Previous Article in Journal
Synergistic Activity of Cefiderocol in Combination with Avibactam, Sulbactam or Tazobactam against Carbapenem-Resistant Gram-Negative Bacteria
Previous Article in Special Issue
Alpha Synuclein Toxicity and Non-Motor Parkinson’s
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

circRNAs as Epigenetic Regulators of Integrity in Blood–Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis

1
Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
2
Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
3
Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
4
Research Department, King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
5
University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
6
LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
7
Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(16), 1316; https://doi.org/10.3390/cells13161316 (registering DOI)
Submission received: 3 July 2024 / Revised: 30 July 2024 / Accepted: 3 August 2024 / Published: 6 August 2024
(This article belongs to the Special Issue Molecular Insights into Neurodegenerative Diseases)

Abstract

:
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease leading to progressive demyelination and neuronal loss, with extensive neurological symptoms. As one of the most widespread neurodegenerative disorders, with an age onset of about 30 years, it turns out to be a socio-health and economic issue, thus necessitating therapeutic interventions currently unavailable. Loss of integrity in the blood–brain barrier (BBB) is one of the distinct MS hallmarks. Brain homeostasis is ensured by an endothelial cell-based monolayer at the interface between the central nervous system (CNS) and systemic bloodstream, acting as a selective barrier. MS results in enhanced barrier permeability, mainly due to the breakdown of tight (TJs) and adherens junctions (AJs) between endothelial cells. Specifically, proinflammatory mediator release causes failure in cytoplasmic exposure of junctions, resulting in compromised BBB integrity that enables blood cells to cross the barrier, establishing iron deposition and neuronal impairment. Cells with a compromised cytoskeletal protein network, fiber reorganization, and discontinuous junction structure can occur, resulting in BBB dysfunction. Recent investigations on spatial transcriptomics have proven circularRNAs (circRNAs) to be powerful multi-functional molecules able to epigenetically regulate transcription and structurally support proteins. In the present review, we provide an overview of the recent role ascribed to circRNAs in maintaining BBB integrity/permeability via cytoskeletal stability. Increased knowledge of the mechanisms responsible for impairment and circRNA’s role in driving BBB damage and dysfunction might be helpful for the recognition of novel therapeutic targets to overcome BBB damage and unrestrained neurodegeneration.

1. Introduction

Multiple sclerosis (MS) is a chronic progressive inflammatory demyelinating disorder of the central nervous system (CNS), leading to severe physical and cognitive disabilities as well as neurological defects [1,2,3,4]. Cognitive impairment is characterized by high variability between patients, including deficits in learning, memory, executive functions, and visuospatial processing [5]. With an age onset of about 30 years, MS is the most common neurodegenerative disease afflicting young adults [6,7]. This condition affects females more than males (sex ratio 2.5:1), with a prevalence of 120 per 100,000 individuals, depending on geographic area [8]. MS etiology remains elusive; however, it can be considered a complex multifactorial disease due to genetic predisposition in combination with hormonal and environmental factors [2,9]. Altogether, these risk factors trigger demyelinating plaques, responsible for the infiltration of proinflammatory T-lymphocytes and macrophages into the CNS across an ineffective blood–brain barrier (BBB), which results in neuronal damage and cognitive impairment [10,11].
One of the notable aspects of MS is the disruption of the BBB, a selective and dynamic endothelial cell monolayer at the interface between the vasculature and the CNS, which ensures the interflow of biological molecules and protects the brain from toxins, microorganisms, and other insults [12]. The BBB physiological functions are ensured by the presence of tight junctions (TJs) and adherens junctions (AJs) between endothelial cells, supported and controlled by cytoskeletal proteins (actin microfilaments, microtubules, and intermediate filaments [12,13]), which preserve homeostasis within the brain microenvironment by tightly regulating paracellular permeability [14].
The rearrangement and destruction of cytoskeletal architecture and cell junctions [12,15], together with the over-activation of the immune system [16], result in BBB integrity loss and subsequent increase in permeability [17], responsible for the initiation and propagation of MS active lesions. Therefore, the opportunity to act on cytoskeletal proteins and fine-tune their levels, restoring the highly complex and regulated organization and their functions, may limit BBB hyperpermeability in MS, as well as other neurodegenerative disorders such as amyotrophic lateral sclerosis, Alzheimer disease, and epilepsy [18,19,20].
In this regard, the main purpose of this review is to provide an overview of current knowledge of circularRNA (circRNA) function and their involvement in cytoskeleton regulation and stability. In fact, over the last decades, exponential advances in transcriptomics have allowed new discoveries in the multilayer regulatory role of circRNAs, a rising class of non-coding RNAs with unique characteristics and several biological functions [21]. CircRNAs, by acting directly on AJs and TJs or by indirectly regulating upstream signal pathways and transcription factors [22,23], could be considered promising drug targets to restore BBB architecture. This could aid in designing novel potential therapeutic strategies for MS in an attempt to improve the life quality and expectancy in MS patients [6,24].

2. Blood–Brain Barrier (BBB) Structure

A healthy brain is protected by the BBB, a very specialized and selective barrier system formed by a monolayer of brain microvascular endothelial cells (BMECs), which line cerebral blood flow, as well as brain microvascular pericytes, their basal lamina, and perivascular astrocytes [25,26] (Figure 1). The barrier controls the entry and the outflow of biological molecules essential for brain metabolism and neuronal functions [12,27]. As a unique part of the neurovascular unit (NVU), the BBB has a crucial role in maintaining brain homeostasis [11] and in protecting the CNS from toxins and pathogen entry [28]. Hence, functional and structural BBB integrity is pivotal to ensure and preserve the brain microenvironment, mediating CNS communication with the rest of the body [11,12].
BMECs are specialized neuronal endothelial cells that strictly limit the movement of substances from blood to CNS, reducing the paracellular flow of hydrophilic molecules via the formation of large numbers of TJs and AJs with adjacent BMECs [12,26] (Figure 1). TJs are localized along the lateral cell membrane to completely block the cleft between cells. Structurally, the complex is composed of transmembrane adhesion proteins, which physically interact with their counterparts on the membranes of adjacent cells and simultaneously connect to the intracellular actin cytoskeleton [14,29]. The TJ transmembrane proteins include the integral membrane proteins (e.g., occludins; claudins; ZO-1), an IgG protein type, and the junctional adhesion molecule (JAM) [14]. Claudins are the main sealing components of TJs, among which claudin-5, the principal constitutive claudin at the BBB [30,31], has the function of limiting paracellular diffusion of molecules larger than 400 Da across the BBB [31,32]. Occludin interacts with scaffold cytoskeleton proteins, contributing to TJ stabilization [33,34]. In fact, in occludin-deficient mice, neurological functions are compromised due to the hyperpermeability of BBB [35]. JAMs interact with cytoskeleton proteins to recruit and assemble signaling complexes, as key regulators of barrier function. One of the roles of JAMs in the TJs of endothelial cells is the regulation of the transendothelial migration of leukocytes to inflammation sites [36].
AJs have a similar organization to TJs. They are formed by the transmembrane proteins, cadherins, responsible for the adhesion of BMECs to cytoskeletal proteins, and catenins, Ca2+-dependent transmembrane proteins involved in supporting cadherin association and regulating out-in signaling processes. V-cadherin is the major transmembrane AJ protein of the BBB [37]. Specifically, catenins are important for the adhesive properties of V-cadherin and for binding actin, controlling BBB leakiness [14].
The other BBB cell types support the barrier structure and enhance its functionality. In detail, microvascular pericytes are essential constituents of brain capillaries and share a basal lamina with BMECs [38,39]. Perivascular astrocytes, as constituents of neuroglia, represent the most abundant cells in the CNS and have several functions, including compartmentalization of the neural parenchyma, ionic homeostatic maintenance of the extracellular space, neurotransmitter uptake, and signaling from neurons to the vasculature [40]. Astrocytes play a critical role in BBB formation and its functional maintenance by providing secreted factors that lead to adequate association between cells of the BBB, and to the formation of strong TJs [41].

BMEC Cytoskeleton

As previously reported, BMECs play multiple roles, including maintaining a flat endothelial morphology and retaining appropriate barrier activity through TJs and AJs [27]. Cytoskeletal proteins are required for all these functions. Similar to other cells, the cytoskeleton of BMECs consists of three filamentous proteins in constant communication with each other: actin microfilaments, microtubules, and intermediate filaments [12,13].
Microfilament proteins are found in endothelial cells as microfilamentous actin (F-Actin) [42]. In the presence of cellular stimuli, actin quickly polymerizes or depolymerizes to perform the required cellular function via proteins that bind it and regulate its action [42,43]. In addition to structural functions, actin plays a fundamental role in cell migration and transport processes, becoming a key factor in BBB permeability [38]. In endothelial cells, actin microfilaments are organized into a ring-like structure near the plasma membrane, forming the cortical actin ring [44], acting as a mechanical stabilizer to the plasma membrane [38,44]. TJs and AJs are attached to the cortical actin ring, allowing cell–cell and cell–matrix interactions and thus permitting endothelial barrier formation and its role, as shown in Figure 2 (upper panel). Specifically, the carboxyl termini of V-cadherin in the cytosolic portion link actin filaments through actin-binding proteins, including vinculin and α-actinin, as well as intracellular anchoring molecules such as p120-catenin, α-catenin, β-catenin, and γ-catenin [38,44,45]. Thus, a barrier with low and regulated permeability requires the cortical actin ring to have a linear and continuous junction structure [12,44], essential for reducing the distance between two adjacent endothelial cells [38,44].
Microtubules (MTs) are the main components of the cytoskeleton. Unlike actin microfilaments, MTs are mainly localized and packed in the middle of the cell and become more thinly distributed closer to the membrane (Figure 2). These cytoskeleton proteins assume a radial distribution since most MTs are strongly attached to centrosomes, although there are also non-centrosomal MTs distributed in the cytosol [46].
MTs are characterized by a highly organized dynamism; in fact, they undergo rapid polymerization or depolymerization following a stimulus, which allows them to grow and shrink in size, respectively. The essential role of MTs in regulating BBB functions has emerged in recent studies, as several MT-associated proteins are involved in junctions and the actin cytoskeleton. MT dynamics closely control BBB permeability by modifying cytoskeletal organization [47].
Intermediate filaments (IFs) are the strongest elements of all three cytoskeleton proteins. Their unique structure is the reason for the high dynamicity and flexibility of IFs, as well as stress resistance [48]. They are ubiquitously spaced throughout the cytoplasm with a higher density close to the nucleus (Figure 2) [49]. The most abundant IF protein in BMECs is vimentin. It participates in cell–cell interactions, immune activation, homeostasis, and tissue repair, providing structural support and mechanical integrity to the cells. In addition to maintaining the cell shape, it plays a fundamental role in protecting the vascular cells and tissues from various mechanical insults such as shear stress and/or contractile forces, strengthening cell attachment to the matrix, and regulating actin microfilaments [50]. Therefore, the unique characteristics of the vimentin network are crucial for the integrity and physiological function of the BBB [51].
Cytoskeleton proteins represent the cement and the bricks of adjacent BMECs, and the lack of a healthy structure of the cytoskeleton network can occur in BBB dysfunctions. This can be efficiently reproduced by in vitro advanced 3D prototypes based on integrated BBB-on-chip models [52], as well as nanoengineering approaches for clinical applications that more closely reflect their in vivo properties and functions [53,54].

3. BBB Dysfunction in MS

The BBB impairment in the pathogenesis of MS basically has a dual effect: early onset of the immune attack, and subsequently, self-sustaining demyelination and neurodegeneration processes in the CNS [55]. The mechanisms causing BBB dysfunction and integrity loss in MS are not fully understood due to the multilayer interplay of direct and/or indirect environmental and lifestyle risk factors. They include dietary habits, gut microbiome, and cigarette smoke [56,57,58,59], as well as a complex genetic scenario with more than 230 trait loci described in genome-wide association studies (GWAS) based on single nucleotide polymorphisms (SNPs) [25,60], besides several other studies [61,62,63,64,65]. Aging and age-related comorbidities are important factors in MS development and progression, and also influence BBB integrity [25,66].
Focal BBB dysfunction occurs early in the pathogenesis of new lesions, indicating that BBB impairment is an early crucial process, together with CNS immune cell infiltration, resulting in myelin damage [67]. This suggests that early diagnosis and timely intervention are strictly necessary to limit disease progression and delay the onset of irreversible neuronal loss.
BBB disruption has several effects: (i) proinflammatory cytokine and chemokine release by CNS-resident cells and infiltrating leukocytes; (ii) influx of immunoglobulins, including autoantibodies, and activation of the complement cascade; (iii) high levels of cytotoxic iron; (iv) ion channel disruption in BMECs, leading to unregulated ion passage and consequent neuronal damage; and (v) thrombin and fibrinogen influx, leading to endothelial cell activation and downstream nitric oxide and reactive oxygen species (ROS) production, as well as downregulation of TJ and AJ molecules [55,64,68,69,70,71,72].
The occurrence of the immune-mediated proinflammatory process favors the “outside-in” theory, according to which inflammation spreads from the periphery towards the CNS. The process is followed by an “inside-out” mechanism self-sustained by the central activation of lymphocytes against self-myelin that return in the peripheral compartment to recruit more proinflammatory lymphocytes, leading to a neuroinflammation-based brain-damaging loop. Both mechanisms are linked to BBB disruption [55,69,73].
Iron overload is a critical issue in BBB impairment diseases [11,63,64]. On one side, the elevated iron levels found in the brains of patients, mainly due to the death of iron-enriched oligodendrocytes and destruction of their myelin sheaths [64], contribute to barrier cell disruption through ferroptosis, a form of iron-dependent cell death due to iron-mediated lipid peroxidation [64,74,75,76,77]. On the other side, the BBB integrity loss can be the reason for the high iron levels, no longer limiting metal ion entry into the brain from the bloodstream [78,79,80].
The iron-driven oxidative stress, together with inflammation, also underlies the mitochondrial injury and the subsequent energy failure of demyelination and neurodegeneration, as in MS [81]. In detail, BBB function and myelin production are energy-intensive processes requiring more ATP and a high number of mitochondria [82]. Hence, the mitochondrial energy imbalance is another key process of BBB dysfunction.
Meanwhile, several MS patients with progressive disease show low levels of serum iron [83], and iron deficiency results in mitochondrial dysfunction and energy failure [84], causing mitochondria distress and needing the metal to make ATP. Accordingly, cells characterized by a “respiratory deficiency” play a role in pathophysiological processes in MS, including BBB integrity loss [85].
The BBB breakdown also elicits the disruption of ion channels in BMECs, resulting in an unregulated passage of ions (e.g., Ca2+; Na+; K+) between the blood and CNS tissue, overall leading to neuronal damage [86]. For instance, in MS, cellular Ca2+-regulating systems are impaired, resulting in Ca2+-dependent synaptic dysfunction, impaired plasticity, and neuronal death [86,87].
Moreover, MS-associated BBB integrity loss is responsible for an influx of thrombin and fibrinogen in the CNS [78]. Accumulation of fibrinogen, converted into fibrin by the action of thrombin [88], increases the activation of microglia and endothelial cells, which induces the further progression of inflammation, oxidative stress, and neuronal injury. In addition, brain fibrinogen impairs remyelination by inhibiting differentiation of oligodendrocyte precursors [89], causing low levels of cross-linked fibrin through the reduced FXIII transglutaminase activity, and may reduce cell adhesion, migration, and endothelial barrier function [90,91,92,93].
The sum of these processes also contributes to cytoskeleton architecture breakdown, promoting stress fiber production and the related downregulation and disruption of junction molecule complexes [94], which in turn negatively affects BBB permeability, resulting in subsequent disruption of brain homoeostasis, neuronal dysfunction, and neurodegeneration [14].

Role of the Cytoskeleton on BBB Hyperpermeability in MS

Cytoskeletal proteins may act differently where BBB dysfunction is concerned. They can directly regulate BBB permeability (paracellular flow), alter transcellular exchange, and modify endothelial metabolism [14,95].
Impaired function of the junctional proteins is driven by changes in protein expression and regulation, or post-translational modifications [14,96]. As a result of different harmful stimuli, the AJs and TJs of adjacent BMECs are damaged, causing BBB failure. Specific MS stressors, such as cytokines, chemokines, free radicals, and iron overload, cause the formation of stress fibers through aberrant actin reorganization in BMECs [97,98]. Enhanced actin activity triggers high cytoskeleton tension, resulting in increased cell spacing that causes direct hyperpermeability of the BBB. Actin reorganization can occur directly from structural changes in F-actins, resulting in the loss of the cortical ring scaffold, or from disturbances in the organization of IF and MT networks [12,97,99], as shown in Figure 2 (lower panel). In turn, the ensuing cytoskeletal disorganization elicits a cellular influx of Ca2+, the production of nitric oxide and ROS, and further formation of stress fibers, triggering a feedback loop capable of amplifying the negative effects on the junctions [12,94,100].
Regarding microfilaments, abnormal regulation of Wnt/β-catenin signaling is involved in the pathogenesis of several neurodegenerative diseases, including MS [101]. Hussain et al. induced endothelial knockout of β-catenin in adult mice and determined its impact on BBB permeability. The researchers noted low mRNA levels of Wnt target genes, indicating dramatic downregulation of endothelial Wnt/β-catenin signaling, leading to both paracellular and transcellular transport alteration [102]. Accordingly, knockdown of alpha-actinin leads to changes in organization of the actin cytoskeleton and loss of cell polarity [103], as occurs in MS [104].
With the aim to investigate cytoskeletal dysfunctions in MS, Nishihara et al. developed BMEC-like iPSCs-derived cells from both healthy controls and MS patients, establishing an in vitro model of BBB that preserves structure and transport characteristics. Unlike healthy BMECs, cells derived from MS patients were larger in size and showed impaired junctional integrity (particularly with V-cadherin, claudin-5, and occludin), impaired barrier properties, and impaired efflux pump activity. In addition, BMEC-like MS-derived cells showed an inflammatory phenotype with increased expression of adhesion molecules and interactions with immune cells [67].
After demonstrating ZO-1 and occludin abnormalities in the white matter of MS patients, Padden et al. described the role of JAM impairment in MS pathogenesis. In MS lesions, although high levels of JAM proteins were found, they are muddled and less organized [105]. In detail, JAM-A alterations mainly affect vessels, while high levels of JAM-B proteins were found in brain parenchyma. However, JAM alterations both affect BBB junctional tightness and leukocyte trafficking [105,106].
The role of IFs in MS endothelial permeability has not been extensively investigated, but studies have mainly involved intracellular filamentous vimentin. First, MS stressors activate various protein kinases (e.g., PKA and PKC), triggering the phosphorylation of the head domain of vimentin and its depolymerization, with consequent BMEC intercellular junction loss. More specifically, when the IF network is altered, the cell loses tensegrity and actin organization, which has significant effects on the disruption of BBB integrity and both transcellular and paracellular permeability [12,107,108]. From a structural point of view, the IFs collapse around the nucleus and redistribute [12,109], as shown in Figure 2 (lower panel).
Several studies show extensive involvement of molecules of the immune response associated with allergy in autoimmune demyelination, as for MS [110,111]. Among these stressors, high levels of histamine neurotransmitter are directly implicated in BBB dysfunction on different cellular and molecular levels [112,113]. In detail, Shasby et al. have shown that high levels of histamine stimulate phosphorylation of AJs (e.g., V-cadherin and catenin) and alter their link to vimentin, hindering in turn AJ architecture [114]. However, the direct relationship between allergies and the worsening of MS symptoms is still under investigation.
Finally, tubulin cytoskeleton dysfunctions are often described as characteristic features of neurodegenerative diseases, including MS. In normal endothelial cells, the MT system has strongly pronounced convergence in the centrosome region. Near the cell edge, MT density decreases significantly. MS is associated with disturbed MT organization due to stress fibers. The rapid depolymerization and disruption of peripheral MTs are essential early events in the complex mechanisms of endothelial barrier dysfunction. Researchers have proven the hypothesis that the entire system of MTs changes, which means that MTs grow out of the centrosome and become significantly shorter [46], as shown in Figure 2 (lower panel). In this regard, Mentor et al. demonstrated that, following treatment with Nocodazole, a MT depolymerizing agent, neuronal endothelial bEnd5 cells exhibit TJ anastomosis misalignments in adjacent cells, failing to establish a well-regulated brain–endothelial barrier [15], endorsing MTs’ indispensable role in the maintenance of regulatory permeability across the BBB.
A fine targeting and controlling of cytoskeletal protein levels may restore their complex and regulated organization. Their rebalancing could mitigate and prevent BBB hyperpermeability, the associated demyelination, and related brain damages.

4. Circular RNAs (circRNAs)

The exponential development of biotechnologies has enabled new discoveries in RNA biology and transcriptomics [56,115]. During the last few decades, the advancement in revolutionary new high-throughput technologies (e.g., next-generation sequencing) and bioinformatics [21] has advanced knowledge of endogenous RNA complexities and transcriptional regulation, first with the identification of microRNAs (miRs) and more recently with a newly recognized large class of non-coding RNAs (ncRNAs) known as circRNAs [116,117,118,119]. Although these species were described for the first time in the 1970s, circRNAs have attracted extensive attention in recent years because of their specific characteristics and functions [23,116,117,118]. Here, we provide an overview of circRNA features, followed by a discussion on their involvement in cytoskeleton architecture impairment.

4.1. CircRNAs: Structure and Functions

CircRNAs differ from other non-coding transcripts due to their unique circular secondary structure, which is created by back-splicing a single-stranded linear transcript, forming a covalent bond, resulting in a closed-loop molecule lacking polyadenylation and capping [118,119,120]. Hence, circRNAs are 2.5–5 times more stable than linear RNAs, due to the absence of exposed ends targetable by exoribonucleases, besides being highly conserved across species and displaying a high degree of cell and tissue specificity, particularly in the brain [63,120,121]. High-throughput RNA-seq studies performed in eukaryotes have identified a large number of circRNAs different in type and length, ranging from approximately 100 to 4000 base pairs [23,118,122].
CircRNAs are considered molecular players with a pivotal role in controlling gene expression at transcriptional and post-transcriptional levels, acting as powerful epigenetic regulators [21,123]. Currently, several circRNA functions have been elucidated, as graphically depicted in Figure 3.
A well-established ability of circRNAs is to act as miR sponges [124] (Figure 3a). In that role, circRNAs are able to specifically sequester miRs to avoid binding to their target mRNAs, thus serving as indirect regulators of specific gene expression networks, influencing the availability and performance of miRs [23,122,125,126].
Although miR sponging is the more well-known mechanism of action, transcriptomic studies have discovered thousands of circRNAs containing a small number of miR binding sites, which suggests that the majority of circRNAs possess other functions and exert additional relevant epigenetic effects [124,127].
Certain circRNAs are able to sequester proteins or translocate them between subcellular compartments, promoting their retention in the cytoplasm and suppressing their downstream functions (Figure 3b). In contrast, other circRNAs facilitate nuclear translocation of several proteins, such as transcription factors, promoting their respective actions (Figure 3c). The mechanisms that allow circRNAs to influence protein trafficking are currently not completely known [125,127,128]. More in-depth research, together with the emergence of spatial transcriptomics [129,130], shows circRNAs as protein scaffolds able to structurally support and facilitate interactions between two or more proteins, for instance co-localizing enzymes and their substrates [131] (Figure 3d).
Despite circRNAs generally being ncRNAs, several studies have provided evidence that some of these circular molecules have the ability to translate into small proteins in a cap-independent and ribosome-entry-site (IRES)-dependent manner [132] (Figure 3e). The functions of many circRNA-derived proteins have yet to be identified, although some have been implicated in cancer progression and neurodegenerative diseases [23,133].
The fascinating description of the circRNA landscape suggests a two-faced nature in these molecules. Specifically, under physiological conditions, circRNAs play essential regulatory roles in many cellular processes, carrying out various biological functions, while when dysregulated, they are implicated in the pathogenesis of several diseases [51], such as neurodegenerative disorders, including MS [134,135,136,137,138].
Accordingly, due to their high stability, cell- and tissue-specific expression, and ability to epigenetically regulate at different levels, circRNAs are considered powerful and promising biomarkers for multiple complex diseases [125,136,139]. In addition, their presence in various body fluids, such as plasma and saliva, allows circRNA detection and analysis in a non-invasive manner, making them even more suitable as biomarkers [140]. Therefore, they are currently being explored as potential high-powered diagnostic markers and target molecules to be overexpressed or knocked down as a therapeutic approach [127,140].
Even though circRNAs have been found to be abundant in the CNS and tend to accumulate with aging, the potential employment of circRNAs as biomarkers in neurodegenerative diseases has not been well explored [118].
Changes in circRNA levels might be the direct cause of MS pathogenesis as well as the downstream effect of MS pathogenic processes, serving as dual indicative biomarkers. Actually, several studies have demonstrated that epigenetic changes and environmental (risk) factors, such as vitamin D deficiency and infections (e.g., Epstein–Barr virus), seem to influence MS susceptibility and progression acting on ncRNA expression, circRNAs included [141,142,143].
Recently, for the first time, studies have demonstrated that circRNAs play a role in BBB breakdown and consequent brain dysfunction by acting on the cytoskeleton proteins [144,145,146].

4.2. CircRNAs as Potential Therapeutic Targets for Cytoskeleton Architecture in BBB Integrity

The role of cytoskeleton proteins and their dynamic contribution to BBB architecture have been discussed in the previous sections. Loss of cytoskeleton homeostasis and the consequent loss of integrity of cellular focal junctions are hallmarks of BBB breakdown, resulting in enhanced barrier permeability, a distinctive feature of MS [14]. The below section aims to illustrate the available knowledge of circRNAs as modulators of dynamic cytoskeletal structures (e.g., AJs, TJs, and microtubules) and their impact on BBB integrity, which currently remains poorly investigated.
Liao et al. performed research to assess the expression profile of circRNAs in HUVEC endothelial cells. RNAseq analysis, followed by RT-qPCR validation of the discovered transcripts, allowed the identification of 16 circRNAs directly or indirectly related to AJs, listed in Table 1 [147]. For instance, five circRNAs were found to be associated with alpha-actinin1 (ACTN1), a cytoskeletal protein isoform found alongside AJs, where it is involved in the cross-linking of actin filaments and their binding to the cell membrane, thus playing a crucial role in the organization of the cytoskeleton framework as well as in the maturation of focal adhesion [148,149]. In detail, hsa_circ_ACTN1_2453, hsa_circ_0002913, and hsa_circ_0032321 negatively affect ACTN1 expression, whereas hsa_circ_ACTN1_2450 and hsa_circ_0007644 promote protein expression [147].
The same study identified the promising hsa_circ_0074158 as a potential therapeutic target, experimentally demonstrating that the knockdown molecule can significantly enhance the expression of V-cadherin, acting on catenin alpha 1 (CTNNA1) [147], a core member of the cadherin-catenin complex that contributes to the arrangement of the junction [158]. V-cadherin disruption leads to AJ breakdown, contributing to endothelial hyperpermeability [159]. In addition, hsa_circ_0074158 downregulation reverses V-cadherin suppression by reducing endothelial hyperpermeability and potentially restoring barrier protection. Consequently, high levels of hsa_circ_0074171, hsa_circ_0008194, and hsa_circ_0007440 negatively affect CTNNA1 expression, destroying barrier integrity [147], and hence potentially being therapeutically targetable.
The tyrosine phosphatase receptor type M proteins (PTPRMs) are key players in the regulation of cell-to-cell adhesions [160], particularly the AJ pathway involved in BBB function [161]. Liao et al. have found that hsa_circ_0004188, hsa_circ_0002872, and hsa_circ_0046813 negatively influence PTPRM, whereas the expressions of hsa_circ_0006114 and hsa_circ_PTPRM_4378 enhance protein function [147].
Furthermore, researchers identified hsa_circ_0008016 and hsa_circ_0005564 as positive modulators of fibroblast growth factor receptor 1 (FGFR1) [147]. This receptor system is involved in the maintenance of vascular BBB integrity via enhancing V-cadherin stabilization at AJs, even though the precise molecular mechanism is not well understood [162,163,164].
As previously discussed, “zippering up” TJs also prevents harmful substances from crossing the BBB and entering the brain [165]. Only a few studies have discovered circRNA involvement in TJ barrier regulation (Table 1). For example, hsa_circ_DLGAP4 is highly expressed in healthy CNS but significantly decreased in neurological disorders, resulting in downregulation of TJ proteins (e.g., ZO-1, claudin-5, and occludin) and associated BBB damage [150,151]. In detail, hsa_circ_DLGAP4 regulates TJ proteins by indirectly acting as an miR-143 sponge. High miR-143 levels have a deleterious effect on BBB integrity, so pathological hsa_circ_DLGAP4 downregulation leads to its failure to sponge miR-143, resulting in TJ and barrier damage [150,166,167].
Likewise, high levels of hsa_circ_USP1 ensure the proper function of ZO-1, claudin-5, and occludin, and in contrast, circRNA downregulation disrupts barrier integrity and increases its permeability by TJ-related protein reduction [152]. Bioinformatics prediction using the CircInteractome database (https://circinteractome.nia.nih.gov/, accessed on 2 August 2024), and subsequent experimental validation, reveal hsa_circ_USP1 as an miR-194-5p sponge, which in turn regulates the transcription factor FLI1 [152].
In contrast, has_circ_HECW2 expression in BMECs promotes the degradation of ZO-1, occludin, and claudin-5. In detail, has_circ_HECW2 competitively sponges miR-30d to up-regulate the expression of autophagy-related 5, which is involved in BBB damage due to TJ depletion. Hence, specific blockage of this circRNA may be envisioned as a potential therapeutic target for the treatment of BBB damage [153,154].
Again, low expression levels of has_circ_7632 and hsa_circ_Cul2 significantly decrease the expression of vimentin [155,156], the most abundant intermediate filament, which plays a key role in BBB permeability, by microtubule and actin reorganization and by direct binding to VE-cadherin or integrin proteins [12]. hsa_circ_2858, instead, acts as an endogenous RNA competitor for the miR-17 family and for miR-93-5p, promoting vascular endothelial growth factor A (VEGFA) expression in BMECs, thus leading to TJ disruption and BBB hyperpermeability [143,153,168].
Particularly noteworthy is the hsa_circ_ZNF609, a microtubule-related circRNA (Table 1). Rossi et al. explain the circRNA molecular mechanism, revealing that circZNF609, widely expressed in the brain, directly binds several mRNAs. Among them, CKAP5 is a key molecule for the stability and dynamic functions of microtubules [157]. Consequently, its downregulation could be partially responsible for cytoskeleton instability and BBB dysfunction.
In summary, the exploration of epigenetic mechanisms that preserve cytoskeleton architecture and ECM dynamics, paves the way for novel attractive circRNA-based therapeutic approaches. Strategies aiming at protecting and preserving BBB integrity and/or reverse endothelial dysfunction may have a great therapeutic potential not only in classical neurodegenerative diseases but also in other complex diseases in which epigenetic mechanisms and dysfunctional endothelial cells play a role [169,170,171].

5. Conclusions, Remarks and Future Directions

Individuals diagnosed with multiple sclerosis (MS) often deal with a diminished quality of life due to physical and cognitive impairments that can significantly impact their daily activities and overall well-being [22]. MS is a prevalent chronic inflammatory neurodegenerative condition leading to a range of debilitating symptoms and challenges for those living with the disease [1,2,9]. Interestingly, recent clinical studies suggest that a multilayer approach, involving physical activity and a healthy diet, may ameliorate disease progression and improve disability outcome, according to the individual genetic background and in line with the P4 medicine ideal [172]. Despite the urgent need for effective and long-lasting treatments to manage MS, such options are currently lacking, leaving patients and healthcare providers with limited options for disease management and symptom control. A key early feature of MS is the compromised integrity of the blood–brain barrier (BBB), a crucial protective barrier in the central nervous system that regulates the passage of substances between the bloodstream and the brain [76]. This disruption in the BBB’s function results in abnormal permeability, potentially allowing harmful immune cells and molecules to enter the brain and contribute to the disease progression. Addressing this distinct characteristic through targeted therapeutic strategies aimed at restoring the integrity of tight and adherens junctions between endothelial cells at the interface with the central nervous system holds promise for developing more effective treatments that could slow or halt the progression of MS and improve the quality of life for those affected by this complex and challenging condition. This review seeks to provide an overview of the significance of cytoskeleton proteins in BBB function and to explore the potential role of circular RNAs (circRNAs) in maintaining cytoskeletal homeostasis and BBB structure. CircRNAs have gained attention in recent years for their regulatory role in gene expression at multiple levels, owing to their stability and tissue-specific expression patterns [115,116,117,118]. They are increasingly recognized as valuable biomarkers for monitoring disease progression and as targets for novel therapeutic interventions [115,116,117,124].
Circular RNAs (circRNAs) have the potential to serve as promising therapeutic targets for enhancing the integrity of the BBB and consequently improving the life expectancy of individuals with MS. CircRNAs can influence the tight junctions (TJs) and adherens junctions (AJs) directly or indirectly by modulating upstream signaling pathways and transcription factors [143,145,147,152,155,156,157,167]. However, our understanding of how circRNAs impact the complex architecture of the BBB is still in its nascent stages, indicating the need for more comprehensive and detailed research in this area to determine suitable druggable targets. To advance this field, extensive high-throughput next-generation sequencing (NGS) studies, in conjunction with sophisticated bioinformatics tools and functional validation experiments, are essential for identifying and characterizing novel circRNAs capable of regulating BBB cytoskeletal proteins. Moreover, the utilization of innovative spatial transcriptomics techniques that specifically target circRNAs could greatly enhance our understanding of these complex biological processes by mapping out the spatial expression patterns of circRNAs within the intricate structural framework of tissues [128]. Despite the inherent challenges associated with this cutting-edge approach, the development of therapeutic strategies focused on circRNAs presents a promising and rapidly expanding frontier in the treatment of MS, aligning seamlessly with the ever-evolving landscape of modern medicine and offering new avenues for more effective interventions.

Author Contributions

Conceptualization, E.D., D.G. and V.T.; writing—original draft preparation, E.D., D.G. and V.T.; writing—review and editing, E.D., F.S., M.V., B.A., M.G., A.V.S., P.S., G.Z., V.T. and D.G.; manuscript revision, E.D., F.S., M.V., B.A., M.G., A.V.S., P.S., G.Z., V.T. and D.G.; funding acquisition, D.G. and V.T. All authors have read and agreed to the published version of the manuscript.

Funding

FAR and FIRD local grants from the University of Ferrara (to D.G. and V.T.); National Recovery and Resilience Plan (NRRP), Mission 04 Component 2 Investment 1.5—NextGenerationEU, Call for tender n. 3277 dated 30/12/2021; Award Number: 0001052 dated 23 June 2022.

Acknowledgments

The authors thank Devahuti R. Chaliha for English proofreading.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Compston, A.; Coles, A. Multiple Sclerosis. Lancet Lond. Engl. 2008, 372, 1502–1517. [Google Scholar] [CrossRef] [PubMed]
  2. Ghasemi, N.; Razavi, S.; Nikzad, E. Multiple Sclerosis: Pathogenesis, Symptoms, Diagnoses and Cell-Based Therapy. Cell J. 2017, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
  3. Soelberg Sorensen, P.; Giovannoni, G.; Montalban, X.; Thalheim, C.; Zaratin, P.; Comi, G. The Multiple Sclerosis Care Unit. Mult. Scler. Houndmills Basingstoke Engl. 2019, 25, 627–636. [Google Scholar] [CrossRef]
  4. Oh, J.; Vidal-Jordana, A.; Montalban, X. Multiple Sclerosis: Clinical Aspects. Curr. Opin. Neurol. 2018, 31, 752–759. [Google Scholar] [CrossRef]
  5. Benedict, R.H.B.; Amato, M.P.; DeLuca, J.; Geurts, J.J.G. Cognitive Impairment in Multiple Sclerosis: Clinical Management, MRI, and Therapeutic Avenues. Lancet Neurol. 2020, 19, 860–871. [Google Scholar] [CrossRef] [PubMed]
  6. McGinley, M.P.; Goldschmidt, C.H.; Rae-Grant, A.D. Diagnosis and Treatment of Multiple Sclerosis: A Review. JAMA 2021, 325, 765–779. [Google Scholar] [CrossRef]
  7. Carotenuto, A.; Cacciaguerra, L.; Pagani, E.; Preziosa, P.; Filippi, M.; Rocca, M.A. Glymphatic System Impairment in Multiple Sclerosis: Relation with Brain Damage and Disability. Brain J. Neurol. 2022, 145, 2785–2795. [Google Scholar] [CrossRef] [PubMed]
  8. Yu, S.; Zeng, M.Y. Sex-Biased Gut Dopamine Signaling in Multiple Sclerosis. Immunity 2023, 56, 2674–2676. [Google Scholar] [CrossRef]
  9. Lublin, F.D.; Häring, D.A.; Ganjgahi, H.; Ocampo, A.; Hatami, F.; Čuklina, J.; Aarden, P.; Dahlke, F.; Arnold, D.L.; Wiendl, H.; et al. How Patients with Multiple Sclerosis Acquire Disability. Brain J. Neurol. 2022, 145, 3147–3161. [Google Scholar] [CrossRef]
  10. Wengler, K.; Ha, J.; Syritsyna, O.; Bangiyev, L.; Coyle, P.K.; Duong, T.Q.; Schweitzer, M.E.; He, X. Abnormal Blood-Brain Barrier Water Exchange in Chronic Multiple Sclerosis Lesions: A Preliminary Study. Magn. Reson. Imaging 2020, 70, 126–133. [Google Scholar] [CrossRef]
  11. Zierfuss, B.; Larochelle, C.; Prat, A. Blood-Brain Barrier Dysfunction in Multiple Sclerosis: Causes, Consequences, and Potential Effects of Therapies. Lancet Neurol. 2024, 23, 95–109. [Google Scholar] [CrossRef]
  12. Bayir, E.; Sendemir, A. Role of Intermediate Filaments in Blood-Brain Barrier in Health and Disease. Cells 2021, 10, 1400. [Google Scholar] [CrossRef]
  13. Hohmann, T.; Dehghani, F. The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019, 8, 362. [Google Scholar] [CrossRef]
  14. Stamatovic, S.M.; Johnson, A.M.; Keep, R.F.; Andjelkovic, A.V. Junctional Proteins of the Blood-Brain Barrier: New Insights into Function and Dysfunction. Tissue Barriers 2016, 4, e1154641. [Google Scholar] [CrossRef]
  15. Mentor, S.; Makhathini, K.B.; Fisher, D. The Role of Cytoskeletal Proteins in the Formation of a Functional In Vitro Blood-Brain Barrier Model. Int. J. Mol. Sci. 2022, 23, 742. [Google Scholar] [CrossRef] [PubMed]
  16. Haase, S.; Linker, R.A. Inflammation in Multiple Sclerosis. Ther. Adv. Neurol. Disord. 2021, 14, 17562864211007687. [Google Scholar] [CrossRef]
  17. Wang, K.; Song, F.; Fernandez-Escobar, A.; Luo, G.; Wang, J.-H.; Sun, Y. The Properties of Cytokines in Multiple Sclerosis: Pros and Cons. Am. J. Med. Sci. 2018, 356, 552–560. [Google Scholar] [CrossRef]
  18. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-Brain Barrier Breakdown in Alzheimer Disease and Other Neurodegenerative Disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef] [PubMed]
  19. Löscher, W.; Friedman, A. Structural, Molecular, and Functional Alterations of the Blood-Brain Barrier during Epileptogenesis and Epilepsy: A Cause, Consequence, or Both? Int. J. Mol. Sci. 2020, 21, 591. [Google Scholar] [CrossRef]
  20. Steinruecke, M.; Lonergan, R.M.; Selvaraj, B.T.; Chandran, S.; Diaz-Castro, B.; Stavrou, M. Blood-CNS Barrier Dysfunction in Amyotrophic Lateral Sclerosis: Proposed Mechanisms and Clinical Implications. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2023, 43, 642–654. [Google Scholar] [CrossRef]
  21. Liu, K.-S.; Pan, F.; Mao, X.-D.; Liu, C.; Chen, Y.-J. Biological Functions of Circular RNAs and Their Roles in Occurrence of Reproduction and Gynecological Diseases. Am. J. Transl. Res. 2019, 11, 1–15. [Google Scholar] [PubMed]
  22. Cashion, J.M.; Young, K.M.; Sutherland, B.A. How Does Neurovascular Unit Dysfunction Contribute to Multiple Sclerosis? Neurobiol. Dis. 2023, 178, 106028. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, X.; Cai, Y.; Xu, J. Circular RNAs: Biogenesis, Mechanism, and Function in Human Cancers. Int. J. Mol. Sci. 2019, 20, 3926. [Google Scholar] [CrossRef] [PubMed]
  24. Lunde, H.M.B.; Assmus, J.; Myhr, K.-M.; Bø, L.; Grytten, N. Survival and Cause of Death in Multiple Sclerosis: A 60-Year Longitudinal Population Study. J. Neurol. Neurosurg. Psychiatry 2017, 88, 621–625. [Google Scholar] [CrossRef] [PubMed]
  25. Ortiz, G.G.; Pacheco-Moisés, F.P.; Macías-Islas, M.Á.; Flores-Alvarado, L.J.; Mireles-Ramírez, M.A.; González-Renovato, E.D.; Hernández-Navarro, V.E.; Sánchez-López, A.L.; Alatorre-Jiménez, M.A. Role of the Blood-Brain Barrier in Multiple Sclerosis. Arch. Med. Res. 2014, 45, 687–697. [Google Scholar] [CrossRef] [PubMed]
  26. Bell, A.H.; Miller, S.L.; Castillo-Melendez, M.; Malhotra, A. The Neurovascular Unit: Effects of Brain Insults During the Perinatal Period. Front. Neurosci. 2019, 13, 1452. [Google Scholar] [CrossRef] [PubMed]
  27. Persidsky, Y.; Ramirez, S.H.; Haorah, J.; Kanmogne, G.D. Blood-Brain Barrier: Structural Components and Function under Physiologic and Pathologic Conditions. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2006, 1, 223–236. [Google Scholar] [CrossRef]
  28. Kadry, H.; Noorani, B.; Cucullo, L. A Blood-Brain Barrier Overview on Structure, Function, Impairment, and Biomarkers of Integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  29. Bazzoni, G.; Dejana, E. Endothelial Cell-to-Cell Junctions: Molecular Organization and Role in Vascular Homeostasis. Physiol. Rev. 2004, 84, 869–901. [Google Scholar] [CrossRef]
  30. Greene, C.; Hanley, N.; Campbell, M. Claudin-5: Gatekeeper of Neurological Function. Fluids Barriers CNS 2019, 16, 3. [Google Scholar] [CrossRef]
  31. Hashimoto, Y.; Campbell, M.; Tachibana, K.; Okada, Y.; Kondoh, M. Claudin-5: A Pharmacological Target to Modify the Permeability of the Blood-Brain Barrier. Biol. Pharm. Bull. 2021, 44, 1380–1390. [Google Scholar] [CrossRef] [PubMed]
  32. Niu, J.; Tsai, H.-H.; Hoi, K.K.; Huang, N.; Yu, G.; Kim, K.; Baranzini, S.E.; Xiao, L.; Chan, J.R.; Fancy, S.P.J. Aberrant Oligodendroglial-Vascular Interactions Disrupt the Blood Brain Barrier Triggering CNS Inflammation. Nat. Neurosci. 2019, 22, 709–718. [Google Scholar] [CrossRef]
  33. Feldman, G.J.; Mullin, J.M.; Ryan, M.P. Occludin: Structure, Function and Regulation. Adv. Drug Deliv. Rev. 2005, 57, 883–917. [Google Scholar] [CrossRef] [PubMed]
  34. Cummins, P.M. Occludin: One Protein, Many Forms. Mol. Cell. Biol. 2012, 32, 242–250. [Google Scholar] [CrossRef]
  35. Sugiyama, S.; Sasaki, T.; Tanaka, H.; Yan, H.; Ikegami, T.; Kanki, H.; Nishiyama, K.; Beck, G.; Gon, Y.; Okazaki, S.; et al. The Tight Junction Protein Occludin Modulates Blood-Brain Barrier Integrity and Neurological Function after Ischemic Stroke in Mice. Sci. Rep. 2023, 13, 2892. [Google Scholar] [CrossRef] [PubMed]
  36. Hartmann, C.; Schwietzer, Y.A.; Otani, T.; Furuse, M.; Ebnet, K. Physiological Functions of Junctional Adhesion Molecules (JAMs) in Tight Junctions. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183299. [Google Scholar] [CrossRef]
  37. Duong, C.N.; Vestweber, D. Mechanisms Ensuring Endothelial Junction Integrity Beyond VE-Cadherin. Front. Physiol. 2020, 11, 519. [Google Scholar] [CrossRef]
  38. Ippolitov, D.; Arreza, L.; Munir, M.N.; Hombach-Klonisch, S. Brain Microvascular Pericytes-More than Bystanders in Breast Cancer Brain Metastasis. Cells 2022, 11, 1263. [Google Scholar] [CrossRef]
  39. Kim, Y.; Park, J.; Choi, Y.K. The Role of Astrocytes in the Central Nervous System Focused on BK Channel and Heme Oxygenase Metabolites: A Review. Antioxidants 2019, 8, 121. [Google Scholar] [CrossRef]
  40. Cabezas, R.; Avila, M.; Gonzalez, J.; El-Bachá, R.S.; Báez, E.; García-Segura, L.M.; Jurado Coronel, J.C.; Capani, F.; Cardona-Gomez, G.P.; Barreto, G.E. Astrocytic Modulation of Blood Brain Barrier: Perspectives on Parkinson’s Disease. Front. Cell. Neurosci. 2014, 8, 211. [Google Scholar] [CrossRef]
  41. Dominguez, R.; Holmes, K.C. Actin Structure and Function. Annu. Rev. Biophys. 2011, 40, 169–186. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, S.H.; Dominguez, R. Regulation of Actin Cytoskeleton Dynamics in Cells. Mol. Cells 2010, 29, 311–325. [Google Scholar] [CrossRef] [PubMed]
  43. Belvitch, P.; Htwe, Y.M.; Brown, M.E.; Dudek, S. Cortical Actin Dynamics in Endothelial Permeability. Curr. Top. Membr. 2018, 82, 141–195. [Google Scholar] [CrossRef] [PubMed]
  44. Lampugnani, M.G.; Zanetti, A.; Breviario, F.; Balconi, G.; Orsenigo, F.; Corada, M.; Spagnuolo, R.; Betson, M.; Braga, V.; Dejana, E. VE-Cadherin Regulates Endothelial Actin Activating Rac and Increasing Membrane Association of Tiam. Mol. Biol. Cell 2002, 13, 1175–1189. [Google Scholar] [CrossRef] [PubMed]
  45. Alieva, I.B. Role of Microtubule Cytoskeleton in Regulation of Endothelial Barrier Function. Biochem. Biokhimiia 2014, 79, 964–975. [Google Scholar] [CrossRef] [PubMed]
  46. Karki, P.; Birukova, A.A. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front. Physiol. 2021, 12, 758313. [Google Scholar] [CrossRef] [PubMed]
  47. Herrmann, H.; Aebi, U. Intermediate Filaments: Structure and Assembly. Cold Spring Harb. Perspect. Biol. 2016, 8, a018242. [Google Scholar] [CrossRef] [PubMed]
  48. Pogoda, K.; Janmey, P.A. Transmit and Protect: The Mechanical Functions of Intermediate Filaments. Curr. Opin. Cell Biol. 2023, 85, 102281. [Google Scholar] [CrossRef] [PubMed]
  49. Ridge, K.M.; Eriksson, J.E.; Pekny, M.; Goldman, R.D. Roles of Vimentin in Health and Disease. Genes Dev. 2022, 36, 391–407. [Google Scholar] [CrossRef]
  50. Alahmari, A. Blood-Brain Barrier Overview: Structural and Functional Correlation. Neural Plast. 2021, 2021, 6564585. [Google Scholar] [CrossRef]
  51. Singh, A.V.; Chandrasekar, V.; Laux, P.; Luch, A.; Dakua, S.P.; Zamboni, P.; Shelar, A.; Yang, Y.; Pandit, V.; Tisato, V.; et al. Micropatterned Neurovascular Interface to Mimic the Blood-Brain Barrier’s Neurophysiology and Micromechanical Function: A BBB-on-CHIP Model. Cells 2022, 11, 2801. [Google Scholar] [CrossRef] [PubMed]
  52. Rathi, S.; Reche, A.; Dhamdhere, N.; Bolenwar, A. Perspectives on the Application of Nanomaterials in Medical and Dental Practices. Cureus 2023, 15, e43565. [Google Scholar] [CrossRef] [PubMed]
  53. Lombardo, L.; Carinci, F.; Martini, M.; Gemmati, D.; Nardone, M.; Siciliani, G. Quantitive Evaluation of Dentin Sialoprotein (DSP) Using Microbeads—A Potential Early Marker of Root Resorption. ORAL Implantol. 2016, 9, 132–142. [Google Scholar] [CrossRef]
  54. Balasa, R.; Barcutean, L.; Mosora, O.; Manu, D. Reviewing the Significance of Blood-Brain Barrier Disruption in Multiple Sclerosis Pathology and Treatment. Int. J. Mol. Sci. 2021, 22, 8370. [Google Scholar] [CrossRef] [PubMed]
  55. Dai, X.; Shen, L. Advances and Trends in Omics Technology Development. Front. Med. 2022, 9, 911861. [Google Scholar] [CrossRef] [PubMed]
  56. Nishanth, K.; Tariq, E.; Nzvere, F.P.; Miqdad, M.; Cancarevic, I. Role of Smoking in the Pathogenesis of Multiple Sclerosis: A Review Article. Cureus 2020, 12, e9564. [Google Scholar] [CrossRef] [PubMed]
  57. Altieri, C.; Speranza, B.; Corbo, M.R.; Sinigaglia, M.; Bevilacqua, A. Gut-Microbiota, and Multiple Sclerosis: Background, Evidence, and Perspectives. Nutrients 2023, 15, 942. [Google Scholar] [CrossRef] [PubMed]
  58. Correale, J.; Hohlfeld, R.; Baranzini, S.E. The Role of the Gut Microbiota in Multiple Sclerosis. Nat. Rev. Neurol. 2022, 18, 544–558. [Google Scholar] [CrossRef] [PubMed]
  59. Cotsapas, C.; Mitrovic, M. Genome-wide Association Studies of Multiple Sclerosis. Clin. Transl. Immunol. 2018, 7, e1018. [Google Scholar] [CrossRef]
  60. Ziliotto, N.; Marchetti, G.; Scapoli, C.; Bovolenta, M.; Meneghetti, S.; Benazzo, A.; Lunghi, B.; Balestra, D.; Laino, L.A.; Bozzini, N.; et al. C6orf10 Low-Frequency and Rare Variants in Italian Multiple Sclerosis Patients. Front. Genet. 2019, 10, 573. [Google Scholar] [CrossRef]
  61. Paraboschi, E.M.; Rimoldi, V.; Soldà, G.; Tabaglio, T.; Dall’Osso, C.; Saba, E.; Vigliano, M.; Salviati, A.; Leone, M.; Benedetti, M.D.; et al. Functional Variations Modulating PRKCA Expression and Alternative Splicing Predispose to Multiple Sclerosis. Hum. Mol. Genet. 2014, 23, 6746–6761. [Google Scholar] [CrossRef] [PubMed]
  62. Paraboschi, E.M.; Cardamone, G.; Rimoldi, V.; Gemmati, D.; Spreafico, M.; Duga, S.; Soldà, G.; Asselta, R. Meta-Analysis of Multiple Sclerosis Microarray Data Reveals Dysregulation in RNA Splicing Regulatory Genes. Int. J. Mol. Sci. 2015, 16, 23463–23481. [Google Scholar] [CrossRef] [PubMed]
  63. Gemmati, D.; Zeri, G.; Orioli, E.; De Gaetano, F.E.; Salvi, F.; Bartolomei, I.; D’Alfonso, S.; Dall’osso, C.; Leone, M.A.; Singh, A.V.; et al. Polymorphisms in the Genes Coding for Iron Binding and Transporting Proteins Are Associated with Disability, Severity, and Early Progression in Multiple Sclerosis. BMC Med. Genet. 2012, 13, 70. [Google Scholar] [CrossRef]
  64. Ferlini, A.; Bovolenta, M.; Neri, M.; Gualandi, F.; Balboni, A.; Yuryev, A.; Salvi, F.; Gemmati, D.; Liboni, A.; Zamboni, P. Custom CGH Array Profiling of Copy Number Variations (CNVs) on Chromosome 6p21.32 (HLA Locus) in Patients with Venous Malformations Associated with Multiple Sclerosis. BMC Med. Genet. 2010, 11, 64. [Google Scholar] [CrossRef] [PubMed]
  65. Knox, E.G.; Aburto, M.R.; Clarke, G.; Cryan, J.F.; O’Driscoll, C.M. The Blood-Brain Barrier in Aging and Neurodegeneration. Mol. Psychiatry 2022, 27, 2659–2673. [Google Scholar] [CrossRef] [PubMed]
  66. Nishihara, H.; Perriot, S.; Gastfriend, B.D.; Steinfort, M.; Cibien, C.; Soldati, S.; Matsuo, K.; Guimbal, S.; Mathias, A.; Palecek, S.P.; et al. Intrinsic Blood-Brain Barrier Dysfunction Contributes to Multiple Sclerosis Pathogenesis. Brain J. Neurol. 2022, 145, 4334–4348. [Google Scholar] [CrossRef] [PubMed]
  67. Salimi, H.; Klein, R.S. Disruption of the Blood-Brain Barrier During Neuroinflammatory and Neuroinfectious Diseases. Neuroimmune Dis. 2019, 195–234. [Google Scholar] [CrossRef]
  68. Luchicchi, A.; Preziosa, P.; ’t Hart, B. Editorial: “Inside-Out” vs “Outside-In” Paradigms in Multiple Sclerosis Etiopathogenesis. Front. Cell. Neurosci. 2021, 15, 666529. [Google Scholar] [CrossRef]
  69. Boscia, F.; Elkjaer, M.L.; Illes, Z.; Kukley, M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front. Cell. Neurosci. 2021, 15, 685703. [Google Scholar] [CrossRef]
  70. Plantone, D.; Inglese, M.; Salvetti, M.; Koudriavtseva, T. A Perspective of Coagulation Dysfunction in Multiple Sclerosis and in Experimental Allergic Encephalomyelitis. Front. Neurol. 2019, 9, 1175. [Google Scholar] [CrossRef]
  71. Yang, S.; Chen, Y.; Deng, X.; Jiang, W.; Li, B.; Fu, Z.; Du, M.; Ding, R. Hemoglobin-Induced Nitric Oxide Synthase Overexpression and Nitric Oxide Production Contribute to Blood-Brain Barrier Disruption in the Rat. J. Mol. Neurosci. MN 2013, 51, 352–363. [Google Scholar] [CrossRef]
  72. Saez-Calveras, N.; Stuve, O. The Role of the Complement System in Multiple Sclerosis: A Review. Front. Immunol. 2022, 13, 970486. [Google Scholar] [CrossRef] [PubMed]
  73. Tisato, V.; Castiglione, A.; Ciorba, A.; Aimoni, C.; Silva, J.A.; Gallo, I.; D’Aversa, E.; Salvatori, F.; Bianchini, C.; Pelucchi, S.; et al. LINE-1 Global DNA Methylation, Iron Homeostasis Genes, Sex and Age in Sudden Sensorineural Hearing Loss (SSNHL). Hum. Genomics 2023, 17, 112. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, X.; Pang, X.; Yeo, A.J.; Xie, S.; Xiang, M.; Shi, B.; Yu, G.; Li, C. The Molecular Mechanisms of Ferroptosis and Its Role in Blood-Brain Barrier Dysfunction. Front. Cell. Neurosci. 2022, 16, 889765. [Google Scholar] [CrossRef] [PubMed]
  75. Van San, E.; Debruyne, A.C.; Veeckmans, G.; Tyurina, Y.Y.; Tyurin, V.A.; Zheng, H.; Choi, S.M.; Augustyns, K.; van Loo, G.; Michalke, B.; et al. Ferroptosis Contributes to Multiple Sclerosis and Its Pharmacological Targeting Suppresses Experimental Disease Progression. Cell Death Differ. 2023, 30, 2092–2103. [Google Scholar] [CrossRef] [PubMed]
  76. Sheykhansari, S.; Kozielski, K.; Bill, J.; Sitti, M.; Gemmati, D.; Zamboni, P.; Singh, A.V. Redox Metals Homeostasis in Multiple Sclerosis and Amyotrophic Lateral Sclerosis: A Review. Cell Death Dis. 2018, 9, 348. [Google Scholar] [CrossRef] [PubMed]
  77. Klistorner, A.; Wang, C.; Yiannikas, C.; Parratt, J.; Dwyer, M.; Barton, J.; Graham, S.L.; You, Y.; Liu, S.; Barnett, M.H. Evidence of Progressive Tissue Loss in the Core of Chronic MS Lesions: A Longitudinal DTI Study. NeuroImage Clin. 2018, 17, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  78. Mills, E.; Dong, X.; Wang, F.; Xu, H. Mechanisms of Brain Iron Transport: Insight into Neurodegeneration and CNS Disorders. Future Med. Chem. 2010, 2, 51. [Google Scholar] [CrossRef]
  79. Tisato, V.; Zuliani, G.; Vigliano, M.; Longo, G.; Franchini, E.; Secchiero, P.; Zauli, G.; Paraboschi, E.M.; Vikram Singh, A.; Serino, M.L.; et al. Gene-Gene Interactions among Coding Genes of Iron-Homeostasis Proteins and APOE-Alleles in Cognitive Impairment Diseases. PLoS ONE 2018, 13, e0193867. [Google Scholar] [CrossRef]
  80. de Barcelos, I.P.; Troxell, R.M.; Graves, J.S. Mitochondrial Dysfunction and Multiple Sclerosis. Biology 2019, 8, 37. [Google Scholar] [CrossRef]
  81. Connor, J.R.; Menzies, S.L. Relationship of Iron to Oligodendrocytes and Myelination. Glia 1996, 17, 83–93. [Google Scholar] [CrossRef]
  82. Tang, C.; Yang, J.; Zhu, C.; Ding, Y.; Yang, S.; Xu, B.; He, D. Iron Metabolism Disorder and Multiple Sclerosis: A Comprehensive Analysis. Front. Immunol. 2024, 15, 1376838. [Google Scholar] [CrossRef] [PubMed]
  83. van Rensburg, S.J.; Peeters, A.V.; van Toorn, R.; Schoeman, J.; Moremi, K.E.; van Heerden, C.J.; Kotze, M.J. Identification of an Iron-Responsive Subtype in Two Children Diagnosed with Relapsing-Remitting Multiple Sclerosis Using Whole Exome Sequencing. Mol. Genet. Metab. Rep. 2019, 19, 100465. [Google Scholar] [CrossRef] [PubMed]
  84. Haider, L. Inflammation, Iron, Energy Failure, and Oxidative Stress in the Pathogenesis of Multiple Sclerosis. Oxid. Med. Cell. Longev. 2015, 2015, 725370. [Google Scholar] [CrossRef]
  85. Cherchi, F.; Bulli, I.; Venturini, M.; Pugliese, A.M.; Coppi, E. Ion Channels as New Attractive Targets to Improve Re-Myelination Processes in the Brain. Int. J. Mol. Sci. 2021, 22, 7277. [Google Scholar] [CrossRef] [PubMed]
  86. Zündorf, G.; Reiser, G. Calcium Dysregulation and Homeostasis of Neural Calcium in the Molecular Mechanisms of Neurodegenerative Diseases Provide Multiple Targets for Neuroprotection. Antioxid. Redox Signal. 2011, 14, 1275–1288. [Google Scholar] [CrossRef] [PubMed]
  87. Gemmati, D.; Vigliano, M.; Burini, F.; Mari, R.; El Mohsein, H.H.A.; Parmeggiani, F.; Serino, M.L. Coagulation Factor XIIIA (F13A1): Novel Perspectives in Treatment and Pharmacogenetics. Curr. Pharm. Des. 2016, 22, 1449–1459. [Google Scholar] [CrossRef] [PubMed]
  88. Alruwaili, M.; Al-Kuraishy, H.M.; Alexiou, A.; Papadakis, M.; ALRashdi, B.M.; Elhussieny, O.; Saad, H.M.; Batiha, G.E.-S. Pathogenic Role of Fibrinogen in the Neuropathology of Multiple Sclerosis: A Tale of Sorrows and Fears. Neurochem. Res. 2023, 48, 3255–3269. [Google Scholar] [CrossRef] [PubMed]
  89. Gemmati, D.; Zeri, G.; Orioli, E.; Mari, R.; Moratelli, S.; Vigliano, M.; Marchesini, J.; Grossi, M.E.; Pecoraro, A.; Cuneo, A.; et al. Factor XIII-A Dynamics in Acute Myocardial Infarction: A Novel Prognostic Biomarker? Thromb. Haemost. 2015, 114, 123–132. [Google Scholar] [CrossRef]
  90. Zamboni, P.; De Mattei, M.; Ongaro, A.; Fogato, L.; Carandina, S.; De Palma, M.; Tognazzo, S.; Scapoli, G.L.; Serino, M.L.; Caruso, A.; et al. Factor XIII Contrasts the Effects of Metalloproteinases in Human Dermal Fibroblast Cultured Cells. Vasc. Endovascular Surg. 2004, 38, 431–438. [Google Scholar] [CrossRef]
  91. Noll, T.; Wozniak, G. Factor XIII and endothelial barrier function. Hamostaseologie 2002, 22, 28–31. [Google Scholar] [CrossRef] [PubMed]
  92. Noll, T.; Wozniak, G.; McCarson, K.; Hajimohammad, A.; Metzner, H.J.; Inserte, J.; Kummer, W.; Hehrlein, F.W.; Piper, H.M. Effect of Factor XIII on Endothelial Barrier Function. J. Exp. Med. 1999, 189, 1373–1382. [Google Scholar] [CrossRef] [PubMed]
  93. Brailoiu, E.; Shipsky, M.M.; Yan, G.; Abood, M.E.; Brailoiu, G.C. Mechanisms of Modulation of Brain Microvascular Endothelial Cells Function by Thrombin. Brain Res. 2017, 1657, 167–175. [Google Scholar] [CrossRef] [PubMed]
  94. Zhao, Y.; Gan, L.; Ren, L.; Lin, Y.; Ma, C.; Lin, X. Factors Influencing the Blood-Brain Barrier Permeability. Brain Res. 2022, 1788, 147937. [Google Scholar] [CrossRef]
  95. Reiche, J.; Huber, O. Post-Translational Modifications of Tight Junction Transmembrane Proteins and Their Direct Effect on Barrier Function. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183330. [Google Scholar] [CrossRef] [PubMed]
  96. Campos, S.B.; Ashworth, S.L.; Wean, S.; Hosford, M.; Sandoval, R.M.; Hallett, M.A.; Atkinson, S.J.; Molitoris, B.A. Cytokine-Induced F-Actin Reorganization in Endothelial Cells Involves RhoA Activation. Am. J. Physiol.-Ren. Physiol. 2009, 296, F487–F495. [Google Scholar] [CrossRef]
  97. Alvarez, J.I.; Cayrol, R.; Prat, A. Disruption of Central Nervous System Barriers in Multiple Sclerosis. Biochim. Biophys. Acta 2011, 1812, 252–264. [Google Scholar] [CrossRef] [PubMed]
  98. Plumb, J.; McQuaid, S.; Mirakhur, M.; Kirk, J. Abnormal Endothelial Tight Junctions in Active Lesions and Normal-Appearing White Matter in Multiple Sclerosis. Brain Pathol. Zurich Switz. 2002, 12, 154–169. [Google Scholar] [CrossRef]
  99. Negri, S.; Faris, P.; Moccia, F. Reactive Oxygen Species and Endothelial Ca2+ Signaling: Brothers in Arms or Partners in Crime? Int. J. Mol. Sci. 2021, 22, 9821. [Google Scholar] [CrossRef]
  100. Ramakrishna, K.; Nalla, L.V.; Naresh, D.; Venkateswarlu, K.; Viswanadh, M.K.; Nalluri, B.N.; Chakravarthy, G.; Duguluri, S.; Singh, P.; Rai, S.N.; et al. WNT-β Catenin Signaling as a Potential Therapeutic Target for Neurodegenerative Diseases: Current Status and Future Perspective. Diseases 2023, 11, 89. [Google Scholar] [CrossRef]
  101. Hussain, B.; Fang, C.; Huang, X.; Feng, Z.; Yao, Y.; Wang, Y.; Chang, J. Endothelial β-Catenin Deficiency Causes Blood-Brain Barrier Breakdown via Enhancing the Paracellular and Transcellular Permeability. Front. Mol. Neurosci. 2022, 15, 895429. [Google Scholar] [CrossRef] [PubMed]
  102. Hamill, K.J.; Hiroyasu, S.; Colburn, Z.T.; Ventrella, R.V.; Hopkinson, S.B.; Skalli, O.; Jones, J.C.R. Alpha Actinin-1 Regulates Cell-Matrix Adhesion Organization in Keratinocytes: Consequences for Skin Cell Motility. J. Investig. Dermatol. 2015, 135, 1043–1052. [Google Scholar] [CrossRef]
  103. Pandey, S.; Dioni, I.; Lambardi, D.; Real-Fernandez, F.; Peroni, E.; Pacini, G.; Lolli, F.; Seraglia, R.; Papini, A.M.; Rovero, P. Alpha Actinin Is Specifically Recognized by Multiple Sclerosis Autoantibodies Isolated Using an N-Glucosylated Peptide Epitope. Mol. Cell. Proteomics MCP 2013, 12, 277–282. [Google Scholar] [CrossRef] [PubMed]
  104. Padden, M.; Leech, S.; Craig, B.; Kirk, J.; Brankin, B.; McQuaid, S. Differences in Expression of Junctional Adhesion Molecule-A and Beta-Catenin in Multiple Sclerosis Brain Tissue: Increasing Evidence for the Role of Tight Junction Pathology. Acta Neuropathol. 2007, 113, 177–186. [Google Scholar] [CrossRef] [PubMed]
  105. Stamatovic, S.M.; Sladojevic, N.; Keep, R.F.; Andjelkovic, A.V. Relocalization of Junctional Adhesion Molecule A during Inflammatory Stimulation of Brain Endothelial Cells. Mol. Cell. Biol. 2012, 32, 3414–3427. [Google Scholar] [CrossRef] [PubMed]
  106. Gregor, M.; Osmanagic-Myers, S.; Burgstaller, G.; Wolfram, M.; Fischer, I.; Walko, G.; Resch, G.P.; Jörgl, A.; Herrmann, H.; Wiche, G. Mechanosensing through Focal Adhesion-Anchored Intermediate Filaments. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2014, 28, 715–729. [Google Scholar] [CrossRef] [PubMed]
  107. Jiu, Y.; Peränen, J.; Schaible, N.; Cheng, F.; Eriksson, J.E.; Krishnan, R.; Lappalainen, P. Vimentin Intermediate Filaments Control Actin Stress Fiber Assembly through GEF-H1 and RhoA. J. Cell Sci. 2017, 130, 892–902. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, T.; Ghamloush, M.M.; Aldawood, A.; Warburton, R.; Toksoz, D.; Hill, N.S.; Tang, D.D.; Kayyali, U.S. Modulating Endothelial Barrier Function by Targeting Vimentin Phosphorylation. J. Cell. Physiol. 2014, 229, 1484–1493. [Google Scholar] [CrossRef]
  109. Fakih, R.; Diaz-Cruz, C.; Chua, A.S.; Gonzalez, C.; Healy, B.C.; Sattarnezhad, N.; Glanz, B.I.; Weiner, H.L.; Chitnis, T. Food Allergies Are Associated with Increased Disease Activity in Multiple Sclerosis. J. Neurol. Neurosurg. Psychiatry 2019, 90, 629–635. [Google Scholar] [CrossRef]
  110. Pedotti, R.; DeVoss, J.J.; Youssef, S.; Mitchell, D.; Wedemeyer, J.; Madanat, R.; Garren, H.; Fontoura, P.; Tsai, M.; Galli, S.J.; et al. Multiple Elements of the Allergic Arm of the Immune Response Modulate Autoimmune Demyelination. Proc. Natl. Acad. Sci. USA 2003, 100, 1867–1872. [Google Scholar] [CrossRef]
  111. Naddafi, F.; Mirshafiey, A. The Neglected Role of Histamine in Alzheimer’s Disease. Am. J. Alzheimers Dis. Other Demen. 2013, 28, 327–336. [Google Scholar] [CrossRef] [PubMed]
  112. Jadidi-Niaragh, F.; Mirshafiey, A. Histamine and Histamine Receptors in Pathogenesis and Treatment of Multiple Sclerosis. Neuropharmacology 2010, 59, 180–189. [Google Scholar] [CrossRef] [PubMed]
  113. Shasby, D.M.; Ries, D.R.; Shasby, S.S.; Winter, M.C. Histamine Stimulates Phosphorylation of Adherens Junction Proteins and Alters Their Link to Vimentin. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002, 282, L1330–L1338. [Google Scholar] [CrossRef] [PubMed]
  114. Dorado, G.; Gálvez, S.; Rosales, T.E.; Vásquez, V.F.; Hernández, P. Analyzing Modern Biomolecules: The Revolution of Nucleic-Acid Sequencing—Review. Biomolecules 2021, 11, 1111. [Google Scholar] [CrossRef] [PubMed]
  115. You, X.; Vlatkovic, I.; Babic, A.; Will, T.; Epstein, I.; Tushev, G.; Akbalik, G.; Wang, M.; Glock, C.; Quedenau, C.; et al. Neural Circular RNAs Are Derived from Synaptic Genes and Regulated by Development and Plasticity. Nat. Neurosci. 2015, 18, 603–610. [Google Scholar] [CrossRef] [PubMed]
  116. Dong, X.; Bai, Y.; Liao, Z.; Gritsch, D.; Liu, X.; Wang, T.; Borges-Monroy, R.; Ehrlich, A.; Serano, G.E.; Feany, M.B.; et al. Circular RNAs in the Human Brain Are Tailored to Neuron Identity and Neuropsychiatric Disease. BioRxiv Prepr. Serv. Biol. 2023, 14, 5327. [Google Scholar] [CrossRef] [PubMed]
  117. Sekar, S.; Liang, W.S. Circular RNA Expression and Function in the Brain. Non-Coding RNA Res. 2019, 4, 23–29. [Google Scholar] [CrossRef]
  118. Zhou, M.; Li, S.; Huang, C. Physiological and Pathological Functions of Circular RNAs in the Nervous System. Neural Regen. Res. 2023, 19, 342–349. [Google Scholar] [CrossRef] [PubMed]
  119. Yu, X.; Liu, H.; Chang, N.; Fu, W.; Guo, Z.; Wang, Y. Circular RNAs: New Players Involved in the Regulation of Cognition and Cognitive Diseases. Front. Neurosci. 2023, 17, 1097878. [Google Scholar] [CrossRef]
  120. Cardamone, G.; Paraboschi, E.M.; Soldà, G.; Cantoni, C.; Supino, D.; Piccio, L.; Duga, S.; Asselta, R. Not Only Cancer: The Long Non-Coding RNA MALAT1 Affects the Repertoire of Alternatively Spliced Transcripts and Circular RNAs in Multiple Sclerosis. Hum. Mol. Genet. 2019, 28, 1414–1428. [Google Scholar] [CrossRef]
  121. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.D.; Gregersen, L.H.; Munschauer, M.; et al. Circular RNAs Are a Large Class of Animal RNAs with Regulatory Potency. Nature 2013, 495, 333–338. [Google Scholar] [CrossRef] [PubMed]
  122. Zeng, X.; Yuan, X.; Cai, Q.; Tang, C.; Gao, J. Circular RNA as an Epigenetic Regulator in Chronic Liver Diseases. Cells 2021, 10, 1945. [Google Scholar] [CrossRef] [PubMed]
  123. Xiao, J.; Joseph, S.; Xia, M.; Teng, F.; Chen, X.; Huang, R.; Zhai, L.; Deng, W. Circular RNAs Acting as miRNAs’ Sponges and Their Roles in Stem Cells. J. Clin. Med. 2022, 11, 2909. [Google Scholar] [CrossRef]
  124. Verduci, L.; Tarcitano, E.; Strano, S.; Yarden, Y.; Blandino, G. CircRNAs: Role in Human Diseases and Potential Use as Biomarkers. Cell Death Dis. 2021, 12, 468. [Google Scholar] [CrossRef] [PubMed]
  125. Salvatori, F.; D’Aversa, E.; Serino, M.L.; Singh, A.V.; Secchiero, P.; Zauli, G.; Tisato, V.; Gemmati, D. miRNAs Epigenetic Tuning of Wall Remodeling in the Early Phase after Myocardial Infarction: A Novel Epidrug Approach. Int. J. Mol. Sci. 2023, 24, 13268. [Google Scholar] [CrossRef] [PubMed]
  126. He, A.T.; Liu, J.; Li, F.; Yang, B.B. Targeting Circular RNAs as a Therapeutic Approach: Current Strategies and Challenges. Signal Transduct. Target. Ther. 2021, 6, 185. [Google Scholar] [CrossRef] [PubMed]
  127. Pisignano, G.; Michael, D.C.; Visal, T.H.; Pirlog, R.; Ladomery, M.; Calin, G.A. Going Circular: History, Present, and Future of circRNAs in Cancer. Oncogene 2023, 42, 2783. [Google Scholar] [CrossRef]
  128. Zhou, Z.; Zhang, J.; Zheng, X.; Pan, Z.; Zhao, F.; Gao, Y. CIRI-Deep Enables Single-Cell and Spatial Transcriptomic Analysis of Circular RNAs with Deep Learning. Adv. Sci. Weinh. Baden-Wurtt. Ger. 2024, 11, e2308115. [Google Scholar] [CrossRef]
  129. Piwecka, M.; Rajewsky, N.; Rybak-Wolf, A. Single-Cell and Spatial Transcriptomics: Deciphering Brain Complexity in Health and Disease. Nat. Rev. Neurol. 2023, 19, 346–362. [Google Scholar] [CrossRef]
  130. Huang, A.; Zheng, H.; Wu, Z.; Chen, M.; Huang, Y. Circular RNA-Protein Interactions: Functions, Mechanisms, and Identification. Theranostics 2020, 10, 3503–3517. [Google Scholar] [CrossRef]
  131. Wang, Y.; Wu, C.; Du, Y.; Li, Z.; Li, M.; Hou, P.; Shen, Z.; Chu, S.; Zheng, J.; Bai, J. Expanding Uncapped Translation and Emerging Function of Circular RNA in Carcinomas and Noncarcinomas. Mol. Cancer 2022, 21, 13. [Google Scholar] [CrossRef] [PubMed]
  132. Lindner, G.; Takenaka, K.; Santucci, K.; Gao, Y.; Janitz, M. Protein-Coding Circular RNAs—Mechanism, Detection, and Their Role in Cancer and Neurodegenerative Diseases. Biochem. Biophys. Res. Commun. 2023, 678, 68–77. [Google Scholar] [CrossRef] [PubMed]
  133. Cardamone, G.; Paraboschi, E.M.; Soldà, G.; Liberatore, G.; Rimoldi, V.; Cibella, J.; Airi, F.; Tisato, V.; Cantoni, C.; Gallia, F.; et al. The Circular RNA Landscape in Multiple Sclerosis: Disease-Specific Associated Variants and Exon Methylation Shape Circular RNA Expression Profile. Mult. Scler. Relat. Disord. 2023, 69, 104426. [Google Scholar] [CrossRef]
  134. Sciaccotta, R.; Murdaca, G.; Caserta, S.; Rizzo, V.; Gangemi, S.; Allegra, A. Circular RNAs: A New Approach to Multiple Sclerosis. Biomedicines 2023, 11, 2883. [Google Scholar] [CrossRef] [PubMed]
  135. D’Anca, M.; Buccellato, F.R.; Fenoglio, C.; Galimberti, D. Circular RNAs: Emblematic Players of Neurogenesis and Neurodegeneration. Int. J. Mol. Sci. 2022, 23, 4134. [Google Scholar] [CrossRef]
  136. Paraboschi, E.M.; Cardamone, G.; Soldà, G.; Duga, S.; Asselta, R. Interpreting Non-Coding Genetic Variation in Multiple Sclerosis Genome-Wide Associated Regions. Front. Genet. 2018, 9, 647. [Google Scholar] [CrossRef] [PubMed]
  137. Cardamone, G.; Paraboschi, E.M.; Rimoldi, V.; Duga, S.; Soldà, G.; Asselta, R. The Characterization of GSDMB Splicing and Backsplicing Profiles Identifies Novel Isoforms and a Circular RNA That Are Dysregulated in Multiple Sclerosis. Int. J. Mol. Sci. 2017, 18, 576. [Google Scholar] [CrossRef]
  138. Hoque, P.; Romero, B.; Akins, R.E.; Batish, M. Exploring the Multifaceted Biologically Relevant Roles of circRNAs: From Regulation, Translation to Biomarkers. Cells 2023, 12, 2813. [Google Scholar] [CrossRef] [PubMed]
  139. Liu, B.; Song, F.; Yang, Q.; Zhou, Y.; Shao, C.; Shen, Y.; Zhao, Z.; Tang, Q.; Hou, Y.; Xie, J. Characterization of Tissue-Specific Biomarkers with the Expression of circRNAs in Forensically Relevant Body Fluids. Int. J. Legal Med. 2019, 133, 1321–1331. [Google Scholar] [CrossRef]
  140. Eslahi, M.; Nematbakhsh, N.; Dastmalchi, N.; Teimourian, S.; Safaralizadeh, R. An Updated Review of Epigenetic-Related Mechanisms and Their Contribution to Multiple Sclerosis Disease. CNS Neurol. Disord. Drug Targets 2023, 22, 381–393. [Google Scholar] [CrossRef]
  141. Munger, K.; Ascherio, A. Understanding the Joint Effects of EBV and Vitamin D in MS. Mult. Scler. Houndmills Basingstoke Engl. 2013, 19, 1554–1555. [Google Scholar] [CrossRef] [PubMed]
  142. Mohammed, E.M. Circular RNA in Multiple Sclerosis: Pathogenicity and Potential Biomarker Development: A Systematic Review. Epigenetics Insights 2023, 16, 25168657231213195. [Google Scholar] [CrossRef]
  143. Yang, R.; Chen, J.; Xu, B.; Yang, B.; Fu, J.; Xiao, S.; Tan, C.; Chen, H.; Wang, X. Circ_2858 Helps Blood-Brain Barrier Disruption by Increasing VEGFA via Sponging miR-93-5p during Escherichia Coli Meningitis. Mol. Ther. Nucleic Acids 2020, 22, 708–721. [Google Scholar] [CrossRef] [PubMed]
  144. Yang, Z.; Huang, C.; Wen, X.; Liu, W.; Huang, X.; Li, Y.; Zang, J.; Weng, Z.; Lu, D.; Tsang, C.K.; et al. Circular RNA Circ-FoxO3 Attenuates Blood-Brain Barrier Damage by Inducing Autophagy during Ischemia/Reperfusion. Mol. Ther. J. Am. Soc. Gene Ther. 2022, 30, 1275–1287. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, X.; Zeng, H.; Lei, L.; Tong, X.; Yang, L.; Yang, Y.; Li, S.; Zhou, Y.; Luo, L.; Huang, J.; et al. Tight Junctions and Their Regulation by Non-Coding RNAs. Int. J. Biol. Sci. 2021, 17, 712–727. [Google Scholar] [CrossRef]
  146. Nelles, D.G.; Hazrati, L.-N. Ependymal Cells and Neurodegenerative Disease: Outcomes of Compromised Ependymal Barrier Function. Brain Commun. 2022, 4, fcac288. [Google Scholar] [CrossRef] [PubMed]
  147. Liao, H.; Chai, Y.; Sun, Y.; Guo, Z.; Wang, X.; Wang, Z.; Wang, Z.; Wang, Z. Hsa_circ_0074158 Regulates the Endothelial Barrier Function in Sepsis and Its Potential Value as a Biomarker. Front. Genet. 2022, 13, 1002344. [Google Scholar] [CrossRef] [PubMed]
  148. Kremerskothen, J.; Teber, I.; Wendholt, D.; Liedtke, T.; Böckers, T.M.; Barnekow, A. Brain-Specific Splicing of Alpha-Actinin 1 (ACTN1) mRNA. Biochem. Biophys. Res. Commun. 2002, 295, 678–681. [Google Scholar] [CrossRef] [PubMed]
  149. Le, S.; Hu, X.; Yao, M.; Chen, H.; Yu, M.; Xu, X.; Nakazawa, N.; Margadant, F.M.; Sheetz, M.P.; Yan, J. Mechanotransmission and Mechanosensing of Human Alpha-Actinin 1. Cell Rep. 2017, 21, 2714–2723. [Google Scholar] [CrossRef]
  150. Bai, Y.; Zhang, Y.; Han, B.; Yang, L.; Chen, X.; Huang, R.; Wu, F.; Chao, J.; Liu, P.; Hu, G.; et al. Circular RNA DLGAP4 Ameliorates Ischemic Stroke Outcomes by Targeting miR-143 to Regulate Endothelial-Mesenchymal Transition Associated with Blood–Brain Barrier Integrity. J. Neurosci. 2018, 38, 32–50. [Google Scholar] [CrossRef]
  151. Feng, Z.; Zhang, L.; Wang, S.; Hong, Q. Circular RNA circDLGAP4 Exerts Neuroprotective Effects via Modulating miR-134-5p/CREB Pathway in Parkinson’s Disease. Biochem. Biophys. Res. Commun. 2020, 522, 388–394. [Google Scholar] [CrossRef] [PubMed]
  152. Gao, Y.; Wu, P.; Ma, Y.; Xue, Y.; Liu, Y.; Zheng, J.; Liu, X.; He, Q.; Ma, J.; Liu, L.; et al. Circular RNA USP1 Regulates the Permeability of Blood-Tumour Barrier via miR-194-5p/FLI1 Axis. J. Cell. Mol. Med. 2020, 24, 342–355. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, R.; Xu, B.; Yang, B.; Fu, J.; Chen, H.; Wang, X. Non-Coding RNAs: The Extensive and Interactive Regulators of the Blood-Brain Barrier Permeability. RNA Biol. 2021, 18, 108–116. [Google Scholar] [CrossRef]
  154. Yang, L.; Han, B.; Zhang, Y.; Bai, Y.; Chao, J.; Hu, G.; Yao, H. Engagement of Circular RNA HECW2 in the Nonautophagic Role of ATG5 Implicated in the Endothelial-Mesenchymal Transition. Autophagy 2018, 14, 404–418. [Google Scholar] [CrossRef] [PubMed]
  155. Qiu, H.; Ni, C.; Jia, C.; Rong, X.; Chu, M.; Wu, R.; Han, B. CircRNA7632 Down-Regulation Alleviates Endothelial Cell Dysfunction in Kawasaki Disease via Regulating IL-33 Expression. Cell Stress Chaperones 2023, 28, 363–374. [Google Scholar] [CrossRef]
  156. Meng, L.; Zhang, Y.; Wu, P.; Li, D.; Lu, Y.; Shen, P.; Yang, T.; Shi, G.; Chen, Q.; Yuan, H.; et al. CircSTX6 Promotes Pancreatic Ductal Adenocarcinoma Progression by Sponging miR-449b-5p and Interacting with CUL2. Mol. Cancer 2022, 21, 121. [Google Scholar] [CrossRef]
  157. Rossi, F.; Beltran, M.; Damizia, M.; Grelloni, C.; Colantoni, A.; Setti, A.; Di Timoteo, G.; Dattilo, D.; Centrón-Broco, A.; Nicoletti, C.; et al. Circular RNA ZNF609/CKAP5 mRNA Interaction Regulates Microtubule Dynamics and Tumorigenicity. Mol. Cell 2022, 82, 75–89.e9. [Google Scholar] [CrossRef]
  158. Zhu, X.; Yang, M.; Zhao, P.; Li, S.; Zhang, L.; Huang, L.; Huang, Y.; Fei, P.; Yang, Y.; Zhang, S.; et al. Catenin α 1 Mutations Cause Familial Exudative Vitreoretinopathy by Overactivating Norrin/β-Catenin Signaling. J. Clin. Investig. 2021, 131, e139869. [Google Scholar] [CrossRef]
  159. Chan, Y.H.; Harith, H.H.; Israf, D.A.; Tham, C.L. Differential Regulation of LPS-Mediated VE-Cadherin Disruption in Human Endothelial Cells and the Underlying Signaling Pathways: A Mini Review. Front. Cell Dev. Biol. 2019, 7, 280. [Google Scholar] [CrossRef]
  160. Sudhir, P.-R.; Lin, S.-T.; Chia-Wen, C.; Yang, S.-H.; Li, A.F.-Y.; Lai, R.-H.; Wang, M.-J.; Chen, Y.-T.; Chen, C.-F.; Jou, Y.-S.; et al. Loss of PTPRM Associates with the Pathogenic Development of Colorectal Adenoma-Carcinoma Sequence. Sci. Rep. 2015, 5, 9633. [Google Scholar] [CrossRef]
  161. Cash, A.; de Jager, C.; Brickler, T.; Soliman, E.; Ladner, L.; Kaloss, A.M.; Zhu, Y.; Pridham, K.J.; Mills, J.; Ju, J.; et al. Endothelial Deletion of EPH Receptor A4 Alters Single-Cell Profile and Tie2/Akap12 Signaling to Preserve Blood-Brain Barrier Integrity. Proc. Natl. Acad. Sci. USA 2023, 120, e2204700120. [Google Scholar] [CrossRef] [PubMed]
  162. Hatanaka, K.; Lanahan, A.A.; Murakami, M.; Simons, M. Fibroblast Growth Factor Signaling Potentiates VE-Cadherin Stability at Adherens Junctions by Regulating SHP2. PLoS ONE 2012, 7, e37600. [Google Scholar] [CrossRef]
  163. Huang, B.; Krafft, P.R.; Ma, Q.; Rolland, W.B.; Caner, B.; Lekic, T.; Manaenko, A.; Le, M.; Tang, J.; Zhang, J.H. Fibroblast Growth Factors Preserve Blood-Brain Barrier Integrity through RhoA Inhibition after Intracerebral Hemorrhage in Mice. Neurobiol. Dis. 2012, 46, 204–214. [Google Scholar] [CrossRef] [PubMed]
  164. Klimaschewski, L.; Claus, P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol. Neurobiol. 2021, 58, 3884–3902. [Google Scholar] [CrossRef]
  165. Kaya, M.; Ahishali, B. Basic Physiology of the Blood-Brain Barrier in Health and Disease: A Brief Overview. Tissue Barriers 2021, 9, 1840913. [Google Scholar] [CrossRef]
  166. Wang, J.; Xu, F.; Zhu, X.; Li, X.; Li, Y.; Li, J. Targeting microRNAs to Regulate the Integrity of the Blood-Brain Barrier. Front. Bioeng. Biotechnol. 2021, 9, 673415. [Google Scholar] [CrossRef] [PubMed]
  167. Bai, Y.; Zhang, Y.; Hua, J.; Yang, X.; Zhang, X.; Duan, M.; Zhu, X.; Huang, W.; Chao, J.; Zhou, R.; et al. Silencing microRNA-143 Protects the Integrity of the Blood-Brain Barrier: Implications for Methamphetamine Abuse. Sci. Rep. 2016, 6, 35642. [Google Scholar] [CrossRef]
  168. Gasparello, J.; d’Aversa, E.; Breveglieri, G.; Borgatti, M.; Finotti, A.; Gambari, R. In Vitro Induction of Interleukin-8 by SARS-CoV-2 Spike Protein Is Inhibited in Bronchial Epithelial IB3-1 Cells by a miR-93-5p agomiR. Int. Immunopharmacol. 2021, 101, 108201. [Google Scholar] [CrossRef]
  169. Tisato, V.; Silva, J.A.; Scarpellini, F.; Capucci, R.; Marci, R.; Gallo, I.; Salvatori, F.; D’Aversa, E.; Secchiero, P.; Serino, M.L.; et al. Epigenetic Role of LINE-1 Methylation and Key Genes in Pregnancy Maintenance. Sci. Rep. 2024, 14, 3275. [Google Scholar] [CrossRef]
  170. Vaccarezza, M.; Papa, V.; Milani, D.; Gonelli, A.; Secchiero, P.; Zauli, G.; Gemmati, D.; Tisato, V. Sex/Gender-Specific Imbalance in CVD: Could Physical Activity Help to Improve Clinical Outcome Targeting CVD Molecular Mechanisms in Women? Int. J. Mol. Sci. 2020, 21, 1477. [Google Scholar] [CrossRef]
  171. Tornese, G.; Iafusco, D.; Monasta, L.; Agnoletto, C.; Tisato, V.; Ventura, A.; Zauli, G.; Secchiero, P. The Levels of Circulating TRAIL at the Onset of Type 1 Diabetes Are Markedly Decreased in Patients with Ketoacidosis and with the Highest Insulin Requirement. Acta Diabetol. 2014, 51, 239–246. [Google Scholar] [CrossRef] [PubMed]
  172. Johannes, C.; Moremi, K.E.; Kemp, M.C.; Whati, L.; Engel-Hills, P.; Kidd, M.; van Toorn, R.; Jaftha, M.; van Rensburg, S.J.; Kotze, M.J. Pathology-Supported Genetic Testing Presents Opportunities for Improved Disability Outcomes in Multiple Sclerosis. Pers. Med. 2023, 20, 107–130. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of BBB structure. As a part of the NVU, the cell-based BBB consists of a monolayer of BMECs that lines the blood flow, supported by brain microvascular pericytes and perivascular astrocytes. TJs and AJs between BMECs strictly limit BBB permeability [created with BioRender.com, accessed on 2 August 2024].
Figure 1. Schematic representation of BBB structure. As a part of the NVU, the cell-based BBB consists of a monolayer of BMECs that lines the blood flow, supported by brain microvascular pericytes and perivascular astrocytes. TJs and AJs between BMECs strictly limit BBB permeability [created with BioRender.com, accessed on 2 August 2024].
Cells 13 01316 g001
Figure 2. Schematic representation of cytoskeleton proteins in healthy and MS BMECs. In healthy conditions, the cortical ring of actin microfilaments supports and stabilizes the cell membrane outwardly, and joins cell–cell TJ (e.g., occludin; JAM) and AJ complexes. MTs, mainly located and packed in the center of the cell, interact with actin microfilaments directly or indirectly through IFs which are located throughout the cytoplasm, with a higher density closer to the nucleus, providing mechanical support and integrity to the cells. In MS, the complex network of IFs and MTs is demolished, along with a loss of actin microfilament organization. Consequently, a breakdown of TJs and AJs occurs, and gaps arise in the intracellular connections, resulting in increased BBB permeability [created with BioRender.com, accessed on 2 August 2024].
Figure 2. Schematic representation of cytoskeleton proteins in healthy and MS BMECs. In healthy conditions, the cortical ring of actin microfilaments supports and stabilizes the cell membrane outwardly, and joins cell–cell TJ (e.g., occludin; JAM) and AJ complexes. MTs, mainly located and packed in the center of the cell, interact with actin microfilaments directly or indirectly through IFs which are located throughout the cytoplasm, with a higher density closer to the nucleus, providing mechanical support and integrity to the cells. In MS, the complex network of IFs and MTs is demolished, along with a loss of actin microfilament organization. Consequently, a breakdown of TJs and AJs occurs, and gaps arise in the intracellular connections, resulting in increased BBB permeability [created with BioRender.com, accessed on 2 August 2024].
Cells 13 01316 g002
Figure 3. Schematic representation of circRNA functions. CircRNAs act as (a) miR and (b) protein sponges suppressing miR and protein downstream functions, (c) transcription regulators facilitating nuclear translocation of transcription factors, (d) protein scaffolds structurally supporting interactions between proteins, and (e) templates for the translation of small proteins [created with BioRender.com, accessed on 2 August 2024].
Figure 3. Schematic representation of circRNA functions. CircRNAs act as (a) miR and (b) protein sponges suppressing miR and protein downstream functions, (c) transcription regulators facilitating nuclear translocation of transcription factors, (d) protein scaffolds structurally supporting interactions between proteins, and (e) templates for the translation of small proteins [created with BioRender.com, accessed on 2 August 2024].
Cells 13 01316 g003
Table 1. List of circRNAs related to the AJ and TJ pathways, TJs, and microtubules.
Table 1. List of circRNAs related to the AJ and TJ pathways, TJs, and microtubules.
circRNA IDProtein TargetRegulationRef.
hsa_circ_0074158CTNNA1Suppressor[147]
hsa_circ_0004188PTPRMSuppressor
hsa_circ_ACTN1_2453ACTN1Suppressor
hsa_circ_0002872PTPRMSuppressor
hsa_circ_0074171CTNNA1Suppressor
hsa_circ_0046813PTPRMSuppressor
hsa_circ_0002913ACTN1Suppressor
hsa_circ_0032321ACTN1Suppressor
hsa_circ_0008194CTNNA1Suppressor
hsa_circ_0007440CTNNA1Suppressor
hsa_circ_0006114PTPRMEnhancer
hsa_circ_0008016FGFR1Enhancer
hsa_circ_PTPRM_4378PTPRMEnhancer
hsa_circ_ACTN1_2450ACTN1Enhancer
hsa_circ_0007644ACTN1Enhancer
hsa_circ_0005564FGFR1Enhancer
hsa_circ_DLGAP4ZO-1, claudin-5, occludinEnhancer[150,151]
hsa_circ_USP1ZO-1, claudin-5, occludinEnhancer[152]
has_circ_HECW2ZO-1, claudin-5, occludinSuppressor[153,154]
has_circ_7632vimentinEnhancer[155]
hsa_circ_Cul2vimentinEnhancer[156]
hsa_circ_2858VEGFAEnhancer[143,153]
hsa_circ_ZNF609CKAP5Enhancer[157]
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

D’Aversa, E.; Salvatori, F.; Vaccarezza, M.; Antonica, B.; Grisafi, M.; Singh, A.V.; Secchiero, P.; Zauli, G.; Tisato, V.; Gemmati, D. circRNAs as Epigenetic Regulators of Integrity in Blood–Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis. Cells 2024, 13, 1316. https://doi.org/10.3390/cells13161316

AMA Style

D’Aversa E, Salvatori F, Vaccarezza M, Antonica B, Grisafi M, Singh AV, Secchiero P, Zauli G, Tisato V, Gemmati D. circRNAs as Epigenetic Regulators of Integrity in Blood–Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis. Cells. 2024; 13(16):1316. https://doi.org/10.3390/cells13161316

Chicago/Turabian Style

D’Aversa, Elisabetta, Francesca Salvatori, Mauro Vaccarezza, Bianca Antonica, Miriana Grisafi, Ajay Vikram Singh, Paola Secchiero, Giorgio Zauli, Veronica Tisato, and Donato Gemmati. 2024. "circRNAs as Epigenetic Regulators of Integrity in Blood–Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis" Cells 13, no. 16: 1316. https://doi.org/10.3390/cells13161316

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