The Role of microRNA in Anaesthetics-induced Brain Injury: A Narrative Review
PDF
Cite
Share
Request
Review Article
VOLUME: 53 ISSUE: 5
P: 187 - 196
October 2025

The Role of microRNA in Anaesthetics-induced Brain Injury: A Narrative Review

Turk J Anaesthesiol Reanim 2025;53(5):187-196
1. Dokuz Eylül University Faculty of Medicine, Department of Anaesthesiology and Reanimation, İzmir, Türkiye
2. Dokuz Eylül University, İzmir Biomedicine and Genome Center, İzmir International Biomedicine and Genome Institute, İzmir, Türkiye
3. Dokuz Eylül University Health Sciences Institute, İzmir Biomedicine and Genome Center, Department of Neuroscience, İzmir, Türkiye
No information available.
No information available
Received Date: 04.09.2024
Accepted Date: 14.01.2025
Online Date: 14.10.2025
Publish Date: 14.10.2025
E-Pub Date: 08.05.2025
PDF
Cite
Share
Request

Abstract

Anaesthetics are commonly used agents during medical interventions and surgeries. Exposure to anaesthetic agents in late intrauterine life or early childhood may cause neurodegeneration in developing brains. Neuroapoptosis and neural inhibition provided by several mechanisms and microRNAs (miRNAs) have crucial roles in this milieu. miRNAs have critical roles in response to anaesthetic exposure. Through this review, we performed a systematic search of the PubMed database for studies on the role of anaesthetics in the brain and their relation with miRNAs. The terms “anesthetic”, “miRNA”, and “brain” were searched. Here we summarized the roles and interactions of miRNAs under exposure to anaesthetics in vivo and in vitro studies. Anaesthetic agents studied included sevoflurane, isoflurane, ketamine, and propofol. Many microRNAs were identified to have regulatory roles in anaesthesia-induced neurotoxicity. The literature study supports the idea that miRNAs play crucial functions in neuroprotection and neurotoxicity in anaesthesia administration. The exact role and implication of miRNA in anaesthesia neurotoxicity needs to be elucidated to gain more knowledge about the area. Several gaps in knowledge should be filled by conducting basic, clinical, and translational analyses in the future to decipher the definite role of miRNAs and their functions in the context of anaesthesia-induced neurotoxicity.

Keywords:
Anaesthesia, anaesthetic agents, miRNA, neurotoxicity

Main Points

• Anaesthetics cause microRNA (miRNA) expression changes in the brain.

• The exact mechanism of anaesthesia-induced neurotoxicity is still not certain. This review summarizes the miRNA expression changes that can be related to anaesthesia-induced neurotoxicity.

• miRNAs are not only related to neurotoxicity. Their expression changes can also result in the neuroprotection, addictive, and antidepressant properties of anaesthetics.

• It is too early to say which miRNA changes its expression after anaesthesia. But further studies and analysis may highlight one or a couple miRNAs related to anaesthesia-induced neurotoxicity. These could be the therapeutic candidates for neuroprotection.

Introduction

Globally, over 300 million surgical operations and diagnostic or interventional tests that require anaesthesia or sedation occur annually. Recent evidence suggests that anaesthetics may cause neurodegeneration in the brain. In this review, our primary goal was to search for articles related to the role of microRNAs (miRNAs) in anaesthesia-induced neurotoxicity (AIN) and our secondary goal was to summarize articles about anaesthesia and miRNA changes in the brain.

In this study, we searched the PubMed database for studies on the role of anaesthetics in the brain and their relationship to miRNAs. The search was conducted in April 2022. The search terms were “anesthetic and miRNA and brain”, “miRNA”, and “brain”. Articles were screened by the authors to exclude duplicates and those that did not meet the inclusion criteria. The inclusion criteria was the articles about anaesthetics and miRNAs in the brain. The exclusion criteria were abstracts, conference lectures, case reports, reviews, biographies, and editorials. In addition, articles not published in English, those for which full texts were not available, and those not related to the brain were excluded. Data regarding the miRNAs involved, their expression status, and their roles in AIN were retrieved (Figure 1). This study aimed to identify miRNAs and their pathways associated with AIN. This review highlights possible mechanisms and miRNAs involved in AIN. This will help researchers to design future studies on this topic and to study possible protective agents.

miRNA

miRNAs are small RNA molecules with a length of 19-25 nucleotides. Their main function is to regulate gene expression post-transcriptionally, and one miRNA can interact with hundreds of mRNAs.1 The first miRNA was discovered in 1993 by Lee et al.2 It has been shown that a smaller RNA product may bind to several sites of the 3′ untranslated region (UTR) of LIN-14 mRNA and mitigate LIN-14 protein expression without affecting its mRNA levels.2

miRNA Biogenesis

Canonical miRNA biogenesis begins with the transcription of miRNA genes into primary miRNAs (pri-miRNAs) by RNA polymerase II. Then, pri-miRNA is cleaved into a precursor RNA, 70-120 nucleotides in length, by microprocessor, a multiprotein complex containing the ribonuclease enzyme Drosha and the RNA-binding protein DiGeorge syndrome critical region 8 (DGCR8). Newly produced pre-miRNAs are transported into the cytoplasm by exportin 5, which is a Ran-dependent nuclear transport receptor protein. Pre-miRNAs mature into 19-25 nucleotide-long duplex miRNAs by removing the terminal loop with Dicer-1, an RNase III enzyme. Duplexes are then separated, and mature miRNAs may interact with Argonaute proteins to form an RNA-inducing silencing complex, which can bind to the 3´ UTR of mRNAs and exert a silencing effect.1

Role of miRNAs in Brain

miRNAs are found in the human brain and have distinct functions. They contribute to development, physiological functions, and human cognition by regulating transcription in specific regions of the brain.3 miR-124, one of the most abundant miRNAs in the brain, affects neural lineage differentiation by downregulating multiple mRNAs. Several studies have demonstrated that miRNAs show expression changes in many neurological diseases such as autism spectrum disorders, schizophrenia, frontotemporal dementia, Alzheimer’s disease, Parkinson’s disease, and neuropsychiatric diseases.3 Discovering miRNA-mRNA interactions in the brain may shed light on drugs that affect the central nervous system, such as anaesthetics.

Sevoflurane

Sevoflurane is a commonly used inhalational anaesthetic, particularly during pediatric induction. Animal studies have widely reported sevoflurane-induced neuroapoptosis and cognitive dysfunction, especially during the brain growth spurt. The exact mechanism of sevoflurane-induced neurotoxicity (SIN) is unclear. In recent years, the roles of miRNAs and long non-coding RNAs (lncRNAs) in SIN have been studied. The hippocampus is the most studied part of the brain because it is closely associated with SIN.

Sevoflurane in vitro Studies

To investigate SIN and the role of miRNAs, E14 mESCs, primary neuron culture, SH-SY5Y neuroblastoma cells, NSC culture from neonatal rat hippocampus, and HN-h, HCN-2, and HEK293 cell lines were used. In these studies, let-7a, miR-27a-3p, miR-188-3p, miR-183, hsa-miR-302e, miR-34a, miR-19-3p, miR-101a-3p, miR-133a, miR-410-3p, miR-214, and miR-128-3p expression were investigated. Except for miR-410-3p, the expression of all miRNAs studied in vitro increased after sevoflurane exposure. Changes in miRNA expression inhibited self-renewal, proliferation, and differentiation and they induced apoptosis and neuroinflammation. Lin-28, PPAR-Ȣ, MDM2, Sox2, BDNF, OXR1, Wnt1, CCNA-2, CXCR5, Sox1, Peg13, and Sox13 were the target genes of miRNAs in vitro studies.4-14 A decrease in the expression of lncRNAs Gm15621 and Rik-203 was found to be related to SIN.11, 15

Sevoflurane in vivo Studies

Eight microarray studies investigated changes in miRNA expression after sevoflurane anaesthesia. Five of these studies used rats. Of these, three focused on the hazardous effects of sevoflurane on children. therefore, they involved 7-day-old (P7) rats in the study, while two studies included adult rats. Two studies included P7 mice, and the newest microarray study chose P5-6 Rhesus monkeys so the Rhesus monkeys can reflect the miRNA changes in the mammalian brain. Although the concentration of sevoflurane was similar between studies (1.9-2.5%), the duration of anaesthesia varied between two and eight hours. The exact duration of anaesthesia for SIN is not clear, but a longer anaesthesia time could be closely related to neurotoxicity. Therefore, the duration of anaesthesia may be associated with changes in the expression of different miRNAs. Brain sections that have been investigated for miRNA expression are essential for understanding miRNAs' role in brain function. Six of the eight microarray studies used the hippocampus, which is closely related to learning and memory disturbances, for miRNA expression. One study selected the left cortex, while another investigated the frontal cortex. The timing of analysis after anaesthesia can result in the detection of different miRNA expression changes. Lin et al.16 showed this in their study. They found that the expression changes of only three miRNAs in the whole brain and four miRNAs in the hippocampus persisted for 2-4 months after sevoflurane anaesthesia. The exact time interval for the transient changes in miRNAs is unknown; therefore, after a few hours, some of the miRNA expression may return to normal. In addition to these methodological differences in microarray studies, changes in miR-125b-5p expression were observed in four studies.17-20 Also, differential expression of miR-129-3p16,17 and miR-44818, 21 has been detected in more than one study after sevoflurane anaesthesia.

Other studies on this topic chose miRNAs that are known to be related to brain function or neurodevelopment and analyzed their expression. Studies have shown that sevoflurane exerts differential effects on miRNAs. However, it can increase the expression of some miRNAs and decrease the expression of others. In a previous study, lncRNA Rik-203’s target miR-4661-3p was investigated in P6 mice, but the researchers could find no change in the expression of miRNA after sevoflurane anaesthesia.11 In contrast, Su et al.12 showed that sevoflurane could decrease miR-410-3p expression and cause apoptosis. Another study chose the target miR-133a of lncRNA Gm15621 and found that its expression increased with increasing concentrations of sevoflurane.15 LncRNA Rik-203 and its target miR-101a-3p were investigated. After sevoflurane anaesthesia, expression of miR-101a-3p was increased, while the expression of Rik-203 decreased.11 Zhao et al.10 investigated the miRNAs that bind to Cyclin A2 protein, which is downregulated after sevoflurane anaesthesia, showing that miR-19-3p was increased. Zhao et al.9 investigated the role of miR-34a in cortical development and found that inhibition of the miRNA decreases SIN. Another study showed that an increase in miR-96 expression is related to apoptosis after sevoflurane anaesthesia.22 A high concentration of sevoflurane (4.8%) was found to be related to increased expression of miR-183, resulting in SIN.6 Wang et al.6 reported that increased expression of miR-188-3p in the hippocampus is related to sevoflurane-induced apoptosis. Likewise, increased expression of miR-27a-3p5 and miR-27b23 induces SIN via different pathways. In addition, a recent study showed that increased expression of miR-21413 plays a role in SIN. Unlike these studies, Shan et al.24 showed that decreased expression of miR-30a, miR-31, miR-190a, and miR-190b is related to SIN in old rats.

SIN Related Pathways

Many pathways have been implicated in SIN; therefore, different pathways related to miRNAs have been investigated. One of the most investigated genes is brain-derived neurotrophic factor (BDNF), which plays an important role in neural differentiation.7, 10, 11, 13, 18

Another commonly studied gene family is the Sox family, which plays a crucial role in cell renewal and differentiation.7, 11, 25 Neuroinflammation is one of the attributed reasons for SIN. miR-410-3p,12 miR-101a-3p,11 and miR-27a-3p5 are the miRNAs found to be related to SIN through neuroinflammation. Other studied pathways include the WNT signaling pathway,9, 26 phosphatidylinositol 3-kinase (PI3K) and protein kinase B signaling pathway,12 insulin-like growth factor 1,22 KEGG pathway,16 MDM2-p53 pathway,6 and LIMK1.23

SIN Related Learning Memory and Function

Learning and memory tests are ways to observe the SIN’s clinical expression and evaluate cognition. The Morris Water Maze (MWM) is the most common learning and memory test, in which an experimental animal learns to locate the swimming platform in a tank filled with water. Sun et al.26 used MWM and showed that the WNT signaling pathway is the most strongly associated pathway to sevoflurane-induced cognitive dysfunction. Another study found a relationship between miR-410-3p and decreased latency time, and increased platform crossing in MWM after sevoflurane anaesthesia.12 Similar to these studies, the link between miRNAs and sevoflurane-induced cognitive dysfunction has been investigated using MWM.5, 6, 9, 10, 22, 23 Zhao et al.10 showed that the upregulation of miR‐19‐3p is associated with sevoflurane-induced learning and memory deficit, using the plus-maze discriminative avoidance task to demonstrate this association. Fujimoto et al.18 reported that sevoflurane anaesthesia could cause subsequent behavioral disorders at open field tests and contextual and cued fear conditioning tests by increasing rno-miR-632.

SIN Related Concentrations in Neurotoxicity

The sevoflurane concentration used in these experiments is usually 1 MAC (2-3%), and the duration of anaesthesia varies between one-six hours.12,13,16-21,24,26 These studies proved that even a relatively short duration of sevoflurane anaesthesia can cause SIN in an experimental setting. Studies that used repeated doses of sevoflurane anaesthesia (1 MAC for 2 hours over 3 consecutive days) also showed changes in the expression of miRNAs in the brain, resulting in SIN.5, 9, 11, 25, 27 Only one study failed to show miRNA expression change with 0.5 and 1 MAC sevoflurane, while 4.8% sevoflurane increased miR-183 expression.6

miRNAs in Neuroprotective Role of Sevoflurane

In addition to its neurotoxic effects, sevoflurane exhibits neuroprotective properties under certain conditions. Shi et al.28 showed that sevoflurane preconditioning decreases the injury of cerebral ischemia by attenuating the upregulation of miR-15b. Similarly, Zhong et al.29 and Ding et al.30 reported the protective effect of sevoflurane on cerebral ischemia by altering the expression of miR-203 and miRNA-490-5p, respectively. Studies in glioma cell cultures have found that sevoflurane can suppress the proliferation, migration, and metastasis of glioma through miR-637,31 miR-124-3p,32 miR-146b-5p,33 miR-761,34 miR-34a-5p,35 and miR-7.36

Isoflurane

Isoflurane is an inhalational anaesthetic, many experimental studies demonstrated that it could cause apoptosis, neurotoxicity, and learning-memory deficits. Rapidly proliferating and aged neurons are particularly vulnerable to the neurotoxic effects of isoflurane. Since the hippocampus is closely related to learning and memory, it is one of the most studied brain regions.

Isoflurane in vitro Studies

Zhang et al.37 showed that isoflurane treatment represses the self-renewal of E14 embryonic stem cells through the mechanism involving miR-9’s direct target, E-cadherin. A study using hippocampal cell culture found that miR-448 increased after isoflurane anaesthesia, resulting in an increase in Bcl-x, a decrease in caspase-3, which causes neuronal apoptosis.38 Yang et al.39 reported that isoflurane can cause neuroapoptosis by decreasing miR-124 and BDNF in primary hippocampal neuron culture. Similarly, the increase in miR-142-5p expression after isoflurane was related to the inhibition of cell viability.40 The PTEN/PI3K/Akt pathway was investigated in a human neuroblastoma cell line, and the authors found that the decrease in miR-214 resulted in neuroapoptosis through this pathway.41

Isoflurane in vivo Studies

In a miRNA microarray study, the hippocampus, was investigated after isoflurane anaesthesia. The authors found that 21 miRNAs increased in expression and 17 miRNAs decreased. They also showed that four miRNAs (miR-146a, miR-9, miR-143, and let-7d) are associated with learning memory deficits in the MWM. Additionally, they reported a relationship between downregulation of let-7d and increased amyloid-β expression.42 Shan et al.24 showed that a decrease in miR-30a, miR-31, miR-190a, and miR-190b expression in the hippocampus and mPFC is related to neuroapoptosis and impaired recognition and spatial memory. Similar to these two studies, it was shown that an increase in miR-448 expression in aged rats was related to learning and memory deficits and neurotoxicity.38

A study found that isoflurane decreased miR-124 expression in the hippocampus, increased EGR1, and caused learning-memory deficits and neuroapoptosis by increasing pro-apoptotic factors (cleaved-caspase-3 and Bax) and decreasing the anti-apoptotic factor Bcl-2 in neonatal rats.39 Two other studies found an increase in miR-142-5p40 and miR-19143 expression in the hippocampus. Li et al.43 showed that BDNF was the target gene of miR-191, whose increased expression results in neurotoxicity and learning-memory deficits.

Li et al.44 showed that overexpression of miR-24 decreases oxidative stress-related molecules such as superoxide dismutase, glutathione peroxidase and catalase, and neuroapoptosis in vivo and in vitro.

Whitaker et al.45 investigated the miRNA expression changes in female and male piglets’ hippocampi after isoflurane anaesthesia. Although they did not find a relationship between miRNA levels and isoflurane-induced neurotoxicity, they reported that there was no significant difference in miRNA expression between sexes. In addition to other isoflurane-related neurotoxicity studies, Fan et al.46 studied diabetic rats and found that miR-140-5p expression increased and caused neuroapoptosis through its target SNX12. In a study investigating the anaesthetic mechanism of isoflurane, the authors showed that among miRNAs, hsa-miR-17-5p had the highest expression level after isoflurane anaesthesia in the cortex. When they investigated sensory perception-related genes, they found that hsa-miR-16-5p, hsa-miR-424-5p, and hsa-miR-497-5p were the miRNAs with the highest expression changes after isoflurane exposure. The authors concluded that the mechanism of isoflurane anaesthesia could be related to the expression changes in miRNAs and related genes.47

Ketamine

Ketamine is commonly used in pediatric surgery. In long-term exposure, ketamine causes hippocampal apoptosis and damages learning memory functions.48 Many studies have been conducted to understand the role of ketamine administration in neurotoxicity and miRNA interactions.

Ketamine in vitro Studies

The functions of miR-107, miR-375, and miR-206 were investigated in vitro. Ketamine upregulates the aforementioned miRNAs, resulting in apoptosis, neurite degeneration, and neural death. Subsequently, the targeted miRNAs were downregulated via a lentiviral system, and the downregulation of miRNAs protected against ketamine-induced neural cell death and neural toxicity. In addition, BDNF was found to be a target gene for miRNAs.48-51 In addition, ketamine downregulated the expression of miR-221-3p and attenuated IFN-α-stimulated NF-κB activation in human astrocytes (HA1800 cells).52

Ketamine in vivo Studies

In miRNA microarray studies, the expression profiles of the hippocampus and prefrontal cortex, have attracted interest under in vivo conditions. miR-331-5p, miR-496-5p, miR-206, miR-98-5p, miR-148a-3p, miR-128-3p, miR-448-3p, miR-764-5p, miR-1264-3p, miR-1298-5p, and miR-1912-3p were differentially expressed in the miRNA microarray profiles.53, 54

Memory performance and cognitive function tests were performed for in vivo cognitive examination to understand the role of miRNAs in ketamine-induced neurotoxicity in the hippocampus. miRNAs overexpressed under ketamine exposure, which cause neuronal injury, were downregulated, or silenced to reverse neuronal injury. Inhibition of miR-124 enhances memory impairment in mice. In adolescent rats, the downregulation of miR-214 results in learning memory impairment. Downregulation of miR-34c improves memory in mice. Downregulation of miR-137 causes significant memory impairment, whereas overexpression of miR-137 rescues memory loss. miR-34a inhibition leads to enhanced memory impairment.49, 55-58

The role of miRNA in Antidepresant Effect of Ketamine

Ketamine has a remarkable and persistent antidepressant effect . A study showed that miRNA-206 might be a therapeutic target for the antidepressant effects of ketamine.59 Another study found that ketamine resulted in increased expression of miR-98-5p, and downregulation of miR-98-5p resulted in a decrease in the antidepressant effect of ketamine in a mouse model.60 One study investigated the alteration of miR-29b-3p in rat brains. A significant increase in miR-29b-3p expression was observed in the prefrontal cortex of normal rats after ketamine exposure. Recombinant adeno-associated virus-mediated overexpression of miR-29b-3p resulted in recognizable relief of depressive behaviors in rats and lower expression of metabotropic glutamate receptor 4 (GRM4).61 After administration of an antidepressant dose of ketamine, the miRNAs 448-3p, 764-5p, 1264-3p, 1298-5p, and 1912-3p clusters were upregulated.62

The role of miRNA in Ketamine Addiction

The hippocampus is a critical area in the brain involved in addiction. Li et al.63 showed hippocampal miR-331-5p was significantly decreased in the ketamine group, and upregulated in the ketamine+rhynchophylline group. In conclusion, miR-331-5p was found to be an important regulator of Nurr1, and rhynchophylline is involved in ketamine addiction, via the miR-331-5p/Nurr1/BDNF pathway or the inhibition of CREB phosphorylation.

The role of BDNF Signaling in miRNA Alteration in the Brain by Ketamine

BDNF is a common target gene of miRNAs. Mir-107 inversely regulates BDNF. siRNA-mediated BDNF inhibition, reversed the protective effect of miR-107 downregulation on ketamine injury.48 Similarly, ketamine upregulated hsa-miR-375 and downregulated BDNF. BDNF is inversely regulated by ketamine-induced neural cell death and toxicity in hESC-derived neurons.51 miR-206 overexpression improved ketamine-induced upregulation of BDNF. This indicates that miRNA-206 may be a therapeutic target for the antidepressant effects of ketamine.59 miR-206 is the target for KCNQ1OT1 and downregulates its expression level, indirectly increasing the expression level of BDNF, and thus has a protective role in neural injury.50

Propofol

Propofol (2,6-diisopropyl phenol) is a common sedative/anaesthetic.64 Propofol acts rapidly and has antiemetic properties.65 Aside from its sedative use, propofol is used in the management of refractory status epilepticus.66 Both advantageous and harmful effects of propofol via miRNAs have been reported in this context.

Propofol in vivo Studies

An important miRNA that has caught the attention of researchers is miR-34a. Inhibition of miR-34a ameliorated propofol-induced impaired learning and memory function, as well as apoptosis, in Sprague-Dawley rats.67 In another study, it was demonstrated that propofol increased neuronal injury in the hippocampus of rats via miR-34a expression, and dexmedetomidine mitigated injury by targeting SIRT1 and PI3K/Akt signaling.68 Propofol injections in seven-day-old Sprague-Dawley rats resulted in miR-132 expression downregulation and a reduction in dendritic spine density in the hippocampus, and thus, impairment of learning and memory.69 Wang et al.70 verified that propofol caused elevated neurocyte damage by diminishing circRNA001372 levels while augmenting IL-1B, IL-6, IL-17, and IL-18. Subsequently, miR-148b-3p, an antagonist of circRNA001372, reversed the possible protective effects of circRNA001372. Propofol was shown to decrease inflammation through the miR-145-3p/NFATc2/NF-κB pathway.71

Propofol in vitro Studies

Several in vitro studies have demonstrated a highly intertwined relationship between propofol and Rno-miR-665. After propofol treatment, Rno-miR-665 was upregulated in primary astrocyte cultures, and it represses BCL2L1; therefore, neuroprotection was decreased.72-75 Li et al.,67 proved that propofol enhanced miR-34a levels with subsequent neurotoxicity, and inhibition of miR-34a rescued negative effects through the MAPK/ERK pathway. A study stated a protective role of propofol. It inhibited pro-inflammatory factors such as NO, TNF-α, and IL-6 via mitigating miR-155 expression, which targets SOCS1.76 In addition, propofol inhibited proinflammatory microglia activation via miR-221 and miR-222 downregulating IRF2 in BV2 microglia.77 miRNA profile screening of primary microglia suggested that propofol inhibited LPS-induced pro-inflammatory responses through the miR-106/Pi3k/Akt pathway.78 Propofol mitigated cerebral ischemia/reperfusion injury by downregulating MALAT1 and upregulating miR-182-5p.79 Moreover, propofol alleviates cerebral ischemia/reperfusion injury via the SNHG14/miR-30b-5p axis.80 Additionally, some screening studies show miRNA expression profiles, all miRNAs altered by propofol.20, 21, 81 In two separate studies, miR-455-3p decreased neurotoxicity induced by propofol via HOTAIR/NLRP182 and EphA4.83 Propofol also blocked apoptosis in hippocampal neurons via the miR-15a-5p/NR2B/ERK1/2 axis.84

Conclusion and Future Perspectives

Every year, many adults and infants are exposed to anaesthetics because of their medical needs. Herein, we compile comprehensive data on the role of miRNA in anaesthetics’ effects (Figure 2). Animal studies demonstrated that anaesthesia induces neuroapoptosis and cognitive dysfunction during brain growth. According to studies on databases, sevoflurane, isoflurane, ketamine, and propofol are the most studied anaesthetic agents in relation to miRNA and neurotoxicity. In the light of the in vivo and in vitro studies, there is no doubt that substantial improvement has been made in understanding the role of miRNAs in anaesthesia neurotoxicity, but we are still in the early days, and so there is a lot of work to be done to fully resolve the neurobiology of miRNAs. Although we know that many miRNAs are related to neurotoxicity in the brain after anaesthesia exposure, the molecular mechanisms and degree of specific miRNAs involved in the onset and progression of these neurotoxicities are unknown.

A remarkable proportion of studies have stated that anaesthetics mostly affect the hippocampal region, where neurodegeneration and neuroapoptosis occur. Cognitive dysfunction and learning memory problems have also been observed.

Rodent studies have shown that sevoflurane anaesthesia can induce neurotoxicity and learning and memory deficits. The results of preclinical studies cannot be applied to clinical practice directly. The duration of anaesthesia, repeated exposure, the dose of the anaesthetic, the use of multiple agents, and the age of the animal are factors influencing cognitive dysfunction in preclinical studies. The results of clinical studies are contradictory.

The altered miRNA expression in the brain after anaesthesia could be an underlying mechanism in the sevoflurane-induced cognitive dysfunction. The studies that investigated the effect of sevoflurane on cognitive dysfunction through miRNAs have varying results. Investigating the miRNA expression in response to sevoflurane anaesthesia has several challenges. First, different methods were used in different studies. Second, some studies investigated the whole miRNA expression in the hippocampus, while others investigated specific miRNA alterations. Third, the duration and concentration of sevoflurane anaesthesia were different in each study. Although additional consistency is needed for more meaningful results, these preclinical studies showed that sevoflurane-induced cognitive dysfunction is related to alterations in miRNA expression. Additionally, the use of miRNA-specific knockout animals in such studies should be considered. miRNAs are becoming biomarkers for the diagnosis and prognosis of other conditions; thus, they can be the targets of therapy for AIN and cognitive dysfunction.

Both in vitro and in vivo studies have investigated the effects of ketamine on neurotoxicity and miRNA interactions. In vivo studies have demonstrated that ketamine causes neurodegeneration in hippocampal neurons, which is followed by an increase in apoptosis and cognitive dysfunction.

One of the critical aspects of miRNAs is the diversity of technologies used for miRNA detection. In contrast to common methods like real-time fluorescent quantitative polymerase chain reaction (PCR) and digital PCR technology, in situ hybridization and new microarray methods have been successfully established and have started to be widely used. More recent advances for miRNA detection include electrochemical detection based on enzymatic signal amplification, rolling ring amplification, and nanoparticle technology.85

Due to scientific and technological achievements, some miRNAs can be used as candidate biomarkers, allowing earlier detection and possibly a higher rate of success for a neuroprotection strategy. Therefore, future work should focus on developing a non-invasive and effective method to deliver miRNA “drugs” into the injured brain to protect it from neurotoxicity.

A comprehensive functional characterization of miRNAs in the context of the interaction among themselves (miRNA/miRNA) with other non-coding RNA species, especially IncRNAs, has attracted attention. This is because carnici carried out an extensive analysis of the transcriptional landscape in mouse brains, and his colleagues demonstrated that a high number of lncRNAs are involved in the regulation of several target genes in the brain.86 Therefore, miRNA-lncRNA interactions and their effect on the target gene are important for miRNA research, making it necessary to elucidate the cause/consequence relationship of their regulation in anaesthesia-related neuroprotection and neurotoxicity.

In conclusion, while the literature supports that miRNAs play crucial roles in neuroprotection and neurotoxicity during anaesthesia administration, the exact role of miRNAs in anaesthesia neurotoxicity needs to be elucidated. Moreover, whether a single miRNA or a combination of miRNAs should be considered the “ideal” therapeutic candidate for neuroprotection and treatment against neurotoxicity requires further studies and more in-depth analysis.

Author Contributions: Concept - E.Ö., Ş.G.; Design - Ş.G.; Data Collection and/or Processing - E.Ö., B.K., B.İ.A., H.A., Ş.G.; Analysis and/or/Interpretation - Ş.G.; Literature Review - E.Ö., B.K., B.İ.A., H.A., Ş.G.; Writing - E.Ö., B.K., B.İ.A.
Declaration of Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

1
O’Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation.Front Endocrinol (Lausanne).2018;9:402.
2
Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14.Cell. 1993;75(5):843-854.
3
Prodromidou K, Matsas R. Species-specific miRNAs in human brain development and disease.Front Cell Neurosci. 2019;13:559.
4
Cai Y, Liu J, Zhang L, et al. Grey matter volume abnormalities in patients with bipolar I depressive disorder and unipolar depressive disorder: a voxel-based morphometry study.Neurosci Bull.2015;31(1):4-12.
5
Lv X, Yan J, Jiang J, Zhou X, Lu Y, Jiang H. MicroRNA-27a-3p suppression of peroxisome proliferator-activated receptor-γ contributes to cognitive impairments resulting from sevoflurane treatment.J Neurochem. 2017;143(3):306-319.
6
Wang L, Zheng M, Wu S, Niu Z. MicroRNA-188-3p is involved in sevoflurane anesthesia-induced neuroapoptosis by targeting MDM2.Mol Med Rep. 2018;17(3):4229-4236.
7
Shao CZ, Xia KP. Sevoflurane anesthesia represses neurogenesis of hippocampus neural stem cells via regulating microRNA-183-mediated NR4A2 in newborn rats.J Cell Physiol. 2019;234(4):3864-3873.
8
Yang L, Shen Q, Xia Y, Lei X, Peng J. Sevofluraneinduced neurotoxicity is driven by OXR1 posttranscriptional downregulation involving hsamiR302e.Mol Med Rep. 2018;18(5):4657-4665.
9
Zhao X, Sun Y, Ding Y, Zhang J, Li K. miR-34a inhibitor may effectively protect against sevoflurane-induced hippocampal apoptosis through the Wnt/β-catenin pathway by targeting Wnt1.Yonsei Med J.2018;59(10):1205-1213.
10
Zhao X, Jin Y, Li H, Jia Y, Wang Y. Sevoflurane impairs learning and memory of the developing brain through post-transcriptional inhibition of CCNA2 via microRNA-19-3p. Aging (Albany NY). 2018;10(12):3794-3805.
11
Zhang L, Yan J, Liu Q, Xie Z, Jiang H. LncRNA Rik-203 contributes to anesthesia neurotoxicity via microRNA-101a-3p and GSK-3β-mediated neural differentiation.Sci Rep.2019;9(1):6822.
12
Su R, Sun P, Zhang D, Xiao W, Feng C, Zhong L. Neuroprotective effect of miR-410-3p against sevoflurane anesthesia-induced cognitive dysfunction in rats through PI3K/Akt signaling pathway via targeting C-X-C motif chemokine receptor 5.Genes Genomics. 2019;41(10):1223-1231.
13
Liu T, Wang B, Li G, et al. Disruption of microRNA-214 during general anaesthesia prevents brain injury and maintains mitochondrial fusion by promoting Mfn2 interaction with Pkm2.J Cell Mol Med.2020;24(23):13589-13599.
14
Jiang Y, Wang Y, Sun Y, Jiang H. Long non-coding RNA Peg13 attenuates the sevoflurane toxicity against neural stem cells by sponging microRNA-128-3p to preserve Sox13 expression.PLoS One. 2020;15(12):e0243644.
15
Zhao Y, Ai Y. Overexpression of lncRNA Gm15621 alleviates apoptosis and inflammation response resulting from sevoflurane treatment through inhibiting miR-133a/Sox4.J Cell Physiol. 2020;235(2):957-965.
16
Lin D, Liu J, Hu Z, Cottrell JE, Kass IS. Neonatal anesthesia exposure impacts brain microRNAs and their associated neurodevelopmental processes.Sci Rep. 2018;8(1):10656.
17
Ye J, Zhang Z, Wang Y, et al. Altered hippocampal microRNA expression profiles in neonatal rats caused by sevoflurane anesthesia: microRNA profiling and bioinformatics target analysis.Exp Ther Med.2016;12(3):1299-1310.
18
Fujimoto S, Ishikawa M, Nagano M, Sakamoto A. Influence of neonatal sevoflurane exposure on nerve development-related microRNAs and behavior of rats.Biomed Res. 2015;36(6):347-355.
19
Zhou X, Xian D, Xia J, et al. MicroRNA-34c is regulated by p53 and is involved in sevoflurane-induced apoptosis in the developing rat brain potentially via the mitochondrial pathway.Mol Med Rep. 2017;15(4):2204-2212.
20
Goto G, Hori Y, Ishikawa M, Tanaka S, Sakamoto A. Changes in the gene expression levels of microRNAs in the rat hippocampus by sevoflurane and propofol anesthesia.Mol Med Rep. 2014;9(5):1715-1722.
21
Lu Y, Jian MY, Ouyang YB, Han RQ. Changes in rat brain MicroRNA expression profiles following sevoflurane and propofol anesthesia.Chin Med J (Engl).2015;128(11):1510-1515.
22
Xu C, Niu JJ, Zhou JF, Wei YS. MicroRNA-96 is responsible for sevoflurane-induced cognitive dysfunction in neonatal rats via inhibiting IGF1R. Brain Res Bull. 2019;144:140-148.
23
Yu Y, Zhang P, Yan J, et al. Sevoflurane induces cognitive impairments via the MiR-27b/LIMK1-signaling pathway in developing rats.Inhal Toxicol.2016;28(14):731-738.
24
Shan L, Ma D, Zhang C, Xiong W, Zhang Y. miRNAs may regulate GABAergic transmission associated genes in aged rats with anesthetics-induced recognition and working memory dysfunction.Brain Res.2017;1670:191-200.
25
Yi X, Cai Y, Zhang N, Wang Q, Li W. Sevoflurane inhibits embryonic stem cell self-renewal and subsequent neural differentiation by modulating the let-7a-Lin28 signaling pathway.Cell Tissue Res. 2016;365(2):319-330.
26
Sun H, Hu H, Xu X, Tao T, Liang Z. Key miRNAs associated with memory and learning disorder upon exposure to sevoflurane determined by RNA sequencing.Mol Med Rep. 2020;22(2):1567-1575.
27
Zhang L, Xue Z, Yan J, Jiang H. LncRNA Rik-203 contributes to sevoflurane induced neurotoxicity?Front Med (Lausanne).2020;7:353.
28
Shi H, Sun BL, Zhang J, et al. miR-15b suppression of Bcl-2 contributes to cerebral ischemic injury and is reversed by sevoflurane preconditioning.CNS Neurol Disord Drug Targets.2013;12(3):381-391.
29
Zhong H, Chen H, Gu C. Sevoflurane post-treatment upregulated miR-203 expression to attenuate cerebral ischemia-reperfusion-induced neuroinflammation by targeting MyD88.Inflammation.2020;43(2):651-663.
30
Ding L, Ning J, Wang Q, Lu B, Ke H. Retracted: sevoflurane improves nerve regeneration and repair of neurological deficit in brain damage rats via microRNA-490-5p/CDK1 axis.Life Sci.2021;271:119111.
31
Yi W, Li D, Guo Y, Zhang Y, Huang B, Li X. Sevoflurane inhibits the migration and invasion of glioma cells by upregulating microRNA-637.Int J Mol Med.2016;38(6):1857-1863.
32
Gao C, Shen J, Meng ZX, He XF. Sevoflurane inhibits glioma cells proliferation and metastasis through miRNA-124-3p/ROCK1 axis.Pathol Oncol Res.2020;26(2):947-954.
33
Zhang L, Wang J, Fu Z, et al. Sevoflurane suppresses migration and invasion of glioma cells by regulating miR-146b-5p and MMP16. Artif Cells Nanomed Biotechnol. 2019;47(1):3306-3314.
34
Xu W, Xue R, Xia R, et al. Sevoflurane impedes the progression of glioma through modulating the circular RNA has_circ_0012129/miR-761/TGIF2 axis.Eur Rev Med Pharmacol Sci.2020;24(10):5534-5548.
35
Zhao H, Xing F, Yuan J, Li Z, Zhang W. Sevoflurane inhibits migration and invasion of glioma cells via regulating miR-34a-5p/MMP-2 axis.Life Sci. 2020;256:117897.
36
Bao X, Peng Y, Shen J, Yang L. Sevoflurane inhibits progression of glioma via regulating the HMMR antisense RNA 1/microRNA-7/cyclin dependent kinase 4 axis.Bioengineered. 2021;12(1):7893-7906.
37
Zhang L, Zhang Y, Hu R, et al. Isoflurane inhibits embryonic stem cell self-renewal and neural differentiation through miR-9/E-cadherin signaling.Stem Cells Dev.2015;24(16):1912-1922.
38
Wu Q, Dai Q, Jiang L, et al. Downregulation of microRNA-448 improves isoflurane-induced learning and memory impairment in rats.Mol Med Rep. 2017;16(2):1578-1583.
39
Yang W, Guo Q, Li J, et al. microRNA-124 attenuates isoflurane-induced neurological deficits in neonatal rats via binding to EGR1. J Cell Physiol. 2019;234(12):23017-23032.
40
Xie C, Wang H, Zhang Y, Wei Y. Neuroprotective effects of miR-142-5p downregulation against isoflurane-induced neurological impairment.Diagn Pathol.2020;15(1):70.
41
Wu Q, Shang Y, Shen T, Liu F, Xu Y, Wang H. Neuroprotection of miR-214 against isoflurane-induced neurotoxicity involves the PTEN/PI3K/Akt pathway in human neuroblastoma cell line SH-SY5Y.Arch Biochem Biophys. 2019;678:108181.
42
Luo T, Yin S, Shi R, et al. miRNA expression profile and involvement of Let-7d-APP in aged rats with isoflurane-induced learning and memory impairment.PLoS One. 2015;10(3):e0119336.
43
Li H, Du M, Xu W, Wang Z. MiR-191 downregulation protects against isoflurane-induced neurotoxicity through targetingBDNF. Toxicol Mech Methods. 2021;31(5):367-373.
44
Li N, Yue L, Wang J, Wan Z, Bu W. MicroRNA-24 alleviates isoflurane-induced neurotoxicity in rat hippocampus via attenuation of oxidative stress.Biochem Cell Biol. 2020;98(2):208-218.
45
Whitaker EE, Wiemann BZ, Xia JC, et al. Distinct, sex-dependent miRNA signatures in piglet hippocampus induced by a clinically relevant isoflurane exposure: a pilot study.J Anesth.2019;33(6):670-679.
46
Fan D, Yang S, Han Y, Zhang R, Yang L. Isoflurane-induced expression of miR-140-5p aggravates neurotoxicity in diabetic rats by targeting SNX12.J Toxicol Sci. 2020;45(2):69-76.
47
Chen Y, Zhou ZF, Wang Y. Prediction and analysis of weighted genes in isoflurane induced general anesthesia based on network analysis.Int J Neurosci. 2020;130(6):610-620.
48
Jiang JD, Zheng XC, Huang FY, Gao F, You MZ, Zheng T. MicroRNA-107 regulates anesthesia-induced neural injury in embryonic stem cell derived neurons.IUBMB Life. 2019;71(1):20-27.
49
Xu H, Zhang J, Zhou W, Feng Y, Teng S, Song X. The role of miR-124 in modulating hippocampal neurotoxicity induced by ketamine anesthesia.Int J Neurosci. 2015;125(3):213-220.
50
Yao Y, Wang X, Gao J. LncRNA KCNQ1OT1 sponges miR-206 to ameliorate neural injury induced by anesthesia via up-regulating BDNF.Drug Des Devel Ther.2020;14:4789-4800.
51
Zhao X, Shu F, Wang X, et al. Inhibition of microRNA-375 ameliorated ketamine-induced neurotoxicity in human embryonic stem cell derived neurons.Eur J Pharmacol. 2019;844:56-64.
52
Feng J, Wang M, Li M, et al. Serum miR-221-3p as a new potential biomarker for depressed mood in perioperative patients.Brain Res.2019;1720:146296.
53
Arqué JM, Sans-Coma V, Durán AC, Cardo M. Origen anómalo de la arteria coronaria izquierda en el tronco pulmonar y su relación con otras anomalías coronarias primarias: estudio experimental [Anomalous origin of the left coronary artery at the pulmonary trunk and its relationship with other primary coronary anomalies: experimental study].Rev Esp Cardiol. 1989;42(6):399-409.
54
Choi Y, Kim B, Ham S, et al. Subanesthetic ketamine rapidly alters medial prefrontal miRNAs involved in ubiquitin-mediated proteolysis.PLoS One.2021;16(8):e0256390.
55
Cao SE, Tian J, Chen S, Zhang X, Zhang Y. Role of miR-34c in ketamine-induced neurotoxicity in neonatal mice hippocampus.Cell Biol Int. 2015;39(2):164-168.
56
Huang C, Zhang X, Zheng J, Chen C, Chen Y, Yi J. Upregulation of miR-137 protects anesthesia-induced hippocampal neurodegeneration.Int J Clin Exp Pathol. 2014;7(8):5000-5007.
57
Jiang XL, Du BX, Chen J, Liu L, Shao WB, Song J. MicroRNA-34a negatively regulates anesthesia-induced hippocampal apoptosis and memory impairment through FGFR1. Int J Clin Exp Pathol. 2014;7(10):6760-6767.
58
Wang J, Zhou M, Wang X, et al. Impact of ketamine on learning and memory function, neuronal apoptosis and its potential association with miR-214 and PTEN in adolescent rats. PLoS One. 2014;9(6):e99855.
59
Yang X, Yang Q, Wang X, et al. MicroRNA expression profile and functional analysis reveal that miR-206 is a critical novel gene for the expression of BDNF induced by ketamine.Neuromolecular Med. 2014;16(3):594-605.
60
Huang C, Wang Y, Wu Z, et al. miR-98-5p plays a critical role in depression and antidepressant effect of ketamine.Transl Psychiatry. 2021;11(1):454.
61
Wan YQ, Feng JG, Li M, et al. Correction: prefrontal cortex miR-29b-3p plays a key role in the antidepressant-like effect of ketamine in rats.Exp Mol Med.2022;54(11):2104-2105.
62
Grieco SF, Velmeshev D, Magistri M, et al. Ketamine up-regulates a cluster of intronic miRNAs within the serotonin receptor 2C gene by inhibiting glycogen synthase kinase-3.World J Biol Psychiatry. 2017;18(6):445-456.
63
Li C, Tu G, Luo C, et al. Effects of rhynchophylline on the hippocampal miRNA expression profile in ketamine-addicted rats.Prog Neuropsychopharmacol Biol Psychiatry. 2018;86:379-389.
64
Stogiannou D, Protopapas A, Protopapas A, Tziomalos K. Is propofol the optimal sedative in gastrointestinal endoscopy?Acta Gastroenterol Belg. 2018;81(4):520-524.
65
Lundström S, Twycross R, Mihalyo M, Wilcock A. Propofol.J Pain Symptom Manage. 2010;40(3):466-470.
66
Brown LA, Levin GM. Role of propofol in refractory status epilepticus.Ann Pharmacother. 1998;32(10):1053-1059.
67
Li GF, Li ZB, Zhuang SJ, Li GC. Inhibition of microRNA-34a protects against propofol anesthesia-induced neurotoxicity and cognitive dysfunction via the MAPK/ERK signaling pathway.Neurosci Lett.2018;675:152-159.
68
Xing N, Xing F, Li Y, et al. Dexmedetomidine improves propofol-induced neuronal injury in rat hippocampus with the involvement of miR-34a and the PI3K/Akt signaling pathway.Life Sci.2020;247:117359.
69
Zhang S, Liang Z, Sun W, Pei L. Repeated propofol anesthesia induced downregulation of hippocampal miR-132 and learning and memory impairment of rats.Brain Res.2017;1670:156-164.
70
Wang M, Suo L, Yang S, Zhang W. CircRNA 001372 reduces inflammation in propofol-induced neuroinflammation and neural apoptosis through PIK3CA/Akt/NF-κB by miRNA-148b-3p.J Invest Surg. 2021;34(11):1167-1177.
71
Cui C, Zhang D, Sun K, et al. Propofol maintains Th17/Treg cell balance and reduces inflammation in rats with traumatic brain injury via the miR1453p/NFATc2/NFκB axis.Int J Mol Med. 2021;48(1):135.
72
Sun WC, Pei L. rno-miR-665 targets BCL2L1 (Bcl-xl) and increases vulnerability to propofol in developing astrocytes.J Neurochem. 2016;138(2):233-242.
73
Sun WC, Liang ZD, Pei L. Propofol-induced rno-miR-665 targets BCL2L1 and influences apoptosis in rodent developing hippocampal astrocytes.Neurotoxicology. 2015;51:87-95.
74
Sun W, Pei L. microRNA expression profiling of propofol-treated developing rat hippocampal astrocytes.DNA Cell Biol. 2015;34(8):511-523.
75
Sun WC, Pei L. Dexmedetomidine attenuates propofol-induced apoptosis of neonatal hippocampal astrocytes by inhibiting the Bcl2l1 signalling pathway.Eur J Neurosci. 2021;54(11):7775-7789.
76
Zheng X, Huang H, Liu J, Li M, Liu M, Luo T. Propofol attenuates inflammatory response in LPS-activated microglia by regulating the miR-155/SOCS1 pathway.Inflammation. 2018;41(1):11-19.
77
Xiao X, Hou Y, Yu W, Qi S. Propofol ameliorates microglia activation by targeting microRNA-221/222-IRF2 axis. J Immunol Res. 2021;2021:3101146.
78
Liu J, Ai P, Sun Y, et al. Propofol inhibits microglial activationvia miR-106b/Pi3k/Akt axis. Front Cell Neurosci. 2021;15:768364.
79
Hu Y, Ye C, Cheng S, Chen J. Propofol downregulates lncRNA MALAT1 to alleviate cerebral ischemia-reperfusion injury.Inflammation. 2021;44(6):2580-2591.
80
Sun B, Ou H, Ren F, Guan Y, Huan Y, Cai H. Propofol protects against cerebral ischemia/reperfusion injury by down-regulating long noncoding RNA SNHG14.ACS Chem Neurosci. 2021;12(16):3002-3014.
81
Jiang C, Logan S, Yan Y, et al. Signaling network between the dysregulated expression of microRNAs and mRNAs in propofol-induced developmental neurotoxicity in mice.Sci Rep. 2018;8(1):14172.
82
Gong H, Wan X, Zhang Y, Liang S. Downregulation of HOTAIR reduces neuronal pyroptosis by targeting miR-455-3p/NLRP1 axis in propofol-treated neurons in vitro.Neurochem Res. 2021;46(5):1141-1150.
83
Zhu X, Li H, Tian M, Zhou S, He Y, Zhou M. miR-455-3p alleviates propofol-induced neurotoxicity by reducing EphA4 expression in developing neurons.Biomarkers. 2020;25(8):685-692.
84
Liu X, Geng J, Guo H, Zhao H, Ai Y. Propofol inhibited apoptosis of hippocampal neurons in status epilepticus through miR-15a-5p/NR2B/ERK1/2 pathway.Cell Cycle. 2020;19(9):1000-1011.
85
Jet T, Gines G, Rondelez Y, Taly V. Advances in multiplexed techniques for the detection and quantification of microRNAs.Chem Soc Rev.2021;50(6):4141-4161.
86
Forero DA, van der Ven K, Callaerts P, Del-Favero J. miRNA genes and the brain: implications for psychiatric disorders. Hum Mutat.2010;31(11):1195-1204.