exposure to pesticides and Parkinson's disease: a systematic review and meta-analysis of cohort studies. Environment international. 2012;46:30-43. 17. Itzhaki RF, Lathe R, Balin BJ, Ball MJ, Bearer EL, Braak H, et al. Microbes and Alzheimer’s disease. Journal of Alzheimer's disease: JAD. 2016;51(4):979. 18. Von Economo C. Encephalitis Lethargica: Its Sequelae and Treatment: Milford; 1931. 19. Neal JB, Bentley IA. Treatment of epidemic encephalitis: a review of the work of the Matheson Commission. Archives of Neurology & Psychiatry. 1932;28(4):897-907. 20. Ravenholt R, Foege W. 1918 influenza, encephalitis lethargica, parkinsonism. The Lancet. 1982;320(8303):860-4. 21. Lo K, Geddes J, Daniels R, Oxford J. Lack of detection of influenza genes in archived formalinfixed, paraffin wax-embedded brain samples of encephalitis lethargica patients from 1916 to 1920. Virchows Archiv. 2003;442(6):591-6. 24

22. McCall S, Henry JM, Reid AH, Taubenberger JK. Influenza RNA not detected in archival brain tissues from acute encephalitis lethargica cases or in postencephalitic Parkinson cases. Journal of Neuropathology & Experimental Neurology. 2001;60(7):696-704. 23. Taubenberger JK, Reid AH, Krafft AE, Bijwaard KE, Fanning TG. Initial genetic characterization of the 1918 “Spanish” influenza virus. science. 1997;275(5307):1793-6. 24. Reid AH, Fanning TG, Hultin JV, Taubenberger JK. Origin and evolution of the 1918 “Spanish” influenza virus hemagglutinin gene. Proceedings of the National Academy of Sciences. 1999;96(4):16516. 25. Reid AH, Fanning TG, Janczewski TA, Taubenberger JK. Characterization of the 1918 “Spanish” influenza virus neuraminidase gene. Proceedings of the National Academy of Sciences. 2000;97(12):6785-90. 26. Anderson L, Vilensky J, Duvoisin R. Neuropathology of acute phase encephalitis lethargica: a review of cases from the epidemic period. Neuropathology and applied neurobiology. 2009;35(5):462-72. 27. Dale RC, Church AJ, Surtees RA, Lees AJ, Adcock JE, Harding B, et al. Encephalitis lethargica syndrome: 20 new cases and evidence of basal ganglia autoimmunity. Brain. 2004;127(1):21-33. 28. Dale RC, Irani SR, Brilot F, Pillai S, Webster R, Gill D, et al. N-methyl-D-aspartate receptor antibodies in pediatric dyskinetic encephalitis lethargica. Annals of neurology. 2009;66(5):704-9. 29. Lopez-Alberola R, Georgiou M, Sfakianakis GN, Singer C, Papapetropoulos S. Contemporary encephalitis lethargica: phenotype, laboratory findings and treatment outcomes. Journal of neurology. 2009;256(3):396-404. 30. Rail D, Scholtz C, Swash M. Post-encephalitic parkinsonism: current experience. Journal of Neurology, Neurosurgery & Psychiatry. 1981;44(8):670-6. 31. Singer HS, Hong JJ, Yoon DY, Williams PN. Serum autoantibodies do not differentiate PANDAS and Tourette syndrome from controls. Neurology. 2005;65(11):1701-7. 32. Vincent A, Buckley C, Schott JM, Baker I, Dewar BK, Detert N, et al. Potassium channel antibody-associated encephalopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain. 2004;127(3):701-12. 33. de Jong MD, Cam BV, Qui PT, Hien VM, Thanh TT, Hue NB, et al. Fatal Avian Influenza A (H5N1) in a Child Presenting with Diarrhea Followed by Coma. New England Journal of Medicine. 2005;352(7):686-91. 34. 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. 2009;106(33):14063-8. 35. Mattos JPd, Rosso ALZd, Corrêa RB, Novis SA. Movement disorders in 28 HIV-infected patients. Arquivos de neuro-psiquiatria. 2002;60(3A):525-30. 36. POSER CM, HUNTLEY CJ, POLAND JD. PARA-ENCEPHALITIC PARKINSONISM. Acta Neurologica Scandinavica. 1969;45(2):199-215. 37. Pranzatelli MR, Mott SH, Pavlakis SG, Conry JA, Tate ED. Clinical spectrum of secondary parkinsonism in childhood: a reversible disorder. Pediatric neurology. 1994;10(2):131-40. 38. Tse W, Cersosimo MG, Gracies J-M, Morgello S, Olanow CW, Koller W. Movement disorders and AIDS: a review. Parkinsonism & related disorders. 2004;10(6):323-34. 39. Walters JH. Postencephalitic Parkinson syndrome after meningoencephalitis due to coxsackie virus group B, type 2. New England Journal of Medicine. 1960;263(15):744-7. 40. Elizan TS, Madden DL, Noble GR, Herrmann KL, Gardner J, Schwartz J, et al. Viral antibodies in serum and CSF of Parkinsonian patients and controls. Archives of neurology. 1979;36(9):529-34. 41. Chen HH, Liu PC, Tsai HH, Yen RF, Liou HH. Re: Wangensteen et al. of a letter on ‘Hepatitis C virus infection: a risk factor for Parkinson's disease.'. Journal of viral hepatitis. 2016;23(7):560-. 42. Tsai H-H, Liou H-H, Muo C-H, Lee C-Z, Yen R-F, Kao C-H. Hepatitis C virus infection as a risk factor for Parkinson disease A nationwide cohort study. Neurology. 2016;86(9):840-6. 43. Wu WYY, Kang KH, Chen SLS, Chiu SYH, Yen AMF, Fann JCY, et al. Hepatitis C virus infection: a risk factor for Parkinson's disease. Journal of viral hepatitis. 2015;22(10):784-91. 25

44. Marttila R, Arstila P, Nikoskelainen J, Halonen P, Rinne U. Viral antibodies in the sera from patients with Parkinson disease. European neurology. 1977;15(1):25-33. 45. Marttila RJ, Rinne UK. Herpes simplex virus antibodies in patients with Parkinson's disease. Journal of the neurological sciences. 1978;35(2):375-9. 46. Marttila RJ, Rinne UK, Halonen P, Madden DL, Sever JL. Herpesviruses and Parkinsonism: herpes simplex virus types 1 and 2, and cytomegalovirus antibodies in serum and CSF. Archives of neurology. 1981;38(1):19-21. 47. Harris MA, Tsui JK, Marion SA, Shen H, Teschke K. Association of Parkinson's disease with infections and occupational exposure to possible vectors. Movement Disorders. 2012;27(9):1111-7. 48. Vlajinac H, Dzoljic E, Maksimovic J, Marinkovic J, Sipetic S, Kostic V. Infections as a risk factor for Parkinson's disease: a case–control study. International Journal of Neuroscience. 2013;123(5):329-32. 49. Bu XL, Yao XQ, Jiao SS, Zeng F, Liu YH, Xiang Y, et al. A study on the association between infectious burden and Alzheimer's disease. European journal of neurology. 2015;22(12):1519-25. 50. Samji T. Influenza A: Understanding the Viral Life Cycle. The Yale Journal of Biology and Medicine. 2009;82(4):153-9. 51. Kumar SP, Chandy ML, Shanavas M, Khan S, Suresh K. Pathogenesis and life cycle of herpes simplex virus infection-stages of primary, latency and recurrence. Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology. 2016;28(4):350-3. 52. Marfurt CF, Rajchert DM. Trigeminal primary afferent projections to “non-trigeminal” areas of the rat central nervous system. Journal of Comparative Neurology. 1991;303(3):489-511. 53. Miller KD, Schnell MJ, Rall GF. Keeping it in check: chronic viral infection and antiviral immunity in the brain. Nature Reviews Neuroscience. 2016;17(12):766-76. 54. Kuiken T, Taubenberger JK. Pathology of human influenza revisited. Vaccine. 2008;26:D59D66. 55. Matsuda K, Shibata T, Sakoda Y, Kida H, Kimura T, Ochiai K, et al. In vitro demonstration of neural transmission of avian influenza A virus. Journal of general virology. 2005;86(4):1131-9. 56. van Riel D, Verdijk R, Kuiken T. The olfactory nerve: a shortcut for influenza and other viral diseases into the central nervous system. The Journal of Pathology. 2015;235(2):277-87. 57. Esiri MM. Herpes simplex encephalitis: an immunohistological study of the distribution of viral antigen within the brain. Journal of the neurological sciences. 1982;54(2):209-26. 58. Shoji H, Koga M, Kusuhara T, Kaji M, Ayabe M, Hino H, et al. Differentiation of herpes simplex virus 1 and 2 in cerebrospinal fluid of patients with HSV encephalitis and meningitis by stringent hybridization of PCR-amplified DNAs. Journal of neurology. 1994;241(9):526-30. 59. Hemling N, Röyttä M, Rinne J, Pöllänen P, Broberg E, Tapio V, et al. Herpesviruses in brains in Alzheimer's and Parkinson's diseases. Annals of neurology. 2003;54(2):267-71. 60. Wetmur JG, Schwartz J, Elizan TS. Nucleic acid homology studies of viral nucleic acids in idiopathic Parkinson's disease. Archives of neurology. 1979;36(8):462-4. 61. Fraser NW, Lawrence WC, Wroblewska Z, Gilden DH, Koprowski H. Herpes simplex type 1 DNA in human brain tissue. Proceedings of the National Academy of Sciences. 1981;78(10):6461-5. 62. Gordon L, McQuaid S, Cosby S. Detection of herpes simplex virus (types 1 and 2) and human herpesvirus 6 DNA in human brain tissue by polymerase chain reaction. Clinical and diagnostic virology. 1996;6(1):33-40. 63. Olsson J, Lövheim H, Honkala E, Karhunen PJ, Elgh F, Kok EH. HSV presence in brains of individuals without dementia: the TASTY brain series. Disease models & mechanisms. 2016;9(11):134955. 64. Wozniak MA, Frost AL, Itzhaki RF. Alzheimer's disease-specific tau phosphorylation is induced by herpes simplex virus type 1. Journal of Alzheimer's Disease. 2009;16(2):341-50. 65. Park CH, Ishinaka M, Takada A, Kida H, Kimura T, Ochiai K, et al. The invasion routes of neurovirulent A/Hong Kong/483/97 (H5N1) influenza virus into the central nervous system after respiratory infection in mice. Archives of Virology. 2002;147(7):1425-36. 26

66. Reinacher M, Bonin J, Narayan O, Scholtissek C. Pathogenesis of neurovirulent influenza A virus infection in mice. Route of entry of virus into brain determines infection of different populations of cells. Lab Invest. 1983;49(6):686-92. 67. 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):2161-9. 68. Yamada T, Yamanaka I, Takahashi M, Nakajima S. Invasion of brain by neurovirulent influenza A virus after intranasal inoculation. Parkinsonism & related disorders. 1996;2(4):187-93. 69. 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. Journal of Neuroscience. 2012;32(5):1545-59. 70. Jamieson GA, Maitland NJ, Wilcock GK, Yates CM, Itzhaki RF. Herpes simplex virus type 1 DNA is present in specific regions of brain from aged people with and without senile dementia of the Alzheimer type. The Journal of Pathology. 1992;167(4):365-8. 71. Nicholson KG, Kent J, Hammersley V, Cancio E. Acute viral infections of upper respiratory tract in elderly people living in the community: comparative, prospective, population based study of disease burden. BMJ. 1997;315(7115):1060-4. 72. De Lau LM, Breteler MM. Epidemiology of Parkinson's disease. The Lancet Neurology. 2006;5(6):525-35. 73. Kleine T, Hackler R, Zöfel P. Age-related alterations of the blood-brain-barrier (bbb) permeability to protein molecules of different size. Zeitschrift fur Gerontologie. 1992;26(4):256-9. 74. Valiathan R, Ashman M, Asthana D. Effects of ageing on the immune system: infants to elderly. Scandinavian journal of immunology. 2016;83(4):255-66. 75. Levine B. Eating Oneself and Uninvited Guests: Autophagy-Related Pathways in Cellular Defense. Cell. 2005;120(2):159-62. 76. Abbas AK, Lichtman AH, Pillai S. Basic immunology: functions and disorders of the immune system: Elsevier Health Sciences; 2014. 77. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nature Reviews Immunology. 2008;8(12):958-69. 78. 6FKURGHU.6ZHHW0-+XPH'$6LJQDOLQWHJUDWLRQEHWZHHQ,)1ȖDQG7/5VLJQDOOLQJSDWKZD\V in macrophages. Immunobiology. 2006;211(6):511-24. 79. Dutton R, Bradley L, Swain S. T cell memory. Annual review of immunology. 1998;16(1):20123. 80. Rajewsky K, Schittek B. Maintenance of B-cell memory by long-lived cells generated from proliferating precursors. Nature. 1990;346(6286):749. 81. Khanna KM, Bonneau RH, Kinchington PR, Hendricks RL. Herpes Simplex Virus-Specific Memory CD8+ T Cells Are Selectively Activated and Retained in Latently Infected Sensory Ganglia. Immunity. 2003;18(5):593-603. 82. Leger AJS, Jeon S, Hendricks RL. Broadening the Repertoire of Functional Herpes Simplex Virus Type 1–Specific CD8+ T Cells Reduces Viral Reactivation from Latency in Sensory Ganglia. The Journal of Immunology. 2013;191(5):2258-65. 83. Benmohamed L, Srivastava R, Khan AA. The Herpes Simplex Virus LAT Gene is Associated with a Broader Repertoire of Virus-Specific Exhausted CD8+ T Cells Retained Within The Trigeminal Ganglia of Latently Infected HLA Transgenic Rabbits. The Journal of Immunology. 2016;196(1 Supplement):79.14-79.14. 84. Halford WP, Gebhardt BM, Carr DJ. Persistent cytokine expression in trigeminal ganglion latently infected with herpes simplex virus type 1. The Journal of Immunology. 1996;157(8):3542-9. 85. Theil D, Derfuss T, Paripovic I, Herberger S, Meinl E, Schueler O, et al. Latent herpesvirus infection in human trigeminal ganglia causes chronic immune response. The American journal of pathology. 2003;163(6):2179-84.

27

86. Perrone LA, Plowden JK, García-Sastre A, Katz JM, Tumpey TM. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS pathogens. 2008;4(8):e1000115. 87. Saunders JAH, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, et al. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson’s disease. Journal of Neuroimmune Pharmacology. 2012;7(4):927-38. 88. Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. T cells from patients ZLWK3DUNLQVRQ¶VGLVHDVHUHFRJQL]HĮ-synuclein peptides. Nature. 2017;546:656. 89. Caggiu E, Paulus K, Arru G, Piredda R, Sechi GP, Sechi LA. Humoral cross reactivity between Į-synuclein and herpes simplex-1 epitope in Parkinson's disease, a triggering role in the disease? Journal of neuroimmunology. 2016;291:110-4. 90. Chang Z. Important aspects of Toll-like receptors, ligands and their signaling pathways. Inflammation research. 2010;59(10):791-808. 91. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-ț%E\7ROO-like receptor 3. Nature. 2001;413(6857):732-8. 92. Matsumoto M, Kikkawa S, Kohase M, Miyake K, Seya T. Establishment of a monoclonal antibody against human Toll-like receptor 3 that blocks double-stranded RNA-mediated signaling. Biochemical and biophysical research communications. 2002;293(5):1364-9. 93. O'Neill LA, Golenbock D, Bowie AG. The history of Toll-like receptors - redefining innate immunity. Nature Reviews Immunology. 2013;13(6):453-60. 94. Bonifati V. Autosomal recessive parkinsonism. Parkinsonism & related disorders. 2012;18:S4S6. 95. Hakimi M, Selvanantham T, Swinton E, Padmore RF, Tong Y, Kabbach G, et al. Parkinson’s disease-linked LRRK2 is expressed in circulating and tissue immune cells and upregulated following recognition of microbial structures. Journal of Neural Transmission. 2011;118(5):795-808. 96. Manzanillo PS, Ayres JS, Watson RO, Collins AC, Souza G, Rae CS, et al. The ubiquitin ligase parkin mediates resistance to intracellular pathogens. Nature. 2013;501:512. 97. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, et al. Mutations in LRRK2 Cause Autosomal-Dominant Parkinsonism with Pleomorphic Pathology. Neuron. 2004;44(4):601-7. 98. Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease. Nature genetics. 2009;41:1303. 99. Guo Y, Deng X, Zheng W, Xu H, Song Z, Liang H, et al. HLA rs3129882 variant in Chinese Han patients with late-onset sporadic Parkinson disease. Neuroscience letters. 2011;501(3):185-7. 100. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nature genetics. 2010;42(9):781-5. 101. Ahmed I, Tamouza R, Delord M, Krishnamoorthy R, Tzourio C, Mulot C, et al. Association between Parkinson's disease and the HLA-DRB1 locus. Movement Disorders. 2012;27(9):1104-10. 102. International Parkinson Disease Genomics C. Imputation of sequence variants for identification of genetic risks for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet. 2011;377(9766):641-9. 103. Nagatsu T, Mogi M, Ichinose H, Togari A. Cytokines in Parkinson's disease. Journal of Neural Transmission Supplementum. 2000;58:143-52. 104. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)-ȕ,/-2, IL-4, IL-6 and transforming growth factor-ĮOHYHOVDUHHOHYDWHGLQYHQWULFXODUFHUHEURVSLQDOIOXLGLQ juvenile parkinsonism and Parkinson's disease. Neuroscience letters. 1996;211(1):13-6. 105. Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, et al. Peripheral cytokines profile in Parkinson’s disease. Brain, Behavior, and Immunity. 2009;23(1):55-63.

28

106. Depino AM, Earl C, Kaczmarczyk E, Ferrari C, Besedovsky H, Del Rey A, et al. Microglial activation with atypical proinflammatory cytokine expression in a rat model of Parkinson's disease. European Journal of Neuroscience. 2003;18(10):2731-42. 107. Koprich JB, Reske-Nielsen C, Mithal P, Isacson O. Neuroinflammation mediated by IL-ȕ increases susceptibility of dopamine neurons to degeneration in an animal model of Parkinson's disease. Journal of Neuroinflammation. 2008;5(1):8. 108. McCabe K, Concannon RM, McKernan DP, Dowd E. Time-course of striatal Toll-like receptor expression in neurotoxic, environmental and inflammatory rat models of Parkinson's disease. Journal of neuroimmunology. 2017;310:103-6. 109. Antrobus R, Boutell C. Identification of a novel higher molecular weight isoform of USP7/HAUSP that interacts with the Herpes simplex virus type-1 immediate early protein ICP0. Virus research. 2008;137(1):64-71. 110. Lin R, Noyce RS, Collins SE, Everett RD, Mossman KL. The herpes simplex virus ICP0 RING finger domain inhibits IRF3-and IRF7-mediated activation of interferon-stimulated genes. Journal of virology. 2004;78(4):1675-84. 111. Preston CM, Harman AN, Nicholl MJ. Activation of interferon response factor-3 in human cells infected with herpes simplex virus type 1 or human cytomegalovirus. Journal of virology. 2001;75(19):8909-16. 112. Thulasi Raman SN, Zhou Y. Networks of Host Factors that Interact with NS1 Protein of Influenza A Virus. Frontiers in Microbiology. 2016;7:654. 113. Wang BX, Wei L, Kotra LP, Brown EG, Fish EN. A Conserved Residue, Tyrosine (Y) 84, in H5N1 Influenza A Virus NS1 Regulates IFN Signaling Responses to Enhance Viral Infection. Viruses. 2017;9(5):107. 114. Pringproa K, Rungsiwiwut R, Tantilertcharoen R, Praphet R, Pruksananonda K, Baumgärtner W, et al. Tropism and Induction of Cytokines in Human Embryonic-Stem Cells-Derived Neural Progenitors upon Inoculation with Highly- Pathogenic Avian H5N1 Influenza Virus. PloS one. 2015;10(8):e0135850. 115. Richart SM. The role of neurons in the herpes simplex virus type 1 infection. 2003. 116. Carr DJ, Noisakran S, Halford WP, Lukacs N, Asensio V, Campbell IL. Cytokine and chemokine production in HSV-1 latently infected trigeminal ganglion cell cultures: effects of hyperthermic stress. Journal of neuroimmunology. 1998;85(2):111-21. 117. Rosato PC, Katzenell S, Pesola JM, North B, Coen DM, Leib DA. Neuronal IFN signaling is dispensable for the establishment of HSV-1 latency. Virology. 2016;497:323-7. 118. Ejlerskov P, Hultberg Jeanette G, Wang J, Carlsson R, Ambjørn M, Kuss M, et al. Lack of Neuronal IFN-ȕ-IFNAR Causes Lewy Body- and Parkinson’s Disease-like Dementia. Cell. 2015;163(2):324-39. 119. Kandel ER, Schwartz JH, Jessell TM, Siegelbaum SA, Hudspeth AJ. Principles of neural science: McGraw-hill New York; 2000. 120. Banati RB, Gehrmann J, Schubert P, Kreutzberg GW. Cytotoxicity of microglia. Glia. 1993;7(1):111-8. 121. Zhang J, Perry G, Smith MA, Robertson D, Olson SJ, Graham DG, et al. Parkinson's disease is associated with oxidative damage to cytoplasmic DNA and RNA in substantia nigra neurons. The American journal of pathology. 1999;154(5):1423-9. 122. Hauwel M, Furon E, Canova C, Griffiths M, Neal J, Gasque P. Innate (inherent) control of brain infection, brain inflammation and brain repair: the role of microglia, astrocytes, "protective" glial stem cells and stromal ependymal cells. Brain research Brain research reviews. 2005;48(2):220-33. Epub 2005/04/27. 123. Farina C, Krumbholz M, Giese T, Hartmann G, Aloisi F, Meinl E. Preferential expression and function of Toll-like receptor 3 in human astrocytes. Journal of neuroimmunology. 2005;159(1):12-9. 124. Bsibsi M, Persoon-Deen C, Verwer RW, Meeuwsen S, Ravid R, Van Noort JM. Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia. 2006;53(7):688-95. 29

125. Carpentier PA, Begolka WS, Olson JK, Elhofy A, Karpus WJ, Miller SD. Differential activation of astrocytes by innate and adaptive immune stimuli. Glia. 2005;49(3):360-74. 126. Jack CS, Arbour N, Manusow J, Montgrain V, Blain M, McCrea E, et al. TLR signaling tailors innate immune responses in human microglia and astrocytes. The Journal of Immunology. 2005;175(7):4320-30. 127. Park C, Lee S, Cho IH, Lee HK, Kim D, Choi SY, et al. TLR3-mediated signal induces proinflammatory cytokine and chemokine gene expression in astrocytes: differential signaling mechanisms of TLR3-induced IP-10 and IL-8 gene expression. Glia. 2006;53(3):248-56. 128. Zhao Y, Rivieccio MA, Lutz S, Scemes E, Brosnan CF. The TLR3 ligand polyI: C downregulates connexin 43 expression and function in astrocytes by a mechanism involving the NF-ț%DQG3,NLQDVH pathways. Glia. 2006;54(8):775-85. 129. Ezan P, André P, Cisternino S, Saubaméa B, Boulay A-C, Doutremer S, et al. Deletion of astroglial connexins weakens the blood–brain barrier. Journal of Cerebral Blood Flow & Metabolism. 2012;32(8):1457-67. 130. Scumpia PO, Kelly KM, Reeves WH, Stevens BR. Double-stranded RNA signals antiviral and inflammatory programs and dysfunctional glutamate transport in TLR3-expressing astrocytes. Glia. 2005;52(2):153-62. 131. Kavouras JH, Prandovszky E, Valyi-Nagy K, Kovacs SK, Tiwari V, Kovacs M, et al. Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation byproducts in mouse P19N neural cell cultures. Journal of Neurovirology. 2007;13(5):416-25. 132. Valyi-Nagy T, Olson SJ, Valyi-Nagy K, Montine TJ, Dermody TS. Herpes simplex virus type 1 latency in the murine nervous system is associated with oxidative damage to neurons. Virology. 2000;278(2):309-21. 133. Menendez CM, Jinkins JK, Carr DJ. Resident T cells are unable to control herpes simplex virus-1 activity in the brain ependymal region during latency. The Journal of Immunology. 2016;197(4):1262-75. 134. Picconi B, Piccoli G, Calabresi P. Synaptic dysfunction in Parkinson’s disease. Synaptic Plasticity: Springer; 2012. p. 553-72. 135. Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S. A pathologic cascade leading to synaptic dysfunction iQĮ-synuclein-induced neurodegeneration. Journal of Neuroscience. 2010;30(24):8083-95. 136. Volpicelli-'DOH\/$/XN.&3DWHO737DQLN6$5LGGOH'06WLHEHU$HWDO([RJHQRXVĮsynuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72(1):57-71. 137. Bernard V, Gardiol A, Faucheux B, Bloch B, Agid Y, Hirsch EC. Expression of glutamate receptors in the human and rat basal ganglia: Effect of the dopaminergic denervation on AMPA receptor gene expression in the striatopallidal complex in parkinson's disease and rat with 6-OHDA lesion. Journal of Comparative Neurology. 1996;368(4):553-68. 138. Day M, Wang Z, Ding J, An X, Ingham CA, Shering AF, et al. Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nature neuroscience. 2006;9(2):251. 139. Kouroupi G, Taoufik E, Vlachos IS, Tsioras K, Antoniou N, Papastefanaki F, et al. Defective synaptic connectivity and axonal neuropathology in a human iPSC-based model of familial Parkinson’s disease. Proceedings of the National Academy of Sciences. 2017;114(18):E3679-E88. 140. Ebrahimie E, Nurollah Z, Ebrahimi M, Hemmatzadeh F, Ignjatovic J. Unique ability of pandemic influenza to downregulate the genes involved in neuronal disorders. Molecular Biology Reports. 2015;42(9):1377-90. 141. Brask J, Chauhan A, Hill RH, Ljunggren H-G, Kristensson K. Effects on synaptic activity in cultured hippocampal neurons by influenza A viral proteins. Journal of Neurovirology. 2005;11(4):395402.

30

142. Zhang H, Li W, Wang G, Su Y, Zhang C, Chen X, et al. The distinct binding properties between avian/human influenza A virus NS1 and Postsynaptic density protein-95 (PSD-95), and inhibition of nitric oxide production. Virology journal. 2011;8(1):298. 143. Prüss H, Finke C, Höltje M, Hofmann J, Klingbeil C, Probst C, et al. N-methyl-D-aspartate receptor antibodies in herpes simplex encephalitis. Annals of neurology. 2012;72(6):902-11. 144. Piacentini R, Puma DDL, Ripoli C, Marcocci ME, De Chiara G, Garaci E, et al. Herpes Simplex Virus type-1 infection induces synaptic dysfunction in cultured cortical neurons via GSK-3 activation and intraneuronal amyloid-ȕSURWHLQDFFXPXODWLRQ6FLHQWLILF5HSRUWV 145. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2016;12(1):1222. 146. Shoji-Kawata S, Levine B. Autophagy, antiviral immunity, and viral countermeasures. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2009;1793(9):1478-84. 147. Yordy B, Iijima N, Huttner A, Leib D, Iwasaki A. A neuron-specific role for autophagy in antiviral defense against herpes simplex virus. Cell host & microbe. 2012;12(3):334-45. 148. Alexander DE, Ward SL, Mizushima N, Levine B, Leib DA. Analysis of the Role of Autophagy in Replication of Herpes Simplex Virus in Cell Culture. Journal of virology. 2007;81(22):12128-34. 149. Tallóczy Z, Virgin I, Herbert, Levine B. PKR-dependent xenophagic degradation of herpes simplex virus type 1. Autophagy. 2006;2(1):24-9. 150. +H%*URVV05RL]PDQ%7KHȖSURWHLQRIKHUSHVVLPSOH[YLUXVFRPSOH[HVZLWK SURWHLQSKRVSKDWDVHĮWRGHSKRVSKRU\ODWHWKHĮVXEXQLWRIWKHHXNDUyotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated protein kinase. Proceedings of the National Academy of Sciences. 1997;94(3):843-8. 151. Mulvey M, Poppers J, Ladd A, Mohr I. A herpesvirus ribosome-associated, RNA-binding protein confers a growth advantage upon mutants deficient in a GADD34-related function. Journal of virology. 1999;73(4):3375-85. 152. Poppers J, Mulvey M, Khoo D, Mohr I. Inhibition of PKR activation by the proline-rich RNA binding domain of the herpes simplex virus type 1 Us11 protein. Journal of virology. 2000;74(23):1121521. 153. Lussignol M, Queval C, Bernet-Camard M-F, Cotte-Laffitte J, Beau I, Codogno P, et al. The herpes simplex virus 1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. Journal of virology. 2013;87(2):859-71. 154. Orvedahl A, Alexander D, Tallóczy Z, Sun Q, Wei Y, Zhang W, et al. HSV-1 ICP34. 5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell host & microbe. 2007;1(1):23-35. 155. Kang R, Zeh H, Lotze M, Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death & Differentiation. 2011;18(4):571-80. 156. Leib DA, Alexander DE, Cox D, Yin J, Ferguson TA. Interaction of ICP34. 5 with Beclin 1 modulates herpes simplex virus type 1 pathogenesis through control of CD4+ T-cell responses. Journal of virology. 2009;83(23):12164-71. 157. Mori I, Goshima F, Imai Y, Kohsaka S, Sugiyama T, Yoshida T, et al. Olfactory receptor neurons prevent dissemination of neurovirulent influenza A virus into the brain by undergoing virus-induced apoptosis. Journal of general virology. 2002;83(9):2109-16. 158. Santana S, Recuero M, Bullido MJ, Valdivieso F, Aldudo J. Herpes simplex virus type I induces the acFXPXODWLRQRILQWUDFHOOXODUȕ-amyloid in autophagic compartments and the inhibition of the nonamyloidogenic pathway in human neuroblastoma cells. Neurobiology of aging. 2012;33(2):430. e19-. e33. 159. Friedman LG, Lachenmayer ML, Wang J, He L, Poulose SM, Komatsu M, et al. Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation RIĮ-synuclein and LRRK2 in the brain. Journal of Neuroscience. 2012;32(22):7585-93. 160. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J-i, Tanida I, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441(7095):880-4.

31

161. Marttila RJ, Rinne UK, Tiilikainen A. Virus antibodies in Parkinson's disease: Herpes simplex and measles virus antibodies in serum and CSF and their relation to HLA types. Journal of the neurological sciences. 1982;54(2):227-38. 162. Fazzini E, Fleming J, Fahn S. Cerebrospinal fluid antibodies to coronavirus in patients with Parkinson's disease. Movement Disorders. 1992;7(2):153-8. 163. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiology of disease. 2006;21(2):404-12. 164. Miklossy J, Doudet D, Schwab C, Yu S, McGeer E, McGeer P. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Experimental neurology. 2006;197(2):275-83. 165. Tesoriero C, Codita A, Zhang M-D, Cherninsky A, Karlsson H, Grassi-Zucconi G, et al. H1N1 influenza virus induces narcolepsy-like sleep disruption and targets sleep–wake regulatory neurons in mice. Proceedings of the National Academy of Sciences. 2016;113(3):E368-E77. 166. Arlehamn CSL, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. Immune response LQ3DUNLQVRQ¶VGLVHDVHGULYHQE\+/$GLVSOD\RIĮ-synuclein peptides. Am Assoc Immnol; 2017. 167. Ng YP, Lee SMY, Cheung TKW, Nicholls JM, Peiris JSM, Ip NY. Avian influenza H5N1 virus induces cytopathy and proinflammatory cytokine responses in human astrocytic and neuronal cell lines. Neuroscience. 2010;168(3):613-23. 168. Takahashi M, Yamada T, Nakanishi K, Fujita K, Nakajima K, Nobusawa E, et al. Influenza a virus infection of primary cultured cells from rat fetal brain. Parkinsonism & related disorders. 1997;3(2):97-102. 169. Schöndorf DC, Aureli M, McAllister FE, Hindley CJ, Mayer F, Schmid B, et al. iPSC-derived neurons from GBA1-associated Parkinson's disease patients show autophagic defects and impaired calcium homeostasis. Nature communications. 2014;5:4028. 170. Gannagé M, Dormann D, Albrecht R, Dengjel J, Torossi T, Rämer PC, et al. Matrix protein 2 of influenza A virus blocks autophagosome fusion with lysosomes. Cell host & microbe. 2009;6(4):367-80. 171. Garcia-Reitböck P, Anichtchik O, Bellucci A, Iovino M, Ballini C, Fineberg E, et al. SNARE protein redistribution and synaptic failure in a transgenic mouse model of Parkinson’s disease. Brain. 2010;133(7):2032-44. 172. Nash J, Johnston T, Collingridge G, Garner C, Brotchie J. Subcellular redistribution of the synapse-associated proteins PSD-95 and SAP97 in animal models of Parkinson’s disease and L-DOPAinduced dyskinesia. The FASEB journal. 2005;19(6):583-5. 173. Fatemi SH, Sidwell R, Kist D, Akhter P, Meltzer HY, Bailey K, et al. Differential expression of synaptosome-associated protein 25 kDa [SNAP-25] in hippocampi of neonatal mice following exposure to human influenza virus in utero. Brain Research. 1998;800(1):1-9. 174. Hacohen Y, Deiva K, Pettingill P, Waters P, Siddiqui A, Chretien P, et al. N-methyl-D-aspartate receptor antibodies in post–herpes simplex virus encephalitis neurological relapse. Movement Disorders. 2014;29(1):90-6.

32