Next Article in Journal
BRCA1, BRCA2 and PALB2 mRNA Expression as Prognostic Markers in Patients with Early Breast Cancer
Next Article in Special Issue
Role of ncRNAs in the Pathogenesis of Sjögren’s Syndrome
Previous Article in Journal
Triple Combinations of Histone Lysine Demethylase Inhibitors with PARP1 Inhibitor–Olaparib and Cisplatin Lead to Enhanced Cytotoxic Effects in Head and Neck Cancer Cells
Previous Article in Special Issue
miR-155 and miR-21 as Diagnostic and Therapeutic Biomarkers for Ulcerative Colitis: There Is Still a Long Way to Go
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Virus-Induced MicroRNA Modulation and Systemic Sclerosis Disease

by
Irene Soffritti
1,2,
Maria D’Accolti
1,2,
Francesca Bini
1,2,
Eleonora Mazziga
1,2,
Dario Di Luca
3,
Clara Maccari
4,
Maria-Cristina Arcangeletti
4 and
Elisabetta Caselli
1,2,*
1
Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
2
CIAS Research Center, University of Ferrara, 44122 Ferrara, Italy
3
Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
4
Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(6), 1360; https://doi.org/10.3390/biomedicines12061360
Submission received: 16 May 2024 / Revised: 6 June 2024 / Accepted: 15 June 2024 / Published: 19 June 2024
(This article belongs to the Special Issue MicroRNA and Its Role in Human Health)

Abstract

:
MicroRNAs (miRNAs) are short noncoding RNA sequences that regulate gene expression at the post-transcriptional level. They are involved in the regulation of multiple pathways, related to both physiological and pathological conditions, including autoimmune diseases, such as Systemic Sclerosis (SSc). Specifically, SSc is recognized as a complex and multifactorial disease, characterized by vascular abnormalities, immune dysfunction, and progressive fibrosis, affecting skin and internal organs. Among predisposing environmental triggers, evidence supports the roles of oxidative stress, chemical agents, and viral infections, mostly related to those sustained by beta-herpesviruses such as HCMV and HHV-6. Dysregulated levels of miRNA expression have been found in SSc patients compared to healthy controls, at both the intra- and extracellular levels, providing a sort of miRNA signature of the SSc disease. Notably, HCMV/HHV-6 viral infections were shown to modulate the miRNA profile, often superposing that observed in SSc, potentially promoting pathological pathways associated with SSc development. This review summarizes the main data regarding miRNA alterations in SSc disease, highlighting their potential as prognostic or diagnostic markers for SSc disease, and the impact of the putative SSc etiological agents on miRNA modulation.

1. Introduction

MicroRNAs (miRNAs) are a class of noncoding RNAs (ncRNAs), 19–25 nucleotides (nts) in length, which can modulate gene expression at the post-transcriptional level and are involved in a broad range of molecular mechanisms, both in physiological and pathological conditions [1]. Their fortuitous discovery in a Caenorhabditis elegans model, more than thirty years ago, opened the way to the studies of antisense RNA–RNA interactions and their regulatory effects on gene expression [2,3].
The synthesis of miRNAs occurs in various steps, finely regulated across different subcellular compartments [4]. Briefly, canonical miRNA biogenesis starts with transcription, mainly of intragenic DNA sequences, by RNA polymerase II [5], leading to long primary miRNAs (pri-miRNAs). Pri-miRNAs are then cleaved by the Drosha-DGCR8 (DiGeorge Syndrome Critical Region 8) nuclear complex to release hairpin-shaped precursor miRNAs (pre-miRNAs, about 60–70 nts in length). These intermediates are exported by the Exportin5-RanGTP complex out of the nucleus, where a second RNase complex, consisting of the ribonuclease Dicer in association with its cofactor named “Transactivation Response element RNA-Binding Protein” (Dicer-TRBP), cleaves the pre-miRNAs into ~22 nt miRNA duplexes, generating mature miRNAs. MiRNAs are able to modulate gene expression through both cis- and trans-regulatory pathways [6]. During the cis-regulation, one of the two strands of the mature miRNA duplex is incorporated into the Argonaute (AGO) proteins (AGO1–4 in humans) to form a miRNA-induced silencing complex (miRISC), which interacts with the 3′ untranslated region (3′ UTR) of target mRNAs to induce mRNA degradation and repress gene expression at the post-transcriptional level [7]. In addition, silencing effects on gene expression have been observed in the case of miRNA interaction with 5′ UTR, coding or promoter sequences [8]. In trans-regulation, miRNAs can inhibit protein translation during the early steps of translation by interacting and/or competing with several factors: RNA-binding proteins or enzymes (e.g., decapping enzymes, deadenylase, 3′ and 5′ exonucleases, and endonucleases) involved in the 43S preinitiation complex formation or affecting post-initiation mechanisms, translation machinery, or post-translational modifiers [6,9].
Moreover, miRNAs biogenesis may also occur via different non-canonical pathways, mainly grouped into Drosha/DGCR8-independent and Dicer-independent pathways, reflecting the dynamism and the flexibility of miRNA machineries [8].
As to the peculiar nomenclature system of mature miRNA, consisting of the suffixes -5p or -3p, it depends on the directionality (5′ end or 3′ end) of the precursor strand included in the miRISC complex; in addition, the miR or miR* annotations indicate the highly expressed “guide strand” (miR) or the less expressed “passenger strand” (miR*), respectively [10].
It has been observed that a single miRNA could have more than 200 target mRNAs, and the expression of a single gene can be regulated by multiple miRNAs [11,12,13].
Since their discovery, a growing body of evidence has supported their key role as epigenetic regulators of a variety of physiological processes, including cell development, proliferation, differentiation, and stress adaptation [14,15,16]. Interestingly, dysregulated miRNAs expression has been observed in pathological conditions, including several types of cancer, metabolic disorders, and cardiovascular and chronic autoimmune diseases (ADs) [6,15].
The latter represent a group of complex and chronic inflammatory pathologies mainly defined by an impairment in autoimmune tolerance and improper immune responses towards healthy tissues [17]. Multiple factors may be associated with the predisposition, development, and/or progression of ADs, including host genetic alteration, persistent immune activation/reactivation, and environmental stimuli, both chemical and biological [18]. Recently, published epidemiological data pointed out that ADs affect around one out of ten individuals (2–10% of the population), showing a significantly higher prevalence in women compared to men (13% and 7%, respectively) [19]. The most relevant ADs include Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), Type 1 Diabetes, and Systemic Sclerosis (SSc) [20]. The diagnosis and treatment of ADs, especially of rare diseases such as SSc, are often hampered by challenges due to the complexity of the identification of initial symptoms; the poor knowledge of the disease; and the lack of rapid, accurate, and universally recognized diagnostic markers. Therefore, in recent years, growing interest has been given to the use of miRNAs as potential biomarkers or innovative therapeutic approaches [6,21,22,23,24].
This review aims to provide an updated overview of miRNA alterations in SSc, highlighting their possible role, and their potential as specific prognostic/diagnostic markers for the disease. Also, the relation between miRNA modulation and the putative SSc etiological agents is explored, particularly focusing on viral agents considered putative SSc triggers.

2. Systemic Sclerosis: An Overview

Systemic Sclerosis (SSc), also known as scleroderma (MIM: 181750; ORPHA: 90291), is a systemic connective autoimmune disease characterized by diffuse fibrosis and vascular abnormalities affecting the connective tissue of skin and internal organs (in particular, the lungs, heart, esophagus, low gastrointestinal tract, and kidneys) [25,26,27,28]. SSc develops as a result of multiple pathogenic mechanisms, primarily the abnormal deposition of the extracellular matrix (ECM) by altered fibroblasts, together with diffuse microangiopathy, immune dysregulation, tissue fibrosis, and apoptosis [26,29,30]. SSc patients are clinically classified based on the European League Against Rheumatism (EULAR) and American College of Rheumatology (ACR) criteria [31,32,33]. Common early clinical manifestations include Raynaud’s phenomenon, digital ulcers, hardening and tightening of the skin, and up to severe lung and heart sclerodermic lesions [28]. Despite the high variability of individual symptoms, the disease is typically categorized as “limited cutaneous” (lcSSc) and “diffuse cutaneous” (dcSSc) based on the extent of skin fibrosis: confined to finger/face/distal extremities with a slow progression in the case of lcSSc or affecting the trunk and proximal extremities with a rapid progression up to the fibrosis of internal organs in the case of dcSSc [26,34,35]. On these bases, the two subgroups present well-recognized patterns in disease severity and prognosis, with a higher risk for dcSSc patients to develop life-threatening complications, including pulmonary arterial hypertension, severe renal crisis, lung fibrosis, and heart and digestive tract involvement [36]. Other classifications also include SSc individuals without identifiable skin involvement (“SSc sine scleroderma”) or with a concurrent other fibrotic disease (“overlap syndrome”) [26,34,37]. A recent epidemiological study estimated the global incidence of the disease as 0.67 million (0.14–1.84) people annually and a prevalence of 18.87 per 100,000 persons (1.55–25.28) [38]. As for other ADs, SSc shows a marked prevalence in the female sex (F:M sex ratio around 4:1) in adults and in high-income level countries [36]. The etiology of SSc is still poorly understood, but several clinical, epidemiological, and experimental findings suggest that it is a multifactorial disease. Accordingly, research is heading towards elucidating several concomitant factors: genetic predisposition, oxidative stresses, and environmental triggers, including both toxic chemical agents (e.g., exposure to solvents and silica dust) and infectious agents [28,39].
So far, several mechanisms related to oxidative stress and redox regulation have been widely reported in SSc disease, related to inappropriate Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS) production, which can lead to pathological tissue damage [40].
In parallel, different infectious agents have been associated with SSc, including bacterial (e.g., Helicobacter pylori and Mycoplasma) and viral agents [28]. Among the viruses, those able to persist and reactivate in infected individuals were particularly claimed as possible triggers, including members of the Orthoherpesviridae family, such as Human Cytomegalovirus (HCMV), Human Herpesvirus-6A (HHV-6A) [28,41,42,43,44], and Epstein–Barr virus (EBV) [45,46,47], and members of the Parvoviridae family—in particular, Human parvovirus B19 (B19V) [48,49,50].
Overall, specific and periodic reactivation of viral infection, increased antibody and cell-mediated immune responses, and molecular mimicry represent the predominant mechanisms by which viral agents may be involved in the onset and progression of SSc [28].
Interestingly, miRNAs could be induced and involved in oxidative stress and viral response processes; in these contexts, their deregulation can trigger or favor the development of multiple pathological pathways. Moreover, the epigenetic regulation driven by miRNAs has been recognized to have a significant role in controlling the main pathological pathways of SSc, including immune responses, autoimmunity, fibrosis, and vasculopathy. The present review aimed to define the miRNome profile characterizing SSc disease at both the tissue/intracellular and extracellular levels. In addition, the effects of two of the key etiological factors of SSc disease, oxidative stress and beta-herpesvirus infection, on miRNA expression and related impacts are presented in Figure 1.

3. MiRNAs and Systemic Sclerosis

MiRNAs can be classified into intracellular miRNAs and extracellular or circulating miRNAs [8]. Inside the cell, miRISC and target mRNA could localize in several subcellular compartments, including the nucleus, rough endoplasmic reticulum, mitochondria, Golgi apparatus, and cytoplasmatic vesicles, such as endosomes and lysosomes [8]. Moreover, extensive literature data have demonstrated miRNA release into the extracellular environment to mediate cell–cell communication, often associated with lipids and proteins, or secreted via exosomes [93,94,95]. Due to their stability in the extracellular environment, their presence has been evidenced in a broad range of biological fluids, including plasma, serum, saliva, tears, bronchial lavage, and breast milk [94,96].
Dysregulated miRNAs in SSc could have an inducing or repressing action, regulating the main pathways of the disease: microvascular changes, fibrosis, and immune alteration. Also, with regards to SSc, altered expression levels of miRNAs have been observed in both sclerodermic tissues and biofluids of SSc patients.

3.1. Alteration of MiRNA Profiles in SSc Tissues

Vascular impairment results are among the early hallmarks of SSc; this process leads to endothelial dysfunction and damage, worsened by tissue oxidative stress, hypoxia, and the recruitment of proinflammatory and profibrotic mediators. The key miRNA involved in SSc-related vasculopathy is miR-193b, which has been observed as decreased in SSc skin and fibroblasts [51]. This miRNA targets the uPA (urokinase-type plasminogen activator) gene, which deregulated expression induces angiogenesis impairment and enhances fibrotic and apoptotic pathways. Blood vessel injuries are typically marked by profound phenotypic and molecular alterations, involving, in particular, vascular smooth muscle cells (VSMCs), which undergo dedifferentiation and acquire a more proliferative and migratory phenotype [51].
The fibrotic process consists of the accumulation of a complex ECM composed of collagen, elastin, and fibronectin. The transforming growth factor (TGF)-β is a key profibrotic mediator, stimulating fibroblast proliferation and accumulation and ECM production [97]. The let-7 family miRNAs are highly conserved across animal species and the first observed miRNAs in humans [98,99]. A miRNA array analysis evidenced a significant downregulation of let-7 family miRNAs, particularly let-7a, in the skin of systemic and localized sclerodermic (LoSc) patients compared to normal and keloid skin samples [52]. Let-7a downmodulation was confirmed both in vitro and in a mouse model of bleomycin-induced dermal sclerosis and resulted in the excessive abnormal expression of type I collagen, typical of a profibrotic effect [52]. Another study reported that five let-7 family members (let-7a, let-7d, let-7e, let-7f, and let-7g) were downregulated in skin biopsies of patients with pulmonary hypertension (PH), one of the most severe complications of SSc. In addition, the let-7b and 7d levels were inversely correlated with the PH severity, highlighting the potential role of these skin miRNAs as biomarkers for PH severity in SSc subjects [53]. In contrast, Li and colleagues reported increased levels of let-7g in skin biopsies of SSc patients, which correlated with SSc pathogenesis [54]. An integrative analysis of miRNA–mRNA expression profiles in the skin of SSc patients highlighted that 21 miRNAs were differentially expressed in SSc compared to healthy controls; altered miRNAs were mainly involved in the Toll-like receptor, TGF-β, and Wnt signaling pathways [55]. Validation experiments confirmed that miR-146b, miR-130b, miR-21, miR-31, and miR-34a were highly expressed in SSc skin tissues and fibroblasts and in ECs after in vitro stimulation with SSc sera. On the contrary, miR-145 and miR-10a showed decreased levels in both SSc skin biopsies and primary skin fibroblasts and were deregulated after stimulation with SSc sera [55]. MiR-27a-3p was decreased both in SSc lung and skin tissues, which promoted fibrotic gene expression by the SPP1/ERK1/2 axis [56]. MiR-150 is another downregulated miRNA in skin and dermal fibroblasts at the plasmatic level [57]. Downregulation of this miRNA is linked to integrin β3-mediated TGF-β activation and led to the overexpression of type I collagen [57]. Zhu and colleagues reported increased levels of miR-21 and decreased amounts of miR-145 and miR-29b in skin biopsies and primary fibroblasts of lSSc and dSSc patients [58]. MiR-21, miR-145, and miR-29b predictively interacted with SMAD7, SMAD3, and COL1A1 mRNAs, respectively, potentially regulating the TGF-β pathway [58,100]. In particular, miR-21 is induced by TGF-β and, in turn, regulates TGF-β-mediated fibrogenic activation of skin fibroblasts by targeting Smad7. MiR-21 expression was also observed to increase in a SSc-bleomycin-induced mouse model, in which antifibrotic treatment with bortezomib restored the miR-21 and Smad7 levels [59]. In addition, miR-21 overexpression also affected SSc lung and dermal fibroblasts [59,60] and regulated Toll-like receptor signaling, suggesting its potential role in immune activation [101]. Both miR-130 and miR-202-3p were upregulated in skin tissue and fibroblasts of SSc patients, showing profibrotic activity by regulating TGF-β via antifibrotic PPARγ inhibition and MMP1 expression, respectively [61,62]. On the contrary, miR-320a has been reported to have an antifibrotic role. Its decreased levels have been observed both in fibrotic SSc patients with interstitial lung disease (ILD) complications and in a bleomycin-induced mouse model [62]. Its decrease in human pulmonary fibroblasts determined the inhibition of type I collagen deposition through the direct regulation of TGFBR2 (transforming growth factor beta receptor 2) and IGF1R (insulin-like growth factor 1 receptor) [63]. Moreover, SSc dermal tissue and primary fibroblasts contained significantly decreased levels of miR-125b and miR-3606-3p [54,64,102]. MiR-125b downregulation induced apoptosis and hindered proliferation and α-SMA expression in dermal fibroblasts cells [102]. MiR-3606-3p targets TGFβ-receptor type 2 (TGFBR2) and has been proposed as an antifibrotic modulator since its overexpression leads to a decline in the Smad2/3 levels and a decrease of type I collagen [64]. MiR-483-5p has been observed typically increased in fibroblasts and endothelial cells, where the expression of fibrosis-related genes is sustained, including collagen-type IV alpha 1 and 2 (COL4A1 and COL4A2) [65,103]. In addition, increased levels of miR-483-5p in endothelial cells lead to the increased transcription of two known myofibroblast differentiation markers (α-SMA and SM22A), promoting the transition to a myofibroblast phenotype [65]. MiR-155 is the second-most relevant miRNA in rheumatic diseases [6]. It was upregulated in SSc and LoSc skin lesions and correlated with the extent of fibrotic tissue damage [66]. Its profibrotic activity lies in the enhancement of Wnt/β-catenin and Akt pathways by the direct targeting of casein kinase 1a (CK1α) and src homology 2-containing inositol phosphatase-1 (SHIP1) [66]. In addition, treatment with antagomir-155 in mice results in a significant decrease of the extent of bleomycin-induced skin fibrosis, highlighting its role as a potential therapeutic target of SSc disease [66]. MiR-155 is also involved in SSc pulmonary fibrosis, being the most expressed miRNA in SSc lung fibroblasts [67]. Several data report its role in regulating both innate and adaptative immune responses [104,105].
The dysregulation of immune responses in SSc disease can be evidenced by the increased presence of inflammatory cells and mediators at the tissue level, together with the enhancement of type I interferon signature and autoimmune responses. MiR-155 SSc-related overexpression is promoted by inflammasome activation and leads to collagen production [67]. MiR-26b-5p, found upregulated in SSc fibroblasts, predictively targets CXCL9 and CXCL13 [68]. Of note, the miR-26b-5p inhibitor hinders the expression of cell fibrosis markers (α-SMA, fibroblast activation protein (FAP), collagen-type I alpha 2 (Col1A2), and Col4A1) through the NF-kB and JAK-STAT pathways in in vitro TGF-β-activated skin fibroblasts. Thus, this miRNA has been suggested as a promising target for SSc therapy [68]. Several miRNAs, including miR-618, miR-126, and miR-139-5p, act by unbalancing the immune response of SSc patients, particularly of plasmacytoid dendritic cells (pDCs). The upregulation of these miRNAs in SSc pDCs induces IFNα production and promotes IFN-inducing genes, causing SSc-pDC imbalance [69,70]. In particular, miR-618 targets an important transcriptional factor for pDC development and activation (IFN regulatory factor 8, IRF-8), and its upregulation in SSc dendritic cells inhibits pDC differentiation and promotes their ability to release IFNα [70]. The MiR-126 and miR-139-5p levels were positively correlated with the increased expression of IFN-responsive genes, suggesting the role of these miRNAs in the interferon response and immune dysregulation in SSc pathogenesis, even at the early stage of the disease [69]. Furthermore, miR-139-5p suppressed the expression of the deubiquitinating enzyme USP24, which is a cofactor for TLR7/9 signaling [69,106].

3.2. Alteration of Circulating MiRNAs in SSc

MiRNAs can be secreted into the extracellular environment, often associated with RNA-binding proteins or inside exosomal vesicles [93,94]. The profile and abundance of specific extracellular miRNAs can reflect the physiological condition of the subject; therefore, they have been widely suggested as potential biomarkers for difficult-to-diagnose diseases. The first data regarding the presence of circulating miRNAs in SSc disease involved miR-29a, which was first quantified in the serum of SSc patients, although no difference between SSc and healthy controls was observed [71]. Interestingly, a significant decrease of this miRNA was evidenced in a group of patients diagnosed as “scleroderma spectrum disorder” (SSD), who do not fit the ACR criteria for SSc but are at risk to develop the disease based on the points system proposed by Ihn et al., which assigns a global score based on five predisposing factors (extent of skin sclerosis, pulmonary abnormalities, antinuclear antibodies, pattern of Raynaud’s phenomenon, and nailfold bleeding) [71]. MiR-29a targets both α1(I) collagen and α2(I) collagen transcripts, so the dysregulation of type I collagens may be triggered by downmodulated miR-29a levels at the SSD stage but maintained by other miRNAs in SSc patients [71].
Further studies have observed a distinctive decreased expression of several miRNAs in different types of biofluids of SSc subjects, proposing their use as useful markers of the disease. Antifibrotic Let-7a showed a significant downregulation in the serum of SSc and LoSc patients compared to healthy controls, showing an inverse correlation with disease severity [52,72]. Similarly, miR-7 and miR-196a were expressed less in LSc sera compared to healthy subjects [72,73]. Of note, data concerning miR-7 expression reported conflicting findings [107]. Specifically, its downmodulation has also been observed locally in LSc dermal fibroblasts, and its general decrease is thought to contribute to LSc pathogenesis through the overexpression of α2(I) collagen protein [72]. On the other side, the microarray analysis reported increased levels of miR-7 in SSc dermal fibroblasts, which determined an antifibrotic effect, maybe as a negative feedback loop, although not sufficient, against excessive fibrogenesis [108]. MiR-196a has been reported under-expressed in LSc skin tissue and serum in vivo, and it is associated with the overexpression of type I collagen [73]. A quantitative PCR screening of 95 miRNAs targeting SSc-related genes by in silico predictive analysis highlighted a remarkable downregulation of about 95% of the tested miRNAs, and in particular, miR-30b were the most heavily decreased in SSc sera compared to healthy specimens, with levels inversely correlated with modified Rodnan skin scores [74]. The downmodulation was also confirmed in bleomycin-treated sclerotic skin in mouse model and in fibrotic skin of SSc patients [74]. This miRNA was observed to regulate PDGFR-β (platelet derived growth factor receptor beta) expression, thus suggesting the role of miR-30b in the pathogenetic PDGFR-β-driven pathway of SSc [74]. MiR-150 was observed underrepresented in SSc sera, compared to healthy individuals, and lower miRNA levels correlated with disease severity [57]. As observed in vitro in dermal fibroblasts, miR-150 acts by regulating integrin b3 and type 1 collagen expression, so its depletion induces an upregulation of these genes implicated in SSc pathogenesis [57].
Conversely, elevated levels of circulating miR-4484 and miR-5196 were detected in the serum of patients with SSc, compared to healthy controls [75,76]. Bioinformatics analysis revealed that miR-4484 could be potentially involved in the TGF-β signaling cascade, ECM-receptor interaction, and MMP expression. Among the latter pathway, the most notable target gene may be MMP-21, which has indeed been observed significantly increased in SSc patients compared to healthy subjects [75]. MiR-5196 exogenous delivery reduced the expression of transcription factor Fra2 (Fos-related antigen-2) and TIMP-1 (tissue inhibitor of metalloproteinases), hinting that it could be useful as a potential regulator of SSc fibrogenesis. In addition, it was also positively correlated with C-reactive protein levels and has been suggested as a biomarker of inflammation in the pathology [75].
Furthermore, other circulating miRNAs resulted upregulated in SSc patients, at plasmatic/serum levels. For example, circulating miR-92a was remarkably elevated in lSSc and dSSc sera and dermal fibroblasts from SSc skin [77]. Authors suggest that miR-92a upregulation may be an effect of TGF-β activation, and the overexpression of this miRNA could induce a downregulation of MMP-1, regulating collagen accumulation [77]. The circulating miR-483-5p was reproducibly observed upregulated in two independent SSc cohorts, including patients with preclinical-SSc symptoms (early SSc) and with localized sclerosis [78]. In particular, its expression correlated with the modified Rodnan skin score (mRSS), in dcSSc subgroup. Its stable expression levels over time, together with its specific upregulation only in SSc disease, and not in other systemic Ads, like SLE and primary SS, suggest its potential role as disease biomarker [78]. Consistently, upregulation of miR-483-5p in fibroblasts and endothelial cells is associated with an increased expression of fibrosis-related genes [65,103].
On the other hand, contrasting results could be observed depending on the type of biofluid investigated, even between serum and plasma samples, as reported for miR-142-3p, a miRNA regulating the expression of integrin αV. Indeed, it was observed significantly higher expressed in serum of both diffuse and limited SSc subjects, compared to healthy controls, and different from other patients’ groups, including SLE, dermatomyositis (DM), or SSD subjects [109]. In contrast, a more recent study reported a decrease of its plasmatic levels in SS patients, compared to controls [110]. In addition to tissue level, miR-21-5p was also found significantly increased at plasmatic level in patients with pulmonary arterial hypertension (APAH), associated with sclerodermic disease [79]. Likewise, upregulated levels of miR-155 were consistently observed also in the sera of SSc patients compared to healthy individuals [80,81]. MiR-155 increase is associated with an early stage of digital microangiopathy, suggesting its potential role as a biomarker for vasculopathy complication on SSc [80].
In order to improve the reliability of circulating miRNAs as potential biomarkers, rather than assessing the abundance and activity of individual molecules, more recent investigations evaluate the expression pattern of multiple miRNAs profile discriminating SSc sera from control individuals, either healthy or with other autoimmune disorders. Following this approach, it has been observed that joint analysis of miR-206 and miR-21 serum levels was more effective to differentiate SSc patients from normal subjects, than single evaluation of miR-206 or miR-21 levels [111]. Another study revealed that miR-223, miR-181b, miR-342-3p, miR-184 were useful in discriminating between diffuse and cutaneous SSc, based on regression analysis and ROC curve evaluation [112]. The analysis of a wider set of miRNAs and study populations evidenced that miRNA-17~92 cluster members (miR-17, miR-20a, miR-92a, and miR-106a) were differentially decreased in SSc sera and can discriminate SSc pathogenic condition from healthy controls. In addition, a set of four opposite regulated miRNAs can be useful in separating SSc (marked by miR-142-3p and miR-223 downmodulation and miR-150 and miR-638 increase) from SLE patients (showing upregulated miR-142-3p and miR-223, together with decrease expression of miR-150, and unaltered miR-638 levels) [110].
MiRNAs-enriched exosomes could be released in the extracellular milieu, having a role in the crosstalk between cells and acting as signaling mediators [95,113]. The number of circulating exosomes was lower in sclerodermic patients, compared to physiological conditions, and a decreased quantity of exosomes has been associated with an increase in vascular complications SSc-related. Considered as a whole, the exosomal fraction isolated from sera of lcSSc and dcSSc patients was found to have an overall profibrotic activity, showing a marked upregulation of 6 profibrotic miRNAs (let-7g, miR-17, miR-23b, miR-155, miR-215, and miR-503), together with decreased levels of 10 antifibrotic miRNAs (let-7a, miR-26b, miR-29b, miR-92a, miR-129, miR-133, miR-140, miR-146a, miR-196a and miR-223) when compared to healthy sera [114]. Interestingly, SSc exosomes induced the expression of profibrotic genes in normal human dermal fibroblasts in vitro treated [114].

4. Interplay between Putative Causal Agents of Viral Origin and MiRNAs in SSc

4.1. Beta-Herpesvirus Infection and MiRNA Dysregulation in SSc

Several infectious agents have been suggested as potential triggering and/or contributing factors in SSc etiology, in particular those able to persist and occasionally reactivate in the host in adulthood [28,115]. In particular, the most widely studied viruses in association with the disease belong to the human Orthoherpesviridae family and Betaherpesvirinae subfamily, including HCMV and HHV-6 [41,44,116,117,118]. Both viruses are highly prevalent in the human population, and, commonly to other herpesviruses, after the primary infection in early childhood, they establish a latent infection in the host, with symptomatic reactivation in immunocompromised adults, where they have been associated with a broad range of autoimmune diseases, including connective tissue diseases [117,118,119,120].
Recent observations have confirmed the high prevalence of beta-herpesvirus infection in sclerodermic patients, highlighting HHV-6/HCMV presence both in the tissue and/or blood level (in particular of HHV-6A, which show a prominent tissue tropism), together with significantly enhanced immune response toward viral antigens, such as UL94 (HCMV) and U94 (HHV-6), compared to the healthy population [41,121,122,123,124,125,126]. In order to understand their potential role and their joint involvement in inducing fibrosis, the effects of in vitro infection of primary human dermal fibroblasts (one of the main cell targets affected in SSc pathology, together with endothelial cells) were evaluated [42,43,44,82]. HHV-6A and HCMV in vitro infections induced a rapid and sustained up-modulation of several profibrotic and pro-apoptotic factors, that resulted even more marked by HHV-6A/HCMV coinfection [43,44]. Interestingly, in vitro single and coinfection by those beta-herpesviruses induced a deep modulation of cell miRNome, starting from viral adsorption and early times post infection as well as later times, until 14 days post-infection (d.p.i.). Of note, HCMV and HHV-6A individual infections induced a marked deregulation of a wide set of miRNAs at each tested time, d.p.i., impacting the expression of up to 43 and 29 miRNAs, respectively [42]. The most altered miRNAs upregulated by both viruses and associated with SSc pathways resulted miR-7, miR-let-7g, miR-92a [42].
All these miRNAs were reported upregulated in SSc dermal fibroblasts, and were associated with fibrogenesis, excessive collagen accumulation and pulmonary involvement SSc-related [54,77,108]. In parallel, beta-herpesviruses infection induced a remarkable downregulation of other miRNAs associated with SSc, namely miR-let-7a, miR-10a, and miR-193 [42]. This negative deregulation trend parallels what has been observed in SSc skin biopsies and primary dermal fibroblasts [51,52,72]. Moreover, HCMV infection determined an upregulation of miR-146b, and a downregulation of miR-29, miR-30b and miR-125b. MiR-146b has been previously observed increased in SSc skin tissues and fibroblasts, as well as in fibroblasts and ECs stimulated with SSc patient sera [55]. MiR-29 has a wide reported antifibrotic activity, and it is involved fibrosis repair at different tissue levels, from skin to internal organs such as heart, lung and kidney [127,128,129]. MiR-30b has been suggested as a marker of disease severity, and its amount is inversely correlated with modified Rodnan skin scores and miR-125b [74,107]. The literature data regarding miR-125b highlighted its decreased levels in SSc skin samples and primary dermal fibroblasts, hinting at its potential role as an antifibrotic and antiapoptotic regulator in the disease [54,102]. Last, miR-20 appeared heavily downregulated by HHV-6A infection [42]. Its lower expression in liver fibrosis induced the activation of the TGF-signaling pathway, driving forward extracellular matrix (ECM) production [130]. Interestingly, signaling pathways potentially regulated by HCMV/HHV-6A-induced miRNAs included those related to the fibrotic process, including TGF-β, Wingless/Int (WNT), and β-catenin [42].
HCMV and HHV-6A viruses, highly prevalent in the human population, are likely to infect or reactivate simultaneously in the host. According to this, their synergistic effect on the modulation of miRNAs in coinfected cells has been investigated [82]. Interestingly, the copresence of HCMV and HHV-6A induced a remarkable dysregulation of fibrosis-associated miRNAs expression compared to what was observed in single-infected primary dermal fibroblasts. Overall, a significant upregulation of several profibrotic miRNAs has been observed, coupled in parallel by a likewise pronounced decrease in miRNAs with antifibrotic activity, thereby supporting the profibrotic influence of HCMV/HHV-6A coinfection. Of note, the joint impact on miRNAs modulation involved various miRNAs associated with SSc disease (such as miR-10a, miR-29, and miR-155) but also with other fibrosis-associated diseases, broadening the potential putative implication of HCMV and HHV-6A coinfection in multiple fibrosis-related diseases.
The miRNAs mainly altered in SSc disease, also highlighting the involvement of HCMV and HHV6-A single or double infections, are summarized in Table 1.

4.2. Other Virus-Induced MiRNA Dysregulation in SSc

Among the potential viruses associated with the development of SSc, EBV was investigated as a possible trigger. EBV is a human herpesvirus belonging to the Gammaherpesvirinae subfamily and was firstly isolated from cultured lymphoblasts derived from a Burkitt’s lymphoma (BL) [132]. Its persistency in latently infected cells has been broadly associated with malignancies, such as endemic BL, Hodgkin’s lymphoma (HL), EBV-positive diffuse large B-cell lymphoma (DLBCL), nasopharyngeal carcinoma (NPC), and EBV-associated gastric carcinoma [133]. In parallel, an increasing body of evidence supports its role in exacerbating and potentially triggering autoimmune and autoinflammatory diseases, including SLE, MS, RA, SS, and even SSc [47,134]. Specifically concerning SSc, EBV infection has been reported since 1981 in some clinical cases [45,46]. Moreover, its infection has been recently detected in monocytes isolated from SSc patients and associated with the activation of Toll-like receptor 8 (TLR8) and the induction of IFN-mediated innate immune response in primary monocytes, suggesting a novel mechanism by which EBV could trigger monocyte inflammation in SSc [47]. Of note, EBV-induced miRNAs were detected and reported to play an important role in maintaining the virus infection and immune evasion [133]. EBV itself encodes for over 40 miRNAs clustered into two genome regions: BART (BamH I-A rightward transcript) and BHRF1 (BamH I-H rightward fragment 1) [135,136,137]. EBV-encoded miRNAs were recognized to support the latent EBV infection, to elude both innate and adaptative immune surveillance of the host, and to promote the neoplastic growth of infected cells [135]. On the other hand, EBV was also reported to induce a deep modulation of multiple host miRNAs, so far mostly recognized as key factors involved in virus-induced oncogenesis and immune escape, including miR-21, the miR-17–92 cluster, and miR-155 [133]. Interestingly, a putative role of EBV-miRNAs in the progression of idiopathic pulmonary fibrosis (IPF) was recently indicated through the induction of epithelial–mesenchymal transition (EMT) in alveolar epithelial cells [138]. Thus, the investigation of EBV-induced miRNAs in a wider range of pathologies, including SSc, could pave the way to a deeper understanding of the mechanisms underlying the autoimmune-based fibrotic processes.
Another virus studied for its potential association with SSc onset is represented by the human Parvovirus B19 (B19V), which is associated with a wide range of pathologies, including erythema infectiosum, arthralgia, fetal death, and autoimmune diseases such as SSc, based on several published studies [139,140,141,142,143,144]. Specifically, SSc patients were observed to show an increased prevalence of B19V viremia and seroprevalence of anti-B19V NS1 antibodies, which are considered a marker of persistent B19V infection [140,141,144]. B19V can infect in vitro dermal fibroblasts and endothelial cells, the main target cells of SSc disease [49,145], together with circulating angiogenic cells (CACs), involved in the vascular regeneration process [146]. At the cellular level, the pathogenic mechanisms by which B19V could cause SSc were linked to the induction of fibrosis (by increased fibroblast activation, migration, and expression of profibrotic factors) [49]; decrease of neovascularization (by the induction of CAC apoptosis) [146], and activation of inflammasome and immune-mediated inflammatory tissue damage [50]. Despite these clues indicating the potential involvement of B19V in the onset or progression of SSc, the role of miRNAs in the virus pathogenesis has been poorly investigated, essentially concerning the regulation of B19V tropism [147,148]; thus, the potential role of B19V-induced miRNAs in SSc has yet to be explored.

4.3. Oxidative Stress and MiRNA Dysregulation in SSc

Notably, oxidative stress is often a result of virus infection and has been reported for many virus types, including influenza viruses, hepatitis B virus, human immunodeficiency virus, human coronaviruses, and herpesviruses [149,150,151,152,153,154]. On the other hand, oxidative stress favors herpesvirus infection [155], triggering an imbalance between the increased production of ROS and reduced antioxidant host responses.
The production of ROS and RNS is part of the physiological processes of cellular metabolism, immune response regulation, and inflammation. In several autoimmune diseases, the abnormal production of ROS and RNS, in the proinflammatory status, leads to permanent tissue damages caused by the chemical interactions between reactive compounds and biomolecules such as DNA, proteins, and lipids [40]. Several literature data have highlighted an increased oxidative stress condition in SSc disease [156,157,158,159]. Briefly, higher levels of several oxidative stress biomarkers, including malondialdehyde (MDA), nitric oxide (NO), asymmetric dimethylarginine, and hydroperoxides (ROOH) have been detected in the circulating blood of SSc patients [160]. Increased levels of nitrated proteins and dimethylarginine (ADMA) were observed in dSSc patients and were associated with disease severity and duration [161]. In parallel, the total antioxidant power (TAP) is increased in the serum of SSc patients in response to the increase in oxidative stress [162]. In addition, the enhanced oxidative stress condition has also been detected in SSc urine samples, harboring high urinary levels of 8-isoprostaglandin-F (8-iso-PGF) and hydroxydeoxyguanosine (8-OHdG), biomarkers of free radical damage and DNA oxidative damage, respectively, compared to healthy subjects [163,164].
Lastly, fibrotic SSc skin harbored increased levels of ROS products, together with enhanced nitrotyrosine staining and the excessive abundance of proteins involved in the oxidative stress response, such as peroxiredoxin1 and carbonyl reductase I, compared to control tissues [161,165,166]. Evidence has confirmed the role of oxidative stress in SSc, although the primary cause leading to ROS production is still unclear; some hypotheses support the primary role of NADPH oxidases (NOX)—in particular, NOX2 and NOX4 [167,168].
Other published data have reported a dysregulation of antioxidant systems in SSc disease, affecting, in particular, the levels of CAT, vitamins C and E, and glutathione (GSH), together with the total antioxidant capacity, which was significantly underrepresented in plasma from SSc patients compared to the controls [162,169,170,171].
At the cellular level, pathways involved in oxidative stress and immune responses are finely regulated by miRNAs and vice versa, and ROS/RSN reactive molecules can affect the miRNA levels, showing a close interconnection and mutual regulation among ROS, miRNAs, and inflammation. Several miRNAs have been associated with oxidative stress and immune dysregulation in SSc pathology. To date, the interconnection between miRNA dysregulation and the impairment of the oxidative stress pathway in SSc diseases are still under active investigation.
The main altered miRNAs in SSc involved in the oxidative stress pathways included miR-21, miR-29a, miR-135b, miR-155, miR-182, miR-193b, and miR-200c. Mir-21, together with miR-200c, were found to be increased in SSc patients [58,59,60,89] and mediate the PDCD4 (programmed cell death protein 4)/PTEN (homologous phosphatase and tensin homolog) pathway, resulting in the inhibition of ROS oxidative stress [88,90]. MiR-29a, found decreased in SSc [58,59,71], and target SIRT1 (silent information regulator factor 2–related enzyme 1) histone deacetylase can deacetylate the FoxO factors and consequently regulate the expression of antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and thioredoxin (TRX) [91]. In addition, miR-29 is also reported to target insulin grow factor 1 (IGF1), which is an upstream activator of the AKT (protein kinase B) signaling pathway, through phosphatidylinositol 4,5-bisphosphate (PIP2) conversion [172]. BMP-7 (bone morphogenetic protein 7), another upstream regulator of the AKT pathways, is instead regulated by miR-135b [92]. Since miR-135b also negatively regulates USP15, a deubiquitinase involved in the Keap1/Nrf2 antioxidative pathway [83], its decreased levels in SSc may also affect the protective effect of this pathway on oxidative stress. MiR-155 has a well-known role as inflamma-miR, finely regulating the NF-κB pathway [173], and its upregulation has been reported in sclerotic lesions of different tissues [66,67,131]. In addition, the knockdown of miR-155 could regulate ROS production, NO generation, and apoptosis via different pathways, including those mediated by PI3K/Akt [174]. As already mentioned, miR-193b contributes to proliferative vasculopathy in SSc, mediated by uPA regulation [51]. Lastly, another miRNA, miR-182, has been described as involved in oxidative stress regulation, being upregulated in SSc patients [85]. It could have a bivalent role, both inhibiting NOX4 and thus decreasing ROS production or upregulating the intracellular redox status via downmodulation of the antioxidant modulator sestrin2 (SESN2) [86,87].

5. Future Directions

The rapid development of omics analysis technologies, including next-generation sequencing and other high-throughput methods, allows the genome-wide investigation of miRNome defining healthy and disease conditions, including Systemic Sclerosis. The characterization of dysregulated miRNA expression levels, which either define SSc pathology or are induced by environmental stimuli, such as oxidative stress and viral infection, provides insight into the molecular mechanisms primarily involved in disease onset, progression, and exacerbation. Among the most advanced analytical methods applied to detect and quantify the miRNAs levels, miRNA sequencing has assumed a growing interest, allowing also the identification of novel miRNAs and pre-miRNAs, unlike the qRT-PCR and miRNA array methods [175]. To date, public databases have more than two thousand miRNAs registered, highlighting the potential of a bioinformatics-integrative approach for genome-wide identification and miRNA–mRNA gene prediction [176,177,178].
A further improvement of the miRNome investigation involves the study of a single cell rather than a bulk of cells constituting tissue biopsies and PBMC fractions. Epigenetic regulation often involves specific cell subpopulations of immune cells or cell types within the same tissue (i.e., ECs vs. myofibroblasts in skin biopsy). Single-cell technologies, such as mass cytometry, cell sorting (FACS), single-cell RNA sequencing, and spatially resolved transcriptomics, represent cutting-edge technologies that enable the comprehensive characterization of dynamic epigenetic changes with single-cell resolution [179,180].
As a result of these new approaches, the increasing knowledge of the miRNA-regulated molecular mechanisms has opened the way to the design of innovative therapeutic approaches. MiRNAs could be investigated as therapeutics (miRNA mimics) or as targets of therapeutics (anti-miRNAs) in order to inhibit the transcription of multiple target genes [181]. Although a promising use of miRNA-based therapeutic tools, the main challenges to overcome include the optimization of proper administration routes, the control of delivery in the blood, the specific targeting of tissues and cell types, and the minimization of adverse effects. Given these multiple aspects still to be optimized, only a few miRNA-based drugs have been evaluated in a clinical test phase so far [181].
On the other hand, circulating miRNAs are stable, less susceptible to degradation than RNA, and easily detectable and quantifiable by sensitive methods in different types of body fluids, first and foremost being blood and plasma [23,79,94,114]. These characteristics make them reliable biomarkers for the diagnosis and discrimination of several autoimmune diseases, including SSc, as demonstrated in several studies so far [79,107,109,110,112,182,183]. However, their use as biomarkers is associated with downsides that need to be addressed—in particular, the choice of biological fluid and the appropriate method of collection, the potential contamination of intracellular RNA, the choice of the most sensitive and potentially standardizable analytical method, and last, the method of data normalization [184,185]. In conclusion, rigorous trials on a large study population will be needed to validate the diagnostic potential of these molecules. Based on the available data so far, general conclusions are still difficult to draw, since a solid interpretation is still hampered by the limitations described for each individual study. Thus, despite the large amount of collected data, future research would be needed to confirm the data in studies conducted in larger cohorts and by using standardized procedures in order to achieve sound and reliable results specifically targeted to SSc and useful for clinical applications.

6. Conclusions

Systemic Sclerosis is a multifaceted autoimmune disease characterized by vascular complications, immune dysregulation, and an excessive fibrosis affecting multiple organs. Although, in recent years, an earlier diagnosis and the routine examination of organ involvement have improved treatment and overall survival, the therapeutic options for SSc are mainly symptom- and organ-based. The little knowledge of the causative factors and the high complexity of the disease result in the lack of an effective treatment for SSc. The research on miRNA modulation in SSc patients, together with that induced in infected cells by the putative SSc causal factors such as beta-herpesviruses, has opened a new perspective on our options to understand the mechanisms by which specific agents could contribute to the onset and progression of the disease. Based on available data, some miRNAs emerged as potential profibrotic factors, both detectable in SSc patients and in beta-herpesvirus infected cells (i.e., miR-let-7, miR-146, and miR-155). From this perspective, miRNAs could serve as novel biomarkers for the rapid end sensitive SSc diagnosis and could represent promising therapeutic targets for SSc treatment.

Author Contributions

Conceptualization, E.C.; methodology, I.S.; software, I.S., M.D., F.B., and E.M.; validation, M.D., F.B., E.M., D.D.L., and C.M.; investigation, I.S., M.D., F.B., E.M., and C.M.; resources, E.C.; data curation, F.B., E.M., D.D.L., and M.-C.A.; writing—original draft preparation, I.S.; writing—review and editing, D.D.L., C.M., M.-C.A., and E.C.; visualization, D.D.L. and M.-C.A.; supervision, E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Shang, R.; Lee, S.; Senavirathne, G.; Lai, E.C. MicroRNAs in Action: Biogenesis, Function and Regulation. Nat. Rev. Genet. 2023, 24, 816–833. [Google Scholar] [CrossRef]
  2. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional Regulation of the Heterochronic Gene Lin-14 by Lin-4 Mediates Temporal Pattern Formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
  3. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans Heterochronic Gene Lin-4 Encodes Small RNAs with Antisense Complementarity to Lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  4. Lee, Y.; Jeon, K.; Lee, J.T.; Kim, S.; Kim, V.N. MicroRNA Maturation: Stepwise Processing and Subcellular Localization. EMBO J. 2002, 21, 4663–4670. [Google Scholar] [CrossRef]
  5. Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef]
  6. Shaikh, F.S.; Siegel, R.J.; Srivastava, A.; Fox, D.A.; Ahmed, S. Challenges and Promise of Targeting MiRNA in Rheumatic Diseases: A Computational Approach to Identify MiRNA Association with Cell Types, Cytokines, and Disease Mechanisms. Front. Immunol. 2023, 14, 1322806. [Google Scholar] [CrossRef]
  7. Huntzinger, E.; Izaurralde, E. Gene Silencing by MicroRNAs: Contributions of Translational Repression and MRNA Decay. Nat. Rev. Genet. 2011, 12, 99–110. [Google Scholar] [CrossRef]
  8. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 388354. [Google Scholar] [CrossRef]
  9. Orang, A.V.; Safaralizadeh, R.; Kazemzadeh-Bavili, M. Mechanisms of MiRNA-Mediated Gene Regulation from Common Downregulation to MRNA-Specific Upregulation. Int. J. Genom. 2014, 2014, 970607. [Google Scholar] [CrossRef]
  10. Desvignes, T.; Batzel, P.; Berezikov, E.; Eilbeck, K.; Eppig, J.T.; McAndrews, M.S.; Singer, A.; Postlethwait, J.H. MicroRNA Nomenclature: A View Incorporating Genetic Origins, Biosynthetic Pathways, and Sequence Variants. Trends Genet. 2015, 31, 613–626. [Google Scholar] [CrossRef]
  11. Lewis, B.P.; Shih, I.H.; Jones-Rhoades, M.W.; Bartel, D.P.; Burge, C.B. Prediction of Mammalian MicroRNA Targets. Cell 2003, 115, 787–798. [Google Scholar] [CrossRef]
  12. Eulalio, A.; Huntzinger, E.; Izaurralde, E. Getting to the Root of MiRNA-Mediated Gene Silencing. Cell 2008, 132, 9–14. [Google Scholar] [CrossRef]
  13. Bushati, N.; Cohen, S.M. MicroRNA Functions. Annu. Rev. Cell Dev. Biol. 2007, 23, 175–205. [Google Scholar] [CrossRef]
  14. Fu, G.; Brkić, J.; Hayder, H.; Peng, C. MicroRNAs in Human Placental Development and Pregnancy Complications. Int. J. Mol. Sci. 2013, 14, 5519–5544. [Google Scholar] [CrossRef]
  15. Paul, P.; Chakraborty, A.; Sarkar, D.; Langthasa, M.; Rahman, M.; Bari, M.; Singha, R.K.S.; Malakar, A.K.; Chakraborty, S. Interplay between MiRNAs and Human Diseases. J. Cell Physiol. 2018, 233, 2007–2018. [Google Scholar] [CrossRef]
  16. Van Rooij, E.; Sutherland, L.B.; Qi, X.; Richardson, J.A.; Hill, J.; Olson, E.N. Control of Stress-Dependent Cardiac Growth and Gene Expression by a MicroRNA. Science 2007, 316, 575–579. [Google Scholar] [CrossRef]
  17. Qiao, Y.-C.; Pan, Y.-H.; Ling, W.; Tian, F.; Chen, Y.-L.; Zhang, X.-X.; Zhao, H.-L. The Yin and Yang of Regulatory T Cell and Therapy Progress in Autoimmune Disease. Autoimmun. Rev. 2017, 16, 1058–1070. [Google Scholar] [CrossRef]
  18. Ceccarelli, F.; Agmon-Levin, N.; Perricone, C. Genetic Factors of Autoimmune Diseases 2017. J. Immunol. Res. 2017, 2017, 2789242. [Google Scholar] [CrossRef]
  19. Conrad, N.; Misra, S.; Verbakel, J.Y.; Verbeke, G.; Molenberghs, G.; Taylor, P.N.; Mason, J.; Sattar, N.; McMurray, J.J.V.; McInnes, I.B.; et al. Incidence, Prevalence, and Co-Occurrence of Autoimmune Disorders over Time and by Age, Sex, and Socioeconomic Status: A Population-Based Cohort Study of 22 Million Individuals in the UK. Lancet 2023, 401, 1878–1890. [Google Scholar] [CrossRef]
  20. Shoenfeld, Y.; Selmi, C.; Zimlichman, E.; Gershwin, M.E. The Autoimmunologist: Geoepidemiology, a New Center of Gravity, and Prime Time for Autoimmunity. J. Autoimmun. 2008, 31, 325–330. [Google Scholar] [CrossRef]
  21. Cipriano, G.L.; Schepici, G.; Mazzon, E.; Anchesi, I. Multiple Sclerosis: Roles of MiRNA, LcnRNA, and CircRNA and Their Implications in Cellular Pathways. Int. J. Mol. Sci. 2024, 25, 2255. [Google Scholar] [CrossRef]
  22. Naithani, U.; Jain, P.; Sachan, A.; Khare, P.; Gabrani, R. MicroRNA as a Potential Biomarker for Systemic Lupus Erythematosus: Pathogenesis and Targeted Therapy. Clin. Exp. Med. 2023, 23, 4065–4077. [Google Scholar] [CrossRef]
  23. Zhang, R.; Wei, Y.; Wang, T.; Nie, X.; Shi, Z.; Deng, Y.; Li, D. Exosomal MiRNAs in Autoimmune Skin Diseases. Front. Immunol. 2023, 14, 1307455. [Google Scholar] [CrossRef]
  24. Nag, S.; Mitra, O.; Tripathi, G.; Samanta, S.; Bhattacharya, B.; Chandane, P.; Mohanto, S.; Sundararajan, V.; Malik, S.; Rustagi, S.; et al. Exploring the Theranostic Potentials of MiRNA and Epigenetic Networks in Autoimmune Diseases: A Comprehensive Review. Immun. Inflamm. Dis. 2023, 11, e1121. [Google Scholar] [CrossRef]
  25. Katsumoto, T.R.; Whitfield, M.L.; Connolly, M.K. The Pathogenesis of Systemic Sclerosis. Annu. Rev. Pathol. 2011, 6, 509–537. [Google Scholar] [CrossRef]
  26. Denton, C.P.; Khanna, D. Systemic Sclerosis. Lancet 2017, 390, 1685–1699. [Google Scholar] [CrossRef]
  27. Rongioletti, F.; Ferreli, C.; Atzori, L.; Bottoni, U.; Soda, G. Scleroderma with an Update about Clinico-Pathological Correlation. G. Ital. Dermatol. Venereol. 2018, 153, 208–215. [Google Scholar] [CrossRef]
  28. Ferri, C.; Arcangeletti, M.-C.; Caselli, E.; Zakrzewska, K.; Maccari, C.; Calderaro, A.; D’accolti, M.; Soffritti, I.; Arvia, R.; Sighinolfi, G.; et al. Insights into the Knowledge of Complex Diseases: Environmental Infectious/Toxic Agents as Potential Etiopathogenetic Factors of Systemic Sclerosis. J. Autoimmun. 2021, 124, 102727. [Google Scholar] [CrossRef]
  29. Boin, F.; Wigley, F.M. Clinical Features and Treatment of Scleroderma. In Kelley’s Textbook of Rheumatology: Volume 1-2, 9th ed.; W.B. Saunders: Philadelphia, PA, USA, 2012; Volume 2, pp. 1366–1403. [Google Scholar] [CrossRef]
  30. Chabaud, S.; Moulin, V.J. Apoptosis Modulation as a Promising Target for Treatment of Systemic Sclerosis. Int. J. Rheumatol. 2011, 2011, 495792. [Google Scholar] [CrossRef]
  31. Araújo, F.C.; Camargo, C.Z.; Kayser, C. Validation of the ACR/EULAR Classification Criteria for Systemic Sclerosis in Patients with Early Scleroderma. Rheumatol. Int. 2017, 37, 1825–1833. [Google Scholar] [CrossRef]
  32. Jordan, S.; Maurer, B.; Toniolo, M.; Michel, B.; Distler, O. Performance of the New ACR/EULAR Classification Criteria for Systemic Sclerosis in Clinical Practice. Rheumatology 2015, 54, 1454–1458. [Google Scholar] [CrossRef]
  33. Hoffmann-Vold, A.-M.; Gunnarsson, R.; Garen, T.; Midtvedt, Ø.; Molberg, Ø. Performance of the 2013 American College of Rheumatology/European League against Rheumatism Classification Criteria for Systemic Sclerosis (SSc) in Large, Well-Defined Cohorts of SSc and Mixed Connective Tissue Disease. J. Rheumatol. 2015, 42, 60–63. [Google Scholar] [CrossRef]
  34. Allanore, Y.; Simms, R.; Distler, O.; Trojanowska, M.; Pope, J.; Denton, C.P.; Varga, J. Systemic Sclerosis. Nat. Rev. Dis. Primers 2015, 1, 15002. [Google Scholar] [CrossRef]
  35. Cutolo, M.; Soldano, S.; Smith, V. Pathophysiology of Systemic Sclerosis: Current Understanding and New Insights. Expert. Rev. Clin. Immunol. 2019, 15, 753–764. [Google Scholar] [CrossRef]
  36. Orphanet: Systemic Sclerosis. Available online: https://www.orpha.net/en/disease/detail/90291 (accessed on 5 March 2024).
  37. De Angelis, R.; Ferri, C.; Giuggioli, D.; Bajocchi, G.; Dagna, L.; Bellando-Randone, S.; Zanframundo, G.; Foti, R.; Cacciapaglia, F.; Cuomo, G.; et al. Systemic Sclerosis Sine Scleroderma: Clinical and Serological Features and Relationship with Other Cutaneous Subsets in a Large Series of Patients from the National Registry ‘SPRING’ of the Italian Society for Rheumatology. RMD Open 2023, 9, e002890. [Google Scholar] [CrossRef]
  38. Tian, J.; Kang, S.; Zhang, D.; Huang, Y.; Zhao, M.; Gui, X.; Yao, X.; Lu, Q. Global, Regional, and National Incidence and Prevalence of Systemic Sclerosis. Clin. Immunol. 2023, 248, 109267. [Google Scholar] [CrossRef]
  39. Thoreau, B.; Chaigne, B.; Renaud, A.; Mouthon, L. Pathophysiology of Systemic Sclerosis. Presse Med. 2021, 50, 104087. [Google Scholar] [CrossRef]
  40. Ibáñez-Cabellos, J.S.; Pallardó, F.V.; García-Giménez, J.L.; Seco-Cervera, M. Oxidative Stress and Epigenetics: MiRNA Involvement in Rare Autoimmune Diseases. Antioxidants 2023, 12, 800. [Google Scholar] [CrossRef]
  41. Caselli, E.; Soffritti, I.; D’Accolti, M.; Bortolotti, D.; Rizzo, R.; Sighinolfi, G.; Giuggioli, D.; Ferri, C. HHV-6A Infection and Systemic Sclerosis: Clues of a Possible Association. Microorganisms 2019, 8, 39. [Google Scholar] [CrossRef]
  42. Soffritti, I.; D’Accolti, M.; Ravegnini, G.; Arcangeletti, M.C.; Maccari, C.; De Conto, F.; Calderaro, A.; Caselli, E. Modulation of Micrornome by Human Cytomegalovirus and Human Herpesvirus 6 Infection in Human Dermal Fibroblasts: Possible Significance in the Induction of Fibrosis in Systemic Sclerosis. Cells 2021, 10, 1060. [Google Scholar] [CrossRef]
  43. Soffritti, I.; D’Accolti, M.; Maccari, C.; Bini, F.; Mazziga, E.; de Conto, F.; Calderaro, A.; Arcangeletti, M.C.; Caselli, E. Human Cytomegalovirus and Human Herpesvirus 6 Coinfection of Dermal Fibroblasts Enhances the Pro-Inflammatory Pathway Predisposing to Fibrosis: The Possible Impact on Systemic Sclerosis. Microorganisms 2022, 10, 1600. [Google Scholar] [CrossRef]
  44. Arcangeletti, M.C.; D’accolti, M.; Maccari, C.; Soffritti, I.; De Conto, F.; Chezzi, C.; Calderaro, A.; Ferri, C.; Caselli, E. Impact of Human Cytomegalovirus and Human Herpesvirus 6 Infection on the Expression of Factors Associated with Cell Fibrosis and Apoptosis: Clues for Implication in Systemic Sclerosis Development. Int. J. Mol. Sci. 2020, 21, 6397. [Google Scholar] [CrossRef]
  45. Longo, F.; Saletta, S.; Lepore, L.; Pennesi, M. Localized Scleroderma after Infection with Epstein-Barr Virus. Clin. Exp. Rheumatol. 1993, 11, 681–683. [Google Scholar]
  46. Urano, J.; Kohno, H.; Watanabe, T. Unusual Case of Progressive Systemic Sclerosis with Onset in Early Childhood and Following Infectious Mononucleosis. Eur. J. Pediatr. 1981, 136, 285–289. [Google Scholar] [CrossRef]
  47. Farina, A.; Peruzzi, G.; Lacconi, V.; Lenna, S.; Quarta, S.; Rosato, E.; Vestri, A.R.; York, M.; Dreyfus, D.H.; Faggioni, A.; et al. Epstein-Barr Virus Lytic Infection Promotes Activation of Toll-like Receptor 8 Innate Immune Response in Systemic Sclerosis Monocytes. Arthritis Res. Ther. 2017, 19, 39. [Google Scholar] [CrossRef]
  48. Magro, C.M.; Nuovo, G.; Ferri, C.; Crowson, A.N.; Giuggioli, D.; Sebastiani, M. Parvoviral Infection of Endothelial Cells and Stromal Fibroblasts: A Possible Pathogenetic Role in Scleroderma. J. Cutan. Pathol. 2004, 31, 43–50. [Google Scholar] [CrossRef]
  49. Arvia, R.; Margheri, F.; Stincarelli, M.A.; Laurenzana, A.; Fibbi, G.; Gallinella, G.; Ferri, C.; Del Rosso, M.; Zakrzewska, K. Parvovirus B19 Activates in Vitro Normal Human Dermal Fibroblasts: A Possible Implication in Skin Fibrosis and Systemic Sclerosis. Revmatologiia 2020, 59, 3526–3532. [Google Scholar] [CrossRef]
  50. Zakrzewska, K.; Arvia, R.; Torcia, M.G.; Clemente, A.M.; Tanturli, M.; Castronovo, G.; Sighinolfi, G.; Giuggioli, D.; Ferri, C. Effects of Parvovirus B19 In Vitro Infection on Monocytes from Patients with Systemic Sclerosis: Enhanced Inflammatory Pathways by Caspase-1 Activation and Cytokine Production. J. Investig. Dermatol. 2019, 139, 2125–2133.e1. [Google Scholar] [CrossRef]
  51. Iwamoto, N.; Vettori, S.; Maurer, B.; Brock, M.; Pachera, E.; Jüngel, A.; Calcagni, M.; Gay, R.E.; Whitfield, M.L.; Distler, J.H.W.; et al. Downregulation of MiR-193b in Systemic Sclerosis Regulates the Proliferative Vasculopathy by Urokinase-Type Plasminogen Activator Expression. Ann. Rheum. Dis. 2016, 75, 303–310. [Google Scholar] [CrossRef]
  52. Makino, K.; Jinnin, M.; Hirano, A.; Yamane, K.; Eto, M.; Kusano, T.; Honda, N.; Kajihara, I.; Makino, T.; Sakai, K.; et al. The Downregulation of MicroRNA Let-7a Contributes to the Excessive Expression of Type I Collagen in Systemic and Localized Scleroderma. J. Immunol. 2013, 190, 3905–3915. [Google Scholar] [CrossRef]
  53. Izumiya, Y.; Jinnn, M.; Kimura, Y.; Wang, Z.; Onoue, Y.; Hanatani, S.; Araki, S.; Ihn, H.; Ogawa, H. Expression of Let-7 Family MicroRNAs in Skin Correlates Negatively with Severity of Pulmonary Hypertension in Patients with Systemic Scleroderma. Int. J. Cardiol. Heart Vasc. 2015, 8, 98–102. [Google Scholar] [CrossRef]
  54. Li, H.; Yang, R.; Fan, X.; Gu, T.; Zhao, Z.; Chang, D.; Wang, W.; Wang, C. MicroRNA Array Analysis of MicroRNAs Related to Systemic Scleroderma. Rheumatol. Int. 2012, 32, 307–313. [Google Scholar] [CrossRef]
  55. Zhou, B.; Zuo, X.X.; Li, Y.S.; Gao, S.M.; Dai, X.D.; Zhu, H.L.; Luo, H. Integration of MicroRNA and MRNA Expression Profiles in the Skin of Systemic Sclerosis Patients. Sci. Rep. 2017, 7, 42899. [Google Scholar] [CrossRef]
  56. Cheng, Q.; Chen, M.; Wang, H.; Chen, X.; Wu, H.; Du, Y.; Xue, J. MicroRNA-27a-3p Inhibits Lung and Skin Fibrosis of Systemic Sclerosis by Negatively Regulating SPP1. Genomics 2022, 114, 110391. [Google Scholar] [CrossRef]
  57. Honda, N.; Jinnin, M.; Kira-Etoh, T.; Makino, K.; Kajihara, I.; Makino, T.; Fukushima, S.; Inoue, Y.; Okamoto, Y.; Hasegawa, M.; et al. MiR-150 Downregulation Contributes to the Constitutive Type I Collagen Overexpression in Scleroderma Dermal Fibroblasts via the Induction of Integrin Β3. Am. J. Pathol. 2013, 182, 206–216. [Google Scholar] [CrossRef]
  58. Zhu, H.; Li, Y.; Qu, S.; Luo, H.; Zhou, Y.; Wang, Y.; Zhao, H.; You, Y.; Xiao, X.; Zuo, X. MicroRNA Expression Abnormalities in Limited Cutaneous Scleroderma and Diffuse Cutaneous Scleroderma. J. Clin. Immunol. 2012, 32, 514–522. [Google Scholar] [CrossRef]
  59. Jafarinejad-Farsangi, S.; Gharibdoost, F.; Farazmand, A.; Kavosi, H.; Jamshidi, A.; Karimizadeh, E.; Noorbakhsh, F.; Mahmoudi, M. MicroRNA-21 and MicroRNA-29a Modulate the Expression of Collagen in Dermal Fibroblasts of Patients with Systemic Sclerosis. Autoimmunity 2019, 52, 108–116. [Google Scholar] [CrossRef]
  60. Yan, Y.M.; Zheng, J.N.; Wu, L.W.; Rao, Q.W.; Yang, Q.R.; Gao, D.; Wang, Q. Prediction of a Competing Endogenous RNA Co-Expression Network by Comprehensive Methods in Systemic Sclerosis-Related Interstitial Lung Disease. Front. Genet. 2021, 12, 633059. [Google Scholar] [CrossRef]
  61. Zhou, B.; Zhu, H.; Luo, H.; Gao, S.; Dai, X.; Li, Y.; Zuo, X. MicroRNA-202-3p Regulates Scleroderma Fibrosis by Targeting Matrix Metalloproteinase 1. Biomed. Pharmacother. 2017, 87, 412–418. [Google Scholar] [CrossRef]
  62. Luo, H.; Zhu, H.; Zhou, B.; Xiao, X.; Zuo, X. MicroRNA-130b Regulates Scleroderma Fibrosis by Targeting Peroxisome Proliferator-Activated Receptor γ. Mod. Rheumatol. 2015, 25, 595–602. [Google Scholar] [CrossRef]
  63. Li, Y.; Huang, J.; Hu, C.; Zhou, J.; Xu, D.; Hou, Y.; Wu, C.; Zhao, J.; Li, M.; Zeng, X.; et al. MicroRNA-320a: An Important Regulator in the Fibrotic Process in Interstitial Lung Disease of Systemic Sclerosis. Arthritis Res. Ther. 2021, 23, 21. [Google Scholar] [CrossRef]
  64. Shi, X.; Liu, Q.; Li, N.; Tu, W.; Luo, R.; Mei, X.; Ma, Y.; Xu, W.; Chu, H.; Jiang, S.; et al. MiR-3606-3p Inhibits Systemic Sclerosis through Targeting TGF-β Type II Receptor. Cell Cycle 2018, 17, 1967–1978. [Google Scholar] [CrossRef]
  65. Mendoza, F.A.; Piera-Velazquez, S.; Farber, J.L.; Feghali-Bostwick, C.; Jiménez, S.A. Endothelial Cells Expressing Endothelial and Mesenchymal Cell Gene Products in Lung Tissue from Patients with Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol. 2016, 68, 210–217. [Google Scholar] [CrossRef]
  66. Yan, Q.; Chen, J.; Li, W.; Bao, C.; Fu, Q. Targeting MiR-155 to Treat Experimental Scleroderma. Sci. Rep. 2016, 6, 20314, Erratum in Sci. Rep. 2016, 6, 21941. [Google Scholar] [CrossRef]
  67. Artlett, C.M.; Sassi-Gaha, S.; Hope, J.L.; Feghali-Bostwick, C.A.; Katsikis, P.D. Mir-155 Is Overexpressed in Systemic Sclerosis Fibroblasts and Is Required for NLRP3 Inflammasome-Mediated Collagen Synthesis during Fibrosis. Arthritis Res. Ther. 2017, 19, 144. [Google Scholar] [CrossRef]
  68. Sun, Y.-H.; Xie, M.; Wu, S.-D.; Zhang, J.; Huang, C.-Z. Identification and Interaction Analysis of Key Genes and MicroRNAs in Systemic Sclerosis by Bioinformatics Approaches. Curr. Med. Sci. 2019, 39, 645–652. [Google Scholar] [CrossRef]
  69. Chouri, E.; Wang, M.; Hillen, M.R.; Angiolilli, C.; Silva-Cardoso, S.C.; Wichers, C.G.K.; van der Kroef, M.; Bekker, C.P.J.; Cossu, M.; van Bon, L.; et al. Implication of MiR-126 and MiR-139-5p in Plasmacytoid Dendritic Cell Dysregulation in Systemic Sclerosis. J. Clin. Med. 2021, 10, 491. [Google Scholar] [CrossRef]
  70. Rossato, M.; Affandi, A.J.; Thordardottir, S.; Wichers, C.G.K.; Cossu, M.; Broen, J.C.A.; Moret, F.M.; Bossini-Castillo, L.; Chouri, E.; van Bon, L.; et al. Association of MicroRNA-618 Expression With Altered Frequency and Activation of Plasmacytoid Dendritic Cells in Patients With Systemic Sclerosis. Arthritis Rheumatol. 2017, 69, 1891–1902. [Google Scholar] [CrossRef]
  71. Kawashita, Y.; Jinnin, M.; Makino, T.; Kajihara, I.; Makino, K.; Honda, N.; Masuguchi, S.; Fukushima, S.; Inoue, Y.; Ihn, H. Circulating MiR-29a Levels in Patients with Scleroderma Spectrum Disorder. J. Dermatol. Sci. 2011, 61, 67–69. [Google Scholar] [CrossRef]
  72. Etoh, M.; Jinnin, M.; Makino, K.; Yamane, K.; Nakayama, W.; Aoi, J.; Honda, N.; Kajihara, I.; Makino, T.; Fukushima, S.; et al. MicroRNA-7 downregulation Mediates Excessive Collagen Expression in Localized Scleroderma. Arch. Dermatol. Res. 2013, 305, 9–15. [Google Scholar] [CrossRef]
  73. Makino, T.; Jinnin, M.; Etoh, M.; Yamane, K.; Kajihara, I.; Makino, K.; Ichihara, A.; Igata, T.; Sakai, K.; Fukushima, S.; et al. Downregulation of MicroRNA-196a in the Sera and Involved Skin of Localized Scleroderma Patients. Eur. J. Dermatol. 2014, 24, 470–476. [Google Scholar] [CrossRef]
  74. Tanaka, S.; Suto, A.; Ikeda, K.; Sanayama, Y.; Nakagomi, D.; Iwamoto, T.; Suzuki, K.; Kambe, N.; Matsue, H.; Matsumura, R.; et al. Alteration of Circulating MiRNAs in SSc: MiR-30b Regulates the Expression of PDGF Receptor β. Rheumatology 2013, 52, 1963–1972. [Google Scholar] [CrossRef]
  75. Rusek, M.; Michalska-Jakubus, M.; Kowal, M.; Bełtowski, J.; Krasowska, D. A Novel MiRNA-4484 Is upregulated on Microarray and Associated with Increased MMP-21 Expression in Serum of Systemic Sclerosis Patients. Sci. Rep. 2019, 9, 14264. [Google Scholar] [CrossRef]
  76. Ciechomska, M.; Zarecki, P.; Merdas, M.; Swierkot, J.; Morgiel, E.; Wiland, P.; Maslinski, W.; Bogunia-Kubik, K. The Role of MicroRNA-5196 in the Pathogenesis of Systemic Sclerosis. Eur. J. Clin. Investig. 2017, 47, 555–564. [Google Scholar] [CrossRef]
  77. Sing, T.; Jinnin, M.; Yamane, K.; Honda, N.; Makino, K.; Kajihara, I.; Makino, T.; Sakai, K.; Masuguchi, S.; Fukushima, S.; et al. MicroRNA-92a Expression in the Sera and Dermal Fibroblasts Increases in Patients with Scleroderma. Rheumatology 2012, 51, 1550–1556. [Google Scholar] [CrossRef]
  78. Chouri, E.; Servaas, N.H.; Bekker, C.P.J.; Affandi, A.J.; Cossu, M.; Hillen, M.R.; Angiolilli, C.; Mertens, J.S.; van den Hoogen, L.L.; Silva-Cardoso, S.; et al. Serum MicroRNA Screening and Functional Studies Reveal MiR-483-5p as a Potential Driver of Fibrosis in Systemic Sclerosis. J. Autoimmun. 2018, 89, 162–170. [Google Scholar] [CrossRef]
  79. Wuttge, D.M.; Carlsen, A.L.; Teku, G.; Wildt, M.; Rådegran, G.; Vihinen, M.; Heegaard, N.H.H.; Hesselstrand, R. Circulating Plasma MicroRNAs in Systemic Sclerosis-Associated Pulmonary Arterial Hypertension. Rheumatology 2021, 61, 309–318. [Google Scholar] [CrossRef]
  80. Wajda, A.; Walczyk, M.; Dudek, E.; Stypińska, B.; Lewandowska, A.; Romanowska-Próchnicka, K.; Chojnowski, M.; Olesińska, M.; Paradowska-Gorycka, A. Serum MicroRNAs in Systemic Sclerosis, Associations with Digital Vasculopathy and Lung Involvement. Int. J. Mol. Sci. 2022, 23, 10731. [Google Scholar] [CrossRef]
  81. Dolcino, M.; Pelosi, A.; Fiore, P.F.; Patuzzo, G.; Tinazzi, E.; Lunardi, C.; Puccetti, A. Gene Profiling in Patients with Systemic Sclerosis Reveals the Presence of Oncogenic Gene Signatures. Front. Immunol. 2018, 9, 449. [Google Scholar] [CrossRef]
  82. Soffritti, I.; D’Accolti, M.; Maccari, C.; Bini, F.; Mazziga, E.; Arcangeletti, M.C.; Caselli, E. Coinfection of Dermal Fibroblasts by Human Cytomegalovirus and Human Herpesvirus 6 Can Boost the Expression of Fibrosis-Associated MicroRNAs. Microorganisms 2023, 11, 412. [Google Scholar] [CrossRef]
  83. Zhang, L.; Gao, X.; Qin, Z.; Shi, X.; Xu, K.; Wang, S.; Tang, M.; Wang, W.; Gao, S.; Zuo, L.; et al. USP15 Participates in DBP-Induced Testicular Oxidative Stress Injury through Regulating the Keap1/Nrf2 Signaling Pathway. Sci. Total Environ. 2021, 783, 146898. [Google Scholar] [CrossRef]
  84. Lv, R.; Du, L.; Zhou, F.; Yuan, X.; Liu, X.; Zhang, L. Rosmarinic Acid Alleviates Inflammation, Apoptosis, and Oxidative Stress through Regulating MiR-155-5p in a Mice Model of Parkinson’s Disease. ACS Chem. Neurosci. 2020, 11, 3259–3266. [Google Scholar] [CrossRef]
  85. Zhang, L.; Wu, H.; Zhao, M.; Lu, Q. Meta-Analysis of Differentially Expressed MicroRNAs in Systemic Sclerosis. Int. J. Rheum. Dis. 2020, 23, 1297–1304. [Google Scholar] [CrossRef]
  86. Lin, M.Y.; Chang, Y.C.; Wang, S.Y.; Yang, M.H.; Chang, C.H.; Hsiao, M.; Kitsis, R.N.; Lee, Y.J. Oncomir Mir-182-5p Enhances Radiosensitivity by Inhibiting the Radiation-Induced Antioxidant Effect through Sesn2 in Head and Neck Cancer. Antioxidants 2021, 10, 1808. [Google Scholar] [CrossRef]
  87. Li, Z.N.; Ge, M.X.; Yuan, Z.F. MicroRNA-182-5p Protects Human Lens Epithelial Cells against Oxidative Stress-Induced Apoptosis by Inhibiting NOX4 and P38 MAPK Signalling. BMC Ophthalmol. 2020, 20, 233. [Google Scholar] [CrossRef]
  88. Shen, Y.; Zhu, Y.; Rong, F. MiR-200c-3p Regulates the Proliferation and Apoptosis of Human Trabecular Meshwork Cells by Targeting PTEN. Mol. Med. Rep. 2020, 22, 1605–1612. [Google Scholar] [CrossRef]
  89. Jiang, Z.; Tao, J.H.; Zuo, T.; Li, X.M.; Wang, G.S.; Fang, X.; Xu, X.L.; Li, X.P. The Correlation between MiR-200c and the Severity of Interstitial Lung Disease Associated with Different Connective Tissue Diseases. Scand. J. Rheumatol. 2017, 46, 122–129. [Google Scholar] [CrossRef]
  90. Qi, H.; Zhang, J.; Shang, Y.; Yuan, S.; Meng, C. Argon Inhibits Reactive Oxygen Species Oxidative Stress via the MiR-21-Mediated PDCD4/PTEN Pathway to Prevent Myocardial Ischemia/Reperfusion Injury. Bioengineered 2021, 12, 5529–5539. [Google Scholar] [CrossRef]
  91. Ding, S.; Liu, D.; Wang, L.; Wang, G.; Zhu, Y. Inhibiting MicroRNA-29a Protects Myocardial Ischemia-Reperfusion Injury by Targeting SIRT1 and Suppressing Oxidative Stress and NLRP3-Mediated Pyroptosis Pathway. J. Pharmacol. Exp. Ther. 2020, 372, 128–135. [Google Scholar] [CrossRef]
  92. Xiao, L.; Luo, D.; Pi, P.; Hu, M.; Li, X.; Yin, Q. Upregulation of MiR-135b Expression Induced by Oxidative Stress Promotes the Apoptosis of Renal Tubular Epithelial Cells under High Glucose Condition. Clin. Exp. Pharmacol. Physiol. 2020, 47, 1410–1419. [Google Scholar] [CrossRef]
  93. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-Mediated Transfer of MRNAs and MicroRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  94. Turchinovich, A.; Weiz, L.; Langheinz, A.; Burwinkel, B. Characterization of Extracellular Circulating MicroRNA. Nucleic Acids Res. 2011, 39, 7223–7233. [Google Scholar] [CrossRef]
  95. Zhang, J.; Li, S.; Li, L.; Li, M.; Guo, C.; Yao, J.; Mi, S. Exosome and Exosomal MicroRNA: Trafficking, Sorting, and Function. Genom. Proteom. Bioinform. 2015, 13, 17–24. [Google Scholar] [CrossRef]
  96. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The MicroRNA Spectrum in 12 Body Fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef]
  97. Wang, R.N.; Green, J.; Wang, Z.; Deng, Y.; Qiao, M.; Peabody, M.; Zhang, Q.; Ye, J.; Yan, Z.; Denduluri, S.; et al. Bone Morphogenetic Protein (BMP) Signaling in Development and Human Diseases. Genes. Dis. 2014, 1, 87–105. [Google Scholar] [CrossRef]
  98. Roush, S.; Slack, F.J. The Let-7 Family of MicroRNAs. Trends Cell Biol. 2008, 18, 505–516. [Google Scholar] [CrossRef]
  99. Pasquinelli, A.E.; Reinhart, B.J.; Slack, F.; Martindale, M.Q.; Kuroda, M.I.; Maller, B.; Hayward, D.C.; Ball, E.E.; Degnan, B.; Müller, P.; et al. Conservation of the Sequence and Temporal Expression of Let-7 Heterochronic Regulatory RNA. Nature 2000, 408, 86–89. [Google Scholar] [CrossRef]
  100. Zhu, H.; Luo, H.; Li, Y.; Zhou, Y.; Jiang, Y.; Chai, J.; Xiao, X.; You, Y.; Zuo, X. MicroRNA-21 in Scleroderma Fibrosis and Its Function in TGF-β-Regulated Fibrosis-Related Genes Expression. J. Clin. Immunol. 2013, 33, 1100–1109. [Google Scholar] [CrossRef]
  101. Quinn, S.R.; O’Neill, L.A. A Trio of MicroRNAs That Control Toll-like Receptor Signalling. Int. Immunol. 2011, 23, 421–425. [Google Scholar] [CrossRef]
  102. Kozlova, A.; Pachera, E.; Maurer, B.; Jüngel, A.; Distler, J.H.W.; Kania, G.; Distler, O. Regulation of Fibroblast Apoptosis and Proliferation by MicroRNA-125b in Systemic Sclerosis. Arthritis Rheumatol. 2019, 71, 2068–2080. [Google Scholar] [CrossRef]
  103. Kubo, M.; Czuwara-Ladykowska, J.; Moussa, O.; Markiewicz, M.; Smith, E.; Silver, R.M.; Jablonska, S.; Blaszczyk, M.; Watson, D.K.; Trojanowska, M. Persistent Downregulation of Fli1, a Suppressor of Collagen Transcription, in Fibrotic Scleroderma Skin. Am. J. Pathol. 2003, 163, 571–581. [Google Scholar] [CrossRef]
  104. Gracias, D.T.; Stelekati, E.; Hope, J.L.; Boesteanu, A.C.; Doering, T.A.; Norton, J.; Mueller, Y.M.; Fraietta, J.A.; Wherry, E.J.; Turner, M.; et al. The MicroRNA MiR-155 Controls CD8+ T Cell Responses by Regulating Interferon Signaling. Nat. Immunol. 2013, 14, 593–602. [Google Scholar] [CrossRef]
  105. Thai, T.H.; Calado, D.P.; Casola, S.; Ansel, K.M.; Xiao, C.; Xue, Y.; Murphy, A.; Frendewey, D.; Valenzuela, D.; Kutok, J.L.; et al. Regulation of the Germinal Center Response by MicroRNA-155. Science 2007, 316, 604–608. [Google Scholar] [CrossRef]
  106. Chiang, C.Y.; Engel, A.; Opaluch, A.M.; Ramos, I.; Maestre, A.M.; Secundino, I.; De Jesus, P.D.; Nguyen, Q.T.; Welch, G.; Bonamy, G.M.C.; et al. Cofactors Required for TLR7- and TLR9-Dependent Innate Immune Responses. Cell Host Microbe 2012, 11, 306–318. [Google Scholar] [CrossRef]
  107. Li, Y.; Huang, J.; Guo, M.; Zuo, X. MicroRNAs Regulating Signaling Pathways: Potential Biomarkers in Systemic Sclerosis. Genom. Proteom. Bioinform. 2015, 13, 234–241. [Google Scholar] [CrossRef]
  108. Kajihara, I.; Jinnin, M.; Yamane, K.; Makino, T.; Honda, N.; Igata, T.; Masuguchi, S.; Fukushima, S.; Okamoto, Y.; Hasegawa, M.; et al. Increased Accumulation of Extracellular Thrombospondin-2 Due to Low Degradation Activity Stimulates Type I Collagen Expression in Scleroderma Fibroblasts. Am. J. Pathol. 2012, 180, 703–714. [Google Scholar] [CrossRef]
  109. Makino, K.; Jinnin, M.; Kajihara, I.; Honda, N.; Sakai, K.; Masuguchi, S.; Fukushima, S.; Inoue, Y.; Ihn, H. Circulating MiR-142-3p Levels in Patients with Systemic Sclerosis. Clin. Exp. Dermatol. 2012, 37, 34–39. [Google Scholar] [CrossRef]
  110. Steen, S.O.; Iversen, L.V.; Carlsen, A.L.; Burton, M.; Nielsen, C.T.; Jacobsen, S.; Heegaard, N.H.H. The Circulating Cell-Free MicroRNA Profile in Systemic Sclerosis Is Distinct from Both Healthy Controls and Systemic Lupus Erythematosus. J. Rheumatol. 2015, 42, 214–221. [Google Scholar] [CrossRef]
  111. Koba, S.; Jinnin, M.; Inoue, K.; Nakayama, W.; Honda, N.; Makino, K.; Kajihara, I.; Makino, T.; Fukushima, S.; Ihn, H. Expression Analysis of Multiple MicroRNAs in Each Patient with Scleroderma. Exp. Dermatol. 2013, 22, 489–491. [Google Scholar] [CrossRef]
  112. Wuttge, D.M.; Carlsen, A.L.; Teku, G.; Steen, S.O.; Wildt, M.; Vihinen, M.; Hesselstrand, R.; Heegaard, N.H.H. Specific Autoantibody Profiles and Disease Subgroups Correlate with Circulating Micro-RNA in Systemic Sclerosis. Rheumatology 2015, 54, 2100–2107. [Google Scholar] [CrossRef]
  113. Chen, X.; Liang, H.; Zhang, J.; Zen, K.; Zhang, C.Y. Secreted MicroRNAs: A New Form of Intercellular Communication. Trends Cell Biol. 2012, 22, 125–132. [Google Scholar] [CrossRef]
  114. Wermuth, P.J.; Piera-Velazquez, S.; Jimenez, S.A. Exosomes Isolated from Serum of Systemic Sclerosis Patients Display Alterations in Their Content of Profibrotic and Antifibrotic MicroRNA and Induce a Profibrotic Phenotype in Cultured Normal Dermal Fibroblasts. Clin. Exp. Rheumatol. 2017, 35, 21. [Google Scholar]
  115. Murdaca, G.; Contatore, M.; Gulli, R.; Mandich, P.; Puppo, F. Genetic Factors and Systemic Sclerosis. Autoimmun. Rev. 2016, 15, 427–432. [Google Scholar] [CrossRef]
  116. Arcangeletti, M.C.; Maccari, C.; Vescovini, R.; Volpi, R.; Giuggioli, D.; Sighinolfi, G.; De Conto, F.; Chezzi, C.; Calderaro, A.; Ferri, C. A Paradigmatic Interplay between Human Cytomegalovirus and Host Immune System: Possible Involvement of Viral Antigen-Driven CD8+ T Cell Responses in Systemic Sclerosis. Viruses 2018, 10, 508. [Google Scholar] [CrossRef]
  117. Broccolo, F.; Drago, F.; Cassina, G.; Fava, A.; Fusetti, L.; Matteoli, B.; Ceccherini-Nelli, L.; Sabbadini, M.G.; Lusso, P.; Parodi, A.; et al. Selective Reactivation of Human Herpesvirus 6 in Patients with Autoimmune Connective Tissue Diseases. J. Med. Virol. 2013, 85, 1925–1934. [Google Scholar] [CrossRef]
  118. Broccolo, F.; Drago, F.; Paolino, S.; Cassina, G.; Gatto, F.; Fusetti, L.; Matteoli, B.; Zaccaria, E.; Parodi, A.; Lusso, P.; et al. Reactivation of Human Herpesvirus 6 (HHV-6) Infection in Patients with Connective Tissue Diseases. J. Clin. Virol. 2009, 46, 43–46. [Google Scholar] [CrossRef]
  119. Álvarez-Lafuente, R.; Fernández-Gutiérrez, B.; De Miguel, S.; Jover, J.A.; Rollin, R.; Loza, E.; Clemente, D.; Lamas, J.R. Potential Relationship between Herpes Viruses and Rheumatoid Arthritis: Analysis with Quantitative Real Time Polymerase Chain Reaction. Ann. Rheum. Dis. 2005, 64, 1357–1359. [Google Scholar] [CrossRef]
  120. Alvarez-Lafuente, R.; Martinez, A.; Garcia-Montojo, M.; Mas, A.; De Las Heras, V.; Dominguez-Mozo, M.I.; Maria Del Carmen, C.; López-Cavanillas, M.; Bartolome, M.; Gomez De La Concha, E.; et al. MHC2TA Rs4774C and HHV-6A Active Replication in Multiple Sclerosis Patients. Eur. J. Neurol. 2010, 17, 129–135. [Google Scholar] [CrossRef]
  121. Ferri, C.; Cazzato, M.; Giuggioli, D.; Sebastiani, M.; Magro, C. Systemic Sclerosis Following Human Cytomegalovirus Infection. Ann. Rheum. Dis. 2002, 61, 937–938. [Google Scholar] [CrossRef]
  122. Lunardi, C.; Bason, C.; Navone, R.; Millo, E.; Damonte, G.; Corrocher, R.; Puccetti, A. Systemic Sclerosis Immunoglobulin G Autoantibodies Bind the Human Cytomegalovirus Late Protein UL94 and Induce Apoptosis in Human Endothelial Cells. Nat. Med. 2000, 6, 1183–1186. [Google Scholar] [CrossRef]
  123. Lunardi, C.; Dolcino, M.; Peterlana, D.; Bason, C.; Navone, R.; Tamassia, N.; Beri, R.; Corrocher, R.; Puccetti, A. Antibodies against Human Cytomegalovirus in the Pathogenesis of Systemic Sclerosis: A Gene Array Approach. PLoS Med. 2006, 3, e2. [Google Scholar] [CrossRef]
  124. Arnson, Y.; Amital, H.; Guiducci, S.; Matucci-Cerinic, M.; Valentini, G.; Barzilai, O.; Maya, R.; Shoenfeld, Y. The Role of Infections in the Immunopathogensis of Systemic Sclerosis-Evidence from Serological Studies. In Proceedings of the Annals of the New York Academy of Sciences. Ann. N. Y. Acad. Sci. 2009, 1173, 627–632. [Google Scholar] [CrossRef]
  125. Marou, E.; Liaskos, C.; Efthymiou, G.; Dardiotis, E.; Daponte, A.; Scheper, T.; Meyer, W.; Hadjigeorgiou, G.; Bogdanos, D.P.; Sakkas, L.I. Increased Immunoreactivity against Human Cytomegalovirus UL83 in Systemic Sclerosis. Clin. Exp. Rheumatol. 2017, 35, S31–S34. [Google Scholar]
  126. Efthymiou, G.; Dardiotis, E.; Liaskos, C.; Marou, E.; Scheper, T.; Meyer, W.; Daponte, A.; Daoussis, D.; Hadjigeorgiou, G.; Bogdanos, D.P.; et al. A Comprehensive Analysis of Antigen-Specific Antibody Responses against Human Cytomegalovirus in Patients with Systemic Sclerosis. Clin. Immunol. 2019, 207, 87–96. [Google Scholar] [CrossRef]
  127. Henry, T.W.; Mendoza, F.A.; Jimenez, S.A. Role of MicroRNA in the Pathogenesis of Systemic Sclerosis Tissue Fibrosis and Vasculopathy. Autoimmun. Rev. 2019, 18, 102396. [Google Scholar] [CrossRef]
  128. Peng, W.J.; Tao, J.H.; Mei, B.; Chen, B.; Li, B.Z.; Yang, G.J.; Zhang, Q.; Yao, H.; Wang, B.X.; He, Q.; et al. MicroRNA-29: A Potential Therapeutic Target for Systemic Sclerosis. Expert. Opin. Ther. Targets 2012, 16, 875–879. [Google Scholar] [CrossRef]
  129. Maurer, B.; Stanczyk, J.; Jüngel, A.; Akhmetshina, A.; Trenkmann, M.; Brock, M.; Kowal-Bielecka, O.; Gay, R.E.; Michel, B.A.; Distler, J.H.W.; et al. MicroRNA-29, a Key Regulator of Collagen Expression in Systemic Sclerosis. Arthritis Rheum. 2010, 62, 1733–1743. [Google Scholar] [CrossRef]
  130. Fu, X.; Qie, J.; Fu, Q.; Chen, J.; Jin, Y.; Ding, Z. MiR-20a-5p/TGFBR2 Axis Affects Pro-Inflammatory Macrophages and Aggravates Liver Fibrosis. Front. Oncol. 2020, 10, 511116. [Google Scholar] [CrossRef]
  131. Eissa, M.G.; Artlett, C.M. The MicroRNA MiR-155 Is Essential in Fibrosis. Noncoding RNA 2019, 5, 23. [Google Scholar] [CrossRef]
  132. Epstein, M.A.; Achong, B.G.; Barr, Y.M. VIRUS PARTICLES IN CULTURED LYMPHOBLASTS FROM BURKITT’S LYMPHOMA. Lancet 1964, 283, 702–703. [Google Scholar] [CrossRef]
  133. Iizasa, H.; Kim, H.; Kartika, A.V.; Kanehiro, Y.; Yoshiyama, H. Role of Viral and Host MicroRNAs in Immune Regulation of Epstein-Barr Virus-Associated Diseases. Front. Immunol. 2020, 11, 367. [Google Scholar] [CrossRef]
  134. Borghol, A.H.; Bitar, E.R.; Hanna, A.; Naim, G.; Rahal, E.A. The Role of Epstein-Barr Virus in Autoimmune and Autoinflammatory Diseases. Crit. Rev. Microbiol. 2024, 1–21. [Google Scholar] [CrossRef]
  135. Kim, H.; Iizasa, H.; Kanehiro, Y.; Fekadu, S.; Yoshiyama, H. Herpesviral MicroRNAs in Cellular Metabolism and Immune Responses. Front. Microbiol. 2017, 8, 274499. [Google Scholar] [CrossRef]
  136. Wang, M.; Yu, F.; Wu, W.; Wang, Y.; Ding, H.; Qian, L. Epstein-Barr Virus-Encoded MicroRNAs as Regulators in Host Immune Responses. Int. J. Biol. Sci. 2018, 14, 565–576. [Google Scholar] [CrossRef]
  137. Skalsky, R.L. MicroRNA-Mediated Control of Epstein-Barr Virus Infection and Potential Diagnostic and Therapeutic Implications. Curr. Opin. Virol. 2022, 56, 101272. [Google Scholar] [CrossRef]
  138. Sakamoto, N.; Yura, H.; Okuno, D.; Miyamura, T.; Hara, A.; Kido, T.; Ishimoto, H.; Ishimatsu, Y.; Mukae, H. The Role of Epstein-Barr Virus-Encoded MicroRNAs in Pulmonary Fibrosis. In ATS 2020 International Conference American Thoracic Society International Conference Meetings Abstracts; American Thoracic Society: New York, NY, USA, 2020; p. A2238. [Google Scholar] [CrossRef]
  139. Kerr, J.R. The Role of Parvovirus B19 in the Pathogenesis of Autoimmunity and Autoimmune Disease. J. Clin. Pathol. 2016, 69, 279–291. [Google Scholar] [CrossRef]
  140. Ferri, C.; Azzi, A.; Magro, C.M. Parvovirus B19 and Systemic Sclerosis. Br. J. Dermatol. 2005, 152, 819–820. [Google Scholar] [CrossRef]
  141. Lundqvist, A.; Isa, A.; Tolfvenstam, T.; Kvist, G.; Broliden, K. High Frequency of Parvovirus B19 DNA in Bone Marrow Samples from Rheumatic Patients. J. Clin. Virol. 2005, 33, 71–74. [Google Scholar] [CrossRef]
  142. Ferri, C.; Zakrzewska, K.; Longombardo, G.; Giuggioli, D.; Storino, F.A.A.; Pasero, G.; Azzi, A. Parvovirus B19 Infection of Bone Marrow in Systemic Sclerosis Patients. Clin. Exp. Rheumatol. 1999, 17, 718–720. [Google Scholar]
  143. Ohtsuka, T.; Yamazaki, S. Increased Prevalence of Human Parvovirus B19 DNA in Systemic Sclerosis Skin. Br. J. Dermatol. 2004, 150, 1091–1095. [Google Scholar] [CrossRef]
  144. Zakrzewska, K.; Corcioli, F.; Carlsen, K.M.; Giuggioli, D.; Fanci, R.; Rinieri, A.; Ferri, C.; Azzi, A. Human Parvovirus B19 (B19V) Infection in Systemic Sclerosis Patients. Intervirology 2009, 52, 279–282. [Google Scholar] [CrossRef]
  145. Zakrzewska, K.; Cortivo, R.; Tonello, C.; Panfilo, S.; Abatangelo, G.; Giuggioli, D.; Ferri, C.; Corcioli, F.; Azzi, A. Human Parvovirus B19 Experimental Infection in Human Fibroblasts and Endothelial Cells Cultures. Virus Res. 2005, 114, 1–5. [Google Scholar] [CrossRef]
  146. Schmidt-Lucke, C.; Zobel, T.; Schrepfer, S.; Kuhl, U.; Wang, D.; Klingel, K.; Becher, P.M.; Fechner, H.; Pozzuto, T.; Van Linthout, S.; et al. Impaired Endothelial Regeneration Through Human Parvovirus B19–Infected Circulating Angiogenic Cells in Patients With Cardiomyopathy. J. Infect. Dis. 2015, 212, 1070–1081. [Google Scholar] [CrossRef]
  147. Berillo, O.; Khailenko, V.; Ivashchenko, A.; Perlmuter-Shoshany, L.; Bolshoy, A. MiRNA and Tropism of Human Parvovirus B19. Comput. Biol. Chem. 2012, 40, 1–6. [Google Scholar] [CrossRef]
  148. Anbarlou, A.; AkhavanRahnama, M.; Atashi, A.; Soleimani, M.; Arefian, E.; Gallinella, G. Possible Involvement of MiRNAs in Tropism of Parvovirus B19. Mol. Biol. Rep. 2016, 43, 175–181. [Google Scholar] [CrossRef]
  149. Gain, C.; Song, S.; Angtuaco, T.; Satta, S.; Kelesidis, T. The Role of Oxidative Stress in the Pathogenesis of Infections with Coronaviruses. Front. Microbiol. 2023, 13, 1111930. [Google Scholar] [CrossRef]
  150. Kavouras, J.; Prandovszky, E.; Valyi-Nagy, K.; Kovacs, S.K.; Tiwari, V.; Kovacs, M.; Shukla, D.; Valyi-Nagy, T. Herpes Simplex Virus Type 1 Infection Induces Oxidative Stress and the Release of Bioactive Lipid Peroxidation By-Products in Mouse P19N Neural Cell Cultures. J. Neurovirol. 2007, 13, 416–425. [Google Scholar] [CrossRef]
  151. Schwarz, K.B. Oxidative Stress during Viral Infection: A Review. Free Radic. Biol. Med. 1996, 21, 641–649. [Google Scholar] [CrossRef]
  152. Speir, E.; Shibutani, T.; Yu, Z.X.; Ferrans, V.; Epstein, S.E. Role of Reactive Oxygen Intermediates in Cytomegalovirus Gene Expression and in the Response of Human Smooth Muscle Cells to Viral Infection. Circ. Res. 1996, 79, 1143–1152. [Google Scholar] [CrossRef]
  153. Prusty, B.K.; Böhme, L.; Bergmann, B.; Siegl, C.; Krause, E.; Mehlitz, A.; Rudel, T. Imbalanced Oxidative Stress Causes Chlamydial Persistence during Non-Productive Human Herpes Virus Co-Infection. PLoS ONE 2012, 7, e47427. [Google Scholar] [CrossRef]
  154. De Francesco, M.A. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses 2024, 16, 133. [Google Scholar] [CrossRef]
  155. Sebastiano, M.; Chastel, O.; De Thoisy, B.; Eens, M.; Costantini, D. Oxidative Stress Favours Herpes Virus Infection in Vertebrates: A Meta-Analysis. Curr. Zool. 2016, 62, 325–332. [Google Scholar] [CrossRef]
  156. Doridot, L.; Jeljeli, M.; Chêne, C.; Batteux, F. Implication of Oxidative Stress in the Pathogenesis of Systemic Sclerosis via Inflammation, Autoimmunity and Fibrosis. Redox Biol. 2019, 25, 101122. [Google Scholar] [CrossRef]
  157. Ames, P.R.J.; Bucci, T.; Merashli, M.; Amaral, M.; Arcaro, A.; Gentile, F.; Nourooz-Zadeh, J.; DelgadoAlves, J. Oxidative/Nitrative Stress in the Pathogenesis of Systemic Sclerosis: Are Antioxidants Beneficial? Free Radic. Res. 2018, 52, 1063–1082. [Google Scholar] [CrossRef]
  158. Vona, R.; Giovannetti, A.; Gambardella, L.; Malorni, W.; Pietraforte, D.; Straface, E. Oxidative Stress in the Pathogenesis of Systemic Scleroderma: An Overview. J. Cell Mol. Med. 2018, 22, 3308–3314. [Google Scholar] [CrossRef]
  159. Piera-velazquez, S.; Jimenez, S.A. Oxidative Stress Induced by Reactive Oxygen Species (ROS) and NADPH Oxidase 4 (NOX4) in the Pathogenesis of the Fibrotic Process in Systemic Sclerosis: A Promising Therapeutic Target. J. Clin. Med. 2021, 10, 4791. [Google Scholar] [CrossRef]
  160. Luo, J.Y.; Liu, X.; Jiang, M.; Zhao, H.P.; Zhao, J.J. Oxidative Stress Markers in Blood in Systemic Sclerosis: A Meta-Analysis. Mod. Rheumatol. 2017, 27, 306–314. [Google Scholar] [CrossRef]
  161. Dooley, A.; Gao, B.; Bradley, N.; Abraham, D.J.; Black, C.M.; Jacobs, M.; Bruckdorfer, K.R. Abnormal Nitric Oxide Metabolism in Systemic Sclerosis: Increased Levels of Nitrated Proteins and Asymmetric Dimethylarginine. Rheumatology 2006, 45, 676–684. [Google Scholar] [CrossRef]
  162. Ogawa, F.; Shimizu, K.; Muroi, E.; Hara, T.; Sato, S. Increasing Levels of Serum Antioxidant Status, Total Antioxidant Power, in Systemic Sclerosis. Clin. Rheumatol. 2011, 30, 921–925. [Google Scholar] [CrossRef]
  163. Volpe, A.; Biasi, D.; Caramaschi, P.; Mantovani, W.; Bambara, L.M.; Canestrini, S.; Ferrari, M.; Poli, G.; Degan, M.; Carletto, A.; et al. Levels of F2-Isoprostanes in Systemic Sclerosis: Correlation with Clinical Features. Rheumatology 2006, 45, 314–320. [Google Scholar] [CrossRef]
  164. Avouac, J.; Borderie, D.; Ekindjian, O.G.; Kahan, A.; Allanore, Y. High DNA Oxidative Damage in Systemic Sclerosis. J. Rheumatol. 2010, 37, 2540–2547. [Google Scholar] [CrossRef]
  165. Bourji, K.; Meyer, A.; Chatelus, E.; Pincemail, J.; Pigatto, E.; Defraigne, J.O.; Singh, F.; Charlier, C.; Geny, B.; Gottenberg, J.E.; et al. High Reactive Oxygen Species in Fibrotic and Nonfibrotic Skin of Patients with Diffuse Cutaneous Systemic Sclerosis. Free Radic. Biol. Med. 2015, 87, 282–289. [Google Scholar] [CrossRef]
  166. Aden, N.; Shiwen, X.; Aden, D.; Black, C.; Nuttall, A.; Denton, C.P.; Leask, A.; Abraham, D.; Stratton, R. Proteomic Analysis of Scleroderma Lesional Skin Reveals Activated Wound Healing Phenotype of Epidermal Cell Layer. Rheumatology 2008, 47, 1754–1760. [Google Scholar] [CrossRef]
  167. Sambo, P.; Baroni, S.S.; Luchetti, M.; Paroncini, P.; Dusi, S.; Orlandini, G.; Gabrielli, A.; Svegliati Baroni, S. Oxidative Stress in Scleroderma Maintenance of Scleroderma Fibroblast Phenotype by the Constitutive Upregulation of Reactive Oxygen Species Generation Through the NADPH Oxidase Complex Pathway. Arthritis Rheum. 2001, 44, 2653–2664. [Google Scholar] [CrossRef]
  168. Svegliati, S.; Spadoni, T.; Moroncini, G.; Gabrielli, A. NADPH Oxidase, Oxidative Stress and Fibrosis in Systemic Sclerosis. Free Radic. Biol. Med. 2018, 125, 90–97. [Google Scholar] [CrossRef]
  169. Balbir-Gurman, A.; Braun-Moscovici, Y.; Livshitz, V.; Schapira, D.; Markovits, D.; Rozin, A.; Boikaner, T.; Nahir, A.M. Antioxidant Status after Iloprost Treatment in Patients with Raynaud’s Phenomenon Secondary to Systemic Sclerosis. Clin. Rheumatol. 2007, 26, 1517–1521. [Google Scholar] [CrossRef]
  170. Musellim, B.; Ikitimur, H.; Uzun, H.; Ongen, G. The Oxidant-Antioxidant Balance in Systemic Sclerosis Cases with Interstitial Lung Involvement. Rheumatol. Int. 2006, 27, 163–167. [Google Scholar] [CrossRef]
  171. Sfrent-Cornateanu, R.; Mihai, C.; Stoian, I.; Lixandru, D.; Bara, C.; Moldoveanu, E. Antioxidant Defense Capacity in Scleroderma Patients. Clin. Chem. Lab. Med. 2008, 46, 836–841. [Google Scholar] [CrossRef]
  172. Wang, Y.; Zhao, R.; Liu, W.; Wang, Z.; Rong, J.; Long, X.; Liu, Z.; Ge, J.; Shi, B. Exosomal CircHIPK3 Released from Hypoxia-Pretreated Cardiomyocytes Regulates Oxidative Damage in Cardiac Microvascular Endothelial Cells via the MiR-29a/IGF-1 Pathway. Oxid. Med. Cell Longev. 2019, 2019. [Google Scholar] [CrossRef]
  173. Markopoulos, G.S.; Roupakia, E.; Tokamani, M.; Alabasi, G.; Sandaltzopoulos, R.; Marcu, K.B.; Kolettas, E. Roles of NF-ΚB Signaling in the Regulation of MiRNAs Impacting on Inflammation in Cancer. Biomedicines 2018, 6, 40. [Google Scholar] [CrossRef]
  174. Chen, Y.; Liu, Y.; Pan, Q.; Zhao, Y.; He, C.; Bi, K.; Chen, Y.; Zhao, B.; Ma, X. MicroRNA-155 Regulates ROS Production, NO Generation, Apoptosis and Multiple Functions of Human Brain Microvessel Endothelial Cells Under Physiological and Pathological Conditions. J. Cell Biochem. 2015, 116, 2870–2881. [Google Scholar] [CrossRef]
  175. Li, N.; You, X.; Chen, T.; Mackowiak, S.D.; Friedländer, M.R.; Weigt, M.; Du, H.; Gogol-Döring, A.; Chang, Z.; Dieterich, C.; et al. Global Profiling of MiRNAs and the Hairpin Precursors: Insights into MiRNA Processing and Novel MiRNA Discovery. Nucleic Acids Res. 2013, 41, 3619–3634. [Google Scholar] [CrossRef]
  176. Creighton, C.J.; Nagaraja, A.K.; Hanash, S.M.; Matzuk, M.M.; Gunaratne, P.H. A Bioinformatics Tool for Linking Gene Expression Profiling Results with Public Databases of MicroRNA Target Predictions. RNA 2008, 14, 2290–2296. [Google Scholar] [CrossRef]
  177. Licursi, V.; Conte, F.; Fiscon, G.; Paci, P. MIENTURNET: An Interactive Web Tool for MicroRNA-Target Enrichment and Network-Based Analysis. BMC Bioinform. 2019, 20, 545. [Google Scholar] [CrossRef]
  178. Jin, J.; Liu, Y.; Tang, Q.; Yan, X.; Jiang, M.; Zhao, X.; Chen, J.; Jin, C.; Ou, Q.; Zhao, J. Bioinformatics-Integrated Screening of Systemic Sclerosis-Specific Expressed Markers to Identify Therapeutic Targets. Front. Immunol. 2023, 14, 1125183. [Google Scholar] [CrossRef]
  179. Hu, Y.; Shen, F.; Yang, X.; Han, T.; Long, Z.; Wen, J.; Huang, J.; Shen, J.; Guo, Q. Single-Cell Sequencing Technology Applied to Epigenetics for the Study of Tumor Heterogeneity. Clin. Epigenetics 2023, 15, 161. [Google Scholar] [CrossRef]
  180. Bheda, P.; Schneider, R. Epigenetics Reloaded: The Single-Cell Revolution. Trends Cell Biol. 2014, 24, 712–723. [Google Scholar] [CrossRef]
  181. Diener, C.; Keller, A.; Meese, E. Emerging Concepts of MiRNA Therapeutics: From Cells to Clinic. Trends Genet. 2022, 38, 613–626. [Google Scholar] [CrossRef]
  182. Jin, F.; Hu, H.; Xu, M.; Zhan, S.; Wang, Y.; Zhang, H.; Chen, X. Serum MicroRNA Profiles Serve as Novel Biomarkers for Autoimmune Diseases. Front. Immunol. 2018, 9, 2381. [Google Scholar] [CrossRef]
  183. Szabo, I.; Muntean, L.; Crisan, T.; Rednic, V.; Sirbe, C.; Rednic, S. Novel Concepts in Systemic Sclerosis Pathogenesis: Role for MiRNAs. Biomedicines 2021, 9, 1471. [Google Scholar] [CrossRef]
  184. Marabita, F.; De Candia, P.; Torri, A.; Tegnér, J.; Abrignani, S.; Rossi, R.L. Normalization of Circulating MicroRNA Expression Data Obtained by Quantitative Real-Time RT-PCR. Brief. Bioinform. 2016, 17, 204–212. [Google Scholar] [CrossRef]
  185. Pritchard, C.C.; Cheng, H.H.; Tewari, M. MicroRNA Profiling: Approaches and Considerations. Nat. Rev. Genet. 2012, 13, 358–369. [Google Scholar] [CrossRef]
Figure 1. Profile of miRNAs deregulated in SSc disease, both at the tissue and extracellular level, and effects of oxidative stress and beta-herpesviruses infection on SSc-associated miRNAs [42,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92] (SSc, Systemic Sclerosis; upward red arrows, up-regulated miRNAs; downward blue arrows, down-regulated miRNAs).
Figure 1. Profile of miRNAs deregulated in SSc disease, both at the tissue and extracellular level, and effects of oxidative stress and beta-herpesviruses infection on SSc-associated miRNAs [42,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92] (SSc, Systemic Sclerosis; upward red arrows, up-regulated miRNAs; downward blue arrows, down-regulated miRNAs).
Biomedicines 12 01360 g001
Table 1. MiRNAs deregulated in SSc disease and potential beta-herpesvirus involvement: expression levels, localization, and functional role.
Table 1. MiRNAs deregulated in SSc disease and potential beta-herpesvirus involvement: expression levels, localization, and functional role.
miRNATissue/Cell TypeEffectExpression
(SSc vs. CTR)
Refs. No.
miR-7 *LSc dermal
fibroblasts, serum
Induces overexpression of α2(I) collagenup/down[72]
miR-10a *Skin biopsy,
fibroblasts
Decreases in EC cells stimulated with SSc serumdown[55]
miR-20 *Dermal fibroblastsActivated TGF-β pathway toward ECM productiondown[130]
miR-21Skin biopsy, lung and dermal fibroblasts, mouse model, plasmaEnhances TGF-β signaling events in SSc fibrosis
targeting SMAD7
Role in immune activation
up[58,79,100,101]
miR-26bFibroblastPromotes cell fibrosis markers expression targeting CXCL9 and CXCL13up[68]
miR-27aLung and skin biopsyPromotes fibrotic gene expression by SPP1/ERK1/2 axisdown[56]
miR-29aSerumDysregulates type I collagen deposition, targeting COL1A1 and COL1Adown[71]
miR-29b *Skin biopsy,
fibroblasts
Regulates TGFβ pathway targeting COL1A1down[58]
miR-30b *Skin biopsy, mouse model, serumPromotes fibrosis by regulating PDGFR-βdown[74]
miR-92a *Dermal fibroblasts,
serum
Decreases MMP-1 expression and control excessive
collagen accumulation
up[77]
miR-125b *Skin biopsy,
fibroblasts
Induces apoptosis, hindered proliferation and α-SMA
expression
down[102]
miR-126pDCsRegulates pDC activation via TLR9-mediated response and IFN signalingup[69]
miR-130Skin biopsy,
fibroblasts
Promotes TGF-β pathway via antifibrotic PPARγ
inhibition
up[62]
miR-135bFibroblast, serumRegulates USP15, involved in Keap1/Nrf2 antioxidative pathwaydown[83]
miR-139pDCsRegulates pDC activation via TLR9-mediated response and IFN signalingup[69]
miR-142Serum, plasmaRegulates the expression of integrin αVup in serum/down in plasma[109,110]
miR-145Skin biopsy,
fibroblasts
Regulates TGF-β pathway targeting SMAD3 down[58]
miR-146b *Skin biopsy,
fibroblasts
Increases in EC cells stimulated with SSc serumup[55]
miR-150Skin biopsy,
fibroblasts, serum
Induces overexpression of type I collagen and integrin β3 by TGF-β activationdown[57]
miR-155 *Skin biopsy, lung
fibroblast, mouse model, serum
Regulates of Wnt/β- catenin and Akt pathways
Enhances inflammation, apoptosis, and oxidative stress
Potential biomarker for vasculopathy
up[66,67,80,81,84,131]
miR-182Dermal fibroblastRegulates oxidative stress with bivalent effect up[85,86,87]
miR-193b *Skin biopsy,
fibroblasts
Regulates proliferative vasculopathy by uPA
overexpression
down[51]
miR-196aLSc skin biopsy,
serum
Induces overexpression of type I collagendown[73]
miR-200cPBMCInhibits ROS oxidative stress via PDCD4/PTENup[88]
miR-202Skin biopsy,
fibroblasts
Promotes fibrosis by regulation of MMP1 expressionup[61]
miR-206SerumCould discriminate SSc from ctr sera, together with miR-21down[111]
miR-320aPulmonary
fibroblasts, mouse model
Inhibits collagen deposition targeting TGFBR2 and IGF1Rdown[63]
miR-483Fibroblasts,
endothelial cells,
serum
Promotes collagen IV-encoding genes COL4A1- COL4A2, and transition of ECs to a myofibroblast phenotypeup[65,78]
miR-618pDCsInhibits pDCs development and induces IFNα secretionup[70]
miR-3606Skin biopsy,
fibroblasts
Hinders TGF-β pathway targeting TGFβ-receptor TGFBR2down[64]
miR-4484SerumInvolved in TGF-β signaling pathway, ECM-receptor
interaction, and MMP expression
up[75]
miR-5196SerumRegulates fibrogenesis targeting FRA2 geneup[76]
let-7 familySkin biopsyInvolved in pulmonary hypertension in SScdown[53]
let-7a *Skin biopsy, serumInduces excessive expression of type I collagendown[52]
let-7g *Skin biopsyCorrelates with SSc pathogenesisup[54]
miR-223
miR-342
miR-181b
miR-184
SerumThis miRNAs cluster could discriminate dSSc
from lSSc sera
down
(dSSc vs. lSSc) up
dSSc vs. lSSc)
[112]
miR-17
miR-20a
miR-92a
miR-106a
SerumThis miRNAs cluster could discriminate SSc
from ctr sera
down[110]
miR-142
miR-223
miR-150
miR-638
SerumThis miRNAs cluster could discriminate SSc
from SLE sera
down
(SSc vs. SLE)
up
(SSc vs. SLE)
[110]
let-7g
miR-17
miR-23b
miR-155
miR-215
miR-503
Serum exosomesprofibrotic miRNAs
SSc exosomes induce a profibrotic phenotype in cultured dermal fibroblasts
up[114]
let-7a
miR-26b
miR-29b
miR-92a
miR-129
miR-133
miR-140
miR-146a
miR-196a
miR-223
Serum exosomesantifibrotic miRNAs
SSc exosomes induce a profibrotic phenotype in cultured dermal fibroblasts
down[114]
* Asterisks indicate miRNAs affected by HHV-6A and/or HCMV infection [42,82] (see Figure 1).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Soffritti, I.; D’Accolti, M.; Bini, F.; Mazziga, E.; Di Luca, D.; Maccari, C.; Arcangeletti, M.-C.; Caselli, E. Virus-Induced MicroRNA Modulation and Systemic Sclerosis Disease. Biomedicines 2024, 12, 1360. https://doi.org/10.3390/biomedicines12061360

AMA Style

Soffritti I, D’Accolti M, Bini F, Mazziga E, Di Luca D, Maccari C, Arcangeletti M-C, Caselli E. Virus-Induced MicroRNA Modulation and Systemic Sclerosis Disease. Biomedicines. 2024; 12(6):1360. https://doi.org/10.3390/biomedicines12061360

Chicago/Turabian Style

Soffritti, Irene, Maria D’Accolti, Francesca Bini, Eleonora Mazziga, Dario Di Luca, Clara Maccari, Maria-Cristina Arcangeletti, and Elisabetta Caselli. 2024. "Virus-Induced MicroRNA Modulation and Systemic Sclerosis Disease" Biomedicines 12, no. 6: 1360. https://doi.org/10.3390/biomedicines12061360

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