Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 7, 2023; 29(33): 4927-4941
Published online Sep 7, 2023. doi: 10.3748/wjg.v29.i33.4927
Non-coding RNAs: The potential biomarker or therapeutic target in hepatic ischemia-reperfusion injury
Jia-Li Shao, Li-Juan Wang, Ji Xiao, Jin-Feng Yang, Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan Province, China
ORCID number: Jia-Li Shao (0000-0002-8291-1849); Li-Juan Wang (0000-0002-1467-7907); Ji Xiao (0000-0002-8726-9285); Jin-Feng Yang (0000-0002-0774-0496).
Author contributions: Shao JL collected the data, prepared the tables and wrote the paper; Wang LJ collected the data; Xiao J revised the paper; Yang JF conceptualized the idea, obtained the funding, and revised the paper; all authors have read and approved the final manuscript.
Supported by the National Natural Science Foundation of China, No. 82070648; and the Science and Technology Innovation Program of Hunan Province, No. 2021SK4014.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Jin-Feng Yang, MD, PhD, Chief Physician, Director, Doctor, Professor, Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283 Tongzipo, Yuelu District, Changsha 410013, Hunan Province, China. yangjinfeng@hnca.org.cn
Received: May 6, 2023
Peer-review started: May 6, 2023
First decision: July 9, 2023
Revised: July 22, 2023
Accepted: August 18, 2023
Article in press: August 18, 2023
Published online: September 7, 2023

Abstract

Hepatic ischemia-reperfusion injury (HIRI) is the major complication of liver surgery and liver transplantation, that may increase the postoperative morbidity, mortality, tumor progression, and metastasis. The underlying mechanisms have been extensively investigated in recent years. Among these, oxidative stress, inflammatory responses, immunoreactions, and cell death are the most studied. Non-coding RNAs (ncRNAs) are defined as the RNAs that do not encode proteins, but can regulate gene expressions. In recent years, ncRNAs have emerged as research hotspots for various diseases. During the progression of HIRI, ncRNAs are differentially expressed, while these dysregulations of ncRNAs, in turn, have been verified to be related to the above pathological processes involved in HIRI. ncRNAs mainly contain microRNAs, long ncRNAs, and circular RNAs, some of which have been reported as biomarkers for early diagnosis or assessment of liver damage severity, and as therapeutic targets to attenuate HIRI. Here, we briefly summarize the common pathophysiology of HIRI, describe the current knowledge of ncRNAs involved in HIRI in animal and human studies, and discuss the potential of ncRNA-targeted therapeutic strategies. Given the scarcity of clinical trials, there is still a long way to go from pre-clinical to clinical application, and further studies are needed to uncover their potential as therapeutic targets.

Key Words: Hepatic ischemia-reperfusion injury, Non-coding RNAs, MicroRNAs, Long non-coding RNAs, Circular RNAs

Core Tip: This review focuses on the recent progress in understanding non-coding RNAs (ncRNAs) in hepatic ischemia-reperfusion injury (HIRI). HIRI can alter ncRNAs expressions, which in turn modulates the pathophysiological processes that contribute to the development of HIRI. Differentially expressed ncRNAs from different sources (the liver tissues, serums and cells) are involved in oxidative stress, inflammatory responses, cell death and so on. ncRNAs are regarded as biomarkers for the diagnosis and assessment of liver damage severity, or as therapeutic targets for HIRI; however, their clinical transformation will still take a long time.



INTRODUCTION

Hepatic ischemia-reperfusion injury (HIRI) is a common clinical issue that occurs during major liver resection, liver transplantation, and liver trauma[1-3]. HIRI usually causes liver injury, early transplantation failure, liver failure and even multiple-organ failure. Although existing studies have shown that several signaling pathways, such as oxidative stress, inflammatory response, and cell death, participate in the pathological process of HIRI[4-6], current treatments and pharmacological approaches cannot completely address this problem. Therefore, much more new molecules need to be explored for the diagnosis and treatment of HIRI.

Non-coding RNAs (ncRNAs) are a cluster of functional RNAs that cannot encode protein[7,8]. In recent years, ncRNAs have become a hot area of research and have been reported to play a significant role in various diseases, including cancers, nervous system diseases, and ischemia/reperfusion injuries[9-11]. ncRNAs mainly consist of microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), which have been shown to modulate genes expression, and participate in many critical biological processes at different levels (e.g. immune responses, oxidative stress reactions, apoptosis, autophagy, and energy metabolism)[12-14]. The role of ncRNAs in HIRI is explored in a few studies, and has attracted the attention of many researchers.

ncRNAs are regarded as potential biomarkers or therapeutic targets for assessing or attenuating HIRI development. As the present studies are mainly in the preclinical stage, and clinical investigations are lacking, there are still large research prospects for the diagnosis, prevention, and treatment of HIRI. This review briefly illustrates the well-studied molecular mechanisms of HIRI and summarizes the relevant ncRNAs and their roles in the pathological process of HIRI to provide a reference for further research.

HIRI AND THE UNDERLYING MECHANISMS

HIRI is usually classified into two types, warm IRI in situ and cold IRI in vitro, and the cells involved are different. Warm IRI is characterized by hepatocellular injury, which is mainly caused by Kupffer cells (KCs) induced oxidative stress and neutrophils recruitment[15]. Cold IRI is associated with sinusoidal endothelial cells (SECs) damage[11]. Although the initial cells are different, they do share common subsequent reactions: activation of cell death programs such as apoptosis, necrosis, pyroptosis, and autophagy, thus contributing to the development of inflammation[1,16-18].

The mechanisms involved in HIRI pathogenesis are multifactorial and complex. Numerous studies have demonstrated several molecular mechanisms that contribute to the development of HIRI[19], such as anaerobic metabolism, immune response[20], microcirculatory dysfunction[21], and gene transcription[6,21].

Oxidative stress

The most widely studied mechanism is oxidative stress, which is defined as an imbalance between the oxidant and antioxidant systems, resulting in tissue damages[22,23]. For instance, CeO2, NO, and chlorogenic acid have a liver-protective effect by reducing oxidative stress during HIRI[20,24,25]. In contrast, Han et al[26] found that hyperglycemia aggravates HIRI by inducing reactive oxygen species (ROS) mediated oxidative stress. ROS are the most critical reactive molecules involved in HIRI. Enormous amounts of ROS are usually generated by mitochondria and KCs during reperfusion, which can result in apoptosis, autophagy, inflammation, protein and DNA damage, and worsened hepatocyte injury[5,22]. Prussian blue scavenger is a potential therapeutic agent for treating HIRI with ROS-scavenging and anti-inflammatory properties[27]. OX40 expression in neutrophils can increase ROS production, which in turn activates neutrophils and, aggravates HIRI[28]. Dugbartey et al[29] designed a reversible redox probe REPOM to monitor ROS during HIRI for early diagnosis and timely intervention. Furthermore, an increasing number of studies on gene therapy for oxidative stress have attracted the attention of researchers in recent years. Therefore, strategies that are aimed at inhibiting oxidative stress or scavenging ROS may alleviate HIRI, and novel antioxidant regulatory molecules are required.

Inflammatory response and immune response

The inflammatory response is another major mechanism underlying HIRI. During HIRI development, KCs and SECs are initially activated[30,31], and generate a range of inflammatory mediators including cytokines, such as PAF, TNFα, and interleukins (e.g., IL-1, IL-6, IL-12, and IL-23), which could lead to the inflammatory response involved in HIRI development[32-34]. In addition, several cytokines (e.g., PAF, leukotriene B4, IL-8, and IL-17) induce neutrophil accumulation, which plays a role in the process of HIRI[35]. Furthermore, activated neutrophils can form neutrophil extracellular traps (NETs) via TLR-dependent pathways, that initiate inflammatory responses during HIRI[36]. This local inflammatory state can cause a systemic inflammatory response, leading to systemic inflammatory response syndrome and even multiple-organ failures.

Both innate and adaptive immune responses play important roles in HIRI[37,38]. KCs can increase the production of damage-associated molecular patterns (DAMPs), such as ATP, histones, high mobility group box 1 (HMGB1), S100, and heat shock proteins, which are released into the circulation and induce cytokine/chemokine storms to attract neutrophils and other immune cells[39]. In contrast, DAMPs can bind to Toll-like receptors (TLRs) and drive immune responses. Targeting the DAMP pathways alleviates HIRI. For example, HMGB1/NLRP3 inflammasome inhibition attenuated HIRI[40]. In addition, the complement system serves as an important contributor to the process of HIRI[41-43]. Therefore, targeting inflammation-oriented therapies may alleviate HIRI.

Cell death

Cell death is a stable pathological indicator of I/R injury, including apoptosis, necrosis, autophagy, pyroptosis, and ferroptosis. Among them, apoptosis, necrosis, and autophagy are the most common types of cell death during HIRI and may share the same stimuli and signaling pathways[44]. For example, hydrogen sulfide (H2S) effectively alleviates HIRI by attenuating hepatocyte apoptosis via inhibition of the endoplasmic reticulum (ER) stress response[22]. Eucommia ulmoides polysaccharide administration notably reduced the area of liver necrosis in a rat HIRI model[45]. Cafestol preconditioning can inhibit apoptosis and autophagy in hepatocytes, thus attenuating HIRI, by suppressing the extracelluar signal-regulated/eroxisome proliferator-activated receptor gamma (ERK/PPARγ) pathway[46]. Moreover, some studies suggest that mitochondrial autophagy is a key pathological mechanism underlying age-dependent hypersensitivity to HIRI[16]. In our previous studies, we found that octreotide pretreatment mitigated HIRI and attenuated kidney injury caused by HIRI by inhibiting hepatocellular apoptosis and enhancing autophagy[47-49]. These three processes usually coexist and participate in the development of HIRI.

The role of pyroptosis in HIRI remains unclear. Pyroptosis is a form of regulated cell death driven by perturbations in extracellular or intracellular homeostasis related to innate immunity. It is usually associated with IL-1β and IL-18 secretion and hence mediates robust pro-inflammatory effects[50]. Adipose-derived mesenchymal stem cells reduce pyroptosis in HIRI by inhibiting the NF-κB pathway and activating the Wnt/β-catenin pathway[51,52]. In the hepatic tissue of HIRI mice, lncRNA KCNQ1OT1 increased, which modulated miR-142a-3p/HMGB1, thereby promoting pyroptosis[15]. Under HIRI condition, IKZF1 increased, but sirtuin 1 (SIRT1) decreased in both human and mouse livers. Further investigation indicated that IKZF1 augmented pyroptosis through negatively regulating SIRT1 expression. Hence, pyroptosis plays an important role in the development of HIRI, and targeting pyroptosis is a promising approach for attenuating HIRI. However, the role of pyroptosis in HIRI requires further investigations.

In addition to the common types of cell death, ferroptosis has recently been reported to participate in HIRI. In 2012, Dixon et al[53] first proposed the definition of ferroptosis, which is characterized by the iron-dependent accumulation of lethal lipid ROS. Then ferroptosis was proved contributed to HIRI pathogenesis. Deferoxamine, (an iron chelator) attenuated HIRI and lipid peroxidation, whereas iron overload was a novel risk factor for HIRI[53]. Bioinformatics analysis was conducted to predict the genes related to ferroptosis in HIRI. Five genes (ATF3, IL6, IL1B, CDKN1A, and PTGS2) and several miRNAs (miR-128-3p and miR-24-3p) were identified[54]. Subsequently, Maresin conjugate in tissue regeneration 1 ameliorated ferroptosis-induced HIRI by promoting nuclear factor erythroid-derived 2-like 2 expression[55]. However, μ opioid receptor alleviated ferroptosis in HIRI via the HIF-1α/ axis[56]. These results indicate that ferroptosis plays a critical role in HIRI.

Other mechanisms

Many other regulatory mechanisms, in addition to those mentioned in previous sections, are involved in the progress of HIRI. For instance, mitochondrial dysfunction (such as mitophagy and impairment of mitochondrial permeability) plays a vital role in HIRI[16]. Acidic microenvironment has been reported to be a key factor affecting HIRI through the regulation of PPAR-γ[26]. Besides, lipid metabolites[57,58], calcium overload[56], adenosine triphosphate depletion[55], gap junctions[6], dysfunctional microcirculation[5,53], and endoplasmic reticulum stress[26,29] can affect the development of HIRI.

miRNAs

miRNAs are a class of endogenous single-chain, small ncRNAs with (21-25 nucleotides in length). The first known miRNAs (lin-4 and let-7) were identified in C. elegans in 1993[59]. Subsequently, more miRNAs have been found in plants, viruses, and animals. miRNAs produced from hairpin-like precursor transcripts are regulators of posttranscriptional and transcriptional gene expression[7,60], and are involved in various biological processes, such as development, apoptosis, metabolism, and proliferation[61]. miRNAs play important roles in human diseases, such as cancer, cardiovascular diseases, and genetic diseases[62,63]. In HIRI, numerous miRNAs have been thus far identified as biomarkers or therapeutic targets in the past decades (shown in Table 1).

Table 1 MicroRNAs and their function in hepatic ischemia-reperfusion injury.
miRNAs
Change
Targets
Effect on HIRI
Models
Ref.
miR-34aUpNrf-2, SIRT1Overexpression aggravates, downregulation alleviatesRats, mice and RAW264.7[66,68-71]
miR-34a-5pUpNF-κB /JNK/ P38Overexpression alleviatesHuman, rats, 7702 cells, AML12 cells[72]
miR-122Up Nrf2, PHD1Overexpression aggravates, overexpression alleviatesRats, mice and human[69-71,78]
miR-370Up TbRII, NF-κB, Blc2/BAXDownregulation alleviatesMice and AML12 [79,80,83]
miR-17Up Stat3Overexpression aggravatesMice and AML12 [96]
miRNA-155Up CD80, CD86, MHC-IIKnock out alleviatesMice, Kupffer cells, AML12 cells, primary hepatocytes[97]
MiR-223UpBiomarkerMice and human[98]
miR-494Up HIF-1α/HO-1, PI3K/Akt pathwayOverexpression alleviatesMice, L02 cells, rats [65,84,85]
miR-27a-5pUpBach1Overexpression alleviates, downregulation aggravatesMice and AML12 cells[99]
miRNA-191Up ZONAB/Cyclin D1Overexpression aggravates, knock out alleviatesMice and LO2 cells[100]
miR-450b-5pUpCRYAB/NF-κB, Akt1/mTORDownregulation alleviatesMice and RAW 264.7 cells[101]
miR-218-5pUp GAB2/PI3K/AKTdownregulation alleviates, overexpression aggravatesmice[102]
miR-494Down PTEN/PI3K/AKTOverexpression alleviatesRats and AML12 cells[86]
miR-146bDown TRAF6, NF-κBDownregulation aggravatesRats[103]
miR-330-3pDown PGAM5Overexpression alleviates, downregulation aggravatesMice and AML12 cells[104]
miR-1246DownIL-6-gp130-STAT3Downregulation aggravatesMice and hUCB-MSCs[105]
miR-142DownHMGB1/TLR4/NF-κBOverexpression alleviates, downregulation aggravatesMice and NCTC 1469 cells [93]
miR-96DownFOXO4Overexpression alleviates, downregulation aggravatesMice and primary hepatocytes[92]
miR-494 Down PTEN/PI3K/AKTOverexpression alleviatesL02 cells; rats, AML12 cells[84,86]
miR-30bDownAtg12-Atg5Downregulation aggravatesMice and AML12[106]
miR-146aDown IRAK1, TRAF6Overexpression alleviatesMice and RAW264.7[107]
miR-124Down Rab38, AKT pathwayOverexpression alleviatesRats and L02 cells[108]
miR-20b-5pDown SIRT1Overexpression alleviatesRats [67]
miRNA-182-5pNo detectTLR4Overexpression alleviatesMice and RAW264.7[109]
miR-192-5pUp/DownZeb2Downregulation alleviatesMice and Hepa1-6 cells, human[90]
125b-5p Down Myd88, c-Fos and A20No functional testsMice[87]
miR-501-3pDownMyd88, c-Fos and A20No functional testsMice[87]
miR-133a-5pDown MAPK6Overexpression alleviates, downregulation aggravatesRats and QSG-7701[95]
miR-214Down TRAF1/ASK1/JNKOverexpression alleviatesMice and AML12 cells[110]
miR-449b-5pDown HMGB1, NF-κBOverexpression alleviatesRats and L02 cells[111]
miR-142-3pDownMARCKSOverexpression alleviates, downregulation aggravatesMice and AML12, HepG2 cells[112]
miR-24-3pDown STINGOverexpression alleviatesMice[113]
miR-9-5pDown CXCR4Overexpression alleviatesLiver sinusoidal endothelial cells [114]
miR-141-3pDownKeap1/Nrf2Overexpression alleviates, downregulation aggravatesHuman, mice and LO2 cells [89]
miR-194Down PHLDA1Overexpression alleviatesMice and RAW 264.7 cells[115]
miR-9-3pDown FNDC3VBOverexpression alleviates, downregulation aggravatesRats[94]
miR-29a-3pNo changeIreb2Overexpression alleviates, downregulation aggravatesRats and BMMSC[116]
miR-124-3pDown TRAF3/CREB, Steap3Overexpression alleviates, downregulation aggravatesMice and Normal BNL Rats, CL.2 hepatocytes, BMMSC [117,118]
miR-140-5pDown CAPN1Overexpression alleviates, downregulation aggravatesMice and AML12 cells[119]

In 2009, Xu et al[64] reported for the first time that under the criteria of fold change > 2 and P value < 0.5, 78 miRNAs (40 down-regulated/38 up-regulated) were identified in the liver upon I/R injury. Among them, four miRNAs (miR-23a, miR-326, miR-346_MM1, and miR-370) were significantly downregulated by ischemic preconditioning (IPC) compared to non-preconditioned controls, implying a potential role of these miRNAs in the protective mechanism of IPC against hepatic injury[64]. Subsequently, more miRNAs were identified and their specific regulatory mechanisms were reported. The rise or fall of miRNAs determines their modulatory effects on HIRI development, and the different target genes of miRNAs allow them to be involved in the different mechanisms underlying HIRI, such as inflammation, apoptosis, and oxidative stress. The relevant information is presented in Tables 1 and 2.

Table 2 MicroRNAs and the involved mechanisms in hepatic ischemia-reperfusion injury.
Mechanisms
miRNAs
Ref.
ApoptosismiR-1, miR-17, miR-133, miR-205, miR-34a, miR-124, miR-146a, miR-494, miR-192-5p, miR-133a-5p, miRNA-155, miR-146b, miR-27a-5p, miR-214, miRNA-191, miR-370, miR-449b-5p, miRNA-142-3p, miRNA-24-3p, miR-9-5p, miR-96, miRNA-141-3p, miR-9-3p, miR-218-5p, miR-124-3p, miR-34a-5p, miR-142, miR-140-5p[68,72,83,84,86,89-95,97,99,100,102,107,108,110-113,114,117,119]
Inflammatory responsesmiRNA-182-5p, miR-370, miR-34a, miR-146a, 125b-5p and miR-501-3p, miRNA-155, miR-146b, miR-148a, miR-1246, MiRNA-142-3p, miRNA-24-3p, miR-9-5p, miR-194, miR-9-3p, miR-124-3p, miR-218-5p, miR-450b-5p, miR-142, miR-140-5p[68,79,80,87,93,94,97,101-103,105,107,109,112-115,117,119,120]
Oxidative stressmiR-34a, miR-122, miR-494, miR-9-3p, miR-218-5p, miR-142[69,70,71,86,93,94,102]
AutophagymiR-17, miR-30b, miR-330-3p[96,104,106]
FerroptosismiR-29a-3p, miR-124-3p[116,118]
miR-34

During HIRI, miR-34 is upregulated and mediates several important signaling pathways involved in various biological processes, such as inflammatory responses and apoptosis. Carbon monoxide (CO) inhalation or p-coumaric acid reduces miR-34a expression in the liver tissue, thus increasing SIRT1, which in turn mitigates HIRI by alleviating inflammatory responses, hepatocellular apoptosis and autophagy[64,65]. Similarly, H2S and crocin exerted hepatoprotective effects in a rat model of HIRI by regulating the miR-34a/Nrf-2 pathway[66,67]. H2S significantly modulated miR-34a expression in hepatocytes, whereas crocin regulated its expression in serum. Both zinc sulfate and gallic acid decreased the miR-34a serum level of miR-34a as an anti-miR to ameliorate HIRI[68-70]. Zheng et al[71] revealed that high miR-34a-5p expression may reduce liver injury during hepatectomy in adults. Further, agomir-miR-34a-5p could attenuate HIRI in rats, and in vitro experiments indicated that miR-34a-5p/HNF4α might be the underlying mechanism.

miR-122

miR-122 is a hepato-specific miRNA that accounts for nearly 70% of the total miRNAs pool in the liver tissue and exerts modulatory effects in liver diseases[71-74]. Recent studies have implicated its role in HIRI. van Caster et al[75] reported that miR-122 is a potential biomarker of warm HIRI of rats. In clinical research, serum miR-122 Levels were significantly elevated in patients with acute liver failure (ALF) than in healthy individuals. Further investigation revealed higher miR-122 levels in the serum and liver tissue of ALF survivors compared with those in the non-recovered patients[75]. Furthermore, miR-122 downregulation is involved in the hepatoprotective effects of crocin, gallic acid, and zinc sulfate[67-69]. However, John et al[76] found that hepatocyte-specific miR122 deletion in mice exacerbated liver injury during HIRI and that nanoparticle-mediated miR122 overexpression attenuated liver injury. These controversial modulatory effects require further investigations.

miR-370

In 2009, miR-370 was detected in a mouse model of HIRI by using miRNA microarrays. Subsequent analysis revealed that miR-370 was upregulated and positively correlated with the severity of ischemic injury[63,77]. Li et al[78] reported that miR-370 was significantly upregulated in a mouse model of HIRI, and that miR-370 inhibition efficiently attenuated liver damage via TbRII. Further investigation revealed that downregulation of miR-370 reduced the levels of proinflammatory cytokines, but had no effect on the apoptosis and proliferation of hepatocytes during HIRI. Moreover, the NF-κB gene was suggested as a potential target of miR-370[78]. Mesenchymal stem cells (MSCs) display immunomodulatory functions and have been proven to alleviate HIRI[79-81]. Zare et al[82] revealed that bone marrow-derived mesenchymal stem cells (BM-MSCs) downregulated miR-370 and inhibited inflammatory responses and apoptosis, thus attenuating liver damage during HIRI.

miR-494

In addition, miR-494 warrants further investigation. In a mouse model of HIRI, miRNA microarrays have indicated a upregulation of miR-494[63]. Besides, miR-494 expression was reported to significantly increase during hypoxia for 4 h in L02 cells, and its overexpression protected against hypoxia-induced apoptosis[82]. Another study suggested in a rat model of HIRI, miR-494 was elevated, and reducing its expression with propofol had a protective effect during HIRI[83,84]. However, we observed an opposite trend in miR-494 expression of in other studies. Su et al[85] showed that miR-494 was downregulated in a rat model of HIRI and H2O2-induced apoptosis in hepatic AML12 cells. Furthermore, overexpression of miR-494 attenuated HIRI by modulating the PTEN/ PI3K/AKT signaling pathway[85]. We speculated that the opposite trend of miR-494 during HIRI might be caused by the duration of ischemia and reperfusion, cell lines, and different models. Based on previous studies, the researchers chosen different durations of ischemia (75 min, 30 min, and 60 min), different durations of reperfusion (2 h, reperfusion moment, 6 h), and different cell models (hypoxia-induced apoptosis in L02 cells and H2O2-induced apoptosis in AML-12 cells) for their studies. Further studies are required to confirm this hypothesis.

In addition to these above miRNAs, many other miRNAs have been identified, including miR-17, miR-155, miR-223, miR-494, miR-27-5p, miR-191, miR-450-5p, and miR-218-5p. Functional tests revealed they might be involved in HIRI development by modulating various biological processes such as cell death, inflammatory immune responses, and oxidative stress by targeting different downstream genes. Tables 1 and 2 present detailed information on the upregulated miRNAs.

The downregulated miRNAs

Several miRNAs were downregulated and exhibited modulatory effects on HIRI. miR-146b, miR-124, miR-20b-5p, miR-133a-5p, miR-449b-5p, miR-9-3p, and miR-124-3p were significantly downregulated in a rat HIRI model[85]. In terms of a mouse HIRI model, the downregulated miRNAs, including miR-330-3p, miR-1246, miR-142, miR-30b, miR-146a, miR-96, 125b-5p , miR-501-3p, miR-214, miR-142-3p, miR-24-3p, miR-141-3p, miR-194, miR-124-3p, miR-140-5p, miR-153-3p, miR-210-5p, miR-107-3p, miR-103-3p, miR-205-5p, miR-296-5p, miR-183-3p, and miR-698-5p were detected[86,87]. Most of these miRNAs were confirmed and their functions were verified using cell models. Detailed information on the downregulated miRNAs is presented in Tables 1 and 2.

Only two miRNAs (miR-141-3p and miR-192-5p) have been identified in humans. Li et al[88] collected serum samples from 27 Liver transplantation patients at different time points (pre-operatively, 4 h after reperfusion, and on postoperative days 1, 2, and 3) and measured the expression of miR-141-3p, ALT, and AST. They found that 4 h after perfusion, miR-141-3p was lower than pre-operation and then gradually increased over time, which manifested a negative correlation with ALT/AST levels[87]. Roy and his colleagues detected miR-192-5p expression in the liver tissues and sera of patients with acute liver injury. The results shown that miR-192-5p decreased in liver samples, but elevated in serum levels from patients with acute hepatic injury; further investigation revealed that miR-192-5p concentrations in serum were positively correlated with AST, ALT, and miR-122 Levels, which might represent a hepatocyte-specific serum biomarker[88].

Of note, several miRNAs have been indicated to participate in the hepatoprotective effect of several pharmacological agents during HIRI, including inhaled anesthetics and propofol. For instance, sevoflurane preconditioning ameliorates liver injury by the inhibitory effects of several miRNAs (e.g., miR-133 and miR-205) on the Akt–GSK–cyclin D1 pathway[89]. Others have reported that sevoflurane preconditioning promotes the expression of miR-96 and inhibits FOXO4, thus alleviating HIRI[90]. In contrast, sevoflurane postconditioning exhibited the same hepatoprotective effect by counteracting miR-142 downregulation induced by I/R[91]. Moreover, isoflurane upregulates miR-9-3p to protect rats from HIRI by inhibiting FNDC3VB[92]. In addition, propofol exhibited protective effects against HIRI in rats by increasing the expression of miR-133a-5p and decreasing that of MAPK6[93].

Collectively, HIRI alters the expression of miRNAs. In turn, differentially expressed miRNAs play vital roles in HIRI development. Currently, a lot of miRNAs have been identified and their specific modulatory roles have been verified. However, it is important to note that human trails are lacking. Future research should focus on clinical transformation, which remains a significant challenge.

LNCRNAS

lncRNAs are a subset of noncoding RNAs with over 200 nucleotides (200 nt) and are localized to both the nucleus and cytoplasm[8,119]. Typically, lncRNAs are transcribed by Pol II, and have 5′-end 7-methyl guanosine (m7G) caps and 3′-end polyadenylated [poly(A)] tails. lncRNAs were considered transcription junks without protein-coding capacity until their modulatory effects on gene expression were established. lncRNAs modulate chromatin structure and function, transcription, post-transcription, and sponge miRNAs by interacting with DNA, RNA and proteins, and in turn participate in diverse cellular processes such as cell differentiation, cell apoptosis, stem cell pluripotency, and stress response[12,120,121]. Recent studies show that lncRNAs can affect various diseases (e.g., nervous disorders, immune systems, and cancers)[122,123]. The role of lncRNAs in the pathophysiology of I/R has been explored in multiple oxygen-dependent organs, such as the heart, brain, and kidney[124-129]. In terms of HIRI, it is still at an early stage. Here, we summarize the lncRNAs that have been reported, and detailed information is shown in Table 3.

Table 3 Long non-coding RNAs and their function in hepatic ischemia-reperfusion injury.
lncRNAs
Change
Targets
Effect on HIRI
Models
Ref.
TUG1Up Brg1, miR-194/SIRT1Alleviate Mice, AML12 cells, WRL-68 cells[133,144]
SNHG1Up miR-186-5p/YY1Alleviate AML12 cells[145]
NEAT1Up-AggravateHL7702 cells[134]
Gm4419UpmiR-455/SOX6 axisAggravateRats and BRL-3A[138]
AK139328UpAkt signaling pathway and NF-κBAggravateMice and primary mouse hepatocyte[130]
AK054386 Up miR-199AggravateMice and BNL-CL2 cells[140]
HOTAIR UpmiR-20b-5p/ATG7AggravateMice and primary mouse hepatocyte[141]
MALAT1UpHMGB1-TLR4AggravateHL7702 cells[135]
Hnf4αosUp Hnf4α/miR-23aAggravateHuman, mice, primary mouse hepatocyte and L02 cells[136]
KCNQ1OT1UpmiR-142a-3p/HMGB1AggravateMice and primary mouse hepatocyte[15]
CCAT1Downcaspase-3, cyclin D1AggravateHL7702 cells [139]
MEG3DownmiR-34a/Nrf2AggravateMice and HL7702 cells[137]

In 2013, Chen et al[130] first revealed that in mouse livers after I/R treatment, 71 upregulated lncRNAs (fold change ≥ 1.5, and P value < 0.5) and 27 downregulated lncRNAs (fold change ≤ 0.7, and P value < 0.5) were identified. Four up-regulated lncRNAs (AK139328, AK087277, AK054386 and AK028007) and six down-regulated lncRNAs (AK143693, NR-028310, NR-015462, NR-036616, ENSMUST00000151138 and AK143294) were validated using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Further investigation suggested that silencing of AK139328 could ameliorate HIRI by activating the Akt/ NF-κB signaling pathway[128]. The same research team detected the lncRNA profile in mouse plasma after HIRI and found that under the same criteria, 64 up-regulated lncRNAs and 244 down-regulated lncRNAs were detected. The authors then conducted a comparative analysis of dysregulated lncRNA profiles between plasma and liver and revealed that all dysregulated lncRNAs in plasma remained either unchanged or absent in mice livers after HIRI, as did dysregulated lncRNAs in the livers, which strongly indicated that the source of these dysregulated lncRNAs may not be restricted to liver cells during HIRI[129]. Another study suggested that blood cells secrete large amounts of lncRNAs during heart failure[130,131].

With the development of novel technologies, an increasing number of differentially expressed lncRNAs have been identified, and their roles have been explored in HIRI models. Current studies indicate that lncRNAs participate in various biological processes involved in HIRI development. A few studies revealed that some lncRNAs, including TUG1[132], NEAT1[133], MALAT1[134] and Hnf4αos[135], could modulate the processes of apoptosis and inflammatory response. Other lncRNAs, such as MEG3[136], Gm4419[137], CCAT1[138] and AK054386[139] were verified to regulate apoptosis, whereas AK139328 was only found to regulate the inflammatory response[130]. In addition to regulating these common biological processes, several lncRNAs participate in uncommon processes. HOTAIR expression in the liver was upregulated in a mouse model of HIRI, and further investigation indicated that HOTAIR regulates hepatocyte autophagy by targeting miR-20b-5p/ATG7[140]. Similarly, HIRI downregulates the expression of KCNQ1OT1 in mice livers, which promotes proliferation and inhibits pyroptosis by serving as a competing endogenous RNA to modulate the miR-142a-3p/HMGB1 axis[5]. Moreover, AK054386 upregulation may lead to sustained ERS and increased cell apoptosis and death in mice HIRI models[139]. Detailed information is shown in Table 4.

Table 4 Long non-coding RNAs and the involved mechanisms in hepatic ischemia-reperfusion injury.
Mechanisms
lncRNAs
Ref.
ApoptosisMEG3, TUG1, NEAT1, Gm4419, CCAT1, AK054386, MALAT1, Hnf4αos[133-140]
Inflammatory responsesTUG1, NEAT1, AK139328, MALAT1, Hnf4αos[130,133-136]
Oxidative stressTUG1[133,144]
AutophagyHOTAIR[141]
Pyroptosis KCNQ1OT1[142]
Endoplasmic reticulum stressAK054386[140]

Li et al[141] successfully constructed HIRI-related lncRNA-miRNA-mRNA networks such as LOC1201029870-miRNA-331-3p/miRNA-128-5p-CDH3/UPK3B and LOC120094223-miRNA-92b-5p-KRT7, which may play an important role in HIRI. However, these specific modulatory mechanisms stay uncovered, and further investigation is needed.

In summary, these studies support the use of lncRNAs as highly attractive targets for diagnosing and treating HIRI. An increasing number of lncRNAs are known to be involved in HIRI. Overexpression and knockdown of lncRNAs attenuated or aggravated the extent of HIRI in vivo and in vitro, respectively, indicating the significance of lncRNAs in HIRI. A comprehensive understanding of lncRNAs in HIRI not only provides a new dimension to the molecular mechanisms, but also paves the way for future treatments. Indeed, future studies need more functional experiments in vivo and in vitro to reveal the specific roles of lncRNAs and to further explore its secretory and transport mechanisms in HIRI.

CIRCULAR RNAS

Circular RNA is a special subclass of ncRNAs characterized by a covalent bond joining the 3′ and 5′ ends generated by the back-splicing of exons[142-144]. Once produced, most circRNAs are exported from the nucleus to the cytoplasm[145]. Compared to their cognate linear RNAs, circRNAs are more stable and are not easily degraded by RNase L, RNase P, or RNase MRP. In addition to regulating transcription, splicing, and chromatin interactions, circRNAs act as decoys for miRNAs and proteins, interact with proteins, and function as templates for translation, and as sources of pseudogene generation[13,144]. circRNAs are involved in various biological processes, including immunity, neuronal function, cell proliferation, and transformation[146-149]. Existing evidence indicates the potential of circRNAs in treating diverse diseases.

Advances in RNA sequencing technologies have allowed the detection and exploration of many circRNAs in various pathological conditions. Currently, circRNAs have been implicated in the process of I/R injury, particularly myocardial I/R injury. For instance, circ_SMG6 deteriorates myocardial I/R injury by activating the miR-138-5p/EGR1/TLR4/TRIF signaling, whereas circ_CNEACR and circ_ACR alleviate myocardial I/R injury by suppressing autophagy[150-152]. Moreover, circ-FoxO3 attenuates blood-brain barrier damage by inhibiting mTORC1 activity during cerebral I/R[153]. circ-AKT3 aggravates renal I/R injury by regulating the miR-144-5p /Wnt/β-catenin pathway[154]. However, only few studies have investigated the role of circRNAs in HIRI.

To date, only two studies have reported the potential role of circRNAs in HIRI. Zhang et al[87] conducted a circRNA microarray in mice for the first time in 2019 and found that, compared to the sham group, 706 circRNAs were differentially expressed in the I/R group, including 213 upregulated and 493 downregulated circRNAs. Compared to the ischemic postconditioning (IPO) group, 641 up-regulated and 252 down-regulated circRNAs were identified in the I/R group (fold change ≥ 2.0 and P value < 0.05). Among these, circRNA_005186 was upregulated in the I/R group, whereas IPO treatment downregulated its expression. A subsequent in vitro experiment showed that circRNA_005186 functioned as a miRNA sponge for miR-124-3p, thereby enhancing Epha2 expression. In other words, the circRNA_005186-miR-124-3p-Epha2 pathway might be a possible protective mechanism of IPO against HIRI[87]. The other five circRNAs (circRNA_011137, circRNA_013703, circRNA_29140, circRNA_36837, and circRNA_43819) were validated by RT-qPCR, and may be the focus of future research. In addition to IPO, IPC can attenuate HIRI. Tian et al[155] reported the circRNA profiles of mice with ischemic livers, with and without IPC. The data revealed that there were 77 circRNAs and 686 mRNAs in the IRI group, 50 circRNAs and 95 mRNAs in the IPC group (fold change ≥ 1.5, and P value < 0.05), respectively, when compared with those in the sham group. Next, they compared the circRNA alterations in the three groups and selected circRNA_017753 for further study. The prediction of the circRNA–miRNA–mRNA pathway implied a potential role of the circRNA_017753–miR-218-5p/miR-7002-3p/miR-7008-3p–Jade1 pathway in the mechanisms of IPC protection in HIRI. However, further investigations are required[155].

Although circRNA research in the field of HIRI is still in its infancy, the present data indicate great prospects for research. With the increasing attention of the scientific community, a thorough understanding of circRNA mechanisms will provide new insights and therapeutic targets for treating HIRI.

ADDITIONAL NCRNAS

Other ncRNAs such as PIWI-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs) and tRNA-derived small RNAs (tsRNAs), have attracted widespread attention in recent years. For instance, some studies have revealed that piRNAs were abnormally expressed and might play a regulatory role in liver cancer, non-alcoholic fatty liver disease and liver injury[156-158]. Moreover, snoRNAs and tsRNAs are involved in several liver diseases as biomarkers and therapeutic targets[159-162]. However, only few studies have investigated their roles in HIRI. As our knowledge of these ncRNAs expands, their potential role in HIRI will be confirmed.

CONCLUSION

Although ncRNAs have not been fully identified in the development of HIRI, current data indicate that ncRNAs are important regulators of various biological processes involved in the pathology of HIRI, and can serve as biomarkers for the diagnosis and assessment of therapeutic targets for treating HIRI. However, we noticed that most of the data were collected from animal studies, and the majority of ncRNAs described in this review were isolated from total liver tissue. So, establishing large clinical trials with diverse sample sources is necessary. Meanwhile, exploring the role of lncRNAs and circRNAs in HIRI is still in the start-up phase, and more attention needs to be paid in the future. In summary, our expanding knowledge of the capabilities of ncRNAs in HIRI will pave the way for novel diagnostic indicators and therapeutic inventions for HIRI.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Joshi MK, India; Katada K, Japan S-Editor: Yan JP L-Editor: A P-Editor: Yu HG

References
1.  Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation--from bench to bedside. Nat Rev Gastroenterol Hepatol. 2013;10:79-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 468]  [Cited by in F6Publishing: 577]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
2.  Teoh NC, Farrell GC. Hepatic ischemia reperfusion injury: pathogenic mechanisms and basis for hepatoprotection. J Gastroenterol Hepatol. 2003;18:891-902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 299]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
3.  Liu H, Man K. New Insights in Mechanisms and Therapeutics for Short- and Long-Term Impacts of Hepatic Ischemia Reperfusion Injury Post Liver Transplantation. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
4.  Huang M, Cai H, Han B, Xia Y, Kong X, Gu J. Natural Killer Cells in Hepatic Ischemia-Reperfusion Injury. Front Immunol. 2022;13:870038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
5.  Han JY, Li Q, Ma ZZ, Fan JY. Effects and mechanisms of compound Chinese medicine and major ingredients on microcirculatory dysfunction and organ injury induced by ischemia/reperfusion. Pharmacol Ther. 2017;177:146-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 105]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
6.  Hernández-Guerra M, Hadjihambi A, Jalan R. Gap junctions in liver disease: Implications for pathogenesis and therapy. J Hepatol. 2019;70:759-772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
7.  Farh KK, Grimson A, Jan C, Lewis BP, Johnston WK, Lim LP, Burge CB, Bartel DP. The widespread impact of mammalian MicroRNAs on mRNA repression and evolution. Science. 2005;310:1817-1821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1125]  [Cited by in F6Publishing: 1141]  [Article Influence: 60.1]  [Reference Citation Analysis (0)]
8.  Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermüller J, Hofacker IL, Bell I, Cheung E, Drenkow J, Dumais E, Patel S, Helt G, Ganesh M, Ghosh S, Piccolboni A, Sementchenko V, Tammana H, Gingeras TR. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316:1484-1488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1800]  [Cited by in F6Publishing: 1838]  [Article Influence: 108.1]  [Reference Citation Analysis (0)]
9.  Ghafouri-Fard S, Shoorei H, Taheri M. Non-coding RNAs participate in the ischemia-reperfusion injury. Biomed Pharmacother. 2020;129:110419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
10.  Slack FJ, Chinnaiyan AM. The Role of Non-coding RNAs in Oncology. Cell. 2019;179:1033-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 872]  [Article Influence: 218.0]  [Reference Citation Analysis (0)]
11.  Salta E, De Strooper B. Noncoding RNAs in neurodegeneration. Nat Rev Neurosci. 2017;18:627-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
12.  Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22:96-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 929]  [Cited by in F6Publishing: 2144]  [Article Influence: 536.0]  [Reference Citation Analysis (0)]
13.  Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 2019;20:675-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1947]  [Cited by in F6Publishing: 2608]  [Article Influence: 521.6]  [Reference Citation Analysis (0)]
14.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 578]  [Cited by in F6Publishing: 628]  [Article Influence: 104.7]  [Reference Citation Analysis (0)]
15.  Liang C, Peng Y, Sun H, Wang L, Jiang L, Zou S. Silencing lncRNA KCNQ1OT1 reduced hepatic ischemia reperfusion injury-induced pyroptosis by regulating miR-142a-3p/HMGB1 axis. Mol Cell Biochem. 2023;478:1293-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
16.  Kim JS, Chapman WC, Lin Y. Mitochondrial Autophagy in Ischemic Aged Livers. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Zou Z, Shang R, Zhou L, Du D, Yang Y, Xie Y, Li Z, Zhao M, Jiang F, Zhang L, Zhou P. The Novel MyD88 Inhibitor TJ-M2010-5 Protects Against Hepatic Ischemia-reperfusion Injury by Suppressing Pyroptosis in Mice. Transplantation. 2023;107:392-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
18.  Yu C, Chen P, Miao L, Di G. The Role of the NLRP3 Inflammasome and Programmed Cell Death in Acute Liver Injury. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
19.  Pretzsch E, Nieß H, Khaled NB, Bösch F, Guba M, Werner J, Angele M, Chaudry IH. Molecular Mechanisms of Ischaemia-Reperfusion Injury and Regeneration in the Liver-Shock and Surgery-Associated Changes. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
20.  Li K, Feng Z, Wang L, Ma X, Liu K, Geng X, Peng C. Chlorogenic Acid Alleviates Hepatic Ischemia-Reperfusion Injury by Inhibiting Oxidative Stress, Inflammation, and Mitochondria-Mediated Apoptosis In Vivo and In Vitro. Inflammation. 2023;46:1061-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  Vollmar B, Menger MD. The hepatic microcirculation: mechanistic contributions and therapeutic targets in liver injury and repair. Physiol Rev. 2009;89:1269-1339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 351]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
22.  Chen L, Lin B, Yang J, Zhong L, Xiong X, Wang X. Hydrogen sulfide alleviates ischemia induced liver injury by repressing the SPHK1/S1P pathway. Ann Transl Med. 2023;11:73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
23.  Elias-Miró M, Jiménez-Castro MB, Rodés J, Peralta C. Current knowledge on oxidative stress in hepatic ischemia/reperfusion. Free Radic Res. 2013;47:555-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 133]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
24.  Galaris D, Barbouti A, Korantzopoulos P. Oxidative stress in hepatic ischemia-reperfusion injury: the role of antioxidants and iron chelating compounds. Curr Pharm Des. 2006;12:2875-2890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 81]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
25.  Gobut H, Kucuk A, Şengel N, Arslan M, Ozdemir C, Mortas T, Kasapbası E, Kurtipek O, Kavutcu M. Effects of cerium oxide (CeO(2)) on liver tissue in liver ischemia-reperfusion injury in rats undergoing desflurane anesthesia. BMC Anesthesiol. 2023;23:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
26.  Han CY, Lim SW, Koo JH, Kim W, Kim SG. PHLDA3 overexpression in hepatocytes by endoplasmic reticulum stress via IRE1-Xbp1s pathway expedites liver injury. Gut. 2016;65:1377-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
27.  Ding W, Duan Y, Qu Z, Feng J, Zhang R, Li X, Sun D, Zhang X, Lu Y. Acidic Microenvironment Aggravates the Severity of Hepatic Ischemia/Reperfusion Injury by Modulating M1-Polarization Through Regulating PPAR-γ Signal. Front Immunol. 2021;12:697362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
28.  Huang Y, Xu Q, Zhang J, Yin Y, Pan Y, Zheng Y, Cai X, Xia Q, He K. Prussian Blue Scavenger Ameliorates Hepatic Ischemia-Reperfusion Injury by Inhibiting Inflammation and Reducing Oxidative Stress. Front Immunol. 2022;13:891351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
29.  Dugbartey GJ, Juriasingani S, Zhang MY, Sener A. H(2)S donor molecules against cold ischemia-reperfusion injury in preclinical models of solid organ transplantation. Pharmacol Res. 2021;172:105842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
30.  Shi Z, Song Y, Gao X, Loor JJ, Aboragah A, Yu H, Fang Z, Zhu Y, Du X, Li X, Gao W, Liu G. Disruption of endoplasmic reticulum homeostasis exacerbates liver injury in clinically ketotic cows. J Dairy Sci. 2021;104:9130-9141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
31.  Hisama N, Yamaguchi Y, Ishiko T, Miyanari N, Ichiguchi O, Goto M, Mori K, Watanabe K, Kawamura K, Tsurufuji S, Ogawa M. Kupffer cell production of cytokine-induced neutrophil chemoattractant following ischemia/reperfusion injury in rats. Hepatology. 1996;24:1193-1198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
32.  Samarasinghe DA, Farrell GC. The central role of sinusoidal endothelial cells in hepatic hypoxia-reoxygenation injury in the rat. Hepatology. 1996;24:1230-1237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 37]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
33.  Zhou W, McCollum MO, Levine BA, Olson MS. Inflammation and platelet-activating factor production during hepatic ischemia/reperfusion. Hepatology. 1992;16:1236-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 57]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
34.  Hoffmann F, Sass G, Zillies J, Zahler S, Tiegs G, Hartkorn A, Fuchs S, Wagner J, Winter G, Coester C, Gerbes AL, Vollmar AM. A novel technique for selective NF-kappaB inhibition in Kupffer cells: contrary effects in fulminant hepatitis and ischaemia-reperfusion. Gut. 2009;58:1670-1678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
35.  Kono H, Fujii H, Ogiku M, Hosomura N, Amemiya H, Tsuchiya M, Hara M. Role of IL-17A in neutrophil recruitment and hepatic injury after warm ischemia-reperfusion mice. J Immunol. 2011;187:4818-4825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
36.  Al Mogrampi S, Boumpoureka C, Afaloniati H, Lagou M, Angelopoulou K, Anestakis D, Tampouratzi ZG, Iliadis S, Antoniadis N, Giakoustidis A, Papalois A, Papadopoulos V, Poutahidis T, Giakoustidis D. Inhibition of γδ-TcR or IL17a Reduces T-Cell and Neutrophil Infiltration after Ischemia/Reperfusion Injury in Mouse Liver. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
37.  Huang H, Tohme S, Al-Khafaji AB, Tai S, Loughran P, Chen L, Wang S, Kim J, Billiar T, Wang Y, Tsung A. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology. 2015;62:600-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 347]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
38.  Zhai Y, Busuttil RW, Kupiec-Weglinski JW. Liver ischemia and reperfusion injury: new insights into mechanisms of innate-adaptive immune-mediated tissue inflammation. Am J Transplant. 2011;11:1563-1569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 304]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
39.  van Golen RF, Reiniers MJ, Olthof PB, van Gulik TM, Heger M. Sterile inflammation in hepatic ischemia/reperfusion injury: present concepts and potential therapeutics. J Gastroenterol Hepatol. 2013;28:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 104]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
40.  Han H, Desert R, Das S, Song Z, Athavale D, Ge X, Nieto N. Danger signals in liver injury and restoration of homeostasis. J Hepatol. 2020;73:933-951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
41.  Du Y, Zhong F, Cheng H, Li T, Chen Y, Tan P, Huang M, Liang T, Liu Y, Xia X, Fu W. The Dietary Supplement γ-Oryzanol Attenuates Hepatic Ischemia Reperfusion Injury via Inhibiting Endoplasmic Reticulum Stress and HMGB1/NLRP3 Inflammasome. Oxid Med Cell Longev. 2021;2021:4628050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
42.  Arumugam TV, Woodruff TM, Stocks SZ, Proctor LM, Pollitt S, Shiels IA, Reid RC, Fairlie DP, Taylor SM. Protective effect of a human C5a receptor antagonist against hepatic ischaemia-reperfusion injury in rats. J Hepatol. 2004;40:934-941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
43.  Diepenhorst GM, de Graaf W, Niessen HW, van Vliet AK, Hack CE, van Gulik TM. Immunoglobulin M, C-reactive protein and complement activation in rat hepatic ischemia-reperfusion injury. Eur Surg Res. 2014;52:50-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
44.  Feng XB, Ke JJ, Rao Y, Zhang ZZ, Wang YL. Effect of complement C1q expression on hepatic ischemia-reperfusion injury in rats. J Huazhong Univ Sci Technolog Med Sci. 2014;34:403-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
45.  Mao B, Yuan W, Wu F, Yan Y, Wang B. Autophagy in hepatic ischemia-reperfusion injury. Cell Death Discov. 2023;9:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
46.  Gao W, Feng Z, Zhang S, Wu B, Geng X, Fan G, Duan Y, Li K, Liu K, Peng C. Anti-Inflammatory and Antioxidant Effect of Eucommia ulmoides Polysaccharide in Hepatic Ischemia-Reperfusion Injury by Regulating ROS and the TLR-4-NF-κB Pathway. Biomed Res Int. 2020;2020:1860637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
47.  Ji J, Wu L, Feng J, Mo W, Wu J, Yu Q, Li S, Zhang J, Dai W, Xu X, Mao Y, Xu S, Chen K, Li J, Guo C. Cafestol preconditioning attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting ERK/PPARγ pathway. Int Immunopharmacol. 2020;84:106529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
48.  Yang J, Sun H, Takacs P, Zhang Y, Liu J, Chang Y, Candiotti KA. The effect of octreotide on hepatic ischemia-reperfusion injury in a rabbit model. Transplant Proc. 2013;45:2433-2438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
49.  Sun H, Zou S, Candiotti KA, Peng Y, Zhang Q, Xiao W, Wen Y, Wu J, Yang J. Octreotide Attenuates Acute Kidney Injury after Hepatic Ischemia and Reperfusion by Enhancing Autophagy. Sci Rep. 2017;7:42701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
50.  Zou S, Sun H, Candiotti KA, Peng Y, Zhang Q, Xiao W, Zhao S, Wu L, Yang J. Octreotide protects against hepatic ischemia/reperfusion injury via HO-1-mediated autophagy. Acta Biochim Biophys Sin (Shanghai). 2018;50:316-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
51.  Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Muñoz-Pinedo C, Nagata S, Nuñez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3672]  [Cited by in F6Publishing: 3674]  [Article Influence: 612.3]  [Reference Citation Analysis (0)]
52.  Piao C, Sang J, Kou Z, Wang Y, Liu T, Lu X, Jiao Z, Wang H. Effects of Exosomes Derived from Adipose-Derived Mesenchymal Stem Cells on Pyroptosis and Regeneration of Injured Liver. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
53.  Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B 3rd, Stockwell BR. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060-1072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4711]  [Cited by in F6Publishing: 8459]  [Article Influence: 704.9]  [Reference Citation Analysis (1)]
54.  Hide D, Ortega-Ribera M, Garcia-Pagan JC, Peralta C, Bosch J, Gracia-Sancho J. Effects of warm ischemia and reperfusion on the liver microcirculatory phenotype of rats: underlying mechanisms and pharmacological therapy. Sci Rep. 2016;6:22107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
55.  Sun S, Xue J, Guo Y, Li J. Bioinformatics analysis of genes related to ferroptosis in hepatic ischemia-reperfusion injury. Front Genet. 2022;13:1072544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
56.  Liu Q, Wu D, Ma Y, Cao Y, Pang Y, Tang M, Pu Y, Zhang T. Intracellular reactive oxygen species trigger mitochondrial dysfunction and apoptosis in cadmium telluride quantum dots-induced liver damage. NanoImpact. 2022;25:100392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
57.  Han X, Wang R, Song X, Yu F, Lv C, Chen L. A mitochondrial-targeting near-infrared fluorescent probe for bioimaging and evaluating endogenous superoxide anion changes during ischemia/reperfusion injury. Biomaterials. 2018;156:134-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 81]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
58.  Monga SP. Lipid metabolic reprogramming in hepatic ischemia-reperfusion injury. Nat Med. 2018;24:6-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
59.  Ke B, Kupiec-Weglinski JW. Lipid Metabolites: The Alarm Signal to Trigger Liver Ischemia-reperfusion Injury. Transplantation. 2018;102:887-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
60.  Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8672]  [Cited by in F6Publishing: 8460]  [Article Influence: 272.9]  [Reference Citation Analysis (0)]
61.  Khraiwesh B, Arif MA, Seumel GI, Ossowski S, Weigel D, Reski R, Frank W. Transcriptional control of gene expression by microRNAs. Cell. 2010;140:111-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 311]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
62.  Bartel DP. Metazoan MicroRNAs. Cell. 2018;173:20-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2329]  [Cited by in F6Publishing: 2287]  [Article Influence: 381.2]  [Reference Citation Analysis (0)]
63.  Mendell JT, Olson EN. MicroRNAs in stress signaling and human disease. Cell. 2012;148:1172-1187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1233]  [Cited by in F6Publishing: 1304]  [Article Influence: 108.7]  [Reference Citation Analysis (0)]
64.  Xu CF, Yu CH, Li YM. Regulation of hepatic microRNA expression in response to ischemic preconditioning following ischemia/reperfusion injury in mice. OMICS. 2009;13:513-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
65.  Kim HJ, Joe Y, Yu JK, Chen Y, Jeong SO, Mani N, Cho GJ, Pae HO, Ryter SW, Chung HT. Carbon monoxide protects against hepatic ischemia/reperfusion injury by modulating the miR-34a/SIRT1 pathway. Biochim Biophys Acta. 2015;1852:1550-1559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
66.  Moradi M, Farbood Y, Mard SA, Dianat M, Goudarzi G, Khorsandi L, Seyedian SS. p-Coumaric acid has pure anti-inflammatory characteristics against hepatopathy caused by ischemia-reperfusion in the liver and dust exposure. Iran J Basic Med Sci. 2023;26:164-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
67.  Huang X, Gao Y, Qin J, Lu S. The role of miR-34a in the hepatoprotective effect of hydrogen sulfide on ischemia/reperfusion injury in young and old rats. PLoS One. 2014;9:e113305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
68.  Akbari G, Mard SA, Dianat M, Mansouri E. The Hepatoprotective and MicroRNAs Downregulatory Effects of Crocin Following Hepatic Ischemia-Reperfusion Injury in Rats. Oxid Med Cell Longev. 2017;2017:1702967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 41]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
69.  Mard SA, Akbari G, Dianat M, Mansouri E. The Effect of Zinc Sulfate on miR-122, miR-34a, Atioxidants, Biochemical and Histopathological Parameters Following Hepatic Ischemia/Reperfusion Injury in Rats. Biol Trace Elem Res. 2019;188:434-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
70.  Akbari G, Savari F, Mard SA, Rezaie A, Moradi M. Gallic acid protects the liver in rats against injuries induced by transient ischemia-reperfusion through regulating microRNAs expressions. Iran J Basic Med Sci. 2019;22:439-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
71.  Zheng X, Wang G, Yuan J, Li N, Yan B, Yan J, Sheng Y. hsa-miR-34a-5p Ameliorates Hepatic Ischemia/Reperfusion Injury Via Targeting HNF4α. Turk J Gastroenterol. 2022;33:596-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
72.  Chang J, Nicolas E, Marks D, Sander C, Lerro A, Buendia MA, Xu C, Mason WS, Moloshok T, Bort R, Zaret KS, Taylor JM. miR-122, a mammalian liver-specific microRNA, is processed from hcr mRNA and may downregulate the high affinity cationic amino acid transporter CAT-1. RNA Biol. 2004;1:106-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 607]  [Cited by in F6Publishing: 632]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
73.  Yin S, Fan Y, Zhang H, Zhao Z, Hao Y, Li J, Sun C, Yang J, Yang Z, Yang X, Lu J, Xi JJ. Differential TGFβ pathway targeting by miR-122 in humans and mice affects liver cancer metastasis. Nat Commun. 2016;7:11012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
74.  Rheault M, Cousineau SE, Fox DR, Abram QH, Sagan SM. Elucidating the distinct contributions of miR-122 in the HCV life cycle reveals insights into virion assembly. Nucleic Acids Res. 2023;51:2447-2463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
75.  Van Caster P, Brandenburger T, Strahl T, Metzger S, Bauer I, Pannen B, Braun S. Circulating microRNA-122, -21 and -223 as potential markers of liver injury following warm ischaemia and reperfusion in rats. Mol Med Rep. 2015;12:3146-3150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
76.  John K, Hadem J, Krech T, Wahl K, Manns MP, Dooley S, Batkai S, Thum T, Schulze-Osthoff K, Bantel H. MicroRNAs play a role in spontaneous recovery from acute liver failure. Hepatology. 2014;60:1346-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
77.  Ju C, Wang M, Tak E, Kim B, Emontzpohl C, Yang Y, Yuan X, Kutay H, Liang Y, Hall DR, Dar WA, Bynon JS, Carmeliet P, Ghoshal K, Eltzschig HK. Hypoxia-inducible factor-1α-dependent induction of miR122 enhances hepatic ischemia tolerance. J Clin Invest. 2021;131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
78.  Li L, Li G, Yu C, Shen Z, Xu C, Feng Z, Zhang X, Li Y. A role of microRNA-370 in hepatic ischaemia-reperfusion injury by targeting transforming growth factor-β receptor II. Liver Int. 2015;35:1124-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
79.  Zhu J, Zhu F, Song W, Zhang B, Zhang X, Jin X, Li H. Altered miR-370 expression in hepatic ischemia-reperfusion injury correlates with the level of nuclear kappa B (NF-κB) related factors. Gene. 2017;607:23-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
80.  Pan GZ, Yang Y, Zhang J, Liu W, Wang GY, Zhang YC, Yang Q, Zhai FX, Tai Y, Liu JR, Zhang Q, Chen GH. Bone marrow mesenchymal stem cells ameliorate hepatic ischemia/reperfusion injuries via inactivation of the MEK/ERK signaling pathway in rats. J Surg Res. 2012;178:935-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
81.  Wu R, Fan X, Wang Y, Shen M, Zheng Y, Zhao S, Yang L. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver Immunity and Therapy. Front Immunol. 2022;13:833878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
82.  Zare MA, Zare A, Azarpira N, Pakbaz S. The protective effect of bone marrow-derived mesenchymal stem cells in liver ischemia/reperfusion injury via down-regulation of miR-370. Iran J Basic Med Sci. 2019;22:683-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
83.  Sun G, Zhou Y, Li H, Guo Y, Shan J, Xia M, Li Y, Li S, Long D, Feng L. Over-expression of microRNA-494 up-regulates hypoxia-inducible factor-1 alpha expression via PI3K/Akt pathway and protects against hypoxia-induced apoptosis. J Biomed Sci. 2013;20:100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
84.  Lv J, Zou X, Yu C, Ou W, Sun C. Effects of propofol on cardiac function and miR-494 expression in rats with hepatic ischemia/reperfusion injury. J Int Med Res. 2021;49:300060521990988.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
85.  Su S, Luo D, Liu X, Liu J, Peng F, Fang C, Li B. miR-494 up-regulates the PI3K/Akt pathway via targetting PTEN and attenuates hepatic ischemia/reperfusion injury in a rat model. Biosci Rep. 2017;37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
86.  Zheng W, Men H, Li J, Xing Y, Wu B, Wang Z, Teng D, Shi Y, Jiang P, Cai J. Global MicroRNA Expression Profiling of Mouse Livers following Ischemia-Reperfusion Injury at Different Stages. PLoS One. 2016;11:e0148677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
87.  Zhang P, Ming Y, Ye Q, Niu Y. Comprehensive circRNA expression profile during ischemic postconditioning attenuating hepatic ischemia/reperfusion injury. Sci Rep. 2019;9:264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
88.  Li T, Chen Q, Dai J, Huang Z, Luo Y, Mou T, Pu J, Yang H, Wei X, Wu Z.   MicroRNA-141-3p Attenuates Oxidative Stress-induced Hepatic Ischemia Reperfusion Injury via Keap1/Nrf2 Pathway. 2021 Preprint.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Li T, Chen Q, Dai J, Huang Z, Luo Y, Mou T, Pu J, Yang H, Wei X, Wu Z. MicroRNA-141-3p attenuates oxidative stress-induced hepatic ischemia reperfusion injury via Keap1/Nrf2 pathway. Mol Biol Rep. 2022;49:7575-7585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
90.  Roy S, Benz F, Alder J, Bantel H, Janssen J, Vucur M, Gautheron J, Schneider A, Schüller F, Loosen S, Luedde M, Koch A, Tacke F, Luedde T, Trautwein C, Roderburg C. Down-regulation of miR-192-5p protects from oxidative stress-induced acute liver injury. Clin Sci (Lond). 2016;130:1197-1207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
91.  Morita T, Ishikawa M, Sakamoto A. Identical MicroRNAs Regulate Liver Protection during Anaesthetic and Ischemic Preconditioning in Rats: An animal study. PLoS One. 2015;10:e0125866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
92.  He B, Yang F, Ning Y, Li Y. Sevoflurane alleviates hepatic ischaemia/reperfusion injury by up-regulating miR-96 and down-regulating FOXO4. J Cell Mol Med. 2021;25:5899-5911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
93.  Xu L, Ge F, Hu Y, Yu Y, Guo K, Miao C. Sevoflurane Postconditioning Attenuates Hepatic Ischemia-Reperfusion Injury by Limiting HMGB1/TLR4/NF-κB Pathway via Modulating microRNA-142 in vivo and in vitro. Front Pharmacol. 2021;12:646307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
94.  Wang H, Guo L, Wang Y, Song S. Isoflurane upregulates microRNA-9-3p to protect rats from hepatic ischemia-reperfusion injury through inhibiting fibronectin type III domain containing 3B. Cell Cycle. 2021;20:1527-1539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
95.  Hao W, Zhao ZH, Meng QT, Tie ME, Lei SQ, Xia ZY. Propofol protects against hepatic ischemia/reperfusion injury via miR-133a-5p regulating the expression of MAPK6. Cell Biol Int. 2017;41:495-504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
96.  Li S, Zhang J, Wang Z, Wang T, Yu Y, He J, Zhang H, Yang T, Shen Z. MicroRNA-17 regulates autophagy to promote hepatic ischemia/reperfusion injury via suppression of signal transductions and activation of transcription-3 expression. Liver Transpl. 2016;22:1697-1709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
97.  Li Y, Ma D, Wang Z, Yang J. MicroRNA-155 Deficiency in Kupffer Cells Ameliorates Liver Ischemia-Reperfusion Injury in Mice. Transplantation. 2017;101:1600-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
98.  Schueller F, Roy S, Loosen SH, Alder J, Koppe C, Schneider AT, Wandrer F, Bantel H, Vucur M, Mi QS, Trautwein C, Luedde T, Roderburg C. miR-223 represents a biomarker in acute and chronic liver injury. Clin Sci (Lond). 2017;131:1971-1987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
99.  Xing Y, Li J, Li SP, Xi J, Ma N, Liu L, Wang JS, Cai JZ. MiR-27a-5p regulates apoptosis of liver ischemia-reperfusion injury in mice by targeting Bach1. J Cell Biochem. 2018;119:10376-10383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
100.  Pan W, Wang L, Zhang XF, Zhang H, Zhang J, Wang G, Xu P, Zhang Y, Hu P, Zhang XD, Du RL, Wang H. Hypoxia-induced microRNA-191 contributes to hepatic ischemia/reperfusion injury through the ZONAB/Cyclin D1 axis. Cell Death Differ. 2019;26:291-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
101.  Huang Z, Mou T, Luo Y, Pu X, Pu J, Wan L, Gong J, Yang H, Liu Y, Li Z, Shen A, Wu Z. Inhibition of miR-450b-5p ameliorates hepatic ischemia/reperfusion injury via targeting CRYAB. Cell Death Dis. 2020;11:455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 42]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
102.  Ji H, Li H, Zhang H, Cheng Z. Role of microRNA2185p in sevofluraneinduced protective effects in hepatic ischemia/reperfusion injury mice by regulating GAB2/PI3K/AKT pathway. Mol Med Rep. 2022;25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
103.  Zhang T, Xiu HH, Liu JX, Ma Y, Xu KQ, Huang WQ. Protective effect of aspirin-triggered resolvin D1 on hepatic ischemia/reperfusion injury in rats: The role of miR-146b. Int Immunopharmacol. 2017;51:140-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
104.  Sun XL, Zhang YL, Xi SM, Ma LJ, Li SP. MiR-330-3p suppresses phosphoglycerate mutase family member 5 -inducted mitophagy to alleviate hepatic ischemia-reperfusion injury. J Cell Biochem. 2019;120:4255-4267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
105.  Xie K, Liu L, Chen J, Liu F. Exosomal miR-1246 derived from human umbilical cord blood mesenchymal stem cells attenuates hepatic ischemia reperfusion injury by modulating T helper 17/regulatory T balance. IUBMB Life. 2019;71:2020-2030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 44]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
106.  Li SP, He JD, Wang Z, Yu Y, Fu SY, Zhang HM, Zhang JJ, Shen ZY. miR-30b inhibits autophagy to alleviate hepatic ischemia-reperfusion injury via decreasing the Atg12-Atg5 conjugate. World J Gastroenterol. 2016;22:4501-4514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
107.  Jiang W, Kong L, Ni Q, Lu Y, Ding W, Liu G, Pu L, Tang W. miR-146a ameliorates liver ischemia/reperfusion injury by suppressing IRAK1 and TRAF6. PLoS One. 2014;9:e101530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 68]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
108.  Li X, Yi S, Deng Y, Cheng J, Wu X, Liu W, Tai Y, Chen G, Zhang Q, Yang Y. MiR-124 protects human hepatic L02 cells from H2O2-induced apoptosis by targeting Rab38 gene. Biochem Biophys Res Commun. 2014;450:148-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
109.  Jiang W, Liu G, Tang W. MicroRNA-182-5p Ameliorates Liver Ischemia-Reperfusion Injury by Suppressing Toll-Like Receptor 4. Transplant Proc. 2016;48:2809-2814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
110.  Huang X, Gao Y, Qin J, Lu S. miR-214 Down-Regulation Promoted Hypoxia/Reoxygenation-Induced Hepatocyte Apoptosis Through TRAF1/ASK1/JNK Pathway. Dig Dis Sci. 2019;64:1217-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
111.  Zhang Y, Lv J, Wu G, Li W, Zhang Z, Lei X. MicroRNA-449b-5p targets HMGB1 to attenuate hepatocyte injury in liver ischemia and reperfusion. J Cell Physiol. 2019;234:16367-16375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
112.  Li Y, Gao M, Xu LN, Yin LH, Qi Y, Peng JY. MicroRNA-142-3p attenuates hepatic ischemia/reperfusion injury via targeting of myristoylated alanine-rich C-kinase substrate. Pharmacol Res. 2020;156:104783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
113.  Shen A, Zheng D, Luo Y, Mou T, Chen Q, Huang Z, Wu Z. MicroRNA-24-3p alleviates hepatic ischemia and reperfusion injury in mice through the repression of STING signaling. Biochem Biophys Res Commun. 2020;522:47-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
114.  Duan Y, Meng Y, Gao Z, Wang X, Zhang H. microRNA-9-5p protects liver sinusoidal endothelial cell against oxygen glucose deprivation/reperfusion injury. Open Life Sci. 2021;16:375-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
115.  Luo YH, Huang ZT, Zong KZ, Cao ZR, Peng DD, Zhou BY, Shen A, Yan P, Wu ZJ. miR-194 ameliorates hepatic ischemia/reperfusion injury via targeting PHLDA1 in a TRAF6-dependent manner. Int Immunopharmacol. 2021;96:107604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
116.  Li X, Wu L, Tian X, Zheng W, Yuan M, Zuo H, Song H, Shen Z. miR-29a-3p in Exosomes from Heme Oxygenase-1 Modified Bone Marrow Mesenchymal Stem Cells Alleviates Steatotic Liver Ischemia-Reperfusion Injury in Rats by Suppressing Ferroptosis via Iron Responsive Element Binding Protein 2. Oxid Med Cell Longev. 2022;2022:6520789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
117.  Wang YL, Zhang Y, Cai DS. Hepatoprotective effects of sevoflurane against hepatic ischemia-reperfusion injury by regulating microRNA-124-3p-mediated TRAF3/CREB axis. Cell Death Discov. 2022;8:105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
118.  Wu L, Tian X, Zuo H, Zheng W, Li X, Yuan M, Song H. miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J Nanobiotechnology. 2022;20:196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 54]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
119.  Yu Q, Chen S, Tang H, Yang H, Zhang J, Shi X, Li J, Guo W, Zhang S. miR1405p alleviates mouse liver ischemia/reperfusion injury by targeting CAPN1. Mol Med Rep. 2021;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
120.  Zheng D, Li Z, Wei X, Liu R, Shen A, He D, Tang C, Wu Z. Role of miR-148a in Mitigating Hepatic Ischemia-Reperfusion Injury by Repressing the TLR4 Signaling Pathway via Targeting CaMKIIα in Vivo and in Vitro. Cell Physiol Biochem. 2018;49:2060-2072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
121.  van Heesch S, van Iterson M, Jacobi J, Boymans S, Essers PB, de Bruijn E, Hao W, MacInnes AW, Cuppen E, Simonis M. Extensive localization of long noncoding RNAs to the cytosol and mono- and polyribosomal complexes. Genome Biol. 2014;15:R6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 276]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
122.  Mercer TR, Mattick JS. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 2013;20:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1037]  [Cited by in F6Publishing: 1087]  [Article Influence: 98.8]  [Reference Citation Analysis (0)]
123.  Noh JH, Kim KM, McClusky WG, Abdelmohsen K, Gorospe M. Cytoplasmic functions of long noncoding RNAs. Wiley Interdiscip Rev RNA. 2018;9:e1471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 256]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
124.  Li W, Wang YY, Xiao L, Ding J, Wang L, Wang F, Sun T. Mysterious long noncoding RNAs and their relationships to human disease. Front Mol Biosci. 2022;9:950408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
125.  Wapinski O, Chang HY. Long noncoding RNAs and human disease. Trends Cell Biol. 2011;21:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1424]  [Cited by in F6Publishing: 1517]  [Article Influence: 116.7]  [Reference Citation Analysis (0)]
126.  Wang K, Liu F, Liu CY, An T, Zhang J, Zhou LY, Wang M, Dong YH, Li N, Gao JN, Zhao YF, Li PF. The long noncoding RNA NRF regulates programmed necrosis and myocardial injury during ischemia and reperfusion by targeting miR-873. Cell Death Differ. 2016;23:1394-1405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 163]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
127.  Liu D, Liu Y, Zheng X, Liu N. c-MYC-induced long noncoding RNA MEG3 aggravates kidney ischemia-reperfusion injury through activating mitophagy by upregulation of RTKN to trigger the Wnt/β-catenin pathway. Cell Death Dis. 2021;12:191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
128.  Jia P, Xu S, Ren T, Pan T, Wang X, Zhang Y, Zou Z, Guo M, Zeng Q, Shen B, Ding X. LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury. Cell Death Dis. 2022;13:562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
129.  Vasudeva K, Dutta A, Munshi A. Role of lncRNAs in the Development of Ischemic Stroke and Their Therapeutic Potential. Mol Neurobiol. 2021;58:3712-3728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
130.  Chen Z, Jia S, Li D, Cai J, Tu J, Geng B, Guan Y, Cui Q, Yang J. Silencing of long noncoding RNA AK139328 attenuates ischemia/reperfusion injury in mouse livers. PLoS One. 2013;8:e80817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
131.  Li D, Chen G, Yang J, Fan X, Gong Y, Xu G, Cui Q, Geng B. Transcriptome analysis reveals distinct patterns of long noncoding RNAs in heart and plasma of mice with heart failure. PLoS One. 2013;8:e77938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 74]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
132.  Gu XX, Xu XX, Liao HH, Wu RN, Huang WM, Cheng LX, Lu YW, Mo J. Dexmedetomidine hydrochloride inhibits hepatocyte apoptosis and inflammation by activating the lncRNA TUG1/miR-194/SIRT1 signaling pathway. J Inflamm (Lond). 2021;18:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
133.  Wang L, Qu P, Yin W, Sun J. Lnc-NEAT1 induces cell apoptosis and inflammation but inhibits proliferation in a cellular model of hepatic ischemia/reperfusion injury. J Int Med Res. 2021;49:300060519887251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
134.  Zhang Y, Zhang H, Zhang Z, Li S, Jiang W, Li X, Lv J. LncRNA MALAT1 cessation antagonizes hypoxia/reoxygenation injury in hepatocytes by inhibiting apoptosis and inflammation via the HMGB1-TLR4 axis. Mol Immunol. 2019;112:22-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
135.  Wang C, Yu H, Lu S, Ke S, Xu Y, Feng Z, Qian B, Bai M, Yin B, Li X, Hua Y, Dong L, Li Y, Zhang B, Li Z, Chen D, Chen B, Zhou Y, Pan S, Fu Y, Jiang H, Wang D, Ma Y. LncRNA Hnf4αos exacerbates liver ischemia/reperfusion injury in mice via Hnf4αos/Hnf4α duplex-mediated PGC1α suppression. Redox Biol. 2022;57:102498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Reference Citation Analysis (0)]
136.  Huang X, Gao Y, Qin J, Lu S. The mechanism of long non-coding RNA MEG3 for hepatic ischemia-reperfusion: Mediated by miR-34a/Nrf2 signaling pathway. J Cell Biochem. 2018;119:1163-1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
137.  Ying D, Zhou X, Ruan Y, Wang L, Wu X. LncRNA Gm4419 induces cell apoptosis in hepatic ischemia-reperfusion injury via regulating the miR-455-SOX6 axis. Biochem Cell Biol. 2020;98:474-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
138.  Zhou Z, Chen Q, Wan L, Zheng D, Li Z, Wu Z. Dexmedetomidine protects hepatic cells against oxygen-glucose deprivation/reperfusion injury via lncRNA CCAT1. Cell Biol Int. 2018;42:1250-1258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
139.  Dai B, Qiao L, Zhang M, Cheng L, Zhang L, Geng L, Shi C, Sui C, Shen W, Sun Y, Chen Q, Hui D, Wang Y, Yang J. lncRNA AK054386 Functions as a ceRNA to Sequester miR-199 and Induce Sustained Endoplasmic Reticulum Stress in Hepatic Reperfusion Injury. Oxid Med Cell Longev. 2019;2019:8189079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
140.  Tang B, Bao N, He G, Wang J. Long noncoding RNA HOTAIR regulates autophagy via the miR-20b-5p/ATG7 axis in hepatic ischemia/reperfusion injury. Gene. 2019;686:56-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
141.  Li X, Su Q, Li W, Zhang X, Ran J. Analysis and identification of potential key genes in hepatic ischemia-reperfusion injury. Ann Transl Med. 2022;10:1375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
142.  Ming N, Na HST, He JL, Meng QT, Xia ZY. Propofol alleviates oxidative stress via upregulating lncRNA-TUG1/Brg1 pathway in hypoxia/reoxygenation hepatic cells. J Biochem. 2019;166:415-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
143.  Sun Q, Gong J, Wu J, Hu Z, Zhang Q, Zhu X. SNHG1-miR-186-5p-YY1 feedback loop alleviates hepatic ischemia/reperfusion injury. Cell Cycle. 2022;21:1267-1279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
144.  Zhang XO, Dong R, Zhang Y, Zhang JL, Luo Z, Zhang J, Chen LL, Yang L. Diverse alternative back-splicing and alternative splicing landscape of circular RNAs. Genome Res. 2016;26:1277-1287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 656]  [Article Influence: 82.0]  [Reference Citation Analysis (0)]
145.  Huang C, Liang D, Tatomer DC, Wilusz JE. A length-dependent evolutionarily conserved pathway controls nuclear export of circular RNAs. Genes Dev. 2018;32:639-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 226]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
146.  Chen LL. The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat Rev Mol Cell Biol. 2020;21:475-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 769]  [Article Influence: 192.3]  [Reference Citation Analysis (0)]
147.  Gao Y, Zhou Y, Wei L, Feng Z, Chen Y, Liu P, Peng Y, Huang Q, Gao L, Liu Y, Han Y, Shen H, Cai C, Zeng S. Hsa_Circ_0066351 Acts as a Prognostic and Immunotherapeutic Biomarker in Colorectal Cancer. Front Immunol. 2022;13:927811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
148.  Xia P, Wang S, Ye B, Du Y, Li C, Xiong Z, Qu Y, Fan Z. A Circular RNA Protects Dormant Hematopoietic Stem Cells from DNA Sensor cGAS-Mediated Exhaustion. Immunity. 2018;48:688-701.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 157]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
149.  Piwecka M, Glažar P, Hernandez-Miranda LR, Memczak S, Wolf SA, Rybak-Wolf A, Filipchyk A, Klironomos F, Cerda Jara CA, Fenske P, Trimbuch T, Zywitza V, Plass M, Schreyer L, Ayoub S, Kocks C, Kühn R, Rosenmund C, Birchmeier C, Rajewsky N. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science. 2017;357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 683]  [Cited by in F6Publishing: 713]  [Article Influence: 101.9]  [Reference Citation Analysis (0)]
150.  Huang C, Qu Y, Feng F, Zhang H, Shu L, Zhu X, Huang G, Xu J. Cardioprotective Effect of circ_SMG6 Knockdown against Myocardial Ischemia/Reperfusion Injury Correlates with miR-138-5p-Mediated EGR1/TLR4/TRIF Inactivation. Oxid Med Cell Longev. 2022;2022:1927260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
151.  Gao XQ, Liu CY, Zhang YH, Wang YH, Zhou LY, Li XM, Wang K, Chen XZ, Wang T, Ju J, Wang F, Wang SC, Wang Y, Chen ZY. The circRNA CNEACR regulates necroptosis of cardiomyocytes through Foxa2 suppression. Cell Death Differ. 2022;29:527-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
152.  Zhou LY, Zhai M, Huang Y, Xu S, An T, Wang YH, Zhang RC, Liu CY, Dong YH, Wang M, Qian LL, Ponnusamy M, Zhang YH, Zhang J, Wang K. The circular RNA ACR attenuates myocardial ischemia/reperfusion injury by suppressing autophagy via modulation of the Pink1/ FAM65B pathway. Cell Death Differ. 2019;26:1299-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 170]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
153.  Yang Z, Huang C, Wen X, Liu W, Huang X, Li Y, Zang J, Weng Z, Lu D, Tsang CK, Li K, Xu A. Circular RNA circ-FoxO3 attenuates blood-brain barrier damage by inducing autophagy during ischemia/reperfusion. Mol Ther. 2022;30:1275-1287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 48]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
154.  Xu Y, Jiang W, Zhong L, Li H, Bai L, Chen X, Lin Y, Zheng D. circ-AKT3 aggravates renal ischaemia-reperfusion injury via regulating miR-144-5p /Wnt/β-catenin pathway and oxidative stress. J Cell Mol Med. 2022;26:1766-1775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
155.  Tian X, Hu Y, Liu Y, Yang Z, Xie H, Zhou L, Zheng S. Circular RNA Microarray Analyses in Hepatic Ischemia-Reperfusion Injury With Ischemic Preconditioning Prevention. Front Med (Lausanne). 2021;8:626948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
156.  Ma X, Huang Y, Ding Y, Shi L, Zhong X, Kang M, Li C. Analysis of piRNA expression spectra in a non-alcoholic fatty liver disease mouse model induced by a methionine- and choline-deficient diet. Exp Ther Med. 2020;19:3829-3839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
157.  Xu J, Yang X, Zhou Q, Zhuang J, Han S. Biological significance of piRNA in liver cancer: a review. Biomarkers. 2020;25:436-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
158.  Xu L, Chen W, Chen J, Jin Y, Ma W, Qi G, Sun X, Luo J, Li C, Zhao K, Zheng Y, Yu D. PIWI-interacting RNA-23210 protects against acetaminophen-induced liver injury by targeting HNF1A and HNF4A. Biochem Pharmacol. 2022;197:114897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
159.  Liu X, Xie W, Meng S, Kang X, Liu Y, Guo L, Wang C. Small Nucleolar RNAs and Their Comprehensive Biological Functions in Hepatocellular Carcinoma. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
160.  Huang P, Tu B, Liao HJ, Huang FZ, Li ZZ, Zhu KY, Dai F, Liu HZ, Zhang TY, Sun CZ. Elevation of plasma tRNA fragments as a promising biomarker for liver fibrosis in nonalcoholic fatty liver disease. Sci Rep. 2021;11:5886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
161.  Zhu J, Cheng M, Zhao X. A tRNA-derived fragment (tRF-3001b) aggravates the development of nonalcoholic fatty liver disease by inhibiting autophagy. Life Sci. 2020;257:118125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
162.  Law PT, Qin H, Ching AK, Lai KP, Co NN, He M, Lung RW, Chan AW, Chan TF, Wong N. Deep sequencing of small RNA transcriptome reveals novel non-coding RNAs in hepatocellular carcinoma. J Hepatol. 2013;58:1165-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 137]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]