Next Article in Journal
Difference between Selenite and Selenate in the Regulation of Growth and Physiological Parameters of Nickel-Exposed Lettuce
Next Article in Special Issue
Neuroprotective Effects of Extracts from Tiger Milk Mushroom Lignosus rhinocerus Against Glutamate-Induced Toxicity in HT22 Hippocampal Neuronal Cells and Neurodegenerative Diseases in Caenorhabditis elegans
Previous Article in Journal
Lymphatic Vessels and Their Surroundings: How Local Physical Factors Affect Lymph Flow
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Relationship of the Mechanisms of the Pathogenesis of Multiple Sclerosis and the Expression of Endogenous Retroviruses

by
Vera R. Lezhnyova
1,
Ekaterina V. Martynova
1,*,
Timur I. Khaiboullin
2,
Richard A. Urbanowicz
3,4,
Svetlana F. Khaiboullina
1,5 and
Albert A. Rizvanov
1
1
Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
2
Republican Clinical Neurological Center, Republic of Tatarstan, 420021 Kazan, Russia
3
Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham NG7 2RD, UK
4
School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
5
Department of Microbiology and Immunology, University of Nevada, Reno, NV 89557, USA
*
Author to whom correspondence should be addressed.
Biology 2020, 9(12), 464; https://doi.org/10.3390/biology9120464
Submission received: 25 November 2020 / Revised: 7 December 2020 / Accepted: 10 December 2020 / Published: 11 December 2020

Abstract

:

Simple Summary

Multiple sclerosis is a neurodegenerative disease of the central nervous system, develops at an early age and often leads to a disability. The etiological cause of the disease has not been fully elucidated, and as a result, no effective treatment is available. This review summarizes the current knowledge about the relationship between the expression of human endogenous retroviruses and the pathogenesis of multiple sclerosis. The epigenetic mechanisms of transcriptional regulation, the role of transcription factors, cytokines, and exogenous viruses are also addressed in this review. The elucidation of the mechanisms of an increase in endogenous retrovirus expression in multiple sclerosis could help to develop therapeutic strategies and novel methods for early diagnosis and treatment of the disease.

Abstract

Two human endogenous retroviruses of the HERV-W family can act as cofactors triggering multiple sclerosis (MS): MS-associated retrovirus (MSRV) and ERVWE1. Endogenous retroviral elements are believed to have integrated in our ancestors’ DNA millions of years ago. Their involvement in the pathogenesis of various diseases, including neurodegenerative pathologies, has been demonstrated. Numerous studies have shown a correlation between the deterioration of patients’ health and increased expression of endogenous retroviruses. The exact causes and mechanisms of endogenous retroviruses activation remains unknown, which hampers development of therapeutics. In this review, we will summarize the main characteristics of human endogenous W retroviruses and describe the putative mechanisms of activation, including epigenetic mechanisms, humoral factors as well as the role of the exogenous viral infections.

1. Endogenous and Exogenous Factors Affecting the Development of MS

In recent years, the incidence and prevalence rates of multiple sclerosis (MS), a neurodegenerative disease, have increased worldwide. MS is a chronic, heterogeneous, immune-associated disease characterized by the formation of loci of inflammation and damage of myelin sheath of nerve fibers in the brain and spinal cord, leading to a persistent disability [1]. There are multiple hypotheses proposed to establish the etiology of MS, showing the multifactorial nature of the disease. There are several risk factors associated with the disease including gender (often diagnosed in women) and age (most common in young age) of the patient [2,3]. Hormone levels have also been implicated in late onset of the disease, as the first episode of MS neurological symptoms is often diagnosed in older women who have been pregnant as compared to those who have not [4,5]. The hormones appear to contribute to decreased relapse rates during pregnancy and reoccurring after the delivery [6]. Numerous risk factors and the multifactorial etiology of the disease makes early diagnosis challenging [7]. This makes development of the etiotropic therapeutics particularly difficult.
Several endogenous and exogenous risk factors have been identified for this disease. One potential endogenous risk factor of the illness is vitamin D deficiency [8]. Vitamin D is synthesized in the skin, where ultraviolet radiation initiates the first non-enzymatic reaction of the synthesis of the precursor of the active form of vitamin D, calcitriol. Numerous studies have shown a correlation between a decrease in the level of ultraviolet radiation with an increase of the latitude of the region of residence and an increase in the risk of MS and relapse of disease [9,10,11]. The role of vitamin D in MS was further confirmed by Ramagopalan et al., as an increased binding of vitamin D receptor (VDR) was found on genes associated with risk of MS [12]. Another endogenous factor contributing to the risk of MS is believed to be immune auto-aggression against self-proteins. It has been shown that cytotoxic T-lymphocytes are involved in the formation of loci of inflammation and demyelination, and are also associated with the severity of MS [13]. In vitro experiments have shown a potential connection between vitamin D and immune responses in MS. It has been demonstrated that human dendritic cells, in the presence of calcitriol, increase expression of CD31, which can reduce their ability to prime CD4+ T lymphocytes [14]. Low levels of vitamin D can disrupt this regulation on CD4+ T lymphocyte activity, leading to an increased production of pro-inflammatory cytokines, often found in MS [15]. The activity of the TAGAP gene (T-Cell Activation RhoGTPase Activating Protein), which is responsible for the Cdc42-dependent maturation of CD4 + T-lymphocytes [16], can be suppressed by direct exposure to calcitriol [17], suggesting an additional mechanism of vitamin D contribution to pathogenesis of MS.
Genetic predisposition and accumulation of mutations in genes associated with MS could play an essential role in pathogenesis of the disease. GWAS (genome-wide association studies) data have shown a missense mutation in the CD226 gene in MS leads to an amino acid change in the DNAM-1 receptor, thereby blocking or changing its function [18]. This receptor is responsible for NK cells’ cytolytic activity upon interaction with the poliovirus receptor (PVR) on the surface of activated CD4+ T lymphocytes in MS [19]. The single-nucleotide polymorphism (rs243324) effects expression in the 5′-regulatory region of the SOCS1 gene [20], which is involved in regulating cytokine signals, and has impaired expression in patients with MS [21].
Exogenous factors also include viral triggers of MS. There are many hypotheses that explain the possible mechanism of MS caused by viruses. HHV-4 (EBV) virus is one of those most commonly linked to MS, as was shown by the theory of molecular mimicry [22,23]. A higher frequency of cross-reactive T cell progenitors to myelin basic protein and HHV-6 in MS patients compared to controls has been shown [24]. In vivo experiments have demonstrated that stimulation of CD4 + CD28null T cells by human cytomegalovirus (CMV), another member of family Herpesviridae, causes them to proliferate, which can initiate migration of lymphocytes and damage to the central nervous system [25].
Other exogenous factors of MS could be bacterial infections, as microbial PAMPs can activate antigen presenting cells (APCs) and subsequently cause proliferation of autoreactive T and B lymphocytes. Similar to the molecular mimicry seen with HHV-4, the epitopes of some bacterial peptides are similar to host antigens and serve to form autoreactive T-cells [26]. Evidence of this was the detection of a significant immune response to the protein Mycobacterium avium subsp. paratuberculosis, which has homology with the C region of the gamma chain of the T cell receptor, in MS [27]. Some studies show a correlation between the incidence of MS and the presence of antibodies to C. pneumoniae, but the exact role of these antibodies in pathogenesis of the disease remains unclear [28,29]. In vivo studies have shown that microbiota from MS twins transplanted to transgenic mice with the T cell receptor specific for myelin oligodendrocyte glycoprotein (MOG), caused a higher level of autoimmunity than the microbiota obtained from healthy twins [30]. These data provide strong evidence on the role of bacteria in the pathogenesis of MS.
Many studies have shown the relationship between various neurodegenerative diseases (MS, amyotrophic lateral sclerosis, Alzheimer’s disease) and expression of endogenous retroviruses genes [31,32]. The expression of endogenous retroviruses can be detected not only in healthy people, for example during the pregnancy [33], but also in various pathologies [34]. Expression of the HERV-W (MS-associated retrovirus env) protein was found in the brain tissue of all studied MS patients during active demyelination at an early stage and during late disease progression [35]. Additionally, the HERV-Wenv protein was found in the serum, infiltrated macrophages, perivascular infiltrates and activated microglial cells of patients with various forms of MS [36]. In a study by Kremer et al., it was found that microglial phagocytosis was inhibited by the HERV-Wenv protein, which in turn leads to the accumulation of myelin debris, which prevents neuronal remyelination [37]. Additionally, this protein reduces the expression of neuroprotective molecules by microglial cells [37]. It is worth noting that HERV-W Env epitopes were discovered on the surface of B cells and monocytes in samples of patients with the active and stable relapsing–remitting form of the disease [38]. The fact that the HERV-Wenv protein is found in the brain tissues, as well as in the serum and cerebrospinal fluid [39] of patients with MS, suggests that it is in volved in the pathogenesis of the disease and should be carefully studied.

2. General Characteristics of HERV-W

In the early 1970s, endogenous retroviral elements were discovered, which, as was shown later, were attained by our ancestors more than 25 million years ago and are constitutively present in the human genome [40,41,42]. These genetic elements were attained in human DNA as the result of the integration of exogenous retroviral RNA followed the reverse transcription [43]. It was suggested that the expression of these viral proteins have had a significant impact on human evolution [32,44]. For example, the glycosylated product of the ERVWE1 gene, located on chromosome 7q21.2 [45], is the fusogenic protein Syncytin-1 (or enverin), which is involved in the fusion of cell membranes during the formation of syncytiotrophoblast during embryogenesis [33].
Human endogenous retroviruses (HERVs) are present in the genome in the form of a provirus and are inherited by vertical gene transfer [46]. From the moment of integration into the human genome, endogenous retroviruses have accumulated many mutations. That is why almost all retroviral inserts currently do not have open reading frames (ORFs), which are necessary for virus replication [47]. All HERVs are grouped into three classes based on the similarity of their sequences with infectious retroviruses [48]. The HERV terminology is based on the type of tRNA that binds to the primer binding site (PBS) [49].
In the context of MS, the study of HERV-W env gene expression is of most interest, since its product can activate the innate immune response through interaction with the TLR-4 receptor [50]. This process contributes to the activation of nitrosative stress, leading to impaired differentiation of oligodendrocytes and impaired remyelination of neurons [51]. In an in vitro study, the expression of the MS-associated retrovirus (MRSV) recombinant protein in the brain microvascular endothelial cells increased the expression of ICAM-1, which is associated with the interaction of the MSRV protein with TLR-4 receptors on the surface of endothelial cells [52]. The HERV-W family has approximately 654 elements that are permanently present in the human genome [53]. In the assembly of the human genome version GRCh37/hg19, 135 pseudogenes and 65 HERV-W proviruses were detected [54]. ORFs for HERV-W env were found on Xq22.3 [55] and 7q21.2 [56], chromosomes encoding the incomplete HERV-W envelope protein and Syncytin-1, respectively. HERV-W env was found in macrophages infiltrating the brain parenchyma and in perivascular infiltrate in patients with MS [36]. The exact reasons for the increased expression of the HERV-W family genes in MS are currently unknown.

3. The Structure of the HERV-W Family and the ERVWE1 Gene

The HERV-W consists of four genes: gag, pro, pol, env (Figure 1). The gag gene encodes the structural components of the matrix, capsid and nucleocapsid, while the pro gene encodes the protease, the pol gene encodes for the reverse transcriptase and integrase, and the env gene encodes the precursor of the envelope glycoprotein. At the 5′ and 3′ ends, these genes are flanked by two identical long terminal repeats (LTR) [57]. LTRs consist of a U3 region containing a primer binding site (PBS), which uses tRNATrp to synthesize negative DNA strands, CAAT, and TATA box binding sites for transcription factors [58]. Additionally, in the U3 region of the 5′LTR ERVWE1, there is a phosphorylated cAMP response element-binding protein (CREB) binding region that is active in all cell types, as well as Sp-1, AP-2 binding sites containing GC-rich domains, GATA, Oct-1 and PPAR-γ/RXR [59]. The repeat (R) sequences region of ERVWE1 contains a CAP transcription initiation site at the 5′ end and a polyadenylation site at the 3′ end [58], and it also serves as the primary site for transcription initiation [59]. Recently, an AG-rich non-coding, 2kb pre-gag region between the 5′-LTR and the ERVWE1 gag gene, whose functions are currently unknown, has been discovered [54]. Additionally, upstream regulatory element (URE), relevant to ERVWE1, includes the trophoblast specific enhancer (TSE) containing AP-2, Sp-1, and GCMa binding sites and flanked by MaLR (mammalian apparent LTR retrotransposon). The cAMP/PKA pathway’s role has also been demonstrated in the regulation of LTR promoter activity in HERV-W env [59]. Interestingly, Schmitt et al. provided evidence of the promoter activity of incomplete LTR sequences [60]. It was also shown that the LTRs with a U3R region were capable of generating antisense transcripts, while LTRs with a U5R region can facilitate sense gene transcription [60].

4. Epigenetic Mechanisms of Regulation of HERV-W Expression

4.1. RNA Interference

Understanding the epigenetic mechanisms helps to establish the relationship between pathological conditions and changes in gene expression regulation. MicroRNAs (miRNAs), which are part of the RNA-induced silencing complex (RISC), can inhibit gene expression at the level of transcription by the RNA interfering mechanism [61] and translational level [62], by suppression of mobile genetic elements replication, including HERV [63]. miRNA can bind to the 3′-end of the untranslated region (UTR) of the target mRNA leading to mRNA cleavage or prevention of subsequent translation [64] (Figure 2).
Currently, there are contradictory data about miRNA effects on development of MS and HERV-W expression. A significant decrease in miRNA-17 and miRNA-20a levels was found in the whole blood from patients with primary progressive (PP), secondary progressive (SP), and relapsing–remitting MS (RRMS) [65]. It was shown that these miRNAs are involved in regulation of genes responsible for T cell activation in MS [65]. Additionally, treatment of RRMS with a monoclonal antibody, natalizumab, decreased miRNA-17 level in CD4+ cells during remission, while it increased during the relapse phase of the disease [66]. Interestingly, the use of natalizumab led to a gradual decrease in the level of MSRV env transcripts and syncytin-1 in peripheral blood mononuclear cells of MS [67]. These conflicting results could be explained by the fact that each study used different patient populations without considering the relapse phase of disease.
Nineteen miRNAs were found to have more than 80% homology to HERV-W [68]. Their expression correlates with the low replication of HERVs genes in healthy tissues [68]. Huang and colleagues found short interfering RNAs (siRNAs) with a high level of time-dependent knockdown effect on syncytin-1 mRNA in vitro [33]. It could be suggested that the mechanism of RNA interference could indirectly affect the expression of HERVs. This assumption is supported by the finding that siRNA to the p65 gene of the NF-κB subunit could reduce URE-LTR activation of the ERVWE1 promoter by TNFα [69].

4.2. Citrullination

Citrullination is one of the mechanisms of post-translational modification of proteins catalyzed by proteins of the Peptidyl Arginine Deiminase (PAD) family. Heterochromatin protein 1 α (HP1α) recognizes methylated histone H3K9 accumulated on the HERV-W/ERVWE1 LTR, which leads to transcription repression (Figure 3A). An elevated level of citrullinated histone H3cit8K9me3 has been detected in the HERV-W/ERVWE1 promoter region in MS [70]. It has also been shown that an increased PADI4 expression could contribute to upregulation of HERV-W/ERVWE1, by converting the H3K9me3 histone to the citrullinated form H3cit8K9me3 (Figure 3B). Local increased citrullination of H3R8 histone, located upstream of H3K9, could inhibit the binding of HP1α to H3K9me3 histone, which has been shown to be associated with increased expression of HERV-W/ERVWE1 [70].
Antibodies to the myelin basic protein (MBP) have been detected in the central nervous system of MS patients [71]. An increase in the level of citrullinated MBP has been detected in the white matter of the brain of MS patients [72,73], ranging from 45% to 80%, compared to 18% found in healthy controls [74,75]. Citrullination changes the conformation of MBP and thus contributes to the formation of a weaker calmodulin binding site [72], which can affect the Ca2+-dependent activation of the large-conductance Ca(2+)-activated K(+) (BKCa) channels involved in depolarization of the cell membrane [73]. These data suggest that the changes in citrullination of MBP could be used as a biomarker for diagnosis of MS as well as for evaluation of the disease progression.
Peptidylarginine deiminase (PADI)-2 and PADI4 proteins responsible for citrullination were found expressed on the myelin sheath, where PADI2 appears to be more citrullinated on the arginine residues as compared to PADI4 [76]. Each substitution of arginine with citrulline causes a change in the MBP charge, which makes it more difficult to interact with the phospholipid layer of the myelin sheath [76]. In MS, the amount of PADI2 and PADI4, and, therefore, citrullinated proteins, has been shown to be increased [76]. A larger amount of PADI2 is localized in the periaxonal regions, which could explain why this end of the oligodendrocyte is the first to undergo to apoptosis [76]. However, it appears that citrullination of MBP has a limited role in activating T lymphocytes, and, therefore, unlikely to play a role in the development of MS [77]. Therefore, the role of citrullination of MBP in the pathogenesis of MS remains unclear.
MS patients have a persistent visual impairment, which is mainly associated with bilateral internuclear ophthalmoplegia and bilateral visual neuropathy [78]. The increased intraocular pressure found in MS [79], may be due to an increased intracellular calcium level. Interestingly, it was shown that an increased intraocular pressure could lead to upregulation of PADI2 expression [80]. Elevated levels of PADI2 and protein deimination were shown to be associated with pathological changes in MS [81], suggesting a role of this protein in pathogenesis of the disease.

4.3. Methylation and Acetylation

DNA methylation at the 5th position of the pyrimidine ring within the cytosine CpG site is a mechanism for gene expression regulation. Hypermethylation of the CpG site in the coding region of the gene can enhance expression [82]. Additionally, hypermethylation in the promoter or enhancer region makes the region heterochromatic, preventing transcription activators to bind to it, which leads to silencing of that gene [83] (Figure 4). Thus, hypermethylation in the LTR region of HERV-W can potentially inhibition the expression of these genes. It has been demonstrated that inactive unmethylated CpG sites in the promoter region can correlate with an increased Lys4 dimethylation in H3 histone, which may indicate that this mechanism could protect DNA from methylation [84]. In MS, a decreased expression of the EHMT2 gene encoding the histone H3 methyltransferase has been demonstrated [85]. Interestingly, methylation of this gene has been shown to be necessary for repression of HERV-W transcription [70]. An increased EHMT2 expression is also found in the late stages of oligodendrocyte differentiation, and shown to contribute to H3K9 methylation and chromatin compaction necessary for differentiation of oligodendrocyte progenitor cells (OPCs) [86], which is impaired in patients with MS [87].
The role of DNA methylation transcription of HERVs has been extensively studied. HERVs’ retroelements that are expressed in many types of tumors are less methylated than those in healthy tissues [88]. Hypomethylation of LTRs has been demonstrated in placental tissues, normally expressing ERVWE1, while in other tissues this region is strongly methylated [89]. Weber et al. showed that the insertion of foreign DNA can affect the methylation of CpG sites in various regions of human genes and change their expression [90]. They later found that transfection of cells with the same foreign DNA did not affect methylation of CpG sites and the level of HERV-W transcription [91]. These results suggest that exogenous retroviral elements are not affecting the expression of endogenous retroviruses by using the CpG site methylation mechanism.
An increased level of expression of DNA methyltransferase-3 (DNMT3B) beta subunits has been detected in MS [92], as has a lower demethylase content in the demyelinated hippocampus compared to the myelinated [92]. As a result of demyelination, a decrease in the methylation state of genes responsible for neuronal death, coupled with an increase in the methylation of genes responsible for neuron survival was demonstrated [92]. It is known that CD8+ T lymphocytes can be locally reactivated at lesions in MS [93]. A study of the degree of methylation of CpG sites in CD4+ and CD8+ T lymphocytes revealed significant hypermethylation in the DNA of CD8+ T lymphocytes in MS, especially at the location upstream of the transcription start site (TSS) and the first exon of genes [94].
Histone acetylation is a mechanism for regulating gene expression. The acetylation of the lysine in the N-terminal residue removes the positive charge from the histone [95], which reduces the strength of its interaction with the negatively charged phosphate groups in the DNA. This results in the chromatin becoming less compact, which facilitates the transcription of genes [95]. Interestingly, treatment of cells with histone deacetylase inhibitors (HDACs) did not increase HERV expression; however, when panobinostat, a histone acetylase inhibitor, was used, expression of HERV-W env was suppressed [96].

5. Changes in LTR Regions and the Effects of Transcription Factors

LTRs are the main regulatory elements of endogenous human retroviruses [58]. Several mutations in the 3’-LTR region are associated with an increased expression of ERVWE1 mRNA as well as protein [97]. For example, the 142T > C mutation was shown to promote the formation of an additional binding site with c-Myb in the 3′-LTR and increase its activity [97]. However, analysis of the expression of proto-oncogenes in the white matter of the MS brain did not find a significant increase in c-Myb expression [98], while expression of c-Fos increased [98]. The increased expression of the HERV-W loci is explained by stimulation of the U3 region initiated by the 5′R LTR region [99]. The number of HERV transcripts from the 3′ end were lower than from the 5′ end, which could be explained by the distance from the U3 initiation site of reverse transcription [57]. It should be noted that intron proviral elements with single LTRs showed a higher level of transcription than elements having both LTRs [57]. Therefore, the role of these regulatory regions in HERV transcription requires further study.

6. The Role of the Immune System in Activation of HERV-W Transcription

Recently, the role of cytokines in the activation of endogenous retroviruses was demonstrated in MS. It was found that cytokines often associated with MS, such as tumor necrosis factor α (TNFα) and interferon-γ (IFN-γ), activate the ERVWE1 promoter [69]. The highest activation of HERV-W expression in glioblastoma cells was detected when exposed to TNFα [69]. The mechanism of transcription activation by this cytokine was shown as associated with an increased binding of p65 of the NF-κB subunit to a sensitive element located in the enhancer region of the ERVWE1 promoter [69].
The permeability of the blood–brain barrier (BBB) is inextricably linked with the relapse in patients with RRMS [100]. The CD9 protein, a tetraspanin family member, the expression of which is increased in the loci of inflammation in MS brains, has been shown to be involved in BBB permeability [101]. Overexpression of CD9 increases interaction with the G protein, which activates the cAMP/PKA signaling pathway, and this in turn leads to activation of Glial Cells Missing Transcription Factor 1 (GCM1), which binds to the regulatory region of ERVWE1 and enhances gene expression [102,103] (Figure 5).

7. Activation of HERV-W Transcription upon Exposure to Exogenous Viruses

The effect of virus infection on the activation of endogenous human virus transcription has been extensively investigated. Studies have shown that human immunodeficiency virus (HIV) is commonly associated with HERV-W expression [104]. It was found that HIV trans-activator of transcription (Tat) protein increases the level of MSRVenv and HERV-Wenv mRNA in blood cells and astrocytes [104]. In monocytes, Tat can stimulate MSRVenv and inhibit ERVWE1, yet when differentiated into macrophages, the expression of both retroviral elements has been demonstrated. In astrocytes, HIV indirectly activates transcription of MSRV and ERVWE1 due to interaction with TLR4 and the induction of TNFα [104]. Therefore, these expression on retroviral elements and worsening MS symptoms could be explained by antiretroviral therapy in patients with HIV [105].
Human cytomegalovirus (HCMV) could also be implicated in activation of HERV-W expression. It was found that when HCMV enters the cell, the reverse transcriptase, required for transcription of HERV, appears to be activated [106]. In vitro experiments have demonstrated the effect of the pleiotropic trans-activator of genes (Infected Cell Polypeptide 0 (ICP0)), which is synthesized shortly after infection with herpes simplex virus 1 (HSV-1), on the transcriptional activation of both HERV-Wenv and MSRVenv genes [107]. Human Herpesvirus 6A (HHV-6A) can activate MSRVenv expression by interacting with the extracellular domains of SCR3 and SCR4 of the isoform of CD46-Cyt1 receptor phosphorylated by tyrosine protein kinase C [108]. However, none of the exogenous viruses have been identified as a sole etiological factor of MS.

8. Conclusions

The pathogenesis of MS is still the subject of debate among scientists and health care providers. Symptomatic and pathogenetic treatment used in MS does not completely abrogate the neurodegeneration and the progression of the disease. Currently, MS is characterized as a multifactorial disease, in which a key role can be attributed to the external and genetic factors. One of the unresolved questions of the disease pathogenesis is the lack of understanding of the mechanism of HERV-W expression in MS, especially in the inflammatory foci within the brain. The elucidation of the mechanisms of an increased HERV-W expression in MS could help to develop therapeutic strategies and novel methods for early diagnosis and treatment of the disease.
In this review, we summarized the mechanisms that can affect the expression of HERV-Wenv and ERVWE1 (Table 1), as well as link them to the pathogenesis of MS. We reviewed the epigenetic mechanisms of transcriptional regulation, the role of transcription factors, cytokines, and exogenous viruses. However, it still remains difficult to accurately identify the cause of the elevated expression of the HERV-W genes in MS. The exact role of these retroviral elements in the pathogenesis of MS is still unknown and therefore further research in this area using multi-cohort studies, GWAS, and in vitro approaches will help to establish the contribution of HERVs in MS pathogenesis.

Author Contributions

Conceptualization, V.R.L., A.A.R., T.I.K.; Writing—original draft preparation, V.R.L., E.V.M.; Writing—review and editing, V.R.L., E.V.M., S.F.K., R.A.U. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the DNA Research Center, Autonomous Nonprofit Organization, Russia and by the Russian Government Program of Competitive Growth.

Conflicts of Interest

The authors declare that they have no conflict of interests.

References

  1. Wallin, M.T.; Culpepper, W.J.; Nichols, E.; Bhutta, Z.A.; Gebrehiwot, T.T.; Hay, S.I.; Khalil, I.A.; Krohn, K.J.; Liang, X.; Naghavi, M.; et al. Global, regional, and national burden of multiple sclerosis 1990–2016: A systematic analysis for the Global Burden of Disease Study. Lancet Neurol. 2019, 18, 269–285. [Google Scholar] [CrossRef] [Green Version]
  2. Rosati, G.; Pinna, L.; Granieri, E.; Aiello, I.; De Bastiani, P.; Tola, R.; Agnetti, V.; Pirisi, A. The distribution of multiple sclerosis in Sardinia. Rivista di Patologia Nervosa e Mentale 1977, 98, 46–64. [Google Scholar]
  3. Houzen, H.; Niino, M.; Hirotani, M.; Fukazawa, T.; Kikuchi, S.; Tanaka, K.; Sasaki, H. Increased prevalence, incidence, and female predominance of multiple sclerosis in northern Japan. J. Neurol. Sci. 2012, 323, 117–122. [Google Scholar] [CrossRef] [PubMed]
  4. McCombe, P.A.; Greer, J.M. Female reproductive issues in multiple sclerosis. Mult. Scler. J. 2013, 19, 392–402. [Google Scholar] [CrossRef] [PubMed]
  5. Nguyen, A.-L.; Vodehnalova, K.; Kalincik, T.; Signori, A.; Havrdova, E.K.; Lechner-Scott, J.; Skibina, O.G.; Eastaugh, A.; Taylor, L.; Baker, J.; et al. Association of Pregnancy with the Onset of Clinically Isolated Syndrome. JAMA Neurol. 2020. [Google Scholar] [CrossRef] [PubMed]
  6. Vukusic, S.; Hutchinson, M.; Hours, M.; Moreau, T.; Cortinovis-Tourniaire, P.; Adeleine, P.; Confavreux, C. Pregnancy and multiple sclerosis (the PRIMS study): Clinical predictors of post-partum relapse. Brain 2004, 127, 1353–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.M.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.S.; et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  8. Smolders, J.; Menheere, P.; Kessels, A.; Damoiseaux, J.; Hupperts, R. Association of vitamin D metabolite levels with relapse rate and disability in multiple sclerosis. Mult. Scler. J. 2008, 14, 1220–1224. [Google Scholar] [CrossRef]
  9. Becker, J.; Callegaro, D.; Lana-Peixoto, M.A.; Talim, N.; Vidaletti, T.; Correa, M.D.P.; Gomes, I. Hypovitaminosis D association with disease activity in relapsing remitting multiple sclerosis in Brazil. J. Neurol. Sci. 2016, 363, 236–239. [Google Scholar] [CrossRef]
  10. Simpson, S., Jr.; Van Der Mei, I.; Lucas, R.M.; Ponsonby, A.-L.; Broadley, S.; Blizzard, L.; Taylor, B.; Dear, K.; Dwyer, T.; Taylor, B.V. Sun exposure across the life course significantly modulates early multiple sclerosis clinical course. Front. Neurol. 2018, 9, 16. [Google Scholar] [CrossRef] [Green Version]
  11. Tremlett, H.; Zhu, F.; Ascherio, A.; Munger, K.L. Sun exposure over the life course and associations with multiple sclerosis. Neurology 2018, 90, e1191–e1199. [Google Scholar] [CrossRef] [PubMed]
  12. Ramagopalan, S.V.; Heger, A.; Berlanga, A.J.; Maugeri, N.J.; Lincoln, M.R.; Burrell, A.; Handunnetthi, L.; Handel, A.E.; Disanto, G.; Orton, S.-M. A ChIP-seq defined genome-wide map of vitamin D receptor binding: Associations with disease and evolution. Genome Res. 2010, 20, 1352–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Peeters, L.M.; Vanheusden, M.; Somers, V.; Van Wijmeersch, B.; Stinissen, P.; Broux, B.; Hellings, N. Cytotoxic CD4+ T Cells Drive Multiple Sclerosis Progression. Front. Immunol. 2017, 8, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Saul, L.; Mair, I.; Ivens, A.; Brown, P.; Samuel, K.; Campbell, J.D.M.; Soong, D.Y.; Kamenjarin, N.; Mellanby, R.J. 1,25-Dihydroxyvitamin D3 Restrains CD4+ T Cell Priming Ability of CD11c+ Dendritic Cells by Upregulating Expression of CD31. Front. Immunol. 2019, 10, 600. [Google Scholar] [CrossRef]
  15. Cao, Y.; Goods, B.A.; Raddassi, K.; Nepom, G.T.; Kwok, W.W.; Love, J.C.; Hafler, D.A. Functional inflammatory profiles distinguish myelin-reactive T cells from patients with multiple sclerosis. Sci. Transl. Med. 2015, 7, 287ra74. [Google Scholar] [CrossRef] [Green Version]
  16. Guo, F.; Hildeman, D.; Tripathi, P.; Velu, C.S.; Grimes, H.L.; Zheng, Y. Coordination of IL-7 receptor and T-cell receptor signaling by cell-division cycle 42 in T-cell homeostasis. Proc. Natl. Acad. Sci. USA 2010, 107, 18505–18510. [Google Scholar] [CrossRef] [Green Version]
  17. Berge, T.; Leikfoss, I.S.; Brorson, I.S.; Bos, S.D.; Page, C.M.; Gustavsen, M.W.; Bjolgerud, A.; Holmoy, T.; Celius, E.G.; Damoiseaux, J.; et al. The multiple sclerosis susceptibility genes TAGAP and IL2RA are regulated by vitamin D in CD4+ T cells. Genes Immun. 2016, 17, 118–127. [Google Scholar] [CrossRef] [Green Version]
  18. Alcina, A.; Vandenbroeck, K.; Otaegui, D.; Saiz, A.; Gonzalez, J.R.; Fernandez, O.; Cavanillas, M.L.; Cenit, M.C.; Arroyo, R.; Alloza, I.; et al. The autoimmune disease-associated KIF5A, CD226 and SH2B3 gene variants confer susceptibility for multiple sclerosis. Genes Immun. 2010, 11, 439–445. [Google Scholar] [CrossRef]
  19. Gross, C.C.; Schulte-Mecklenbeck, A.; Rünzi, A.; Kuhlmann, T.; Posevitz-Fejfár, A.; Schwab, N.; Schneider-Hohendorf, T.; Herich, S.; Held, K.; Konjević, M. Impaired NK-mediated regulation of T-cell activity in multiple sclerosis is reconstituted by IL-2 receptor modulation. Proc. Natl. Acad. Sci. USA 2016, 113, E2973–E2982. [Google Scholar] [CrossRef] [Green Version]
  20. Vandenbroeck, K.; Alvarez, J.; Swaminathan, B.; Alloza, I.; Comabella, M.; Urcelay, E.; Comabella, M.; Alcina, A.; Fedetz, M.; Ortiz, A.M.; et al. A cytokine gene screen uncovers SOCS1 as genetic risk factor for multiple sclerosis. Genes Immun. 2011, 13, 21–28. [Google Scholar] [CrossRef] [Green Version]
  21. Toghi, M.; Taheri, M.; Arsang-Jang, S.; Ohadi, M.; Mirfakhraie, R.; Mazdeh, M.; Sayad, A. SOCS gene family expression profile in the blood of multiple sclerosis patients. J. Neurol. Sci. 2017, 375, 481–485. [Google Scholar] [CrossRef] [PubMed]
  22. Lünemann, J.D.; Jelčić, I.; Roberts, S.; Lutterotti, A.; Tackenberg, B.; Martin, R.; Münz, C. EBNA1-specific T cells from patients with multiple sclerosis cross react with myelin antigens and co-produce IFN-γ and IL-2. J. Exp. Med. 2008, 205, 1763–1773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Angelini, D.F.; Serafini, B.; Piras, E.; Severa, M.; Coccia, E.M.; Rosicarelli, B.; Ruggieri, S.; Gasperini, C.; Buttari, F.; Centonze, D.; et al. Increased CD8+ T Cell Response to Epstein-Barr Virus Lytic Antigens in the Active Phase of Multiple Sclerosis. PLOS Pathog. 2013, 9, e1003220. [Google Scholar] [CrossRef] [PubMed]
  24. Tejada-Simon, M.V.; Zang, Y.C.Q.; Hong, J.; Rivera, V.M.; Zhang, J. Cross-reactivity with myelin basic protein and human herpesvirus-6 in multiple sclerosis. Ann. Neurol. 2003, 53, 189–197. [Google Scholar] [CrossRef] [PubMed]
  25. Vanheusden, M.; Broux, B.; Welten, S.P.M.; Peeters, L.M.; Panagioti, E.; Van Wijmeersch, B.; Somers, V.; Stinissen, P.; Arens, R.; Hellings, N. Cytomegalovirus infection exacerbates autoimmune mediated neuroinflammation. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
  26. Oldstone, M.B. Molecular Mimicry: Its Evolution from Concept to Mechanism as a Cause of Autoimmune Diseases. Monoclon. Antibodies Immunodiagn. Immunother. 2014, 33, 158–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Cossu, D.; Cocco, E.; Paccagnini, D.; Masala, S.; Ahmed, N.; Frau, J.; Marrosu, M.G.; Sechi, L.A. Association of Mycobacterium avium subsp. paratuberculosis with Multiple Sclerosis in Sardinian Patients. PLOS ONE 2011, 6, e18482. [Google Scholar] [CrossRef]
  28. Ivanova, M.V.; Kolkova, N.I.; Morgunova, E.Y.; Pashko, Y.P.; Zigangirova, N.A.; Zakharova, M.N. Role of Chlamydia in Multiple Sclerosis. Bull. Exp. Biol. Med. 2015, 159, 646–648. [Google Scholar] [CrossRef]
  29. Kazemi, S.; Aghaee, B.L.; Soltanian, A.R.; Mazdeh, M.; Taheri, M.; Alikhani, M.Y. Investigation of Chlamydia pneumoniae Infection in Patients with Multiple Sclerosis: A Case-Control Study. Avicenna J. Clin. Microb. Infect. 2020, 7, 36–39. [Google Scholar] [CrossRef]
  30. Berer, K.; Gerdes, L.A.; Cekanaviciute, E.; Jia, X.; Xiao, L.; Xia, Z.; Liu, C.; Klotz, L.; Stauffer, U.; Baranzini, S.E. Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proc. Natl. Acad. Sci. USA 2017, 114, 10719–10724. [Google Scholar] [CrossRef] [Green Version]
  31. Mameli, G.; Poddighe, L.; Astone, V.; Delogu, G.; Arru, G.; Sotgiu, S.; Serra, C.; Dolei, A. Novel reliable real-time PCR for differential detection of MSRVenv and syncytin-1 in RNA and DNA from patients with multiple sclerosis. J. Virol. Methods 2009, 161, 98–106. [Google Scholar] [CrossRef] [PubMed]
  32. Küry, P.; Nath, A.; Créange, A.; Dolei, A.; Marche, P.; Gold, J.; Giovannoni, G.; Hartung, H.-P.; Perron, H. Human Endogenous Retroviruses in Neurological Diseases. Trends Mol. Med. 2018, 24, 379–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Huang, Q.; Li, J.; Wang, F.; Oliver, M.T.; Tipton, T.; Gao, Y.; Jiang, S.-W. (Albert) Syncytin-1 modulates placental trophoblast cell proliferation by promoting G1/S transition. Cell. Signal. 2013, 25, 1027–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Fischer, S.; Echeverría, N.; Moratorio, G.; Landoni, A.I.; Dighiero, G.; Cristina, J.; Oppezzo, P.; Moreno, P. Human endogenous retrovirus np9 gene is over expressed in chronic lymphocytic leukemia patients. Leuk. Res. Rep. 2014, 3, 70–72. [Google Scholar] [CrossRef] [Green Version]
  35. Van Horssen, J.; Van Der Pol, S.; Nijland, P.; Amor, S.; Perron, H. Human endogenous retrovirus W in brain lesions: Rationale for targeted therapy in multiple sclerosis. Mult. Scler. Relat. Disord. 2016, 8, 11–18. [Google Scholar] [CrossRef] [Green Version]
  36. Perron, H.; Germi, R.; Bernard, C.; Garcia-Montojo, M.; Deluen, C.; Farinelli, L.; Faucard, R.; Veas, F.; Stefas, I.; O Fabriek, B.; et al. Human endogenous retrovirus type W envelope expression in blood and brain cells provides new insights into multiple sclerosis disease. Mult. Scler. J. 2012, 18, 1721–1736. [Google Scholar] [CrossRef]
  37. Kremer, D.; Gruchot, J.; Weyers, V.; Oldemeier, L.; Göttle, P.; Healy, L.; Jang, J.H.; Xu, Y.K.T.; Volsko, C.; Dutta, R. pHERV-W envelope protein fuels microglial cell-dependent damage of myelinated axons in multiple sclerosis. Proc. Natl. Acad. Sci. USA 2019, 116, 15216–15225. [Google Scholar] [CrossRef] [Green Version]
  38. Brudek, T.; Christensen, T.; Aagaard, L.; Petersen, T.; Hansen, H.J.; Møller-Larsen, A. B cells and monocytes from patients with active multiple sclerosis exhibit increased surface expression of both HERV-H Env and HERV-W Env, accompanied by increased seroreactivity. Retrovirology 2009, 6, 104. [Google Scholar] [CrossRef] [Green Version]
  39. Arru, G.; Mameli, G.; Astone, V.; Serra, C.; Huang, Y.-M.; Link, H.; Fainardi, E.; Castellazzi, M.; Granieri, E.; Fernandez, M.; et al. Multiple Sclerosis and HERV-W/MSRV: A Multicentric Study. Int. J. Biomed. Sci. 2007, 3, 292–297. [Google Scholar]
  40. Venter, J.C.; Adams, M.D.; Myers, E.W.; Li, P.W.; Mural, R.J.; Sutton, G.G.; Smith, H.O.; Yandell, M.; Evans, C.A.; Holt, R.A.; et al. The Sequence of the Human Genome. Sci. Transl. Med. 2001, 291, 1304–1351. [Google Scholar] [CrossRef] [Green Version]
  41. Villesen, P.; Aagaard, L.; Wiuf, C.; Pedersen, F.S. Identification of endogenous retroviral reading frames in the human genome. Retrovirology 2004, 1, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Oja, M.; Sperber, G.O.; Blomberg, J.; Kaski, S. Self-Organizing Map-Based Discovery and Visualization of Human Endogenous Retroviral Sequence Groups. Int. J. Neural Syst. 2005, 15, 163–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Grandi, N.; Tramontano, E. Human Endogenous Retroviruses Are Ancient Acquired Elements Still Shaping Innate Immune Responses. Front. Immunol. 2018, 9, 2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Santoni, F.; Guerra, J.; Luban, J. HERV-H RNA is abundant in human embryonic stem cells and a precise marker for pluripotency. Retrovirology 2012, 9, 111. [Google Scholar] [CrossRef] [Green Version]
  45. Bonnaud, B.; Beliaeff, J.; Bouton, O.; Oriol, G.; Duret, L.; Mallet, F. Natural history of the ERVWE1 endogenous retroviral locus. Retrovirology 2005, 2, 57. [Google Scholar] [CrossRef] [Green Version]
  46. Boeke, J.; Stoye, J. Retrotransposons, Endogenous Retroviruses, and the Evolution of Retroelements; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 1997. [Google Scholar]
  47. Vargiu, L.; Rodriguez-Tomé, P.; Sperber, G.O.; Cadeddu, M.; Grandi, N.; Blikstad, V.; Tramontano, E.; Blomberg, J. Classification and characterization of human endogenous retroviruses; mosaic forms are common. Retrovirology 2016, 13, 1–29. [Google Scholar] [CrossRef] [Green Version]
  48. Griffiths, D.J. Endogenous retroviruses in the human genome sequence. Genome Biol. 2001, 2, 1017. [Google Scholar] [CrossRef]
  49. Nelson, P.N.; Carnegie, P.; Martin, J.; Ejtehadi, H.D.; Hooley, P.; Roden, D.; Rowland-Jones, S.; Warren, P.; Astley, J.; Murray, P.G. Demystified…Human endogenous retroviruses. Mol. Pathol. 2003, 56, 11. [Google Scholar] [CrossRef] [Green Version]
  50. Rolland, A.; Jouvin-Marche, E.; Viret, C.; Faure, M.; Perron, H.; Marche, P.N. The Envelope Protein of a Human Endogenous Retrovirus-W Family Activates Innate Immunity through CD14/TLR4 and Promotes Th1-Like Responses. J. Immunol. 2006, 176, 7636–7644. [Google Scholar] [CrossRef]
  51. Kremer, D.; Schichel, T.; Förster, M.; Tzekova, N.; Bernard, C.; Van Der Valk, P.; Van Horssen, J.; Hartung, H.-P.; Perron, H.; Küry, P. Human endogenous retrovirus type W envelope protein inhibits oligodendroglial precursor cell differentiation. Ann. Neurol. 2013, 74, 721–732. [Google Scholar] [CrossRef]
  52. Duperray, A.; Barbe, D.; Raguenez, G.; Weksler, B.B.; Romero, I.A.; Couraud, P.-O.; Perron, H.; Marche, P.N. Inflammatory response of endothelial cells to a human endogenous retrovirus associated with multiple sclerosis is mediated by TLR4. Int. Immunol. 2015, 27, 545–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Pavlíček, A.; Pačes, J.; Elleder, D.; Hejnar, J. Processed pseudogenes of human endogenous retroviruses generated by LINEs: Their integration, stability, and distribution. Genome Res. 2002, 12, 391–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Grandi, N.; Cadeddu, M.; Blomberg, J.; Tramontano, E. Contribution of type W human endogenous retroviruses to the human genome: Characterization of HERV-W proviral insertions and processed pseudogenes. Retrovirology 2016, 13, 1–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Laufer, G.; Mayer, J.; Mueller, B.F.; Mueller-Lantzsch, N.; Ruprecht, K. Analysis of transcribed human endogenous retrovirus W env loci clarifies the origin of multiple sclerosis-associated retrovirus envsequences. Retrovirology 2009, 6, 37. [Google Scholar] [CrossRef] [Green Version]
  56. Voisset, C.; Bouton, O.; Bedin, F.; Duret, L.; Mandrand, B.; Mallet, F.; Paranhos-Baccalà, G. Chromosomal Distribution and Coding Capacity of the Human Endogenous Retrovirus HERV-W Family. AIDS Res. Hum. Retroviruses 2000, 16, 731–740. [Google Scholar] [CrossRef] [Green Version]
  57. Li, F.; Nellåker, C.; Yolken, R.; Karlsson, H. A systematic evaluation of expression of HERV-W elements; influence of genomic context, viral structure and orientation. BMC Genom. 2011, 12, 22. [Google Scholar] [CrossRef] [Green Version]
  58. Blond, J.-L.; Besème, F.; Duret, L.; Bouton, O.; Bedin, F.; Perron, H.; Mandrand, B.; Mallet, F. Molecular Characterization and Placental Expression of HERV-W, a New Human Endogenous Retrovirus Family. J. Virol. 1999, 73, 1175–1185. [Google Scholar] [CrossRef] [Green Version]
  59. Prudhomme, S.; Oriol, G.; Mallet, F. A Retroviral Promoter and a Cellular Enhancer Define a Bipartite Element Which Controls env ERVWE1 Placental Expression. J. Virol. 2004, 78, 12157–12168. [Google Scholar] [CrossRef] [Green Version]
  60. Schmitt, K.; Richter, C.; Backes, C.; Meese, E.; Ruprecht, K.; Mayer, J. Comprehensive Analysis of Human Endogenous Retrovirus Group HERV-W Locus Transcription in Multiple Sclerosis Brain Lesions by High-Throughput Amplicon Sequencing. J. Virol. 2013, 87, 13837–13852. [Google Scholar] [CrossRef] [Green Version]
  61. Place, R.F.; Li, L.-C.; Pookot, D.; Noonan, E.J.; Dahiya, R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc. Natl. Acad. Sci. USA 2008, 105, 1608–1613. [Google Scholar] [CrossRef] [Green Version]
  62. Bagga, S.; Bracht, J.; Hunter, S.; Massirer, K.; Holtz, J.; Eachus, R.; Pasquinelli, A.E. Regulation by let-7 and lin-4 miRNAs Results in Target mRNA Degradation. Cell 2005, 122, 553–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhang, S.; Li, J.; Li, J.; Yang, Y.; Kang, X.; Li, Y.; Wu, X.; Zhu, Q.; Zhou, Y.; Hu, Y. Up-regulation of microRNA-203 in influenza A virus infection inhibits viral replication by targeting DR1. Sci. Rep. 2018, 8, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  65. Cox, M.B.; Cairns, M.J.; Gandhi, K.S.; Carroll, A.P.; Moscovis, S.; Stewart, G.J.; Broadley, S.; Scott, R.J.; Booth, D.R.; Lechner-Scott, J.; et al. MicroRNAs miR-17 and miR-20a Inhibit T Cell Activation Genes and Are Under-Expressed in MS Whole Blood. PLOS ONE 2010, 5, e12132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Meira, M.; Sievers, C.; Hoffmann, F.; Rasenack, M.; Kuhle, J.; Derfuss, T.; Kappos, L.; Lindberg, R.L.P. Unraveling Natalizumab Effects on Deregulated miR-17 Expression in CD4+T Cells of Patients with Relapsing-Remitting Multiple Sclerosis. J. Immunol. Res. 2014, 2014, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Arru, G.; Leoni, S.; Pugliatti, M.; Mei, A.; Serra, C.; Delogu, L.G.; Manetti, R.; Dolei, A.; Sotgiu, S.; Mameli, G. Natalizumab inhibits the expression of human endogenous retroviruses of the W family in multiple sclerosis patients: A longitudinal cohort study. Mult. Scler. J. 2013, 20, 174–182. [Google Scholar] [CrossRef]
  68. Hakim, S.T.; Alsayari, M.; McLean, D.C.; Saleem, S.; Addanki, K.C.; Aggarwal, M.; Mahalingam, K.; Bagasra, O. A large number of the human microRNAs target lentiviruses, retroviruses, and endogenous retroviruses. Biochem. Biophys. Res. Commun. 2008, 369, 357–362. [Google Scholar] [CrossRef]
  69. Mameli, G.; Astone, V.; Khalili, K.; Serra, C.; Sawaya, B.E.; Dolei, A. Regulation of the syncytin-1 promoter in human astrocytes by multiple sclerosis-related cytokines. Virology 2007, 362, 120–130. [Google Scholar] [CrossRef] [Green Version]
  70. Sharma, P.; Azebi, S.; England, P.; Christensen, T.; Møller-Larsen, A.; Petersen, T.; Batsche, E.; Muchardt, C. Citrullination of Histone H3 Interferes with HP1-Mediated Transcriptional Repression. PLoS Genet. 2012, 8, e1002934. [Google Scholar] [CrossRef] [Green Version]
  71. Warren, K.G.; Catz, I. Autoantibodies to myelin basic protein within multiple sclerosis central nervous system tissue. J. Neurol. Sci. 1993, 115, 169–176. [Google Scholar] [CrossRef]
  72. Libich, D.S.; Hill, C.M.; Bates, I.R.; Hallett, F.R.; Armstrong, S.; Siemiarczuk, A.; Harauz, G. Interaction of the 18.5-kD isoform of myelin basic protein with Ca2+-calmodulin: Effects of deimination assessed by intrinsic Trp fluorescence spectroscopy, dynamic light scattering, and circular dichroism. Protein Sci. 2003, 12, 1507–1521. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, H.; Jo, S.; Park, C.-S.; Song, H.-J.; Park, Z.-Y. Myelin basic protein as a binding partner and calmodulin adaptor for the BKCa channel. Proteomics 2007, 7, 2591–2602. [Google Scholar] [CrossRef] [PubMed]
  74. Moscarello, M.A.; Wood, D.D.; Ackerley, C.; Boulias, C. Myelin in multiple sclerosis is developmentally immature. J. Clin. Investig. 1994, 94, 146–154. [Google Scholar] [CrossRef] [PubMed]
  75. Wood, D.D.; Moscarello, M.A.; Bilbao, J.M.; O’Connors, P. Acute multiple sclerosis (marburg type) is associated with developmentally immature myelin basic protein. Ann. Neurol. 1996, 40, 18–24. [Google Scholar] [CrossRef]
  76. Wood, D.D.; A Ackerley, C.; Brand, B.V.D.; Zhang, L.; Raijmakers, R.; Mastronardi, F.G.; A Moscarello, M. Myelin localization of peptidylarginine deiminases 2 and 4: Comparison of PAD2 and PAD4 activities. Lab. Investig. 2008, 88, 354–364. [Google Scholar] [CrossRef] [Green Version]
  77. Faigle, W.; Cruciani, C.; Wolski, W.; Roschitzki, B.; Puthenparampil, M.; Tomas-Ojer, P.; Sellés-Moreno, C.; Zeis, T.; Jelcic, I.; Schaeren-Wiemers, N.; et al. Brain Citrullination Patterns and T Cell Reactivity of Cerebrospinal Fluid-Derived CD4+ T Cells in Multiple Sclerosis. Front. Immunol. 2019, 10, 540. [Google Scholar] [CrossRef] [Green Version]
  78. Jasse, L.; Vukusic, S.; Durand-Dubief, F.; Vartin, C.; Piras, C.; Bernard, M.; Pélisson, D.; Confavreux, C.; Vighetto, A.; Tilikete, C. Persistent visual impairment in multiple sclerosis: Prevalence, mechanisms and resulting disability. Mult. Scler. J. 2013, 19, 1618–1626. [Google Scholar] [CrossRef]
  79. Bazelier, M.T.; Mueller-Schotte, S.; Leufkens, H.G.; Uitdehaag, B.M.; Van Staa, T.P.; De Vries, F. Risk of cataract and glaucoma in patients with multiple sclerosis. Mult. Scler. J. 2011, 18, 628–638. [Google Scholar] [CrossRef]
  80. Bhattacharya, S.K.; Bhat, M.B.; Takahara, H. Modulation of Peptidyl Arginine Deiminase 2 and Implication for Neurodegeneration. Curr. Eye Res. 2006, 31, 1063–1071. [Google Scholar] [CrossRef]
  81. Bhattacharya, S.K.; Sinicrope, B.; Rayborn, M.E.; Hollyfield, J.G.; Bonilha, V.L. Age-related reduction in retinal deimination levels in the F344BN rat. Aging Cell 2008, 7, 441–444. [Google Scholar] [CrossRef] [Green Version]
  82. Yang, X.; Han, H.; De Carvalho, D.D.; Lay, F.D.; Jones, P.A.; Liang, G. Gene Body Methylation Can Alter Gene Expression and Is a Therapeutic Target in Cancer. Cancer Cell 2014, 26, 577–590. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, P.S.; Teaberry, V.S.; Bland, A.E.; Huang, Z.; Whitaker, R.S.; Baba, T.; Fujii, S.; Secord, A.A.; Berchuck, A.; Murphy, S.K. Elevated MAL expression is accompanied by promoter hypomethylation and platinum resistance in epithelial ovarian cancer. Int. J. Cancer 2010, 126, 1378–1389. [Google Scholar] [PubMed]
  84. Weber, M.; Hellmann, I.; Stadler, M.B.; Ramos, L.; Pääbo, S.; Rebhan, M.; Schübeler, D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat. Genet. 2007, 39, 457–466. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, X.; Deng, F.-Y.; Lü, X.; Lei, S.-F. Susceptibility Genes for Multiple Sclerosis Identified in a Gene-Based Genome-Wide Association Study. J. Clin. Neurol. 2015, 11, 311–318. [Google Scholar] [CrossRef] [Green Version]
  86. Douvaras, P.; Rusielewicz, T.; Kim, K.H.; Haines, J.D.; Casaccia, P.; Fossati, V. Epigenetic Modulation of Human Induced Pluripotent Stem Cell Differentiation to Oligodendrocytes. Int. J. Mol. Sci. 2016, 17, 614. [Google Scholar] [CrossRef] [Green Version]
  87. Cui, Q.-L.; Kuhlmann, T.; Miron, V.E.; Leong, S.Y.; Fang, J.; Gris, P.; Kennedy, T.E.; Almazan, G.; Antel, J. Oligodendrocyte Progenitor Cell Susceptibility to Injury in Multiple Sclerosis. Am. J. Pathol. 2013, 183, 516–525. [Google Scholar] [CrossRef]
  88. Szpakowski, S.; Sun, X.; Lage, J.M.; Dyer, A.; Rubinstein, J.; Kowalski, D.; Sasaki, C.; Costa, J.; Lizardi, P.M. Loss of epigenetic silencing in tumors preferentially affects primate-specific retroelements. Gene 2009, 448, 151–167. [Google Scholar] [CrossRef] [Green Version]
  89. Matoušková, M.; Blažková, J.; Pajer, P.; Pavlíček, A.; Hejnar, J. CpG methylation suppresses transcriptional activity of human syncytin-1 in non-placental tissues. Exp. Cell Res. 2006, 312, 1011–1020. [Google Scholar] [CrossRef]
  90. Weber, S.; Hofmann, A.; Herms, S.; Hoffmann, P.; Doerfler, W. Destabilization of the human epigenome: Consequences of foreign DNA insertions. Epigenomics 2015, 7, 745–755. [Google Scholar] [CrossRef] [Green Version]
  91. Weber, S.; Jung, S.; Doerfler, W. DNA methylation and transcription in HERV (K, W, E) and LINE sequences remain unchanged upon foreign DNA insertions. Epigenomics 2016, 8, 157–165. [Google Scholar] [CrossRef]
  92. Chomyk, A.M.; Volsko, C.; Tripathi, A.; Deckard, S.A.; Trapp, B.D.; Fox, R.J.; Dutta, R. DNA methylation in demyelinated multiple sclerosis hippocampus. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Van Nierop, G.P.; Van Luijn, M.M.; Michels, S.; Samira, S.; Melief, M.-J.; Janssen, M.; Langerak, A.W.; Ouwendijk, W.J.D.; Hintzen, R.Q.; Verjans, G.M. Phenotypic and functional characterization of T cells in white matter lesions of multiple sclerosis patients. Acta Neuropathol. 2017, 134, 383–401. [Google Scholar] [CrossRef] [PubMed]
  94. Bos, S.D.; Page, C.M.; Andreassen, B.K.; Elboudwarej, E.; Gustavsen, M.W.; Briggs, F.B.; Quach, H.; Leikfoss, I.S.; Bjølgerud, A.; Berge, T.; et al. Genome-Wide DNA Methylation Profiles Indicate CD8+ T Cell Hypermethylation in Multiple Sclerosis. PLoS ONE 2015, 10, e0117403. [Google Scholar] [CrossRef] [PubMed]
  95. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
  96. Hurst, T.; Pace, M.J.; Katzourakis, A.; Phillips, R.; Klenerman, P.; Frater, J.; Magiorkinis, G. Human endogenous retrovirus (HERV) expression is not induced by treatment with the histone deacetylase (HDAC) inhibitors in cellular models of HIV-1 latency. Retrovirology 2016, 13, 10. [Google Scholar] [CrossRef] [Green Version]
  97. Yu, H.; Liu, T.; Zhao, Z.; Chen, Y.; Zeng, J.; Liu, S.; Zhu, F. Mutations in 3′-long terminal repeat of HERV-W family in chromosome 7 upregulate syncytin-1 expression in urothelial cell carcinoma of the bladder through interacting with c-Myb. Oncogene 2013, 33, 3947–3958. [Google Scholar] [CrossRef]
  98. Yu, J.S.; Hayashi, T.; Seboun, E.; Sklar, R.M.; Doolittle, T.H.; Hauser, S.L. Fos RNA accumulation in multiple sclerosis white matter tissue. J. Neurol. Sci. 1991, 103, 209–215. [Google Scholar] [CrossRef]
  99. Gimenez, J.; Montgiraud, C.; Pichon, J.-P.; Bonnaud, B.; Arsac, M.; Ruel, K.; Bouton, O.; Mallet, F. Custom human endogenous retroviruses dedicated microarray identifies self-induced HERV-W family elements reactivated in testicular cancer upon methylation control. Nucleic Acids Res. 2010, 38, 2229–2246. [Google Scholar] [CrossRef]
  100. Cramer, S.P.; Simonsen, H.; Frederiksen, J.; Rostrup, E.; Larsson, H.B.W. Abnormal blood–brain barrier permeability in normal appearing white matter in multiple sclerosis investigated by MRI. NeuroImage Clin. 2014, 4, 182–189. [Google Scholar] [CrossRef] [Green Version]
  101. Schenk, G.J.; Dijkstra, S.; Hof, A.J.V.H.; Van Der Pol, S.M.A.; Drexhage, J.A.R.; Van Der Valk, P.; Reijerkerk, A.; Van Horssen, J.; De Vries, H.E. Roles for HB-EGF and CD9 in multiple sclerosis. Glia 2013, 61, 1890–1905. [Google Scholar] [CrossRef]
  102. Muroi, Y.; Sakurai, T.; Hanashi, A.; Kubota, K.; Nagaoka, K.; Imakawa, K. CD9 regulates transcription factor GCM1 and ERVWE1 expression through the cAMP/protein kinase A signaling pathway. Reproduction 2009, 138, 945–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Fei, F.; Li, C.; Wang, X.; Du, J.; Liu, K.; Li, B.; Yao, P.; Li, Y.; Zhang, S. Syncytin 1, CD9, and CD47 regulating cell fusion to form PGCCs associated with cAMP/PKA and JNK signaling pathway. Cancer Med. 2019, 8, 3047–3058. [Google Scholar] [CrossRef] [PubMed]
  104. Uleri, E.; Mei, A.; Mameli, G.; Poddighe, L.; Serra, C.; Dolei, A. HIV Tat acts on endogenous retroviruses of the W family and this occurs via Toll-like receptor 4: Inference for neuroAIDS. Aids 2014, 28, 2659–2670. [Google Scholar] [CrossRef] [PubMed]
  105. Anand, P.; Saylor, D. Multiple sclerosis and HIV: A case of multiple sclerosis-immune reconstitution inflammatory syndrome associated with antiretroviral therapy initiation. Int. J. STD AIDS 2018, 29, 929–932. [Google Scholar] [CrossRef] [PubMed]
  106. Assinger, A.; Yaiw, K.-C.; Göttesdorfer, I.; Leib-Mösch, C.; Söderberg-Nauclér, C. Human Cytomegalovirus (HCMV) induces Human Endogenous Retrovirus (HERV) transcription. Retrovirology 2013, 10, 132. [Google Scholar] [CrossRef] [Green Version]
  107. Ruprecht, K.; Obojes, K.; Wengel, V.; Gronen, F.; Kim, K.S.; Perron, H.; Schneider-Schaulies, J.; Rieckmann, P. Regulation of human endogenous retrovirus W protein expression by herpes simplex virus type 1: Implications for multiple sclerosis. J. Neurovirol. 2006, 12, 65–71. [Google Scholar] [CrossRef]
  108. Charvet, B.; Reynaud, J.M.; Gourru-Lesimple, G.; Perron, H.; Marche, P.N.; Horvat, B. Induction of Proinflammatory Multiple Sclerosis-Associated Retrovirus Envelope Protein by Human Herpesvirus-6A and CD46 Receptor Engagement. Front. Immunol. 2018, 9, 2803. [Google Scholar] [CrossRef]
Figure 1. The structure of the HERV-W family and the ERVWE1 gene. (A) The HERV-W family includes four genes, with an up and down stream LTRs. These LTRs consist of U3, R and U5 region. The U3 region of 5′-LTR consist of tRNATrp-binding PBS, CAAT and TATA box sites. The gag gene encodes elements of the matrix, capsid and nucleocapsid, while pro gene encodes for protease. Viral pol gene encodes for reverse transcriptase and integrase. Finally, the envelop protein is coded by the env gene. (B) The ERVWE1 gene is “domesticated” and retains the ability to express genes of the HERV-W family. It consists of the env gene, called Syncitin-1, with an ORF and is also surrounded by two LTR from 5′ and 3′ ends. Trophoblast-specific enhancer (TSE) containing AP-2, Sp-1 and GCMa binding sites is located upstream of the 5′-LTR. The U3 region of 5′-LTR contains PPAR-γ, Oct-1, GATA, AP-2, Sp-1, CREB, CAAT and TATA box binding sites. R region contains a CAP transcription initiation site at the 5′ end and a polyadenylation site at the 3′ end. The R region of the 3′-LTR includes a polyadenylation site. The whole ERVWE1 gene is flanked by the MaLR retrotransposon.
Figure 1. The structure of the HERV-W family and the ERVWE1 gene. (A) The HERV-W family includes four genes, with an up and down stream LTRs. These LTRs consist of U3, R and U5 region. The U3 region of 5′-LTR consist of tRNATrp-binding PBS, CAAT and TATA box sites. The gag gene encodes elements of the matrix, capsid and nucleocapsid, while pro gene encodes for protease. Viral pol gene encodes for reverse transcriptase and integrase. Finally, the envelop protein is coded by the env gene. (B) The ERVWE1 gene is “domesticated” and retains the ability to express genes of the HERV-W family. It consists of the env gene, called Syncitin-1, with an ORF and is also surrounded by two LTR from 5′ and 3′ ends. Trophoblast-specific enhancer (TSE) containing AP-2, Sp-1 and GCMa binding sites is located upstream of the 5′-LTR. The U3 region of 5′-LTR contains PPAR-γ, Oct-1, GATA, AP-2, Sp-1, CREB, CAAT and TATA box binding sites. R region contains a CAP transcription initiation site at the 5′ end and a polyadenylation site at the 3′ end. The R region of the 3′-LTR includes a polyadenylation site. The whole ERVWE1 gene is flanked by the MaLR retrotransposon.
Biology 09 00464 g001
Figure 2. The mechanism of RNA interference. The Dicer enzyme cleaves specific to HERV-W double-stranded miRNA and promotes the loading of each strand into the RISC complex. Dicer binds to the transactivating response RNA-binding protein (TRBP) to facilitate the transfer of dsRNA fragments to Argonaute 2 (Ago2). The least stable 5′ end RNA strand is integrated with the Ago2 protein in RISC, and the second passenger strand is degraded. The RISC complex with guide strand miRNA complementary binds to the 3′-LTR of the mRNA HERV-W and silences it.
Figure 2. The mechanism of RNA interference. The Dicer enzyme cleaves specific to HERV-W double-stranded miRNA and promotes the loading of each strand into the RISC complex. Dicer binds to the transactivating response RNA-binding protein (TRBP) to facilitate the transfer of dsRNA fragments to Argonaute 2 (Ago2). The least stable 5′ end RNA strand is integrated with the Ago2 protein in RISC, and the second passenger strand is degraded. The RISC complex with guide strand miRNA complementary binds to the 3′-LTR of the mRNA HERV-W and silences it.
Biology 09 00464 g002
Figure 3. The mechanism of regulation of HERV-W transcription by histone citrullination. (A) Repression of HERV-W transcription upon the interaction of HP1α with histone H3K9 in LTR. (B) Enhanced expression of HERV-W due to the formation of citrullinated H3K9 histone by PADI4.
Figure 3. The mechanism of regulation of HERV-W transcription by histone citrullination. (A) Repression of HERV-W transcription upon the interaction of HP1α with histone H3K9 in LTR. (B) Enhanced expression of HERV-W due to the formation of citrullinated H3K9 histone by PADI4.
Biology 09 00464 g003
Figure 4. Regulation of HERV-W transcription by methylation of CpG sites in the promoter region.
Figure 4. Regulation of HERV-W transcription by methylation of CpG sites in the promoter region.
Biology 09 00464 g004
Figure 5. ERVWE1 expression mechanism due to activation of cAMP/PKA signaling pathway by CD9.
Figure 5. ERVWE1 expression mechanism due to activation of cAMP/PKA signaling pathway by CD9.
Biology 09 00464 g005
Table 1. Possible downregulation and upregulation mechanisms HERV-W expression.
Table 1. Possible downregulation and upregulation mechanisms HERV-W expression.
FactorsDownregulation MechanismReferences
RNA interferencemiRNA with homology to HERV-W: hsa-miR-376b, hsa-miR-22, hsa-miR-574, hsa-miR-570, hsa-miR-198, MIMAT0000228, hsa-miR-493-3p, MIMAT0003161, hsa-miR-214, MIMAT0000271, hsa-miR-101, hsa-miR-296, hsa-miR-31, hsa-miR-659, hsa-miR-185, hsa-miR-202, hsa-miR-122a, hsa-miR-24, hsa-miR-506, hsa-miR-632, hsa-miR-376a, hsa-miR-326.
siRNA to the p65 gene of the NF-κB subunit reduces URE-LTR activation of the ERVWE1 promoter by TNFα.
[68,69]
DNA methylationHypermethylation in the LTR region of HERV-W leads to repression of HERV-W expression.[83]
Upregulation mechanism
CitrullinationIncreased level of PADI4 leads to formation of H3cit8K9me3 histone in promotor region of ERVWE1. Local increased citrullination of H3R8 histone localized above H3K9 was associated with increased expression of HERV-W/ERVWE1.[70]
Changes in regulatory regionThe 142T > C mutation promotes the formation of an additional binding site with c-Myb in 3′-LTR of ERVWE1 and due to this activation of its promoter activity.[97]
Activation by immune systemTNFα and IFN-γ activate the ERVWE1 promoter.
TNFα promotes increased binding of p65 of the NF-κB subunit to a sensitive element located in the enhancer region of the ERVWE1 promoter.
CD9 through cAMP/PKA signaling pathway leads to the activation of GCM1, which binds to the regulatory region of ERVWE1 and enhances gene expression.
[69,102,103]
Exogenous virusesTat protein of HIV activate transcription of MSRV and ERVWE1 in in astrocytes through TLR-4 and TNFα.
HCMV activate reverse transcriptase necessary for transcription of HERV.
ICP0 protein of HSV-1 activate transcription of the HERV-Wenv and MSRVenv genes.
HHV-6A activates MSRVenv expression by interacting with the isoform of CD46-Cyt1 receptor.
[104,106,107,108]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lezhnyova, V.R.; Martynova, E.V.; Khaiboullin, T.I.; Urbanowicz, R.A.; Khaiboullina, S.F.; Rizvanov, A.A. The Relationship of the Mechanisms of the Pathogenesis of Multiple Sclerosis and the Expression of Endogenous Retroviruses. Biology 2020, 9, 464. https://doi.org/10.3390/biology9120464

AMA Style

Lezhnyova VR, Martynova EV, Khaiboullin TI, Urbanowicz RA, Khaiboullina SF, Rizvanov AA. The Relationship of the Mechanisms of the Pathogenesis of Multiple Sclerosis and the Expression of Endogenous Retroviruses. Biology. 2020; 9(12):464. https://doi.org/10.3390/biology9120464

Chicago/Turabian Style

Lezhnyova, Vera R., Ekaterina V. Martynova, Timur I. Khaiboullin, Richard A. Urbanowicz, Svetlana F. Khaiboullina, and Albert A. Rizvanov. 2020. "The Relationship of the Mechanisms of the Pathogenesis of Multiple Sclerosis and the Expression of Endogenous Retroviruses" Biology 9, no. 12: 464. https://doi.org/10.3390/biology9120464

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