Next Article in Journal
Fullerene Rosette: Two-Dimensional Interactive Nanoarchitectonics and Selective Vapor Sensing
Previous Article in Journal
CD24: A Marker for an Extended Expansion Potential of Urothelial Cancer Cell Organoids In Vitro?
Previous Article in Special Issue
Stress-Induced Depression and Alzheimer’s Disease: Focus on Astrocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of miRNAs in Dexmedetomidine’s Neuroprotective Effects against Brain Disorders

by
Codrin-Constantin Burlacu
1,
Maria-Adriana Neag
2,*,
Andrei-Otto Mitre
1,
Alexandru-Constantin Sirbu
2,
Andrei-Vlad Badulescu
1 and
Anca-Dana Buzoianu
2
1
Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
2
Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(10), 5452; https://doi.org/10.3390/ijms23105452
Submission received: 30 March 2022 / Revised: 10 May 2022 / Accepted: 11 May 2022 / Published: 13 May 2022
(This article belongs to the Special Issue Molecular Mechanisms and Pharmacological Targeting of Neuroprotection)

Abstract

:
There are limited neuroprotective strategies for various central nervous system conditions in which fast and sustained management is essential. Neuroprotection-based therapeutics have become an intensively researched topic in the neuroscience field, with multiple novel promising agents, from natural products to mesenchymal stem cells, homing peptides, and nanoparticles-mediated agents, all aiming to significantly provide neuroprotection in experimental and clinical studies. Dexmedetomidine (DEX), an α2 agonist commonly used as an anesthetic adjuvant for sedation and as an opioid-sparing medication, stands out in this context due to its well-established neuroprotective effects. Emerging evidence from preclinical and clinical studies suggested that DEX could be used to protect against cerebral ischemia, traumatic brain injury (TBI), spinal cord injury, neurodegenerative diseases, and postoperative cognitive disorders. MicroRNAs (miRNAs) regulate gene expression at a post-transcriptional level, inhibiting the translation of mRNA into functional proteins. In vivo and in vitro studies deciphered brain-related miRNAs and dysregulated miRNA profiles after several brain disorders, including TBI, ischemic stroke, Alzheimer’s disease, and multiple sclerosis, providing emerging new perspectives in neuroprotective therapy by modulating these miRNAs. Experimental studies revealed that some of the neuroprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, such as lipopolysaccharides induced neuroinflammation, β-amyloid induced dysfunction, brain ischemic-reperfusion injury, and anesthesia-induced neurotoxicity models. This review aims to outline the neuroprotective mechanisms of DEX in brain disorders by modulating miRNAs. We address the neuroprotective effects of DEX by targeting miRNAs in modulating ischemic brain injury, ameliorating the neurotoxicity of anesthetics, reducing postoperative cognitive dysfunction, and improving the effects of neurodegenerative diseases.

1. Introduction

Brain injury refers to a multitude of conditions, which are often life-threatening, lead to chronic disability, and subsequently high socioeconomic costs, while therapeutic strategies often have limited effectiveness. Of this wider category, the most debilitating are traumatic and ischemic brain injuries, with neuroprotective agents most commonly used in this setting [1]. Neurodegenerative disorders including Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD), which are responsible for long-term disability and cognitive impairment, have become a growing cause of morbidity and mortality in both elderly and young patients [2,3]. Neuroinflammatory processes contribute to the initiation and progression of these neurodegenerative diseases, as well as to neurodegeneration and postoperative cognitive impairments related to surgical stress and anesthetics toxicity [4,5,6].
Neuroscience confronts with limited neuroprotective strategy for multifold CNS conditions, for which a fast and sustained management, initially established is essential. Neuroprotection-based therapeutics have become an intensively researched topic in the neuroscience field, with multiple novel promising agents, from natural products to mesenchymal stem cells, homing peptides, and nanoparticles-mediated agents, all aiming to significantly provide neuroprotection in experimental and clinical studies [7,8,9,10]. Despite a growing understanding of this process, the vast majority of agents that showed promising effects in preclinical studies have failed to demonstrate a clear clinical benefit [11].
In this context, dexmedetomidine (DEX), typically used as a sedative, anesthetic adjuvant, and opioid-sparing medication, stands out due to its established neuroprotective effects, which seem to translate well from animal models to a clinical setting [12,13,14]. While its applications in anesthesiology and pain management are consistent with its pharmacological activity as a selective α2-adrenoceptor agonist, the precise mechanisms of DEX-induced neuroprotection are mostly unclear. In addition to the neuroprotective effects of its sympatholytic, α2-agonist, activity, several neuroprotective mechanisms confirmed in animal brain disease models involve anti-inflammatory effects through the inhibition of TLR4/NF-κB, JAK2-STAT3, activation of PI3K/Akt and ERK1/2 pathways, and inhibition of apoptosis and autophagy through hypoxia-inducible factor 1-α (HIF-1α) upregulation [12,15,16,17,18,19].
MicroRNAs (miRNAs), small (18–22 nucleotides), non-coding ribonucleic acid (RNA) molecules, regulate gene expression at the post-transcriptional level, by interacting with the 3′ UTR of mRNAs, thus silencing the translation of mRNA into functional proteins [20,21]. miRNAs regulate a variety of biological processes in a cell-specific manner, including stem cell proliferation and cell differentiation, apoptosis, as well as malignancy, cardiovascular disease, neurodegeneration, and autoimmunity [22,23].
In vivo and in vitro studies deciphered brain-related miRNAs and dysregulated miRNA profiles after several brain disorders, including ischemic stroke, AD, MS, and brain tumors, emerging new perspectives in neuroprotective therapies by miRNAs modulation [24,25,26,27].
Experimental studies revealed that some of the neuroprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, such as neuroinflammation induced by lipopolysaccharides (LPS), β-amyloid, brain ischemic-reperfusion injury and anesthesia-induced neurotoxicity models [28,29,30,31].
On the other hand, miRNAs can greatly influence drug response, either by modulating pharmacokinetics and pharmacodynamic, or by influencing the pathological process itself [32]. In the case of DEX, multiple clinical studies revealed changes in miRNA expression upon DEX treatment, associated with different pharmacology features of DEX [33].
An in-depth understanding at a molecular level of the mechanistic data of DEX in the context of neuroprotection and the variability of DEX response will further provide new perspectives on neurotherapeutic strategy. This review aims to outline the neuroprotective mechanisms of DEX in brain disorders by modulating miRNAs, alongside the miRNAs involved in the pharmacological profile of DEX. We address the neuroprotective effects of DEX by targeting miRNAs in modulating ischemic brain injury, ameliorating the neurotoxicity of anesthetics, reducing postoperative cognitive dysfunction, and improving the effects of neurodegenerative diseases.

2. Pharmacological Features of Dexmedetomidine

DEX is the S-enantiomer of medetomidine, a sedative agent used in veterinary medicine [34]. It was first approved as an FDA drug in 1999 and was used as a sedative in patients in the intensive care unit (ICU). Subsequently, in 2011, DEX was approved by the European Union. Nowadays, this substance is a well-known drug used for sedation and anesthesia [35].
It acts as an agonist with a high affinity for α2 adrenergic receptors (AR) [36]. Like clonidine, DEX is an imidazole derivate, but has a higher selectivity to α2 receptors than this [34,37]. There are three types of α2AR (α2A, α2B, α2C) that are important targets in the treatment of pain or diseases such as high blood pressure, hyperactivity disorder, etc. [38].
More than 90% of the DEX administered is protein bound. DEX is metabolized in the liver by hydroxylation and glucuronidation by CYP2A6, UGT2B10, and UGT1A4. The resulting metabolites are considered inactive, being eliminated mainly renally and a small percentage in the feces [39]. Notably, the pharmacokinetics of dexmedetomidine is not influenced by patients’ renal function, allowing DEX anesthesia in patients with renal failure [40,41].
The sedative effect depends on the plasma concentration. At low plasma concentrations (0.2–0.3 ng/mL) DEX causes mild/moderate sedation, while at high levels (1.9 ng/mL) it is responsible for deep sedation [39].
Drug interactions may occur when DEX is used concomitantly with other medicines. Butorphanol, as an opioid analgesic, exerts antinociception and sedation via acting on MORs, KORs, and DORs, but when used concomitantly with DEX, could suggest a synergistic antinociception and sedation activity in the treatment of acute nociceptive pain [42]. Parecoxib, a cycloxygenase-2-inhibitor, commonly used as a perioperative clinical anesthetic agent in combination with DEX, is rapidly converted in Valdecoxib, its active metabolite [43,44,45]. As DEX inhibits cytochrome P450 and parecoxib’s metabolism depends on CYP3A4 and CYP2C9, cotreatment with DEX and parecoxib will further lead to an inhibition on valdecoxib’s metabolism, its active metabolite, and subsequent increased exposure time to valdecoxib [46].
Alongside propofol and midazolam, DEX is widely used for sedation of critically ill patients who are undergoing mechanical ventilation. Compared to these, DEX does not act through gama aminobutyric acid (GABA) pathway like propofol and midazolam [36]. DEX has sedative and analgesic effects with a different mechanism of action compare with opiates, a well-known class of drugs with analgesic effects [47].
One of the major advantages of DEX compared with GABA receptor agonists is its safety profile against respiration depression [48]. Another advantage of DEX is that if it is given concomitantly with opiates it reduces the required dose of opiates in ICU patients [49].
Among α2-agonists, DEX has shown to have a greater efficacy than clonidine as premedication for bupivacaine spinal block and rarer hemodynamic adverse effects (hypotension and rebound hypertension) than clonidine for the sedation of ICU patients requiring endotracheal intubation [50,51].
Multiple clinical studies evidenced the potential of perineural DEX, used in spinal anesthesia and brachial plexus block, to prolong the motor and sensory block, as well as time to first analgesic request, suggesting DEX as an alternative to opioids [52].
The main side effects of DEX are hypotension and bradycardia. They often have no clinical impact and do not require treatment. However, if treatment is needed, dose reduction of DEX, and administration of fluids or vasopressors are recommended [53]. In a meta-analysis including 516 patients from nine clinical trials, Abdallah et al., have been shown reversible bradycardia, with no hypotension and respiratory depression, after intrathecally or peripherally infusion of DEX [52].
The main effects of DEX are mentioned in Table 1.
The beneficial effect of DEX is dose-dependent. For example, after general anesthesia, there are many unwanted symptoms such as high blood pressure, tachycardia, agitation, shivering, or coughing. Thus, different doses of DEX (1 µg/kg, 0.5 µg/kg, and 0.25 µg/kg), administered in bolus at the end of the surgery, were tested to observe the reduction of the above-mentioned symptoms. The results showed that a higher dose (1 µg/kg) significantly decreases all symptoms, the middle dose (0.5 µg/kg) was effective, except for cough, and the lower dose (0.25 µg/kg) caused hypotension, but did not delay extubation [65].
Multiple disease models revealed a different miRNA expression profiling upon DEX treatment, suggesting the role of miRNAs in the organ-protective effects of DEX (Table 2). Therefore, some of the cytoprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, neuroinflammation induced by LPS and β-amyloid, lung injury in chronic obstructive pulmonary disease, and ischemia/reperfusion injury in the myocardium and liver [28,29,66,67,68].
We performed KEGG pathway analysis using miRNet and identified the following pathways as being influenced by the upregulated miRNAs upon DEX treatment: Neurotrophin signaling pathway (adjusted p-value = 3.27 × 10−13), T cell receptor signaling pathway (adjusted p-value = 1.73 × 10−9), MAPK signaling pathway (adjusted p-value = 7.37 × 10−9), p53 signaling pathway (adjusted p-value = 1.25 × 10−7), Chemokine signaling pathway (adjusted p-value = 6.3 × 10−7), apoptosis (adjusted p-value = 5.4 × 10−6), Jak-STAT signaling pathway (adjusted p-value = 6.19 × 10−6), Toll-like receptor signaling pathway (1.72 × 10−5), axon guidance (adjusted p-value = 2.35 × 10−5), VEGF signaling pathway (adjusted p-value = 7.38 × 10−5), mTOR (adjusted p-value = 1.64 × 10−4), Wnt signaling pathway (adjusted p-value = 6.93 × 10−4), TGF-beta signaling pathway (adjusted p-value = 2.6 × 10−3); Leukocyte transendothelial migration (adjusted p-value = 7.5 × 10−3), NOD-like receptor signaling pathway (adjusted p-value = 9.56 × 10−3), and Natural killer cell mediated cytotoxicity (adjusted p-value = 1.2 × 10−2).
According to KEGG pathway analysis, the common role in the DEX-upregulated miRNAs is represented by anti-inflammatory, anti-apoptotic, and oxidative stress-suppressing signaling pathways, which provide neuroprotection in brain injury models. The neuroprotection effects of DEX by influencing miRNAs are mediated by all these pathways retrieved by KEGG pathway analysis and will be further described in this review.
Some of the most essential miRNAs listed in Table 2, which are involved in DEX-induced neuroprotection are represented by miR-7-5p, miR-214-5p, miR-29a-3p, and miR-381. They directly mediate neuroprotective effects of DEX and also strengthen the beneficial effects of DEX in different brain disease models. The other microRNAs pose an indirect role in DEX-induced neuroprotection, indirectly mediating several neuroprotective pathways.

3. Evidence of Dexmedetomidine as a Neuroprotective Agent

Even if DEX has been successfully used as an agent for sedation and anesthesia, more and more studies are pointing towards its potential use as a neuroprotective agent. Its role in neuroprotection has been underlined in several experimental and clinical studies, and even though there are several explanations for the mechanisms that are not perfectly understood. The neuroprotective effects of DEX have been ascertained in multiple animal models of brain injury, such as ischemic brain injury, traumatic brain injury (TBI), neurodegenerative diseases, and postoperative cognitive disorders [13].
The main neuroprotective mechanism, resulted from the intrinsic sympatholytic activity of DEX as α2-agonist, consisting of negatively regulation of catecholamine releasing, by activation of α2AR in locus coeruleus, thus hampering cerebral vascular spasms and brain damage [84].
Studies have so far observed that DEX reduces pro-inflammatory cytokines. Previous experimental studies have proven that it reduces interleukin 6 (IL-6) release through Interleukin 1-β (IL1-β) suppression [85,86]. DEX also attenuates neuroinflammation through the cholinergic anti-inflammatory pathway, enhancing the discharge of the cervical vagus, thus decreasing the systemic cytokine levels [87]. Another neuroprotective effect is proceeded by reducing pro-inflammatory effects of TNF-α and nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) [88]. By binding to α2 receptors of neurons, DEX reduced the release of catecholamines and modulated receptors’ sensitivity and release of glutamate. Glutamate release is also reduced by the suppression of voltage-dependent calcium channels but also by activated protein kinase activity [89]. DEX also enhances the expression of the excitatory amino acid transporters 1 and 3, glutamate transporters that remove glutamate from the synaptic cleft and extra synaptic sites by reuptake in neurons and glial cells [90,91]. Otherwise, DEX could inhibit extracellular glutamate by hampering the response of glutamate channels to depolarization and could also inhibit the neurocyte’s absorption ability of glutamate [84].
In response to apoptosis, DEX modulates pro and anti-apoptotic proteins, including Bcl-2 and Bcl-xl [92]. Neuronal hypoxic damage can cause adaptive reactions by increasing the HIF-1α and the vascular endothelial growth factor (VEGF) to protect the neuronal cell from oxygen and glucose depravation (OGD). By binding to the I2 imidazoline receptor, DEX upregulates the expression of both, VEGF and HIF-1α, promoting neuroprotective effects [93]. Other neuroprotective mechanisms of DEX consist of anti-inflammatory effects by inhibiting TLR4/NF-κB, JAK2-STAT3 pathways, and anti-apoptotic effects by activating PI3K/Akt, ERK1/2 pathways, and anti-apoptotic and -autophagic effects through HIF-1α upregulation [12,15,16,17,18].
Besides the experimental evidence that supports the neuroprotection of DEX, there are also several clinical studies that confirm its benefits in the clinical settings. In clinical randomized trials, DEX provided neuroprotection in septic patients, improving significantly the mortality rates up to 70% compared to lorazepam sedated patients, and has also reduced the incidence and duration of delirium in elderly ICU patients, candidates for non-cardiac surgeries, compared to placebo [94,95,96]. These might be explained by the different immune-modulatory profiles of anesthetic agents. Compared to benzodiazepines and midazolam, which impaired neutrophils and macrophages activity, DEX exerts stimulatory effects on macrophage phagocytosis, with no influence on neutrophil activity, thus exerting protective effects [97,98]. Moreover, in the prophylaxis of postsurgical delirium in elderly patients after coronary artery bypass grafting, the DEX treatment group had lower mortality, a shorter ICU stay, and a lower opioid consumption [99].
In 160 patients undergoing elective cranial surgery, DEX decreased postoperative neurological complications and postoperative delirium. or saline combined with goal-directed hemodynamic therapy to evaluate the neuroprotective effect [100]. DEX sustain recovery of cognition functions in 49 patients after carotid endarterectomy, lowering cerebral neuroinflammatory markers, IL-6, TNF-α, lactate content, and increasing brain-derived neurotrophic factor (BDNF) levels after 24 h of the procedure [101]. Blood samples of 60 glioma patients who underwent craniotomy revealed a decreased level of IL-6, TNF-α, neuron-specific enolase, S100 β, malondialdehyde, and increased level of superoxide dismutase after DEX pretreatment, suggesting the anti-inflammatory and anti-oxidative role of DEX against brain damage [102].

4. Short Overview of miRNAs as Therapeutic and Diagnostic Tools in Brain Disorders

In the recent years, increasing evidence highlights the regulatory roles of miRNAs behind protein expression in developmental processes such as: stem cell division, cell differentiation, proliferation, and apoptosis, defining new mechanisms involved in different pathological conditions, including cancer, cardiovascular, neurodegenerative diseases, immune system disorders [22,23].
The first well-documented disease, where miRNAs have been shown to play a regulatory role was chronic lymphocytic leukemia, with the extension of the area of experimental research in multiple pathological settings: cancer, cardiovascular, autoimmune disorders, and neurological disorders [22,103]. Extracellular miRNAs have been found in a remarkably stable state in serum or plasma, resistant to RNase and unfortunate physical conditions due to different carriers such as microvesicles and exosomes that mediate miRNA transport [104,105]. Thus, they might serve as potential clinical biomarkers, for multiple conditions including cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases [106].
In neuroscience, miRNAs gained attention in neurological disorders as emerging new promising biomarkers to estimate the progression and extension of multiple disorders, as well as new therapeutic strategies aimed to protect the brain and sustain recovery from brain damage [107,108]. Certain blood miRNAs emerged as biomarkers for ischemic stroke, intracerebral hemorrhage, and seizures, such as: miR-210, -125a-5p, -miR-155, and miR-362-3p, -125b-5p, miR-143-3p, -miR-298 [109,110,111].
Ponnusamy et al., deciphered the role of miRNAs involved in normal brain development, including the neurodevelopment processes of myelination and synaptogenesis, vasculo-genesis, angiogenesis then continuing with normal functioning processes of the brain of synaptic plasticity and memory [112].
Following hypoxic/ischemic injury, miRNAs modulate a chain of inflammatory responses, leading to microglial activation, cytokine production, apoptosis, energy balance, and cell apoptosis; all these events are influenced by miRNAs [24].
MiRNAs dysregulation profile of miRNAs has been ascertained in ischemic brain injury, TBI, neurodegenerative diseases, i.e., AD, HD, PD, prion diseases, brain tumors, cerebral infections, and hypoxic-ischemic encephalopathy [113,114]. As key mediators in multifold pathological conditions, miRNAs have been widely exploited as theragnostic tools in cerebral ischemia, further providing a promising neuroprotective strategy in ischemic stroke. Using microarray and stem-loop Real-Time PCR analysis to measure miRNA expression, of the 157 miRNAs found to be regulated in 19 patients with ischemic stroke, 138 miRNAs exhibited upregulated levels and 19 miRNAs exhibited downregulated levels [115].
MiRNAs expression profiling has been shown to change significantly in a time-dependent manner. For example, miR-290 and miR-494 exhibited differential changes in their expression at 24 and 48 h after the stroke event, while miR-150, -195, and -320 reversed their expression profiling compared to the previous ones. The number of dysregulated miRNAs decreased significantly from 106 after 24 h of stroke induction to 82 after 48 h of stroke event, suggesting the dynamic of miRNAs expressing profiling in promoting neuroprotective mechanisms of the ischemic brain [116]. The level of miR-223-3p, miR-155-5p, miR-3473, and miR-448-5p in the blood serum, liver, and spleen, have been changed in time-dependent manner in ischemic stroke rat model, with differential expressed miRNAs in the brain between different anatomical areas [117].
MiRNAs biosynthesis complexes are influenced by pathological conditions. For instance, Ago and Dicer enzymes have been shown to be upregulated upon ischemic insult after 24 h and Dicer, XPO5, and TRBP increased in the spleen after 72 h [117]. Moreover, miRNA–RISC complex configurations are modulated by ischemic/hypoxic conditions [118].
Understanding the involvement of miRNAs in the complex array of a neuropathophysiological chain of reactions reveals new potential therapeutic targets of miRNAs in brain injuries. Using bioinformatics analysis, Chenjing et al., mapped the dysregulated profile of miRNAs in AD, consisting of 27 dysregulated miRNAs, involved in pathogenesis processes of AD, i.e., amyloidogenesis, inflammation, neurite differentiation, degradation of neurons, and neuronal survival and proliferation [119]. In preclinical models of intracerebral hemorrhage, several miRNAs have been related to beneficial neuroprotective effects, such as miR-223,-7, -let-7a, -23b, -126-3p, -132, -140-5p, -146a, -152, -181c, -183-5p and -194-5p, whereas others have been associated with deleterious effects, including miR-222, -494, -23a-3p both acting on neuroinflammatory signaling pathways [120].
MiRNAs-based agents—agomirs and antagomirs—chemically modified oligonucleotides modulate target miRNAs, by replacing or silencing miRNAs, depending on the ‘regulation’ state of these miRNAs. In this way, we can replace a low concentration of a beneficial miRNA or inhibit a highly expressed miRNA related to deleterious effects, using these agents [107,121]. Thus, miR-153 and miR-205 have been identified as potential therapeutic targets in PD, as miR-153 and miR-205 mimic targeting a-synuclein and LRRK2 protein in PD [122,123].
Otherwise, blocking the expression of some miRNAs involved in detrimental effects of brain injury processes could be used as a therapeutic strategy [124]. For instance, intracerebroventricular administration of anti-miR-103-1 in the ischemic stroke rat model upregulates the expression of Na+/Ca2+ exchanger, which was related to the maintaining of ionic homeostasis in the ischemic brain [125]. MiR-181a antagomir provides a new promising therapeutic strategy in stroke patients for long-lasting recovery of motor function and coordination.
Notably, the miRNAs expression profiling changes during pathological events depending on the intensity of the injurious stimulus. For instance, miR-181a expression level was upregulated in ischemic stroke tissue, increased levels being associated with increased injury and oxidative stress [126]. In the penumbra zone of ischemic stroke, miR-181a has been shown to be downregulated, thus protecting neurons and glial cells [127]. By the same mechanism, ischemic/hypoxia preconditioning downregulated miR-181a expression in oxygen-glucose deprivation/reperfusion (OGD/R) neuron model and MCAO rat model, protecting cerebral tissue against ischemia/reperfusion (I/R) injury, by inhibiting apoptosis and pyroptosis of neurons and enhancing the protective function of neuronal mitochondria [127].

5. Regulation Production of miRNAs after Dexmedetomidine Treatment

5.1. miRNA Biogenesis

The multistep process of miRNA biogenesis begins with DNA transcription of the miRNA-coding genes from introns of protein-coding genes and intergenic miRNAs regions. This will further lead to pri-miRNA transcript, and the formation of pre-miRNA [21,128]. Then, the pre-miRNA is exported into the cytoplasm and processed by RNase III Dicer, producing the mature miRNA duplex [129]. Formation of the RNA-induced silencing complex (RISC), the silencing inductors of mRNA, requires loading the miRNA duplex into the Argonaut (AGO) family of proteins [21,130]. By base pairing the 5′ end with 3′ untranslated regions (3′-UTRs) of mRNAs, 7-methylguanosine from mRNA complex interacts with Ago proteins from the RISC complexes of miRNAs, inducing miRNA-mediated gene silencing [131,132]. The formation process of miRNAs is mediated by specific enzymes: RNA polymerase II, micro-processor complex (Ribonuclease III enzyme, Drosha, and DGCR8), Exportin 5 transporter, RNase III Dicer, and Ago2 [21,133]. Depending on what region has bound, miRNAs could induce both silencing gene expression, by binding to 5′ UTR or coding regions or inducing transcription by the interaction with the promoter region [134,135].

5.2. DEX-Dependent Transcription Controlling Systems of the miRNAs

MiRNAs biosynthesis complexes are influenced by several pathological conditions. Under hypoxic conditions, manifold mechanisms that regulate miRNA biogenesis machinery are triggered at different levels: transcription, maturation, and function levels [136].
Different molecular mechanisms might be involved in DEX-induced signals on miRNAs biogenesis. Hypoxia, which contributes to the pathogenesis of ischemic-hypoxic brain injuries, neuroinflammatory diseases, and neurodegenerative diseases, has a direct impact on enzymatic complexes of miRNA biogenesis [136,137]. By inducing the expression of HIF, p53, and NF-kB pathway, hypoxia could stimulate miRNA transcription, thus influencing the expression level of miRNAs [136]. Multiple anti-inflammatory effects of DEX proceed via regulating the NF-kB signaling pathways and EGR1/p53 pathway [70,74]. By modulating NF-kB and p53-associated pathways, DEX might interfere with the synthesis process of the altered brain-related miRNA in different brain disorders associated with hypoxic conditions. Drosha, DRG8 complexes are suppressed under hypoxia, thus inhibiting the cleavage of pri-miRNA into pre-miRNA [117,136]. Moreover, miRNA–RISC complexes are stimulated by hypoxia, leading to mRNA degradation and translation repression in brain injuries [136]. Therefore, DEX might counteract the inhibitory or stimulatory effects of hypoxia on miRNA biogenesis complexes, which might lead to favorable effects in brain disorders. α and β AR activate transcription of IL-6, and TNF-α in vivo astrocyte cultured models of rat spinal cord, suggesting the potential of adrenoceptors to induce transcription activation of different factors [138,139]. Dexmedetomidine poses transcriptional activation roles in astrocyte cultures, increasing TNF-α mRNA, which leads to JNK and ERK phosphorylation [139].
DEX, α2 AR agonist, by binding to G-protein-coupled receptors (GPCRs) potentiates guanine-nucleotide regulatory binding proteins (G inhibitory protein), which leads to decreased cAMP level. [13] By affecting GPCRs, DEX might influence the miRNA expression, by controlling the transcription systems of miRNA.
Multiple studies revealed that miRNAs are affected after stimulating G-protein-coupled receptors (GPCRs), several miRNAs acting as downstream targets of GPCRs [140]. In breast cancer and hepatocarcinoma, miR-144 is regulated by GPER via modulating PI3K/ERK1/2/Elk1 pathway [141]. miR-148a acts as a bidirectional modulating molecule, as E2-GPER downregulates miR-148a and miR-148 downregulates HOTAIR, non-coding RNA [142].
The controlling systems that affect different steps in miRNA synthesis, maturation, and function after DEX treatment are not fully understood. The research studies mentioned in this review are focused on the level of expression of miRNAs after DEX administration, specific mRNA targets of miRNA, and associated biological effects.
More research studies to depict the biological systems that affect the up- and down-regulation of miRNAs in response to DEX treatment will be needed.

6. Translation Relevance of miRNA in Dexmedetomidine-Mediated Neuroprotection

6.1. Neuroprotective Profile of Dexmedetomidine in Brain Disorders

Over the last years, the exploring of multiple neuroprotection agents for targeting brain damage tissue in experimental animal studies, turn out unsuccessfully from bench to bedside in brain disorders research [143]. Emerging evidence from preclinical and clinical studies demonstrated the beneficial use of DEX as a neuroprotective agent against cerebral ischemia, TBI, spinal cord injury, neurodegenerative diseases, and postoperative cognitive disorders [13].
Furthermore, emerging preclinical studies have confirmed the hypothesis that DEX regulates miRNA expression in brain injuries (Figure 1). An in-depth understanding of the miRNA expression profile upon DEX administration is essential for emerging new circulatory biomarkers to estimate the neuroprotective potential of DEX, as well as developing new miRNAs-based therapeutics, which could enhance DEX’s pharmacological profile in protecting the brain.

6.2. Modulation of Ischemic Brain Injury

DEX activation could affect miRNA-mediated neuropathological responses under ischemic conditions by regulating direct or indirect pathways. There are several hypotheses suggesting that DEX treatment can affect miRNA-mediated neuroinflammatory and apoptotic responses, including the circ-CDR1as/miR-28-3p/TRAF3 pathway in mouse hippocampal neuronal cells exposed to H/R insult, miRNA-214/ROCK1/NF-κB pathway in rat cerebral ischemia-reperfusion injury (CI/RI), SNHG11/miR-324-3p/VEGFA and miRNA-10b-5p/BDNF pathway in MCAO ischemic rat model and neuron cell induced to OGD/R dysfunction.
As an endogenous RNA, which competes with miRNAs for the post-transcriptional regulation role, circ-CDR1as mediate cell viability and apoptosis, being involved in numerous nervous system conditions, including cerebral I/R, PD, and AD [144,145,146,147]. TRAF3 belongs to the TRAF adaptor protein family, regulating several cellular activities in multifold disorders and promoting apoptosis and inflammatory responses in vivo and vitro models of I/R [148]. Treatment with DEX after hypoxia-reperfusion injury induces neuronal protection by up-regulating miR-28-3p expression, and attenuating circ-CDR1as expression, thus reversing inflammatory responses and apoptotic rate in hippocampal neurons. Moreover, by upregulating miR-28-3p, DEX could mitigate TRAF3 overexpression, which has been associated with negative responses to brain I/R injury [148].
Liu et al., demonstrated the therapeutic effects of DEX in protecting neurons against ischemic damage by overexpressing miR-214, thus disrupting ROCK1 expression and dampening of NF-κB pathway, which are responsible for neuroinflammatory responses [149]. The level of miR-205-5p has been found to be low-expressed in a rat I/R injury model after MCAO and cultured hippocampal cells exposed to H/R injury [30]. Glutathione levels, altered malondialdehyde levels, and cytokine production have been shown to be attenuated upon DEX treatment [150]. DEX could alleviate neuroinflammation and oxidative stress associated with I/R injury by upregulating miR-205-5p, thereby decreasing HMGB1 expression and increasing anti-oxidative protein levels in rat brains [30].
Both DEX and miRNAs pose important roles in apoptotic signaling pathways by regulating many pro-apoptotic proteins. For instance, DEX negatively regulated cleaved caspase-3 and increased Bcl-2 expression in H2O2-induced dysfunction of PC12 cells [151]. DEX confers protection against hypoxia-induced injury in PC12 cells by inhibiting miR-134, thus reversing the increased level of cleaved caspase-3 and decreased Bcl-2 expression, related to elevated apoptotic rate [151].
Small nucleolar RNA host gene 11 (SNHG11) has been revealed to be significantly expressed in vivo and in vitro models of I/R injury, with its overexpressed level being associated with increased cell apoptosis and reduced cell viability [152]. I/R injury resulted in a low expression level of miR-324-3p, with upregulated SNHG11 and VEGFA levels in OGD/R induced neuronal injury on SH-SY5Y cells [152]. In this context, miR-324-3p targets SNHG11 and VEGFA to strengthen the neuroprotective effects of DEX in OGD/R-induced neuronal injury [152]. By inhibiting miR-199a expression, DEX exerts neuroprotection in rat CI/RI, restoring neuronal structure and improving modified neurological and cognitive functions.
By promoting neural stem cell proliferation and differentiation to neurons miR-381, its regenerative potential in multiple neurological disorders, such as AD, HD, PD, and spinal cord injury is revealed [153]. Interferon regulatory factors 4 and 5 (IRF5, IRF4) regulatory axis modulates microglial pro- and anti-inflammatory responses, corresponding to different cytokine production. IRF4 has been shown to regulate anti-inflammatory pathways related to microglia activation in the ischemic stroke model [154]. Treatment with DEX after MCAO rat models and primary neuron cells exposed to OGD increased miR-381 expression, which was associated with declined IRF4–IL-9 expression, leading to reduced apoptosis rate and improved neurological function [73].
BDNF, a crucial growth factor in the differentiation and survival of neurons during the development stage of the brain, prevented neuronal death in several hypoxic or excitotoxic models in the neonatal animal brain [155,156]. In a mouse model of brain excitotoxicity, DEX and BDNF exerted neuroprotective effects via the ERK1/2 pathway, DEX increasing the astrocyte expression of BDNF by subsequent activation of astrocyte’s α2-adrenergic receptors [155]. Upregulation of miR-10b-5p has been associated with neuronal apoptosis in ischemic brain injury by negatively regulating BDNF expression, with reversing the miR-10b-5p expression upon DEX treatment [157]. The protective mechanism induced by DEX has been mediated by the SNHG16-miR-10b-5p-BDNF pathway [157].
DEX poses important regulatory roles in hypoxic/ischemic injury by regulating the expression of inflammatory and pro-apoptotic factors in the neonatal brain [150,158].
In hypoxic ischemic brain damage (HIBD), MALAT-1 was highly expressed in the mice brain, leading to downregulation of miR-429 and ultimately increased WNT1 expression, which stimulates apoptosis of hippocampal neurons [159]. Otherwise, MALAT-1 silencing enhances the neuroprotective effect of DEX on HIBD by up-regulating miR-429, subsequently leading to the downregulation of WNT1 [159]. DEX elicited neuroprotective effects in neonatal ischemic brain injury by increasing expression of miR-128, co-treatment with both DEX and ago-miR-128 significantly increasing neuroprotection against brain injury and decreasing nerve cell apoptosis rate [83]. Moreover, upregulation of miR-128 promoted by DEX sustained down-regulating of WNT1, which will further lead to a decreased apoptotic rate of neurons [83].
In H2O2-induced neuronal injury, DEX promoted the expression of miR-223-3p and inhibited its target TIAL1. Pretreatment with DEX of neuroblastoma cells exposed to OGD has been proved to inhibit cell apoptosis, and sustain cell proliferation, possibly mediated by suppressing miR-29b [160]. In H2O2-induced injury, DEX protected PC12 cells from oxidative damage by down-regulating miR-199a, thereby promoting an increase in HIF-1α and activating the neuroprotective pathways of PI3K/AKT/mTOR and Wnt/b-catenin pathways [161].
ERK pathway serves important regulatory roles in neuron proliferation, and apoptosis, participating in some of the signaling cascades of DEX to induce neuroprotection [162,163]. According to Zhu et al., downregulation of miR-155, mediated by DEX treatment in OGD/R injury of rat hippocampal neurons, promotes the ERK1/2 pathway and enhances matrix metalloproteinases, thereby inhibiting apoptosis and oxidative stress [164].
Histone Deacetylase 4 (HDAC4) mediates epigenetic mechanisms and acts as transcriptional repressor to maintain normal transcriptional gene transcription. In the brain, HDAC displays neuroprotective effects in AD, and ischemic stroke, and interacting with miRNAs [165]. MiR-29a-3p promotes the neuroprotective effects of DEX on HIBD rats by interfering with neuron inflammation and apoptosis in the hippocampal region, by inhibiting HDAC4 [72].
Maternally expressed gene 3 (MEG3), long noncoding RNA, emerged as a competing endogenous RNA for miRNAs in experimental disease models. MEG3 suppressing, or miR-129-5p upregulation enhanced the neuroprotection effect of DEX on neonatal mice with HIBD, hence reducing neuronal apoptosis rate and sustaining cerebral function recovery [166]. Microglial activation represents the first initiator of neuroinflammation in response to ischemic brain injury, subsequently leading to neuronal damage [167,168]. Moreover, microglial activation might be modulated by miRNAs [24]. In spinal cord ischemia-reperfusion injury, DEX suppressed the activation of microglial cells via upregulation of let-7a-1/2-3p and downregulation of the HMGB1 pathway, model, which has been related to improved nerve motor function [169].

6.3. Decreasing the Neurotoxicity of Anesthetics

In neurotoxicity models of anesthetics, DEX could modulate pro-apoptotic and anti-apoptotic signaling pathways [69]. DEX treatment regulates neuron apoptosis against anesthetics-induced neurotoxicity in multiple toxicity models of sevoflurane, ketamine, and isoflurane, in both, immature and adult brains [170,171,172]. Besides the anesthetic toxicity associated with surgical procedures, oxygen, used mainly in neonatal intensive care, has been associated with disruption of intracellular hemostasis reaction and subsequently activation of oxidative stress and inflammation, which were responsible for long-term cognitive dysfunction and neurodegeneration [150]. In a hyperoxia induced-neurodegeneration model, DEX promotes neuroprotective effects on neonatal rat brain injury by restoring altered glutathione levels and suppressing lipid peroxidation, counteracting deleterious effects of oxidative stress [150].
Furthermore, an alteration in brain-related miRNAs has been shown after anesthetic exposure [173]. Ropivacaine, used as a local anesthetic, could exert neurotoxic effects on neurocyte cells [174]. In a model of ropivacaine-induced neurocyte injury, ropivacaine attenuates miR-381 expression and enhances LRRC expression, inducing apoptotic and anti-proliferative effects on neurocyte cells [31]. Cotreatment with DEX upregulated miR-381, with suppression of LRRC and activation of SDF-1/CXCR4 pathway [31]. In human neuroblastoma SH-SY5Y cells, DEX reversed the apoptotic effect of bupivacaine in a dose-dependent manner by decreasing the levels of pro-apoptosis associated proteins Bax and cleaved caspase-3 and reducing ROS production [69].
Parthanatos, a caspase-independent cell death was linked to the overactivation of PARP-1 [175]. The involvement of parthanatos in brain disorders has been revealed in cerebral ischemia, glioma cells, and other CNS-associated pathological processes [176,177]. To pose the DEX mediated effect against nervous cells parthanatos, a toxicity model of Bupivacaine on human neuroblastoma cells was created [69].
Thus, DEX inhibited bupivacaine-induced PARP1 expression and increased miR-7-5p expression, attenuating the oxidative stress. Moreover, miR-7-5p upregulation protects against parthanatos by stabilizing the mitochondrial membrane potential and antioxidative enzymes, which were disrupted by bupivacaine [69]. Some of DEX’s neuroprotective properties in the neurotoxicity model of sevoflurane- were mediated by miR-330-3p/ULK1 pathway, thus inhibiting cell apoptosis and facilitating mitophagy [178]. MiR-129 has been shown to target Toll-like receptor 4 (TLR4). Up-regulation of miR-129 in SEV-induced post-operative cognitive dysfunction (POCD) rats inhibited TLR4 expression and prevented NF-κB p65 phosphorylation, thus alleviating cognitive impairments in rats, and hindering neuronal apoptosis [70].

6.4. DEX-Mediated Neuroprotection against Postoperative Cognitive Dysfunctions

Several neurodegenerative diseases are linked to neuroinflammation, with a strong correlation between neuroinflammation and increased peripheral proinflammatory cytokines being evidenced [179,180]. During surgery, the release into circulation of a large amount of pro-inflammatory cytokines, e.g., IL1-β and TNF-α, and subsequent activation of the immune system were correlated with neuronal dysfunction and death, leading to postoperative cognitive impairments [76].
Neuroinflammation induced by LPS treatment is followed by changes in the expression profiles of miR 124, 132, 134, and 155 in a time-dependent manner in different tissues, including the hippocampus, cortex, and plasma [76]. Treatment with DEX attenuates LPS-induced upregulation expression of these miRNAs at different time points, with different and fluctuated levels of miRNAs expression [76].
DEX administrated alone could significantly change the expression levels of miRNAs in the brain tissue under normoxic conditions, with more enhanced regulation of miRNAs in the neuroinflammatory context [76]. Therefore, DEX alone promotes the same ‘increasing’ trend in the expression of miR-124, 132, 134, and 155, as in the LPS-induced neuroinflammation model [76]. MiR-155 has been revealed to negatively regulate the BBB in a rat model of LPS-induced neuroinflammation [181]. Otherwise, DEX attenuated disruption of BBB during neuroinflammation [182]. In this context, Paeschke et al., hypothesized a possible involvement of DEX in attenuating BBB disruption by modulating miR-155 [76]. In the blood samples of 40 patients who underwent off-pump coronary artery bypass grafting, circulating miRNAs exhibited downregulated levels of miR-320 after adjunct anesthesia with DEX, with high expression of neuroglobin (NGB), suggesting the role of miR-320/NGB pathways in enhancing the proliferation activity of neuronal cells, associated with reduced POCD [77].
Recent findings provide insight into the neuroprotective and organ-protective mechanism of DEX by restoring the autophagic flux in the spleen, attenuating the PARP cleavage induced by LPS in the spleen, and regulating the cholinergic anti-inflammatory pathway [75]. DEX induces neuroprotection by attenuating LPS-induced upregulation of miR-30a-5p and downregulation of miR-21-5p and miR-204-5p in the hippocampus. Interestingly, DEX did not suppress the alteration of miRNA expression in the cortex induced by LPS, but regulated the expression of miR-204-5p, miR-Let-7a-5p, and miR-21-5p in the spleen [75].
DEX treatment downregulates miR-29b expression, thus affecting cell viability, and inhibiting cell apoptosis in the OGD-induced neuronal injury [160]. Up-regulation of miR-340 expression in primary microglia cultured cells (BV2 microglia cells) under treatment of LPS enhanced the anti-inflammatory effect of DEX via inhibiting the NF-κB pathway. Furthermore, DEX promotes phagocytosis in LPS-induced microglia cell dysfunction [28]. In POCD-operated mice anesthetized with sevoflurane, DEX administration resulted in enhanced miR-381 expression and inhibition of EGR1/p53 pathways, related to decreased rate of hippocampal neuron apoptosis, DNA damage, neuroinflammation, and cognitive impairment [74].

6.5. Modulation of Neurodegenerative Disorders

Neuroinflammation triggered by abnormal protein complexes’ formation represents a critical step in the initiation and progression of neurodegenerative disorders [183]. The neuroprotective therapy aimed to counteract the immunological and neurodegenerative reactions in AD, PD, MS is based on the modulation of microglial cell activation and regulation of the releasing of pro-inflammatory mediators [184].
The anti-inflammatory and immunomodulatory effects of DEX in neurodegenerative disease models proceed by suppressing both cytokine production and microglia activation [185]. MOG-induced animal experimental autoimmune encephalomyelitis (EAE) reproduces infiltration of spinal cord with microglia cells, clinically mimicking MS. In C57BL/6 mouse-induced EAE, DEX reduces microglial infiltration in the spinal cord, by suppressing the expression of CXCR7, a chemokine receptor expressed on the surface of microglia, thus inhibiting chemotaxis of microglia triggered by SDF-1 or I-TAC. By binding to α2 AR, DEX induces desensitization of microglial CXCR7 in the mouse model of EAE. Thus, DEX attenuated the clinical severity of EAE, suggesting that MS could benefit from the immunopharmacological effects of DEX [185].
Inflammasomes, multiprotein complexes responsible for the synthesis of pro-inflammatory cytokines, IL-1β and IL-18, in response to ROS production, modulate the initial stages of neuroinflammation during multiple neurodegenerative diseases (AD, TBI, MS) [186,187]. Nod-like receptor (NLR) family proteins, essential components of the inflammasome, are specific sensors or receptors which recognize multiple damage signals, such as pro-caspase-1, and ASC (the apoptotic speck-like protein containing a caspase recruitment domain) [188,189]. In neuroinflammation models, NLRP inflammasome induces activation of caspase-1, thus releasing IL-1β and IL-18, which are responsible for further recruitment of microglia and astrocyte cells [186,190]. In vivo rat model of rheumatoid arthritis (RA), DEX suppresses NLRC5, therefore reducing cytokine levels of IL-1β, IL-6, IL-17A, and TNF-α. In human RA, DEX inhibits migration, invasion, and inflammation of fibroblast-like synoviocytes by decreasing the NLRC5 level, which will further inhibit NF-κB pathway [191]. Therefore, modulation of inflammasome activation by DEX treatment could be a promising neuroprotective strategy in neurodegenerative diseases by inhibiting NLRP inflammasomes.
Macrophage migration inhibitory factor (MIF), an immunoregulatory cytokine, produced by both innate and adaptive immune systems has been shown to be involved in the pathogenesis of multiple autoimmune disorders, exerting both immunoinflammatory and immunomodulatory effects in neurodegenerative disease models [192,193]. Immunopharmacological effects of MIF include increasing the TLR4 expression, inducing phagocytosis, and Th1 immune response, and stimulating cytokine-producing [194,195]. MIF poses enzymatic, endocrine, and cytokine-like roles, by binding to CD74 cell surface receptor and C-X-C chemokine receptors (CXCR2, CXCR4, CXCR7) [195]. MIF primarily displays pro-inflammatory effects, by activating pro-inflammatory mediators, such as IL-6 and TNF-α, and also inducing activation of inflammasome [196]. In a mouse model of PD, the upregulated profile of MIF exerts an antiapoptotic and autophagy-inducer role, suggesting the beneficial role of MIF in PD [197]. D-dopachrome tautomerase (D-DT) (also referred to as MIF2), the structural homologue of MIF, exerts synergic functions similar to MIF, both contributing to the progression of MS in males [198]. Benedek et al., found high levels of MIF and D-DT in MS male and female patients, with higher levels of CD74 in females than in males with increased MS disease severity [198]. In an EAE mouse model, mice lacking MIF or D-DT have been shown less severe symptoms of EAE. As MIF and D-DT are emerging as disease modifiers in MS, immunopharmacological therapy aimed to target CD74, the transducer of MIF signaling, might be a promising neuroprotective therapy in inhibiting the clinical progression of MS [198,199]. Given the anti-inflammatory and immunomodulatory profile of DEX, future studies to evaluate the impact of DEX on MIF and D-DT will be needed.
Moreover, immunoinflammatory disorders such as major depressive disorder characterized by high levels of inflammatory biomarkers have been shown reduced MIF levels after antidepressants, suggesting the pathogenic role of MIF in the induction and maintenance of these disorders [200].
Although multiple studies have shown the effect of DEX on decreasing pro-inflammatory cytokines (IL1-β and TNF-α) in animal models of neuroinflammation and neurodegenerative disorders, there is no data regarding the influence of DEX on anti-inflammatory cytokines that might play a protective role in these diseases [71,76]. Larger studies to explore the immunomodulatory role of DEX on anti-inflammatory cytokines associated with neuroprotection during neurodegenerative disorders including TGF beta, IL1 receptor antagonist, IL10, IL13, IL35, and IL37 should also be proposed.
Increasing evidence supported the contribution of miRNA dysregulation in the development and progression of neurodegenerative diseases such as AD, ALS, and HD [201,202,203].
In post-mortem analysis of PD patients, Hoss et al., have found a number of 125 miRNAs to be significantly altered in the prefrontal cortex [25].
In an APPswe/PS1dE9 mouse model of AD, DEX administration decreases the apoptosis rate of hippocampal neurons, and neuroinflammatory and oxidative stress-related processes, which were associated with up-regulated miR-214–5p and down-regulated SUZ12 [71]. The expression level of Yes-associated protein 1 (YAP1) and Jagged 1 (JAG1), have been shown to be up-regulated in AD patients, with miRNAs modulating the interaction between YAP1 and JAG1 [204,205,206]. In Aβ1-42-injected mice, DEX improved hippocampal neuron apoptosis and attenuated cognitive impairment by enhancing miR-129 which subsequently disrupt the interaction between YAP1 and JAG1, protecting against AD [207]. Upregulating miR-151-3p promotes anti-apoptotic and anti-oxidative effects of DEX on Aβ-treated neuronal cells by modulating DAPK-1 and TP53 signaling pathways [29]. Interestingly, DEX stimulates pro-apoptotic signal by activation of the ERK1/2 pathway, but TP53, the downstream effector of this pathways, abrogated this effect, suggesting a balance between pro-apoptotic and anti-apoptotic pathways [29].
An overview of the miRNAs involved in the neuroprotective mechanisms of DEX is illustrated in Figure 2.

7. MiRNAs in Pharmacological Response of Dexmedetomidine and Drug Response Variability

7.1. Involvement of MiRNAs in Drug Response Variability

miRNAs are increasingly being researched in the pathogenesis of multiple diseases, but the study of their involvement in drug response variability is essential to strengthening the effectiveness of the existing therapies [208].
Interindividual variability of drug response influences the efficacy and toxicity of drugs, and is controlled by both genetic and environmental factors [209,210]. Pharmacoepigenomics identifies the variability in gene expression related to drug response, and proceeds via methylation, histone modification, and miRNAs at the transcriptional and post-transcriptional levels [211].
Pharmacological responses of drugs are also controlled by miRNAs, as different expression levels of miRNAs have been revealed after several drugs’ administration [212].
MiRNAs could affect epigenetic regulation of genes in the expression of drug transporters, such as ABC and SLC transporters, and drug-metabolizing enzymes (DMEs), including cytochrome P450s (CYP450s) [212]. CYP2C9 responsible for the metabolism of warfarin and phenytoin, has been revealed to be suppressed after overexpression of miR-128-3p in HepaRG cells [213]. MiR-297 sensitizes colorectal multidrug resistance cancer cells to vincristine, oxaliplatin, and 5-fluorouracil, by reducing ABCC2 protein expression [214]. Another mechanism that could explain the regulation of drug transporters and DMEs consists in modulation of the expression of nuclear receptors, such as xenobiotic-sensing nuclear receptors family such as pregnane X receptor (PXR), hepatocyte nuclear factor 4 alpha (HNF4α), peroxisome proliferator-activated receptor alpha (PPARα) and nuclear hormone receptors, including glucocorticoid receptor (GR), estrogen receptor 1 (ESR1) [212]. The miRNAs expression profiling has shown significant changes after drug treatment. For instance, increases in serum levels of miR-451a and decreases in the serum levels of miRNA-34a-5p and miRNA-221-3p have been revealed in 84 depressed patients after treatment with paroxetine [215].
Besides this, miRNA polymorphisms or miRSNPs play critical roles in gene regulation, consisting of polymorphisms at the binding sites of miRNAs or at the level of miRNA sequences during biogenesis processes [212].
Numerous polymorphisms of miRNAs emerged as potential prognostic biomarkers and disease-modifying agents in cancer chemotherapy. LCS6 rs61764370 T > G, the polymorphism within let-7 binding site in the KRAS gene was associated with poor prognosis and response to cetuximab-irinotecan therapy in metastatic colorectal cancer [216]. Two polymorphisms, pri-miR26a-1rs7372209, and pri-miR-100rs1834306 were associated with different responses to chemotherapy treatment in metastatic colon cancer in response to 5-fluorouracil and irinotecan [217].
MiRNAs might be suitable biomarkers of individual drug response due to their stability in many body fluids, as well as their simple availability and reliable measurement [208]. Numerous promising genomic biomarkers to estimate drug responses are emerging for several disorders. For instance, miR-301 levels were elevated in non-respondent schizophrenic patients after haloperidol treatment [218]. In rheumatoid arthritis (RA) patients, analysis of blood samples indicated a high level of miR-125b-5p expression in good response patients, outlining the role of miR-125b-5p as predicting biomarker in response to Rituximab treatment [219].

7.2. MiRNAs in Dexmedetomidine’s Pharmacological Profiles

DEX exhibited significant differences in miRNAs expression, emerging promising miRNAs biomarkers to identify the interindividual variability of drug response. Mounting evidence supports the role of DEX in modulation of miRNAs expression for promoting specific effects in multiple organs, but the role of miRNAs in the variability of drug response was not fully explored.
A higher miRNA-183 expression in peripheral blood of 80 patients who underwent laparoscopic cholecystectomy and oophorocystectomy has been confirmed after treatment with DEX in day surgery, which was correlated with improving the remifentanil-related hyperalgesia [220]. In AD mice, miRNA-214-5p enhances the cerebral protection of DEX against neuronal injury, attenuating the cognitive impairment of AD mice [71]. Moreover, miRNA-151-3p promoted anti-apoptotic and anti-oxidative mechanisms against Aβ-treated neuronal cells, by modulating DAPK-1 and TP53 signaling pathways [29].
α2-adrenergic receptors (α2AAR) play significant regulatory roles in depressive responses, including postpartum depressive symptoms (PDS) pathophysiology, which were associated with an elevation in presynaptic α2AAR.
miRSNPs poses also an important role in α2AAR gene transcription, with important clinical significance in decreasing PDS incidence [221]. In a randomized clinical study, DEX attenuates PDS incidence in 600 women following cesarean section, by stimulating α2 adrenergic receptors [222].
α2AAR gene polymorphisms have been shown to play important roles in 103 PDS patients, who underwent cesarean section. α2AAR rs13306146 A > G polymorphism resulted in a decreased binding ability of miR-646 to the α2AAR gene at the 3′UTR, leading to a reduced α2AAR gene expression [221]. Cai et al., in a predictive study of response to DEX treatment, proposed miR-30a-5p, miR-101-3p, miR-140-3p, and miR-141-3p as predictive serum biomarkers for preoperative sedation outcome in 133 pediatric patients [33]. A higher plasma level of miR-101-3p and miR-140-3p was observed in respondents, while a downregulated expression of miR-101-3p and miR-30a-5p was associated with DEX adverse effects, i.e., hypotension and bradycardia [33]. Another study identified 12 differentially expressed human miRNAs in three patients after DEX treatment, five of that were upregulated (hsa-miR-4508, hsa-miR-novel-chr8_87373, hsa-miR-30a3p, hsa-miR-novel-chr16_26099, hsa-miR-4306 and seven downregulated (hsa-miR-744-5p, hsamiR-320a, hsa-miR-novel-chr9_90035, hsa-miR-101-3p, hsa-miR-150-5p, hsa-miR-342-3p, and hsa-miR-140-3p) [223]. In terms of DMEs of DEX, miR-141-3p down-regulates UDP-Glucuronosyltransferase (UGT) 1A4 enzyme UGT1A4, an important metabolizer of DEX treatment. A summary of promising miRNAs described so far in clinical setup, which might be used as predictive human biomarkers for DEX response can be viewed in Table 3.
Given the limitations of genomic analysis studies, aimed to estimate the circulating level of miRNA in response to DEX treatment, such as software predictions, lack of healthy sample subjects, and a more in-depth analysis of differential expression of miRNA depending on conditions of the health of patients will need to be conducted.
Therefore, miRNAs might be promising tools in estimating the interindividual variability of DEX response and also the safety and efficacy of DEX treatment in both healthy and disease conditions. Analyzing the circulating miRNAs in clinical studies in different conditions of the health, could translate precise genomic biomarkers of DEX response into clinical setups.

8. Potential Clinical Applications and Therapeutic Targets

With the knowledge advancement in the field of miRNAs, more and more studies outline the potential of miRNAs as peripheral biomarkers and disease-modifying agents in multiple brain disorders [137,224].
The applicability of miRNAs-based biomarkers in brain disorders has been ascertained in plasma, serum, cerebrospinal fluid, and other biological fluids [225]. Due to their stability in serum and other body fluids, and their correlation with disease prognosis, miRNAs were proposed as predictive biomarkers in disorders and drug response [208].
Understanding the role of miRNAs involved in the variability of drug response in terms of DEX efficacy and safety might strengthen the existing pharmacological profile of DEX. Moreover, targeting the miRNAs involved in the protective effects of DEX in multiple disorders, using specific miRNAs-based agents, termed agomirs and antagomirs, represents a promising therapeutic strategy for multiple clinical applications of DEX. Thus, we could supplementarily regulate the existing miRNAs regulated by DEX itself, therefore achieving a powerful effect of DEX against brain injuries.
On the other hand, we could modulate miRNAs responsible for DEX pharmacological responses, by using agomirs and antagomirs, reducing adverse effects and strengthen the existing DEX effects, thus achieving a personalized therapy for patients, which are candidates for DEX therapy. Thus, downregulating the overexpressed miRNAs associated with non-respondent patients after DEX therapy, using miRNAs blockers (antagomirs), or miRNA mimics (agomirs) might be a promising therapeutic tool.
The translation into practice of miRNAs-based agents in clinics for DEX treatment comes with pearls and pitfalls. From the miRNAs expression changes in a time-related manner to the variations depending on the intensity of the stimulus to the age and sex differences, all of which are part of the hard-to-control variables of the expression profiling of miRNAs [226]. Another impediment stems from the necessity of achieving an effective delivery system to address improper distribution, accumulation in non-target organs, non-specific absorption, toxicity, and rapid circulation degradation by systemic [227,228,229].
Besides the aforementioned considerations, we must take into consideration the limitations of animal studies, including low-quality scores, lack of data from females, old animals, and experimental models of comorbid animals [230].
More research studies will be needed to elucidate the molecular mechanisms of DEX in the neuroprotection context, and emerging new signaling pathways which could interact with miRNAs. Moreover, advancing in miRNA technology for developing new stable and feasible miRNAs-based biomarkers and therapeutic agents will further reinforce the existing DEX therapy against brain injury and avoid variability of DEX response and adverse effects. A better understanding of miRNAs-based gene regulation of DEX treatment could strengthen the existing neuroprotective effects of DEX treatment against brain injuries and could also provide future directions in individualized drug therapy.

Author Contributions

Conceptualization, C.-C.B., M.-A.N., A.-O.M., A.-C.S., A.-V.B. and A.-D.B.; writing—original draft preparation, C.-C.B., M.-A.N., A.-O.M., A.-C.S., A.-V.B. and A.-D.B.; writing—review and editing, C.-C.B., M.-A.N. and A.-O.M., supervision M.-A.N. and A.-D.B.; project administration, M.-A.N.; funding acquisition, C.-C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Iuliu Hatieganu University of Medicine and Pharmacy, internal grant number 35.171/17.12.2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Farrell, D.; Bendo, A.A. Perioperative Management of Severe Traumatic Brain Injury: What Is New? Curr. Anesthesiol. Rep. 2018, 8, 279–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Erkkinen, M.G.; Kim, M.-O.; Geschwind, M.D. Clinical Neurology and Epidemiology of the Major Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2018, 10, a033118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wallin, M.T.; Culpepper, W.J.; Nichols, E.; Bhutta, Z.A.; Gebrehiwot, T.T.; Hay, S.I.; Khalil, I.A.; Krohn, K.J.; Liang, X.; Naghavi, M.; et al. Global, Regional, and National Burden of Multiple Sclerosis 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 269–285. [Google Scholar] [CrossRef] [Green Version]
  4. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS Neurodegenerative Diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [Green Version]
  5. Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.-S.; Knapp, D.J.; Crews, F.T. Systemic LPS Causes Chronic Neuroinflammation and Progressive Neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef] [Green Version]
  6. Fidalgo, A.R.; Cibelli, M.; White, J.P.M.; Nagy, I.; Maze, M.; Ma, D. Systemic Inflammation Enhances Surgery-Induced Cognitive Dysfunction in Mice. Neurosci. Lett. 2011, 498, 63–66. [Google Scholar] [CrossRef]
  7. He, W.; Zhang, Z.; Sha, X. Nanoparticles-Mediated Emerging Approaches for Effective Treatment of Ischemic Stroke. Biomaterials 2021, 277, 121111. [Google Scholar] [CrossRef]
  8. Hong, H.-Y.; Choi, J.S.; Kim, Y.J.; Lee, H.Y.; Kwak, W.; Yoo, J.; Lee, J.-T.; Kwon, T.-H.; Kim, I.-S.; Han, H.-S.; et al. Detection of Apoptosis in a Rat Model of Focal Cerebral Ischemia Using a Homing Peptide Selected from in Vivo Phage Display. J. Control. Release Off. J. Control. Release Soc. 2008, 131, 167–172. [Google Scholar] [CrossRef]
  9. Paudyal, A.; Ghinea, F.S.; Driga, M.P.; Fang, W.-H.; Alessandri, G.; Combes, L.; Degens, H.; Slevin, M.; Hermann, D.M.; Popa-Wagner, A. P5 Peptide-Loaded Human Adipose-Derived Mesenchymal Stem Cells Promote Neurological Recovery After Focal Cerebral Ischemia in a Rat Model. Transl. Stroke Res. 2021, 12, 125–135. [Google Scholar] [CrossRef]
  10. Rehman, M.U.; Wali, A.F.; Ahmad, A.; Shakeel, S.; Rasool, S.; Ali, R.; Rashid, S.M.; Madkhali, H.; Ganaie, M.A.; Khan, R. Neuroprotective Strategies for Neurological Disorders by Natural Products: An Update. Curr. Neuropharmacol. 2019, 17, 247–267. [Google Scholar] [CrossRef]
  11. Chamorro, Á.; Lo, E.H.; Renú, A.; van Leyen, K.; Lyden, P.D. The Future of Neuroprotection in Stroke. J. Neurol. Neurosurg. Psychiatry 2021, 92, 129–135. [Google Scholar] [CrossRef]
  12. Bozorgi, H.; Zamani, M.; Motaghi, E.; Eslami, M. Dexmedetomidine as an Analgesic Agent with Neuroprotective Properties: Experimental and Clinical Aspects. J. Pain Palliat. Care Pharmacother. 2021, 35, 215–225. [Google Scholar] [CrossRef]
  13. Liaquat, Z.; Xu, X.; Zilundu, P.L.M.; Fu, R.; Zhou, L. The Current Role of Dexmedetomidine as Neuroprotective Agent: An Updated Review. Brain Sci. 2021, 11, 846. [Google Scholar] [CrossRef]
  14. Wang, Y.; Han, R.; Zuo, Z. Dexmedetomidine-Induced Neuroprotection: Is It Translational? Transl. Perioper. Pain Med. 2016, 1, 15–19. [Google Scholar]
  15. Hoffman, W.E.; Kochs, E.; Werner, C.; Thomas, C.; Albrecht, R.F. Dexmedetomidine Improves Neurologic Outcome from Incomplete Ischemia in the Rat. Reversal by the Alpha 2-Adrenergic Antagonist Atipamezole. Anesthesiology 1991, 75, 328–332. [Google Scholar] [CrossRef]
  16. Cheng, J.; Zhu, P.; Qin, H.; Li, X.; Yu, H.; Yu, H.; Peng, X. Dexmedetomidine Attenuates Cerebral Ischemia/Reperfusion Injury in Neonatal Rats by Inhibiting TLR4 Signaling. J. Int. Med. Res. 2018, 46, 2925–2932. [Google Scholar] [CrossRef]
  17. Zhu, Y.-M.; Wang, C.-C.; Chen, L.; Qian, L.-B.; Ma, L.-L.; Yu, J.; Zhu, M.-H.; Wen, C.-Y.; Yu, L.-N.; Yan, M. Both PI3K/Akt and ERK1/2 Pathways Participate in the Protection by Dexmedetomidine against Transient Focal Cerebral Ischemia/Reperfusion Injury in Rats. Brain Res. 2013, 1494, 1–8. [Google Scholar] [CrossRef]
  18. Luo, C.; Ouyang, M.-W.; Fang, Y.-Y.; Li, S.-J.; Zhou, Q.; Fan, J.; Qin, Z.-S.; Tao, T. Dexmedetomidine Protects Mouse Brain from Ischemia-Reperfusion Injury via Inhibiting Neuronal Autophagy through Up-Regulating HIF-1α. Front. Cell. Neurosci. 2017, 11, 197. [Google Scholar] [CrossRef] [Green Version]
  19. Lankadeva, Y.R.; Ma, S.; Iguchi, N.; Evans, R.G.; Hood, S.G.; Farmer, D.G.S.; Bailey, S.R.; Bellomo, R.; May, C.N. Dexmedetomidine Reduces Norepinephrine Requirements and Preserves Renal Oxygenation and Function in Ovine Septic Acute Kidney Injury. Kidney Int. 2019, 96, 1150–1161. [Google Scholar] [CrossRef]
  20. Bartel, D.P. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  21. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [Green Version]
  22. Tüfekci, K.U.; Oner, M.G.; Meuwissen, R.L.J.; Genç, S. The Role of MicroRNAs in Human Diseases. Methods Mol. Biol. 2014, 1107, 33–50. [Google Scholar] [CrossRef]
  23. Ardekani, A.M.; Naeini, M.M. The Role of MicroRNAs in Human Diseases. Avicenna J. Med. Biotechnol. 2010, 2, 161–179. [Google Scholar] [PubMed]
  24. Chen, Y.-M.; He, X.-Z.; Wang, S.-M.; Xia, Y. δ-Opioid Receptors, MicroRNAs, and Neuroinflammation in Cerebral Ischemia/Hypoxia. Front. Immunol. 2020, 11, 421. [Google Scholar] [CrossRef] [PubMed]
  25. Hoss, A.G.; Labadorf, A.; Beach, T.G.; Latourelle, J.C.; Myers, R.H. MicroRNA Profiles in Parkinson’s Disease Prefrontal Cortex. Front. Aging Neurosci. 2016, 8, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Nuzziello, N.; Vilardo, L.; Pelucchi, P.; Consiglio, A.; Liuni, S.; Trojano, M.; Liguori, M. Investigating the Role of MicroRNA and Transcription Factor Co-Regulatory Networks in Multiple Sclerosis Pathogenesis. Int. J. Mol. Sci. 2018, 19, 3652. [Google Scholar] [CrossRef] [Green Version]
  27. Petrescu, G.E.D.; Sabo, A.A.; Torsin, L.I.; Calin, G.A.; Dragomir, M.P. MicroRNA Based Theranostics for Brain Cancer: Basic Principles. J. Exp. Clin. Cancer Res. 2019, 38, 231. [Google Scholar] [CrossRef] [Green Version]
  28. Bao, Y.; Zhu, Y.; He, G.; Ni, H.; Liu, C.; Ma, L.; Zhang, L.; Shi, D. Dexmedetomidine Attenuates Neuroinflammation In LPS-Stimulated BV2 Microglia Cells Through Upregulation Of MiR-340. Drug Des. Devel. Ther. 2019, 13, 3465–3475. [Google Scholar] [CrossRef] [Green Version]
  29. Guo, Y.; Wu, Y.; Li, N.; Wang, Z. Up-Regulation of MiRNA-151-3p Enhanced the Neuroprotective Effect of Dexmedetomidine against β-Amyloid by Targeting DAPK-1 and TP53. Exp. Mol. Pathol. 2021, 118, 104587. [Google Scholar] [CrossRef]
  30. Yang, J.-J.; Zhao, Y.-H.; Yin, K.-W.; Zhang, X.-Q.; Liu, J. Dexmedetomidine Inhibits Inflammatory Response and Oxidative Stress through Regulating MiR-205-5p by Targeting HMGB1 in Cerebral Ischemic/Reperfusion. Immunopharmacol. Immunotoxicol. 2021, 43, 478–486. [Google Scholar] [CrossRef]
  31. Xue, Y.; Xu, T.; Jiang, W. Dexmedetomidine Protects PC12 Cells from Ropivacaine Injury through MiR-381/LRRC4 /SDF-1/CXCR4 Signaling Pathway. Regen. Ther. 2020, 14, 322–329. [Google Scholar] [CrossRef]
  32. Ciccacci, C.; Rufini, S.; Politi, C.; Novelli, G.; Forte, V.; Borgiani, P. Could MicroRNA Polymorphisms Influence Warfarin Dosing? A Pharmacogenetics Study on Mir133 Genes. Thromb. Res. 2015, 136, 367–370. [Google Scholar] [CrossRef]
  33. Cai, X.; Li, B.; Wei, W.; Guan, Y.; Bai, X.; Huang, M.; Huang, Y.; Rong, L.; Song, X.; Zhong, G. Circulating MicroRNA-30a-5p, MicroRNA-101-3p, MicroRNA-140-3p and MicroRNA-141-3p as Potential Biomarkers for Dexmedetomidine Response in Pediatric Patients. Eur. J. Clin. Pharmacol. 2021, 77, 1853–1859. [Google Scholar] [CrossRef]
  34. Scott-Warren, V.L.; Sebastian, J. Dexmedetomidine: Its Use in Intensive Care Medicine and Anaesthesia. BJA Educ. 2016, 16, 242–246. [Google Scholar] [CrossRef] [Green Version]
  35. Li, A.; Yuen, V.M.Y.; Goulay-Dufay, S.; Kwok, P.C.L. Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Drug Dev. Ind. Pharm. 2016, 42, 1917–1927. [Google Scholar] [CrossRef]
  36. Shehabi, Y.; Howe, B.D.; Bellomo, R.; Arabi, Y.M.; Bailey, M.; Bass, F.E.; Bin Kadiman, S.; McArthur, C.J.; Murray, L.; Reade, M.C.; et al. Early Sedation with Dexmedetomidine in Critically Ill Patients. N. Engl. J. Med. 2019, 380, 2506–2517. [Google Scholar] [CrossRef]
  37. Kim, K.H. Safe Sedation and Hypnosis Using Dexmedetomidine for Minimally Invasive Spine Surgery in a Prone Position. Korean J. Pain 2014, 27, 313–320. [Google Scholar] [CrossRef]
  38. Yuan, D.; Liu, Z.; Kaindl, J.; Maeda, S.; Zhao, J.; Sun, X.; Xu, J.; Gmeiner, P.; Wang, H.-W.; Kobilka, B.K. Activation of the A2B Adrenoceptor by the Sedative Sympatholytic Dexmedetomidine. Nat. Chem. Biol. 2020, 16, 507–512. [Google Scholar] [CrossRef]
  39. Weerink, M.A.S.; Struys, M.M.R.F.; Hannivoort, L.N.; Barends, C.R.M.; Absalom, A.R.; Colin, P. Clinical Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Clin. Pharmacokinet. 2017, 56, 893–913. [Google Scholar] [CrossRef] [Green Version]
  40. De Wolf, A.M.; Fragen, R.J.; Avram, M.J.; Fitzgerald, P.C.; Rahimi-Danesh, F. The Pharmacokinetics of Dexmedetomidine in Volunteers with Severe Renal Impairment. Anesth. Analg. 2001, 93, 1205–1209. [Google Scholar] [CrossRef]
  41. Zhong, W.; Zhang, Y.; Zhang, M.-Z.; Huang, X.-H.; Li, Y.; Li, R.; Liu, Q.-W. Pharmacokinetics of Dexmedetomidine Administered to Patients with End-Stage Renal Failure and Secondary Hyperparathyroidism Undergoing General Anaesthesia. J. Clin. Pharm. Ther. 2018, 43, 414–421. [Google Scholar] [CrossRef]
  42. Ahsan, M.Z.; Khan, F.U.; Zhao, M.-J.; Wang, Y.-X. Synergistic Interaction between Butorphanol and Dexmedetomidine in Antinociception. Eur. J. Pharm. Sci. 2020, 149, 105322. [Google Scholar] [CrossRef]
  43. Saccomanni, G.; Giorgi, M.; Del Carlo, S.; Manera, C.; Saba, A.; Macchia, M. Simultaneous Detection and Quantification of Parecoxib and Valdecoxib in Canine Plasma by HPLC with Spectrofluorimetric Detection: Development and Validation of a New Methodology. Anal. Bioanal. Chem. 2011, 401, 1677–1684. [Google Scholar] [CrossRef]
  44. Fenton, C.; Keating, G.M.; Wagstaff, A.J. Valdecoxib: A Review of Its Use in the Management of Osteoarthritis, Rheumatoid Arthritis, Dysmenorrhoea and Acute Pain. Drugs 2004, 64, 1231–1261. [Google Scholar] [CrossRef]
  45. Du, X.; Song, F.; Zhang, X.; Ma, S. Protective Efficacy of Combined Use of Parecoxib and Dexmedetomidine on Postoperative Hyperalgesia and Early Cognitive Dysfunction after Laparoscopic Cholecystectomy for Elderly Patients. Acta Cir. Bras. 2019, 34, e201900905. [Google Scholar] [CrossRef]
  46. Hu, J.; Lv, B.; Guo, W.; Wang, B.; Miao, D.; Qiu, X.; Chen, X. Effects of Dexmedetomidine on the Pharmacokinetics of Parecoxib and Its Metabolite Valdecoxib in Beagles by UPLC-MS/MS. BioMed Res. Int. 2020, 2020, e1563874. [Google Scholar] [CrossRef]
  47. Kaye, A.D.; Chernobylsky, D.J.; Thakur, P.; Siddaiah, H.; Kaye, R.J.; Eng, L.K.; Harbell, M.W.; Lajaunie, J.; Cornett, E.M. Dexmedetomidine in Enhanced Recovery After Surgery (ERAS) Protocols for Postoperative Pain. Curr. Pain Headache Rep. 2020, 24, 21. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, C.; Chen, Q.; Wang, P.; Jin, W.; Zhong, C.; Ge, Z.; Xu, K. The Effect of Dexmedetomidine as a Sedative Agent for Mechanically Ventilated Patients With Sepsis: A Systematic Review and Meta-Analysis. Front. Med. 2021, 8, 2444. [Google Scholar] [CrossRef] [PubMed]
  49. Xiang, H.; Hu, B.; Li, Z.; Li, J. Dexmedetomidine Controls Systemic Cytokine Levels through the Cholinergic Anti-Inflammatory Pathway. Inflammation 2014, 37, 1763–1770. [Google Scholar] [CrossRef] [PubMed]
  50. Srivastava, U.; Sarkar, M.E.; Kumar, A.; Gupta, A.; Agarwal, A.; Singh, T.K.; Badada, V.; Dwivedi, Y. Comparison of Clonidine and Dexmedetomidine for Short-Term Sedation of Intensive Care Unit Patients. Indian J. Crit. Care Med. 2014, 18, 431–436. [Google Scholar] [CrossRef] [Green Version]
  51. Reddy, V.S.; Shaik, N.A.; Donthu, B.; Reddy Sannala, V.K.; Jangam, V. Intravenous Dexmedetomidine versus Clonidine for Prolongation of Bupivacaine Spinal Anesthesia and Analgesia: A Randomized Double-Blind Study. J. Anaesthesiol. Clin. Pharmacol. 2013, 29, 342–347. [Google Scholar] [CrossRef]
  52. Abdallah, F.W.; Brull, R. Facilitatory Effects of Perineural Dexmedetomidine on Neuraxial and Peripheral Nerve Block: A Systematic Review and Meta-Analysis. Br. J. Anaesth. 2013, 110, 915–925. [Google Scholar] [CrossRef] [Green Version]
  53. Keating, G.M. Dexmedetomidine: A Review of Its Use for Sedation in the Intensive Care Setting. Drugs 2015, 75, 1119–1130. [Google Scholar] [CrossRef]
  54. Giovannitti, J.A.; Thoms, S.M.; Crawford, J.J. Alpha-2 Adrenergic Receptor Agonists: A Review of Current Clinical Applications. Anesth. Prog. 2015, 62, 31–38. [Google Scholar] [CrossRef] [Green Version]
  55. Kim, D.; Jeong, J.S.; Park, H.; Sung, K.-S.; Choi, S.J.; Gwak, M.S.; Kim, G.S.; Hahm, T.S.; Ko, J.S. Postoperative Pain Control after the Use of Dexmedetomidine and Propofol to Sedate Patients Undergoing Ankle Surgery under Spinal Anesthesia: A Randomized Controlled Trial. J. Pain Res. 2019, 12, 1479–1487. [Google Scholar] [CrossRef] [Green Version]
  56. Yuki, K. The Immunomodulatory Mechanism of Dexmedetomidine. Int. Immunopharmacol. 2021, 97, 107709. [Google Scholar] [CrossRef]
  57. Li, B.; Li, Y.; Tian, S.; Wang, H.; Wu, H.; Zhang, A.; Gao, C. Anti-Inflammatory Effects of Perioperative Dexmedetomidine Administered as an Adjunct to General Anesthesia: A Meta-Analysis. Sci. Rep. 2015, 5, 12342. [Google Scholar] [CrossRef] [Green Version]
  58. Zhang, L.; Xiao, F.; Zhang, J.; Wang, X.; Ying, J.; Wei, G.; Chen, S.; Huang, X.; Yu, W.; Liu, X.; et al. Dexmedetomidine Mitigated NLRP3-Mediated Neuroinflammation via the Ubiquitin-Autophagy Pathway to Improve Perioperative Neurocognitive Disorder in Mice. Front. Pharmacol. 2021, 12, 1143. [Google Scholar] [CrossRef]
  59. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  60. Feng, X.; Guan, W.; Zhao, Y.; Wang, C.; Song, M.; Yao, Y.; Yang, T.; Fan, H. Dexmedetomidine Ameliorates Lipopolysaccharide-Induced Acute Kidney Injury in Rats by Inhibiting Inflammation and Oxidative Stress via the GSK-3β/Nrf2 Signaling Pathway. J. Cell. Physiol. 2019, 234, 18994–19009. [Google Scholar] [CrossRef]
  61. Tas, N.; Altinbas, A.; Noyan, T.; Kokturk, S.; Ayhan, S.; Canakci, E. Acute Acetaminophene-Induced Hepatotoxicity and Nephrotoxicity; Therapeutic Effect of Dexmedetomidine. Bratisl. Lek. Listy 2019, 120, 270–276. [Google Scholar] [CrossRef] [PubMed]
  62. Morelli, A.; Sanfilippo, F.; Arnemann, P.; Hessler, M.; Kampmeier, T.G.; D’Egidio, A.; Orecchioni, A.; Santonocito, C.; Frati, G.; Greco, E.; et al. The Effect of Propofol and Dexmedetomidine Sedation on Norepinephrine Requirements in Septic Shock Patients: A Crossover Trial. Crit. Care Med. 2019, 47, e89. [Google Scholar] [CrossRef] [PubMed]
  63. Bafna, U.; Sharma, P.; Singhal, R.K.; Gurjar, S.S.; Bhargava, S.K. Comparison of Hypotensive Properties of Dexmedetomidine versus Clonidine for Induced Hypotension during Functional Endoscopic Sinus Surgery: A Randomised, Double-Blind Interventional Study. Indian J. Anaesth. 2021, 65, 579–585. [Google Scholar] [CrossRef] [PubMed]
  64. Cioccari, L.; Luethi, N.; Bailey, M.; Shehabi, Y.; Howe, B.; Messmer, A.S.; Proimos, H.K.; Peck, L.; Young, H.; Eastwood, G.M.; et al. The Effect of Dexmedetomidine on Vasopressor Requirements in Patients with Septic Shock: A Subgroup Analysis of the Sedation Practice in Intensive Care Evaluation [SPICE III] Trial. Crit. Care 2020, 24, 441. [Google Scholar] [CrossRef]
  65. Aouad, M.T.; Zeeni, C.; Al Nawwar, R.; Siddik-Sayyid, S.M.; Barakat, H.B.; Elias, S.; Yazbeck Karam, V.G. Dexmedetomidine for Improved Quality of Emergence From General Anesthesia: A Dose-Finding Study. Anesth. Analg. 2019, 129, 1504–1511. [Google Scholar] [CrossRef]
  66. Li, N.; Ouyang, B.S.; Liu, L.; Lin, C.S.; Xing, D.D.; Liu, J. Dexmedetomidine Protected COPD-Induced Lung Injury by Regulating MiRNA-146a. Bratisl. Lek. Listy 2016, 117, 539–542. [Google Scholar] [CrossRef] [Green Version]
  67. Zheng, X.; Li, J.; Fan, Q.; Zhao, X.; Chen, K. Dexmedetomidine Alleviates Myocardial Ischemia/Reperfusion-Induced Injury and Ca2+ Overload via the MicroRNA-346-3p/CaMKIId Axis. Int. J. Cardiol. 2021, 338, 185–195. [Google Scholar] [CrossRef]
  68. Ma, X.-G.; Liu, Y.; Xue, M.-X. [Dexmedetomidine alleviates hepatic ischemia-reperfusion injury by regulating MALAT1/miR-126-5p/HMGB1 axis]. Sheng Li Xue Bao 2021, 73, 253–262. [Google Scholar]
  69. Zheng, T.; Zheng, C.; Gao, F.; Huang, F.; Hu, B.; Zheng, X. Correction to: Dexmedetomidine Suppresses Bupivacaine-Induced Parthanatos in Human SH-SY5Y Cells via the MiR-7-5p/PARP1 Axis-Mediated ROS. Naunyn. Schmiedebergs Arch. Pharmacol. 2021, 394, 1055–1056. [Google Scholar] [CrossRef]
  70. Wei, W.; Sun, Z.; He, S.; Zhang, W.; Chen, S. Protective Role of Dexmedetomidine against Sevoflurane-Induced Postoperative Cognitive Dysfunction via the MicroRNA-129/TLR4 Axis. J. Clin. Neurosci. 2021, 92, 89–97. [Google Scholar] [CrossRef]
  71. Hu, G.; Shi, Z.; Shao, W.; Xu, B. MicroRNA-214-5p Involves in the Protection Effect of Dexmedetomidine against Neurological Injury in Alzheimer’s Disease via Targeting the Suppressor of Zest 12. Brain Res. Bull. 2022, 178, 164–172. [Google Scholar] [CrossRef]
  72. Huang, W.; Xiao, F.; Huang, W.; Wei, Q.; Li, X. MicroRNA-29a-3p Strengthens the Effect of Dexmedetomidine on Improving Neurologic Damage in Newborn Rats with Hypoxic-Ischemic Brain Damage by Inhibiting HDAC4. Brain Res. Bull. 2021, 167, 71–79. [Google Scholar] [CrossRef]
  73. Fang, H.; Li, H.-F.; Yan, J.-Y.; Yang, M.; Zhang, J.-P. Dexmedetomidine-up-Regulated MicroRNA-381 Exerts Anti-Inflammatory Effects in Rats with Cerebral Ischaemic Injury via the Transcriptional Factor IRF4. J. Cell. Mol. Med. 2021, 25, 2098–2109. [Google Scholar] [CrossRef]
  74. Wang, Y.-L.; Zhang, Y.; Cai, D.-S. Dexmedetomidine Ameliorates Postoperative Cognitive Dysfunction via the MicroRNA-381-Mediated EGR1/P53 Axis. Mol. Neurobiol. 2021, 58, 5052–5066. [Google Scholar] [CrossRef]
  75. Kho, W.; von Haefen, C.; Paeschke, N.; Nasser, F.; Endesfelder, S.; Sifringer, M.; González-López, A.; Lanzke, N.; Spies, C.D. Dexmedetomidine Restores Autophagic Flux, Modulates Associated MicroRNAs and the Cholinergic Anti-Inflammatory Pathway upon LPS-Treatment in Rats. J. Neuroimmune Pharmacol. 2021. [Google Scholar] [CrossRef]
  76. Paeschke, N.; Von Haefen, C.; Endesfelder, S.; Sifringer, M.; Spies, C.D. Dexmedetomidine Prevents Lipopolysaccharide-Induced MicroRNA Expression in the Adult Rat Brain. Int. J. Mol. Sci. 2017, 18, 1830. [Google Scholar] [CrossRef] [Green Version]
  77. Gao, Y.; Zhu, X.; Yu, H.; Wang, W.; Wang, Y.; Li, F.; Teng, J. Regulatory Role of MicroRNA-320 during off-Pump Coronary Artery Bypass Grafting with Dexmedetomidine Adjunct Anesthesia. Exp. Ther. Med. 2021, 22, 1201. [Google Scholar] [CrossRef]
  78. Sun, L.; Zhou, J.; Sun, C. MicroRNA-211-5p Enhances Analgesic Effect of Dexmedetomidine on Inflammatory Visceral Pain in Rats by Suppressing ERK Signaling. J. Mol. Neurosci. 2019, 68, 19–28. [Google Scholar] [CrossRef]
  79. Wang, L.; Tang, S.; Wang, Z.; Chen, H.; Rajcha, S.S.; Qian, J. The Administration of Dexmedetomidine Changes MicroRNA Expression Profiling of Rat Hearts. Biomed. Pharmacother. 2019, 120, 109463. [Google Scholar] [CrossRef]
  80. Pardo, P.S.; Hajira, A.; Boriek, A.M.; Mohamed, J.S. MicroRNA-434-3p Regulates Age-Related Apoptosis through EIF5A1 in the Skeletal Muscle. Aging 2017, 9, 1012–1029. [Google Scholar] [CrossRef] [Green Version]
  81. Wong, L.L.; Rademaker, M.T.; Saw, E.L.; Lew, K.S.; Ellmers, L.J.; Charles, C.J.; Richards, A.M.; Wang, P. Identification of Novel MicroRNAs in the Sheep Heart and Their Regulation in Heart Failure. Sci. Rep. 2017, 7, 8250. [Google Scholar] [CrossRef]
  82. Wang, J.; Dai, E.; Su, Z. Dexmedetomidine Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Regulation of MicroRNA-223-3p and HDAC4; Social Science Research Network: Rochester, NY, USA, 2020. [Google Scholar]
  83. Fang, H.; Li, H.-F.; Yang, M.; Wang, R.-R.; Wang, Q.-Y.; Zheng, P.-C.; Zhang, F.-X.; Zhang, J.-P. MicroRNA-128 Enhances Neuroprotective Effects of Dexmedetomidine on Neonatal Mice with Hypoxic-Ischemic Brain Damage by Targeting WNT1. Biomed. Pharmacother. 2019, 113, 108671. [Google Scholar] [CrossRef]
  84. Bao, N.; Tang, B. Organ-Protective Effects and the Underlying Mechanism of Dexmedetomidine. Mediators Inflamm. 2020, 2020, 6136105. [Google Scholar] [CrossRef]
  85. Tanabe, K.; Matsushima-Nishiwaki, R.; Kozawa, O.; Iida, H. Dexmedetomidine Suppresses Interleukin-1β-Induced Interleukin-6 Synthesis in Rat Glial Cells. Int. J. Mol. Med. 2014, 34, 1032–1038. [Google Scholar] [CrossRef] [Green Version]
  86. Zhang, X.; Wang, J.; Qian, W.; Zhao, J.; Sun, L.; Qian, Y.; Xiao, H. Dexmedetomidine Inhibits Tumor Necrosis Factor-Alpha and Interleukin 6 in Lipopolysaccharide-Stimulated Astrocytes by Suppression of c-Jun N-Terminal Kinases. Inflammation 2014, 37, 942–949. [Google Scholar] [CrossRef]
  87. Zhu, Y.-J.; Peng, K.; Meng, X.-W.; Ji, F.-H. Attenuation of Neuroinflammation by Dexmedetomidine Is Associated with Activation of a Cholinergic Anti-Inflammatory Pathway in a Rat Tibial Fracture Model. Brain Res. 2016, 1644, 1–8. [Google Scholar] [CrossRef]
  88. Wang, L.; Liu, H.; Zhang, L.; Wang, G.; Zhang, M.; Yu, Y. Neuroprotection of Dexmedetomidine against Cerebral Ischemia-Reperfusion Injury in Rats: Involved in Inhibition of NF-ΚB and Inflammation Response. Biomol. Ther. 2017, 25, 383–389. [Google Scholar] [CrossRef] [Green Version]
  89. Chiu, K.-M.; Lin, T.-Y.; Lu, C.-W.; Wang, S.-J. Inhibitory Effect of Glutamate Release from Rat Cerebrocortical Nerve Terminals by A2 Adrenoceptor Agonist Dexmedetomidine. Eur. J. Pharmacol. 2011, 670, 137–147. [Google Scholar] [CrossRef]
  90. Wang, X.; Shan, Y.; Tang, Z.; Gao, L.; Liu, H. Neuroprotective Effects of Dexmedetomidine against Isoflurane-Induced Neuronal Injury via Glutamate Regulation in Neonatal Rats. Drug Des. Devel. Ther. 2018, 13, 153–160. [Google Scholar] [CrossRef] [Green Version]
  91. Peng, M.; Ling, X.; Song, R.; Gao, X.; Liang, Z.; Fang, F.; Cang, J. Upregulation of GLT-1 via PI3K/Akt Pathway Contributes to Neuroprotection Induced by Dexmedetomidine. Front. Neurol. 2019, 10, 1041. [Google Scholar] [CrossRef] [Green Version]
  92. Han, J.-H.; Kim, D.-O.; Yi, J.-W.; Park, S.-W.; Kang, W.-J.; Choi, Y.-K.; Kim, S.-H.; Ko, I.-G.; Jin, J.-J.; Kim, S.-E.; et al. Dexmedetomidine, α2 -Adrenoceptor Agonist, Does Not Induce Apoptosis in the Brachial Plexus of Rats. Anim. Cells Syst. 2014, 18, 407–415. [Google Scholar] [CrossRef] [Green Version]
  93. Zhang, F.; Ding, T.; Yu, L.; Zhong, Y.; Dai, H.; Yan, M. Dexmedetomidine Protects against Oxygen–Glucose Deprivation-Induced Injury through the I2 Imidazoline Receptor-PI3K/AKT Pathway in Rat C6 Glioma Cells. J. Pharm. Pharmacol. 2011, 64, 120–127. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, Y.; Ma, L.; Gao, M.; Guo, W.; Ma, Y. Dexmedetomidine Reduces Postoperative Delirium after Joint Replacement in Elderly Patients with Mild Cognitive Impairment. Aging Clin. Exp. Res. 2016, 28, 729–736. [Google Scholar] [CrossRef] [PubMed]
  95. Su, X.; Meng, Z.-T.; Wu, X.-H.; Cui, F.; Li, H.-L.; Wang, D.-X.; Zhu, X.; Zhu, S.-N.; Maze, M.; Ma, D. Dexmedetomidine for Prevention of Delirium in Elderly Patients after Non-Cardiac Surgery: A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2016, 388, 1893–1902. [Google Scholar] [CrossRef] [Green Version]
  96. Pandharipande, P.P.; Sanders, R.D.; Girard, T.D.; McGrane, S.; Thompson, J.L.; Shintani, A.K.; Herr, D.L.; Maze, M.; Ely, E.W. MENDS investigators Effect of Dexmedetomidine versus Lorazepam on Outcome in Patients with Sepsis: An a Priori-Designed Analysis of the MENDS Randomized Controlled Trial. Crit. Care 2010, 14, R38. [Google Scholar] [CrossRef] [Green Version]
  97. Finnerty, M.; Marczynski, T.J.; Amirault, H.J.; Urbancic, M.; Andersen, B.R. Benzodiazepines Inhibit Neutrophil Chemotaxis and Superoxide Production in a Stimulus Dependent Manner; PK-11195 Antagonizes These Effects. Immunopharmacology 1991, 22, 185–193. [Google Scholar] [CrossRef]
  98. Miles, B.A.; Lafuse, W.P.; Zwilling, B.S. Binding of Alpha-Adrenergic Receptors Stimulates the Anti-Mycobacterial Activity of Murine Peritoneal Macrophages. J. Neuroimmunol. 1996, 71, 19–24. [Google Scholar] [CrossRef]
  99. Shokri, H.; Ali, I. A Randomized Control Trial Comparing Prophylactic Dexmedetomidine versus Clonidine on Rates and Duration of Delirium in Older Adult Patients Undergoing Coronary Artery Bypass Grafting. J. Clin. Anesth. 2020, 61, 109622. [Google Scholar] [CrossRef]
  100. Chen, P.-H.; Tsuang, F.-Y.; Lee, C.-T.; Yeh, Y.-C.; Cheng, H.-L.; Lee, T.-S.; Chang, Y.-W.; Cheng, Y.-J.; Wu, C.-Y. Neuroprotective Effects of Intraoperative Dexmedetomidine versus Saline Infusion Combined with Goal-Directed Haemodynamic Therapy for Patients Undergoing Cranial Surgery: A Randomised Controlled Trial. Eur. J. Anaesthesiol. 2021, 38, 1262–1271. [Google Scholar] [CrossRef]
  101. Ge, Y.; Li, Q.; Nie, Y.; Gao, J.; Luo, K.; Fang, X.; Wang, C. Dexmedetomidine Improves Cognition after Carotid Endarterectomy by Inhibiting Cerebral Inflammation and Enhancing Brain-Derived Neurotrophic Factor Expression. J. Int. Med. Res. 2019, 47, 2471–2482. [Google Scholar] [CrossRef]
  102. Luo, X.; Zheng, X.; Huang, H. Protective Effects of Dexmedetomidine on Brain Function of Glioma Patients Undergoing Craniotomy Resection and Its Underlying Mechanism. Clin. Neurol. Neurosurg. 2016, 146, 105–108. [Google Scholar] [CrossRef]
  103. Calin, G.A.; Liu, C.-G.; Sevignani, C.; Ferracin, M.; Felli, N.; Dumitru, C.D.; Shimizu, M.; Cimmino, A.; Zupo, S.; Dono, M.; et al. MicroRNA Profiling Reveals Distinct Signatures in B Cell Chronic Lymphocytic Leukemias. Proc. Natl. Acad. Sci. USA 2004, 101, 11755–11760. [Google Scholar] [CrossRef] [Green Version]
  104. Zhang, J.; Li, S.; Li, L.; Li, M.; Guo, C.; Yao, J.; Mi, S. Exosome and Exosomal MicroRNA: Trafficking, Sorting, and Function. Genom. Proteom. Bioinform. 2015, 13, 17–24. [Google Scholar] [CrossRef] [Green Version]
  105. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of MicroRNAs in Serum: A Novel Class of Biomarkers for Diagnosis of Cancer and Other Diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef] [Green Version]
  106. Huang, W. MicroRNAs: Biomarkers, Diagnostics, and Therapeutics. Methods Mol. Biol. 2017, 1617, 57–67. [Google Scholar] [CrossRef]
  107. Basak, I.; Patil, K.S.; Alves, G.; Larsen, J.P.; Møller, S.G. MicroRNAs as Neuroregulators, Biomarkers and Therapeutic Agents in Neurodegenerative Diseases. Cell. Mol. Life Sci. CMLS 2016, 73, 811–827. [Google Scholar] [CrossRef]
  108. Alawneh, K.Z.; Raffee, L.A.; Alshehabat, M.A.M.; Haddad, H.; Jaradat, S.A. Characterizing and Profiling MicroRNAs in Dogs Undergoing Induced Ischemic Brain Stroke After Middle Cerebral Artery Occlusion Under Fluoroscopic Guidance. Vasc. Health Risk Manag. 2021, 17, 543–550. [Google Scholar] [CrossRef]
  109. Liu, D.-Z.; Tian, Y.; Ander, B.P.; Xu, H.; Stamova, B.S.; Zhan, X.; Turner, R.J.; Jickling, G.; Sharp, F.R. Brain and Blood MicroRNA Expression Profiling of Ischemic Stroke, Intracerebral Hemorrhage, and Kainate Seizures. J. Cereb. Blood Flow Metab. 2010, 30, 92–101. [Google Scholar] [CrossRef]
  110. Zeng, L.; Liu, J.; Wang, Y.; Wang, L.; Weng, S.; Tang, Y.; Zheng, C.; Cheng, Q.; Chen, S.; Yang, G.-Y. MicroRNA-210 as a Novel Blood Biomarker in Acute Cerebral Ischemia. Front. Biosci. Elite Ed. 2011, 3, 1265–1272. [Google Scholar] [CrossRef]
  111. Tiedt, S.; Prestel, M.; Malik, R.; Schieferdecker, N.; Duering, M.; Kautzky, V.; Stoycheva, I.; Böck, J.; Northoff, B.H.; Klein, M.; et al. RNA-Seq Identifies Circulating MiR-125a-5p, MiR-125b-5p, and MiR-143-3p as Potential Biomarkers for Acute Ischemic Stroke. Circ. Res. 2017, 121, 970–980. [Google Scholar] [CrossRef]
  112. Ponnusamy, V.; Yip, P.K. The Role of MicroRNAs in Newborn Brain Development and Hypoxic Ischaemic Encephalopathy. Neuropharmacology 2019, 149, 55–65. [Google Scholar] [CrossRef]
  113. Ma, F.; Zhang, X.; Yin, K.-J. MicroRNAs in Central Nervous System Diseases: A Prospective Role in Regulating Blood-Brain Barrier Integrity. Exp. Neurol. 2020, 323, 113094. [Google Scholar] [CrossRef]
  114. Saugstad, J.A. MicroRNAs as Effectors of Brain Function with Roles in Ischemia and Injury, Neuroprotection, and Neurodegeneration. J. Cereb. Blood Flow Metab. 2010, 30, 1564–1576. [Google Scholar] [CrossRef]
  115. Tan, K.S.; Armugam, A.; Sepramaniam, S.; Lim, K.Y.; Setyowati, K.D.; Wang, C.W.; Jeyaseelan, K. Expression Profile of MicroRNAs in Young Stroke Patients. PLoS ONE 2009, 4, e7689. [Google Scholar] [CrossRef] [Green Version]
  116. Jeyaseelan, K.; Lim, K.Y.; Armugam, A. MicroRNA Expression in the Blood and Brain of Rats Subjected to Transient Focal Ischemia by Middle Cerebral Artery Occlusion. Stroke 2008, 39, 959–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Voelz, C.; Ebrahimy, N.; Zhao, W.; Habib, P.; Zendedel, A.; Pufe, T.; Beyer, C.; Slowik, A. Transient Focal Cerebral Ischemia Leads to MiRNA Alterations in Different Brain Regions, Blood Serum, Liver, and Spleen. Int. J. Mol. Sci. 2021, 23, 161. [Google Scholar] [CrossRef] [PubMed]
  118. Bertero, T.; Rezzonico, R.; Pottier, N.; Mari, B. Impact of MicroRNAs in the Cellular Response to Hypoxia. Int. Rev. Cell Mol. Biol. 2017, 333, 91–158. [Google Scholar] [CrossRef] [PubMed]
  119. Sun, C.; Liu, J.; Duan, F.; Cong, L.; Qi, X. The Role of the MicroRNA Regulatory Network in Alzheimer’s Disease: A Bioinformatics Analysis. Arch. Med. Sci. 2021, 18, 206–222. [Google Scholar] [CrossRef] [PubMed]
  120. Kashif, H.; Shah, D.; Sukumari-Ramesh, S. Dysregulation of MicroRNA and Intracerebral Hemorrhage: Roles in Neuroinflammation. Int. J. Mol. Sci. 2021, 22, 8115. [Google Scholar] [CrossRef]
  121. Wang, H.-W.; Noland, C.; Siridechadilok, B.; Taylor, D.W.; Ma, E.; Felderer, K.; Doudna, J.A.; Nogales, E. Structural Insights into RNA Processing by the Human RISC-Loading Complex. Nat. Struct. Mol. Biol. 2009, 16, 1148–1153. [Google Scholar] [CrossRef] [Green Version]
  122. Doxakis, E. Post-Transcriptional Regulation of α-Synuclein Expression by Mir-7 and Mir-153. J. Biol. Chem. 2010, 285, 12726–12734. [Google Scholar] [CrossRef] [Green Version]
  123. Cho, H.J.; Liu, G.; Jin, S.M.; Parisiadou, L.; Xie, C.; Yu, J.; Sun, L.; Ma, B.; Ding, J.; Vancraenenbroeck, R.; et al. MicroRNA-205 Regulates the Expression of Parkinson’s Disease-Related Leucine-Rich Repeat Kinase 2 Protein. Hum. Mol. Genet. 2013, 22, 608–620. [Google Scholar] [CrossRef] [Green Version]
  124. Abbasi-Habashi, S.; Jickling, G.C.; Winship, I.R. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front. Neurol. 2021, 12, 746486. [Google Scholar] [CrossRef]
  125. Vinciguerra, A.; Formisano, L.; Cerullo, P.; Guida, N.; Cuomo, O.; Esposito, A.; Di Renzo, G.; Annunziato, L.; Pignataro, G. MicroRNA-103-1 Selectively Downregulates Brain NCX1 and Its Inhibition by Anti-MiRNA Ameliorates Stroke Damage and Neurological Deficits. Mol. Ther. J. Am. Soc. Gene Ther. 2014, 22, 1829–1838. [Google Scholar] [CrossRef] [Green Version]
  126. Xu, L.-J.; Ouyang, Y.-B.; Xiong, X.; Stary, C.M.; Giffard, R.G. Post-Stroke Treatment with MiR-181 Antagomir Reduces Injury and Improves Long-Term Behavioral Recovery in Mice after Focal Cerebral Ischemia. Exp. Neurol. 2015, 264, 1–7. [Google Scholar] [CrossRef] [Green Version]
  127. Zhuo, Y.; Chen, W.; Li, W.; Huang, Y.; Duan, D.; Ge, L.; He, J.; Liu, J.; Hu, Z.; Lu, M. Ischemic-Hypoxic Preconditioning Enhances the Mitochondrial Function Recovery of Transplanted Olfactory Mucosa Mesenchymal Stem Cells via MiR-181a Signaling in Ischemic Stroke. Aging 2021, 13, 11234–11256. [Google Scholar] [CrossRef]
  128. Bartel, D.P. MicroRNAs: Target Recognition and Regulatory Functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
  129. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The Nuclear RNase III Drosha Initiates MicroRNA Processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  130. MacFarlane, L.-A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar] [CrossRef] [Green Version]
  131. Lytle, J.R.; Yario, T.A.; Steitz, J.A. Target MRNAs Are Repressed as Efficiently by MicroRNA-Binding Sites in the 5’ UTR as in the 3’ UTR. Proc. Natl. Acad. Sci. USA 2007, 104, 9667–9672. [Google Scholar] [CrossRef] [Green Version]
  132. Kiriakidou, M.; Tan, G.S.; Lamprinaki, S.; De Planell-Saguer, M.; Nelson, P.T.; Mourelatos, Z. An MRNA M7G Cap Binding-like Motif within Human Ago2 Represses Translation. Cell 2007, 129, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  133. Treiber, T.; Treiber, N.; Meister, G. Regulation of MicroRNA Biogenesis and Its Crosstalk with Other Cellular Pathways. Nat. Rev. Mol. Cell Biol. 2019, 20, 5–20. [Google Scholar] [CrossRef]
  134. Forman, J.J.; Legesse-Miller, A.; Coller, H.A. A Search for Conserved Sequences in Coding Regions Reveals That the Let-7 MicroRNA Targets Dicer within Its Coding Sequence. Proc. Natl. Acad. Sci. USA 2008, 105, 14879–14884. [Google Scholar] [CrossRef] [Green Version]
  135. Zhang, J.; Zhou, W.; Liu, Y.; Liu, T.; Li, C.; Wang, L. Oncogenic Role of MicroRNA-532-5p in Human Colorectal Cancer via Targeting of the 5’UTR of RUNX3. Oncol. Lett. 2018, 15, 7215–7220. [Google Scholar] [CrossRef] [Green Version]
  136. Nallamshetty, S.; Chan, S.Y.; Loscalzo, J. Hypoxia: A Master Regulator of MicroRNA Biogenesis and Activity. Free Radic. Biol. Med. 2013, 64, 20–30. [Google Scholar] [CrossRef] [Green Version]
  137. Minhas, G.; Mathur, D.; Ragavendrasamy, B.; Sharma, N.K.; Paanu, V.; Anand, A. Hypoxia in CNS Pathologies: Emerging Role of MiRNA-Based Neurotherapeutics and Yoga Based Alternative Therapies. Front. Neurosci. 2017, 11, 386. [Google Scholar] [CrossRef]
  138. Morimoto, K.; Eguchi, R.; Kitano, T.; Otsuguro, K.-I. Alpha and Beta Adrenoceptors Activate Interleukin-6 Transcription through Different Pathways in Cultured Astrocytes from Rat Spinal Cord. Cytokine 2021, 142, 155497. [Google Scholar] [CrossRef]
  139. Morimoto, K.; Kitano, T.; Eguchi, R.; Otsuguro, K.-I. Bidirectional Modulation of TNF-α Transcription via α- and β-Adrenoceptors in Cultured Astrocytes from Rat Spinal Cord. Biochem. Biophys. Res. Commun. 2020, 528, 78–84. [Google Scholar] [CrossRef]
  140. Nohata, N.; Goto, Y.; Gutkind, J.S. Onco-GPCR Signaling and Dysregulated Expression of MicroRNAs in Human Cancer. J. Hum. Genet. 2017, 62, 87–96. [Google Scholar] [CrossRef]
  141. Vivacqua, A.; De Marco, P.; Santolla, M.F.; Cirillo, F.; Pellegrino, M.; Panno, M.L.; Abonante, S.; Maggiolini, M. Estrogenic Gper Signaling Regulates Mir144 Expression in Cancer Cells and Cancer-Associated Fibroblasts (Cafs). Oncotarget 2015, 6, 16573–16587. [Google Scholar] [CrossRef] [Green Version]
  142. Tao, S.; He, H.; Chen, Q. Estradiol Induces HOTAIR Levels via GPER-Mediated MiR-148a Inhibition in Breast Cancer. J. Transl. Med. 2015, 13, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Luo, Y.; Tang, H.; Li, H.; Zhao, R.; Huang, Q.; Liu, J. Recent Advances in the Development of Neuroprotective Agents and Therapeutic Targets in the Treatment of Cerebral Ischemia. Eur. J. Med. Chem. 2019, 162, 132–146. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, Z.; Wang, Y.; Li, F.; Liu, X.; Zhang, H.; Zhu, Z.; Huang, H.; Xu, X. Circ-Camk4 Involved in Cerebral Ischemia/Reperfusion Induced Neuronal Injury. Sci. Rep. 2020, 10, 7012. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, H.; Wang, H.; Shang, H.; Chen, X.; Yang, S.; Qu, Y.; Ding, J.; Li, X. Circular RNA Circ_0000950 Promotes Neuron Apoptosis, Suppresses Neurite Outgrowth and Elevates Inflammatory Cytokines Levels via Directly Sponging MiR-103 in Alzheimer’s Disease. Cell Cycle Georget. Tex 2019, 18, 2197–2214. [Google Scholar] [CrossRef]
  146. 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]
  147. Chen, H.; Gu, B.; Zhao, X.; Zhao, Y.; Huo, S.; Liu, X.; Lu, H. Circular RNA Hsa_circ_0007364 Increases Cervical Cancer Progression through Activating Methionine Adenosyltransferase II Alpha (MAT2A) Expression by Restraining MicroRNA-101-5p. Bioengineered 2020, 11, 1269–1279. [Google Scholar] [CrossRef]
  148. Wang, J.; Wang, Y. Circular RNA Cerebellar Degeneration-Related Protein 1 Antisense RNA (Circ-CDR1as) Downregulation Induced by Dexmedetomidine Treatment Protects Hippocampal Neurons against Hypoxia/Reoxygenation Injury through the MicroRNA-28-3p (MiR-28-3p)/Tumor Necrosis Factor Receptor-Associated Factor-3 (TRAF3) Axis. Bioengineered 2021, 12, 10512–10524. [Google Scholar] [CrossRef]
  149. Liu, W.; Shao, C.; Zang, C.; Sun, J.; Xu, M.; Wang, Y. Protective Effects of Dexmedetomidine on Cerebral Ischemia/Reperfusion Injury via the MicroRNA-214/ROCK1/NF-ΚB Axis. BMC Anesthesiol. 2021, 21, 203. [Google Scholar] [CrossRef]
  150. Sifringer, M.; von Haefen, C.; Krain, M.; Paeschke, N.; Bendix, I.; Bührer, C.; Spies, C.D.; Endesfelder, S. Neuroprotective Effect of Dexmedetomidine on Hyperoxia-Induced Toxicity in the Neonatal Rat Brain. Oxid. Med. Cell. Longev. 2015, 2015, 530371. [Google Scholar] [CrossRef] [Green Version]
  151. Li, Y.-J.; Zhang, D.-Z.; Xi, Y.; Wu, C.-A. Protective Effect of Dexmedetomidine on Neuronal Hypoxic Injury through Inhibition of MiR-134. Hum. Exp. Toxicol. 2021, 40, 2145–2155. [Google Scholar] [CrossRef]
  152. Chen, Y.; Fan, Z.; Wu, Q. Dexmedetomidine Improves Oxygen-Glucose Deprivation/Reoxygenation (OGD/R)-Induced Neurological Injury through Regulating SNHG11/MiR-324-3p/VEGFA Axis. Bioengineered 2021, 12, 4794–4804. [Google Scholar] [CrossRef]
  153. Shi, X.; Yan, C.; Liu, B.; Yang, C.; Nie, X.; Wang, X.; Zheng, J.; Wang, Y.; Zhu, Y. MiR-381 Regulates Neural Stem Cell Proliferation and Differentiation via Regulating Hes1 Expression. PLoS ONE 2015, 10, e0138973. [Google Scholar] [CrossRef]
  154. Mamun, A.A.; Chauhan, A.; Qi, S.; Ngwa, C.; Xu, Y.; Sharmeen, R.; Hazen, A.L.; Li, J.; Aronowski, J.A.; McCullough, L.D.; et al. Microglial IRF5-IRF4 Regulatory Axis Regulates Neuroinflammation after Cerebral Ischemia and Impacts Stroke Outcomes. Proc. Natl. Acad. Sci. USA 2020, 117, 1742–1752. [Google Scholar] [CrossRef]
  155. Degos, V.; Charpentier, T.L.; Chhor, V.; Brissaud, O.; Lebon, S.; Schwendimann, L.; Bednareck, N.; Passemard, S.; Mantz, J.; Gressens, P. Neuroprotective Effects of Dexmedetomidine against Glutamate Agonist-Induced Neuronal Cell Death Are Related to Increased Astrocyte Brain-Derived Neurotrophic Factor Expression. Anesthesiology 2013, 118, 1123–1132. [Google Scholar] [CrossRef]
  156. Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef]
  157. Wang, L.; Liu, W.; Zhang, Y.; Hu, Z.; Guo, H.; Lv, J.; Du, H. Dexmedetomidine Had Neuroprotective Effects on Hippocampal Neuronal Cells via Targeting LncRNA SHNG16 Mediated MicroRNA-10b-5p/BDNF Axis. Mol. Cell. Biochem. 2020, 469, 41–51. [Google Scholar] [CrossRef] [Green Version]
  158. Shan, Y.; Yang, F.; Tang, Z.; Bi, C.; Sun, S.; Zhang, Y.; Liu, H. Dexmedetomidine Ameliorates the Neurotoxicity of Sevoflurane on the Immature Brain Through the BMP/SMAD Signaling Pathway. Front. Neurosci. 2018, 12, 964. [Google Scholar] [CrossRef]
  159. Fang, H.; Li, H.-F.; He, M.-H.; Yan, J.-Y.; Yang, M.; Zhang, F.-X.; Wang, R.-R.; Wang, Q.-Y.; Zhang, J.-P. Long Non-Coding RNA MALAT1 Sponges MicroRNA-429 to Regulate Apoptosis of Hippocampal Neurons in Hypoxic-Ischemic Brain Damage by Regulating WNT1. Brain Res. Bull. 2019, 152, 1–10. [Google Scholar] [CrossRef]
  160. Huang, Z.; Liu, G.; Zeng, Q.; Gao, R.; Zhang, S.; Wang, L.; Liu, B.; Yu, Y.; Zhao, A.; Li, R.; et al. MiR-29b Expression Is Associated with a Dexmedetomidine-Mediated Protective Effect against Oxygen-Glucose Deprivation-Induced Injury to SK-N-SH Cells in Vitro. Cell Biol. Int. 2018, 42, 344–352. [Google Scholar] [CrossRef]
  161. Wu, L.; Xi, Y.; Kong, Q. Dexmedetomidine Protects PC12 Cells from Oxidative Damage through Regulation of MiR-199a/HIF-1α. Artif. Cells Nanomed. Biotechnol. 2020, 48, 506–514. [Google Scholar] [CrossRef]
  162. Dahmani, S.; Rouelle, D.; Gressens, P.; Mantz, J. Effects of Dexmedetomidine on Hippocampal Focal Adhesion Kinase Tyrosine Phosphorylation in Physiologic and Ischemic Conditions. Anesthesiology 2005, 103, 969–977. [Google Scholar] [CrossRef]
  163. Reyland, M.E.; Barzen, K.A.; Anderson, S.M.; Quissell, D.O.; Matassa, A.A. Activation of PKC Is Sufficient to Induce an Apoptotic Program in Salivary Gland Acinar Cells. Cell Death Differ. 2000, 7, 1200–1209. [Google Scholar] [CrossRef]
  164. Zhu, Y.-S.; Liu, Z.; Min, J.; Xiong, Y.-F. Dexmedetomidine Reduces the Apoptosis of Rat Hippocampal Neurons via Mediating ERK1/2 Signal Pathway by Targeting MiR-155. Acta Histochem. 2021, 123, 151734. [Google Scholar] [CrossRef]
  165. Mielcarek, M.; Zielonka, D.; Carnemolla, A.; Marcinkowski, J.T.; Guidez, F. HDAC4 as a Potential Therapeutic Target in Neurodegenerative Diseases: A Summary of Recent Achievements. Front. Cell. Neurosci. 2015, 9, 42. [Google Scholar] [CrossRef] [Green Version]
  166. Zhou, X.-M.; Liu, J.; Wang, Y.; Zhang, M.-H. Silencing of Long Noncoding RNA MEG3 Enhances Cerebral Protection of Dexmedetomidine against Hypoxic-Ischemic Brain Damage in Neonatal Mice by Binding to MiR-129-5p. J. Cell. Biochem. 2019, 120, 7978–7988. [Google Scholar] [CrossRef]
  167. Rayasam, A.; Hsu, M.; Kijak, J.A.; Kissel, L.; Hernandez, G.; Sandor, M.; Fabry, Z. Immune Responses in Stroke: How the Immune System Contributes to Damage and Healing after Stroke and How This Knowledge Could Be Translated to Better Cures? Immunology 2018, 154, 363–376. [Google Scholar] [CrossRef] [Green Version]
  168. Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/Macrophage Polarization Dynamics Reveal Novel Mechanism of Injury Expansion after Focal Cerebral Ischemia. Stroke 2012, 43, 3063–3070. [Google Scholar] [CrossRef] [Green Version]
  169. Ta Na, H.S.; Nuo, M.; Meng, Q.-T.; Xia, Z.-Y. The Pathway of Let-7a-1/2-3p and HMGB1 Mediated Dexmedetomidine Inhibiting Microglia Activation in Spinal Cord Ischemia-Reperfusion Injury Mice. J. Mol. Neurosci. 2019, 69, 106–114. [Google Scholar] [CrossRef]
  170. Perez-Zoghbi, J.F.; Zhu, W.; Grafe, M.R.; Brambrink, A.M. Dexmedetomidine-Mediated Neuroprotection against Sevoflurane-Induced Neurotoxicity Extends to Several Brain Regions in Neonatal Rats. Br. J. Anaesth. 2017, 119, 506–516. [Google Scholar] [CrossRef] [Green Version]
  171. Sanders, R.D.; Sun, P.; Patel, S.; Li, M.; Maze, M.; Ma, D. Dexmedetomidine Provides Cortical Neuroprotection: Impact on Anaesthetic-Induced Neuroapoptosis in the Rat Developing Brain. Acta Anaesthesiol. Scand. 2010, 54, 710–716. [Google Scholar] [CrossRef]
  172. Duan, X.; Li, Y.; Zhou, C.; Huang, L.; Dong, Z. Dexmedetomidine Provides Neuroprotection: Impact on Ketamine-Induced Neuroapoptosis in the Developing Rat Brain. Acta Anaesthesiol. Scand. 2014, 58, 1121–1126. [Google Scholar] [CrossRef] [PubMed]
  173. Twaroski, D.; Bosnjak, Z.J.; Bai, X. MicroRNAs: New Players in Anesthetic-Induced Developmental Neurotoxicity. Pharm. Anal. Acta 2015, 6, 357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hong, B.; Lim, C.S.; Kim, Y.-H.; Lee, J.U.; Kim, Y.M.; Jung, C.; Jo, Y. Comparison of Topical Ropivacaine with and without Ketamine on Post-Surgical Pain in Children Undergoing Tonsillectomy: A Randomized Controlled Double-Blind Study. J. Anesth. 2017, 31, 559–564. [Google Scholar] [CrossRef] [PubMed]
  175. David, K.K.; Andrabi, S.A.; Dawson, T.M.; Dawson, V.L. Parthanatos, a Messenger of Death. Front. Biosci. Landmark Ed. 2009, 14, 1116–1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Li, W.-H.; Yang, Y.-L.; Cheng, X.; Liu, M.; Zhang, S.-S.; Wang, Y.-H.; Du, G.-H. Baicalein Attenuates Caspase-Independent Cells Death via Inhibiting PARP-1 Activation and AIF Nuclear Translocation in Cerebral Ischemia/Reperfusion Rats. Apoptosis Int. J. Program. Cell Death 2020, 25, 354–369. [Google Scholar] [CrossRef] [PubMed]
  177. Ma, D.; Lu, B.; Feng, C.; Wang, C.; Wang, Y.; Luo, T.; Feng, J.; Jia, H.; Chi, G.; Luo, Y.; et al. Deoxypodophyllotoxin Triggers Parthanatos in Glioma Cells via Induction of Excessive ROS. Cancer Lett. 2016, 371, 194–204. [Google Scholar] [CrossRef] [PubMed]
  178. Xu, S.; Gao, R.; Chen, L. Dexmedetomidine Regulates Sevoflurane-Induced Neurotoxicity through the MiR-330-3p/ULK1 Axis. J. Biochem. Mol. Toxicol. 2021, 35, e22919. [Google Scholar] [CrossRef] [PubMed]
  179. Biesmans, S.; Meert, T.F.; Bouwknecht, J.A.; Acton, P.D.; Davoodi, N.; De Haes, P.; Kuijlaars, J.; Langlois, X.; Matthews, L.J.R.; Ver Donck, L.; et al. Systemic Immune Activation Leads to Neuroinflammation and Sickness Behavior in Mice. Mediators Inflamm. 2013, 2013, 271359. [Google Scholar] [CrossRef]
  180. Lee, J.W.; Lee, Y.K.; Yuk, D.Y.; Choi, D.Y.; Ban, S.B.; Oh, K.W.; Hong, J.T. Neuro-Inflammation Induced by Lipopolysaccharide Causes Cognitive Impairment through Enhancement of Beta-Amyloid Generation. J. Neuroinflamm. 2008, 5, 37. [Google Scholar] [CrossRef] [Green Version]
  181. Lopez-Ramirez, M.A.; Wu, D.; Pryce, G.; Simpson, J.E.; Reijerkerk, A.; King-Robson, J.; Kay, O.; de Vries, H.E.; Hirst, M.C.; Sharrack, B.; et al. MicroRNA-155 Negatively Affects Blood-Brain Barrier Function during Neuroinflammation. FASEB J. 2014, 28, 2551–2565. [Google Scholar] [CrossRef]
  182. Ning, Q.; Liu, Z.; Wang, X.; Zhang, R.; Zhang, J.; Yang, M.; Sun, H.; Han, F.; Zhao, W.; Zhang, X. Neurodegenerative Changes and Neuroapoptosis Induced by Systemic Lipopolysaccharide Administration Are Reversed by Dexmedetomidine Treatment in Mice. Neurol. Res. 2017, 39, 357–366. [Google Scholar] [CrossRef]
  183. Kwon, H.S.; Koh, S.-H. Neuroinflammation in Neurodegenerative Disorders: The Roles of Microglia and Astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
  184. Wyss-Coray, T.; Mucke, L. Inflammation in Neurodegenerative Disease—A Double-Edged Sword. Neuron 2002, 35, 419–432. [Google Scholar] [CrossRef] [Green Version]
  185. Huang, Y.; Hu, S.; Li, Y.; Xue, D.; Wu, X. Dexmedetomidine, an Alpha 2a Adrenergic Receptor Agonist, Mitigates Experimental Autoimmune Encephalomyelitis by Desensitization of CXCR7 in Microglia. Biochemistry 2018, 57, 4197–4205. [Google Scholar] [CrossRef]
  186. Freeman, L.C.; Ting, J.P.-Y. The Pathogenic Role of the Inflammasome in Neurodegenerative Diseases. J. Neurochem. 2016, 136 (Suppl. 1), 29–38. [Google Scholar] [CrossRef]
  187. Schweneker, K.; Gorka, O.; Schweneker, M.; Poeck, H.; Tschopp, J.; Peschel, C.; Ruland, J.; Gross, O. The Mycobacterial Cord Factor Adjuvant Analogue Trehalose-6,6′-Dibehenate (TDB) Activates the Nlrp3 Inflammasome. Immunobiology 2013, 218, 664–673. [Google Scholar] [CrossRef]
  188. Meihe, L.; Shan, G.; Minchao, K.; Xiaoling, W.; Peng, A.; Xili, W.; Jin, Z.; Huimin, D. The Ferroptosis-NLRP1 Inflammasome: The Vicious Cycle of an Adverse Pregnancy. Front. Cell Dev. Biol. 2021, 9, 2137. [Google Scholar] [CrossRef]
  189. Kaushal, V.; Dye, R.; Pakavathkumar, P.; Foveau, B.; Flores, J.; Hyman, B.; Ghetti, B.; Koller, B.H.; LeBlanc, A.C. Neuronal NLRP1 Inflammasome Activation of Caspase-1 Coordinately Regulates Inflammatory Interleukin-1-Beta Production and Axonal Degeneration-Associated Caspase-6 Activation. Cell Death Differ. 2015, 22, 1676–1686. [Google Scholar] [CrossRef] [Green Version]
  190. Thomas, P.G.; Dash, P.; Aldridge, J.R.; Ellebedy, A.H.; Reynolds, C.; Funk, A.J.; Martin, W.J.; Lamkanfi, M.; Webby, R.J.; Boyd, K.L.; et al. The Intracellular Sensor NLRP3 Mediates Key Innate and Healing Responses to Influenza A Virus via the Regulation of Caspase-1. Immunity 2009, 30, 566–575. [Google Scholar] [CrossRef] [Green Version]
  191. Ji, Y.-R.; Chen, Y.; Chen, Y.-N.; Qiu, G.-L.; Wen, J.-G.; Zheng, Y.; Li, X.-F.; Cheng, H.; Li, Y.-H.; Li, J. Dexmedetomidine Inhibits the Invasion, Migration, and Inflammation of Rheumatoid Arthritis Fibroblast-like Synoviocytes by Reducing the Expression of NLRC5. Int. Immunopharmacol. 2020, 82, 106374. [Google Scholar] [CrossRef]
  192. Petralia, M.C.; Battaglia, G.; Bruno, V.; Pennisi, M.; Mangano, K.; Lombardo, S.D.; Fagone, P.; Cavalli, E.; Saraceno, A.; Nicoletti, F.; et al. The Role of Macrophage Migration Inhibitory Factor in Alzheimer’s Disease: Conventionally Pathogenetic or Unconventionally Protective? Molcules 2020, 25, 291. [Google Scholar] [CrossRef] [Green Version]
  193. Tilstam, P.V.; Qi, D.; Leng, L.; Young, L.; Bucala, R. MIF Family Cytokines in Cardiovascular Diseases and Prospects for Precision-Based Therapeutics. Expert Opin. Ther. Targets 2017, 21, 671–683. [Google Scholar] [CrossRef]
  194. Stosic-Grujicic, S.; Stojanovic, I.; Nicoletti, F. MIF in Autoimmunity and Novel Therapeutic Approaches. Autoimmun. Rev. 2009, 8, 244–249. [Google Scholar] [CrossRef]
  195. Günther, S.; Fagone, P.; Jalce, G.; Atanasov, A.G.; Guignabert, C.; Nicoletti, F. Role of MIF and D-DT in Immune-Inflammatory, Autoimmune, and Chronic Respiratory Diseases: From Pathogenic Factors to Therapeutic Targets. Drug Discov. Today 2019, 24, 428–439. [Google Scholar] [CrossRef]
  196. Jankauskas, S.S.; Wong, D.W.L.; Bucala, R.; Djudjaj, S.; Boor, P. Evolving Complexity of MIF Signaling. Cell. Signal. 2019, 57, 76–88. [Google Scholar] [CrossRef]
  197. Li, S.; Nie, K.; Zhang, Q.; Guo, M.; Qiu, Y.; Li, Y.; Gao, Y.; Wang, L. Macrophage Migration Inhibitory Factor Mediates Neuroprotective Effects by Regulating Inflammation, Apoptosis and Autophagy in Parkinson’s Disease. Neuroscience 2019, 416, 50–62. [Google Scholar] [CrossRef]
  198. Benedek, G.; Meza-Romero, R.; Jordan, K.; Zhang, Y.; Nguyen, H.; Kent, G.; Li, J.; Siu, E.; Frazer, J.; Piecychna, M.; et al. MIF and D-DT Are Potential Disease Severity Modifiers in Male MS Subjects. Proc. Natl. Acad. Sci. USA 2017, 114, E8421–E8429. [Google Scholar] [CrossRef] [Green Version]
  199. Macrophage Migration Inhibitory Factor Potentiates Autoimmune-Mediated Neuroinflammation—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/23797673/ (accessed on 29 March 2022).
  200. Petralia, M.C.; Mazzon, E.; Fagone, P.; Basile, M.S.; Lenzo, V.; Quattropani, M.C.; Bendtzen, K.; Nicoletti, F. Pathogenic Contribution of the Macrophage Migration Inhibitory Factor Family to Major Depressive Disorder and Emerging Tailored Therapeutic Approaches. J. Affect. Disord. 2020, 263, 15–24. [Google Scholar] [CrossRef]
  201. Lee, S.-T.; Chu, K.; Im, W.-S.; Yoon, H.-J.; Im, J.-Y.; Park, J.-E.; Park, K.-H.; Jung, K.-H.; Lee, S.K.; Kim, M.; et al. Altered MicroRNA Regulation in Huntington’s Disease Models. Exp. Neurol. 2011, 227, 172–179. [Google Scholar] [CrossRef]
  202. Figueroa-Romero, C.; Hur, J.; Lunn, J.S.; Paez-Colasante, X.; Bender, D.E.; Yung, R.; Sakowski, S.A.; Feldman, E.L. Expression of MicroRNAs in Human Post-Mortem Amyotrophic Lateral Sclerosis Spinal Cords Provides Insight into Disease Mechanisms. Mol. Cell. Neurosci. 2016, 71, 34–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Putteeraj, M.; Fairuz, Y.M.; Teoh, S.L. MicroRNA Dysregulation in Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2017, 16, 1000–1009. [Google Scholar] [CrossRef] [PubMed]
  204. Ren, K.; Li, T.; Zhang, W.; Ren, J.; Li, Z.; Wu, G. MiR-199a-3p Inhibits Cell Proliferation and Induces Apoptosis by Targeting YAP1, Suppressing Jagged1-Notch Signaling in Human Hepatocellular Carcinoma. J. Biomed. Sci. 2016, 23, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Xu, M.; Zhang, D.-F.; Luo, R.; Wu, Y.; Zhou, H.; Kong, L.-L.; Bi, R.; Yao, Y.-G. A Systematic Integrated Analysis of Brain Expression Profiles Reveals YAP1 and Other Prioritized Hub Genes as Important Upstream Regulators in Alzheimer’s Disease. Alzheimers Dement. 2018, 14, 215–229. [Google Scholar] [CrossRef]
  206. Guo, H.; Tian, J.; Zhu, J.; Li, L.; Sun, K.; Shao, S.; Cui, G. Electroacupuncture Suppressed Neuronal Apoptosis and Improved Cognitive Impairment in the AD Model Rats Possibly via Downregulation of Notch Signaling Pathway. Evid. Based Complement. Alternat. Med. 2015, 2015, e393569. [Google Scholar] [CrossRef]
  207. Sun, W.; Zhao, J.; Li, C. Dexmedetomidine Provides Protection Against Hippocampal Neuron Apoptosis and Cognitive Impairment in Mice with Alzheimer’s Disease by Mediating the MiR-129/YAP1/JAG1 Axis. Mol. Neurobiol. 2020, 57, 5044–5055. [Google Scholar] [CrossRef]
  208. Latini, A.; Borgiani, P.; Novelli, G.; Ciccacci, C. MiRNAs in Drug Response Variability: Potential Utility as Biomarkers for Personalized Medicine. Pharmacogenomics 2019, 20, 1049–1059. [Google Scholar] [CrossRef]
  209. Thummel, K.E.; Lin, Y.S. Sources of Interindividual Variability. Methods Mol. Biol. 2014, 1113, 363–415. [Google Scholar] [CrossRef]
  210. Turner, R.M.; Park, B.K.; Pirmohamed, M. Parsing Interindividual Drug Variability: An Emerging Role for Systems Pharmacology. Wiley Interdiscip. Rev. Syst. Biol. Med. 2015, 7, 221–241. [Google Scholar] [CrossRef]
  211. Baer-Dubowska, W.; Majchrzak-Celińska, A.; Cichocki, M. Pharmocoepigenetics: A New Approach to Predicting Individual Drug Responses and Targeting New Drugs. Pharmacol. Rep. 2011, 63, 293–304. [Google Scholar] [CrossRef]
  212. Li, M.-P.; Hu, Y.-D.; Hu, X.-L.; Zhang, Y.-J.; Yang, Y.-L.; Jiang, C.; Tang, J.; Chen, X.-P. MiRNAs and MiRNA Polymorphisms Modify Drug Response. Int. J. Environ. Res. Public Health 2016, 13, 1096. [Google Scholar] [CrossRef] [Green Version]
  213. Yu, D.; Green, B.; Marrone, A.; Guo, Y.; Kadlubar, S.; Lin, D.; Fuscoe, J.; Pogribny, I.; Ning, B. Suppression of CYP2C9 by MicroRNA Hsa-MiR-128-3p in Human Liver Cells and Association with Hepatocellular Carcinoma. Sci. Rep. 2015, 5, 8534. [Google Scholar] [CrossRef]
  214. Xu, K.; Liang, X.; Shen, K.; Cui, D.; Zheng, Y.; Xu, J.; Fan, Z.; Qiu, Y.; Li, Q.; Ni, L.; et al. MiR-297 Modulates Multidrug Resistance in Human Colorectal Carcinoma by down-Regulating MRP-2. Biochem. J. 2012, 446, 291–300. [Google Scholar] [CrossRef]
  215. Kuang, W.-H.; Dong, Z.-Q.; Tian, L.-T.; Li, J. MicroRNA-451a, MicroRNA-34a-5p, and MicroRNA-221-3p as Predictors of Response to Antidepressant Treatment. Braz. J. Med. Biol. Res. 2018, 51, e7212. [Google Scholar] [CrossRef] [Green Version]
  216. Graziano, F.; Canestrari, E.; Loupakis, F.; Ruzzo, A.; Galluccio, N.; Santini, D.; Rocchi, M.; Vincenzi, B.; Salvatore, L.; Cremolini, C.; et al. Genetic Modulation of the Let-7 MicroRNA Binding to KRAS 3’-Untranslated Region and Survival of Metastatic Colorectal Cancer Patients Treated with Salvage Cetuximab-Irinotecan. Pharm. J. 2010, 10, 458–464. [Google Scholar] [CrossRef] [Green Version]
  217. Boni, V.; Zarate, R.; Villa, J.C.; Bandrés, E.; Gomez, M.A.; Maiello, E.; Garcia-Foncillas, J.; Aranda, E. Role of Primary MiRNA Polymorphic Variants in Metastatic Colon Cancer Patients Treated with 5-Fluorouracil and Irinotecan. Pharm. J. 2011, 11, 429–436. [Google Scholar] [CrossRef]
  218. Alacam, H.; Akgun, S.; Akca, H.; Ozturk, O.; Kabukcu, B.B.; Herken, H. MiR-181b-5p, MiR-195-5p and MiR-301a-3p Are Related with Treatment Resistance in Schizophrenia. Psychiatry Res. 2016, 245, 200–206. [Google Scholar] [CrossRef]
  219. Duroux-Richard, I.; Pers, Y.-M.; Fabre, S.; Ammari, M.; Baeten, D.; Cartron, G.; Touitou, I.; Jorgensen, C.; Apparailly, F. Circulating MiRNA-125b Is a Potential Biomarker Predicting Response to Rituximab in Rheumatoid Arthritis. Mediators Inflamm. 2014, 2014, 342524. [Google Scholar] [CrossRef]
  220. Luo, Y.; Wang, L.; Li, G.W.; Yang, Z.Y.; Lin, X.Z. [Dexmedetomidine up-regulates peripheral blood microRNA 183 to improve remifentanil-related hyperalgesia]. Zhonghua Yi Xue Za Zhi 2021, 101, 1246–1249. [Google Scholar] [CrossRef]
  221. Duan, K.M.; Fang, C.; Yang, S.Q.; Yang, S.T.; Xiao, J.D.; Chang, H.; Lin, G.X.; Zhang, L.B.; Peng, M.C.; Liu, Z.Q.; et al. Genetic Polymorphism of Rs13306146 Affects A2AAR Expression and Associated With Postpartum Depressive Symptoms in Chinese Women Who Received Cesarean Section. Front. Genet. 2021, 12, 959. [Google Scholar] [CrossRef]
  222. Yu, H.-Y.; Wang, S.-Y.; Quan, C.-X.; Fang, C.; Luo, S.-C.; Li, D.-Y.; Zhen, S.-S.; Ma, J.-H.; Duan, K.-M. Dexmedetomidine Alleviates Postpartum Depressive Symptoms Following Cesarean Section in Chinese Women: A Randomized Placebo-Controlled Study. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2019, 39, 994–1004. [Google Scholar] [CrossRef]
  223. Yang, X.; Chen, H.; Chen, Y.; Birnbaum, Y.; Liang, R.; Ye, Y.; Qian, J. Circulating MiRNA Expression Profiling and Target Prediction in Patients Receiving Dexmedetomidine. Cell. Physiol. Biochem. 2018, 50, 552–568. [Google Scholar] [CrossRef] [PubMed]
  224. Pignataro, G. Emerging Role of MicroRNAs in Stroke Protection Elicited by Remote Postconditioning. Front. Neurol. 2021, 12, 1833. [Google Scholar] [CrossRef] [PubMed]
  225. Joilin, G.; Leigh, P.N.; Newbury, S.F.; Hafezparast, M. An Overview of MicroRNAs as Biomarkers of ALS. Front. Neurol. 2019, 10, 186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Banerjee, A.; Chokkalla, A.K.; Shi, J.J.; Lee, J.; Venna, V.R.; Vemuganti, R.; McCullough, L.D. Microarray Profiling Reveals Distinct Circulating MiRNAs in Aged Male and Female Mice Subjected to Post-Stroke Social Isolation. Neuromol. Med. 2021, 23, 305–314. [Google Scholar] [CrossRef]
  227. Baumann, V.; Winkler, J. MiRNA-Based Therapies: Strategies and Delivery Platforms for Oligonucleotide and Non-Oligonucleotide Agents. Future Med. Chem. 2014, 6, 1967–1984. [Google Scholar] [CrossRef] [Green Version]
  228. Li, G.; Morris-Blanco, K.C.; Lopez, M.S.; Yang, T.; Zhao, H.; Vemuganti, R.; Luo, Y. Impact of MicroRNAs on Ischemic Stroke: From Pre- to Post-Disease. Prog. Neurobiol. 2018, 163–164, 59–78. [Google Scholar] [CrossRef]
  229. Cuomo, O.; Cepparulo, P.; Anzilotti, S.; Serani, A.; Sirabella, R.; Brancaccio, P.; Guida, N.; Valsecchi, V.; Vinciguerra, A.; Molinaro, P.; et al. Anti-MiR-223-5p Ameliorates Ischemic Damage and Improves Neurological Function by Preventing NCKX2 Downregulation after Ischemia in Rats. Mol. Ther. Nucleic Acids 2019, 18, 1063–1071. [Google Scholar] [CrossRef] [Green Version]
  230. Dirnagl, U. Bench to Bedside: The Quest for Quality in Experimental Stroke Research. J. Cereb. Blood Flow Metab. 2006, 26, 1465–1478. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Changes in brain-related miRNAs expression after DEX treatment associated with neuroprotective effects in brain injuries. Upward and downward arrows represent the up-/down-regulation of miRNAs in response to DEX treatment. References: Decreasing the Anesthetics Neurotoxicity: [31,69,144,145]; Alleviating Postoperative Cognitive Dysfunctions: [28,70,74,75,76,146]; Modulation of Neurodegenerative Disorders: [29,71,147,148]; Modulation of Ischemic Brain Injury: [30,72,73,83,149,150,151,152,153,154,155,156,157,158,159,160,161,162]; Isolation of RNA from hippocampal neuron cells and brain homogenate was performed by using Trizol reagent method and total RNA was measured by using small RNA sequencing or real-time quantitative PCR (RTqPCR). The RNA quality and quantity of the RNA were analyzed with a spectrophotometer. The differentially expressed microRNAs were screened out by using Microarray analysis. In this context, the experimental studies aimed to investigate the neuroprotective potential of DEX by targeting miRNAs expression have been addressed to four topics, which will further explore: (1) modulation of ischemic brain injury, (2) decreasing the neurotoxicity of anesthetics, (3) reducing the postoperative cognitive dysfunction, including delirium or cognitive dysfunction, and (4) modulation of neurodegenerative diseases.
Figure 1. Changes in brain-related miRNAs expression after DEX treatment associated with neuroprotective effects in brain injuries. Upward and downward arrows represent the up-/down-regulation of miRNAs in response to DEX treatment. References: Decreasing the Anesthetics Neurotoxicity: [31,69,144,145]; Alleviating Postoperative Cognitive Dysfunctions: [28,70,74,75,76,146]; Modulation of Neurodegenerative Disorders: [29,71,147,148]; Modulation of Ischemic Brain Injury: [30,72,73,83,149,150,151,152,153,154,155,156,157,158,159,160,161,162]; Isolation of RNA from hippocampal neuron cells and brain homogenate was performed by using Trizol reagent method and total RNA was measured by using small RNA sequencing or real-time quantitative PCR (RTqPCR). The RNA quality and quantity of the RNA were analyzed with a spectrophotometer. The differentially expressed microRNAs were screened out by using Microarray analysis. In this context, the experimental studies aimed to investigate the neuroprotective potential of DEX by targeting miRNAs expression have been addressed to four topics, which will further explore: (1) modulation of ischemic brain injury, (2) decreasing the neurotoxicity of anesthetics, (3) reducing the postoperative cognitive dysfunction, including delirium or cognitive dysfunction, and (4) modulation of neurodegenerative diseases.
Ijms 23 05452 g001
Figure 2. Molecular pathways shared between miRNAs and their associated signaling pathways for promoting dexmedetomidine’s neuroprotective effects. Brain injuries that involved miRNA regulation, in which dexmedetomidine poses neuroprotection refer to: ischemic brain injury, anesthetics-induced neurotoxicity, postoperative cognitive dysfunction, and neurodegenerative diseases. Dexmedetomidine regulates a variety of neuropathogenic pathways which involves miRNAs, including: neuroinflammation, oxidative stress, and apoptosis, with a few contributions of mitophagy, autophagy, parthanatos, and mitochondrial-associated processes. miRNAs color-coded in blue represent those that are upregulated after DEX treatment in brain disorders, and miRNAs in red represent those that are downregulated after DEX treatment.
Figure 2. Molecular pathways shared between miRNAs and their associated signaling pathways for promoting dexmedetomidine’s neuroprotective effects. Brain injuries that involved miRNA regulation, in which dexmedetomidine poses neuroprotection refer to: ischemic brain injury, anesthetics-induced neurotoxicity, postoperative cognitive dysfunction, and neurodegenerative diseases. Dexmedetomidine regulates a variety of neuropathogenic pathways which involves miRNAs, including: neuroinflammation, oxidative stress, and apoptosis, with a few contributions of mitophagy, autophagy, parthanatos, and mitochondrial-associated processes. miRNAs color-coded in blue represent those that are upregulated after DEX treatment in brain disorders, and miRNAs in red represent those that are downregulated after DEX treatment.
Ijms 23 05452 g002
Table 1. Main effects of dexmedetomidine administration.
Table 1. Main effects of dexmedetomidine administration.
EffectMechanism/ConsequencesReferences
Sedativeactivates central pre- and postsynaptic α2ARs in the locus coeruleus
decreases norepinephrine release
mimics physiological sleep
[39]
Analgesicactivates α2ARs in the spinal cord–dorsal horn (locus coeruleus)
decreases the release of substance P
[54,55]
Anti-inflammatory↓ TNF-α, IL-6, IL-8, IL-1β
↑ IL-10
↑ NF-κB and CD4 cells
↓ CD8 cells
↓ plasma level of epinephrine, norepinephrine, and cortisol
increases the degradation of NLRP3 inflammasome (a molecule with a role in activating caspase-1 and secreting proinflammatory cytokines such as IL-1β or IL-18)
[49,56,57,58,59]
Antioxidantdecreased ROS and increased SOD, GSH, and CAT after LPS or acetaminophen-induced oxidative stress[60,61]
Increases
blood pressure after rapid bolus injection
changes the expression α2 AR from the subtype α2A to a1 and α2B.
α2A ARs are involved in vasodilation by NO production
activates phospholipase A and increases the vascular response to angiotensin and catecholamines.
[54,62]
Decreases blood pressure after intravenous continuous infusionactivates presynaptic α2ARs
decreases norepinephrine release
decreases sympathetic outflow
[63,64]
Abbreviations: α2ARs, α2 adrenergic receptor; CAT, Catalase; CD 4, 8, cluster of differentiation 4, 8; GSH, Glutathione; IL-6, -8, -1β, 10, 18, Interleukin 6, -8, -1β, 10; LPS, Lipopolysaccharide; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3, NLR family pyrin domain containing 3; NO, nitric oxide; ROS, Reactive oxygen species; SOD, Superoxide dismutase; TNF-α, Tumor necrosis factor-α.
Table 2. End-organ effects of DEX.
Table 2. End-organ effects of DEX.
StudymiRNARole of miRNAEffect of DEX on miRNABiological Effect of miRNAReferences
In vitro
Human neuroblastoma SH-SY5Y cells—bupivacaine-induced
Neuron injury
miR-7-5pImproved mitochondrial membrane potential
Increased antioxidative enzymes and reduced apoptosis
Impaired bupivacaine-induced
parthanatos
UpregulatedInhibiting PARP1
↓ SOD1, CAT
↑ GPx
↓ Bax, cleaved caspase 3
[69]
In vivo
Sprague-Dawley rats—postoperative cognitive dysfunction induced by sevoflurane
miR-129involved in cognitive dysfunction-related diseasesIncreases expression in hippocampus and cortical neuronsInhibiting TLR4
and NF-κB p65 phosphorylation
[70]
In vivo:
APPswe/PS1dE9-induced AD mice
miR-214-5pinvolved in reducing apoptosis
neuroinflammation and oxidative stress of hippocampal neurons
Attenuating cognitive impairments of AD mice
Upregulated expression in hippocampal neuronsDownregulated SUZ12[71]
In vivo
Newborn Sprague-Dawley rats—induced HIBD
miR-29a-3pElevated MiR-29a-3p reduces inflammation and apoptosis rate of hippocampal neurons
increase spatial learning and memory abilities
Improved brain atrophy and alleviated pathological
changes in ischemic rat brain
UpregulatedNegatively regulating HDAC4
↓ IL-6, IL-8, and TNFα
↑ IL-10
[72]
In vivo
Wistar rats—induced CI/R injury
In vitro
Rat hippocampal neurons cells
miR-205-5pMay inhibit the inflammatory response and oxidative stress
reduced brain infraction volume, improved neurological function score in acute cerebral ischemia induced in rats
Upregulated expression in hippocampal neuronsNegatively regulating HMGB1
↑ SOD,
↓ ROS, MDA
↑ Nrf2, GR, GPX, HO-1, CAT
↓ IL-1β, IL-6, and TNF-α
[30]
In vivo
Sprague-Dawley rats
—CI/R injury
In vitro
Primary hippocampal neuron cell culture exposed to OGD/R treatment
miR-381inhibited inflammation response and neuron cell apoptosis
improved neurological function
UpregulatedInhibiting IRF4-IL-9 [73]
In vivo
C57BL/6 mice—sevoflurane-induced
POCD
In vitro
primary hippocampal neurons from newborn
mice
involved in apoptosis, neuroinflammation, and DNA damage repair
Alleviates Cognitive Dysfunction
UpregulatedInhibiting EGR1/p53
Disrupts interaction of EGR1 with p53
Decreasing EGR1-mediated p53 transcription
[74]
In vitro cell culture (Pheochromocytoma cell line—PC12)—ropivacaine-induced neuronal injurymay reduce the proliferation and apoptosis of ropivacaine-induced PC12 cellsUpregulatedNegatively regulating LRRC4 expression
↓ Cleaved-Caspase-3
↑ Bcl-2
[31]
In vivo
Wistar rats—lipopolysaccharide (LPS)-induced inflammation
miR-21-5pinvolved in inflammation, autophagy, and apoptosisDecreased the expression in the spleen and hippocampusAttenuated PARP cleavage in the spleen[75]
miR-204-5p
miR-30a-5p
In vivo
Wistar rats—LPS-induced neuroinflammation
miR-124involved in neuronal differentiationDecreased the expression in the hippocampus and cortexTargeted SHIP1,
SOCS1
[76]
miR-132involved in the inflammatory response in alveolar macrophages
potentiated cholinergic anti-inflammatory pathway
miR-155involved in pro- and anti-inflammatory mechanisms
In vivo
40 patients undergoing off-pump coronary artery bypass grafting
miR-320aassociated with cell proliferationDecreased miRNA-320 expression level in the blood of patients after treatment with DEXStimulated NGB expression[77]
In vivo
Adult male Sprague-Dawley rats- chronic inflammatory visceral pain
miR-211Down-regulated in TNBS-induced chronic inflammatory visceral painUpregulated after DEX treatmentNegatively regulating ERK expression
↓ IL-1β, TNF-α, and IL-6
[78]
In vivo
Adult male Sprague-Dawley rats
In vitro
primary myoblasts from young mice

In vivo
Coopworth ewes sheep
rno-miR-434-3pmiR-434-3p protects myocytes from apoptosisUpregulatedTargeted EIF5A G2E3, DCAF6, and TMEM68
↓ caspases-3, −8 and −9
[79,80]
rno-miR-3596dTargeted G2E3, DCAF6, TMEM68, ATAD2B, NPY1R, SRSF1, ITGA6, MORC3, and RSF1
rno-miR-496-5pControlled mTOR pathway
Targeted ATAD2B, NPY1R, SRSF1, ITGA6, MORC3, and RSF1
rno-miR-7a-2-3pNegatively regulating PARP expression
rno-miR-702-3p
rno-miR-208b-3pUpregulated in heart failureDownregulatedTargeted CSNK2A2/NLK
Promoted Wnt/β-catenin pathway
↑ Bcl-2
[79,81]
In vivo
Sprague-Dawley rats—MI/R injury
miR-346-3pmiRNA-346
suppresses infarct size
inhibits myocardial cell apoptosis
may protect against MI/R injury
Upregulated the expressionNegatively regulated CAMK2D expression
↓ NF-κB
↓ NLRP3 inflammasome
[67]
In vivo
LPS-induced acute lung injury of mice
miR-223-3pThe absence is associated with severe lung inflammation
pulmonary up-regulation of this miRNA in mice may provide protection during acute lung injury due to various causes (eg. mechanical ventilation)
UpregulatedNegatively regulated HDAC4 expression
↓ TLR4
↓ NF-κB
[82]
In vivo
neonatal mice model—induced HIBD
mmu-miR-128Increased the neuroprotective effects of DEX against ischemic brain injury
attenuated nerve cell apoptosis
enhanced learning and memory abilities
reduced left-brain water content
UpregulatedNegatively regulating WNT1 expression[83]
Abbreviations: AD, Alzheimer’s Disease; ATAD2B, ATPase Family AAA Domain Containing 2B; Bax, BCL2 Associated X, Apoptosis Regulator; Bcl-2, B-cell lymphoma; CAT, Catalase; CAMK2D, Calcium/calmodulin dependent protein kinase II delta; CI/R, cerebral ischemia/reperfusion; CSNK2A2, Casein Kinase 2 Alpha 2; DCAF6, DDB1 And CUL4 Associated Factor 6; EGR1, Early Growth Response 1; EIF5A, Eukaryotic Translation Initiation Factor 5A; ERK, extracellular signal-regulated kinases; G2E3, G2/M-Phase Specific E3 Ubiquitin Protein Ligase; GR, Glutathione reductase; GPX1, glutathione peroxidase-1; HDAC4, Histone deacetylase 4; HIBD, Hypoxic Ischemic Brain Damage; HMBG1, High mobility group box 1; HO-1, Heme oxygenase-1; IRF4, Interferon Regulatory Factor 4; IL-1β, -6, Interleukin -1β, -6; ITGA6, Integrin Subunit Alpha 6; LPS, Lipopolysaccharide; LRRC4, Leucine Rich Repeat Containing 4; MCAO, Middle Cerebral Artery Occlusion; MI/R, myocardial ischemia/reperfusion; MORC3, MORC Family CW-Type Zinc Finger 3; NGB, Neuroglobin; NLK, nemo-like kinase; NLRP3, NLR family pyrin domain containing; NPY1R, Neuropeptide Y Receptor Y1; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, Nuclear factor-erythroid factor 2-related factor 2; OGD/R, oxygen-glucose deprivation/Reperfusion; p53, Tumor protein P53; PARP1, Poly(ADP-ribose) polymerase 1; POCD, Postoperative Cognitive Disorder; ROS, Reactive oxygen species; RSF1, Remodeling And Spacing Factor 1; SHIP1, phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase 1; SOCS1, suppressor of cytokine signaling 1; SOD, Superoxide dismutase; SRSF1, Serine And Arginine Rich Splicing Factor 1; SUZ12, Suppressor of ZESTE 12; TMEM68, Transmembrane Protein 68; TLR4, Toll-like receptor 4; TNBS, trinitrobenzene sulfonic acid, TNF-α, Tumor necrosis factor-α.
Table 3. Circulating human miRNAs expression profiling in DEX treatment, emerging as promising genomic biomarkers of DEX response in clinical studies.
Table 3. Circulating human miRNAs expression profiling in DEX treatment, emerging as promising genomic biomarkers of DEX response in clinical studies.
miRNAsType of StudyInterventionObservationsReferences
miR-32040 patientsOff-pump coronary artery bypass graftingDownregulated levels after treatment[77]
miR-18380 patientsLaparoscopic cholecystectomy and oophorocystectomy surgeryOverexpressed after treatment[220]
miR-646--α2AAR s13306146 polymorphism568 cesarean section patientsChinese women who received cesarean sectionmiR-646 level
in α2AAR s13306146 polymorphism
affects α2AAR Postpartum Depressive Symptoms
[221]
miR-30a-5p, -101-3p, -140-3p and -141-3p133 pediatric patientsPreoperative sedation for different proceduresIncreased levels of miR-101-3p and 140-3p in respondents
and downregulated levels of miR-101-3p and miR-30a-5p in hypotension and bradycardia patients
[33]
hsa-miR-4508, -novel-chr8_87373, -30a-3p, -novel-chr16_26099, -4306, -744-5p, -320a, -novel-chr9_90035, -101-3p, -150-5p, -342-3p, and-140-3pthree patientsElective surgeryFive miRNAs upregulated (hsa-miR-4508, -novel-chr8_87373, -30a-3p, -novel-chr16_26099, -4306,) and seven miRNAs downregulated (hsa-miR-744-5p, -320a, -novel-chr9_90035, -101-3p, -150-5p, -342-3p, and -140-3p) after DEX treatment[223]
Abbreviations: α2AAR, α2-adrenergic receptors; miRNAs, miRs; DEX, dexmedetomidine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Burlacu, C.-C.; Neag, M.-A.; Mitre, A.-O.; Sirbu, A.-C.; Badulescu, A.-V.; Buzoianu, A.-D. The Role of miRNAs in Dexmedetomidine’s Neuroprotective Effects against Brain Disorders. Int. J. Mol. Sci. 2022, 23, 5452. https://doi.org/10.3390/ijms23105452

AMA Style

Burlacu C-C, Neag M-A, Mitre A-O, Sirbu A-C, Badulescu A-V, Buzoianu A-D. The Role of miRNAs in Dexmedetomidine’s Neuroprotective Effects against Brain Disorders. International Journal of Molecular Sciences. 2022; 23(10):5452. https://doi.org/10.3390/ijms23105452

Chicago/Turabian Style

Burlacu, Codrin-Constantin, Maria-Adriana Neag, Andrei-Otto Mitre, Alexandru-Constantin Sirbu, Andrei-Vlad Badulescu, and Anca-Dana Buzoianu. 2022. "The Role of miRNAs in Dexmedetomidine’s Neuroprotective Effects against Brain Disorders" International Journal of Molecular Sciences 23, no. 10: 5452. https://doi.org/10.3390/ijms23105452

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