Next Article in Journal
Antiviral Activity of 3D, a Butene Lactone Derivative Against Influenza A Virus In Vitro and In Vivo
Previous Article in Journal
VOC 202012/01 Variant Is Effectively Neutralized by Antibodies Produced by Patients Infected before Its Diffusion in Italy
Previous Article in Special Issue
Human Cytomegalovirus and Autoimmune Diseases: Where Are We?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story

1
Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale (UPO), “Maggiore della Carità” University Hospital, Via P. Solaroli 17, 28100 Novara, Italy
2
Center for Translational Research on Autoimmune and Allergic Disease-CAAD, 28100 Novara, Italy
3
Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10126 Turin, Italy
4
Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy
*
Author to whom correspondence should be addressed.
Viruses 2021, 13(2), 277; https://doi.org/10.3390/v13020277
Submission received: 1 January 2021 / Revised: 6 February 2021 / Accepted: 7 February 2021 / Published: 11 February 2021
(This article belongs to the Special Issue Viruses and Autoimmunity)

Abstract

:
A causal link between viral infections and autoimmunity has been studied for a long time and the role of some viruses in the induction or exacerbation of systemic lupus erythematosus (SLE) in genetically predisposed patients has been proved. The strength of the association between different viral agents and SLE is variable. Epstein–Barr virus (EBV), parvovirus B19 (B19V), and human endogenous retroviruses (HERVs) are involved in SLE pathogenesis, whereas other viruses such as Cytomegalovirus (CMV) probably play a less prominent role. However, the mechanisms of viral–host interactions and the impact of viruses on disease course have yet to be elucidated. In addition to classical mechanisms of viral-triggered autoimmunity, such as molecular mimicry and epitope spreading, there has been a growing appreciation of the role of direct activation of innate response by viral nucleic acids and epigenetic modulation of interferon-related immune response. The latter is especially important for HERVs, which may represent the molecular link between environmental triggers and critical immune genes. Virus-specific proteins modulating interaction with the host immune system have been characterized especially for Epstein–Barr virus and explain immune evasion, persistent infection and self-reactive B-cell “immortalization”. Knowledge has also been expanding on key viral proteins of B19-V and CMV and their possible association with specific phenotypes such as antiphospholipid syndrome. This progress may pave the way to new therapeutic perspectives, including the use of known or new antiviral drugs, postviral immune response modulation and innate immunity inhibition. We herein describe the state-of-the-art knowledge on the role of viral infections in SLE, with a focus on their mechanisms of action and potential therapeutic targets.

1. Introduction

Autoimmune disorders (AIDs) have a multifactorial pathogenesis, characterized by an interplay between immune dysregulation, environmental factors, and a genetic background [1].
Systemic lupus erythematosus (SLE) is a multisystem AID predominantly affecting women of child-bearing age, with a chronic relapsing–remitting course. Key aspects of SLE pathophysiology include impaired clearance of nucleic acids (NA), enhanced type I Interferon (IFN) response, abnormality in B cell tolerance and production of multiple autoantibodies, with immunocomplex formation and deposition causing progressive organ damage.
Antinuclear antigens (ANA), antidouble stranded DNA (dsDNA) and anti-Smith (anti-Sm) antibodies are serological hallmarks of SLE.
Clinical manifestations are variable. Constitutional, mucocutaneous and muscoloskeletal symptoms usually represent the earliest signs of disease. However, any apparatus can be involved, from the cardiovascular to the central nervous system, and an interval of years can exist between the onset of different symptoms [1,2].
Lupus nephritis (LN) develops in around 50% of patients and can progress to end-stage renal disease in 10% of them, significantly increasing disease morbidity and mortality [3].
Despite immunosuppressive therapy, currently based on hydroxychloroquine, mycophenolate mofetil, cyclophosphamide and steroids [1], one third of patients require multiple treatment cycles due to resistant or relapsing forms [4], with a heavy impact on all aspects of their lives [5]. The increasingly widespread use of biologics (Rituximab and Belimumab) is only partially overcoming these limits [1].
The establishment of a causal link between infections and autoimmunity has been largely evaluated in a host of clinical studies, proving the role of infectious agents in the induction, as well as in the progression or flareups of SLE. However, we are still far from an in depth understanding of microbial–host interactions in its pathogenesis.
An underlying trigger for SLE has remained elusive, and multiple interacting environmental and genetic factors likely contribute to onset and perpetuation. Among environmental influences, infectious agents appear to play a pivotal role in driving autoimmunity [6,7,8,9,10,11].
On the other hand, chronic immunosuppressive therapy significantly increases the risk of infections in SLE [12] and viral infections or reactivation can be severe and even life-threatening in this setting [13]. Thus, the relationship between viruses and SLE is complex and multifaceted.
The aim of this review is to provide state-of-the-art knowledge on the role of viral infections in triggering SLE onset and flares and in modulating its phenotype and course, with a focus on viral mechanisms of interaction with host immune system. The impact of immunosuppressive therapy on the risk of developing viral infections in SLE patients is beyond the scope of this work.
The PubMed library was searched from inception to December 2020, using a combination of Medical Subject Headings (MeSH) and keywords related to SLE, autoimmunity, AIDs and viruses, including: Epstein–Barr virus (EBV), Parvovirus B19 (B19V), Retroviruses (RVs), Human Endogenous Retroviruses (HERV), Human Immunodeficiency Virus (HIV), Torque Teno Virus (TTV), Cytomegalovirus (CMV). We also checked the references of relevant articles.

2. General Mechanisms of Virus-Induced Autoimmunity

Association of SLE with viral infection has been proposed on the basis of some key observations, which can be summarized as follows: (a) it can occur during chronic viral infections; (b) SLE onset of can be concomitant with that of a viral infection; (c) experimental evidence suggests that specific viral infections can trigger it.
In general, viral infections can interact with the host immune system through several mechanisms which ultimately lead to the loss of tolerance, the production of autoantibodies, the tissue deposition of immune complexes and consequent tissue damage. These are: structural or functional molecular mimicry, epitope spreading, superantigen production, bystander activation, altered apoptosis and clearance deficit, epigenetic factors, persistent or recurrent viral infection, and innate immunity activation with type I IFN production [14,15,16,17,18,19,20,21,22,23,24,25,26].
All these mechanisms, described in Table 1, have been potentially implicated in the pathogenesis of SLE but are shared by many other AIDs.
Molecular mimicry and epitope spreading are classical, well-documented mechanisms of autoimmunity induction, as confirmed in a recent study which showed massive peptide sharing between EBV epitopes and human proteins [15].
Over the last decade, however, epigenetic mechanisms have been highlighted as an important modality of the interaction between viruses and the immune system in SLE, as they allow viruses to create gene expression profiles predisposing the host to autoimmunity. They include DNA methylation changes in SLE-susceptibility genes (e.g., IFN-related genes), histone modifications and microRNA (miRNA) modulation [24,25,26]. These mechanisms are especially exploited by HERVs, as detailed in a specific paragraph.
Persistent viral infection can be due to viruses with the ability to evade immune clearance indefinitely, such as herpesviruses and hepatitis C virus (HCV). This determines chronic inflammation and polyclonal T- and B-cell proliferation, eventually leading to emergence of self-reactive clones and autoantibodies production. Many non-retrovirus RNA viruses cause “within host” persistent infections which can occasionally trigger AIDs. Specific strategies to achieve viral persistence have been partly elucidated for HCV, a virus associated with several autoimmune manifestations and possibly also with SLE, and will be analyzed in a specific paragraph (Section 3.1) [27,28].
Recurrent viral infections role in autoimmunity has been highlighted by recent evidence on type 1 diabetes (T1D) pathogenesis. Human pancreatic β-cells can sustain recurrent enteroviral infections, including RNA-virus Coxsackieviruses B (CVB), which can induce an adaptive response with paradoxical production of “facilitating antibodies” (antibody-dependent enhancement, or ADE). When CVB infection recurs, ADE effect facilitates monocyte recruitment, the type I IFN response and the dissemination of infection. Subsequent HERV-W envelope protein (HERV-W-Env) transactivation further promotes pancreatic autoimmune attack, leading to T1D [29]. Recurrent herpetic infections also appear to play a role in several postinfectious autoimmune neurological disorders. Herpes Simplex virus (HSV) infection can lead to Herpes Simplex Virus encephalitis, which evolves in autoimmune encephalitis in up to 27% of cases, while Varicella Zoster Virus (VZV) has been associated to Neuromyelitis Optica spectrum disorders [30].
Activation of innate immunity is another important mechanism facilitating autoimmunity (Table 1). Viral nucleic acids (NAs) and other pathogen- or damage-associated molecular patterns (PAMPs and DAMPs, respectively) interact with a variety of pattern recognition receptors (PRRs) such as toll-like receptors (TLRs), the nucleotide binding and oligomerization domain receptors (NLRs), retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation associated gene 5 (MDA-5), known as RIG-I-like receptors (RLRs), and cyclic GMP-AMP synthase (cGAS). After binding viral NAs, these complexes can be incorporated into apoptotic blebs, promoting the activation of dendritic cells (DCs) and B cells, or activating multiple intracellular pathways converging on the expression of proinflammatory cytokines and type I IFN response genes in plasmacytoid DCs (pDCs) [31], as shown in Figure 1.
Thus TLRs, RLR and the cGAS receptor systems play an essential role in host defense against viral infections, but at the risk of potential autoreactivity against abundant self-NAs.
Of interest is the fact that the expression of TLR-7 and TLR-9 by B cells is associated with the production of typical SLE autoantibodies (anti-dsDNA, antiribonucleoproteins) [32,33].
Finally, direct cytotoxicity of viruses with a specific tropism for a certain tissue can result in development of an AID as a consequence of cell destruction. For example, herpesviruses are neurotropic and can kill central nervous system cells causing several AIDs [34] and, as already mentioned, CVB and HERV-W-Env exert a direct cytotoxic effect towards pancreatic β-cells, contributing to T1DM pathogenesis [29].

3. Role of Specific Viral Infections in SLE

Many viruses have been linked to the pathogenesis of SLE, particularly EBV, B19V, RVs and CMV. Among these, EBV has the strongest evidence as an etiological candidate, and a definite association seems to exist also with B19V and RV. The strength of association is variable for other viruses [14].

3.1. Epstein–Barr Virus (EBV)

The general features of EBV are outlined in Table 2. EBV is present in a latent form in the memory B cells of 95% of the world population. After being transmitted by saliva, it first infects pharyngeal epithelial cells and then resting B-lymphocytes, T lymphocytes, NK cells and neutrophils [35]. While a primary infection is usually asymptomatic (during childhood) or causes a self-limiting disease, infectious mononucleosis (IM) (in adolescence), the virus is also associated with several neoplasms and AIDs [6,7,8,9,10,11].
An in depth analysis of EBV’s molecular biology is beyond the scope of this work, but some of its features are relevant to understand the association between EBV and autoimmunity [55,56,57,58]. Briefly, EBV is characterized by a peculiar ability to shift between an active lytic cycle and a latent state from which it can reactivate, depending on its interaction with host immune cells [59]. A second key feature is that the virus can deeply alter physiological B cell differentiation to determine life-long infection in memory B cells [58,59,60].
These two phases are characterized by the expression of different viral proteins, some of which are involved in immune system evasion and autoimmunity induction (Table 3):
(a)
Lytic phase. It occurs during primary infection and subsequent EBV reactivations from the latent state. The viral DNA is replicated and most viral genes are expressed, allowing spread to other cells. Key lytic phase proteins are ZEBRA protein (BZLF-1), which can deregulate immune surveillance [61]; an IL-10-homologue virokine, which inhibits IFN-γ production, CD8+ cytotoxic T-cells and MHC-I expression [62]; restricted early antigen (EA/R), a viral homologue of Bcl-2 oncogene which makes infected epithelial and B-cells resistant to apoptosis [63].
(b)
Latent phase. It develops after primary infection, as a result of the host’s EBV-specific immune response. The expression of latent phase viral proteins is epigenetically controlled and results in an almost complete silencing of the EBV genome in memory B lymphocytes [64,65], thus avoiding recognition by T cells. Only a few genes are expressed [66], coding for three latent membrane proteins (LMPs)—LMP-1, LMP-2A and LMP-2B—and 5 EBV nuclear antigens (EBNA)—EBNA 1, -2, -3A, -3B and -3C. This limited set of proteins is critical for self-reactive memory B lymphocyte “immortalization”, autoimmunity induction [14] and EBV-driven oncogenesis [59].
LMP-1 and LMP-2 both help EBV gain access to the memory B cell pool and act as survival signals for EBV-infected lymphocytes, mimicking signals from CD40 transduction pathway and antigen-binding to B-cell receptor (BCR), respectively [66,67].
LMP2-A has been associated with the evasion of affinity-based selection in germinal centers [67].
LMP-1 has more complex functions, highly relevant to autoimmunity: it induces BAFF (B-cell activating factor of the tumor necrosis family), which rescues self-reactive B cells, it can determine a SLE-like form in transgenic mice [77] and has been associated with type I IFN pathway activation in SLE [68]. Of interest is the fact that there is a mutual interaction between LMP-1 expression and IFN-α levels. On the one hand, EBV-DNA and RNA can induce the production of IFN-α by pDCs through TLR 9- and 7-mediated stimulation of LMP-1 [69,70,71]; on the other hand, IFN-α and other proinflammatory cytokines can induce LMP-1 expression in EBV-infected B-cells. Recent evidence has also highlighted the role of LMP-1 in inducing Programmed Death-Ligand 1 (PD-L1) overexpression in EBV-infected cells such as neutrophils [35]. Taken together, these data highlight the involvement of LMP-1 in key pathways modulating SLE activity: IFN-α, TLRs and PD-1/PD-1L system (Figure 1).
An increased expression of LMP-1 and EBV-encoded RNA-1 (EBER-1) has also been found in the renal tissue of patients with LN as compared to HCs and minimal change nephropathy patients [72] and a correlation between the degree of LMP-1 expression and the histological severity of LN has been suggested [73].
EBNA proteins also play a key role in EBV latent phase and autoimmunity induction.
EBNA-1 is the only viral antigen which acts as a replication factor during latent infection and it can be the only EBV protein expressed in resting B cells. It is not degraded by proteasome and consequently cannot be presented at the cell surface, becoming invisible to immune system. EBNA-1 cross-reacts with several self-antigens and can induce autoimmunity through molecular mimicry with C1q and epitope spreading [74,75].
EBNA-2 controls the expression of all other latent viral genes and blocks replication in most EBV-infected cells. Genomewide association studies (GWAS) have shown a high number of associations (>2.200) between hundreds of transcription factors (TFs) and many of the genetic loci increasing the risk of SLE. Of interest is the fact that around 50% of these loci can be occupied by EBNA-2, suggesting a dominant role of this protein in modulating susceptibility not only to SLE but also to other AIDs, such as rheumatoid arthritis (RA) and multiple sclerosis (MS), which could be classified as “EBNA-2 disorders” [76].

Alterations of EBV-Specific Immune Response in SLE

Peculiar alterations in EBV-specific immune response are found in SLE (Table 4).
Both lytic and latent EBV proteins elicit a strong T- and B-cell response. The expansion of EBV-infected B cells is physiologically controlled by CD8+ cytotoxic T lymphocytes (CTL). EBV-specific CD8+ CTL are instead dysfunctional in SLE and exhibit an abnormally reduced production of cytokines when stimulated with EBV [78,79,80]. This inadequate response favors recurrent lytic EBV replication, resulting in a chronically increased blood viral load and enhanced cell death [75,81,82]. T-cell exhaustion due to increased expression of PD-1 and PD-1L is probably an important mechanism through which EBV persistent infection can blunt CTL response and evade immune control [83]. A decreased Th17 and Treg response may be another key EBV-induced alteration [84].
Persistently active viral cycles increase the risk for both EBV-associated malignant transformation and AID in genetically predisposed individuals. An inverse correlation exists between impaired function of EBV-specific CTL, serum levels of antibodies against lytic antigen (anti-EA/D) and disease activity (SLEDAI)/autoantibodies [78,79,80]. This reflects an unbalanced B/T cell-mediated response towards EBV and other viruses, predisposing the host to an enhanced frequency of EBV reactivations and autoimmunity induction.
The impaired control of EBV by CTL and persistent latent EBV infection constitute the primum movens of a broad spectrum of immunological alterations which characterize SLE (Table 4).
The most immediate one is that elevated EBV viral loads are found in peripheral B cells [85] and in PBMC of SLE patients, with a 10-to 40-fold increase compared to HCs, regardless of immunosuppressive therapy [82,86,87].
Furthermore, the aberrant expression of key viral mRNAs related to proteins in both the lytic (BZLF-1, BLLF-1) and latent phases (LMP-1, LMP-2 and EBNA-1 in PBMCs) suggests intense EBV reactivation and enhanced survival of EBV-infected lymphocytes [85,88].
Persistently active EBV infection due to inefficient T cell response appears to be associated with a shift towards an amplified humoral response against EA/D and other viral antigens [63,80,89]. Several features of this abnormal humoral response to EBV are typical of SLE (Table 4).
First of all, the seroprevalence of anti-EBV antibodies is very high in SLE [90,91,92]. Two recent metanalysis have confirmed a strong association between EBV seropositivity (EBNA-1 and VCA IgG) and SLE onset [93,94], indicating that prior EBV infection is a prerequisite for developing SLE, at least in some ethnic groups.
Second, the humoral response is qualitatively different from that of HCs in several respects. These peculiarities were demonstrated in the clinical studies summarized in Table 4.
Among these, it is interesting to note that the prevalence of an IgA response against EBV antigens is elevated in SLE patients compared to HCs and also other AIDs [99,100].
Furthermore, the coexistence of multiple antibody isotypes against EA/D is significantly more frequent in SLE. In a study by Draborg, 65% of SLEs tested positive for two or more isotypes of antibodies against EA/D, whereas none of the HCs were positive for three isotypes, only 10% were positive for two isotypes and 65% of them had no antibodies against EA at all [89]. These alterations likely reflect the chronic production of antibodies against cells expressing EA/D, EBV-infected cell lysis and the release of EA/D and intracellular antigens. Depending on the type of infected cell, this could result in the enhanced production of either IgA (epithelial cells) or IgG (lymphocytes) against EA/D, in addition to autoantibodies (EA/D is bound to dsDNA, functioning as EBV-DNA polymerase). Thus, the presence of multiple anti-EA/D antibody isotypes could reflect a more disseminated infection, with a higher risk of cell lysis and autoimmunity induction.
In conclusion, the role of EBV in the pathogenetic mechanisms of SLE is supported by very high seroprevalence of antibodies against EBV, increased viral loads and peculiar alterations of the EBV-specific immune response. Overall, these data indicate that previous exposure to EBV represents a triggering factor for autoantibody generation and clinical disease onset in genetically predisposed individuals.
The presence of EBV proteins in renal tissue from patients with LN confirms direct involvement of this virus in organ damage.

3.2. Parvovirus B19 (B19V)

B19V is a small single-strand DNA virus with a worldwide distribution. Its general features are outlined in Table 2. Although it usually has a benign, self-limiting course, B19V proteins can induce SLE and other AIDs [106,107].
Most viral proteins are involved in inflammatory and autoimmune disorders:
  • VP-1, which includes a “VP-1 unique region” (VP-1u) characterized by a phospholipase A2-like activity, probably playing a role in viral entry into the cell [108] and the generation of inflammatory mediators (leukotrienes and prostaglandins) and phospholipids epitopes, potentially triggering antiphospholipid (aPL) antibodies [109]. Of note is the fact that VP1-unique region contains many epitopes recognized by neutralizing antibodies, which determine life-long protection [106].
  • NS-1, a cytotoxic, nonstructural protein which can transactivate several promoters, also enhancing retroviral replication, induces IL6 gene expression and stimulates polyclonal B-cell activation [110]; the expression of NS-1 probably marks complicated or chronic B19V infections [111].
B19V has been associated with immune-mediated manifestations such as different types of glomerulonephritis (GN) (mainly acute postinfectious, membranoproliferative, SLE-like GN and minimal change disease) [112,113]. However, B19V seroprevalence among patients who undergo kidney biopsy is similar to the overall population and the mechanism of renal damage is still undefined, although it is likely mediated by endothelial injury [113].
As for systemic autoimmunity, acute B19V infection can transiently mimic full-blown SLE (facial rash, photosensitivity, acute polyarthritis, cytopenia) [114] or even trigger or exacerbate full SLE [115,116] and can be accompanied by a broad spectrum of autoantibodies, including antinuclear antigen antibodies (ANA), anti-dsDNA, rheumatoid factor and APL antibodies [115,117,118].
The abnormal apoptosis of B19V infected bone marrow (BM) erythroblast, with consequent production of many altered self-antigens, has been proposed as a possible mechanism favoring these autoimmune processes [119]
A specific aspect is B19V’s role in antiphospholipid syndrome (APS) pathogenesis. APS refers to the association of thrombosis and/or pregnancy morbidity with antiphospholipid (aPL) antibodies and can be either primary or develop within the setting of SLE (secondary APS) [120].
Although the transient appearance of APL antibodies is in general quite common during infections, the association between B19V, isolated APL antibodies (Lupus Anticoagulant or LAC, anti-β 2-glycoprotein I, anticardiolipin) and even full-blown APS has been consistently reported over the last two decades [121,122].
In a recent study by Hod T. et al. a correlation between viral serology and clinical and serological parameters was ruled out in the general SLE population, but a higher B19V seroprevalence (both IgG and IgM) was found in a subset of SLE patients with APS. These patients also had a higher titer of IgG antibodies to B19V compared to those without APS, suggesting a link between previous viral exposure and APS [42]. The role of protein VP1u as an antigen triggering APS-like syndromes has been established [109].
Recent studies also indicate that B19V is associated with a specific SLE complication, dilated cardiomyopathy (DCM), probably through an increase of Th-17-related cytokines, as prevalence of anti-B19-NS1 and anti-B19-VP1u IgG was significantly higher in patients with DCM than in the general SLE population [123].
In conclusion, although no study has formally proved a direct causal role of B19V in AIDs so far, as available evidence is largely based on small case series [109], there appears to be sufficient epidemiological evidence to conclude that B19V infection can trigger SLE and modulate its clinical expression [124]. The pathogenetic mechanisms of B19V infection (the promotion of erythroblast apoptosis and the expression of viral proteins VP-1 and NS-1, both characterized by important immunomodulatory properties) support its role as an inducer of a broad range of autoimmune phenomena, ranging from isolated autoantibodies to definite SLE and APS.

3.3. Retroviruses

Both endogenous and exogenous RVs (Table 2) can stimulate the immune system and have been associated with SLE. Three elements indicating a relationship between RVs and SLE have long been recognized [125,126]:
(a)
The presence of retroviral-like particles in the tissues of SLE patients;
(b)
The presence of antiretroviral antibodies in an important proportion of SLE patients;
(c)
Clinically overlapping features between retroviral infections and SLE.
However, the relationship between RVs and SLE is multifaceted and must be differentiated according to the type of RV. While the association with HTLV-1 is still undefined and debated, being largely based on small studies or case-reports, the one between HIV and SLE is definite and plausibly supported by clinical and translational elements.
As for HERV, a large body of evidence points to an important role in mechanisms of autoimmunity, ranging from molecular mimicry to the activation of intracellular sensors by viral RNA and epigenetic modulation of immune response genes.
We will therefore separately analyze the different types of RVs.

3.3.1. Human Endogenous RVs (HERVs)

Up to 200 families of HERVs have been described and some of them (HRES-1, ERV-3, HERV-E 4-1, HERV-K10 and HERV-K18) have been especially implicated in SLE [54,127,128,129,130].
HERVs are ancient RVs which are stably integrated in host mammalian genome. Endogenous retroviral sequences (ERS) account for up to 8% of it and probably represent the remnants of exogenous RVs which integrated into the genome and became trapped during evolution, due to mutations of essential genes [54,131]. Environmental factors (such as infections, estrogens, drugs/chemicals, UVB exposure) can epigenetically reactivate the transcription of HERV-related sequences. Hence, HERV may represent the missing molecular link between the genetic predisposition to autoimmunity and external triggers [25]. Epigenetic changes are especially important as transducers of external stimuli: the genome is overall undermethylated in SLE, causing increased transcription of IFN-responsive genes and related ERSs [132].
While exogenous RVs are infectious and require a replication cycle to integrate proviral DNA into the host genome, endogenous RVs are genetically transmitted through the germline as proviral DNA. Despite some beneficial effects (e.g., the epigenetic modulation of immune response genes to fight exogenous viruses), HERV elevated transcription and anti-HERV antibody reactivity are strongly implicated in SLE pathogenesis [54,125,126]. GWAS has identified 124 unique ERV loci that are significantly more expressed in the PBMCs of SLE patients [133]. For example, HTLV-1-related ERS (HRES-1) is integrated at chromosome 1q42, a SLE-susceptibility locus, and codes for HRES/Rab4, a GTPase markedly overexpressed in T cells of SLE patients [134].
HERVs can trigger autoimmunity through several mechanisms [125,126]:
(a)
They produce neosynthesized viral antigens, which can stimulate autoimmune response through the molecular mimicry of self-proteins, determining the production of polyclonal antibodies and nephritogenic immunocomplexes; in some cases, they act as superantigens.
(b)
They can directly stimulate intracellular sensor molecules (e.g., TLRs) through their NAs, triggering inflammatory cascades.
(c)
They promote the transcription of IFN-γ-related and other immune-related genes. This epigenetic control is often mediated by HERV retroviral long terminal repeats (LTRs) domains, which are co-opted by their mammalian hosts as specific regulatory sequences.
Over the last decade, significant evidence has been accumulating on each of these mechanisms, which we will analyze in more detail.
(a)
Molecular mimicry and production of pathogenic antibodies.
HERV proteins are characterized by strong cross-reactivity to self-antigens and molecular mimicry is well documented [99]. For example, HTLV-1-related endogenous sequence (HRES-1), can code for HRES-1/p28, a 28-kDa nuclear autoantigen targeted by cross-reactive antiviral antibodies [135].
This protein is characterized by a sequence which harbors a triplet of highly charged amino acids, which is also found in the gag-like region of 70K U1snRNP SLE autoantigen. These cross-reactive epitopes can drive the formation of antinuclear autoantibodies [129]. Furthermore, HRES-1/Rab 4 overexpression interferes with endosomal recycling of CD3/TCRζ chain in lupus T cells, determining a critical loss of expression of this receptor, and lowering T-cells’ threshold for activation (“lupus T cells”) [136,137].
In addition to exerting molecular mimicry, viral antigens can also trigger the production of antibodies and immune complexes, directly contributing to organ damage. Anti-HERV-K envelope antibodies have been identified in SLE and the envelope glycoprotein 70 (gp70) and viral NA-protein complexes are targeted by antibodies as a result of TLR-7 activation. Retroviral gp70-anti-gp70 immune complexes are nephritogenic in murine models of LN [138]. The synergistic effect of three different gp70 loci (Sgp3,4 and 5) enhances the expression of HERVs and of this protein [139] and germline deletions of SLE. Susceptibility Locus Sgp3, which encodes the HERV suppressors named “SNERV” 1 and 2, indirectly determines autoantigen gp70 overproduction and LN in SLE-prone mice [140].
Furthermore, the antibody-induced extensive internalization of retroviral envelope glycoproteins initiates signaling cascades leading to the activation of lymphocytes in a murine model [141].
  • (b) Direct stimulation of intracellular sensor molecules by viral NAs.
Several NA-sensing systems are highly expressed in SLE PBMCs and can be activated by RVs RNA. Upon binding with viral NA they trigger a variety of pathways, all converging on type I IFN response [32].
RVs can be detected by several systems such as endosomal TLRs, which can recognize viral dsRNA (TLR-3) and ssRNA (TLR-7 and -8), in addition to dsDNA (TLR-9) [142].
The NAs of RVs can also sensitize other PRRs in the cytosol besides TLRs in the endosomes [143] (Figure 2).
While NA-sensing system is physiologically required to control viral infections, an excessive activation can drive SLE progression by enhancing the production of key cytokines, such as IL17A [144]. For example, a deficit in apoptosis can favor cell engulfment with HERV-derived DNA and abnormally amplify this process [125,145].
  • (c) Epigenetic control of host immune genes and other mechanisms of immunomodulation.
HERV sequences coding for viral genes gag, pol and env are flanked by long terminal repeats (LTRs).
LTRs are often present as solitary sequences, spread in high numbers of copies throughout the human genome and likely control the transcription of adjacent genes. A part of them have preserved regulatory activity, as they contain promoter/enhancer sequences which generally have a minor impact on gene transcription. However, alterations in the epigenetic mechanisms which normally control them can pathologically increase their activity. This epigenetic reset can occur in autoimmunity and cancer and promote the transcription of LTR-dependent immune genes and the reactivation of HERV. Exogenous viral infections (HIV, HTLV-1, influenza A virus, herpesviruses) can also rescue HERVs from their inactive status [125].
For example, MER 41, an LTR belonging to a family of HERVs, is a unique STAT1-binding site near the IFN-γ-inducible Absent in Melanoma 2 (AIM2) gene and acts as its promoter. AIM2 codes for a protein which is a key foreign DNA sensor, connected to innate response (AIM2 inflammasome) and SLE pathogenesis (Figure 2). Thus, uncontrolled retroviral regulatory sequences can represent a reservoir of IFN-inducible enhancers [131].
The role of different LTRs in modulating expression of both HERV genes and their molecular targets is further underlined in another setting. Increased HERV-E clone 4-1 mRNA expression was found in SLE CD4+ T-cells and PBMCs, secondary to a variety of factors: activation of Ca++/calcineurin (CaN)/Nuclear factor of activated T cells 1 (NFAT1), due to UVB or foreign infections with high IL6 and TNFα levels; Estrogen receptor-α (ER-α) signaling pathway; abnormal DNA hypomethylation of HERV-E clone 4-1 5’LTR. Once expressed, this HERV clone 3’LTR restrains miR-302d activity (an inhibitory miRNA, abnormally downregulated in SLE monocytes), activating a cascade which leads to DNA hypomethylation, IL17 release from CD4+ T-cells and type I IFN response [146,147,148]. Increased expression of HERV-E clone 4-1 gag transcripts has been associated with immunological activity and anti-U1RNP and anti-Sm autoantibodies [149]. This cascade is outlined in Figure 2.
Overexpression and LTR polymorphisms of another viral protein, HRES-1/Rab4, which regulates the surface expression of CD4 on T cells through increased lysosomal degradation of CD4 and CD3/TCRζ, have been associated with T cell activation upon TCR stimulation. Similar to HERV-E clone 4-1, also HRES-1/Rab4 expression is induced by UVB, Estrogen and even by TAT protein of HIV-1, suggesting interactions between different RVs [129,134].
Another modality of the epigenetic modulation of immune gene expression occurs through Long Interspersed Nuclear Elements (LINEs), retrotransposable elements coding for endonuclease and retrotranscriptase. Due to their autonomous capacity to replicate and shift along the genome they can control in cis expression of adjacent key genes (innate immunity, apoptosis), in a similar way to intragenic LTRs [125]. Furthermore, their DNA and RNA transcripts can activate TLR-7 and -9, initiating pathways which converge on IFN gene transcription, especially in LN [150]. LINEs sequences are also derepressed by hypomethylation, as shown in SLE neutrophils and PBMC, and this process correlates with immunological activity [151,152].
In addition to epigenetic control, HERV can exert immunomodulation through proteins with superantigen properties. For example, the env protein of HERV-W (syncytin) and HERV-K18.1 may act as superantigens and strongly stimulate T lymphocyte proliferation [153].
However, HERVs may also play an immunosuppressive role in SLE. A HERV locus coding for envelope protein 59 negatively correlates with interleukin-6 (IL6) and TLR-7 expression. This protein contained an immunosuppressive domain (ISU) which showed strong anti-inflammatory properties in animal models and also in ex vivo experiments in SLE patients, suggesting a potential therapeutic application [154,155,156].

3.3.2. Exogenous RVs: Human T-Cell Lymphotropic Virus Type I (HTLV-1) and Human Immunodeficiency Virus (HIV)

Exogenous retroviruses, like HIV and HTLV-1, have been both associated with SLE-like manifestations or to a fair SLE diagnosis. Molecular mimicry has long been proposed as a pathogenetic mechanism, as cross-reactivity of HTLV-1 and HIV gag antigens with self-proteins exists.

HTLV-1

HTLV-1 can determine adult T cell leukemia/lymphomas and HTLV-1–associated myelopathy/tropical spastic paraparesis (HAM/TSP), which has been associated with several AIDs including SLE [51,157].
Anti-HTLV-1 antibodies have been detected in variable proportions of SLE patients and a single study suggested that seropositive patients could have a later-onset, milder disease [158].
HTLV-1 has the potential to induce SLE through molecular mimicry between a 28Kda protein coded by HRES-1, which acts as a nuclear autoantigen, and HTLV-1 gag p24 protein [159].
In addition, HTLV-1 codes for several regulatory proteins which modulate inflammatory pathways upregulating T CD4+ Th1 lymphocytes and downregulating Treg [51].

HIV

Epidemiological data suggest an association between HIV and SLE. In a large nationwide French study, a wide spectrum of autoimmune manifestations has been reported to occur during HIV infection, with an overall prevalence of 4.13%; they mainly included arthralgias or arthritis, psoriasis, sarcoidosis, RA and ankylosing spondylitis, while the prevalence of SLE was lower. These disorders mainly occurred in patients on antiretroviral therapy with undetectable HIV viremia, on average more than 10 years after HIV diagnosis [160]. Full-blown SLE appearing in HIV-positive patients, mostly Black Africans, have also been reported in smaller studies [161,162].
Furthermore, “LN-like” proliferative GN has been described as part of the spectrum of HIV-associated nephropathy (HIVAN). The availability of HAART may have contributed to the moderate prevalence of these manifestations over the last few decades and to remission induction of SLE-like GN in some cases [154,163].
Several other elements also support the plausibility of an association between HIV and SLE.
These two conditions share common clinical and laboratory manifestations and are both characterized by an activation of type-I IFN-mediated response, which is essential to counteract HIV infection but also represents a molecular “signature” of SLE. In HIV polyclonal proliferation of B cells is associated with hypergammaglobulinemia and production of broadly neutralizing antibodies against Env epitopes. Some of them, such as CH98, cross-react with ds-DNA. Interestingly, the subset of anti-dsDNA-positive HIV patients appears to be less prone to AIDS progression, suggesting that an amplified humoral response may be protective, at the price of an increased risk of inducing autoantibody production [164]. Anti-Sm and, more commonly, ANA and anti-aPL antibodies are also found in HIV [165].
Another shared key alteration is an impaired autophagy, with consequent accumulation of senescent or damaged cell components. Autophagy is a crucial mechanism to control HIV infection, through stimulation of TLR-7 and TRL-9 and activation of intracellular inflammatory pathways; however, HIV proteins can blunt this process to prevent clearance of virions, thus creating a predisposing condition for autoantigen exposal and SLE induction [166,167]. Of interest is the fact that the mammalian Target Of Rapamycin (mTOR) inhibitor rapamycin induces autophagy, with beneficial effects on both conditions [168].

3.4. Torque Teno Virus (TTV)

TTV is single-stranded circular DNA Anellovirus, originally named after the initials of the first patient (TT), then “transfusion-transmitted” (it was initially associated with posttransfusion hepatitis) and, more recently, “Torque Teno” virus (Table 2). It is considered a commensal human virus, ubiquitously infecting human populations by early infancy and has not been causally associated with any disease until recently.
However, increased TTV load is common in AIDS patients and immunosuppressed solid organ transplant (SOT) recipients and this virus is also probably involved in SLE pathogenesis, even though Anelloviruses may represent a generic marker of immune dysfunction and immunodeficiency [44,169,170].
TTV-DNA was actually detected more frequently in SLE patients than in HCs and RA [129]. TTV PCR-positive SLE patients have a significant prevalence (21%–52%) of antibodies against HRES-1/p28 and molecular mimicry between this antigen and 70KU1snRNP may contribute to the generation of nuclear autoantibodies [159]. In addition, HRES-1/p28 cross-reacts with TTV peptide ORF2a [43,44,135] and also with EBV antigens (EBV-LF3 and EBNA 3 C) [171,172].
Finally, TTV-DNA could also activate innate immunity through TLR-9 [169].

3.5. Cytomegalovirus (CMV)

Human CMV is a member of the β-subgroup of the herpesvirus family, commonly acquired during childhood, with absent or self-limiting symptoms (Table 2). It is characterized by several features which can explain its involvement in autoimmunity and are similar to those of EBV: lytic replication in multiple tissues, lifelong persistence through periods of latency and intermitting reactivation, extensive manipulation of adaptive and innate immunity [173,174,175] (Figure 1).
A causal relationship between CMV and SLE and other AIDs is suggested by the literature. However, a clear association of CMV seroprevalence and disease has not been firmly established yet. Growing evidence suggests that CMV can have multiple interactions with the host immune system and could play a contributive role in flareups and possibly in conferring SLE specific-clinical features, such as associated secondary APS and Raynaud’s phenomenon. Population-based, long-term prospective studies are needed to better elucidate its impact on disease course [174].
Some key viral proteins mediate immunopathogenic mechanisms of CMV-induced autoimmunity:
  • Phosphoprotein 65 (pp65422–439). This protein contains an epitope region which has a homology with TATA-box binding protein associated factor 9 (TAF9134–144). Consequently, antibodies against pp65422–439 cross-react with TAF9134–144 and also with ANA and anti-dsDNA in immunized BALB/c mice, in which they determine typical histological LN lesions. Furthermore, SLE patients appear to have higher anti-TAF9 antibody titers compared to HCs. Overall pp65 acts can induce autoantibodies production in susceptible animals [176,177].
  • UL44 is nonstructural intranuclear protein which is essential for CMV replication. A human monoclonal antibody from CMV seropositive donor immunoprecipitates UL44 with other SLE nuclear antigens including nucleolin, dsDNA and ku70. UL44 appears to be relocated at cell surface complexed with these molecules during CMV-induced apoptosis, suggesting a mechanism of bystander autoantigen presentation [178].
  • US31 is highly expressed in PBMC of SLE patients and can skew macrophage differentiation toward an M1 phenotype, activating inflammation through a direct interaction with NF-κB2 [179].
Potential cross-reactivity of CMV-specific T cells with La (SS-B) protein, a ribonucleoprotein associated with RNA polymerase, has been shown in childhood onset SLE [104,180].
Furthermore, CMV has the potential to extensively manipulate host T cell function during active infection. In vitro studies showed that CMV antigens can increase CD4+/CD8+ T-cell ratio in SLE PBMCs and their IL-4 and IFN-γ production, driving the development of a large pool of memory T-cells which can facilitate autoimmunity [181].
However, other studies showed that the dysfunctional EBV-specific CD4+ T-cell response observed in SLE patients does not seem to apply to CMV to the same extent [182] and that CMV-specific CD8+ T-cell responses appear preserved [183]. Overall, these data suggest that CMV probably plays a less important role in the development or exacerbation of SLE than EBV. Although the SLE-intrinsic dysregulation in B- and T-lymphocytes’ responses is the main reason for skewed EBV and CMV-specific T response [137], the intensity of immunosuppressive therapy also crucially modulates the risk of herpesviruses reactivation [184].
A peculiar aspect of CMV is that it can drive expansion of a highly cytotoxic, immunosenescence-associated subset of CD4+ T-cells lacking CD28 (CD28nullCD4+). This T cell subset is resistant to CD4+CD25+T reg-mediated inhibition and to apoptosis and is probably mainly CMV-specific. While its frequency is usually below 1% even in very old people, CMV infection can increase it more than 10-fold. The accumulation of CMV-specific CD28null infiltrates was demonstrated in disease affected tissues in SLE and other AIDs and has also been associated with atherosclerotic vascular complications in RA [185].
Finally, due to its tropism for endothelial cells, CMV has been implicated in the pathogenesis of vascular damage in systemic sclerosis (SSc), through molecular mimicry of normally expressed endothelial cell surface molecules [186], Raynaud’s phenomenon [187] and APS [188,189].

3.6. Other Viruses

We will separately analyze other RNA- and DNA-viruses potentially associated with SLE.

3.6.1. HCV, Measles, Influenza A and Dengue Virus

HCV is a hepatotropic RNA-virus supposed to have an additional extrahepatic tropism, including that for B lymphocytes. This virus determines a chronic infection in 70% of patients, evading innate and adaptive immunity and promoting B lymphocyte proliferation and a low-grade, chronic systemic inflammation. HCV strategies to achieve viral persistence include: hijacking cellular factors such as miRNA that bind its genome protecting it from degradation, inhibiting NA detection by innate immunity sensors, limiting type I IFN response activation and repressing viral transcription and replication, thus allowing host cell survival [28,190].
Chronic HCV infection has been associated with a large spectrum of extrahepatic autoimmune manifestations, including cryoglobulinemic vasculitis, Sjögren’s syndrome, RA and SLE. Direct-acting antiviral agents and new B-cell-depleting or B-cell-modulating monoclonal antibodies will probably expand treatment options for HCV-related AIDs [191].
In a recent study on patients with SLE, RA and systemic sclerosis, IgG-seropositivity for measles virus (MeV) was associated with significantly higher titers of natural autoantibodies against citrate synthase, which have a modulating effect on the immune system [192].
A recent Korean study showed a significant association between seasonal influenza infection and flares in SLE patients [193]. A massive peptide overlap between five common human viruses (Influenza A virus, Borna disease virus, MeV, mumps virus and rubella virus) and human proteome emerged. Sequence similarity may explain autoimmune cross-reactions during immune responses directed towards viral infections or due to immunizations [194]. Furthermore, Influenza A virus can also rescue HERVs from their inactive status [125].
The relationship between Dengue virus (DENV) and SLE is complex and still remains to be elucidated. On the one hand, DENV has been anecdotally reported to trigger SLE and LN [46]; on the other hand, IgG that can cross-neutralize DENV are often found in SLE sera, probably explaining rarity of severe forms in SLE patients [195].

3.6.2. Human Papillomavirus

Human Papillomavirus (HPV) is a dsDNA virus which has recently been proposed to play a role in SLE pathogenesis on the basis of several elements: HPV seroprevalence is increased among SLE patients, SLE development post-HPV vaccination has been reported and a large peptide overlap has emerged between HPV L1 proteins and many human proteins such as lupus Ku autoantigen proteins p86 and p70, lupus brain antigen 1 homolog and others. This further supports the hypothesis of cross-reactivity as primum movens for SLE onset and the concept of SLE as “an autoimmune mosaic syndrome” [196].

4. Future Perspectives

The expanding knowledge of the role of viral infections or reactivation as a trigger for AID onset or as modifiers of disease severity and course is paving the way for alternative approaches in the management of SLE. As viral infections and autoimmunity share activation of overlapping pathogenetic pathways, there is a rationale in investigating the use of antiviral drugs as part of SLE therapy, along with modulation of virally induced immune response. Progress in these research areas will be analyzed. Although most data are preliminary and deserve to be confirmed, they represent frontiers which may lead to new therapeutic, diagnostic, and prognostic tools.

4.1. Anti-EBV Therapies

An interesting potential therapeutic strategy could be that of modulating anti-EBV response. This approach has been investigated mainly in the setting of EBV-associated cancer, with the rationale of exploiting EBV presence in tumor cells to target them selectively [197,198]; however, it could also open new perspectives on AIDs.
Inhibitors of EBV reactivation and replication include drugs already licensed for the treatment of other herpesviruses and nutritional constituents [199].
The adoptive transfer of EBV-specific transgenic T cells has been therapeutically explored with clinical success in EBV-associated malignancies [200].

4.2. Anti-B19V Therapies

New antiviral strategies have been explored in recent years, including: hydroxyurea; cidofovir and brincidofovir, nucleotide analogues both characterized by a broad-range activity against DNA-viruses; coumarin derivatives and flavonoid molecules [201].
B19V nuclear localization signals (NLSs), which are critical to guide the virus into the nucleus, are promising therapeutic targets. Therapeutic vaccines or antiviral drugs interfering with them could block infection [202].

4.3. Antiretroviral Therapies

Reverse transcriptase inhibitors represent the cornerstone of antiretroviral agents employed in HIV therapy. A large study on over 20,000 patients from Taiwan has shown that HAART therapy could protect from SLE onset [203] and blunt reactivation of some HERVs [204], thus interfering with an important mechanism of epigenetic modulation in SLE.

4.4. T Lymphocyte Modulating Therapies

Therapies targeting CD28null T cells, a subset strongly associated with CMV infection, appear promising as they seem to represent a common pathogenic mechanism shared by SLE and other AIDs [185]. For example, anti-TNFα and methotrexate therapy reduce expansion of CD28null CD4+ T-cells in RA and lipid-lowering rosuvastatin induces their apoptosis in patients with coronary heart disease. Antiviral ganciclovir has also proved effective in this regard in ANCA-associated vasculitis, reinforcing the concept that the subclinical reactivation of CMV is crucial in driving expansion of this T cell subset [205]. A randomized controlled proof-of-concept trial aimed at studying the immunomodulating effect of Valaciclovir in vasculitis is under way [206].
Another potential approach is based on the modulation of CD8+ T-cell exhaustion. This process can develop during chronic viral infection, due to persistent antigen stimulation and inadequate costimulation, and it prevents viral clearance (e.g., HCV) or adequate response to vaccination (e.g., Influenza). This process can be induced by the virus itself, such as in the case of EBV, as a tool to evade immune system [83]. On the other hand, T cell exhaustion is associated with better outcome in several AIDs [207].
Interestingly, kidney-infiltrating T cells in LN have a hypofunctional profile caused by an “exhausted” transcriptional signature, possibly representing a tissue defense mechanism to suppress T cell response [208].
A selective induction of this T cell phenotype might represent a new therapeutic tool to blunt autoimmunity, although its potentially detrimental effect on an associated chronic viral infection requires further study. Of interest is the fact that an anti-CD3+ therapy (teplizumab) to promote islet-specific CD8+ T-cell exhaustion showed a beneficial role in type 1 diabetes [209].

4.5. Inhibition of TLRs and Other Innate Immunity Modulators

Production of IFN-α and other cytokines by activated pDCs, a key process in SLE pathogenesis, can be determined by EBV-binding to class II MHC molecules, followed by endosomal uptake and TLR-9 (EBV-DNA) or TLR-7 engagement (EBV-encoded RNA, EBER) [69,70,71,144].
NA-sensing TLRs may therefore represent an interesting therapeutic target and oligonucleotide-based TLR antagonists have already proved capable of inhibiting EBV-DNA/RNA induced IFN-α production by human pDCs [145].
Another potential target could be Cathelicidin LL37, an antimicrobial and immunomodulatory peptide which can form complexes with the DNA and autoantibodies (LL37-DNA/anti-DNA). These are endocytosed by pDCs and recognized by TLR-9, leading to DC activation [210].

4.6. Inhibition of miRNAs

An interesting therapeutic target is represented by miR-30e. This is induced by HBV and other viral infections and targets multiple negative regulators of innate immune signaling, thus activating immune response. The inhibition of miR-30e in mouse models and also in SLE patients (for example with anti-HBV treatment) has significantly reduced type I IFN response [211].

4.7. Analysis and Modulation of Microbiota

Important research efforts focusing on how human commensal microbes trigger systemic autoimmunity have been made over the last decade [212]. Gut microbial composition and function are markedly altered in untreated SLE patients compared with HCs, having an autoimmunogenic and proinflammatory profile characterized by a consistently reduced Firmicutes-to-Bacteroidetes ratio [213,214].
Gut bacteria can either promote autoimmunity themselves through molecular mimicry, sharing protein epitopes with the Ro60 autoantigen or native DNA, or modulate the risk of autoimmunity-associated viral infections. For example, Staphylococcus Aureus colonization reduced the time to CMV acquisition in a prospective birth cohort followed up for 10 years [215]. On the other hand, microbiota has its own virobiota, in the form of bacteriophages which regulate microbiota composition, providing selective advantages to certain strains [216].
These data suggest that the interplay between infections and the immune system should be studied over time by defining individual exposure to bacterial and viral infections and their mutual interactions, long before the clinical onset of autoimmunity.
Finally, a recent study showed that PBMC virome of SLE patients has a significantly increased diversity compared to HCs and that viral taxa can reliably discriminate cases from controls. These SLE-associated virome signatures might represent a source of biomarkers to predict disease course and outcome [217].
Taken together, these data seem to provide a rationale for microbiome-modulating approaches as adjuvant therapies of AIDs [218].

4.8. Antiviral Vaccines

Another potential approach could be that of directly targeting replication of viruses involved in autoimmunity through vaccines. Vaccination against EBV or EBV-activated HERV in early childhood, at least in groups at high risk, could in theory help prevent the development of SLE at a later age, as epidemiological evidence suggests that a more advanced age of EBV infection might be associated with increased viral replication and risk of autoimmunity induction [219]. The availability of serological or genetic biomarkers to predict risk of evolution towards full-blown AID would be essential for this strategy. However, the vaccine itself could entail risks of autoimmunity if the patient has an intrinsic defect in suppression of postviral inflammation, and not only an abnormal viral replication. This concern has been limiting this approach. Reports of post-HPV vaccination autoimmune phenomena including SLE underlie the need for low-similarity peptide antigens in order to prevent the risk of adverse events triggered by immune cross-reactivity [220].

5. Conclusions

Viral infections represent in general a key environmental factor in SLE pathogenesis and can play an important role in triggering disease onset and flareups or in modulating clinical phenotype. Some viruses such as EBV, B19V and HERV can contribute to triggering SLE onset, whereas others, such as CMV, probably modulate disease course, having a more controversial role. Over the last decade, growing evidence has clarified a broad spectrum of viral immune-modulating mechanisms, which deeply alter the host immune system and interact with SLE-intrinsic alterations in B and T cell-mediated responses, as in the case of EBV-driven autoreactive B cell “immortalization”. Classical mechanisms of viral-triggered autoimmunity, such as molecular mimicry, have been further confirmed and a multitude of peptide overlap between viral and self-antigens has widened the concept of SLE as “an autoimmune mosaic syndrome”. Research on the epigenetic modulation of IFN-responsive genes by reactivated HERVs has shed light on this missing link between environmental stimuli and the genome. The relationship between microbiota, virobiota and SLE susceptibility genes is another area of ongoing research. New biomarkers and innovative therapeutic approaches may derive from this progress, including manipulation of innate immunity response, T-lymphocyte subpopulations, and epigenetic mechanisms of viral control on SLE susceptibility genes.
Even if their impact on autoimmunity has been recognized for a long time, viruses are acquiring an increasingly important role as new players in SLE. A better understanding of the interactions between viral proteins and NAs and the host immune system could help improve the management of this heterogeneous and challenging disease.

Authors Contributions

M.Q. and V.C. designed and wrote the initial manuscript; G.M. designed the Figures and Tables; C.B. and M.D.A. integrated the virological aspects; all authors critically revised, discussed and edited the article until it reached its current form. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partially funded by the Italian Ministry of Education, University and Research (MIUR) program “Departments of Excellence 2018–2022”, AGING Project—Department of Translational Medicine, University of Piemonte Orientale (UPO) and by local grants from the University of Piemonte Orientale (UPO, FAR) to M.Q. and V.C.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AIDautoimmune disorders
ADEantibody-dependent enhancement
ANAantinuclear antigens
APCantigen-presenting cell
aPLantiphospholipid
APSantiphospholipid syndrome
BAFFB-lymphocyte activating factor of the TNF family
BCRB-cell receptor
BMBone Marrow
BMTBone Marrow Transplant
cGAScyclic GMP-AMP synthase
CMVCytomegalovirus
CTLCytotoxic T lymphocytes
DAMPDamage Associated Molecular Patterns
DCDendritic Cell
DENVDengue Virus
dsDNAdouble-stranded DNA
EA/DDiffuse early antigen
EA/RRestricted early antigen
EBEREbstein-Barr encoded RNA-1
EBNAEbstein-Barr nuclear antigen
EBVEbstein-Barr Virus
ENV gp70Envelop glycoprotein 70
EREstrogen Receptor
ERSEndogenous Retroviral Sequence
GNglomerulonephritis
CVBCoxsackieviruses B (CVB)
GWASgenomewide association study
HAARTHighy Active Antiretroviral Therapy
HAM/TSPHTLV-1–associated myelopathy/tropical spastic paraparesis
HCVHepatitis C Virus
HERVsHuman Endogenous Retroviruses
HIVHuman Immunodeficiency Virus
HIVANHuman Immunodeficiency Virus Associated-Nephropathy
HPVHuman Papillomavirus
HRES-1HTLV-1-related endogenous sequence
IFNInterferon
IMInfectious Mononucleosis
IVIgIntravenous Immunoglobulins
LACLupus Anticoagulant
LINEsLong Interspersed Nuclear Elements
LNLupus Nephritis
LTRLong Terminal Repeat
MAVSmitochondrial antiviral signaling protein
MDA-5melanoma differentiation associated gene 5
MHCmajor histocompatibility complex
MeVmeasles virus
MSMultiple Sclerosis
NANucleic Acid
NFATNuclear Factor of activated T cells
NLSNuclear Localization Signal
miRNAmicroRNA
mTORmammalian Target of Rapamycin
PAMPsPathogen Associated Molecular Patterns
PBMCperipheral blood mononuclear cell
pDCplasmacytoid dendritic cell
PD-1LProgrammed Death Ligand-1
pp65422–439phosphoprotein 65422–439
PRRsPattern Recognition Receptors
B19VParvovirus B19
RARheumatoid Arthritis
RIG-Iretinoic acid-inducible gene-I
RLRsRIG-I-like receptors
RVsRetroviruses
SLELupus Erythematosus Systemicus
SOTSolid Organ Transplant
SScSystemic Sclerosis
TAF9134–144 TATA-box binding protein associated factor
T1Dtype 1 diabetes
TCRT-cell receptor
TFTranscription Factor
ThT helper
TLRToll-like receptor
T-regT-regulatory cells
VCAViral Capsid Antigen
VP-1uViral Protein-1 unique region

References

  1. 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]
  2. Aringer, M.; Johnson, S.R. Classifying and Diagnosing Systemic Lupus Erythematosus in the 21st Century. Rheumatology 2020, 59, v4–v11. [Google Scholar] [CrossRef] [PubMed]
  3. Parikh, S.V.; Almaani, S.; Brodsky, S.; Rovin, B.H. Update on Lupus Nephritis: Core Curriculum 2020. Am. J. Kidney Dis. 2020, 76, 265–281. [Google Scholar] [CrossRef] [PubMed]
  4. Lateef, A.; Petri, M. Unmet Medical Needs in Systemic Lupus Erythematosus. Arthritis Res. Ther. 2012, 14 (Suppl. 4), S4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Macejova, Z.; Madarasova Geckova, A.; Husarova, D.; Zarikova, M.; Kotradyova, Z. Living with Systematic Lupus Erythematosus: A Profile of Young Female Patients. Int. J. Environ. Res. Public Health 2020, 17, 1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Esposito, S.; Bosis, S.; Semino, M.; Rigante, D. Infections and Systemic Lupus Erythematosus. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1467–1475. [Google Scholar] [CrossRef]
  7. Nelson, P.; Rylance, P.; Roden, D.; Trela, M.; Tugnet, N. Viruses as Potential Pathogenic Agents in Systemic Lupus Erythematosus. Lupus 2014, 23, 596–605. [Google Scholar] [CrossRef]
  8. Rigante, D.; Mazzoni, M.B.; Esposito, S. The Cryptic Interplay between Systemic Lupus Erythematosus and Infections. Autoimmun. Rev. 2014, 13, 96–102. [Google Scholar] [CrossRef]
  9. Rigante, D.; Esposito, S. Infections and Systemic Lupus Erythematosus: Binding or Sparring Partners? Int. J. Mol. Sci. 2015, 16, 17331–17343. [Google Scholar] [CrossRef] [Green Version]
  10. 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] [Green Version]
  11. Lossius, A.; Johansen, J.N.; Torkildsen, Ø.; Vartdal, F.; Holmøy, T. Epstein-Barr Virus in Systemic Lupus Erythematosus, Rheumatoid Arthritis and Multiple Sclerosis—Association and Causation. Viruses 2012, 4, 3701–3730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yuan, Q.; Xing, X.; Lu, Z.; Li, X. Clinical Characteristics and Risk Factors of Infection in Patients with Systemic Lupus Erythematosus: A Systematic Review and Meta-Analysis of Observational Studies. Semin. Arthritis Rheum. 2020, 50, 1022–1039. [Google Scholar] [CrossRef] [PubMed]
  13. Vassia, V.; Croce, A.; Ravanini, P.; Leutner, M.; Saglietti, C.; Fangazio, S.; Quaglia, M.; Smirne, C. Unusual Presentation of Fatal Disseminated Varicella Zoster Virus Infection in a Patient with Lupus Nephritis: A Case Report. BMC Infect. Dis. 2020, 20, 538. [Google Scholar] [CrossRef]
  14. Pan, Q.; Liu, Z.; Liao, S.; Ye, L.; Lu, X.; Chen, X.; Li, Z.; Li, X.; Xu, Y.-Z.; Liu, H. Current Mechanistic Insights into the Role of Infection in Systemic Lupus Erythematosus. Biomed. Pharmacother. 2019, 117, 109122. [Google Scholar] [CrossRef] [PubMed]
  15. Kanduc, D.; Shoenfeld, Y. From Anti-EBV Immune Responses to the EBV Diseasome via Cross-Reactivity. Glob. Med. Genet. 2020, 7, 51–63. [Google Scholar] [CrossRef]
  16. Dreyfus, D.H.; Farina, A.; Farina, G.A. Molecular Mimicry, Genetic Homology, and Gene Sharing Proteomic “Molecular Fingerprints” Using an EBV (Epstein-Barr Virus)-Derived Microarray as a Potential Diagnostic Method in Autoimmune Disease. Immunol. Res. 2018, 66, 686–695. [Google Scholar] [CrossRef] [Green Version]
  17. 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] [Green Version]
  18. Migliorini, P.; Baldini, C.; Rocchi, V.; Bombardieri, S. Anti-Sm and Anti-RNP Antibodies. Autoimmunity 2005, 38, 47–54. [Google Scholar] [CrossRef]
  19. Cornaby, C.; Gibbons, L.; Mayhew, V.; Sloan, C.S.; Welling, A.; Poole, B.D. B Cell Epitope Spreading: Mechanisms and Contribution to Autoimmune Diseases. Immunol. Lett. 2015, 163, 56–68. [Google Scholar] [CrossRef] [PubMed]
  20. 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]
  21. Barrat, F.J.; Su, L. A Pathogenic Role of Plasmacytoid Dendritic Cells in Autoimmunity and Chronic Viral Infection. J. Exp. Med. 2019, 216, 1974–1985. [Google Scholar] [CrossRef] [Green Version]
  22. Fitzsimmons, L.; Kelly, G.L. EBV and Apoptosis: The Viral Master Regulator of Cell Fate? Viruses 2017, 9, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zan, H.; Tat, C.; Casali, P. MicroRNAs in Lupus. Autoimmunity 2014, 47, 272–285. [Google Scholar] [CrossRef]
  24. Lanata, C.M.; Chung, S.A.; Criswell, L.A. DNA Methylation 101: What Is Important to Know about DNA Methylation and Its Role in SLE Risk and Disease Heterogeneity. Lupus Sci. Med. 2018, 5, e000285. [Google Scholar] [CrossRef] [Green Version]
  25. Wang, Z.; Chang, C.; Peng, M.; Lu, Q. Translating Epigenetics into Clinic: Focus on Lupus. Clin. Epigenetics 2017, 9, 78. [Google Scholar] [CrossRef] [Green Version]
  26. Long, H.; Yin, H.; Wang, L.; Gershwin, M.E.; Lu, Q. The Critical Role of Epigenetics in Systemic Lupus Erythematosus and Autoimmunity. J. Autoimmun. 2016, 74, 118–138. [Google Scholar] [CrossRef] [PubMed]
  27. Koyanagi, N.; Kawaguchi, Y. Evasion of the Cell-Mediated Immune Response by Alphaherpesviruses. Viruses 2020, 12, 1354. [Google Scholar] [CrossRef]
  28. Randall, R.E.; Griffin, D.E. Within Host RNA Virus Persistence: Mechanisms and Consequences. Curr. Opin. Virol. 2017, 23, 35–42. [Google Scholar] [CrossRef] [Green Version]
  29. Levet, S.; Charvet, B.; Bertin, A.; Deschaumes, A.; Perron, H.; Hober, D. Human Endogenous Retroviruses and Type 1 Diabetes. Curr. Diab. Rep. 2019, 19, 141. [Google Scholar] [CrossRef] [Green Version]
  30. Blackburn, K.M.; Wang, C. Post-Infectious Neurological Disorders. Ther. Adv. Neurol. Disord. 2020, 13, 1756286420952901. [Google Scholar] [CrossRef] [PubMed]
  31. Kontaki, E.; Boumpas, D.T. Innate Immunity in Systemic Lupus Erythematosus: Sensing Endogenous Nucleic Acids. J. Autoimmun. 2010, 35, 206–211. [Google Scholar] [CrossRef] [PubMed]
  32. Crowl, J.T.; Gray, E.E.; Pestal, K.; Volkman, H.E.; Stetson, D.B. Intracellular Nucleic Acid Detection in Autoimmunity. Annu. Rev. Immunol. 2017, 35, 313–336. [Google Scholar] [CrossRef]
  33. Hejrati, A.; Rafiei, A.; Soltanshahi, M.; Hosseinzadeh, S.; Dabiri, M.; Taghadosi, M.; Taghiloo, S.; Bashash, D.; Khorshidi, F.; Zafari, P. Innate Immune Response in Systemic Autoimmune Diseases: A Potential Target of Therapy. Inflammopharmacology 2020, 28, 1421–1438. [Google Scholar] [CrossRef]
  34. Smatti, M.K.; Cyprian, F.S.; Nasrallah, G.K.; Al Thani, A.A.; Almishal, R.O.; Yassine, H.M. Viruses and Autoimmunity: A Review on the Potential Interaction and Molecular Mechanisms. Viruses 2019, 11, 762. [Google Scholar] [CrossRef] [Green Version]
  35. Curran, C.S.; Gupta, S.; Sanz, I.; Sharon, E. PD-1 Immunobiology in Systemic Lupus Erythematosus. J. Autoimmun. 2019, 97, 1–9. [Google Scholar] [CrossRef] [PubMed]
  36. James, J.A.; Kaufman, K.M.; Farris, A.D.; Taylor-Albert, E.; Lehman, T.J.; Harley, J.B. An Increased Prevalence of Epstein-Barr Virus Infection in Young Patients Suggests a Possible Etiology for Systemic Lupus Erythematosus. J. Clin. Investig. 1997, 100, 3019–3026. [Google Scholar] [CrossRef] [Green Version]
  37. Arbuckle, M.R.; Gross, T.; Scofield, R.H.; Hinshaw, L.B.; Chang, A.C.; Taylor, F.B.; Harley, J.B.; James, J.A. Lupus Humoral Autoimmunity Induced in a Primate Model by Short Peptide Immunization. J. Investig. Med. 1998, 46, 58–65. [Google Scholar]
  38. Dostál, C.; Newkirk, M.M.; Duffy, K.N.; Palecková, A.; Bosák, V.; Cerná, M.; Zd’arský, E.; Zvárová, J. Herpes Viruses in Multicase Families with Rheumatoid Arthritis and Systemic Lupus Erythematosus. Ann. N. Y. Acad. Sci. 1997, 815, 334–337. [Google Scholar] [CrossRef]
  39. Britt, W.J.; Prichard, M.N. New Therapies for Human Cytomegalovirus Infections. Antivir. Res. 2018, 159, 153–174. [Google Scholar] [CrossRef]
  40. Chen, J.; Zhang, H.; Chen, P.; Lin, Q.; Zhu, X.; Zhang, L.; Xue, X. Correlation between Systemic Lupus Erythematosus and Cytomegalovirus Infection Detected by Different Methods. Clin. Rheumatol. 2015, 34, 691–698. [Google Scholar] [CrossRef]
  41. Hsu, T.C.; Tsay, G.J. Human Parvovirus B19 Infection in Patients with Systemic Lupus Erythematosus. Rheumatology 2001, 40, 152–157. [Google Scholar] [CrossRef] [Green Version]
  42. Hod, T.; Zandman-Goddard, G.; Langevitz, P.; Rudnic, H.; Grossman, Z.; Rotman-Pikielny, P.; Levy, Y. Does Parvovirus Infection Have a Role in Systemic Lupus Erythematosus? Immunol. Res. 2017, 65, 447–453. [Google Scholar] [CrossRef] [PubMed]
  43. Gergely, P.; Pullmann, R.; Stancato, C.; Otvos, L.; Koncz, A.; Blazsek, A.; Poor, G.; Brown, K.E.; Phillips, P.E.; Perl, A. Increased Prevalence of Transfusion-Transmitted Virus and Cross-Reactivity with Immunodominant Epitopes of the HRES-1/P28 Endogenous Retroviral Autoantigen in Patients with Systemic Lupus Erythematosus. Clin. Immunol. 2005, 116, 124–134. [Google Scholar] [CrossRef]
  44. Gergely, P.; Perl, A.; Poór, G. Possible Pathogenic Nature of the Recently Discovered TT Virus: Does It Play a Role in Autoimmune Rheumatic Diseases? Autoimmun. Rev. 2006, 6, 5–9. [Google Scholar] [CrossRef]
  45. Stölzel, U.; Schuppan, D.; Tillmann, H.L.; Manns, M.P.; Tannapfel, A.; Doss, M.O.; Zimmer, T.; Köstler, E. Autoimmunity and HCV Infection in Porphyria Cutanea Tarda: A Controlled Study. Cell. Mol. Biol. 2002, 48, 43–47. [Google Scholar]
  46. Talib, S.; Bhattu, S.; Bhattu, R.; Deshpande, S.; Dahiphale, D. Dengue Fever Triggering Systemic Lupus Erythematosus and Lupus Nephritis: A Case Report. Int. Med. Case Rep. J. 2013, 6, 71–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Rajadhyaksha, A.; Mehra, S. Dengue Fever Evolving into Systemic Lupus Erythematosus and Lupus Nephritis: A Case Report. Lupus 2012, 21, 999–1002. [Google Scholar] [CrossRef]
  48. Emlen, W.; Niebur, J.; Kadera, R. Accelerated in Vitro Apoptosis of Lymphocytes from Patients with Systemic Lupus Erythematosus. J. Immunol. 1994, 152, 3685–3692. [Google Scholar]
  49. Carter, C.J. Extensive Viral Mimicry of 22 AIDS-Related Autoantigens by HIV-1 Proteins and Pathway Analysis of 561 Viral/Human Homologues Suggest an Initial Treatable Autoimmune Component of AIDS. FEMS Immunol. Med. Microbiol. 2011, 63, 254–268. [Google Scholar] [CrossRef] [Green Version]
  50. Getts, D.R.; Chastain, E.M.L.; Terry, R.L.; Miller, S.D. Virus Infection, Antiviral Immunity, and Autoimmunity. Immunol. Rev. 2013, 255, 197–209. [Google Scholar] [CrossRef] [Green Version]
  51. Quaresma, J.A.S.; Yoshikawa, G.T.; Koyama, R.V.L.; Dias, G.A.S.; Fujihara, S.; Fuzii, H.T. HTLV-1, Immune Response and Autoimmunity. Viruses 2015, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  52. Pullmann, R.; Bonilla, E.; Phillips, P.E.; Middleton, F.A.; Perl, A. Haplotypes of the HRES-1 Endogenous Retrovirus Are Associated with Development and Disease Manifestations of Systemic Lupus Erythematosus. Arthritis Rheum. 2008, 58, 532–540. [Google Scholar] [CrossRef] [PubMed]
  53. Stetson, D.B.; Ko, J.S.; Heidmann, T.; Medzhitov, R. Trex1 Prevents Cell-Intrinsic Initiation of Autoimmunity. Cell 2008, 134, 587–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Perl, A.; Fernandez, D.; Telarico, T.; Phillips, P.E. Endogenous Retroviral Pathogenesis in Lupus. Curr. Opin. Rheumatol. 2010, 22, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Draborg, A.H.; Duus, K.; Houen, G. Epstein-Barr Virus and Systemic Lupus Erythematosus. Clin. Dev. Immunol. 2012, 2012, 370516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. James, J.A.; Robertson, J.M. Lupus and Epstein-Barr. Curr. Opin. Rheumatol. 2012, 24, 383–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Johnson, K.E.; Tarakanova, V.L. Gammaherpesviruses and B Cells: A Relationship That Lasts a Lifetime. Viral Immunol. 2020, 33, 316–326. [Google Scholar] [CrossRef]
  58. Buschle, A.; Hammerschmidt, W. Epigenetic Lifestyle of Epstein-Barr Virus. Semin. Immunopathol. 2020, 42, 131–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Münz, C. Latency and Lytic Replication in Epstein-Barr Virus-Associated Oncogenesis. Nat. Rev. Microbiol. 2019, 17, 691–700. [Google Scholar] [CrossRef] [Green Version]
  60. Niller, H.H.; Wolf, H.; Minarovits, J. Regulation and Dysregulation of Epstein-Barr Virus Latency: Implications for the Development of Autoimmune Diseases. Autoimmunity 2008, 41, 298–328. [Google Scholar] [CrossRef]
  61. Germini, D.; Sall, F.B.; Shmakova, A.; Wiels, J.; Dokudovskaya, S.; Drouet, E.; Vassetzky, Y. Oncogenic Properties of the EBV ZEBRA Protein. Cancers 2020, 12, 1479. [Google Scholar] [CrossRef]
  62. 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] [Green Version]
  63. Khanim, F.; Dawson, C.; Meseda, C.A.; Dawson, J.; Mackett, M.; Young, L.S. BHRF1, a Viral Homologue of the Bcl-2 Oncogene, Is Conserved at Both the Sequence and Functional Level in Different Epstein-Barr Virus Isolates. J. Gen. Virol. 1997, 78 Pt 11, 2987–2999. [Google Scholar] [CrossRef] [Green Version]
  64. Rasmussen, N.S.; Draborg, A.H.; Nielsen, C.T.; Jacobsen, S.; Houen, G. Antibodies to Early EBV, CMV, and HHV6 Antigens in Systemic Lupus Erythematosus Patients. Scand. J. Rheumatol. 2015, 44, 143–149. [Google Scholar] [CrossRef] [Green Version]
  65. Babcock, G.J.; Hochberg, D.; Thorley-Lawson, A.D. The Expression Pattern of Epstein-Barr Virus Latent Genes in Vivo Is Dependent upon the Differentiation Stage of the Infected B Cell. Immunity 2000, 13, 497–506. [Google Scholar] [CrossRef] [Green Version]
  66. Arcipowski, K.M.; Bishop, G.A. Roles of the Kinase TAK1 in TRAF6-Dependent Signaling by CD40 and Its Oncogenic Viral Mimic, LMP1. PLoS ONE 2012, 7, e42478. [Google Scholar] [CrossRef]
  67. Minamitani, T.; Yasui, T.; Ma, Y.; Zhou, H.; Okuzaki, D.; Tsai, C.-Y.; Sakakibara, S.; Gewurz, B.E.; Kieff, E.; Kikutani, H. Evasion of Affinity-Based Selection in Germinal Centers by Epstein-Barr Virus LMP2A. Proc. Natl. Acad. Sci. USA 2015, 112, 11612–11617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Han, L.; Zhang, Y.; Wang, Q.; Xin, M.; Yang, K.; Lei, K.; Sun, M. Epstein-Barr Virus Infection and Type I Interferon Signature in Patients with Systemic Lupus Erythematosus. Lupus 2018, 27, 947–954. [Google Scholar] [CrossRef]
  69. Quan, T.E.; Roman, R.M.; Rudenga, B.J.; Holers, V.M.; Craft, J.E. Epstein-Barr Virus Promotes Interferon-Alpha Production by Plasmacytoid Dendritic Cells. Arthritis Rheum. 2010, 62, 1693–1701. [Google Scholar] [CrossRef] [Green Version]
  70. Valente, R.M.; Ehlers, E.; Xu, D.; Ahmad, H.; Steadman, A.; Blasnitz, L.; Zhou, Y.; Kastanek, L.; Meng, B.; Zhang, L. Toll-like Receptor 7 Stimulates the Expression of Epstein-Barr Virus Latent Membrane Protein 1. PLoS ONE 2012, 7, e43317. [Google Scholar] [CrossRef] [Green Version]
  71. Murayama, G.; Furusawa, N.; Chiba, A.; Yamaji, K.; Tamura, N.; Miyake, S. Enhanced IFN-α Production Is Associated with Increased TLR7 Retention in the Lysosomes of Palasmacytoid Dendritic Cells in Systemic Lupus Erythematosus. Arthritis Res. Ther. 2017, 19, 234. [Google Scholar] [CrossRef] [Green Version]
  72. Yu, X.-X.; Yao, C.-W.; Tao, J.-L.; Yang, C.; Luo, M.-N.; Li, S.-M.; Liu, H.-F. The Expression of Renal Epstein-Barr Virus Markers in Patients with Lupus Nephritis. Exp. Ther. Med. 2014, 7, 1135–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ding, Y.; He, X.; Liao, W.; Yi, Z.; Yang, H.; Xiang, W. The Expression of EBV-Encoded LMP1 in Young Patients with Lupus Nephritis. Int. J. Clin. Exp. Med. 2015, 8, 6073–6078. [Google Scholar]
  74. 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] [PubMed]
  75. McClain, M.T.; Poole, B.D.; Bruner, B.F.; Kaufman, K.M.; Harley, J.B.; James, J.A. An Altered Immune Response to Epstein-Barr Nuclear Antigen 1 in Pediatric Systemic Lupus Erythematosus. Arthritis Rheum. 2006, 54, 360–368. [Google Scholar] [CrossRef] [PubMed]
  76. Harley, J.B.; Chen, X.; Pujato, M.; Miller, D.; Maddox, A.; Forney, C.; Magnusen, A.F.; Lynch, A.; Chetal, K.; Yukawa, M.; et al. Transcription Factors Operate across Disease Loci, with EBNA2 Implicated in Autoimmunity. Nat. Genet. 2018, 50, 699–707. [Google Scholar] [CrossRef] [PubMed]
  77. Peters, A.L.; Stunz, L.L.; Meyerholz, D.K.; Mohan, C.; Bishop, G.A. Latent Membrane Protein 1, the EBV-Encoded Oncogenic Mimic of CD40, Accelerates Autoimmunity in B6.Sle1 Mice. J. Immunol. 2010, 185, 4053–4062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Draborg, A.H.; Jacobsen, S.; Westergaard, M.; Mortensen, S.; Larsen, J.L.; Houen, G.; Duus, K. Reduced Response to Epstein-Barr Virus Antigens by T-Cells in Systemic Lupus Erythematosus Patients. Lupus Sci. Med. 2014, 1, e000015. [Google Scholar] [CrossRef] [Green Version]
  79. Draborg, A.H.; Sandhu, N.; Larsen, N.; Lisander Larsen, J.; Jacobsen, S.; Houen, G. Impaired Cytokine Responses to Epstein-Barr Virus Antigens in Systemic Lupus Erythematosus Patients. J. Immunol. Res. 2016, 2016, 6473204. [Google Scholar] [CrossRef]
  80. Draborg, A.; Izarzugaza, J.M.G.; Houen, G. How Compelling Are the Data for Epstein-Barr Virus Being a Trigger for Systemic Lupus and Other Autoimmune Diseases? Curr. Opin. Rheumatol. 2016, 28, 398–404. [Google Scholar] [CrossRef] [Green Version]
  81. Tsokos, G.C.; Magrath, I.T.; Balow, J.E. Epstein-Barr Virus Induces Normal B Cell Responses but Defective Suppressor T Cell Responses in Patients with Systemic Lupus Erythematosus. J. Immunol. 1983, 131, 1797–1801. [Google Scholar]
  82. Kang, I.; Quan, T.; Nolasco, H.; Park, S.-H.; Hong, M.S.; Crouch, J.; Pamer, E.G.; Howe, J.G.; Craft, J. Defective Control of Latent Epstein-Barr Virus Infection in Systemic Lupus Erythematosus. J. Immunol. 2004, 172, 1287–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Larsen, M.; Sauce, D.; Deback, C.; Arnaud, L.; Mathian, A.; Miyara, M.; Boutolleau, D.; Parizot, C.; Dorgham, K.; Papagno, L.; et al. Exhausted Cytotoxic Control of Epstein-Barr Virus in Human Lupus. PLoS Pathog. 2011, 7, e1002328. [Google Scholar] [CrossRef] [PubMed]
  84. Su, R.; Li, Z.; Wang, Y.; Liu, Y.; Zheng, X.; Gao, C.; Li, X.; Wang, C. Imbalance between Th17 and Regulatory T Cells in Patients with Systemic Lupus Erythematosus Combined EBV/CMV Viraemia. Clin. Exp. Rheumatol. 2020, 38, 864–873. [Google Scholar] [PubMed]
  85. Gross, A.J.; Hochberg, D.; Rand, W.M.; Thorley-Lawson, D.A. EBV and Systemic Lupus Erythematosus: A New Perspective. J. Immunol. 2005, 174, 6599–6607. [Google Scholar] [CrossRef]
  86. Moon, U.Y.; Park, S.J.; Oh, S.T.; Kim, W.-U.; Park, S.-H.; Lee, S.-H.; Cho, C.-S.; Kim, H.-Y.; Lee, W.-K.; Lee, 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] [Green Version]
  87. Yu, S.-F.; Wu, H.-C.; Tsai, W.-C.; Yen, J.-H.; Chiang, W.; Yuo, C.-Y.; Lu, S.-N.; Chiang, L.-C.; Chen, C.-J. Detecting Epstein-Barr Virus DNA from Peripheral Blood Mononuclear Cells in Adult Patients with Systemic Lupus Erythematosus in Taiwan. Med. Microbiol. Immunol. 2005, 194, 115–120. [Google Scholar] [CrossRef]
  88. Poole, B.D.; Templeton, A.K.; Guthridge, J.M.; Brown, E.J.; Harley, J.B.; James, J.A. Aberrant Epstein-Barr Viral Infection in Systemic Lupus Erythematosus. Autoimmun. Rev. 2009, 8, 337–342. [Google Scholar] [CrossRef] [Green Version]
  89. Draborg, A.H.; Jørgensen, J.M.; Müller, H.; Nielsen, C.T.; Jacobsen, S.; Iversen, L.V.; Theander, E.; Nielsen, L.P.; Houen, G.; Duus, K. Epstein-Barr Virus Early Antigen Diffuse (EBV-EA/D)-Directed Immunoglobulin A Antibodies in Systemic Lupus Erythematosus Patients. Scand. J. Rheumatol. 2012, 41, 280–289. [Google Scholar] [CrossRef]
  90. 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]
  91. Vista, E.S.; Weisman, M.H.; Ishimori, M.L.; Chen, H.; Bourn, R.L.; Bruner, B.F.; Hamijoyo, L.; Tanangunan, R.D.; Gal, N.J.; Robertson, J.M.; et al. Strong Viral Associations with SLE among Filipinos. Lupus Sci. Med. 2017, 4, e000214. [Google Scholar] [CrossRef]
  92. Cui, J.; Yan, W.; Xu, S.; Wang, Q.; Zhang, W.; Liu, W.; Ni, A. Anti-Epstein-Barr Virus Antibodies in Beijing during 2013-2017: What We Have Found in the Different Patients. PLoS ONE 2018, 13, e0193171. [Google Scholar] [CrossRef] [Green Version]
  93. Hanlon, P.; Avenell, A.; Aucott, L.; Vickers, M.A. Systematic Review and Meta-Analysis of the Sero-Epidemiological Association between Epstein-Barr Virus and Systemic Lupus Erythematosus. Arthritis Res. Ther. 2014, 16, R3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. 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. Exp. Med. 2019, 19, 23–36. [Google Scholar] [CrossRef] [Green Version]
  95. Esen, B.A.; Yılmaz, G.; Uzun, S.; Ozdamar, M.; Aksözek, A.; Kamalı, S.; Türkoğlu, S.; Gül, A.; Ocal, L.; Aral, O.; et al. Serologic Response to Epstein-Barr Virus Antigens in Patients with Systemic Lupus Erythematosus: A Controlled Study. Rheumatol. Int. 2012, 32, 79–83. [Google Scholar] [CrossRef] [PubMed]
  96. Chougule, D.; Nadkar, M.; Rajadhyaksha, A.; Pandit-Shende, P.; Surve, P.; Dawkar, N.; Khadilkar, P.; Patwardhan, M.; Kaveri, S.; Ghosh, K.; et al. 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] [PubMed]
  97. Rasmussen, N.S.; Nielsen, C.T.; Houen, G.; Jacobsen, S. Humoral Markers of Active Epstein-Barr Virus Infection Associate with Anti-Extractable Nuclear Antigen Autoantibodies and Plasma Galectin-3 Binding Protein in Systemic Lupus Erythematosus. Lupus 2016, 25, 1567–1576. [Google Scholar] [CrossRef] [PubMed]
  98. Trier, N.H.; Draborg, A.H.; Sternbæk, L.; Troelsen, L.; Larsen, J.L.; Jacobsen, S.; Houen, G. EBNA1 IgM-Based Discrimination Between Rheumatoid Arthritis Patients, Systemic Lupus Erythematosus Patients and Healthy Controls. Antibodies 2019, 8, 35. [Google Scholar] [CrossRef] [Green Version]
  99. Chen, C.-J.; Lin, K.-H.; Lin, S.-C.; Tsai, W.-C.; Yen, J.-H.; Chang, S.-J.; Lu, S.-N.; Liu, H.-W. High Prevalence of Immunoglobulin A Antibody against Epstein-Barr Virus Capsid Antigen in Adult Patients with Lupus with Disease Flare: Case Control Studies. J. Rheumatol. 2005, 32, 44–47. [Google Scholar] [PubMed]
  100. Sternbæk, L.; Draborg, A.H.; Østerlund, M.T.; Iversen, L.V.; Troelsen, L.; Theander, E.; Nielsen, C.T.; Jacobsen, S.; Houen, G. Increased Antibody Levels to Stage-Specific Epstein-Barr Virus Antigens in Systemic Autoimmune Diseases Reveal a Common Pathology. Scand. J. Clin. Lab. Investig. 2019, 79, 7–16. [Google Scholar] [CrossRef]
  101. Watad, A.; Azrielant, S.; Bragazzi, N.L.; Sharif, K.; David, P.; Katz, I.; Aljadeff, G.; Quaresma, M.; Tanay, G.; Adawi, M.; et al. Seasonality and Autoimmune Diseases: The Contribution of the Four Seasons to the Mosaic of Autoimmunity. J. Autoimmun. 2017, 82, 13–30. [Google Scholar] [CrossRef] [PubMed]
  102. McClain, M.T.; Heinlen, L.D.; Dennis, G.J.; Roebuck, J.; Harley, J.B.; James, J.A. Early Events in Lupus Humoral Autoimmunity Suggest Initiation through Molecular Mimicry. Nat. Med. 2005, 11, 85–89. [Google Scholar] [CrossRef] [PubMed]
  103. Jog, N.R.; Young, K.A.; Munroe, M.E.; Harmon, M.T.; Guthridge, J.M.; Kelly, J.A.; Kamen, 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] [PubMed] [Green Version]
  104. Agmon-Levin, N.; Dagan, A.; Peri, Y.; Anaya, J.-M.; Selmi, C.; Tincani, A.; Bizzaro, N.; Stojanovich, L.; Damoiseaux, J.; Cohen Tervaert, J.W.; et al. The Interaction between Anti-Ro/SSA and Anti-La/SSB Autoantibodies and Anti-Infectious Antibodies in a Wide Spectrum of Auto-Immune Diseases: Another Angle of the Autoimmune Mosaic. Clin. Exp. Rheumatol. 2017, 35, 929–935. [Google Scholar] [PubMed]
  105. Maślińska, M. The Role of Epstein-Barr Virus Infection in Primary Sjögren’s Syndrome. Curr. Opin. Rheumatol. 2019, 31, 475–483. [Google Scholar] [CrossRef]
  106. Young, N.S.; Brown, K.E. Parvovirus B19. N. Engl. J. Med. 2004, 350, 586–597. [Google Scholar] [CrossRef]
  107. Ganaie, S.S.; Qiu, J. Recent Advances in Replication and Infection of Human Parvovirus B19. Front. Cell. Infect. Microbiol. 2018, 8, 166. [Google Scholar] [CrossRef] [Green Version]
  108. Dorsch, S.; Liebisch, G.; Kaufmann, B.; von Landenberg, P.; Hoffmann, J.H.; Drobnik, W.; Modrow, S. The VP1 Unique Region of Parvovirus B19 and Its Constituent Phospholipase A2-like Activity. J. Virol. 2002, 76, 2014–2018. [Google Scholar] [CrossRef] [Green Version]
  109. Lin, C.-Y.; Chiu, C.-C.; Cheng, J.; Lin, C.-Y.; Shi, Y.-F.; Tsai, C.-C.; Tzang, B.-S.; Hsu, T.-C. Antigenicity Analysis of Human Parvovirus B19-VP1u Protein in the Induction of Anti-Phospholipid Syndrome. Virulence 2018, 9, 208–216. [Google Scholar] [CrossRef]
  110. Moffatt, S.; Tanaka, N.; Tada, K.; Nose, M.; Nakamura, M.; Muraoka, O.; Hirano, T.; Sugamura, K. A Cytotoxic Nonstructural Protein, NS1, of Human Parvovirus B19 Induces Activation of Interleukin-6 Gene Expression. J. Virol. 1996, 70, 8485–8491. [Google Scholar] [CrossRef] [Green Version]
  111. Mitchell, L.A. Parvovirus B19 Nonstructural (NS1) Protein as a Transactivator of Interleukin-6 Synthesis: Common Pathway in Inflammatory Sequelae of Human Parvovirus Infections? J. Med. Virol. 2002, 67, 267–274. [Google Scholar] [CrossRef]
  112. Marco, H.; Guermah, I.; Matas, L.; Hernández, A.; Navarro, M.; Lopez, D.; Bonet, J. Postinfectious Glomerulonephritis Secondary to Erythrovirus B19 (Parvovirus B19): Case Report and Review of the Literature. Clin. Nephrol. 2016, 85, 238–244. [Google Scholar] [CrossRef]
  113. Kauffmann, M.; Bobot, M.; Daniel, L.; Torrents, J.; Knefati, Y.; Moranne, O.; Burtey, S.; Zandotti, C.; Jourde-Chiche, N. Parvovirus B19 Infection and Kidney Injury: Report of 4 Cases and Analysis of Immunization and Viremia in an Adult Cohort of 100 Patients Undergoing a Kidney Biopsy. BMC Nephrol. 2020, 21, 260. [Google Scholar] [CrossRef] [PubMed]
  114. Sève, P.; Ferry, T.; Koenig, M.; Cathebras, P.; Rousset, H.; Broussolle, C. Lupus-like Presentation of Parvovirus B19 Infection. Semin. Arthritis Rheum. 2005, 34, 642–648. [Google Scholar] [CrossRef] [PubMed]
  115. Severin, M.C.; Levy, Y.; Shoenfeld, Y. Systemic Lupus Erythematosus and Parvovirus B-19: Casual Coincidence or Causative Culprit? Clin. Rev. Allergy Immunol. 2003, 25, 41–48. [Google Scholar] [CrossRef]
  116. Ramos-Casals, M.; Cuadrado, M.J.; Alba, P.; Sanna, G.; Brito-Zerón, P.; Bertolaccini, L.; Babini, A.; Moreno, A.; D’Cruz, D.; Khamashta, M.A. Acute Viral Infections in Patients with Systemic Lupus Erythematosus: Description of 23 Cases and Review of the Literature. Medicine 2008, 87, 311–318. [Google Scholar] [CrossRef] [PubMed]
  117. Page, C.; François, C.; Goëb, V.; Duverlie, G. Human Parvovirus B19 and Autoimmune Diseases. Review of the Literature and Pathophysiological Hypotheses. J. Clin. Virol. 2015, 72, 69–74. [Google Scholar] [CrossRef]
  118. Aslanidis, S.; Pyrpasopoulou, A.; Kontotasios, K.; Doumas, S.; Zamboulis, C. Parvovirus B19 Infection and Systemic Lupus Erythematosus: Activation of an Aberrant Pathway? Eur. J. Intern. Med. 2008, 19, 314–318. [Google Scholar] [CrossRef]
  119. Kerr, J.R.; Barah, F.; Mattey, D.L.; Laing, I.; Hopkins, S.J.; Hutchinson, I.V.; Tyrrell, D.A.J. Circulating Tumour Necrosis Factor-Alpha and Interferon-Gamma Are Detectable during Acute and Convalescent Parvovirus B19 Infection and Are Associated with Prolonged and Chronic Fatigue. J. Gen. Virol. 2001, 82, 3011–3019. [Google Scholar] [CrossRef]
  120. Petri, M. Antiphospholipid Syndrome. Transl. Res. 2020, 225, 70–81. [Google Scholar] [CrossRef]
  121. Kalt, M.; Gertner, E. Antibodies to Beta 2-Glycoprotein I and Cardiolipin with Symptoms Suggestive of Systemic Lupus Erythematosus in Parvovirus B19 Infection. J. Rheumatol. 2001, 28, 2335–2336. [Google Scholar]
  122. Lehmann, H.W.; Plentz, A.; von Landenberg, P.; Küster, R.-M.; Modrow, S. Different Patterns of Disease Manifestations of Parvovirus B19-Associated Reactive Juvenile Arthritis and the Induction of Antiphospholipid-Antibodies. Clin. Rheumatol. 2008, 27, 333–338. [Google Scholar] [CrossRef]
  123. Chen, D.-Y.; Chen, Y.-M.; Tzang, B.-S.; Lan, J.-L.; Hsu, T.-C. Th17-Related Cytokines in Systemic Lupus Erythematosus Patients with Dilated Cardiomyopathies: A Possible Linkage to Parvovirus B19 Infection. PLoS ONE 2014, 9, e113889. [Google Scholar] [CrossRef] [PubMed]
  124. Jia, X.; Gong, L.; Huang, G.; Zhang, W. Review of correlation between human parvovirus B19 and autoimmune disease etiology. Chin. J. Cell. Mol. Immunol. 2020, 36, 75–80. [Google Scholar]
  125. Talotta, R.; Atzeni, F.; Laska, M.J. Retroviruses in the Pathogenesis of Systemic Lupus Erythematosus: Are They Potential Therapeutic Targets? Autoimmunity 2020, 53, 177–191. [Google Scholar] [CrossRef] [PubMed]
  126. Greenig, M. HERVs, Immunity, and Autoimmunity: Understanding the Connection. PeerJ 2019, 7, e6711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Adelman, M.K.; Marchalonis, J.J. Endogenous Retroviruses in Systemic Lupus Erythematosus: Candidate Lupus Viruses. Clin. Immunol. 2002, 102, 107–116. [Google Scholar] [CrossRef]
  128. Katoh, I.; Kurata, S.-I. Association of Endogenous Retroviruses and Long Terminal Repeats with Human Disorders. Front. Oncol. 2013, 3, 234. [Google Scholar] [CrossRef] [Green Version]
  129. Perl, A.; Colombo, E.; Dai, H.; Agarwal, R.; Mark, K.A.; Banki, K.; Poiesz, B.J.; Phillips, P.E.; Hoch, S.O.; Reveille, J.D. Antibody Reactivity to the HRES-1 Endogenous Retroviral Element Identifies a Subset of Patients with Systemic Lupus Erythematosus and Overlap Syndromes. Correlation with Antinuclear Antibodies and HLA Class II Alleles. Arthritis Rheum. 1995, 38, 1660–1671. [Google Scholar] [CrossRef]
  130. Mustelin, T.; Ukadike, K.C. How Retroviruses and Retrotransposons in Our Genome May Contribute to Autoimmunity in Rheumatological Conditions. Front. Immunol. 2020, 11, 593891. [Google Scholar] [CrossRef]
  131. Chuong, E.B.; Elde, N.C.; Feschotte, C. Regulatory Evolution of Innate Immunity through Co-Option of Endogenous Retroviruses. Science 2016, 351, 1083–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Blank, M.; Shoenfeld, Y.; Perl, A. Cross-Talk of the Environment with the Host Genome and the Immune System through Endogenous Retroviruses in Systemic Lupus Erythematosus. Lupus 2009, 18, 1136–1143. [Google Scholar] [CrossRef]
  133. Tokuyama, M.; Kong, Y.; Song, E.; Jayewickreme, T.; Kang, I.; Iwasaki, A. ERVmap Analysis Reveals Genome-Wide Transcription of Human Endogenous Retroviruses. Proc. Natl. Acad. Sci. USA 2018, 115, 12565–12572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Godavarthy, A.; Kelly, R.; Jimah, J.; Beckford, M.; Caza, T.; Fernandez, D.; Huang, N.; Duarte, M.; Lewis, J.; Fadel, H.J.; et al. Lupus-Associated Endogenous Retroviral LTR Polymorphism and Epigenetic Imprinting Promote HRES-1/RAB4 Expression and MTOR Activation. JCI Insight 2020, 5, e134010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Perl, A.; Nagy, G.; Koncz, A.; Gergely, P.; Fernandez, D.; Doherty, E.; Telarico, T.; Bonilla, E.; Phillips, P.E. Molecular Mimicry and Immunomodulation by the HRES-1 Endogenous Retrovirus in SLE. Autoimmunity 2008, 41, 287–297. [Google Scholar] [CrossRef] [Green Version]
  136. Fernandez, D.R.; Telarico, T.; Bonilla, E.; Li, Q.; Banerjee, S.; Middleton, F.A.; Phillips, P.E.; Crow, M.K.; Oess, S.; Muller-Esterl, W.; et al. Activation of Mammalian Target of Rapamycin Controls the Loss of TCRzeta in Lupus T Cells through HRES-1/Rab4-Regulated Lysosomal Degradation. J. Immunol. 2009, 182, 2063–2073. [Google Scholar] [CrossRef] [Green Version]
  137. Mak, A.; Kow, N.Y. The Pathology of T Cells in Systemic Lupus Erythematosus. J. Immunol. Res. 2014, 2014, 419029. [Google Scholar] [CrossRef]
  138. Baudino, L.; Yoshinobu, K.; Morito, N.; Santiago-Raber, M.-L.; Izui, S. Role of Endogenous Retroviruses in Murine SLE. Autoimmun. Rev. 2010, 10, 27–34. [Google Scholar] [CrossRef] [PubMed]
  139. Ito, K.; Baudino, L.; Kihara, M.; Leroy, V.; Vyse, T.J.; Evans, L.H.; Izui, S. Three Sgp Loci Act Independently as Well as Synergistically to Elevate the Expression of Specific Endogenous Retroviruses Implicated in Murine Lupus. J. Autoimmun. 2013, 43, 10–17. [Google Scholar] [CrossRef] [Green Version]
  140. Treger, R.S.; Pope, S.D.; Kong, Y.; Tokuyama, M.; Taura, M.; Iwasaki, A. The Lupus Susceptibility Locus Sgp3 Encodes the Suppressor of Endogenous Retrovirus Expression SNERV. Immunity 2019, 50, 334–347.e9. [Google Scholar] [CrossRef] [Green Version]
  141. Panova, V.; Attig, J.; Young, G.R.; Stoye, J.P.; Kassiotis, G. Antibody-Induced Internalisation of Retroviral Envelope Glycoproteins Is a Signal Initiation Event. PLoS Pathog. 2020, 16, e1008605. [Google Scholar] [CrossRef] [PubMed]
  142. Browne, E.P. The Role of Toll-Like Receptors in Retroviral Infection. Microorganisms 2020, 8, 1787. [Google Scholar] [CrossRef]
  143. Barral, P.M.; Sarkar, D.; Su, Z.; Barber, G.N.; DeSalle, R.; Racaniello, V.R.; Fisher, P.B. Functions of the Cytoplasmic RNA Sensors RIG-I and MDA-5: Key Regulators of Innate Immunity. Pharmacol. Ther. 2009, 124, 219–234. [Google Scholar] [CrossRef] [Green Version]
  144. Shehab, M.; Sherri, N.; Hussein, H.; Salloum, N.; Rahal, E.A. Endosomal Toll-Like Receptors Mediate Enhancement of Interleukin-17A Production Triggered by Epstein-Barr Virus DNA in Mice. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  145. Miyake, K.; Shibata, T.; Ohto, U.; Shimizu, T.; Saitoh, S.-I.; Fukui, R.; Murakami, Y. Mechanisms Controlling Nucleic Acid-Sensing Toll-like Receptors. Int. Immunol. 2018, 30, 43–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Wu, Z.; Mei, X.; Zhao, D.; Sun, Y.; Song, J.; Pan, W.; Shi, W. DNA Methylation Modulates HERV-E Expression in CD4+ T Cells from Systemic Lupus Erythematosus Patients. J. Dermatol. Sci. 2015, 77, 110–116. [Google Scholar] [CrossRef]
  147. Wang, X.; Zhao, C.; Zhang, C.; Mei, X.; Song, J.; Sun, Y.; Wu, Z.; Shi, W. Increased HERV-E Clone 4-1 Expression Contributes to DNA Hypomethylation and IL-17 Release from CD4+ T Cells via MiR-302d/MBD2 in Systemic Lupus Erythematosus. Cell Commun. Signal. 2019, 17, 94. [Google Scholar] [CrossRef] [Green Version]
  148. Talotta, R.; Atzeni, F.; Laska, M.J. The Contribution of HERV-E Clone 4-1 and Other HERV-E Members to the Pathogenesis of Rheumatic Autoimmune Diseases. APMIS 2020, 128, 367–377. [Google Scholar] [CrossRef] [PubMed]
  149. Piotrowski, P.C.; Duriagin, S.; Jagodzinski, P.P. Expression of Human Endogenous Retrovirus Clone 4-1 May Correlate with Blood Plasma Concentration of Anti-U1 RNP and Anti-Sm Nuclear Antibodies. Clin. Rheumatol. 2005, 24, 620–624. [Google Scholar] [CrossRef]
  150. Mavragani, C.P.; Sagalovskiy, I.; Guo, Q.; Nezos, A.; Kapsogeorgou, E.K.; Lu, P.; Liang Zhou, J.; Kirou, K.A.; Seshan, S.V.; Moutsopoulos, H.M.; et al. Expression of Long Interspersed Nuclear Element 1 Retroelements and Induction of Type I Interferon in Patients With Systemic Autoimmune Disease. Arthritis Rheumatol. 2016, 68, 2686–2696. [Google Scholar] [CrossRef]
  151. Huang, X.; Su, G.; Wang, Z.; Shangguan, S.; Cui, X.; Zhu, J.; Kang, M.; Li, S.; Zhang, T.; Wu, F.; et al. Hypomethylation of Long Interspersed Nucleotide Element-1 in Peripheral Mononuclear Cells of Juvenile Systemic Lupus Erythematosus Patients in China. Int. J. Rheum. Dis. 2014, 17, 280–290. [Google Scholar] [CrossRef]
  152. Sukapan, P.; Promnarate, P.; Avihingsanon, Y.; Mutirangura, A.; Hirankarn, N. Types of DNA Methylation Status of the Interspersed Repetitive Sequences for LINE-1, Alu, HERV-E and HERV-K in the Neutrophils from Systemic Lupus Erythematosus Patients and Healthy Controls. J. Hum. Genet. 2014, 59, 178–188. [Google Scholar] [CrossRef]
  153. Emmer, A.; Staege, M.S.; Kornhuber, M.E. The Retrovirus/Superantigen Hypothesis of Multiple Sclerosis. Cell. Mol. Neurobiol. 2014, 34, 1087–1096. [Google Scholar] [CrossRef] [PubMed]
  154. Belgaumkar, V.A.; Chavan, R.B.; Suryataley, P.R.; Salunke, A.S.; Patil, P.P.; Borade, S.M. Systemic Lupus Erythematosus in HIV: An Insight into Clinical Implications and Management. Indian J. Sex. Transm. Dis. AIDS 2019, 40, 64–66. [Google Scholar] [CrossRef] [PubMed]
  155. Laska, M.J.; Troldborg, A.; Hauge, E.-M.; Bahrami, S.; Stengaard-Pedersen, K. Human Endogenous Retroviral Genetic Element With Immunosuppressive Activity in Both Human Autoimmune Diseases and Experimental Arthritis. Arthritis Rheumatol. 2017, 69, 398–409. [Google Scholar] [CrossRef] [PubMed]
  156. Bahrami, S.; Gryz, E.A.; Graversen, J.H.; Troldborg, A.; Stengaard Pedersen, K.; Laska, M.J. Immunomodulating Peptides Derived from Different Human Endogenous Retroviruses (HERVs) Show Dissimilar Impact on Pathogenesis of a Multiple Sclerosis Animal Disease Model. Clin. Immunol. 2018, 191, 37–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Wakui, H.; Masai, R.; Okuyama, S.; Ohtani, H.; Komatsuda, A.; Toyoshima, I.; Watanabe, S.; Sawada, K.-I. Development of Lupus Nephritis in a Patient with Human T-Cell Lymphotropic Virus Type I-Associated Myelopathy. Am. J. Kidney Dis. 2005, 46, e25–e29. [Google Scholar] [CrossRef]
  158. Akimoto, M.; Matsushita, K.; Suruga, Y.; Aoki, N.; Ozaki, A.; Uozumi, K.; Tei, C.; Arima, N. Clinical Manifestations of Human T Lymphotropic Virus Type I-Infected Patients with Systemic Lupus Erythematosus. J. Rheumatol. 2007, 34, 1841–1848. [Google Scholar]
  159. Banki, K.; Maceda, J.; Hurley, E.; Ablonczy, E.; Mattson, D.H.; Szegedy, L.; Hung, C.; Perl, A. Human T-Cell Lymphotropic Virus (HTLV)-Related Endogenous Sequence, HRES-1, Encodes a 28-KDa Protein: A Possible Autoantigen for HTLV-I Gag-Reactive Autoantibodies. Proc. Natl. Acad. Sci. USA 1992, 89, 1939–1943. [Google Scholar] [CrossRef] [Green Version]
  160. Lebrun, D.; Hentzien, M.; Cuzin, L.; Rey, D.; Joly, V.; Cotte, L.; Allavena, C.; Dellamonica, P.; Servettaz, A.; Bani-Sadr, F.; et al. Epidemiology of Autoimmune and Inflammatory Diseases in a French Nationwide HIV Cohort. AIDS 2017, 31, 2159–2166. [Google Scholar] [CrossRef]
  161. Gindea, S.; Schwartzman, J.; Herlitz, L.C.; Rosenberg, M.; Abadi, J.; Putterman, C. Proliferative Glomerulonephritis in Lupus Patients with Human Immunodeficiency Virus Infection: A Difficult Clinical Challenge. Semin. Arthritis Rheum. 2010, 40, 201–209. [Google Scholar] [CrossRef]
  162. Wiegersma, J.S.; Franssen, C.F.M.; Diepstra, A. Nephrotic Syndrome Due to Lupus-like Glomerulonephritis in an HIV-Positive Patient. Neth. J. Med. 2017, 75, 412–414. [Google Scholar]
  163. Hamid, C.K.; Hameed, R.A.; Khaliq, B.I.; Manzoor, R.; Hamid, C.Q. HIV Associated Lupus like Nephropathy. Ethiop. J. Health Sci. 2014, 24, 277–283. [Google Scholar] [CrossRef] [Green Version]
  164. Bonsignori, M.; Wiehe, K.; Grimm, S.K.; Lynch, R.; Yang, G.; Kozink, D.M.; Perrin, F.; Cooper, A.J.; Hwang, K.-K.; Chen, X.; et al. An Autoreactive Antibody from an SLE/HIV-1 Individual Broadly Neutralizes HIV-1. J. Clin. Investig. 2014, 124, 1835–1843. [Google Scholar] [CrossRef] [PubMed]
  165. Yao, Q.; Frank, M.; Glynn, M.; Altman, R.D. Rheumatic Manifestations in HIV-1 Infected in-Patients and Literature Review. Clin. Exp. Rheumatol. 2008, 26, 799–806. [Google Scholar]
  166. Nardacci, R.; Ciccosanti, F.; Marsella, C.; Ippolito, G.; Piacentini, M.; Fimia, G.M. Role of Autophagy in HIV Infection and Pathogenesis. J. Intern. Med. 2017, 281, 422–432. [Google Scholar] [CrossRef] [PubMed]
  167. Qi, Y.-Y.; Zhou, X.-J.; Zhang, H. Autophagy and Immunological Aberrations in Systemic Lupus Erythematosus. Eur. J. Immunol. 2019, 49, 523–533. [Google Scholar] [CrossRef] [Green Version]
  168. Oaks, Z.; Winans, T.; Huang, N.; Banki, K.; Perl, A. Activation of the Mechanistic Target of Rapamycin in SLE: Explosion of Evidence in the Last Five Years. Curr. Rheumatol. Rep. 2016, 18, 73. [Google Scholar] [CrossRef] [Green Version]
  169. Lolomadze, E.A.; Rebrikov, D.V. Constant Companion: Clinical and Developmental Aspects of Torque Teno Virus Infections. Arch. Virol. 2020, 165, 2749–2757. [Google Scholar] [CrossRef] [PubMed]
  170. Webb, B.; Rakibuzzaman, A.; Ramamoorthy, S. Torque Teno Viruses in Health and Disease. Virus Res. 2020, 285, 198013. [Google Scholar] [CrossRef]
  171. Garbuglia, A.R.; Iezzi, T.; Capobianchi, M.R.; Pignoloni, P.; Pulsoni, A.; Sourdis, J.; Pescarmona, E.; Vitolo, D.; Mandelli, F. Detection of TT Virus in Lymph Node Biopsies of B-Cell Lymphoma and Hodgkin’s Disease, and Its Association with EBV Infection. Int. J. Immunopathol. Pharmacol. 2003, 16, 109–118. [Google Scholar] [CrossRef] [PubMed]
  172. Sospedra, M.; Zhao, Y.; zur Hausen, H.; Muraro, P.A.; Hamashin, C.; de Villiers, E.-M.; Pinilla, C.; Martin, R. Recognition of Conserved Amino Acid Motifs of Common Viruses and Its Role in Autoimmunity. PLoS Pathog. 2005, 1, e41. [Google Scholar] [CrossRef] [Green Version]
  173. Dioverti, M.V.; Razonable, R.R. Cytomegalovirus. Microbiol. Spectr. 2016, 4, 97–125. [Google Scholar] [CrossRef] [PubMed]
  174. Halenius, A.; Hengel, H. Human Cytomegalovirus and Autoimmune Disease. Biomed. Res. Int. 2014, 2014, 472978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Yamazaki, S.; Endo, A.; Iso, T.; Abe, S.; Aoyagi, Y.; Suzuki, M.; Fujii, T.; Haruna, H.; Ohtsuka, Y.; Shimizu, T. Cytomegalovirus as a Potential Trigger for Systemic Lupus Erythematosus: A Case Report. BMC Res. Notes 2015, 8, 487. [Google Scholar] [CrossRef] [Green Version]
  176. 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] [PubMed] [Green Version]
  177. 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]
  178. 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]
  179. 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-ΚB2 Activation. Cell Death Dis. 2018, 9, 104. [Google Scholar] [CrossRef] [Green Version]
  180. Rozenblyum, E.V.; Allen, U.D.; Silverman, E.D.; Levy, D.M. Cytomegalovirus Infection in Childhood-Onset Systemic Lupus Erythematosus. Int. J. Clin. Rheumtol. 2013, 8, 137–146. [Google Scholar] [CrossRef]
  181. 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]
  182. Draborg, A.H.; Rasmussen, N.S.; Larsen, J.L.; Jørgensen, C.S.; Sandhu, N.; Skogstrand, K.; Jacobsen, S.; Houen, G. Immune Responses to an Early Lytic Cytomegalovirus Antigen in Systemic Lupus Erythematosus Patients: T-Cell Responses, Cytokine Secretions and Antibody Status. PLoS ONE 2018, 13, e0193244. [Google Scholar] [CrossRef] [Green Version]
  183. Wu, C.-S.; Chyuan, I.-T.; Chiu, Y.-L.; Chen, W.-L.; Shen, C.-Y.; Hsu, P.-N. Preserved Specific Anti-Viral T-Cell Response but Associated with Decreased Lupus Activity in SLE Patients with Cytomegalovirus Infection. Rheumatology 2020, 59, 3340–3349. [Google Scholar] [CrossRef] [PubMed]
  184. Cassaniti, I.; Cavagna, L.; Calarota, S.A.; Adzasehoun, K.M.G.; Comolli, G.; Montecucco, C.; Baldanti, F. Evaluation of EBV- and HCMV-Specific T Cell Responses in Systemic Lupus Erythematosus (SLE) Patients Using a Normalized Enzyme-Linked Immunospot (ELISPOT) Assay. J. Immunol. Res. 2019, 2019, 4236503. [Google Scholar] [CrossRef]
  185. Bano, A.; Pera, A.; Almoukayed, A.; Clarke, T.H.S.; Kirmani, S.; Davies, K.A.; Kern, F. CD28 Null CD4 T-Cell Expansions in Autoimmune Disease Suggest a Link with Cytomegalovirus Infection. F1000Resarch 2019, 8, 327. [Google Scholar] [CrossRef]
  186. Dolcino, M.; Puccetti, A.; Barbieri, A.; Bason, C.; Tinazzi, E.; Ottria, A.; Patuzzo, G.; Martinelli, N.; Lunardi, C. Infections and Autoimmunity: Role of Human Cytomegalovirus in Autoimmune Endothelial Cell Damage. Lupus 2015, 24, 419–432. [Google Scholar] [CrossRef] [PubMed]
  187. Stratta, P.; Canavese, C.; Ciccone, G.; Santi, S.; Quaglia, M.; Ghisetti, V.; Marchiaro, G.; Barbui, A.; Fop, F.; Cavallo, R.; et al. Correlation between Cytomegalovirus Infection and Raynaud’s Phenomenon in Lupus Nephritis. Nephron 1999, 82, 145–154. [Google Scholar] [CrossRef]
  188. Labarca, J.A.; Rabaggliati, R.M.; Radrigan, F.J.; Rojas, P.P.; Perez, C.M.; Ferrés, M.V.; Acuna, G.G.; Bertin, P.A. Antiphospholipid Syndrome Associated with Cytomegalovirus Infection: Case Report and Review. Clin. Infect. Dis. 1997, 24, 197–200. [Google Scholar] [CrossRef] [Green Version]
  189. Avcin, T.; Toplak, N. Antiphospholipid Antibodies in Response to Infection. Curr. Rheumatol. Rep. 2007, 9, 212–218. [Google Scholar] [CrossRef] [PubMed]
  190. Sarnow, P.; Sagan, S.M. Unraveling the Mysterious Interactions Between Hepatitis C Virus RNA and Liver-Specific MicroRNA-122. Annu. Rev. Virol. 2016, 3, 309–332. [Google Scholar] [CrossRef]
  191. Rosenthal, E.; Cacoub, P. Extrahepatic Manifestations in Chronic Hepatitis C Virus Carriers. Lupus 2015, 24, 469–482. [Google Scholar] [CrossRef] [PubMed]
  192. Böröcz, K.; Simon, D.; Erdő-Bonyár, S.; Kovács, K.T.; Tuba, É.; Czirják, L.; Németh, P.; Berki, T. Relationship between Natural and Infection-Induced Antibodies in Systemic Autoimmune Diseases (SAD): SLE, SSc and RA. Clin. Exp. Immunol. 2020, 203, 32–40. [Google Scholar] [CrossRef]
  193. Joo, Y.B.; Lim, Y.-H.; Kim, K.-J.; Park, K.-S.; Park, Y.-J. Association of Influenza Infection with Hospitalisation-Related Systemic Lupus Erythematosus Flares: A Time Series Analysis. Clin. Exp. Rheumatol. 2020. [Google Scholar]
  194. Kanduc, D. The Comparative Biochemistry of Viruses and Humans: An Evolutionary Path towards Autoimmunity. Biol. Chem. 2019, 400, 629–638. [Google Scholar] [CrossRef] [PubMed]
  195. Zainal, N.; Tan, K.-K.; Johari, J.; Hussein, H.; Wan Musa, W.R.; Hassan, J.; Lin, Y.-S.; AbuBakar, S. Sera of Patients with Systemic Lupus Erythematosus Cross-Neutralizes Dengue Viruses. Microbiol. Immunol. 2018, 62, 659–672. [Google Scholar] [CrossRef]
  196. Segal, Y.; Dahan, S.; Calabrò, M.; Kanduc, D.; Shoenfeld, Y. HPV and Systemic Lupus Erythematosus: A Mosaic of Potential Crossreactions. Immunol. Res. 2017, 65, 564–571. [Google Scholar] [CrossRef]
  197. Wilson, J.B.; Manet, E.; Gruffat, H.; Busson, P.; Blondel, M.; Fahraeus, R. EBNA1: Oncogenic Activity, Immune Evasion and Biochemical Functions Provide Targets for Novel Therapeutic Strategies against Epstein-Barr Virus- Associated Cancers. Cancers 2018, 10, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Yiu, S.P.T.; Dorothea, M.; Hui, K.F.; Chiang, A.K.S. Lytic Induction Therapy against Epstein-Barr Virus-Associated Malignancies: Past, Present, and Future. Cancers 2020, 12, 2142. [Google Scholar] [CrossRef]
  199. Kerr, J.R. Epstein-Barr Virus (EBV) Reactivation and Therapeutic Inhibitors. J. Clin. Pathol. 2019, 72, 651–658. [Google Scholar] [CrossRef]
  200. Münz, C. Redirecting T Cells against Epstein-Barr Virus Infection and Associated Oncogenesis. Cells 2020, 9, 1400. [Google Scholar] [CrossRef]
  201. Manaresi, E.; Gallinella, G. Advances in the Development of Antiviral Strategies against Parvovirus B19. Viruses 2019, 11, 659. [Google Scholar] [CrossRef] [Green Version]
  202. Liu, P.; Chen, S.; Wang, M.; Cheng, A. The Role of Nuclear Localization Signal in Parvovirus Life Cycle. Virol. J. 2017, 14, 80. [Google Scholar] [CrossRef] [PubMed]
  203. Yen, Y.-F.; Chuang, P.-H.; Jen, I.-A.; Chen, M.; Lan, Y.-C.; Liu, Y.-L.; Lee, Y.; Chen, Y.-H.; Chen, Y.-M.A. Incidence of Autoimmune Diseases in a Nationwide HIV/AIDS Patient Cohort in Taiwan, 2000-2012. Ann. Rheum. Dis. 2017, 76, 661–665. [Google Scholar] [CrossRef]
  204. Contreras-Galindo, R.; Dube, D.; Fujinaga, K.; Kaplan, M.H.; Markovitz, D.M. Susceptibility of Human Endogenous Retrovirus Type K to Reverse Transcriptase Inhibitors. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [Green Version]
  205. Chanouzas, D.; Sagmeister, M.; Faustini, S.; Nightingale, P.; Richter, A.; Ferro, C.J.; Morgan, M.D.; Moss, P.; Harper, L. Subclinical Reactivation of Cytomegalovirus Drives CD4+CD28null T-Cell Expansion and Impaired Immune Response to Pneumococcal Vaccination in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. J. Infect. Dis. 2019, 219, 234–244. [Google Scholar] [CrossRef] [PubMed]
  206. Chanouzas, D.; Dyall, L.; Nightingale, P.; Ferro, C.; Moss, P.; Morgan, M.D.; Harper, L. Valaciclovir to Prevent Cytomegalovirus Mediated Adverse Modulation of the Immune System in ANCA-Associated Vasculitis (CANVAS): Study Protocol for a Randomised Controlled Trial. Trials 2016, 17, 338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. McKinney, E.F.; Lee, J.C.; Jayne, D.R.W.; Lyons, P.A.; Smith, K.G.C. T-Cell Exhaustion, Co-Stimulation and Clinical Outcome in Autoimmunity and Infection. Nature 2015, 523, 612–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Tilstra, J.S.; Avery, L.; Menk, A.V.; Gordon, R.A.; Smita, S.; Kane, L.P.; Chikina, M.; Delgoffe, G.M.; Shlomchik, M.J. Kidney-Infiltrating T Cells in Murine Lupus Nephritis Are Metabolically and Functionally Exhausted. J. Clin. Investig. 2018, 128, 4884–4897. [Google Scholar] [CrossRef]
  209. Linsley, P.S.; Long, S.A. Enforcing the Checkpoints: Harnessing T-Cell Exhaustion for Therapy of T1D. Curr. Opin. Endocrinol. Diabetes Obes. 2019, 26, 213–218. [Google Scholar] [CrossRef] [PubMed]
  210. Moreno-Angarita, A.; Aragón, C.C.; Tobón, G.J. Cathelicidin LL-37: A New Important Molecule in the Pathophysiology of Systemic Lupus Erythematosus. J. Transl. Autoimmun. 2020, 3, 100029. [Google Scholar] [CrossRef]
  211. Mishra, R.; Bhattacharya, S.; Rawat, B.S.; Kumar, A.; Kumar, A.; Niraj, K.; Chande, A.; Gandhi, P.; Khetan, D.; Aggarwal, A.; et al. MicroRNA-30e-5p Has an Integrated Role in the Regulation of the Innate Immune Response during Virus Infection and Systemic Lupus Erythematosus. iScience 2020, 23, 101322. [Google Scholar] [CrossRef]
  212. Bogdanos, D.P.; Sakkas, L.I. From Microbiome to Infectome in Autoimmunity. Curr. Opin. Rheumatol. 2017, 29, 369–373. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, B.; Jia, X.-M.; Xu, J.-Y.; Zhao, L.-D.; Ji, J.-Y.; Wu, B.-X.; Ma, Y.; Li, H.; Zuo, X.-X.; Pan, W.-Y.; et al. The Gut Microbiota of Non-Treated Patients with SLE Defines an Autoimmunogenic and Proinflammatory Profile. Arthritis Rheumatol. 2020. [Google Scholar] [CrossRef] [PubMed]
  214. Kim, J.-W.; Kwok, S.-K.; Choe, J.-Y.; Park, S.-H. Recent Advances in Our Understanding of the Link between the Intestinal Microbiota and Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2019, 20, 4871. [Google Scholar] [CrossRef] [Green Version]
  215. Carvalho-Queiroz, C.; Johansson, M.A.; Persson, J.-O.; Jörtsö, E.; Kjerstadius, T.; Nilsson, C.; Saghafian-Hedengren, S.; Sverremark-Ekström, E. Associations between EBV and CMV Seropositivity, Early Exposures, and Gut Microbiota in a Prospective Birth Cohort: A 10-Year Follow-Up. Front. Pediatr. 2016, 4, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Steed, A.L.; Stappenbeck, T.S. Role of Viruses and Bacteria-Virus Interactions in Autoimmunity. Curr. Opin. Immunol. 2014, 31, 102–107. [Google Scholar] [CrossRef] [Green Version]
  217. Guo, G.; Ye, L.; Shi, X.; Yan, K.; Huang, J.; Lin, K.; Xing, D.; Ye, S.; Wu, Y.; Li, B.; et al. Dysbiosis in Peripheral Blood Mononuclear Cell Virome Associated With Systemic Lupus Erythematosus. Front. Cell. Infect. Microbiol. 2020, 10, 131. [Google Scholar] [CrossRef]
  218. Marietta, E.; Mangalam, A.K.; Taneja, V.; Murray, J.A. Intestinal Dysbiosis in, and Enteral Bacterial Therapies for, Systemic Autoimmune Diseases. Front. Immunol. 2020, 11, 573079. [Google Scholar] [CrossRef]
  219. Dreyfus, D.H. Autoimmune Disease: A Role for New Anti-Viral Therapies? Autoimmun. Rev. 2011, 11, 88–97. [Google Scholar] [CrossRef]
  220. Bragazzi, N.L.; Bridgewood, C.; Sharif, K.; Kamal, M.; Amital, H.; Watad, A.; Shoenfeld, Y. HPV Vaccines and Lupus: Current Approaches towards Preventing Adverse Immune Cross-Reactivity. Expert Rev. Vaccines 2019, 18, 31–42. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Multiple intracellular pathways triggered by Cytomegalovirus (CMV) and Epstein–Barr virus (EBV) infection converge on expression of proinflammatory cytokines and type I Interferon (IFN) response genes. (1) CMV glycoproteins B and H (gB/gH) engage surface TLR-2 and trigger an inflammatory pathway leading to AP-1 activation; (2) cytosolic CMV-DNA interacts with cyclic GMP-AMP synthase (cGAS) and initiates a cascade that targets NFkB and IRF3; (3) CMV nuclear material trafficked to the endosome triggers TLR-7 signaling, leading to IRF-3, -5 and -7 activation; (4) in EBV latency phase, LMP1 induces expression of IR-5, -7 and NFkB; (5) cytosolic EBER-1 is recognised by RIG-1 and MDA-5 and associates with IPS-1 via CARD-like domains, leading to NFkB and IRF3 activation; (6) EBV induces IRF2 and activates ISRE, which codes for EBNA proteins. Legend: MyD88: Myeloid differentiation primary response 88, IKK complex: IkB kinase complex, TRAF: TNF receptor associated factor, AP-1: activator protein-1, EBER-1: EBV-encoded RNA-1; ER: endoplasmic reticulum, cGAS: cyclic GMP-AMP synthase, cGAMP: cyclic guanosine monophosphate-adenosine monophosphate, STING: cyclic GMP-AMP receptor stimulator of interferon genes, TBK: TANK-binding kinase, IRF: interferon regulator factor, LMP1: Epstein–Barr virus latent membrane protein 1, RIG-1: retinoic acid-inducible gene 1, CARD: caspase activation and recruitment domain, MDA5: melanoma differentiation-associated protein-5, IPS-1: interferon-beta promoter stimulator 1, ISRE: IFN-stimulated response element.
Figure 1. Multiple intracellular pathways triggered by Cytomegalovirus (CMV) and Epstein–Barr virus (EBV) infection converge on expression of proinflammatory cytokines and type I Interferon (IFN) response genes. (1) CMV glycoproteins B and H (gB/gH) engage surface TLR-2 and trigger an inflammatory pathway leading to AP-1 activation; (2) cytosolic CMV-DNA interacts with cyclic GMP-AMP synthase (cGAS) and initiates a cascade that targets NFkB and IRF3; (3) CMV nuclear material trafficked to the endosome triggers TLR-7 signaling, leading to IRF-3, -5 and -7 activation; (4) in EBV latency phase, LMP1 induces expression of IR-5, -7 and NFkB; (5) cytosolic EBER-1 is recognised by RIG-1 and MDA-5 and associates with IPS-1 via CARD-like domains, leading to NFkB and IRF3 activation; (6) EBV induces IRF2 and activates ISRE, which codes for EBNA proteins. Legend: MyD88: Myeloid differentiation primary response 88, IKK complex: IkB kinase complex, TRAF: TNF receptor associated factor, AP-1: activator protein-1, EBER-1: EBV-encoded RNA-1; ER: endoplasmic reticulum, cGAS: cyclic GMP-AMP synthase, cGAMP: cyclic guanosine monophosphate-adenosine monophosphate, STING: cyclic GMP-AMP receptor stimulator of interferon genes, TBK: TANK-binding kinase, IRF: interferon regulator factor, LMP1: Epstein–Barr virus latent membrane protein 1, RIG-1: retinoic acid-inducible gene 1, CARD: caspase activation and recruitment domain, MDA5: melanoma differentiation-associated protein-5, IPS-1: interferon-beta promoter stimulator 1, ISRE: IFN-stimulated response element.
Viruses 13 00277 g001
Figure 2. Human Exogenous and Endogenous RVs initiate proinflammatory intracellular pathways converging on critical immune genes involved in SLE pathogenesis. Exogenous RVs RNA and retrotranscribed DNA can activate different cellular sensors located at plasmatic (1) and endosomal membranes (2) or in the cytosol (3), triggering a proinflammatory response (Panel A). HERVs similarly activate endosomal TLRs and also mediate the effects of environmental factors on critical immune genes: estrogens, UVB and infections can increase HERV-E clone 4-1 mRNA expression in SLE CD4+ T-cells and PBMCs, activating a cascade which leads to DNA hypomethylation and type I IFN response (Panel B). Legend: MyD88: Myeloid differentiation primary response 88, TRIF: TIR-domain-containing adapter-inducing interferon-β, TRAM: TRIF-related adaptor molecule, AP-1: activator protein-1, cGAS: cyclic GMP-AMP synthase, cGAMP: cyclic guanosine monophosphate-adenosine monophosphate, STING: cyclic GMP-AMP receptor stimulator of interferon genes, TBK: TANK-binding kinase, IRF: interferon regulator factor, RIG-1: retinoic acid-inducible gene 1, MDA5: melanoma differentiation-associated protein-5, AIM2: absent in melanoma 2, DAI: DNA-dependent activator of interferon regulatory factor, IFI16: γ-interferon-inducible protein IFI-16, MAVS: mitochondrial antiviral signaling protein, LGP2: the laboratory of genetics and physiology 2, NFAT1: nuclear factor of activated T cells 1, MBD2: methyl-CpG-binding domain protein 2,UVB: ultraviolet beams, ERα: estrogen receptor alpha.
Figure 2. Human Exogenous and Endogenous RVs initiate proinflammatory intracellular pathways converging on critical immune genes involved in SLE pathogenesis. Exogenous RVs RNA and retrotranscribed DNA can activate different cellular sensors located at plasmatic (1) and endosomal membranes (2) or in the cytosol (3), triggering a proinflammatory response (Panel A). HERVs similarly activate endosomal TLRs and also mediate the effects of environmental factors on critical immune genes: estrogens, UVB and infections can increase HERV-E clone 4-1 mRNA expression in SLE CD4+ T-cells and PBMCs, activating a cascade which leads to DNA hypomethylation and type I IFN response (Panel B). Legend: MyD88: Myeloid differentiation primary response 88, TRIF: TIR-domain-containing adapter-inducing interferon-β, TRAM: TRIF-related adaptor molecule, AP-1: activator protein-1, cGAS: cyclic GMP-AMP synthase, cGAMP: cyclic guanosine monophosphate-adenosine monophosphate, STING: cyclic GMP-AMP receptor stimulator of interferon genes, TBK: TANK-binding kinase, IRF: interferon regulator factor, RIG-1: retinoic acid-inducible gene 1, MDA5: melanoma differentiation-associated protein-5, AIM2: absent in melanoma 2, DAI: DNA-dependent activator of interferon regulatory factor, IFI16: γ-interferon-inducible protein IFI-16, MAVS: mitochondrial antiviral signaling protein, LGP2: the laboratory of genetics and physiology 2, NFAT1: nuclear factor of activated T cells 1, MBD2: methyl-CpG-binding domain protein 2,UVB: ultraviolet beams, ERα: estrogen receptor alpha.
Viruses 13 00277 g002
Table 1. General mechanisms of virus-induced autoimmunity.
Table 1. General mechanisms of virus-induced autoimmunity.
MechanismDescriptionReferences
Molecular mimicryViral antigens with structural similarity with self-antigens can be presented to and activate autoreactive T-lymphocytes.[8,9,14,15,16,17,18]
Epitope-spreadingOver time, persistent viral infection elicits autoantibodies directed not only towards initial antigens but also multiple epitopes of the same antigens or even different antigens, increasing breadth of immune response.[8,9,14,19]
Superantigen productionSuperantigens lack antigenic specificity and bind to T-cell receptor (TCR) and major histocompatibility complex (MHC) class II molecules, activating T-lymphocytes with a wide range of specificities.[8,9,14]
Bystander activation Release of cytokine by antigen-presenting cells (APCs) or virus-specific T- lymphocytes activates neighboring preprimed autoreactive T-lymphocytes.[8,9,14,20]
Altered apoptosis and clearance deficitViral infections can increase cell apoptosis, with activation of T-helper 17 (Th17) and release of nondigested nuclear material; if a clearance deficit is present, this process may stimulate autoreactive B-lymphocytes survival.[8,9,14,22]
Epigenetic factorsDNA methylation, histone modifications and RNA-based mechanisms are the three main epigenetic modalities which allow viruses to modulate expression of genes involved in immune response.[8,9,14,23,24,25,26]
Persistent or recurrent viral infectionPersistent infection by lymphotropic viruses can stimulate expansion of polyclonal lymphocytes, leading to autoantibody production
Recurrent infections can trigger “facilitating antibodies” which enhance inflammation and antigen exposure in subsequent infective episodes, leading to autoimmunity (e.g.,T1D)
[8,9,14,27,28,29,30]
Innate immunity activationViral DNA/RNA bind to different PRRs which initiate pathways leading to type I IFN response[31,32,33]
Direct cytotoxicityViruses can infect and directly kill target cells, causing AIDs[29,34]
Table 2. General features of the viruses putatively involved in systemic lupus erythematosus (SLE) pathogenesis.
Table 2. General features of the viruses putatively involved in systemic lupus erythematosus (SLE) pathogenesis.
VirusFamily
(Type of Genome)
Proposed Pathogenetic Mechanism in SLETherapeutic ApproachesReferences
Epstein–Barr virus (EBV, HHV4)Herpesviridae (dsDNA, linear)Molecular mimicry, epitope spreadingSynthetic nucleoside analogs (acyclovir/ganciclovir) effective against lytic infection only, not recommended. Corticosteroids possibly beneficial in patients with airway defects or EBV-induced autoimmune complications. [36,37,38]
Cytomegalovirus (CMV, HHV5)Herpesviridae (dsDNA, linear)Epitope spreadingSynthetic nucleosides (ganciclovir or valganciclovir) drugs of choice for serious infections or treatment of immunocompromised hosts. Nucleotide (cidofovir) or pyrophosphate analogs (foscarnet) second-choice drugs.[39,40]
Parvovirus B19 (B19V)Parvoviridae (positive or negative ssDNA, linear)Molecular mimicrySupportive care with transfusion (severe anemia, chronic hemolytic disorders).
Reduction of immunosuppression.
[41,42]
Torque Teno Virus (TTV)Anelloviridae (negative ssDNA, circular)Molecular mimicryNot sensitive to current antiviral prophylaxis/therapy. Viral load reduction observed in HIV pts undergoing Highly active antiretroviral therapy (HAART). IFN associated with viral
clearance during treatment of coinfecting
hepatitis viruses
[43,44]
Hepatitis C virus (HCV)Flaviviridae (positive ssRNA)Epitope spreadingDirect-acting antivirals against viral proteases or polymerases. Ribavirin and IFN as second options.[45]
Dengue virus (DENV)Flaviviridae (positive ssRNA)Epitope spreadingNo specific treatment [46,47]
Retroviruses(RVs)Exogenous retroviruses
(HIV, HTLV)
Retroviridae (positive diploid ssRNA, linear)Dysregulation of apoptosis and molecular mimicry (HIV), regulation of CD4 expression (HTLV-1)Highly active antiretroviral therapy (HAART) (HIV); nucleoside/nucleotide reverse transcriptase inhibitors associated with IFN useful in HTLV-associated haematological diseases, even if prone to relapses. Insufficient evidence to support use of antiretroviral therapy for the treatment of HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP)[48,49,50,51]
Human Endogenous Retroviruses
(HERV)
Retroviridae (integrated in the host genome)Molecular mimicry, defects in
IFN-stimulatory DNA pathways
HAART therapy for HIV may also blunt activation of some HERVs[43,52,53,54]
Table 3. EBV proteins involved in immune system evasion and autoimmunity induction.
Table 3. EBV proteins involved in immune system evasion and autoimmunity induction.
ProteinMechanismReferences
Viral IL-10 homologueIt inhibits IFN-γ production, CD8+ cytotoxic T-cells and MHC-I expression[62]
EA/RIt is a viral Bcl-2 homologue which confers resistance to apoptosis[64]
LMP-1 and LMP-2AThey rescue infected B cells from apoptosis and are involved in molecular mimicry.
LMP-1 triggers IFN-α production by pDCs through TLRs and
mediates PDL-1 overexpression in neutrophils.
LMP-1 renal expression is increased in LN and correlates with severity.
[17,66,67,68,69,70,71,72,73]
EBNA-1It can induce autoimmunity through molecular mimicry with C1q[74,75]
EBNA-2It is a TF which controls all other latent viral genes.
50% of SLE-predisposing loci can be occupied by EBNA-2, suggesting a key role in AIDs
[76]
Table 4. Peculiar alterations of EBV-specific immune response in SLE.
Table 4. Peculiar alterations of EBV-specific immune response in SLE.
Type of Alteration CommentReferences
Reduced EBV-specific CD8+ lymphocyte response and increased CD4+ lymphocyte responseImpaired cytotoxic potential of EBV-specific CD8+ lymphocytes is due to a SLE-intrinsic defect and is probably the primum movens of altered immune response [78,79,80,81,82,83]
Decreased Th17 and Treg responseImbalance between Th17 and T reg is a major cause of AIDs and decrease in Th17 may be an important feature of EBV/CMV infection.[84]
Elevated EBV viral load in B cells and PBMC and aberrant expression of viral mRNAs of lytic (BZLF-1, BLLF-1) and latent phase proteins (LMP-1, LMP-2 and EBNA-1) It is due to impaired control of EBV infection by CTLs, with frequent viral reactivations (BZLF-1, BLLF-1) and abnormal latency state (LMP-1, LMP-2,
EBNA-1)
[82,85,86,87,88,89]
Elevated EBV seroprevalenceEBV seroprevalence is very high especially in certain populations, in which prior EBV infection appears necessary to develop SLE[90,91,92,93,94]
Elevated titers of antibody against early lytic (EA/D; EA/R) and latent (EBNA) EBV antigens This could represent an enhanced compensatory humoral response secondary to an inadequate T-cell control of a chronic EBV infection, with frequent reactivations. It can be associated with production of autoantibodies. [75,89,91,95,96,97,98]
Elevated prevalence of IgA against EA/D antigen and coexistence of different EBV-specific immunoglobulin isotypesIt may indicate disseminated EBV infection, with higher lytic rate of epithelial cells (IgA) and lymphocytes (IgG).[89,99,100]
Temporal relationship between anti-EBV antibodies and SLE manifestationsAnti-EBV humoral response can precede SLE onset or flareups.
It identifies first-degree unaffected family members of SLE patients who are at risk of transitioning to SLE
[75,101,102,103]
Coexistence of anti-EBV antibodies and SLE-specific autoantibodies Cross-reactivity between EBV- and self-antigens explains associations between anti-EA/D and anti-Ro/anti-La antibodies, also observed in Sjögren’s syndrome, and between anti-EBNA-1 and anti-C1q.[74,93,104,105]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Quaglia, M.; Merlotti, G.; De Andrea, M.; Borgogna, C.; Cantaluppi, V. Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story. Viruses 2021, 13, 277. https://doi.org/10.3390/v13020277

AMA Style

Quaglia M, Merlotti G, De Andrea M, Borgogna C, Cantaluppi V. Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story. Viruses. 2021; 13(2):277. https://doi.org/10.3390/v13020277

Chicago/Turabian Style

Quaglia, Marco, Guido Merlotti, Marco De Andrea, Cinzia Borgogna, and Vincenzo Cantaluppi. 2021. "Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story" Viruses 13, no. 2: 277. https://doi.org/10.3390/v13020277

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