key: cord-0025488-pg84zx56 authors: Zingale, Valeria Domenica; Gugliandolo, Agnese; Mazzon, Emanuela title: MiR-155: An Important Regulator of Neuroinflammation date: 2021-12-22 journal: Int J Mol Sci DOI: 10.3390/ijms23010090 sha: d42e9e9366a8e3d2f57feaf39bf46307f3deac81 doc_id: 25488 cord_uid: pg84zx56 MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression at the post-transcriptional level and that play an important role in many cellular processes, including modulation of inflammation. MiRNAs are present in high concentrations in the central nervous system (CNS) and are spatially and temporally expressed in a specific way. Therefore, an imbalance in the expression pattern of these small molecules can be involved in the development of neurological diseases. Generally, CNS responds to damage or disease through the activation of an inflammatory response, but many neurological disorders are characterized by uncontrolled neuroinflammation. Many studies support the involvement of miRNAs in the activation or inhibition of inflammatory signaling and in the promotion of uncontrolled neuroinflammation with pathological consequences. MiR-155 is a pro-inflammatory mediator of the CNS and plays an important regulatory role. The purpose of this review is to summarize how miR-155 is regulated and the pathological consequences of its deregulation during neuroinflammatory disorders, including multiple sclerosis, Alzheimer’s disease and other neuroinflammatory disorders. Modulation of miRNAs’ expression could be used as a therapeutic strategy in the treatment of pathological neuroinflammation. MicroRNAs (miRNAs) are a large group of small endogenous molecules of singlestrand non-coding RNA, approximately 21-25 nucleotides in length, and their fundamental role is to regulate gene expression at the post-transcriptional level. They act as negative regulators by binding through a specific region (seed) the 3 UTR of a mRNA target and determining its degradation or the inhibition of its translation [1] . The first miRNA was discovered by studying the development of nematode Caenorhabditis elegans with the identification of the developmental regulator LIN-4 [2] . In 2000, the second miRNA let-7 was discovered in C. elegans, and it was found to be preserved in many organisms, including humans, suggesting that this class of small regulatory RNA has a more general role in biology [3] . Gene-expression regulation mediated by miRNAs seems to affect more than 30% of human genes. It has been shown that each miRNA may regulate different mRNA targets and that multiple miRNAs may regulate the same mRNA. This allows us to consider miR-NAs as elements of control of numerous pathways, which regulate fundamental cellular processes, such as cell cycle regulation, cell proliferation, differentiation and apoptosis [4] . It has been shown that alterations in miRNAs expression may be involved in the onset and progression of several diseases, such as cancer, cardiovascular and neurodegenerative diseases [5] . Various studies have documented the importance of miRNAs in inflammatory conditions and in various central nervous system (CNS) pathological conditions, including neuroinflammation, neurodegeneration and autoimmune diseases [6] . Given the importance of immune-inflammatory processes in neurological diseases, research in recent years has focused on understanding the mechanisms involved and possible therapeutic targets. Degradation of the messenger, which occurs in the case of perfect complementarity between the two sequences [11] . • Inhibition of translation, that occurs in the presence of mismatches between the two sequences [12] . MiRNAs are present in high concentrations in the CNS and are expressed in a spatially and temporally specific way; the gene silencing mediated by these small molecules seems to be fundamental in all the stages concerning the development of brain and the maintenance of homeostasis. Post-transcriptional regulation mediated by miRNAs modulates processes such as neurogenesis, neuronal differentiation, synaptic plasticity, gliogenesis and myelin repair; therefore, an imbalance in the expression pattern of these small molecules can be involved in neurological pathologies [13] , abnormalities of neurological development, neurodegenerative and autoimmune processes [14] . Numerous evidences support the involvement of miRNAs in the modulation of inflammatory signaling within the CNS. Depending on their activity, miRNAs can promote or reduce inflammatory signaling, and in conditions of uncontrolled neuroinflammation, they can aggravate or improve pathological consequences [15] . Then the neuroinflammatory response is the result of the synergistic or antagonistic activity of several miRNAs with pro-inflammatory action (e.g., miR-155, miR-27b and miR-326) and anti-inflammatory activity (e.g., miR-124, miR-146a, miR-21 and miR-223) or by mixed immunomodulators, such as let-7 family [16] . The miRNAs in the neuroinflammatory process can regulate the activation of microglia and astrocytes and also control the activity of peripheral immune cells, such as neutrophils, macrophages, leukocytes and T and B cells [17] . In the context of neuroinflammation, the regulation modes of several miRNAs have been analyzed; among the most important are miR-155, miR-146a, miR-124, miR-21 and let-7 [15] . Some miRNAs modulators of inflammation were evaluated in microglia cultures, demonstrating their role in the inflammatory activation of microglia in CNS. In particular, miR-689, miR-124 and miR-155 were the most strongly associated with pro-inflammatory signaling and microglia activation phenotype. After exposure to lipopolysaccharide (LPS), cultured microglia showed increased expression of miR-155, but reduced expression of miR-689 and miR-124 [18] . Recent studies have revealed that miRNAs are involved in the differentiation of T and B cells, in the signaling of transduction by Toll-like receptors (TLRs) and in the production of cytokines. For example, the expression of miR-146a, miR-155 and miR-132 is altered in response to LPS, Tumor Necrosis Factor (TNF-α), and interleukin (IL)-1β. It has been observed that miR-146 plays an anti-inflammatory role in reducing excessive inflammation, while miR-155 plays a pro-inflammatory role in regulating inflammatory cytokines, such as interferon (IFN)-λ and IFN-β [19] . Their deregulated expression can lead to the uncontrolled proliferation of immune cells and the activation of inflammatory pathways, leading to the development of pathological processes [20] . The term "neuroinflammation" refers to an inflammatory response within the CNS, with the aim of promoting cellular homeostasis in physiological and pathological conditions [21] . Under physiological conditions, the CNS produces pro-inflammatory factors in response to different injuries. In this case, the activation of inflammatory process has a neuroprotective effect and promotes tissue repair [22] . However, in pathological conditions, uncontrolled neuroinflammation can cause tissue damage and become a major component of many neurodegenerative diseases. In fact, the uncontrolled activation of inflammatory processes determine an excessive activation of glial cells, leading to the production of pro-inflammatory cytokines, such as IL-1β, IL-6 and TNF-α; chemokines; secondary messengers, including nitric oxide (NO); prostaglandins; and reactive oxygen species (ROS) [23, 24] . In addition, the activation of inflammatory processes cause the loss of the blood-brain barrier (BBB) and the infiltration of peripheral immune cells [25] . In response to injury, the inflammatory response is activated by glial cells residing in the CNS, particularly microglia and astrocytes. Microglial activation provides the first line of defense when injuries or diseases occurs. Under normal conditions, microglia are in a "quiescent" or "resting" state, where they actively supervise the microenvironment and help maintain cerebral homeostasis [26] . The activation of microglia occurs in the presence of infections and tissue damage and is accompanied by several morphological changes; in fact, they pass from a branched to an amoeboid morphology that allows motility and phagocytosis. Microglia can attack healthy neurons through phagocytosis or secretion of apoptotic factors [23] . Depending on the nature of the signals, activated microglia can differentiate into classical phenotype M1 (pro-inflammatory) or alternative phenotype M2 (anti-inflammatory) [27] . M2 microglia release anti-inflammatory and protective cytokines, such as Transforming Growth Factor β (TGF-β), IL-10, IL-4 and IL-13, which play important roles in wound healing and tissue repair. Conversely, M1 microglia releases an excess of inflammatory mediators, such as ROS, superoxide anion, matrix metalloproteinase (MMP)-9 and pro-inflammatory cytokines, such as TNF-α, IL-6 and IL-1β [28] . Microglia activation and the resulting morphological and functional changes have been observed during almost all neuropathological conditions, including neurodegenerative diseases, infections, strokes, tumors and brain injuries [29] . As a result of uncontrolled processes of neuroinflammation, these cells can remain active for long periods, contributing to neurodegeneration through the continuous release of cytokines and neurotoxic molecules. Inhibition of pro-inflammatory mediators produced by microglial cells could be considered a valid therapeutic approach to reduce the progression of neurodegenerative diseases [30] . Astrocytes are another type of glial cells that play an important role in influencing the inflammatory response of the CNS in case of stress or diseases [31] . Under normal conditions, astrocytes help to maintain homeostasis through interactions with the neuronal signaling system, providing metabolic support, regulating synaptogenesis and through the clearance of neurotransmitters. They also regulate the extracellular space volume and modulate the synaptic plasticity [32] . Astrocytes can promote or reduce neuroinflammation through the release of pro-inflammatory and anti-inflammatory molecules, and acting as functional barriers to the CNS parenchyma [33] . Astrocytes and microglia express different types of receptors, including TLRs, whose activation triggers the neuroinflammatory reaction [34] . Among these, of particular importance is the TLR4, which is considered a key receptor of pro-inflammatory signaling. Its activation induces the release of TNF-α and IL-1β, which activate the inflammatory signaling cascade [30] . Endothelial cells and peripheral immune cells also play a role in the propagation of these inflammatory signals [21] . Neuroinflammation is also regulated through complex signaling cascades between different cell types within the CNS, such as the neurovascular unit (NVU). The NVU is a structure composed of neurons, astrocytes, extracellular matrix and the microvascular endothelial brain, which play a key role in controlling the neuroinflammatory process [35] . This network allows intercommunication between blood vessels and neurons within the CNS and allows regulation of blood flow, brain development, BBB permeability, elimination of toxic by-products and immune surveillance [36] . Thus, neuroinflammation is mediated through the complex interaction between CNS cells and peripheral cells. Understanding the cellular and molecular mechanisms that control neuroinflammation is a field of research that has generated much interest in the discovery of new therapeutic approaches. Among the central regulators of these processes, there are miRNAs, which, if deregulated, may contribute to the progression of the disease, or may reflect a homeostatic attempt of the CNS to prevent and restore normal conditions [15] . Their role in initiating and maintaining neuroinflammation is well reported. MiR-155 is encoded by the MIR-155 host gene, mir155hg, also called the B-cell Integration Cluster (BIC) gene [37] . The BIC gene is composed of three exons within a 13 kb region located in human chromosome 21q21 and produces a pri-miR-155 of 1500 bp transcript in exon 3, which is processed to the mature miR-155. Based on the processing of pre-miR-155 (65 bp), the mature sequence of miR-155-5p or miR-155-3p is obtained [38, 39] . The involvement of miR-155 in inflammatory processes was first suggested in a study of human B-cell lymphoma where its expression was found to be significantly elevated [40] . MiR-155 is highly conserved and plays an important role in the immune system of mammals; numerous studies support the involvement of miR-155 in regulating the differentiation of helper T cells and in the regulation of the response by macrophages through the production of cytokines [41] . In mouse macrophages knockout (KO) for miR-155, a reduction in inflammatory signaling was observed. In addition, there was a 72% reduction in the activity of the inflammatory genes iNOS (inducible nitric oxide synthase), IL-1β and TNF-α after stimulation with IFN-γ + LPS. These data support the role of miR-155 in inflammatory macrophage signaling [42] . Later studies have shown the presence of miR-155 in numerous tissues, including the brain, confirming the pro-inflammatory action of this miRNA in both the peripheral immune system and the CNS [43] . MiR-155 is considered a pro-inflammatory mediator of the CNS and results upregulated in the brain of patients affected by many neurodegenerative diseases. Its action is induced in microglia and macrophages through the nuclear factor κB (NF-κB), after stimulation of the TLR and release of the pro-inflammatory cytokine IFN-γ [19, 44, 45] . MiR-155 induces neuroinflammation through the inhibition of factors involved in the inflammatory process. The involvement of miR-155 determines a reduction of endogenous anti-inflammatory response resulting in increased inflammation. Some targets of miR-155 include anti-inflammatory regulators, such as Suppressor of Cytokine Signaling (SOCS1), a negative regulator of cytokines [43] ; SH2 Domain-Containing Inositol 5 -Phosphatase1 (SHIP1), a negative regulator of TNF-α [46] ; and IL-13 receptor alpha 1 (IL13Rα1) [47] . Cardoso et al. showed the pro-inflammatory role of miR-155 following microglia activation, suggesting that post-transcriptional modulation of SOCS-1 determines the progression of the immune response. SOCS-1 is part of a group of proteins inhibiting the cytokine signal translation pathways. In fact, it plays an important role in regulating immune response through direct inhibition of Janus tyrosine kinase (JAK) and consequent inhibition of signal transducer and transcription factor activator (STAT). In vitro studies in N9 microglia cells exposed to LPS evidenced the upregulation of miR-155 and the reduction in SOCS-1 levels. In addition, anti-miR-155 oligonucleotides decreased the expression of inflammatory cytokines IL-6, IFN-β and TNF-α and the production of NO with subsequent reduction of neuronal death. These results suggest that miR-155 inhibition induces neuronal protection and miR-155 could be a target for controlling neuronal inflammation [43] . In addition, miR-155 is involved in the gene regulation of astrocytes. Activation of these cells induces upregulation of miR-155, which, by inhibiting the mRNA of SOCS-1, determines the high production of pro-inflammatory cytokines [48] . Stimulation of RAW264.7 and THP-1 cells with LPS results in increased miR-155 expression levels correlated with SOCS-1 repression and increased TNF-α and IL-6 production. Treatment with curcumin significantly reduced miR-155 levels and cytokine production, indicating miR-155 as a potential target for suppressing inflammatory responses. Curcumin inhibits miR-155, TNF-α and IL-6 through the PI3K-AKT pathway, suggesting a crucial role of this pathway in the regulation of miR-155 [49] . MiR-155 was one of the first miRNAs to be related to the activation processes of the phenotype M1 of microglia. Resting microglia cells are characterized by low levels of miR-155 expression. On the other hand, in the presence of strong inflammatory stimuli, microglia assume an M1 phenotype, characterized by high levels of miR-155 expression. Upregulation of miR-155 is believed to be crucial for the establishment of this phenotype, since this miRNA acts directly on anti-inflammatory molecules, such as SOCS-1, leading to overregulation of the various inflammatory mediators characteristic of the M1 phenotype [50] . In microglia, miR-155 also acts through the involvement of the transcriptional factor p53 (tumor protein P53), and c-Maf (musculoaponeurotic fibrosarcoma). Moreover, p53 is activated in microglia by ROS, DNA damage or cell stress associated with CNS disease and injury and promotes the expression of the BIC gene coding for miR-155. MiR-155 degrades c-Maf, known for its anti-inflammatory activity, thus promoting pro-inflammatory processes of microglia. The correlation between p53, miR-155 and c-Maf was analyzed in adult murine microglia after induced neuroinflammation through the middle cerebral artery occlusion (MCAO) for 15 min. Two more miRNAs, miR-34a and miR-145, were considered. Activation of p53 induces miR-155, miR-145 and miR-34a. While miR-155 acts on the c-Maf factor, miR-145 and miR-34a act on Twist2 (Twist Family BHLH Transcription Factor 2), an activator of the c-Maf expression. Both pathways downregulate the expression of the c-Maf anti-inflammatory transcription factor [51] . AD is an aging-associated progressive neurodegenerative condition characterized by memory loss, impaired cognitive functions and behavioral changes [52] . It is characterized by the presence of senile plaques caused by extracellular deposits of beta-amyloid peptide (Aβ) and neurofibrillar tangles consisting of accumulations of the hyperphosphorylated protein Tau [53] . Neuroinflammation plays an important role in the pathogenesis and progression of AD. Increased microglia activation has been observed, resulting in the release of inflammatory mediators and progressive neuronal degeneration [54] . The release of ROS by microglia activates the NF-κB dependent signaling pathway, which amplifies the inflammatory response through the production of a large number of inflammatory factors. In addition, activation of the NF-κB pathway induces upregulation of the site 1 amyloid precursor protein splitting enzyme (BACE1) and promotes the production of large quantities of Aβ [55] . Experimental studies concerning miR-155 in AD are summarized in Table 1 . Many studies have focused on the aggregation of Aβ, in particular, Aβ42, which seems to be the main cause of AD development. In AD, the peptide Aβ42 is present in high quantities and initiates the mechanism of polymerization by forming neurotoxic lamellar structures [56] . Anti-miR-155: