Next Article in Journal
Structural and Functional Disparities within the Human Gut Virome in Terms of Genome Topology and Representative Genome Selection
Previous Article in Journal
Systematic Genomic Surveillance of SARS-CoV-2 at Xiamen International Airport and the Port of Xiamen Reveals the Importance of Incoming Travelers in Lineage Diversity
Previous Article in Special Issue
Risk Factors Associated with Opportunistic Infections among People Living with HIV/AIDS and Receiving an Antiretroviral Therapy in Gabon, Central Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them?

by
Maria Antonia De Francesco
Department of Molecular and Translational Medicine, Institute of Microbiology, University of Brescia-ASST Spedali Civili, 25123 Brescia, Italy
Viruses 2024, 16(1), 133; https://doi.org/10.3390/v16010133
Submission received: 9 November 2023 / Revised: 6 December 2023 / Accepted: 15 January 2024 / Published: 17 January 2024
(This article belongs to the Special Issue Opportunistic Viral Infections)

Abstract

:
Alzheimer’s disease and Parkinson’s disease represent the most common forms of cognitive impairment. Multiple sclerosis is a chronic inflammatory disease of the central nervous system responsible for severe disability. An aberrant immune response is the cause of myelin destruction that covers axons in the brain, spinal cord, and optic nerves. Systemic lupus erythematosus is an autoimmune disease characterized by alteration of B cell activation, while Sjögren’s syndrome is a heterogeneous autoimmune disease characterized by altered immune responses. The etiology of all these diseases is very complex, including an interrelationship between genetic factors, principally immune associated genes, and environmental factors such as infectious agents. However, neurodegenerative and autoimmune diseases share proinflammatory signatures and a perturbation of adaptive immunity that might be influenced by herpesviruses. Therefore, they might play a critical role in the disease pathogenesis. The aim of this review was to summarize the principal findings that link herpesviruses to both neurodegenerative and autoimmune diseases; moreover, briefly underlining the potential therapeutic approach of virus vaccination and antivirals.

1. Introduction

The Herpesviridae family, which includes three subfamilies (α-, β- and γ-herpesviruses), is constituted of enveloped double-stranded DNA viruses distributed worldwide and responsible for various clinical pictures [1]. Herpes simplex virus 1, herpes simplex virus 2 (HSV-1, HSV-2), and varicella-zoster virus (VZV), which belong to the α-virus subfamily, are associated with herpes labialis, genital herpes, chickenpox, and herpes zoster, respectively. Cytomegalovirus (CMV), HHV-6, and HHV-7 are β-herpesviruses, which are associated with congenital/neonatal infections and skin rash (roseola), respectively. Epstein–Barr virus (EBV), the etiological agent of infectious mononucleosis, and Kaposi sarcoma-associated herpesvirus (HHV-8) belong to γ-herpesviruses [1,2]. All of them give rise to lifelong infections in the affected subjects persisting in a latent phase with periodic reactivation.
Neurodegenerative and autoimmune diseases share a disruption of immune system functions characterized by inflammatory and autoimmune processes. It has also been found that Alzheimer’s disease and multiple sclerosis exhibit a common pro-inflammatory signature [3]. Furthermore, an aberrant activation of inflammasome proteins, important players in innate immunity and inflammation, has been detected in both neurodegenerative and autoimmune diseases such as Alzheimer’s disease, Parkinson’s disease, systemic lupus erythematosus, and multiple sclerosis [4]. Moreover, an important role has been attributed to the B cells in the pathophysiology of both autoimmune and neurodegenerative diseases [5]. The biological characteristic involved in the establishment of herpesvirus latency supports the hypothesis that they might be implicated in the pathogenesis of some neurodegenerative and autoimmune diseases.
They might be the nexus between immune dysfunctions and genetic factors contributing to the pathology of these diseases.
Therefore, this review summarizes the findings supporting a role played by herpesviruses in these diseases and explores all the mechanisms establishing a link between them.

2. Neurodegenerative Diseases

Neurodegenerative processes are the most frequent cause of cognitive decline.
To date, the etiology of Alzheimer’s disease (AD) and Parkinson’s disease (PD), the most common forms of dementia, is not yet well understood. Different risk factors have been associated with both diseases, such as cigarette smoking, caffeine intake, chemical exposure, and diet [6,7]. Furthermore, bacteria, viruses, and lastly prions have been included as risk factors [8,9,10].
AD and PD affect >50 million and >10 million people worldwide, respectively [11,12,13]. Clinically, affected patients manifest behavioral and cognitive alterations with a change in personality [14,15] and autonomic dysfunction [16]. The pathologic mechanism of both diseases involves the accumulation of misfolded proteins in the brain [17,18].
Alzheimer’s disease is characterized by an extracellular deposition of the amyloid b peptide (Aβ) that exists in different forms after the cleavage of the amyloid protein precursor (APP) [19]. This protein plays an important role in central nervous system (CNS) homeostasis because it is involved in synapsis, calcium homeostasis, metal ion capture, and neurogenesis [20]. A high production of Aβ-peptide leads to the formation of neurofibrillary tangles (NFTs) composed of the hyperphosphorylated Tau protein (p-Tau) [21,22]. This results in the destabilization of associated microtubules, synaptic loss, and neurodegeneration [23]. Besides the formation of amyloid plaques and NFTs, AD pathologic mechanisms include oxidative stress and the alteration of calcium homeostasis [24,25], defective autophagy [26], mitochondrial dysfunction [27], and neuroinflammation [28,29].
When the disease is acquired before the age of 65, it is considered as early onset AD (EOAD) and it is associated with mutations in genes coding APP, presenilin-1 (PSEN1), and presenilin-2 (PSEN2) [30].
Parkinson’s disease is characterized by a loss of dopaminergic neurons, which are localized mostly in the substantia nigra, but also in the dorsal motor nucleus of the vagus and peripheral neurons [31,32], and by the intraneural deposition of aggregates constituted of Lewy bodies, which are formed by α-synuclein and ubiquitin, and by chronic neuroinflammation [33].
An interrelation of genetic, aging, and environmental factors might have a role in their accumulation in neurons [33,34], such as the D620N mutation in a gene and the vacuolar sorting protein 35 (VPS35), involved in PD pathogenesis [35,36,37]. VPS35 belongs to the retromer complex, which acts by forming vesicles that transport cargo molecules from the endosome membrane to the trans-Golgi network (TGN) [38,39]. The D620N mutation was associated with a perturbation of cargo molecule trafficking, a reduction in autophagy, and an accumulation of α-synuclein [40,41,42].

2.1. Herpesviruses’ Role in the Pathogenesis of Neurodegenerative Diseases

The postulated hypotheses consist of both a direct and indirect effect on brain. The first assumes a direct viral infection of the brain through the blood and the blood–brain barrier (BBB). In addition to these infection routes, viral entry may occur via monocyte-macrophage/microglia cells [43], the circumventricular organs (CVOs) [44,45], the olfactory bulb, and the peripheral nerves [46].
After entering the central nervous system, viruses start to replicate and activate an inflammatory response that induces microglia cells to produce inflammatory cytokines [47,48] associated with neuronal death also in the substantia nigra (PD) and to neurogenesis [47]. Then, the production of inflammatory cytokines after viral infection leads to the increased expression of the b- and g-secretase enzymes responsible for the cleavage of APP in Aβ-peptide [49] and to the increase in kinases activity related to Tau hyperphosphorylation [50,51].
The indirect effect is based on the viral induction of peripheral inflammation, a role mostly played in AD. The brain interacts with the periphery through the neuronal and humoral pathways [47]. In PD pathogenesis, autoimmune mechanisms induced by molecular mimicry between some herpesviruses and alpha-synuclein are postulated [52] (Figure 1 and Figure 2).

2.2. Research Data That Establish a Link between AD and Herpesviruses

Numerous studies support the involvement of HSV-1 in AD. For example, it was demonstrated that mice infected with HSV-1 showed an accumulation of Aβ-peptide and cognitive impairment [53,54,55].
Moreover, various research papers have shown the presence of HSV-1 DNA in temporal and frontal brain of AD patients compared to matched controls [56,57,58]. In particular, the virus’ genome is principally localized in amyloid plaques [59].
Furthermore, various studies have supported evidence that HSV-1 might induce both direct and indirect inflammation [47,48,49,50,51].
Then, it was shown that HSV-1 induced the production of both Aβ and tau protein in human neural cells [55]; the infection of neuronal and glial cells has been shown to modulate autophagy, which might lead to the accumulation of both Aβ and tau protein [60,61,62,63]. HSV-1 infection seems to influence the autophagic process through the binding of the HSV1 ICP34.5 (infected cell protein 34.5) to Beclin1, a protein involved in the synthesis and maturation of autophagosomes [64]. Therefore, this interaction blocks the host autophagy response [65]. Moreover, the binding of the HSV-1 US11 protein to the protein kinase PKR prevents its activity [66]. Next, it has been shown that HSV-1 infection increases the production of reactive oxygen species (ROS) [67], which in turn enhances the impairment of the autophagic process determined by the HSV-1 infection [68] creating a vicious circle.
Patients carrying the apolipoprotein E (APOE) E4 allele, a genetic risk factor for AD, had a greater risk of developing AD [69,70]; in addition, HSV-1 DNA has more often been detected in the brains of these subjects [71]. Serological studies have also detected the presence of Aβ and HSV-1 cross-reactive antibodies in patients with AD [72,73], and, furthermore, two prospective studies involving 512 and >8000 subjects, respectively, showed that increased levels of IgM antibodies against HSV-1,2 and symptomatic infection with these viruses were associated with a hazard ratio (HR) of 2.55 and a three-fold increased relative risk, respectively, of developing dementia later in life [74,75]. Furthermore, different studies have supported an important role of EBV and HHV-6 in AD pathogenesis. In fact, EBV DNA has been found in the blood of 45% of patients with AD; then, the virus genome has been found in the brain of 6% of AD patients carrying the pathogenic apolipoprotein E (APOE) E4 allele [76].
Furthermore, EBV induces an inflammatory environment as demonstrated by the evidence that EBV reactivation from the latency phase gives rise to a systemic immune response, with an increase in the inflammation processes associated with cognitive decline in older age [77]; further, that B lymphocytes derived from a patient with AD and infected with EBV produce high levels of TNF-α in vitro, leading to accumulation of Aβ protein and to the hyperphosphorylation of the tau protein [78,79]. Subsequently, CD8 effector memory CD45RA+ (TEMRA) cells have been found to be responsible for proinflammatory and cytotoxic functions in AD patients and to play an important role in disease progression; two antigens of EBV (the Epstein–Barr nuclear antigen 3 and the trans-activator protein BZLF1) have been identified as stimulators of this adaptive immunity in AD patients [80,81].
The EBV protein, BNLF-2a, has been found to be involved in AD progression because it blocks the transporter protein (TAP) associated with antigen processing and decreases the expression of the major histocompatibility complexes (MHC)-I and II, which might determine the increase in neuronal cells and viral polypeptides in the environment [82,83].
Additionally, HHV-6 might promote neuroinflammation. In fact, HHV6-infected primary glial cell cultures produced high levels of proinflammatory cytokines [84]; furthermore, microglial and T cells infected with HHV6 showed an increased expression of Aβ 1-42, tau and ApoE proteins [85,86].
Furthermore, autophagy was reduced following the infection of astrocytoma cells and primary neurons with HHV-6 and this induced an increase in the misfolding of the Aβ protein and hyperphosphorylation of the tau protein [87,88,89].
A study comparing the transcriptome of white blood cells (WBCs) infected with HHV6 to the transcriptome of WBCs derived from AD patients showed the sharing of 95 differentially expressed genes, mostly involved in antigen presentation by MHC class II antigens [90]. The same study showed that the host response against CMV, EBV, and HHV6 involved oxidative stress mechanisms activating sirtuin-1 and the peroxisome proliferator-activated receptor-g coactivator (PGC)-1 pathway [90].
However, the first evidence that HHV6 DNA was present at higher levels in AD brains than in healthy controls [91] was confuted by other successive studies employing the same RNA data set [92,93,94].
On the other hand, few epidemiological and serological studies reported an association between AD and VZV and CMV.
Epidemiological studies involving 80,000 and 3384 patients with herpes zoster showed a significantly increased risk (HR_1.11; 95% CI, 1.04 to 1.17 and HR_2.83; 95% CI, 1.83 to 4.37, respectively) of developing dementia during aging [95,96]; therefore, VZV might contribute to AD pathogenesis through its binding to the insulin-degrading enzyme (IDE), a zinc metalloprotease associated with Aβ degradation, inducing an impairment of its activity [97,98].
Epidemiological studies showed an association between CMV and moderate–severe dementia [99]. Longitudinal studies showed that higher percentages of patients with increased CMV blood markers exhibited an impairment of cognitive functions after a 4–5-year period [76,100]. Finally, it was shown that an increased density of neurofibrillary tangles is associated with the plasmatic levels of CMV IgG antibodies [101].

2.3. Research Data That Establish a Link between PD and Herpesviruses

Different immunological and virological studies correlate HSV1, EBV, HHV-6, and CMV to PD.
A molecular mimicry between HSV-1 and α-synuclein produces autoimmune responses which trigger aggregation of α-synuclein, target neurons of substantia nigra, and induce subsequent neuronal degeneration [102,103]; therefore, the production of TNF-α following HSV-1 infection might contribute to PD pathogenesis. In fact, dopaminergic neurons are very susceptible to TNF-α activity, which induces neuronal death [103].
Molecular mimicry between the C-terminal region of α-synuclein and a repeat region in latent membrane protein 1 (LMP1) encoded by EBV produces oligomerization of α-synuclein [104].
CMV reactivations could expedite the onset of PD inducing a neuroinflammatory environment with the production of inflammatory cytokines by dendritic cells [105]; furthermore, these cells might present antigens derived from dopaminergic neurons, which might be responsible for autoimmune response to neuromelanin [106].
HHV-6 might contribute to PD pathogenesis by direct CNS entry, immunologically mediated mechanisms or inducing parainfectious cytotoxic changes [107]. Finally, a study that analyzed the transcriptome datasets from seropositive or seronegative patients for CMV, EBV, and HHV6 and PD patients showed that patients infected with these herpesviruses shared the differentially expressed genes BCL6, GYG1, RBCK1, TIMP2, and CIRBP with PD patients [90]. In particular, TIMPs (tissue inhibitors of metalloproteinases) are inhibitors of matrix metalloproteinases (MMPs), the altered expression of which is related to neuroinflammation and neuronal death [108].
The principal findings supporting a role for different herpesviruses in the etiology of the described neurodegenerative diseases are summarized in Table 1.

3. Autoimmune Diseases

At present, 80 autoimmune diseases are known to affect about 5% of the general population [109]; because most of them have unknown etiology, different factors such as genetics, environment, age, and viruses are considered to be triggers of aberrant immune responses [110,111]. Among the many known autoimmune diseases, only three (multiple sclerosis, systemic lupus erythematosus, and Sjögren’s syndrome) have been included in this review because of the association found between them and herpesviruses.
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system responsible for severe disability in young adults [112]. The estimated prevalence is 2.8 million people worldwide [113]. A dysregulated immune response is at the root of myelin destruction that covers axons in the brain, spinal cord, and optic nerves leading to demyelination and axonal degeneration [112]. The disease starts with a relapsing–remitting clinical form and becomes progressively chronic in later clinical phases [114]. The etiology of MS is heterogeneous including an interrelationship between genetic factors, principally immune associated genes, and environmental factors such as infectious agents, vitamin D deficiency, sun exposure, obesity, and smoking [115,116,117].
Systemic lupus erythematosus (SLE) is a composite autoimmune disease [118] characterized by the alteration of B cell activation leading to the production of multiple autoantibodies, dysregulation of T cell function with impairment of cell-mediated immunity [119], impaired clearance of nucleic acids, and increased Type 1 IFN response [120]. Characteristic serologic markers of SLE are antinuclear antigens (ANA), anti-double-stranded DNA (dsDNA) and anti-Smith (anti-Sm) autoantibodies [120]. Clinical symptoms range from initial musculoskeletal and mucocutaneous symptoms to the later involvement of any system [121,122]. Genetic and environmental factors such as infectious agents have been recognized to play an important role in its pathogenesis [123,124,125].
Sjögren’s syndrome (SS) is a heterogeneous autoimmune disease [126] characterized by inflammatory infiltration and autoimmune response against exocrine glands [127,128], aberrant polyclonal B cell activation with the production of autoimmune antibodies against ribonucleoproteins [129,130]. Its prevalence is estimated between 0.2 and 0.5% and it mostly affects middle-aged women [131]. At onset, clinical manifestations of primary Sjögren’s syndrome include chronic fatigue and mucosal dryness of the mouth and eyes. The progression of primary Sjögren’s syndrome to systemic disease involves different organs leading to interstitial lung disease, autoimmune cholangitis, hepatitis, vasculitis, and alteration of both the peripheral and central nervous system [132].
Like in other autoimmune diseases, in Sjögren’s syndrome different factors involving genetics, immune responses, and the environment play important roles in disease development [132].

3.1. Herpesviruses’ Role in the Pathogenesis of Autoimmune Diseases

Different mechanisms are postulated as possible effects of viruses in generating a disruption of immune system response, but the principals involve molecular mimicry, bystander activation, and epitope spreading.
Molecular mimicry implies a structural, functional, or genetic similarity between viral proteins and host proteins. This similarity between virus and host can generate immune responses by activating autoreactive T and B cells capable of destroying both self and non-self-antigens [133,134,135].
Bystander activation consists of a production of a pro-inflammatory environment with activation of dendritic cells and autoreactive naïve T cells inducing damage to healthy cells and the release of self-antigens able to activate autoimmune reactions [136].
Epitope spreading relies on the release of self-antigens during the course of a viral infection and on the activation of autoreactive cells, which target self-epitopes [137] (Figure 3 and Figure 4).

3.2. Research Data That Establish a Link between Multiple Sclerosis and Herpesviruses

Several studies have shown a direct and indirect role of HSV-1 in inducing demyelination.
Some studies reported that HSV-1 DNA has been more frequently detected in the peripheral blood and in the brain of MS patients than in the brain of controls [138,139].
Moreover, HSV-1 infection of mice induced both CNS demyelination and inflammation [140]. Interestingly, it was found that these effects were related to the murine strains. Similarly, it was found that children that lack the DRB1*15 allele and had HSV-1 seropositivity were associated with an increased risk of MS [141], underlining that genetic background is important for the development of MS [142].
Furthermore, it has been shown that HSV-1 might induce an increase in brain–blood barrier (BBB) permeability during both acute infection and latency. Microglia infected by HSV-1 release inflammatory cytokines such as TNF-a, IL-1b and IFN-g, able to increase the expression of ICAM1 and NO levels altering endothelial cell function [143]. In addition, chronic immune responses in infected neuronal cells are present during HSV-1 latency in the trigeminal ganglia [144] and they might generate a persistent inflammatory environment leading to the production of autoreactive T cells [145]. Finally, it was found that HSV-1 infection generated mitochondrial dysfunction and the following release of reactive oxygen species (ROS) is associated with neurodegenerative processes [146,147,148]. Therefore, the interference of HSV-1 with the autophagy in the CNS could lead to the accumulation of cellular proteins and of myelin debris [149].
Then, another way by which the virus could have a role in MS pathogenesis is via molecular mimicry. In fact, autoreactive T cells might induce CNS inflammation by binding epitopes in the brain, which share molecular mimicry with viral antigens. To support this hypothesis, different studies have shown the presence of T cells and antibodies able to cross-react with the HSV-1 protein UL15, with the antigen myelin basic protein, and with the HSV-1 glycoprotein B and a brain epitope, respectively [150,151].
Among the herpesviruses, EBV is one that has been investigated the most for its role in triggering MS. Different mechanisms have been considered, such as molecular mimicry, defective EBV immune responses, and EBV-induced inflammation.

3.2.1. Molecular Mimicry

Different EBV antigens share epitopes similar to host proteins, leading to both cross-reactive humoral and cellular immune responses. Different studies have shown that T cells autoreactive to basic myelin proteins derived from MS patients were cross-reactive to several viral peptides such as EBNA-1 [152,153]. Furthermore, structurally related pairs of peptides from EBNA-1 and b-synuclein, a protein present in the brain and involved in MS, have been identified [154]. Then, molecular mimicry between anoctamin 2 (ANO2), a chloride channel in the brain, and EBNA-1 has been associated with an increased risk of MS [155]. Other cross-reactivities have been found between EBNA-1 and α-cristallin B chain (CRYAB) [156] and the glial cell adhesion molecule [157]. Additionally, molecular mimicry has been found between EBV lytic proteins BHRF1 and BPLF1 and the self-peptide derived from the RAS guanyl releasing protein 2 (RASGRP2) [153], expressed both in B cells and in neurons, and responsible for generating autoreactive T cells. These autoreactive CD4+ T cells enter the brain, where they induce an inflammatory response leading to demyelination and axonal damage [158].

3.2.2. Defective EBV Immune Responses

Generally, cytotoxic CD8+T cells control EBV-infected B cells, but this mechanism is defective in MS patients [159]. So, virus-infected B cells become resistant to apoptosis [154] and generate co-stimulator signals that activate autoreactive T cells. Then, B cells can contribute to disease progression by acting directly in the CNS. Even if perivascular MS brain lesions show a low number of B cells, their presence is high in the MS meningeal site and is associated with cortical damage [160]. Antibodies produced by B cells, that have been boosted in the CNS, are oligoclonal IgG against EBV [161]. Higher titers of EBNA-1 antibodies have been found in both the serum and CSF of MS patients and they have been associated with a higher risk of MS [162,163], a risk that is more elevated in subjects with HLA class II DR2b (DRB1*1501 b, DRA1*0101 a) [164]. Furthermore, a prospective study showed a four-fold increase in anti-EBNA 2 antibody titers in MS patients [165]. In large epidemiological studies, EBV seroconversion has been found to precede the outcome of MS clinical signs [166] and another large prospective study found that all of the MS patients included in the analysis were positive for EBV infection [167].
All of these serological studies support evidence for dysregulated EBV immune responses in MS.

3.2.3. EBV Induced Inflammation

EBV remains in a latent state in memory B cells, which exhibit a pro-inflammatory phenotype. Therefore, they produce various inflammatory cytokines both in meninges and in regional lymph nodes, in addition to an inflammation process mediated by the release of exosomes containing EBV-encoded immunomodulatory RNAs (EBER1, miRNA) [168]. These exosomes stimulate cellular functions such as dendritic cells’ antiviral inflammatory activity [168], while miRNAs affect genes associated with MS risk [169]. Additionally, the subsequently created inflammatory environment triggers a BBB permeability that allows anti-EBV immune cells across, the activation of both microglia and astrocytes, and a possible oligodendrocyte dysfunction, which together contribute to neuronal destruction [170,171,172,173,174].
Different studies have demonstrated an association between HHV-6 and MS. Indeed, higher levels of HHV-6 mRNA and viral proteins have been detected in MS plaques rather than in the white matter in the brain of patients compared to controls [175], in particular in oligodendrocytes. Higher levels of the HHV-6 genome have been found mostly in acute rather than chronic lesions in MS patients who did not receive immunomodulatory therapies [176]. A strong association with an OR of 6.7 and a 95% CI of 4.8 to 8.6 (p < 0.00001) [177] has been found between HHV-6 and MS in a meta-analysis of 39 studies. Furthermore, an association has been found between HHV6 reactivation and disease activity in relapsing–remitting MS (RRMS) and secondary progressive MS (SPMS) [178]. Then, different studies have shown that serological response against HHV-6 is higher in MS patients than in controls [179,180]. In addition, a Swedish study analyzing 8742 MS subjects and 7215 matched controls showed an increase in IgG response against the IE1A peptide of HHV-6A [181]. Then, an increase in HHV-6-specific antibodies was associated with clinical relapses [182], while raised anti-HHV-6 early antigen (p41/38) IgM antibodies have been shown in patients with RRMS as compared to patients affected by other MS subtypes or other neurological/autoimmune diseases [183]. Oligoclonal bands against HHV-6 have been detected in the CSF of MS patients underlining the production of HHV-6-specific IgG [184]; furthermore, intrathecal HHV-6 antibodies have been more frequently identified in RRMS and chronic progressive MS than in other neurological diseases [185].
Additionally, a sequence similarity has been identified between the U24 protein of HHV-6 (residues 4–10) and MBP (residues 96–102) [186]. Furthermore, the frequency of circulating T cells recognizing both HHV-6 U24 and MBP is higher in MS patients than in controls [186,187]. Furthermore, HHV-6-infected T cells induce the increased production of different inflammatory cytokines, which in turn correlates with the severity and progression of MS [188]. In addition, HHV-6 infection of oligodendrocyte precursor cells affects the remyelination process in MS patients [189], impairing the differentiation and migration process of these cells [190]. Furthermore, for HHV-6, a role as trans-activator of latent viruses such as EBV or endogenous retroviruses [191,192], which also play an important role in MS pathogenesis, has been proposed. Lastly, increased levels of a soluble CD46 receptor, a complement system regulator, and the human receptor for HHV-6, have been found in the CSF and blood of MS patients [193], leading to the hypothesis that HHV-6 infection, through the engagement of CD46, might induce exaggerated activation of the complement system which could contribute to MS pathogenesis [194]. Another supposition was that during its replication, HHV-6 might incorporate different host antigens, therefore provoking autoimmune responses [194].
The role of HCMV in MS is quite controversial, because several studies proposed both a deleterious and protective HCMV role.
In fact, some studies have shown higher loads of HCMV genome in MS patients than healthy controls [195,196]; furthermore, HCMV has been found in the plaques and the CSF of MS patients [197].
Then, it has been shown that reactivation of HCMV was associated with a deterioration of the course of MS in some subjects [198].
Moreover, a similarity has been shown between the HCMV antigen (UL86981-1003) and myelin oligodendrocyte glycoprotein (MOG) (residues 34–56), supporting the hypothesis about the generation of autoreactive T cells [199].
By contrast, other studies support a beneficial role of HCMV in MS. In fact, a recent study has shown a lower prevalence of IgM against HCMV in MS patients than in controls. Furthermore, the patients had relapsing MS, underlining that HCMV has a part in reducing disease severity [200].
Furthermore, it has been shown that HCMV infection is related to a decreased production of inflammatory cytokines in progressive MS, confirming a protective role of HCMV in MS [201].
Few epidemiological and serological studies shown an association between VZV and MS.
A study conducted in Taiwan showed that patients with herpes zoster had a risk for MS of 3.63-fold higher than controls [202]; furthermore, MS patients showed specific antibodies against VZV [203]. In addition, a meta-analysis involving 2266 MS patients and 1818 healthy subjects showed that VZV seropositivity was higher in MS patients than in controls (OR = 4.47, p < 0.001) [204]. Moreover, high loads of VZV DNA have been detected in the CSF and PBMCs of MS patients [205]. Finally, a recent Mendelian randomization analysis was used to study the association between VZV and MS by using summary statistics from genome-wide association studies (GWAS). The results supported a significant association between genetically predicted chickenpox and the risk of MS with an OR of 35.27 (CI = 22.97–54.17, p = 1.46 × 10−59) [206].

3.3. Research Data That Establish a Link between Systemic Lupus Erythematosus and Herpesviruses

Serological studies have found a strong association between SLE and EBV and, to a lesser extent, between SLE and CMV. In particular, SLE patients presented an increased reactivation of EBV, as supported by the detection in their blood of high loads of EBV DNA [207] and a significantly higher OR for the presence of IgG against VCA and EA in SLE patients than in controls (OR = 2.06, 95% CI= 1.30–3.26, p = 0.002 and OR = 7.70, 95% CI= 4.64–12.76, p < 0.001, respectively) [208].
Different studies have detected higher anti-EBV antibody titers in SLE patients compared to healthy subjects [209,210,211]; however, a significant correlation between EBV serology and single nucleotide polymorphisms (SNPs) in genes related to SLE has been found [212], underlining the important role played by genetic factors in SLE pathogenesis. In addition, different cross-reactivities have been detected between EBV antigens and SLE autoantigens, leading to the production of autoantibodies such as anti-EBNA 1 antibodies that cross-react with autoantigens of SLE (SmB, SmD and Ro) [213]. Then, anti-EBNA1 antibodies have been found to cross-react with dsDNA in mice [214] and with the C1q complement component [211]. In fact, SLE patients who had a seropositivity against EBNA348, a peptide of EBNA1, showed higher titers of anti-C1q [215]. A cross-reactivity has been further found between EBNA2 and the antigenic terminal domain of the SLE antigen SmD1 [216]. In addition, it has been shown that EP4, a peptide from EBV EA, induced an increase in anti-SmD and anti-Ro and correlated with the SLE disease activity index (SLEDAI) [217]. Finally, EBV IL10, a viral EBV gene expressed during its lytic cycle, is homologous of IL10 and, because it engages the same receptor, it inhibits all the immune responses regulated by IL10 [218].
vIL-10 has been detected at higher concentrations in the plasma of SLE patients than in controls [219] and it has been correlated to an increased production of inflammatory cytokine leading to a defective clearance of infected cells and to increased antigen presentation that may generate autoimmune responses [218,220].
On the other hand, for CMV, a similarity has been detected between an epitope of phosphoprotein 65 and TATA-box-binding protein associated factor 9 (TAF9134-144) leading to the production of antibodies cross-reacting with both proteins, ANA and anti-ds DNA in mice [221]. Then, an increase in antibody titers against TAF9 has been shown in SLE patients [222]. Furthermore, a monoclonal antibody against the CMV UL44 immunoprecipitated both viral UL44 and some SLE antigens such as nucleoli, dsDNA, and ku70 [223]; and, SLE patients have displayed an increased level of CMV US31 leading to macrophage differentiation and inflammation activation [224].
A cross-reactivity between CMV-specific T cells and the La protein has been detected at the beginning of SLE in childhood [225]. Moreover, CMV antigens may induce, in the PBMCs of SLE patients, an increase in IFN-gamma and IL-4 cytokines with a greater expansion of memory T cells potentially involved in autoimmune events [226].
There is no evidence that HHV-6 has a role in the occurrence of SLE. The only hypothesized role indicated for this herpesvirus was the establishment of a coinfection with EBV determining a synergistic effect that might generate an activation of polyclonal B lymphocytes and a perturbation of immune functions [208].

3.4. Research Data That Establish a Link between Sjögren’s Syndrome and Herpesviruses

Different studies have shown an association between SS and EBV, VZV, and HHV-6.
In fact, some studies have shown a correlation between EBV reactivation in SS patients and B cell polyclonal activation, which contributes to autoantibody production [227,228]. Then, it has been shown that EBV infection plays a role in B and T cell differentiation in an effector phenotype related to SS. Lymphocyte activation at ectopic germinal center, induced by follicular T helper and cytotoxic cells, and probably stimulated by EBV, might give rise to autoimmune epithelitis [229]. High levels of EBV DNA have been found in the salivary gland tissue and PBMCs of SS patients [230]; further, high levels of anti-EA have been found in SS patients [231]. By analyzing the gene expression of PBMCs derived from both SS patients and healthy controls, it has been shown that several differentially expressed genes were shared, including abnormal signaling pathways of T and B cell receptors and virus-correlated pathways [232]. Finally, similarities between viral EBNA 2 and the Ro-60 antigen and between EBER-1 and the La antigen have been detected [233].
A recent nation-wide, population-based, case–control study including 5751 SS patients and 28,755 matched controls showed a significant correlation between herpes zoster exposure and SS risk, which was greater when the interval between the last visit for herpes zoster infection and the index date was <3 months (OR adjusted for comorbidities = 3.13, 95%CI = 2.20–4.45) [234].
Higher titers of anti-HHV-6 have been found in a group of SS patients with rheumatoid arthritis than in controls [235]; however, even if HHV-6 might contribute to SS development, the contrary is also true. Namely, the autoimmune disease might lead to the reactivation of HHV-6 [236].
To date, it is not known if CMV might contribute to SS development. The virus reactivation observed in SS patients might be due to the use of pronounced immunosuppression [237].
The principal findings supporting a role for different herpesviruses in the etiology of the described autoimmune diseases are summarized in Table 2.

4. Relationship between Herpesviruses, Neurological Disorders and Autoimmune Diseases

From the high number of reported studies, how can we relate the same herpesviruses to pathologies as distinct as neurodegenerative and autoimmune diseases? All of them share the biological characteristic to persist in the host in a latency state interspaced with periodic reactivation.
This periodic reactivation might lead to chronic pathological conditions. Is the virus’ reactivation a consequence of the immune dysregulation linked to these diseases or is the virus infection a trigger of neurodegenerative and autoimmune diseases? The starting point is quite controversial and probably only longitudinal studies might clarify this issue. However, a recent study found 45 significant associations between the exposure to a viral infection and the risk of contracting a neurodegenerative disease later in life [238]. Among the viral infections, viral encephalitis and varicella zoster are included, supporting the results of previous studies, which found associations between HSV encephalitis and AD [239,240], HSV and MS [241], EBV and dementia [242], and EBV and MS [166].
Is there a common pathogenesis mechanism involved in both neurodegenerative and autoimmune diseases induced by herpesviruses?
Molecular mimicry was one of the mechanisms postulated for PD pathogenesis involving HSV1 and EBV [102,103,104]; the same mechanism was invoked for MS involving HSV1 [150,151], EBV [154,155,156,157,158] and HCMV [199], and also for SS involving EBV [233]. Molecular mimicry was finally detected between EBV and CMV proteins and self-antigens in SLE [213,214,215,221,225].
Another mechanism underlying pathogenesis in neurodegenerative and autoimmune diseases is herpesvirus-induced peripheral and central inflammation. This was reported for HSV-1 [47,48,49,50,51], EBV [77,78,79], and HHV-6 [84] in AD; for HSV1 [103], CMV [105], and HHV6 [106] in PD; for HSV1 [140,143], and EBV [168,169,170,171,172,173,174] in MS, and for EBV [218,220] and CMV [226] in SLE.
What is the important trigger that directs the development of a neurodegenerative or an autoimmune disease from the same herpesvirus?
It is likely that host-specific factors play a pivotal role in the regulation of virus virulence. Given the ubiquity of herpesviruses compared to the lower frequency of these diseases, it is obvious that not all herpesvirus-infected subjects develop neurodegenerative or autoimmune diseases. There may be both virus- and host-related factors that have an influence on the host immune response and/or on the susceptibility to virus-induced pathological effects. A recognized important host-related factor is genetic background, supported by evidence of specific host genetic loci that increase the risk of the development of neurodegenerative or autoimmune diseases, such as the presence of APOE4, related to increased AD susceptibility [243]. Then, a recent study investigating the shared genetic susceptibility between AD and MS detected 16 shared loci, with 8 of them able to have concordant effects both on AD and MS [244]. These genes were involved in molecular signaling pathways related to inflammation and the structural organization of neurons. Therefore, such genetic factors could contribute in a determining way to what fate will befall infected subjects, in terms of developing one or the other pathologic condition.

5. Conclusions

The etiology of both neurodegenerative and autoimmune diseases is complex and heterogenous.
However, even if it is not possible to discriminate whether herpesviruses are the cause or the effect of these diseases, the overlapping of pathogenetic pathways between virus infections and neurodegenerative and autoimmune processes constitutes a motivation to use antiviral therapies as treatment approach for these diseases. To date, in spite of promising results [244,245], few data are available on the efficacy of antiviral therapies.
Then, vaccination has also been proposed to mitigate the possible risk of developing neurodegenerative diseases by using available or future vaccines against herpesviruses. An efficacious viral vaccine might reduce virus diffusion and limit altered immune responses, that have in turn a consequent role on disease pathogenesis because both inflammation and dysregulated immune responses are known to contribute greatly to these pathologies. Some results support this hypothesis. In fact, varicella-zoster vaccination has been associated with a reduced risk of dementia, AD, and PD in both the United States and Wales [246,247,248,249,250].
Future research is needed to fully understand the correlation between herpesviruses and host-specific factors and their impact on the disease development, and might lead to the identification of novel targets to prevent or slow the progression of both neurodegenerative and autoimmune diseases.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Lan, K.; Luo, M.H. Herpesviruses: Epidemiology, pathogenesis, and interventions. Virol. Sin. 2017, 32, 347–348. [Google Scholar] [CrossRef] [PubMed]
  2. Nath, A. Herpes viruses, Alzheimer’s disease, and related dementias: Unifying or confusing hypothesis? Neurotherapeutics 2019, 16, 180–181. [Google Scholar] [CrossRef] [PubMed]
  3. Rossi, B.; Santos-Lima, B.; Terrabuio, E.; Zenaro, E.; Constantin, G. Common Peripheral Immunity Mechanisms in Multiple Sclerosis and Alzheimer’s Disease. Front. Immunol. 2021, 12, 639369. [Google Scholar] [CrossRef]
  4. Bulté, D.; Rigamonti, C.; Romano, A.; Mortellaro, A. Inflammasomes: Mechanisms of Action and Involvement in Human Diseases. Cells 2023, 12, 1766. [Google Scholar] [CrossRef] [PubMed]
  5. Sabatino, J.J.; Pröbstel, A.K.; Zamvil, S.S. B cells in autoimmune and neurodegenerative central nervous system disease. Nat. Rev. Neurosci. 2019, 20, 728–745. [Google Scholar] [CrossRef]
  6. Chen, H.; Ritz, B. The search for environmental causes of Parkinson’s disease: Moving forward. J. Parkinons Dis. 2018, 8, S9–S17. [Google Scholar] [CrossRef]
  7. Marras, C.; Canning, C.G.; Goldman, S.M. Environment, lifestyle, and Parkinson’s disease: Implications for prevention in the next decade. Mov. Disord. 2019, 34, 801–811. [Google Scholar] [CrossRef]
  8. Patrick, K.L.; Bell, S.L.; Weindel, C.G.; Watson, R.O. Exploring the “Multiple-hit hypothesis” of neurodegenerative disease: Bacterial infection comes up to bat. Front. Cell. Infect. Microbiol. 2019, 9, 138. [Google Scholar] [CrossRef]
  9. Wouk, J.; Zendrini Rechenchonski, D.; Cerqueira Dias Rodrigues, B.; Vicente Ribelato, E.; Faccin-Galhardi, L.C. Viral infections and their relationship to neurological disorders. Arch. Virol. 2021, 166, 733–753. [Google Scholar] [CrossRef]
  10. Duyckaerts, C.; Clavaguera, F.; Potier, M.C. The prion-like propagation hypothesis in Alzheimer’s and Parkinson’s disease. Curr. Opin. Neurol. 2019, 32, 266–271. [Google Scholar] [CrossRef]
  11. Shi, M.; Li, C.; Tain, X.; Chu, F.; Zhu, J. Can control infections slow down the progression of Alzheimer’s disease? Talking about the role of infections in Alzheimer’s disease. Front. Aging Neurosci. 2021, 13, 434. [Google Scholar] [CrossRef]
  12. Patterson, C. World Alzheimer Report 2018; Alzheimer’s Disease International: London, UK, 2018. [Google Scholar]
  13. Parkinsons’s Foundation. Available online: https://www.parkinson.org/understandingParkinsons/Statistics (accessed on 2 August 2020).
  14. de Lau, L.M.; Breteler, M.M. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006, 5, 525–535. [Google Scholar] [CrossRef]
  15. Blennow, K.; Zetterberg, H. Biomarkers for Alzheimer’s disease: Current status and prospects for the future. J. Intern. Med. 2018, 284, 643–663. [Google Scholar] [CrossRef] [PubMed]
  16. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef]
  17. Ross, C.A.; Poirier, M.A. Protein aggregation and neurodegenerative disease. Nat. Med. 2004, 10, S10–S17. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, X.; Huang, T.; Bu, G.; Xu, H. Dysregulation of protein trafficking in neurodegeneration. Mol. Neurodegener. 2014, 9, 31. [Google Scholar] [CrossRef] [PubMed]
  19. Cipriani, G.; Dolciotti, C.; Picchi, L.; Bonuccelli, U. Alzheimer and his disease: A brief history. Neurol. Sci. 2011, 32, 275–279. [Google Scholar] [CrossRef]
  20. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
  21. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef]
  22. Lee, V.M.; Goedert, M.; Trojanowski, J.Q. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 2001, 24, 1121–1159. [Google Scholar] [CrossRef]
  23. Kolarova, M.; Garcia-Sierra, F.; Bartos, A.; Ricny, J.; Ripova, D. Structure and pathology of tau protein in Alzheimer disease. Int. J. Alzheimers Dis. 2012, 2012, 731526. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, J.M.; Sun, C. Calcium and neurogenesis in Alzheimer’s disease. Front. Neurosci. 2010, 4, 194. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.G.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta 2014, 8, 1240–1247. [Google Scholar] [CrossRef] [PubMed]
  26. Boland, B.; Kumar, A.; Lee, S.; Platt, F.M.; Wegiel, J.; Yu, W.H.; Nixon, R.A. Autophagy induction and autophagosome clearance in neurons: Relationship to autophagic pathology in Alzheimer’s disease. J. Neurosci. 2008, 28, 6926–6937. [Google Scholar] [CrossRef] [PubMed]
  27. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef]
  28. Welikovitch, L.A.; Do Carmo, S.; Magloczky, Z.; Malcolm, J.C.; Loke, J.; Klein, W.L.; Freund, T.; Cuello, A.C. Early intraneuronal amyloid triggers neuron derived inflammatory signaling in APP transgenic rats and human brain. Proc. Natl. Acad. Sci. USA 2020, 117, 6844–6854. [Google Scholar] [CrossRef]
  29. Madore, C.; Yin, Z.; Leibowitz, J.; Butovsky, O. Microglia, lifestyle stress, and neurodegeneration. Immunity 2020, 52, 222–240. [Google Scholar] [CrossRef]
  30. Laval, K.; Enquist, L.W. The potential role of herpes simplex virus type 1 and neuroinflammation in the pathogenesis of Alzheimer’s disease. Front. Neurol. 2021, 12, 458. [Google Scholar] [CrossRef]
  31. Grosch, J.; Winkler, J.; Kohl, Z. Early degeneration of both dopaminergic and serotnergic axons—A common mechanism in Parkinson’s disease. Front. Cell Neurosci. 2016, 10, 293. [Google Scholar] [CrossRef]
  32. Braak, H.; Del Tredici, K.; Rüb, U.; de Vos, R.A.I.; Steur, E.N.H.J.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  33. Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, Pathophysiology. Clin. Geriatr. Med. 2020, 36, 1–12. [Google Scholar] [CrossRef] [PubMed]
  34. Pang, S.Y.; Ho, P.W.; Liu, H.F.; Leung, C.T.; Li, L.; Chang, E.E.S.; Ramsden, D.B.; Ho, S.L. The Interplay of Aging, Genetics and Environmental Factors in the Pathogenesis of Parkinson’s Disease. Transl. Neurodegener. 2019, 8, 23. [Google Scholar] [CrossRef] [PubMed]
  35. Small, S.A.; Kent, K.; Pierce, A.; Leung, C.; Kang, M.S.; Okada, H.; Honig, L.; Vonsattel, J.P.; Kim, T.W. Model-Guided MicroarrayImplicates the Retromer Complex in Alzheimer’s Disease. Ann. Neurol. 2005, 58, 909–919. [Google Scholar] [CrossRef] [PubMed]
  36. Wen, L.; Tang, F.L.; Hong, Y.; Luo, S.W.; Wang, C.L.; He, W.; Shen, C.; Jung, J.U.; Xiong, F.; Lee, D.H.; et al. Vps35 HaploinsufficiencyIncreases Alzheimer’s Disease Neuropathology. J. Cell Biol. 2011, 195, 765–779. [Google Scholar] [CrossRef]
  37. Li, J.G.; Chiu, J.; Pratico, D. Full Recovery of the Alzheimer’s Disease Phenotype by Gain of Function of Vacuolar Protein Sorting 35. Mol. Psychiatry 2020, 25, 2630–2640. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, K.E.; Healy, M.D.; Collins, B.M. Towards a Molecular Understanding of Endosomal Trafficking by Retromer and Retriever. Traffic 2019, 20, 465–478. [Google Scholar] [CrossRef] [PubMed]
  39. Cullen, P.J.; Steinberg, F. To Degrade or Not to Degrade: Mechanisms and Significance of Endocytic Recycling. Nat. Rev. Mol. Cell Biol. 2018, 19, 679–696. [Google Scholar] [CrossRef] [PubMed]
  40. Temkin, P.; Lauffer, B.; Jager, S.; Cimermancic, P.; Krogan, N.J.; von Zastrow, M. Snx27 Mediates Retromer Tubule Entry and Endosome-to-Plasma Membrane Trafficking of Signalling Receptors. Nat. Cell Biol. 2011, 13, 715–721. [Google Scholar] [CrossRef]
  41. Tang, F.L.; Erion, J.R.; Tian, Y.; Liu, W.; Yin, D.M.; Ye, J.; Tang, B.; Mei, L.; Xiong, W.C. for Endosome-to-Golgi Retrieval of Lamp2a, a Receptor of Chaperone-Mediated Autophagy That Is Critical for Alpha-Synuclein Degradation and Prevention of Pathogenesis of Parkinson’s Disease. J. Neurosci. 2015, 35, 10613–10628. [Google Scholar] [CrossRef]
  42. Cui, Y.; Yang, Z.; Flores-Rodriguez, N.; Follett, J.; Ariotti, N.; Wall, A.A.; Parton, R.G.; Teasdale, R.D. Formation of Retromer Transport Carriers Is Disrupted by the Parkinson Disease-Linked Vps35 D620n Variant. Traffic 2021, 22, 123–136. [Google Scholar] [CrossRef]
  43. Dando, S.J.; Mackay-Sim, A.; Norton, R.; Currie, B.J.; St John, J.A.; Ekberg, J.A.K.; Batzloff, M.; Ulett, G.C.; Beacham, I.R. Pathogens penetrating the central nervous system: Infection pathways and the cellular and molecular mechanisms of invasion. Clin. Microbiol. Rev. 2014, 27, 691–726. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, K.S. Mechanisms of microbial traversal of the blood-brain barrier. Nat. Rev. Microbiol. 2008, 6, 625–634. [Google Scholar] [CrossRef]
  45. D’ Mello, C.; Swain, M.G. Immune-to-brain communication pathways in inflammation-associated sickness and depression. In Current Topics in Behavioral Neurosciences; Dantzer, R., Capuron, L., Eds.; Springer: Cham, Switzerland, 2017; pp. 73–94. [Google Scholar]
  46. McGavern, D.B.; Kang, S.S. Illuminating viral infections in the nervous system. Nat. Rev. Immunol. 2011, 11, 318–329. [Google Scholar] [CrossRef] [PubMed]
  47. Xie, J.; Van Hoecke, L.; Vandenbroucke, R. The impact of systemic inflammation on Alzheimer’s disease pathology. Front. Immunol. 2021, 12, 79867. [Google Scholar] [CrossRef] [PubMed]
  48. Kirkley, K.S.; Popichak, K.A.; Afzali, M.F.; Legare, M.E.; Tjalkens, R.B. Microglia amplify inflammatory activation of astrocytes in manganese neurotoxicity. J. Neuroinflamm. 2017, 14, 99. [Google Scholar] [CrossRef]
  49. Kaur, D.; Sharma, V.; Deshmukh, R. Activation of microglia and astrocytes: A roadway to neuroinflammation and Alzheimer’s disease. Inflammopharmacology 2019, 27, 663–677. [Google Scholar] [CrossRef]
  50. Kitazawa, M.; Cheng, D.; Tsukamoto, M.R.; Koike, M.A.; Wes, P.D.; Vasilevko, V.; Cribbs, D.H.; LaFerla, F.M. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal beta-catenin pathway function in an Alzheimer’s disease model. J. Immunol. 2011, 187, 6539. [Google Scholar] [CrossRef]
  51. Quintanilla, R.A.; Orellana, D.I.; González-Billault, C.; Maccioni, R.B. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp. Cell Res. 2004, 295, 245–257. [Google Scholar] [CrossRef]
  52. Woulfe, J.M.; Gray, M.T.; Gray, D.A.; Munoz, D.G.; Middeldorp, J.M. Hypothesis: A role for EBV-induced molecular mimicry in Parkinson’s disease. Parkinsonism Relat. Disord. 2014, 20, 685–694. [Google Scholar] [CrossRef]
  53. De Chiara, G.; Piacentini, R.; Fabiani, M.; Mastrodonato, A.; Marcocci, M.E.; Limingi, D.; Napoletani, G.; Protto, V.; Coluccio, P.; Celestino, I.; et al. Recurrent herpes simplex virus-1 infection induces hallmarks of neurodegeneration and cognitive deficits in mice. PLoS Pathog. 2019, 15, e1007617. [Google Scholar] [CrossRef]
  54. Ezzat, K.; Pernemalm, M.; Palsson, S.; Roberts, T.C.; Jarver, P.; Dondalska, A.; Bestas, B.; Sobkowiak, M.J.; Levanen, B.; Skold, M.; et al. The viral protein corona directs viral pathogenesis and amyloid aggregation. Nat. Commun. 2019, 10, 2331. [Google Scholar] [CrossRef] [PubMed]
  55. Wozniak, M.A.; Frost, A.L.; Itzaki, R.F. Alzheimer’s disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J. Alzheimers Dis. 2009, 16, 341–350. [Google Scholar] [CrossRef]
  56. Itzhaki, R.F.; Golde, T.E.; Heneka, M.T.; Readhead, B. Do infections have a role in the pathogenesis of Alzheimer disease? Nat. Rev. Neurol. 2020, 1, 193–197. [Google Scholar] [CrossRef]
  57. Jamieson, G.A.; Maitland, N.J.; Craske, J.; Wilcock, G.K.; Itzhaki, R.F. Detection of herpes simplex virus type 1 DNA sequences in normal and Alzheimer’s disease brain using polymerase chain reaction. Biochem. Soc. Trans. 1991, 19, 122S. [Google Scholar] [CrossRef]
  58. Itzhaki, R.F.; Lin, W.R.; Shang, D.; Wilcock, G.K.; Faragher, B.; Jamieson, G.A. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet 1997, 349, 241–244. [Google Scholar] [CrossRef] [PubMed]
  59. Wozniak, M.A.; Mee, A.P.; Itzhaki, R.F. Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaques. J. Pathol. 2009, 217, 131–138. [Google Scholar] [CrossRef] [PubMed]
  60. Zare-Shahabadi, A.; Masliah, E.; Johnson, G.V.W.; Rezaei, N. Autophagy in Alzheimer’s disease. Rev. Neurosci. 2015, 26, 385–395. [Google Scholar] [CrossRef]
  61. Wang, Y.; Mandelkow, E. Degradation of tau protein by autophagy and proteasomal pathways. Biochem. Soc. Trans. 2012, 40, 644–652. [Google Scholar] [CrossRef]
  62. Kerr, J.S.; Adriaanse, B.A.; Greig, N.H.; Mattson, M.P.; Cader, M.Z.; Bohr, V.A.; Fang, E.F. Mitophagy and Alzheimer’s disease: Cellular and molecular mechanisms. Trends Neurosci. 2017, 40, 151–166. [Google Scholar] [CrossRef]
  63. Uddin, M.S.; Stachowiak, A.; Mamun, A.A.; Tzvetkov, N.T.; Takeda, S.; Atanasov, A.G.; Bergantin, L.B.; Abdel-Daim, M.M.; Stankiewicz, A.M. Autophagy and Alzheimer’s disease: From molecular mechanisms to therapeutic implications. Front. Aging Neurosci. 2018, 10, 04. [Google Scholar] [CrossRef]
  64. Hill, S.M.; Wrobel, L.; Rubinsztein, D.C. Post-translational modifications of Beclin 1 provide multiple strategies for autophagy regulation. Cell Death Differ. 2019, 26, 617–629. [Google Scholar] [CrossRef]
  65. Orvedahl, A.; Alexander, D.; Talloczy, Z.; Sun, Q.; Wei, Y.; Zhang, W.; Burns, D.; Leib, D.A.; Levine, B. HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe 2007, 1, 23–35. [Google Scholar] [CrossRef] [PubMed]
  66. Lussignol, M.; Queval, C.; Bernet-Camard, M.F.; Cotte-Laffitte, J.; Beau, I.; Codogno, P.; Esclatine, A. The herpes simplex virus1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. J. Virol. 2013, 87, 859–871. [Google Scholar] [CrossRef] [PubMed]
  67. Kavouras, J.H.; Prandovszky, E.; Valyi-Nagy, K.; Kovacs, S.K.; Tiwari, V.; Kovacs, M.; Shukla, D.; Valyi-Nagy, T. Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation by-products in mouse P19N neural cell cultures. J. Neurovirol. 2007, 13, 416–425. [Google Scholar] [CrossRef]
  68. Santana, S.; Sastre, I.; Recuero, M.; Bullido, M.J.; Aldudo, J. Oxidative stress enhances neurodegeneration markers induced by herpes simplex virus type 1 infection in human neuroblastoma cells. PLoS ONE 2013, 8, e75842. [Google Scholar] [CrossRef] [PubMed]
  69. Mahley, R.W.; Rall, S.C., Jr. Apolipoprotein E: Far more than a lipid transport protein. Annu. Rev. Genom. Hum. Genet. 2000, 1, 507–537. [Google Scholar] [CrossRef]
  70. Kuhlmann, I.; Minihane, A.M.; Huebbe, P.; Nebel, A.; Rimbach, G. Apolipoprotein E genotype and hepatitis C, HIV and herpes simplex disease risk: A literature review. Lipids Health Dis. 2010, 9, 8. [Google Scholar] [CrossRef] [PubMed]
  71. Lovheim, H.; Norman, T.; Weidung, B.; Olsson, J.; Josefsson, M.; Adolfsson, R.; Nyberg, L.; Elgh, F. Herpes simplex virus, APOE epsilon4, and cognitive decline in old age: Results from the Betula Cohort Study. J. Alzheimers Dis. 2019, 67, 211–220. [Google Scholar] [CrossRef] [PubMed]
  72. Cheng, S.B.; Ferland, P.; Webster, P.; Bearer, E.L. Herpes simplex virus dances with amyloid precursor protein while exiting the cell. PLoS ONE 2011, 6, e17966. [Google Scholar] [CrossRef]
  73. Vojdani, A.; Vojdani, E. Amyloid-Beta1-42 cross-reactive antibody prevalent in human sera may contribute to intraneuronal deposition of A-Beta-P-42. Int. J. Alzheimers Dis. 2018, 2018, 1672568. [Google Scholar]
  74. Letenneur, L.; Peres, K.; Fleury, H.; Garrigue, I.; Barberger-Gateau, P.; Helmer, C.; Orgogozo, J.M.; Gauthier, S.; Dartigues, J.F. Seropositivity to herpes simplex virus antibodies and risk of Alzheimer’s disease: A population-based cohort study. PLoS ONE 2008, 3, e3637. [Google Scholar] [CrossRef]
  75. Tzeng, N.S.; Chung, C.H.; Lin, F.H.; Chiang, C.P.; Yeh, C.B.; Huang, S.Y.; Lu, R.B.; Chang, H.A.; Kao, Y.C.; Yeh, H.W.; et al. Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections—A nationwide, population-based cohort study in Taiwan. Neurotherapeutics 2018, 15, 417–429. [Google Scholar] [CrossRef]
  76. Carbone, I.; Lazzarotto, T.; Ianni, M.; Porcellini, E.; Forti, P.; Masliah, E.; Gabrielli, L.; Licastro, F. Herpes virus in Alzheimer’s disease: Relation to progression of the disease. Neurobiol. Aging 2014, 35, 122–129. [Google Scholar] [CrossRef]
  77. Shim, S.M.; Cheon, H.S.; Jo, C.; Koh, Y.H.; Song, J.; Jeon, J.P. Elevated Epstein-Barr Virus Antibody Level is Associated with Cognitive Decline in the Korean Elderly. J. Alzheimers Dis. 2017, 55, 293–301. [Google Scholar] [CrossRef]
  78. Dezfulian, M. A new Alzheimer’s disease cell model using B cells to induce beta amyloid plaque formation and increase TNF alpha expression. Int. Immunopharmacol. 2018, 59, 106–112. [Google Scholar] [CrossRef] [PubMed]
  79. Ounanian, A.; Guilbert, B.; Seigneurin, J.M. Characteristics of Epstein-Barr virus transformed B cell lines from patients with Alzheimer’s disease and age-matched controls. Mech. Ageing Dev. 1992, 63, 105–116. [Google Scholar] [CrossRef] [PubMed]
  80. Gate, D.; Saligrama, N.; Leventhal, O.; Yang, A.C.; Unger, M.S.; Middeldorp, J.; Chen, K.; Lehallier, B.; Channappa, D.; De Los Santos, M.B.; et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 2020, 577, 399–404. [Google Scholar] [CrossRef]
  81. Hur, J.Y.; Frost, G.R.; Wu, X.; Crump, C.; Pan, S.J.; Wong, E.; Barros, M.; Li, T.; Nie, P.; Zhai, Y.; et al. The innate immunity protein IFITM3 modulates gamma-secretase in Alzheimer’s disease. Nature 2020, 586, 735–740. [Google Scholar] [CrossRef]
  82. Tiwari, D.; Singh, V.K.; Baral, B.; Pathak, D.K.; Jayabalan, J.; Kumar, R.; Tapryal, S.; Jha, H.C. Indication of Neurodegenerative Cascade Initiation by Amyloid-like Aggregate-Forming EBV Proteins and Peptide in Alzheimer’s Disease. ACS Chem. Neurosci. 2021, 12, 3957–3967. [Google Scholar] [CrossRef] [PubMed]
  83. Wycisk, A.I.; Lin, J.; Loch, S.; Hobohm, K.; Funke, J.; Wieneke, R.; Koch, J.; Skach, W.R.; Mayerhofer, P.U.; Tampe, R. Epstein-Barr viral BNLF2a protein hijacks the tail-anchored protein insertion machinery to block antigen processing by the transport complex TAP. J. Biol. Chem. 2011, 286, 41402–41412. [Google Scholar] [CrossRef] [PubMed]
  84. Reynaud, J.M.; Jegou, J.F.; Welsch, J.; Horvat, B. Human herpesvirus 6A infection in CD46 transgenic mice: Viral persistence in the brain and increased production of proinflammatory chemokines via TLR9. J. Virol. 2014, 88, 5421–5436. [Google Scholar] [CrossRef] [PubMed]
  85. Rizzo, R.; Bortolotti, D.; Gentili, V.; Rotola, A.; Bolzani, S.; Caselli, E.; Tola, M.R.; Di Luca, D. KIR2DS2/KIR2DL2/HLA-C1 haplotype is associated with Alzheimer’s disease: Implication for the role of herpesvirus infections. J. Alzheimers Dis. 2019, 67, 1379–1389. [Google Scholar] [CrossRef]
  86. Bortolotti, D.; Gentili, V.; Rotola, A.; Caselli, E.; Rizzo, R. HHV-6 infection induces amyloid-beta expression and activation of microglial cells. Alzheimers Res. Ther. 2019, 11, 104. [Google Scholar] [CrossRef] [PubMed]
  87. Romeo, M.A.; Masuelli, L.; Gaeta, A.; Nazzari, C.; Granato, M.; Gilardini Montani, M.S.; Faggioni, A.; Cirone, M. Impact of HHV-6A and HHV-6B lytic infection on autophagy and endoplasmic reticulum stress. J. Gen. Virol. 2019, 100, 89–98. [Google Scholar] [CrossRef] [PubMed]
  88. Romeo, M.A.; Gilardini Montani, M.S.; Gaeta, A.; D’Orazi, G.; Faggioni, A.; Cirone, M. HHV-6A infection dysregulates autophagy/UPR interplay increasing beta amyloid production and tau phosphorylation in astrocytoma cells as well as in primary neurons, possible molecular mechanisms linking viral infection to Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165647. [Google Scholar] [CrossRef] [PubMed]
  89. Romeo, M.A.; Gilardini Montani, M.S.; Falcinelli, L.; Gaeta, A.; Nazzari, C.; Faggioni, A.; Cirone, M. HHV-6B reduces autophagy and induces ER stress in primary monocytes impairing their survival and differentiation into dendritic cells. Virus Res. 2019, 273, 197757. [Google Scholar] [CrossRef] [PubMed]
  90. Costa Sa, A.C.; Madsen, H.; Brown, J.R. Shared molecular signatures across neurodegenerative diseases and herpes virus infections highlights potential mechanisms for maladaptive innate immune responses. Sci. Rep. 2019, 9, 8795. [Google Scholar] [CrossRef] [PubMed]
  91. Readhead, B.; Haure-Mirande, J.V.; Funk, C.C.; Richards, M.A.; Shannon, P.; Haroutunian, V.; Sano, M.; Liang, W.S.; Beckmann, N.D.; Price, N.D.; et al. Multiscale analysis of independent Alzheimer’s cohorts finds disruption of molecular, genetic, and clinical networks by human herpesvirus. Neuron 2018, 99, 64–82. [Google Scholar] [CrossRef] [PubMed]
  92. Allnutt, M.A.; Johnson, K.; Bennett, D.A.; Connor, S.M.; Troncoso, J.C.; Pletnikova, O.; Albert, M.S.; Resnick, S.M.; Scholz, S.W.; De Jager, P.L.; et al. Human herpesvirus 6 detection in Alzheimer’s disease cases and controls across multiple cohorts. Neuron 2020, 105, 1027–1035. [Google Scholar] [CrossRef] [PubMed]
  93. Jeong, H.H.; Liu, Z. Are HHV-6A and HHV-7 really more abundant in Alzheimer’s disease? Neuron 2019, 104, 1034–1035. [Google Scholar] [CrossRef] [PubMed]
  94. Chorlton, S.D. Reanalysis of Alzheimer’s brain sequencing data reveals absence of purported HHV-6A and HHV-7. J. Bioinform. Comput. Biol. 2020, 18, 2050012. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, V.C.; Wu, S.I.; Huang, K.Y.; Yang, Y.H.; Kuo, T.Y.; Liang, H.Y.; Huang, K.L.; Gossop, M. Herpes zoster and dementia: A nationwide population-based cohort study. J. Clin. Psychiatry 2018, 79, 16m11312. [Google Scholar] [CrossRef]
  96. Tsai, M.C.; Cheng, W.L.; Sheu, J.J.; Huang, C.C.; Shia, B.C.; Kao, L.T.; Lin, H.C. Increased risk of dementia following herpes zoster ophthalmicus. PLoS ONE 2017, 12, e0188490. [Google Scholar] [CrossRef] [PubMed]
  97. Li, Q.; Ali, M.A.; Wang, K.; Sayre, D.; Hamel, F.G.; Fisher, E.R.; Bennett, R.G.; Cohen, J.I. Insulin degrading enzyme induces a con-formational change in Varicella-Zoster Virus gE, and enhances virus infectivity and stability. PLoS ONE 2010, 5, e11327. [Google Scholar]
  98. Bernstein, H.; Keilhoff, G.; Dobrowolny, H.; Steiner, J. Binding varicella zoster virus: An underestimated facet of insulin degrading enzyme’s implication for Alzheimer´s disease pathology? Eur. Arch. Psychiatry Clin. Neurosci. 2020, 270, 495–496. [Google Scholar] [CrossRef] [PubMed]
  99. Lee, K.H.; Kwon, D.E.; Do Han, K.; La, Y.; Han, S.H. Association between cytomegalovirus end-organ diseases and moderate-to severe dementia: A population-based cohort study. BMC Neurol. 2020, 20, 216. [Google Scholar] [CrossRef] [PubMed]
  100. Aiello, A.E.; Haan, M.N.; Blythe, L.; Moore, K.; Gonzalez, J.M.; Jagust, W. The Influence of latent viral infection on rate of cognitive decline over 4 years. J. Am. Geriatr. Soc. 2006, 54, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
  101. Lurain, N.S.; Hanson, B.A.; Martinson, J.; Leurgans, S.E.; Landay, A.L.; Bennett, D.A.; Schneider, J.A. Virological and immunological characteristics of human cytomegalovirus infection associated with Alzheimer disease. J. Infect. Dis. 2013, 208, 564–572. [Google Scholar] [CrossRef] [PubMed]
  102. Caggiu, E.; Paulus, K.; Arru, G.; Piredda, R.; Sechi, G.P.; Sechi, A. Humoral cross reactivity between α-synuclein and herpes simplex-1 epitope in Parkinson’s disease, a triggering role in the disease? J. Neuroimmunol. 2016, 291, 110–114. [Google Scholar] [CrossRef] [PubMed]
  103. Caggiu, E.; Arru, G.; Hosseini, S.; Niegowska, M.; Sechi, G.; Zarbo, I.R.; Sechi, L.A. Inflammation, infectious triggers, and Parkinson’s disease. Front. Neurol. 2019, 10, 122. [Google Scholar] [CrossRef]
  104. Woulfe, J.; Hoogendoorn, H.; Tarnopolsky, M.; Muñoz, D.G. Monoclonal antibodies against Epstein-Barr virus cross-react with α-synuclein in human brain. Neurology 2000, 55, 1398–1440. [Google Scholar] [CrossRef] [PubMed]
  105. Goldeck, D.; Maetzler, W.; Berg, D.; Oettinger, L.; Pawelec, G. Altered dendritic cell subset distribution in patients with Parkinson’s disease: Impact of cmv serostatus. J. Neuroimmunol. 2016, 290, 60–65. [Google Scholar] [CrossRef] [PubMed]
  106. Koutsilieri, E.; Lutz, M.B.; Scheller, C. Autoimmunity, dendritic cells and relevance for Parkinson’s disease. J. Neural Transm. 2013, 120, 75–81. [Google Scholar] [CrossRef] [PubMed]
  107. Cury, R.G.; Lopez, W.O.C. Bilateral striatal lesion due to herpesvirus-6 infection. J. Neurol. Sci. 2015, 358, 538–539. [Google Scholar] [CrossRef] [PubMed]
  108. Dzwonek, J.; Rylski, M.; Kaczmarek, L. Matrix metalloproteinases and their endogenous inhibitors in neuronal physiology of the adult brain. FEBS Lett. 2004, 567, 129–135. [Google Scholar] [CrossRef]
  109. GBD 2016 Neurology Collaborators. Global, regional, and national burden of multiple sclerosis 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 269–285. [Google Scholar] [CrossRef] [PubMed]
  110. Ercolini, A.M.; Miller, S.D. The role of infections in autoimmune disease. Clin. Exp. Immunol. 2009, 155, 1–15. [Google Scholar] [CrossRef]
  111. Temajo, N.O.; Howard, N. The mosaic of environment involvement in autoimmunity: The abrogation of viral latency by stress, a non-infectious environmental agent, is an intrinsic prerequisite prelude before viruses can rank as infectious environmental agents that trigger autoimmune diseases. Autoimmun. Rev. 2014, 13, 635–640. [Google Scholar] [PubMed]
  112. Dimitrov, L.G. What’ s new in multiple sclerosis. Br. J. Gen. Pract. 2014, 64, 612.e3. [Google Scholar] [CrossRef] [PubMed]
  113. Walton, C.; King, R.; Rechtman, L.; Kaye, W.; Leray, E.; Marrie, R.A.; Robertson, N.; La Rocca, N.; Uitdehaag, B.; van der Mai, I.; et al. Rising prevalence of multiple sclerosis worldwide: Insights from the Atlas of MS, third edition. Mult. Scler. 2020, 26, 1816–1821. [Google Scholar] [CrossRef]
  114. Confavreux, C.; Vukusic, S. The clinical course of multiple sclerosis. Handb. Clin. Neurol. 2014, 122, 343–369. [Google Scholar] [PubMed]
  115. Kular, L.; Liu, Y.; Ruhrmann, S.; Zheleznyakova, G.; Marabita, F.; Gomez-Cabrero, D.; James, T.; Ewing, E.; Lindén, M.; Górnikiewicz, B.; et al. DNA methylation as a mediator of HLA-DRB1* 15: 01 and a protective variant in multiple sclerosis. Nat. Commun. 2018, 9, 2397. [Google Scholar] [CrossRef]
  116. Thakolwiboon, S.; Zhao-Fleming, H.; Karukote, A.; Pachariyanon, P.; Williams, H.G.; Avila, M. Regional differences in the association of cytomegalovirus seropositivity and multiple sclerosis: A systematic review and meta-analysis. Mult. Scler. Relat. Disord. 2020, 45, 102393. [Google Scholar] [CrossRef] [PubMed]
  117. Brodin, P.; Jojic, V.; Gao, T.; Bhattacharya, S.; Lopez Angel, C.J.; Furan, D.; Shen-Orr, S.; Dekker, C.L.; Swan, G.E.; Butte, A.J. Variation in the human immune system is largely driven by non- heritable influences. Cell 2015, 160, 37–47. [Google Scholar] [CrossRef]
  118. Mok, C.C.; Lau, C.S. Pathogenesis of systemic lupus erythematosus. J. Clin. Pathol. 2003, 56, 481–490. [Google Scholar] [CrossRef]
  119. Li, Z.X.; Zeng, S.; Wu, H.X.; Zhou, Y. The risk of systemic lupus erythematosus associated with Epstein-Barr virus infection. A systematic review and meta-analysis. Clin. Experiment. Med. 2019, 19, 23–36. [Google Scholar] [CrossRef] [PubMed]
  120. Quaglia, M.; Merlotti, G.; DeAndrea, M.; Borgogna, C.; Cantaluppi, V. Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story. Viruses 2021, 13, 277. [Google Scholar] [CrossRef] [PubMed]
  121. Luo, S.; Long, H.; Lu, Q. Recent Advances in Understanding Pathogenesis and Therapeutic Strategies of Systemic Lupus Erythematosus. Int. Immunopharmacol. 2020, 89, 107028. [Google Scholar] [CrossRef]
  122. Aringer, M.; Johnson, S.R. Classifying and Diagnosing Systemic Lupus Erythematosus in the21st Century. Rheumatology 2020, 59, v4–v11. [Google Scholar] [CrossRef] [PubMed]
  123. Sutanto, H.; Yuliasih, Y. Disentangling the Pathogenesis of Systemic Lupus Erythematosus: Close Ties between Immunological, Genetic and Environmental Factors. Medicina 2023, 59, 1033. [Google Scholar] [CrossRef]
  124. Rigante, D.; Esposito, S. Infections and Systemic Lupus Erythematosus: Binding or Sparring Partners? Int. J. Mol. Sci. 2015, 16, 17331–17343. [Google Scholar] [CrossRef]
  125. Illescas-Montes, R.; Corona-Castro, C.C.; Melguizo-Rodríguez, L.; Ruiz, C.; Costela-Ruiz, V.J. Infectious Processes and Systemic Lupus Erythematosus. Immunology 2019, 158, 153–160. [Google Scholar] [CrossRef] [PubMed]
  126. Mariette, X.; Criswell, L.A. Primary Sjogren’s Syndrome. N. Engl. J. Med. 2018, 378, 931–939. [Google Scholar] [CrossRef] [PubMed]
  127. Martin-Nares, E.; Hernandez-Molina, G. Novel autoantibodies in Sjogren’s syndrome: A comprehensive review. Autoimmun. Rev. 2019, 18, 192–198. [Google Scholar] [CrossRef] [PubMed]
  128. Mavragani, C.P.; Moutsopoulos, H.M. Sjogren’s syndrome. Annu. Rev. Pathol. 2014, 9, 273–285. [Google Scholar] [CrossRef] [PubMed]
  129. Parisis, D.; Chivasso, C.; Perret, J.; Soyfoo, M.S.; Delporte, C. Current state of knowledge on primary sjögren’s syndrome, an autoimmune exocrinopathy. J. Clin. Med. 2020, 9, 2299. [Google Scholar] [CrossRef]
  130. Fisher, B.A.; Jonsson, R.; Daniels, T.; Bombardieri, M.; Brown, R.M.; Morgan, P.; Bombardieri, S.; Ng, W.F.; Tzioufas, A.G.; Vitali, C.; et al. Standardisation of labial salivary gland histopathology in clinical trials in primary Sjögren’s syndrome. Ann. Rheum. Dis. 2017, 76, 1161–1168. [Google Scholar] [CrossRef] [PubMed]
  131. Pontarini, E.; Lucchesi, D.; Bombardieri, M. Current views on the pathogenesis of Sjögren’s syndrome. Curr. Opin. Rheumatol. 2018, 30, 215–221. [Google Scholar] [CrossRef]
  132. Maslinska, M.; Kostyra-Grabczak, K. The role of virus infections in Sjögren’s syndrome. Front. Immunol. 2022, 13, 823659. [Google Scholar] [CrossRef]
  133. Cusick, M.F.; Libbey, J.E.; Fujinami, R.S. Molecular mimicry as a mechanism of autoimmune disease. Clin. Rev. Allerg. Immunol. 2012, 42, 102–111. [Google Scholar] [CrossRef] [PubMed]
  134. Tapryal, S.; Gaur, V.; Kaur, K.J.; Salunke, D.M. Structural evaluation of a mimicry-recognizing paratope: Plasticity in antigen-antibody interactions manifests in molecular mimicry. J. Immunol. 2013, 191, 456–463. [Google Scholar] [CrossRef] [PubMed]
  135. Dreyfus, D.H. Gene sharing between Epstein-Barr virus and human immune response genes. Immunol. Res. 2017, 65, 37–45. [Google Scholar] [CrossRef]
  136. Pacheco, Y.; Acosta-Ampudia, Y.; Monsalve, D.M.; Chang, C.; Gershwin, M.E.; Anaya, J.M. Bystander activation and autoimmunity. J. Autoimmun. 2019, 103, 102301. [Google Scholar] [CrossRef] [PubMed]
  137. Powell, A.M.; Black, M.M. Epitope spreading: Protection from pathogens, but propagation of autoimmunity? Clin. Exp. Dermatol. 2001, 26, 427–433. [Google Scholar] [CrossRef] [PubMed]
  138. Ferrante, P.; Mancuso, R.; Pagani, E.; Guerini, F.R.; Calvo, M.G.; Saresella, M.; Speciale, L.; Caputo, D. Molecular evidences for a role of HSV-1 in multiple sclerosis clinical acute attack. J. Neurovirol. 2000, 6, S109–S114. [Google Scholar] [PubMed]
  139. Sanders, V.; Waddell, A.; Felisan, S.; Li, X.; Conrad, A.; Tourtellotte, W. Herpes simplex virus in postmortem multiple sclerosis brain tissue. Arch. Neurol. 1996, 53, 125–133. [Google Scholar] [CrossRef]
  140. Boukhvalova, M.S.; Mortensen, E.; Mbaye, A.; Lopez, D.; Kastrukoff, L.; Blanco, J.C.G. Herpes simplex virus 1 induces brain inflammation and multifocal demyelination in the cotton rat sigmodon hispidus. J. Virol. 2020, 94, 10–1128. [Google Scholar] [CrossRef]
  141. Waubant, E.; Mowry, E.M.; Krupp, L.; Chitnis, T.; Yeh, E.A.; Kuntz, N.; Ness, J.; Chabas, D.; Strober, J.; McDonald, J.; et al. Common viruses associated with lower pediatric multiple sclerosis risk. Neurology 2011, 76, 1989–1995. [Google Scholar] [CrossRef]
  142. Kastrukoff, L.F.; Lau, A.S.; Thomas, E.E. The effect of mouse strain on herpes simplex virus type 1 (HSV-1) infection of the central nervous system (CNS). Herpesviridae 2012, 3, 4. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, Y.C.; Bang, D.; Lee, S.; Lee, K.H. The effect of herpesvirus infection on the expression of cell adhesion molecules on cultured human dermal microvascular endothelial cells. J. Dermatol. Sci. 2000, 24, 38–47. [Google Scholar] [CrossRef] [PubMed]
  144. Held, K.; Eiglmeier, I.; Himmelein, S.; Sinicina, I.; Brandt, T.; Theil, D.; Dornmair, K.; Derfuss, T. Clonal expansions of CD8þ T cells in latently HSV-1-infected human trigeminal ganglia. J. Neurovirol. 2012, 18, 62–68. [Google Scholar] [CrossRef] [PubMed]
  145. Savarin, C.; Bergmann, C.C. Viral-induced suppression of self-reactive T cells: Lessons from neurotropic coronavirus-induced demyelination. J. Neuroimmunol. 2017, 308, 12–16. [Google Scholar] [CrossRef] [PubMed]
  146. Sethi, D.K.; Gordo, S.; Schubert, D.A.; Wucherpfennig, K.W. Cross reactivity of a human autoimmune TCR is dominated by a single TCR loop. Nat. Commun. 2013, 4, 2623. [Google Scholar] [CrossRef]
  147. Cortese, I.; Capone, S.; Luchetti, S.; Cortese, R.; Nicosia, A. Cross-reactive phage displayed mimotopes lead to the discovery of mimicry between HSV-1 and a brain-specific protein. J. Neuroimmunol. 2001, 113, 119–128. [Google Scholar] [CrossRef]
  148. Kozin, M.S.; Kulakova, O.G.; Favorova, O.O. Involvement of mitochondria in neurodegeneration in multiple sclerosis. Biochemist 2018, 83, 813–830. [Google Scholar] [CrossRef] [PubMed]
  149. Qi, X.; Lewin, A.S.; Sun, L.; Hauswirth, W.W.; Guy, J. Mitochondrial protein nitration primes neurodegeneration in experimental autoimmune encephalomyelitis. J. Biol. Chem. 2006, 281, 31950–31962. [Google Scholar] [CrossRef] [PubMed]
  150. Mahad, D.; Ziabreva, I.; Lassmann, H.; Turnbull, D. Mitochondrial defects in acute multiple sclerosis lesions. Brain 2008, 131, 1722–1735. [Google Scholar] [CrossRef] [PubMed]
  151. Liang, P.; Le, W. Role of autophagy in the pathogenesis of multiple sclerosis. Neurosci. Bull. 2015, 31, 435–444. [Google Scholar] [CrossRef] [PubMed]
  152. Wucherpfennig, K.W.; Strominger, J.L. Molecular mimicry in T cell-mediated autoimmunity: Viral peptides activate human T cell clones specific for myelin basic protein. Cell 1995, 80, 695–705. [Google Scholar] [CrossRef]
  153. Lunemann, J.D.; Jelcić, I.; Roberts, S.; Lutterotti, A.; Tackenberg, B.; Martin, R.; Münz, C. EBNA1-specific T cells from patients with multiple sclerosis cross react with myelin antigens and co-produce IFN-gamma and IL-2. J. Exp. Med. 2008, 205, 1763–1773. [Google Scholar] [CrossRef]
  154. Ramasamy, R.; Mohammed, F.; Meier, U.C. HLA DR2b-Binding Peptides From Human Endogenous Retrovirus Envelope, Epstein-Barr Virus and Brain Proteins in the Context of Molecular Mimicry in Multiple Sclerosis. Immunol. Lett. 2020, 217, 15–24. [Google Scholar] [CrossRef] [PubMed]
  155. Tengvall, K.; Huang, J.; Hellstrom, C.; Kammer, P.; Bistrom, M.; Ayoglu, B.; Bomfim, I.L.; Stridh, P.; Butt, J.; Brenner, N.; et al. Molecular Mimicry Between Anoctamin 2 and Epstein-Barr Virus Nuclear Antigen 1 Associates With Multiple Sclerosis Risk. Proc. Natl. Acad. Sci. USA 2019, 116, 16955–16960. [Google Scholar] [CrossRef] [PubMed]
  156. van Sechel, A.C.; Bajramovic, J.J.; van Stipdonk, M.J.; Persoon-Deen, C.; Geutskens, S.B.; van Noort, J.M. EBV-induced expression and HLA-DR-restricted presentation by human B cells of alpha B-crystallin, a candidate autoantigen in multiple sclerosis. J. Immunol. 1999, 162, 129–135. [Google Scholar] [CrossRef]
  157. Lanz, T.V.; brewer, R.C.; Ho, P.P.; Moon, J.S.; Jude, K.M.; Fernandez, D.; Fernandes, R.A.; Gomez, A.M.; Nadj, G.S.; Bartley, C.M.; et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022, 603, 321–327. [Google Scholar] [CrossRef] [PubMed]
  158. Jelcic, I.; Al Nimer, F.; Wang, J.; Lentsch, V.; Planas, R.; Jelcic, I.; Madjovski, A.; Ruhrmann, S.; Faigle, W.; Frauenknecht, K.; et al. Memory B cells activate brain-homing, autoreactive CD4(+) T cells in multiple sclerosis. Cell 2018, 175, 85–100. [Google Scholar] [CrossRef]
  159. Nociti, V.; Frisullo, G.; Marti, A.; Luigetti, M.; Iorio, R.; Patanella, A.K.; Bianco, A.; Tonali, P.A.; Grillo, R.L.; Sabatelli, M.; et al. Epstein-Barr virus antibodies in serum and cerebrospinal fluid from multiple sclerosis, chronic inflammatory demyelinating polyradiculoneuropathy and amyotrophic lateral sclerosis. J. Neuroimmunol. 2010, 225, 149–152. [Google Scholar] [CrossRef] [PubMed]
  160. Kuchroo, V.K.; Weiner, H.L. How does Epstein-Barr virus trigger MS? Immunity 2022, 55, 390–392. [Google Scholar] [CrossRef]
  161. Castellazzi, M.; Contini, C.; Tamborino, C.; Fasolo, F.; Roversi, G.; Seraceni, S.; Rizzo, R.; Baldi, E.; Tola, M.R.; Bellini, T.; et al. Epstein-Barr virus specific intrathecal oligoclonal IgG production in relapsing-remitting multiple sclerosis is limited to a subset of patients and is composed of low-affinity antibodies. J. Neuroinflamm. 2014, 11, 188. [Google Scholar] [CrossRef] [PubMed]
  162. Ascherio, A.; Munger, K.L.; Lunemann, J.D. The initiation and prevention of multiple sclerosis. Nat. Rev. Neurol. 2012, 8, 602–612. [Google Scholar] [CrossRef] [PubMed]
  163. Hedström, A.K.; Michel, A.; Butt, J.; Brenner, N.; Hillert, J.; Waterboer, T.; Kockum, I.; Olsson, T.; Alfresdsson, L. High levels of Epstein-Barr virus nuclear antigen-1-specific antibodies and infectious mononucleosis act both independently and synergistically to increase multiple sclerosis risk. Front. Neurol. 2019, 10, 1368. [Google Scholar] [CrossRef]
  164. Disanto, G.; Hall, C.; Lucas, R.; Ponsonby, A.L.; Berlanga-Taylor, A.J.; Giovannoni, G.; the Ausimmune Investigator Group. Assessing Interactions Between HLA-DRB1*15 and Infectious Mononucleosis on the Risk of Multiple Sclerosis. Mult. Scler. 2013, 19, 1355–1358. [Google Scholar] [CrossRef] [PubMed]
  165. Ahmed, S.I.; Aziz, K.; Gul, A.; Samar, S.S.; Bareeqa, S.B. Risk of multiple sclerosis in Epstein-Barr virus infection. Cureus 2019, 11, e5699. [Google Scholar] [CrossRef]
  166. Bjornevik, K.; Cortese, M.; Healy, B.C.; Kuhle, J.; Mina, M.J.; Leng, Y.; Elledge, S.J.; Niebuhr, D.W.; Scher, A.I.; Munger, K.L.; et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375, 296–301. [Google Scholar] [CrossRef]
  167. Deeba, E.; Koptides, D.; Gaglia, E.; Constantinou, A.; Lambrianides, A.; Pantzaris, M.; Krashias, G.; Christodoulou, C. Evaluation of Epstein-Barr virus-specific antibodies in Cypriot multiple sclerosis patients. Mol. Immunol. 2019, 105, 270–275. [Google Scholar] [CrossRef] [PubMed]
  168. Baglio, S.R.; van Eijndhoven, M.A.J.; Koppers-Lalic, D.; Berenguer, J.; Lougeed, S.M.; Gibbs, S.; Leveille, N.; Rinkel, R.N.P.M.; Hopmans, E.S.; Swaminathan, S.; et al. Sensing of latent EBV infection through exosomal transfer of 5′pppRNA. Proc. Natl. Acad. Sci. USA 2016, 113, E587–E596. [Google Scholar] [CrossRef] [PubMed]
  169. Afrasiabi, A.; Fewings, N.L.; Schibeci, S.D.; Keane, J.T.; Booth, D.R.; Parnell, G.P.; Swaminathan, S. The interaction of human and Epstein-Barr virus miRNAs with multiple sclerosis risk loci. Int. J. Mol. Sci. 2021, 22, 2927. [Google Scholar] [CrossRef]
  170. Absinta, M.; Maric, D.; Gharagozloo, M.; Garton, T.; Smith, M.D.; Jin, J.; Fitzgerald, K.C.; Song, A.; Liu, P.; Lin, J.P.; et al. A Lymphocyte–Microglia–Astrocyte Axis in Chronic Active Multiple Sclerosis. Nature 2021, 597, 709–714. [Google Scholar] [CrossRef]
  171. Horng, S.; Therattil, A.; Moyon, S.; Gordon, A.; Kim, K.; Argaw, A.T.; Hara, Y.; Marian, J.N.; Sawai, S.; Flodby, P.; et al. Astrocytic Tight Junctions Control Inflammatory CNS Lesion Pathogenesis. J. Clin. Investig. 2017, 127, 3136–3151. [Google Scholar] [CrossRef]
  172. Ponath, G.; Park, C.; Pitt, D. The Role of Astrocytes in Multiple Sclerosis. Front. Immunol. 2018, 9, 217. [Google Scholar] [CrossRef]
  173. Dong, Y.; Yong, V.W. When Encephalitogenic T Cells Collaborate with Microglia in Multiple Sclerosis. Nat. Rev. Neurol. 2019, 15, 704–717. [Google Scholar] [CrossRef] [PubMed]
  174. Kunkl, M.; Frascolla, S.; Amormino, C.; Volpe, E.; Tuosto, L. T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis. Cells 2020, 9, 482. [Google Scholar] [CrossRef] [PubMed]
  175. Opsahl, M.L.; Kennedy, P.G.E. Early and late HHV-6 gene transcripts in multiple sclerosis lesions and normal appearing white matter. Brain 2005, 128, 516–527. [Google Scholar] [CrossRef] [PubMed]
  176. Goodman, A.D.; Mock, D.J.; Powers, J.M.; Baker, J.V.; Blumberg, B.M. Human herpesvirus 6 genome and antigen in acute multiple sclerosis lesions. J. Infect. Dis. 2003, 187, 1365–1376. [Google Scholar] [CrossRef] [PubMed]
  177. Pormohammad, A.; Azimi, T.; Falah, F.; Faghihloo, E. Relationship of human herpes virus 6 and multiple sclerosis: A systematic review and meta-analysis. J. Cell Physiol. 2018, 233, 2850–2862. [Google Scholar] [CrossRef]
  178. Chapenko, S.; Millers, A.; Nora, Z.; Logina, I.; Kukaine, R.; Murovska, M. Correlation Between HHV-6 Reactivation and Multiple Sclerosis Disease Activity. J. Med. Virol. 2003, 69, 111–117. [Google Scholar] [CrossRef]
  179. Ramroodi, N.; Sanadgol, N.; Ganjali, Z.; Niazi, A.A.; Sarabandi, V.; Moghtaderi, A. Monitoring of active human herpes virus 6 infection in Iranian patients with different subtypes of multiple sclerosis. J. Pathog. 2013, 2013, 194932. [Google Scholar] [CrossRef]
  180. Bistrom, M.; Jons, D.; Engdahl, E.; Gustafsson, R.; Huang, J.; Brenner, N.; Butt, J.; AlonsoMagdalena, L.; Gunnarsson, M.; Vrethem, M.; et al. Epstein-Barr virus infection after adolescence and human herpesvirus 6A as risk factors for multiple sclerosis. Eur. J. Neurol. 2021, 28, 579–586. [Google Scholar] [CrossRef] [PubMed]
  181. Engdahl, E.; Gustafsson, R.; Huang, J.; Bistrom, M.; Lima Bomfim, I.; Stridh, P.; Khademi, M.; Brenner, N.; Butt, J.; Michel, A.; et al. Increased serological response against human herpesvirus 6A is associated with risk for multiple sclerosis. Front. Immunol. 2019, 10, 2715. [Google Scholar] [CrossRef]
  182. Ortega-Madueño, I.; Garcia-Montojo, M.; Dominguez-Mozo, M.I.; GarciaMartinez, A.; Arias-Leal, A.M.; Casanova, I.; Arroyo, R.; Alvarez-Lafuente, R. Anti-Human Herpesvirus 6A/B IgG Correlates with Relapses and Progression in Multiple Sclerosis. PLoS ONE 2014, 9, e104836. [Google Scholar] [CrossRef]
  183. Soldan, S.S.; Berti, R.; Salem, N.; Secchiero, P.; Flamand, L.; Calabresi, P.A.; Brennan, M.B.; Maloni, H.W.; McFarland, H.F.; Lin, H.C.; et al. Association of Human Herpes Virus 6 (HHV-6) With Multiple Sclerosis: Increased IgM Response to HHV-6 Early Antigen and Detection of Serum HHV-6 DNA. Nat. Med. 1997, 3, 1394–1397. [Google Scholar] [CrossRef]
  184. Rand, K.H.; Houck, H.; Denslow, N.D.; Heilman, K.M. Epstein-Barr virus nuclear antigen-1 (EBNA-1) associated oligoclonal bands in patients with multiple sclerosis. J. Neurol. Sci. 2000, 173, 32–39. [Google Scholar] [CrossRef] [PubMed]
  185. Derfuss, T.; Hohlfeld, R.; Meinl, E. Intrathecal antibody (IgG) production against human herpesvirus type 6 occurs in about 20% of multiple sclerosis patients and might be linked to a polyspecific B-cell response. J. Neurol. 2005, 252, 968–971. [Google Scholar] [CrossRef] [PubMed]
  186. Tejada-Simon, M.V.; Zang, Y.C.; Hong, J.; Rivera, V.M.; Zhang, J.Z. Cross-Reactivity with Myelin Basic Protein and Human Herpesvirus-6 in Multiple Sclerosis. Ann. Neurol. 2003, 53, 189–197. [Google Scholar] [CrossRef]
  187. Cheng, W.; Ma, Y.; Gong, F.; Hu, C.; Qian, L.; Huang, Q.; Yu, Q.; Zhang, J.; Chen, S.; Liu, Z.; et al. Cross-reactivity of autoreactive T cells with MBP and viral antigens in patients with MS. Front. Biosci. 2012, 17, 1648–1658. [Google Scholar] [CrossRef]
  188. Mayne, M.; Cheadle, C.; Soldan, S.S.; Cermelli, C.; Yamano, Y.; Akhyani, N.; Nagel, J.E.; Taub, D.D.; Becker, K.G.; Jacobson, S. Gene expression profile of herpesvirus infected T cells obtained using immune microarrays: Induction of proinflammatory mechanisms. J. Virol. 2001, 75, 11641–11650. [Google Scholar] [CrossRef]
  189. Dietrich, J.; Blumberg, B.M.; Roshal, M.; Baker, J.V.; Hurley, S.D.; Mayer-Proschel, M.; Mock, D.J. Infection with an endemic human herpesvirus disrupts critical glial precursor cell properties. J. Neurosci. 2004, 24, 4875–4883. [Google Scholar] [CrossRef] [PubMed]
  190. Franklin, R.J.M.; Gallo, V. The translational biology of remyelination: Past, present, and future. Glia 2014, 62, 1905–1915. [Google Scholar] [CrossRef]
  191. Cuomo, L.; Angeloni, A.; Zompetta, C.; Cirone, M.; Calogero, A.; Frati, L.; Ragona, G.; Faggioni, A. Human Herpesvirus 6 Variant A, But Not Variant B, Infects EBV-Positive B Lymphoid Cells, Activating the Latent EBV Genome Through a BZLF-1-Dependent Mechanism. AIDS Res. Hum. Retroviruses 1995, 11, 1241–1245. [Google Scholar] [CrossRef]
  192. Turcanova, V.L.; Bundgaard, B.; Hollsberg, P. Human Herpesvirus-6B Induces Expression of the Human Endogenous Retrovirus K18-Encoded Superantigen. J. Clin. Virol. 2009, 46, 15–19. [Google Scholar] [CrossRef]
  193. Soldan, S.S.; Fogdell-Hahn, A.; Brennan, M.B.; Mittleman, B.B.; Ballerini, C.; Massacesi, L.; Seya, T.; McFarland, H.F.; Jacobson, S. Elevated serum and cerebrospinal fluid levels of soluble human herpesvirus type 6 cellular receptor, membrane cofactor protein, in patients with multiple sclerosis. Ann. Neurol. 2001, 50, 486–493. [Google Scholar] [CrossRef]
  194. Sanadgol, N.; Ramroodi, N.; Ahmadi, G.A.; Komijani, M.; Moghtaderi, A.; Bouzari, M.; Rezaei, M.; Kardi, M.T.; Dabiri, S.; Moradi, M.; et al. Prevalence of Cytomegalovirus Infection and Its Role in Total Immunoglobulin Pattern in Iranian Patients with Different Subtypes of Multiple Sclerosis. New Microbiol. 2011, 34, 263–274. [Google Scholar] [PubMed]
  195. Najafi, S.; Ghane, M.; Poortahmasebi, V.; Jazayeri, S.M.; Yousefzadeh-Chabok, S. Prevalence of Cytomegalovirus in Patients with Multiple Sclerosis: A Case-Control Study in Northern Iran. Jundishapur. J. Microbiol. 2016, 9, e36582. [Google Scholar] [CrossRef] [PubMed]
  196. Smyk, D.S.; Alexander, A.K.; Walker, M.; Walker, M. Acute disseminated encephalomyelitis progressing to multiple sclerosis: Are infectious triggers involved? Immunol. Res. 2014, 60, 16–22. [Google Scholar] [CrossRef] [PubMed]
  197. Vanheusden, M.; Stinissen, P.; ’t Hart, B.A.; Hellings, N. Cytomegalovirus: A Culprit or Protector in Multiple Sclerosis? Trends Mol. Med. 2015, 21, 16–23. [Google Scholar] [CrossRef]
  198. Clerico, M.; De Mercanti, S.; Artusi, C.A.; Durelli, L.; Naismith, R.T. Active CMV Infection in Two Patients with Multiple Sclerosis Treated with Alemtuzumab. Mult. Scler. 2017, 23, 874–876. [Google Scholar] [CrossRef]
  199. Zheng, M.M.; Zhang, X.H. Cross-reactivity between human cytomegalovirus peptide 981-1003 and myelin oligodendroglia glycoprotein peptide 35-55 in experimental autoimmune encephalomyelitis in Lewis rats. Biochem. Biophys. Res. Commun. 2014, 443, 1118–1123. [Google Scholar] [CrossRef]
  200. Maple, P.A.C.; Tanasescu, R.; Gran, B.; Constantinescu, C.S. A Different Response to Cytomegalovirus (CMV) and Epstein-Barr Virus (EBV) Infection in UK People with Multiple Sclerosis (PwMS) Compared to Controls. J. Infect. 2020, 80, 320–325. [Google Scholar] [CrossRef]
  201. Zabalza, A.; Vera, A.; Alari-Pahissa, E.; Munteis, E.; Moreira, A.; Yélamos, J.; Llop, M.; López-Botet, M.; Martínez-Rodríguez, J.E. Impact of Cytomegalovirus Infection on B Cell Differentiation and Cytokine Production in Multiple Sclerosis. J. Neuroinflamm. 2020, 17, 161. [Google Scholar] [CrossRef]
  202. Kang, J.-H.; Sheu, J.-J.; Kao, S.; Lin, H.C. Increased risk of multiple sclerosis following herpes zoster: A nationwide, population-based study. J. Infect. Dis. 2011, 204, 188–192. [Google Scholar] [CrossRef]
  203. Jarius, S.; Eichhorn, P.; Franciotta, D.; Petereit, H.F.; Akman-Demir, G.; Wick, M.; Wildemann, B. The MRZ reaction as a highly specific marker of multiple sclerosis: Re-evaluation and structured review of the literature. J. Neurol. 2017, 264, 453–466. [Google Scholar] [CrossRef]
  204. Rice, E.M.; Thakolwiboon, S.; Avila, M. Geographic heterogeneity in the association of varicella-zoster virus seropositivity and multiple sclerosis: A systematic review and meta-analysis. Mult. Scler. Relat. Disord. 2021, 53, 103024. [Google Scholar] [CrossRef] [PubMed]
  205. Sotelo, J.; Ordoñez, G.; Pineda, B.; Flores, J. The participation of varicella zoster virus in relapses of multiple sclerosis. Clin. Neurol. Neurosurg. 2014, 119, 44–48. [Google Scholar] [CrossRef] [PubMed]
  206. Zhu, G.; Zhou, S.; Xu, Y.; Gao, R.; Zhang, M.; Zeng, Q.; Su, W.; Wang, R. Chickenpox and multiple sclerosis: A Mendelian randomization study. J. Med. Virol. 2022, 95, e28315. [Google Scholar] [CrossRef]
  207. Moon, U.Y.; Park, S.J.; Oh, S.T.; Kim, W.U.; Park, S.H.; Lee., S.H.; Choo, C.S.; Kim, H.Y.; Lee, W.K.; Leel, S.K. Patients with systemic lupus erythematosus have abnormally elevated Epstein-Barr virus load in blood. Arthritis Res. Ther. 2004, 6, R295–R302. [Google Scholar] [CrossRef]
  208. Chen, X.; Li, H.; Wu, C.; Zhang, Y. Epstein‒Barr virus and human herpesvirus 6 infection in patients with systemic lupus erythematosus. Virol. J. 2023, 20, 29. [Google Scholar] [CrossRef] [PubMed]
  209. Berkun, Y.; Zandman-Goddard, G.; Barzilai, O.; Boaz, M.; Sherer, Y.; Larida, B.; Blank, M.; Anaya, J.M.; Shoenfeld, Y. Infectious antibodies in systemic lupus erythematosus patients. Lupus 2009, 18, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
  210. James, J.A.; Neas, B.R.; Moser, K.L.; Hall, T.; Bruner, G.R.; Sestak, A.L.; Harley, J.B. Systemic lupus erythematosus in adults is associated with previous Epstein-Barr virus exposure. Arthritis Rheum. 2001, 44, 1122–1126. [Google Scholar] [CrossRef]
  211. Chougule, D.; Nadkar, M.; Rajadhyaksha, A.; Khadilkar, P.; Patwardhan, M.; Kaveri, S.; Ghosh, K.; Pradhan, V. Association of clinical and serological parameters of systemic lupus erythematosus patients with Epstein-Barr virus antibody profile. J. Med. Virol. 2018, 90, 559–563. [Google Scholar] [CrossRef]
  212. Jog, N.R.; Young, K.A.; Munroe, M.E.; Harmon, M.T.; Guthridge, J.M.; Kelly, J.A.; Kame, D.L.; Gilkeson, G.S.; Weisman, M.H.; Karp, D.R.; et al. Association of Epstein-Barr virus serological reactivation with transitioning to systemic lupus erythematosus in at-risk individuals. Ann. Rheum. Dis. 2019, 78, 1235–1241. [Google Scholar] [CrossRef]
  213. James, J.A.; Harley, J.B.; Scofield, R.H. Epstein-Barr virus and systemic lupus erythematosus. Curr. Opin. Rheumatol. 2006, 18, 462–467. [Google Scholar] [CrossRef]
  214. Yadav, P.; Tran, H.; Ebegbe, R.; Gottlieb, P.; Wei, H.; Lewis, R.H.; Mumbey-Wafula, A.; Kaplan, A.; Kholdarova, E.; Spatz, L. Antibodies elicited in response to EBNA-1 may cross-react with dsDNA. PLoS ONE 2011, 6, e14488. [Google Scholar] [CrossRef] [PubMed]
  215. Csorba, K.; Schirmbeck, L.A.; Tuncer, E.; Ribi, C.; Roux-Lombard, P.; Chizzolini, C.; Huynh-Do, U.; Vanhecke, D.; Trendelenburg, M. Anti-C1q Antibodies as Occurring in Systemic Lupus Erythematosus Could Be Induced by an Epstein-Barr Virus-Derived Antigenic Site. Front. Immunol. 2019, 10, 2619. [Google Scholar] [CrossRef]
  216. Incaprera, M.; Rindi, L.; Bazzichi, A.; Garzelli, C. Potential role of the Epstein– Barr virus in systemic lupus erythematosus autoimmunity. Clin. Exp. Rheumatol. 1998, 16, 289–294. [Google Scholar]
  217. Tu, J.; Wang, X.; Geng, G.; Xue, X.; Lin, X.; Zhu, X.; Sun, L. The Possible Effect of B-Cell Epitopes of Epstein-Barr Virus Early Antigen, Membrane Antigen, Latent Membrane Protein-1, and -2A on Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 187. [Google Scholar] [CrossRef]
  218. Jog, N.R.; James, J.A. Epstein Barr Virus and Autoimmune Responses in Systemic Lupus Erythematosus. Front. Immunol. 2021, 11, 623944. [Google Scholar] [CrossRef]
  219. Jog, N.R.; Chakravarty, E.F.; Guthridge, J.M.; James, J.A. Epstein Barr Virus Interleukin 10 Suppresses Anti-inflammatory Phenotype in Human Monocytes. Front. Immunol. 2018, 9, 2198. [Google Scholar] [CrossRef]
  220. Stewart, J.P.; Rooney, C.M. The interleukin-10 homolog encoded by Epstein-Barr virus enhances the reactivation of virus-specific cytotoxic T cell and HLA unrestricted killer cell responses. Virology 1992, 191, 773–782. [Google Scholar] [CrossRef]
  221. HoHsieh, A.; Wang, C.M.; Wu, Y.-J.J.; Chen, A.; Chang, M.-I.; Chen, J.-Y. B Cell Epitope of Human Cytomegalovirus Phosphoprotein 65 (HCMV Pp65) Induced Anti-DsDNA Antibody in BALB/c Mice. Arthritis Res. Ther. 2017, 19, 65. [Google Scholar] [CrossRef]
  222. Hsieh, A.-H.; Kuo, C.-F.; Chou, I.-J.; Tseng, W.-Y.; Chen, Y.-F.; Yu, K.-H.; Luo, S.-F. Human Cytomegalovirus Pp65 Peptide-Induced Autoantibodies Cross-Reacts with TAF9 Protein and Induces Lupus-like Autoimmunity in BALB/c Mice. Sci. Rep. 2020, 10, 9662. [Google Scholar] [CrossRef]
  223. Neo, J.Y.J.; Wee, S.Y.K.; Bonne, I.; Tay, S.H.; Raida, M.; Jovanovic, V.; Fairhurst, A.-M.; Lu, J.; Hanson, B.J.; MacAry, P.A. Characterisation of a Human Antibody That Potentially Links Cytomegalovirus Infection with Systemic Lupus Erythematosus. Sci. Rep. 2019, 9, 9998. [Google Scholar] [CrossRef]
  224. Guo, G.; Ye, S.; Xie, S.; Ye, L.; Lin, C.; Yang, M.; Shi, X.; Wang, F.; Li, B.; Li, M.; et al. The Cytomegalovirus Protein US31 Induces Inflammation through Mono-Macrophages in Systemic Lupus Erythematosus by Promoting NF-KB2 Activation. Cell Death Dis. 2018, 9, 104. [Google Scholar] [CrossRef] [PubMed]
  225. Rozenblyum, E.V.; Allen, U.D.; Silverman, E.D.; Levy, D.M. Cytomegalovirus Infection in Childhood-Onset Systemic Lupus Erythematosus. Int. J. Clin. Rheumatol. 2013, 8, 137–146. [Google Scholar] [CrossRef]
  226. Janahi, E.M.A.; Das, S.; Bhattacharya, S.N.; Haque, S.; Akhter, N.; Jawed, A.; Wahid, M.; Mandal, R.K.; Lohani, M.; Areeshi, M.Y.; et al. Cytomegalovirus Aggravates the Autoimmune Phenomenon in Systemic Autoimmune Diseases. Microb. Pathog. 2018, 120, 132–139. [Google Scholar] [CrossRef] [PubMed]
  227. Kivity, S.; Arango, M.T.; Ehrenfeld, M.; Tehori, O.; Shoenfeld, Y.; Anaya, J.M.; Agmon-Levin, N. Infection and autoimmunity in Sjogren’s syndrome: A clinical study and comprehensive review. J. Autoimmun. 2014, 51, 17–22. [Google Scholar] [CrossRef]
  228. Sanosyan, A.; Daien, C.; Nutz, A.; Bollore, K.; Bedin, A.S.; Morel, J.; Zimmermann, V.; Nocturne, G.; Peries, M.; Guigue, N.; et al. Discrepancy of Serological and Molecular Patterns of Circulating Epstein-Barr Virus Reactivation in Primary Sjögren’s Syndrome. Front. Immunol. 2019, 10, 1153. [Google Scholar] [CrossRef]
  229. Barcelos, F.; Martins, C.; Monteiro, R.; Cardigos, J.; Prussiani, T.; Sitima, M.; Alves, N.; Vaz-Patto, J.; Cunha-Branco, J.; Borrego, L.M. Association between EBV serological patterns and lymphocytic profle of SjS patients support a virally triggered autoimmune epithelitis. Sci. Rep. 2021, 11, 4082. [Google Scholar] [CrossRef] [PubMed]
  230. Saito, I.; Servenius, B.; Compton, T.; Fox, R.I. Detection of Epstein-Barr virus DNA by polymerase chain reaction in blood and tissue biopsies from patients with Sjögren’s syndrome. J. Exp. Med. 1989, 169, 2191–2198. [Google Scholar] [CrossRef] [PubMed]
  231. Casanova, J.L.; Abel, L. Mechanisms of viral inflammation and disease in humans. Science 2021, 374, 1080–1086. [Google Scholar] [CrossRef]
  232. Hou, X.; Hong, X.; Ou, M.; Meng, S.; Wang, T.; Liao, S.; He, J.; Yu, H.; Liu, L.; Yin, L.; et al. Analysis of Gene Expression and TCR/B Cell Receptor Profiling of Immune Cells in Primary Sjogren’s Syndrome by Single-Cell Sequencing. J. Immunol. 2022, 209, 238–249. [Google Scholar] [CrossRef]
  233. Maslinska, M. The role of Epstein-Barr virus infection in primary Sjogren’s syndrome. Curr. Opin. Rheumatol. 2019, 31, 475–483. [Google Scholar] [CrossRef]
  234. Huang, P.J.; Lin, S.P.; Wu, C.Y.; Liu, Y.T.; Chen, H.H. Association between a History of herpes zoster and the risk of Sjögren’s syndrome: A nationwide, population-based, case– control study. BMJ Open 2022, 12, e061962. [Google Scholar] [CrossRef]
  235. Newkirk, M.M.; Duffy, K.N.W.; Lecrerc, J.; Lambert, N.; Shiroky, J.B. Detection of cytomegalovirus, Epstein-Barr and herpes virus—6 in patients with rheumatoid arthritis with or without sjogren’s syndrome. Rheumatology 1994, 33, 317–322. [Google Scholar] [CrossRef]
  236. Broccolo, F.; Fusetti, L.; Ceccherini-Nelli, L. Possible role of human herpesvirus 6 as a trigger of autoimmune disease. Sci. World J. 2013, 24, 867389. [Google Scholar] [CrossRef]
  237. Takizawa, Y.; Inokuma, S.; Tanaka, Y.; Saito, K.; Atsumi, T.; Hirakata, M.; Kameda, H.; Hirohata, S.; Kondo, H.; Kumagai, S.; et al. Clinical characteristics of cytomegalovirus infection in rheumatic diseases: Multicentre survey in a large patient population. Rheumatology 2008, 47, 1373–1378. [Google Scholar] [CrossRef]
  238. Leonard, L.; Blauwendraat, C.; Iwaki, H.; Johnson, N.; Bandres-Ciga, S.; Ferrucci, L.; Faghri, F.; Singleton, A.B.; Nalls, M.A. Virus exposure and neurodegenerative disease risk across national biobanks. Neuron 2023, 111, 1086–1093. [Google Scholar]
  239. Itzaki, R.F.; Tabet, N. Herpes simplex encephalitis and Alzheimer’s disease: Is there a link? J. Neurol. Sci. 2017, 380, 20–21. [Google Scholar] [CrossRef]
  240. Marcocci, M.E.; Napoletani, G.; Protto, V.; Kolesova, O.; Piacentini, R.; Li Puma, D.D.; Lomonte, P.; Grassi, C.; Palamara, A.T.; De Chiara, G. Herpes simplex virus-1 in the brain: The dark side of a sneaky infection. Trends Microbiol. 2020, 28, 808–820. [Google Scholar] [CrossRef]
  241. Huang, S.Y.; Yang, Y.X.; Kuo, K.; Li, H.Q.; Shen, X.N.; Chen, S.D.; Cui, M.; Tan, L.; Dong, J.T.; Yu, J.T. Herpesvirus infections and Alzheimer’s disease: A Mendelian randomization study. Alzheimers Res. Ther. 2021, 13, 158. [Google Scholar] [CrossRef]
  242. Xu, L.; Zhang, L.J.; Yang, L.; Yang, C.S.; Yi, M.; Zhang, S.N.; Wang, N.; Huang, C.N.; Liu, M.Q. Positive association of herpes simplex virus-IgG with multiple sclerosis: A systematic review and meta-analysis. Mult. Scler. Relat. Disord. 2021, 47, 102633. [Google Scholar] [CrossRef]
  243. Safieh, M.; Korczyn, A.D.; Michaelson, D.M. ApoE4: An emerging therapeutic target for Alzheimer’s disease. BMC Med. 2019, 17, 64. [Google Scholar] [CrossRef]
  244. Fominykh, V.; Shadrin, A.A.; Jaholkowski, P.P.; Bahrami, S.; Athanasiu, L.; Wightman, D.P.; Uffelmann, E.; Posthuma, D.; Selbaek, G.; Dale, A.M.; et al. Shared genetic loci between Alzheimer’s disease and multiple sclerosis: Crossroads between neurodegeneration and immune system. Neurobiol. Dis. 2023, 183, 106174. [Google Scholar] [CrossRef] [PubMed]
  245. Iqbal, U.H.; Zeng, E.; Pasinetti, G.M. The use of antimicrobial and antiviral drugs in Alzheimer’s disease. Int. J. Mol. Sci. 2020, 21, 4920. [Google Scholar] [CrossRef] [PubMed]
  246. Hui, Z.; Zhijun, Y.; Yushan, Y.; Liping, C.; Yiying, Z.; Difan, Z.; Chunglit, C.T.; Wei, C. The combination of acyclovir and dexamethasone protects against Alzheimer’s disease-related cognitive impairments in mice. Psychopharmacology 2020, 237, 1851–1860. [Google Scholar] [CrossRef]
  247. Schnier, C.; Janbek, J.; Lathe, R.; Haas, J. Reduced dementia incidence after Varicella zoster vaccination in Wales 2013–2020. Alzheimers Dement. 2022, 8, e12293. [Google Scholar] [CrossRef] [PubMed]
  248. Scherrer, J.F.; Salas, J.; Wiemken, T.L.; Hoft, D.F.; Jacobs, C.; Morley, J.E. Impact of herpes zoster vaccination on incident dementia: A retrospective study in two patient cohorts. PLoS ONE 2021, 16, e0257405. [Google Scholar] [CrossRef]
  249. Lehrer, S.; Rheinstein, P.H. Herpes zoster vaccination reduces risk of dementia. In Vivo 2021, 35, 3271–3275. [Google Scholar] [CrossRef]
  250. Lophatananon, A.; Mekli, K.; Cant, R.; Burns, A.; Dobson, C.; Itzhaki, R.; Muir, K. Shingles, Zostavax vaccination and risk of developing dementia: A nested case–control study—Results from the UK Biobank cohort. BMJ Open 2021, 11, e045871. [Google Scholar] [CrossRef]
Figure 1. Role of herpesviruses in Alzheimer’s disease. Where there is no explanation next to it, the arrow with the tip indicates activation, the arrow without the tip indicates inhibition. The red up arrow indicates increase, while the red down arrow indicates decrease. Abbreviations: ROS, reactive oxygen species; TAP, transporter protein; NFTs, neurofibrillar tangles.
Figure 1. Role of herpesviruses in Alzheimer’s disease. Where there is no explanation next to it, the arrow with the tip indicates activation, the arrow without the tip indicates inhibition. The red up arrow indicates increase, while the red down arrow indicates decrease. Abbreviations: ROS, reactive oxygen species; TAP, transporter protein; NFTs, neurofibrillar tangles.
Viruses 16 00133 g001
Figure 2. Role of herpesviruses in Parkinson’s disease.
Figure 2. Role of herpesviruses in Parkinson’s disease.
Viruses 16 00133 g002
Figure 3. Role of herpesviruses in multiple sclerosis.
Figure 3. Role of herpesviruses in multiple sclerosis.
Viruses 16 00133 g003
Figure 4. Role of herpesviruses in systemic lupus erythematosus and Sjogren’s syndrome.
Figure 4. Role of herpesviruses in systemic lupus erythematosus and Sjogren’s syndrome.
Viruses 16 00133 g004
Table 1. Summary of findings supporting a role of herpesviruses in neurodegenerative diseases.
Table 1. Summary of findings supporting a role of herpesviruses in neurodegenerative diseases.
DiseaseResultStudy TypeRefs
ADThe presence of Aβ and HSV-1 cross-reactive antibodies in patients with ADComparative study[73]
An increased level of IgM antibodies against HSV-1,2 was associated with an increased risk of developing dementiaProspective cohort studies[74,75]
AD-derived B lymphocytes infected with EBV produced high levels of TNF-α in vitroCell model studies[78,79]
EBV antigens stimulated cytotoxic and proinflammatory functions by CD45RA+ cellsProspective cohort and knockout studies[80,81]
Increased CMV blood markers associated with cognitive declineLongitudinal, prospective cohort studies [76,100]
Levels of CMV IgG antibodies associated with increased neurofibrillary tanglesLongitudinal study[101]
Patients with herpes zoster showed a significantly increased risk of developing ADRetrospective cohort studies[95,96]
Studies showed an association between CMV and moderate–severe dementiaPopulation cohort study[99]
HSV-1 induced neuroinflammationIn vitro model studies[47,48,49,50,51]
HSV-1 induced the production of both Aβ and the tau protein in human neural cellsIn vitro infection studies[55]
HSV-1 DNA was present in the brain of AD patientsMolecular studies[57,58]
EBV was present in the blood and brains of AD patientsComparative and molecular study[76]
EBV reactivation has been associated with cognitive declineLongitudinal study[77]
EBV BNLF-2a has been associated with AD progressionBiochemical studies[82,83]
HHV-6-infected microglia showed an accumulation of Aβ and tau proteinsIn vivo and in vitro infection model[85,86]
HHV-6 reduced autophagyIn vitro infection model[87,88,89]
HHV-6-infected patients and AD patients shared 95 differentially expressed genesComputational analysis study[90]
Binding of VZV to the insulin-degrading enzymeIn vitro infection model[97,98]
PDMolecular mimicry between HSV-1 and α-synucleinSeroprevalence study[102]
Molecular mimicry between EBV LMP1 and α-synucleinSeroprevalence study[104]
CMV-, EBV-, and HHV-6-infected patients shared several differentially expressed genes with AD patientsComputational analysis study[90]
AD, Alzheimer’s disease; PD, Parkinson’s disease, LMP-1, latent membrane protein-1.
Table 2. Summary of findings supporting a role for herpesviruses in autoimmune diseases.
Table 2. Summary of findings supporting a role for herpesviruses in autoimmune diseases.
DiseaseResultStudy TypeRefs
MSMolecular mimicry between HSV-1 protein and myelin basic proteinBiochemical study[150]
Molecular mimicry between EBV protein LMP-1 and different proteins involved in MS pathogenesisCase–control studies[152,153,154,155]
Molecular mimicry between EBV proteins BHRF1 and BPLF1 with a protein present in neuronsEpitope discover approach and cell immunity analysis[158]
Defective cytotoxic T cells control of EBV in MSSeroprevalence study[159]
Oligoclonal IgG against EBV have been detected in the brain of MS patientsComparative study[161]
Higher titers of EBV antibodies have been detected in MS patientsCase control study[164]
Serological response against HHV-6 was higher in MS patientsMolecular, case–control studies[179,180,181,182,183]
Oligoclonal IgG against HHV-6 have been detected in the brains of MS patientsSeroprevalence study[184]
Molecular mimicry between HHV-6 protein U24 and myelin basic proteinSeroprevalence study[186]
Increased level of sCD46, the receptor for HHV-6 has been detected in MS patients Immunological study[193]
Molecular mimicry between HCMV antigen (UL86981-1003) and myelin oligodendrocyte glycoprotein (MOG) In vivo experimental model[199]
Patients with herpes zoster have been associated with a higher risk of MSPopulation-based study and computational GWAS[202,206]
HSV-1 DNA in the blood and in the brains of MS patientsMolecular studies[138,139]
HSV-1 infection-induced CNS demyelination and neuroinflammationIn vitro and in vivo infection studies[140,141,142,143,145]
EBV-induced an inflammatory environmentIn vitro infection and molecular studies[168,169,170,171,172,173]
High load of HCMV genome has been detected in MS patientsCase–control study [195]
High load of VZV genome has been detected in MS patientsMolecular study[205]
SLESerological response against EBV was higher in SLE patientsCase–control studies[209,210,211]
EBV antibodies cross-reacted with autoantigens of SLESeroprevalence studies[215,216,217]
Similarity between CMV antigens and autoantigens of SLEAnimal model and in vitro infection studies[222,223]
High load of EBV DNA was detected in SLE patientsMolecular study[207]
vIL-10, a viral EBV protein, has been detected at higher concentrations in plasma of SLE patientsIn vitro infection model[219]
SSHigher titers of anti-HHV-6 have been found in a group of SS patientsSeroprevalence study[235]
A significant correlation between herpes zoster exposure and SS risk has been detectedPopulation-based case–control study[234]
High levels of EBV DNA have been detected in SS patientsMolecular study [230]
Increased EBV reactivation has been observed in SS patientsObservational study[228]
MS, multiple sclerosis; SLE, systemic lupus erythematosus; SS, Sjögren’s syndrome; LMP-1, latent membrane protein-1; anti-EA, anti-early antigen.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

De Francesco, M.A. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses 2024, 16, 133. https://doi.org/10.3390/v16010133

AMA Style

De Francesco MA. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses. 2024; 16(1):133. https://doi.org/10.3390/v16010133

Chicago/Turabian Style

De Francesco, Maria Antonia. 2024. "Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them?" Viruses 16, no. 1: 133. https://doi.org/10.3390/v16010133

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop