Open Access

An update on the functional roles of long non‑coding RNAs in ischemic injury (Review)

  • Authors:
    • Yanqun Cao
    • Jia Liu
    • Quzhe Lu
    • Kai Huang
    • Baolin Yang
    • James Reilly
    • Na Jiang
    • Xinhua Shu
    • Lei Shang
  • View Affiliations

  • Published online on: May 18, 2022     https://doi.org/10.3892/ijmm.2022.5147
  • Article Number: 91
  • Copyright : © Cao et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ischemic injuries result from ischemia and hypoxia in cells. Tissues and organs receive an insufficient supply of nutrients and accumulate metabolic waste, which leads to the development of inflammation, fibrosis and a series of other issues. Ischemic injuries in the brain, heart, kidneys, lungs and other organs can cause severe adverse effects. Acute renal ischemia induces acute renal failure, heart ischemia induces myocardial infarction and cerebral ischemia induces cerebrovascular accidents, leading to loss of movement, consciousness and possibly, life‑threatening disabilities. Existing evidence suggests that long non‑coding RNAs (lncRNAs) are regulatory sequences involved in transcription, post‑transcription, epigenetic regulation and multiple physiological processes. lncRNAs have been shown to be differentially expressed following ischemic injury, with the severity of the ischemic injury being affected by the upregulation or downregulation of certain types of lncRNA. The present review article provides an extensive summary of the functional roles of lncRNAs in ischemic injury, with a focus on the brain, heart, kidneys and lungs. The present review mainly summarizes the functional roles of lncRNA MALAT1, lncRNA MEG3, lncRNA H19, lncRNA TUG1, lncRNA NEAT1, lncRNA AK139328 and lncRNA CAREL, among which lncRNA MALAT1, in particular, plays a crucial role in ischemic injury and is currently a hot research topic.

1. Introduction

Ischemic injury is caused by insufficient blood perfusion in cells, tissues and organs due to a limited blood supply. Given that blood is the carrier of oxygen, the main cause of ischemia is hypoxia. However, ischemia also involves an insufficient supply of nutrients and clearance of metabolic wastes, which leads to inflammation, acidosis and an electrolyte imbalance. Ischemic injuries to the brain, heart, kidneys, lungs and other organs can cause severe damage (1). For instance, stroke is an acute brain injury that involves brain damage and neurological dysfunction, and is associated with a high mortality or disability rate globally. Acute renal ischemia can cause acute renal failure, while heart ischemia can cause myocardial infarction (2-4).

Long non-coding RNAs (lncRNAs) refer to the transcripts of non-coding RNAs that are >200 nucleotides in length (5). Previous research defined lncRNAs as by-products of transcription; however, recent research has found that they regulate various physiological and pathophysiological processes (6). lncRNAs regulate gene expression at the epigenetic, transcriptional, post-transcriptional and chromatin remodeling levels (6). They also activate or inhibit the expression of target genes by directly binding them or recruiting transcription factors (5).

Recently, research on lncRNAs has become a hot topic, with several studies suggesting that lncRNAs are involved in ischemic injuries (7,8). For instance, experimental models (9) have found that lncRNA expression differs between stroke patients and healthy individuals, and that genetic variations of lncRNA loci are associated with an increased risk of suffering from a stroke (2). lncRNAs are highly specific in the central nervous system and stroke is known to induce changes in several lncRNAs in the brain. Therefore, lncRNAs play a crucial role in the complex pathological processes associated with stroke, and may regulate the development of the central nervous system and disease progression (10). During the tra/rensplantation of lungs, kidneys and other organs, ischemia/reperfusion (I/R) is a critical factor affecting the success rate of organ transplantation (11,12). The present review article discusses the functional roles of lncRNAs in ischemic injuries in different organs.

2. Synthesis of lncRNAs

RNA-producing genes can be divided into two main types: Protein-coding genes and non-protein-coding genes. Of the 180,000 transcripts in human cells, ~20,000 are protein-coding and the remaining 160,000 are non-coding transcripts (6). Non-coding RNAs are divided into two groups based on their size: Short non-coding RNAs (<200 nt), e.g., microRNAs (miRNAs/miRs) and long non-coding RNAs (≥200 bp), such as lncRNAS (13). Recent estimates suggest there are >100,000 lncRNAs (14,15). In most cases, mRNA and lncRNA are similar in several aspects of their biogenesis, although there are differences between them (5,16). Similar to mRNAs, the synthesis of lncRNAs requires one of the two strands of DNA as a template. The majority of lncRNAs are transcribed by RNA polymerase II (pol II), m7G capped at 5′-end and polyadenylated at 3′-end (17). In contrast to mRNAs, IncRNAs are not well conserved in evolution, consist of fewer exons and are expressed at relatively low levels; large proportions of IncRNAS are also retained in the nucleus (18).

Increasing evidence has indicated that lncRNAs play a crucial role in regulating gene expression, such as gene silencing or gene activation. Furthermore, the regulation of lncRNAs can affect the transcription, splicing, translation, output, import and stability of mRNAs (19). The role of lncRNAs in the process of ischemic injury will be discussed in detail in the following sections.

3. Role of lncRNAs in cerebral ischemic injury

Cerebral ischemic injuries, including ischemic stroke and cerebral ischemia-reperfusion injuries, can lead to severe brain dysfunction, disability and a high mortality. lncRNAs are highly expressed in the brain, indicating that they may be involved in the physiological and pathological processes of the brain, such as cerebral ischemic injury, neurodegeneration, neurodevelopment and plasticity (20,21). A number of studies have demonstrated that lncRNAs play a central role in ischemic brain injuries (Table I).

Table I

Role of lncRNAs in cerebral ischemic injuries.

Table I

Role of lncRNAs in cerebral ischemic injuries.

lncRNAEffect on miRNAs and signaling pathways (Refs.)Effect on inflammationEffect on apoptosisEffect on autophagyEffect on other aspectsEffect on cerebral ischemic injury
KCNQ1OT1miR-200a/FOXO3/ATG7 (51)
MALAT1 miR-30a/Beclin1/autophagy (27)
miR-145/AQP42 (28)
miR-211-5p (52)
miR-142-3p/SIRT1 (53)Acts as ceRNA for miR-142-3p
miR-375/PDE4D (54)
miR-181c-5p/HMGB1 (29)Acts as ceRNA for miR-181c-5p
miR-195a-5p/HMGA1 (55)Sponging miR-195a-5p to upregulate HMGA1
miR-182-5p/TLR4 (56)
miR-375/PDE4D (54)
Bim and E-selectin (25)
ERK/MAPK (57)
MyD88/IRAK1/TRAF6 (58)
AQP4 (59)
Gm11974miR-766-3p/NR3C2 (46)
miR-122-5P/SEMA3A (60)
MEG3miR-485/AIM2 (35)
miR-378/GRB2 (61)Protect neurons
miR-181c-5p/ATG7 (62)
NF-κB (63)
p53/GPX4 (64)Mediate ferroptosis of brain microvascular
endothelial cells
H19miR-29b/Akt3/mTOR (40)Acts as a miR-29b sponge
miR-138-5p/p65 (65)
miRNA-29b/SIRT1/PGC-1α (66)
miR-107 (41)
miR-1306-5p/BCL2L13 (67)Acts as a miR-1306-5p sponge
miR-19a-3p/PTEN (68)
DUSP5-ERK1/2 (69)
IGF1/mTOR (42)Inhibits axon sprouting and functional recovery
RORmiR-135a-5p/ROCK1/2 (70)
NespasModulates TNFα-induced apoptosis (71)
SNHG12miR-199a/SIRT1/AMPK (72)
Inhibits TRIM8-related K63-linked
polyubiquitination of TAK1 (73)
Acts as an autophagy inducer (74)
SIRT1/FOXO3a (75)
SNHG14miR-136-5p/ROCK1 (76)
miR-182-5p/BINP3 (77)
miR-181c-5p/BMF (78)
miR-199b/AQP4 (79)
MIATmiR-204-5p/HMGB1 (80)
miR-211/GDNF (48)
REDD1 (81)
TUG1miR-145a-5p (82)
miR-204-5p/COX2 (83)Sponging miR-204-5p
miR-145/AQP4 (84)Sponging miR-145
miR-200a-3p/NLRP3 (45)
miR-142-3p (85)
miR-493-3p or miR-410-3p (44)Sponging miR-493-3p/miR-410-3p
RMSTmiR-150 (86)Inhibits cell proliferation
Protects against MCAO-induced
ischemic stroke (87)
N1LRp53 (86)
CHRFmiR-126/SOX6 (47)
Inhibits p53 phosphorylation (88)
Oprm1miR-155/GATA3 (49)

[i] lncRNA, long non-coding RNA; MALAT1, metastatic-associated lung adenocarcinoma transcript 1; TUG1, taurine up-regulated gene 1; I/R, ischemia/reperfusion; MEG3, maternal expression of gene 3; MCAO, middle cerebral artery occlusion; KCNQ1OT1, potassium voltage-gated channel subfamily Q member 1 opposite strand 1; SNHG12, small nuclear RNA host gene 12; SNHG14, small nuclear RNA host gene 14; MIAT, myocardial infarction associated transcript; RMST, rhabdomyosarcoma 2-associated transcript; N1LR, novel I/R-induced lncRNA; CHRF, cardiac hypertrophy-related factor; Oprm1, opioid receptor Mu 1. The '↓' symbol implies the downregulation of inflammation, apoptosis, etc.; and the '↑' symbol implies thje upregulation of inflammation, apoptosis, etc.

Metastatic-associated lung adenocarcinoma transcript 1 (MALAT1)

Cerebral ischemic stroke is a main cause of mortality and long-term disability globally (5) and is one of the most common cerebral diseases, particularly among the elderly. MALAT1 is closely associated with abnormal cellular signaling, as well as with the occurrence, development and response to the treatment of human diseases. It has been shown that MALAT1 plays a role in the pathophysiology of a number of diseases and may be a therapeutic or prognostic biological target (22,23). A previous study demonstrated that MALAT1 was strongly induced in anoxic mouse organs, particularly in the spleen, kidneys, testicles, lungs and brain (24).

In 2017, Zhang et al (25) explored the relevant mechanisms of MALAT1 in regulating cerebral vascular lesions in ischemic stroke. These authors established a mouse model of transient focal cerebral ischemia through intracavitary middle cerebral artery occlusion (MCAO) and then evaluated the infarct volume, neurological deficits and sensorimotor function. Additionally, they simulated ischemic injury in vitro by exposing rat brain microvascular endothelial cells (BMECs) to oxygen-glucose deprivation (OGD) for 16 h. RNA sequencing revealed that the levels of MALAT1 in the OGD-exposed BMEC cells and in ischemic cerebral microvessels were significantly increased, suggesting that the upregulation of MALAT1 may play a crucial role in cerebrovascular pathologies induced by ischemic stroke (25). Furthermore, fluorescein-labeled MALAT1-Gapmer was introduced into mouse BMECs in in vitro culture to achieve targeted MALAT1 degradation, resulting in a significant reduction in the changes of OGD-induced MALAT1 levels, accompanied by increased cerebrovascular endothelial cell death and programmed death-activating factor caspase-3 activity. In addition, MALAT1 knockout mice and litter control mice were briefly treated with MCAO for 1 h and perfused for 24 h. The results revealed that MALAT1 knockout mice displayed a larger infarct volume and significantly more severe neurological deficits. These results suggest that the loss of MALAT1 exacerbates ischemic brain injury. In subsequent experiments, the researchers found that MALAT1 bound directly to Bim (a pro-apoptotic regulator) and e-selectin. The silencing of MALAT1 increased the expression of Bim and pro-inflammatory cytokines. Cerebrovascular endothelial inflammation and subsequent endothelial function damage are associated with ischemic brain injuries (21).

Similarly, in 2018, Zhang et al (26) also explored the mechanisms of MALAT1 in ischemic stroke. The mouse dual-2-min homolog (MDM2) gene encodes the E3 ubiquitin ligase of p53 in mitotic cells, which is an oncoprotein that blocks the transcriptional activation mediated by p53 tumor suppressors. MALAT1 was found to be highly expressed in ischemic stroke samples, including human brain microvascular endothelial cells (HBMECs). MALAT1 significantly promoted the occurrence of an ischemic stroke by regulating MDM2 expression and activating the p53 signaling pathway. Subsequently, Zhang et al (26) found that MALAT1 and MDM2 were highly expressed in HBMECs and that both MALAT1 and MDM2 were localized in the nucleus following OGD/reoxygenation (OGD/R). The expression of the p53 signaling pathway-related proteins was highly increased in HBMECs exposed to OGD/R. Furthermore, the knockdown of MALAT1 effectively reduced the expression of MDM2, while the knockdown of MDM2 had no effect on the expression of MALAT1. These results revealed that MALAT1 regulated and promoted the expression of MDM2 following OGD/R. OGD/R also promoted apoptosis through the p53 signaling pathway, while the silencing of MALAT1 and MDM2 inhibited this effect. MCAO/reperfusion (MCAO/R) in mouse brains also caused a similar effect on expression of related proteins and lncRNAs (26).

Additionally, several studies have indicated that MALAT1 plays a crucial role in the progression of ischemic brain injury. In 2017, Guo et al (27) found that MALAT1, the autophagy-related microtubule-related proteins light chain 3-I (LC3-I)/LC3-II and Beclin1 were upregulated in the cortical neurons of mice following MCAO/R. Furthermore, neuronal cell death was significantly increased following OGD compared with the sham group; the downregulation of MALAT1 significantly reversed this effect. Additionally, the downregulation of MALAT1 significantly inhibited the increase in LC3-I/LC3-II and Beclin1 levels induced by OGD (27). This indicates that the downregulation of MALAT1 can inhibit the occurrence of autophagy and ischemic injury. It was also demonstrated that the downregulation of MALAT1 alleviated neuronal cell death by regulating the Beclin1-dependent autophagy of miR-30a during an ischemic stroke (27).

In 2020, Wang et al (28) established the OGD/R an in vitro astrocyte model and an MCAO/R in vivo mouse model. In both models, the level of MALAT1 was significantly upregulated and the expression of aquaporin 4 (AQP4) was significantly increased. Following intervention with MALAT1 siRNA, the cell survival rate was increased and apoptosis was reduced. Further experiments revealed that MALAT1 siRNA could not reduce the aggravation of the injury induced by a miR-145 inhibitor. This indicates that MALAT1 silencing protects the cerebral ischemia-reperfusion injury through miR-145. These results suggest that MALAT1 positively regulates AQP4 expression through miR-145 and promotes cerebral ischemia-reperfusion injury (28). Cao et al (29) also found that the depletion of MALAT1 suppressed inflammatory injury in the ischemic brain and that the overexpression of MALAT1 had an adverse effect; in fact, MALAT1 positively regulates the high-mobility Group Box 1 (HMGB1) to promote inflammation by acting as a competing endogenous RNA (ceRNA) to inhibit the function of miR-181C-5p (29). Another study demonstrated that MALAT1 also inhibited PI3K/AKT signaling by sponging miR-126 to promote the OGD-induced apoptosis of cerebral microvascular endothelial cells (30). Thus, MALAT1 may be a novel target for the treatment of cerebral ischemic stroke.

Nevertheless, a few studies have reported inconsistent results concerning the effects of MALAT1 in ischemic brain injuries. For example, polydatin (a natural product) upregulated MALAT1 expression and activated the C/EBP/MALAT1/CREB/PGC-1/PPAR pathway to protect the brain microvascular integrity and decrease the damage caused by cerebral stroke (31). MALAT1 can protect cerebral micro-vascular endothelial cells from OGD injuries by sponging miR-26b and upregulating the expression of ULK2 (12,32).

Maternal expression of gene 3 (MEG3)

MEG3 plays an antitumor role in various types of cancer by regulating the major tumor suppressor genes, p53 and Rb, inhibiting angiogenesis-related factors and controlling miRNAs (33). Recent experiments have also explored the mechanisms of MEG3 involved in cerebral ischemic injuries.

In 2019, You and You (34) found that MEG3 was upregulated in rat models of MCAO. The rats with MACO exhibited evident nervous system injuries, infarction areas, an increase in the blood-brain barrier (BBB) permeability, water content, neuronal apoptosis and necrosis. An increase was also observed in the levels of the Wnt/β-catenin signaling pathway key proteins, including brain-derived neurotrophic factor (BDNF), the nerve growth factor (NGF) and basic fibroblast growth factor (bFGF). The depletion of MEG3 by siRNA (si-MEG3) resulted in alleviated neurological damage, reduced infarct area, water content, BBB permeability, neuronal apoptosis and necrosis, and significantly increased neurogenesis, whereas the overexpression of MEG3 exerted opposite effects. Following treatment with the classic Wnt pathway inhibitor, DKK1, the effect of si-MEG3 was reversed. The effect of MEG3 was also reversed following treatment with LiCl, a classical Wnt pathway activator. These experimental results suggest that the downregulation of the lncRNA MEG3 activates the Wnt/β-catenin signaling pathway, which further promotes nerve cell growth in rats following cerebral I/R injury, and alleviates nervous tissue injuries (34).

In 2020, Liang et al (35) reported that MEG3 was significantly upregulated and miR-485 was significantly downregulated in brain tissues following cerebral I/R, suggesting that MEG3 and miR-485 may be involved in the regulation of brain I/R. The pyroptosis in the brains of rats wit h MCAO was significantly higher than that in the sham-operated group, while the expression of absent in melanoma 2 (AIM2), an inflammatory factor closely related to pyroptosis, was also significantly increased in the I/R group. The protein levels of several key components of the AIM2 inflammatory signal pathway, including AIM2, lysed caspase-1 (c-caspase-1), IL-1β and IL-18 were significantly increased in the brains of rats in the I/R group, suggesting that I/R in the brain activates AIM2 inflammasome signaling, and subsequently induces pyroptosis and the secretion of inflammatory cytokines (35). Further data indicated that MEG3 negatively regulated the expression of miR-485, which can directly bind the 3′-UTR of AIM2 to inhibit its expression. Furthermore, the knockdown of MEG3 inhibited OGD/R-induced pyroptosis by directly increasing the level of miR-485 (35). Other studies have reported similar observations. For example, several researchers have found that MEG3 exacerbrates oxygen-deficient damage in PC12 cells by targeting miR-147 (36). MEG3 silencing also protects PC12 cells from hypoxic damage by sponging miR-21 and regulating both the PI3K/AKT and NF-κB pathways (37). MEG3 silencing can also enhance the cerebral protective effect of dexmedetomidine against hypoxic-ischemic brain injuries in newborn mice via the upregulation of miR-129-5p, leading to decreased neuronal cell death and cerebral atrophy (38).

These studies also demonstrate that the regulation of brain neurogenesis by targeting miRNAs is a common action form of MEG3. The data suggest that MEG3 inhibits AIM2 expression by sponging miR-485 during the I/R process in the brain, leading to pyroptosis and inflammation (31), thus providing a novel direction for the targeted therapy of ischemic stroke.

lncRNA H19

lncRNA H19 was previously misidentified as 'transcriptional noise' (39). Existing studies have found that H19 plays a critical role in the pathogenesis of various central nervous system diseases, such as Parkinson's disease, Alzheimer's disease, cerebral ischemia, cerebral hemorrhage and neuroblastoma (39). A number of studies have been conducted to investigate the mechanisms of the role of H19 in the process of ischemic brain injury.

In a study in 2020, the researchers established a neonatal rat model of hypoxic-ischemic encephalopathy (HIE), which exhibited an increased area of cerebral infarction, apoptosis and impaired neurological function in neonatal rats with HIE (40). That study confirmed that the overexpression of H19 functioned as a sponge for miR-29b and attenuated neural damage and reduced autophagy in brain tissue in neonatal rats with HIE by upregulating the Akt3/mTOR pathway. The effect produced by the overexpression of H19 could also be partially reversed by autophagy activators, which suggests that H19 affects autophagy (40). Furthermore, research on hypoxic-ischemic brain injury has confirmed that H19 overexpression is able to decrease miR-107 expression and increase VEGF expression, which results in the inhibition of neuronal apoptosis and the alleviation of cognitive dysfunction (41).

The two aforementioned studies demonstrate the positive role of H19 overexpression in cerebral I/R; other studies have demonstrated that H19 knockdown is capable of ischemic brain injury-induced inflammation and neurological damage. For example, the knockdown of lncRNA H19 has been shown to promotes axon sprouting and functional recovery following cerebral ischemic stroke (42). Another study established an in vivo model of I/R using rats with MCAO and an in vitro model of cells exposed to OGD/R and revealed that the knockdown of H19 promoted cell proliferation, decreased the rate of apoptosis, and alleviated the inflammatory status following OGD/R (39). That study also revealed that H19 was involved in this process by regulating the miR-138-5p/p65 axis (39). Similarly, another study established in vitro and in vivo models of cerebral I/R (40). The results of that study confirmed that lncRNA H19 knockdown attenuated the OGD-mediated downregulation of miR-29b, SIRT1 and PGC-1α expression levels, which in turn ameliorated the OGD-induced increase in apoptosis and the concentration of inflammatory cytokines (40). Another study also demonstrated that H19 functioned as a sponge for miR-1306-5p and triggered cerebral I/R-induced injury, and that the knockdown of H19 alleviated injury (41). Blocking the lncRNA H19-miR-19a-Id2 axis has also been found to attenuate hypoxia/ischemia-induced neuronal injuries (43). Other pathways in which H19 is involved are listed in Table I, which also fully illustrates the close association of H19 with cerebral I/R.

The existing studies suggest that H19 plays a crucial role in cerebral I/R and is involved in various mechanisms, such as autophagy, apoptosis and inflammation.

lncRNA taurine upregulated gene 1 (TUG1)

lncRNA TUG1 is one of the first lncRNAs identified to be associated with human diseases, and is known to be involved in and to regulate the biological processes of a number of human diseases. Several studies have been conducted to investigate its mechanisms of action in cerebral ischemic injury.

Several studies have described that TUG1 functions by sponging miRNAs. For example, a study published in 2022 demonstrated that TUG1 sponged miR-204-5p and induced the downregulation of miR-204-5p in the blood of patients with ischemic stroke and in the brains of rats with MCAO/R (43). By contrast, the intracerebral injection of miR-204-5p in rats significantly decreased COX2, IL-1β, TNFα and PGE2 expression, increased IL-10 expression and ameliorated brain injury, inhibited apoptosis, and significantly decreased brain infarcts in rats with MCAO/R. That study confirmed that the TUG1/miR-204-5p/COX2 axis is one of the pathways involved in cerebral ischemic injury associated with TUG1 (43).

A study published in 2020 also reported that TUG1 promoted inflammation and apoptosis by sponging miR-145 and thereby upregulating AQP4 expression in an in vitro cellular model of OGD/R and a rat model of MCAO (44). TUG1 has also been shown to activate the JNK and p38 MAPK signaling pathways by sponging miR-493-3p or miR-410-3p (44). In addition, TUG1 can also promote inflammation in brain ischemic injury through other miRNA (miR-145a-5p or miR-200a-3p)-mediated signaling pathways (45).

All the aforementioned studies have demonstrated that TUG1 negatively affects ischemic injury, increases the expression of inflammatory factors and the occurrence of apoptosis, and that these pathways are dependent on many targeted miRNAs.

Other lncRNAs

Additionally, a large number of studies have examined the involvement of other lncRNAs in cerebral ischemic injuries. Similarly, the majority of these lncRNAs aggravate or protect ischemic brain injuries by directly or indirectly acting on miRNAs and associated signaling pathways. For example, the knockdown of lncRNA Gm11974 has been shown to provide protection against cerebral I/R through the miR-766-3p/NR3C2 axis (46). The lncRNA SNHG12 is a potent autophagy inducer that exerts neuroprotective effects against cerebral I/R injuries (46). The lncRNA CHRF modulates the progression of cerebral I/R injuries via the miR-126/SOX6 signaling pathway (47).

Both the upregulation and downregulation of several lncRNAs can influence ischemic brain injuries. For example, the overexpression of lncRNA MIAT has been found to reduce neuronal apoptosis in a neonatal rat model of hypoxic-ischemic injury through the miR-211/GDNF pathway (48). The overexpression of the lncRNA Oprm1 and Rian attenuate cell apoptosis induced by cerebral ischemia-reperfusion injuries through the Oprm1/miR-155/GATA3 and Rian/miR-144-3p/GATA3 axes, respectively (49,50).

On the whole, a number of lncRNAs are involved in cerebral ischemic injuries and some of these are listed in Table I (25,27-29,35,40-42,44-49,51-88).

4. Role of lncRNAs in heart ischemic injury

Ischemic heart disease mainly refers to myocardial ischemia induced by coronary artery obstruction or stenosis. It often leads to myocardial infarction, left ventricular aneurysm, ventricular septal defect and mitral insufficiency, which is common among middle-aged and older individuals. Myocardial infarction is caused by vascular stenosis, blocked circulation blood flow and insufficient myocardial blood supply due to atherosclerosis in the coronary artery wall. The high incidence of myocardial infarction is a major human health concern. Therefore, it is of utmost clinical significance to comprehensively understand the relevant mechanisms underlying ischemic heart diseases.

lncRNA AK139328 and lncRNA CAREL

In recent years, IncRNAs have been reported to be associated with ischemic heart diseases. Similar to ischemic brain injuries, lncRNAs can promote or prevent the occurrence of ischemic heart injuries via acting on various miRNAs. It has been shown that patients with diabetes display a higher risk of ischemic heart disease and myocardial I/R injury (89). Yu et al (90) investigated the effect of the lncRNA AK139328 on myocardial I/R injury in diabetic mice. First, the researchers established I/R models in normal mice and diabetic mice. Compared to other RNAs, AK139328 was the most notably upregulated in DM and the expression levels of phosphocreatine kinase (CK), creatine kinase myocardial band (CK-MB) and lactate dehydrogenase (LDH) (enzymes associated with myocardial injury) were higher following I/R in the diabetic mice compared with the normal mice. The downregulation of AK139328 significantly inhibited the expression of CK, CK-MB and LDH. While I/R induced the apoptosis of myocardial cells of both normal and diabetic mice, the rate of apoptosis was significantly higher in the myocardial cells of diabetic mice. The downregulation of AK139328 significantly inhibited the activity of caspase-3 in DM and inhibited the apoptosis of cardiomyocytes. Myocardial I/R injuries promoted the expression of the autophagy-related proteins, Atg7, Atg5 and LC3-II/LC3-I, and decreased p62 expression; downregulation of AK139328 reversed the changes in the expression of these proteins and inhibited autophagy. In fact, the knockdown of AK139328 significantly alleviated I/R injury in diabetic mice by regulating miR-204-3p expression (86).

Another study explored the influence of the lncRNA CAREL on heart regeneration, demonstrating that the overexpression of CAREL in cardiomyocytes decreased the division and proliferation of cardiomyocytes, and attenuated heart regeneration after injury. By contrast, the silencing of CAREL significantly promoted cardiac regeneration and improved cardiac function following myocardial infarction in both neonatal and adult mice. In fact, CAREL functioned as the ceRNA for miR-296 which suppressed the expression of its target genes, Trp53inp1 and Itm2a. Furthermore, the overexpression of miR-296 significantly increased the replication of myocardial cells and heart regeneration following injury (91).

lncRNA nuclear enriched transcript 1 (NEAT1)

In recent studies, the function of lncRNA NEAT1 in myocardial I/R injuries has been described. For example, a study in published 2020 demonstrated that NEAT1 was highly expressed in the blood of patients with myocardial infarction and in mouse cardiomyocytes (92). lncRNA NEAT1 inhibited the expression of miR-378a-3p, which in turn inhibited the expression of Atg12 and affected autophagy in cardiomyocytes. It was also found that lncRNA NEAT1 significantly promoted the proliferation and migration of cardiomyocytes and had a protective effect against myocardial injury (92).

In addition to its functions in autophagy, NEAT1 affects cardiac functions, infarct size and myocardial apoptosis. The knockdown of NEAT1 has been shown to attenuate myocardial I/R injury through the miR-140/RhoA axis (93). This demonstrates a negative effect of NEAT1 on myocardial injury, an observation that was confirmed by another study, wherein the inhibition of NEAT1 reduced apoptosis and also attenuated the hypoxia-induced inhibition of proliferation (94). In another study, it was demonstrated that in lipopolysaccharide-induced myocardial injury, the knockdown of NEAT1 significantly ameliorated I/R-induced cardiac insufficiency, oxidative stress and inflammatory response, and also alleviated lipopolysaccharide-induced myocardial injury by inhibiting the TLR2/NF-κB signaling pathway (91).

The existing studies indicate that NEAT1 is involved in various pathways, such as apoptosis, autophagy, inflammatory response and oxidative stress. NEAT1 plays a double-sided role in ischemic injury of the heart, and may thus provide novel strategies for the clinical targeting of myocardial injury.

Other lncRNAs

There are other lncRNAs that have also been implicated in ischemic heart injuries (Table II) (92-121). Several studies found that lncRNAs play a key role in alleviating myocardial infarction injuries and myocardial ischemia-reperfusion injuries. These findings provide new insight into the pathophysiology of acute myocardial I/R injuries, and may lead to the identification of novel biomarkers and therapeutic targets for the detection and treatment of acute myocardial infarction. The pharmacological and genetic manipulation of lncRNAs has the therapeutic potential to improve the clinical outcomes of patients with acute myocardial infarction (122).

Table II

Role of lncRNAs in ischemic heart injury.

Table II

Role of lncRNAs in ischemic heart injury.

lncRNAEffect on miRNAs and signaling pathways (Refs.)Effect on inflammationEffect on apoptosisEffect on autophagyEffect on ischemic heart injuryEffect on other aspects
NEAT1miR-27b/PINK1 (95)
miR-129-5p (94)Suppresses proliferation
miR-378a-3p/Atg12 (92)
miR-140/RhoA (93)
TLR2/NF-κB signaling pathway (96)
Decreases pri-miRNA processing (97)
H19miR-22-3P (98)
miR-22-3p/KDM3A (99)
miR-675/PPARα (100)
miR-29b-3p (101)
YB-1 protein (102)Results in cardiac dilatation, and cardiac fibrosis
Increases the stability of nucleolin protein (103)Is involved in myocardial ischemic preconditioning (IP)
TUG1miR-132-3p/HDAC3 (104)
miR-142-3p (105)
miR-9a-5p/KLF5 (106)
miR-29a-3p (107)
miR-532-5p/Sox8 (108)
MALAT1miR-20b (109)
miR-133 (110)
miR-320/Pten (111)
β-catenin (112)
ANRILmiR-7-5p/SIRT1 (113)
HOTAIRmiR-125/mmp2 (114)
GAS5miR-142-5p (115)
CAIFp53 (116)
XISTmiR-133a/SOCS2 (117)
HIF1A-AS1miRNA-204/SOCS2 (118)Promotes ventricular remodeling
LETmiR-138 (118)
Gm2691PI3K/AKT (119)
MIATNF-κB (120)
RMRPmiR-206/ATG3 (121)

[i] lncRNA, long non-coding RNA; MALAT1, metastatic-associated lung adenocarcinoma transcript 1; TUG1, taurine up-regulated gene 1. NEAT1, nuclear enriched abundant transcript 1; ANRIL, antisense non-coding RNA in the INK4 locus; HOTAIR, HOX transcript antisense RNA; GAS5, growth arrest-specific transcript 5; CAIF, cardiac autophagy inhibitory factor; XIST, X-inactive specific transcript; LET, low expression in tumor; MIAT, myocardial infarction associated transcript; RMRP, RNA component of mitochondria RNA processing endoribonuclease. The '↓' symbol implies the downregulation of inflammation, apoptosis, etc.; and the '↑' symbol implies thje upregulation of inflammation, apoptosis, etc.

5. Role of lncRNAs in renal ischemic injury

An ischemic renal injury refers to damage caused by severe renal ischemia resulting from severe stenosis or obstruction of the main renal artery or its branches. Acute renal ischemia and renal I/R injuries are the main causes of acute renal failure. Chronic ischemia often leads to chronic ischemic nephropathy in middle-aged and elderly patients (123). Several studies have demonstrated that various lncRNAs play a potential role in renal I/R injuries (124,125).

lncRNA MALAT1

The findings of recent studies analyzing the role of MALAT1 in renal ischemic injuries have been inconsistent. For example, one study demonstrated that expression of MALAT1 was significantly increased in mouse kidneys following I/R injury and in human proximal renal tubular epithelial cells (HK2) under hypoxic conditions (111). The knockdown of MALAT1 significantly increased both the expression of NF-κB and HIF-1α, and the secretion of IL-6 and TNFα in hypoxic HK2 cells. This suggests that the knockdown of MALAT1 expression can promote hypoxia-induced inflammation in HK2 cells (111).

Nevertheless, other studies have suggested that MALAT1 may have no effect on renal I/R injury. For instance, Kölling et al (126) found that the level of MALAT1 was increased in kidney biopsies and plasma from patients with acute kidney injury; MALAT1 expression was also increased in hypoxic mouse kidney tissues, endothelial cells and tubular epithelial cells. However, MALAT1 knockout mice exhibited the same degree of extramedullary injury, capillary thinning, fibrosis, inflammatory cell infiltration, inflammatory gene expression and proliferation as that in wild-type mice under unilateral renal I/R injuries. MALAT1 knockout mice and wild-type mice also exhibited similar renal dysfunction in bilateral kidney I/R injuries (126). However, a review of the role of MALAT1 in vascular and psychosomatic diseases stated that most experimental studies have shown that the downregulation of MALAT1 leads to more pronounced atherosclerotic lesions and aggravates renal and other organ damage after ischemia (127).

I/R injury is not only a main cause of acute kidney injury, but also a critical risk factor for delayed or non-functional graft function in renal transplantation. In 2019, Su et al (11), using microarray data, found that MALAT1 was upregulated in I/R injury; MALAT1 expression was increased 3.79-fold in the kidneys of mice with I/R injury when compared with sham-operated mice. The researchers also detected a 40.4% decrease in the expression of miR-139-5p in the kidneys of the mice with I/R injury compared with the sham-operated mice, which is closely related to cell proliferation (11). The afore-mentioned data indicate that both MALAT1 and miR-139-5p are involved in renal I/R injury, although the specific association between them has not yet been fully elucidated.

Other lncRNAs

A number of studies have demonstrated that other lncRNAs play a crucial role in renal ischemic injuries. Several studies have analyzed the miRNA axis under renal ischemic injuries. For example, lncRNA NEAT1 has been shown to promote hypoxia-induced renal tubular epithelial apoptosis through the downregulation of miR-27a-3p (128). The lncRNA GAS5 has also been found to promote apoptosis by competing with endogenous RNA for miR-21 via thrombospondin 1 in acute ischemic kidney injury (129). Furthermore, another study demonstrated that the downregulation of lncRNA TUG1 attenuated inflammation and apoptosis in renal tubular epithelial cells induced by ischemia-reperfusion by sponging miR-449b-5p via HMGB1 and MMP2 (130).

Other studies have confirmed that lncRNAs can regulate renal ischemic injuries by activating other signal pathways. For example, LINC00520, which targets miR-27b-3p, regulates the expression of oncostatin M receptor to promote the development of acute kidney injuries through the PI3K/AKT signaling pathway (131). The lncRNA np_5318 promotes renal I/R injuries through the TGF-β/Smad signaling pathway (132). The lncRNA XLOC_032768 protects renal tubular epithelial cells against apoptosis in renal I/R injuries by regulating FNDC3B/TGF-β1 signaling pathway (133). Additionally, a previous clinical study demonstrated that changes in the concentration of circulating lncRNAs in the plasma of patients with acute kidney injury were predictive of cohort mortality (134). This also reveals the practical application of lncRNAs in clinical ischemic injuries.

6. Role of lncRNAs in lung ischemic injury

Lung I/R is a common type of ischemic injury, as well as a common post-operative complication of lung transplantation with high morbidity and mortality rates. The elucidation of the mechanisms of pulmonary ischemic injury is of utmost significance for the clinical diagnosis and treatment of the condition. However, only a few studies to date have analyzed the role of lncRNAs in pulmonary ischemic injury, at least to the best of our knowledge.

In line with its effects on the brain, heart and kidneys, it is also assumed that MALAT1 plays a crucial role in ischemic lung injuries. In 2019, Wei et al (12) explored the role of MALAT1 in inflammatory injuries following lung transplantation I/R (LTIR). The expression of MALAT1 was significantly increased in the lung tissues of rats with LTIR. The expression of IL-8 was the highest among all examined inflammatory factors and was significantly higher in the serum and in bronchoalveolar lavage fluid of rats with LTIR compared with the control animals. The knockdown of MALAT1 significantly reduced the expression of IL-8 (mRNA and protein) and suppressed the apoptosis of pulmonary epithelial cells. In fact, MALAT1 was directly bound to p300 and activated Il-8 transcription. The silencing of MALAT1 alleviated inflammatory injury in rats with LTIR by suppressing IL-8 expression, and inhibiting neutrophil activation and infiltration (8).

Primary graft dysfunction (PGD) is an acute lung injury caused by I/R injury, which is the main cause of early morbidity and mortality following transplantation (135). A previous study found that lncRNA X-inactive-specific transcript (XIST) upregulated IL-12A expression by binding to miR-21, thus inducing the formation of neutrophil extracellular traps (NETs) and accelerated PGD following lung transplantation (136). In that study, miR-21 expression was significantly reduced in patients with PGD following lung transplantation compared with non-PGD patients, while its expression level was inversely associated with the PGD grade. Moreover, the level of the pro-inflammatory cytokines, IL-6, CXCL10, CCL2, and the chemokine, IL-8, in patients with PGD was higher than that in patients without PGD. An elevated expression of XIST was also detected in the alveolar lavage fluid from patients with PGD (136). During acute immune responses, neutrophils migrate to the site of inflammation and decompose their nuclear inclusions to form NETs, which are a possible risk factor for PGD. XIST was found to negatively correlate with miR-21, but to positively correlate with IL-12A. Silencing XIST also downregulated the expression of IL-12A by upregulating miR-21, inhibiting the formation of NETs, and ultimately reducing PGD following lung transplantation (136).

7. Overview of lncRNAs in ischemic injuries

A large number of in vitro and in vivo experiments have demonstrated that multiple lncRNAs are involved in the ischemic injury to different organs, among which MALAT1, H19, MEG3 and TUG1 are the most commonly described lncRNAs. Some lncRNAs exhibit similar expression patterns in ischemic cell and animal models and in the plasma of ischemic patients (Table SI), while the knockdown/overexpression of lncRNAs in animal models recapitulates the pathology of ischemic patients, suggesting that lncRNAs may contribute to the formation and progression of ischemic injury in patients. However, the mechanisms and associated signaling pathways of the majority of lncRNAs in ischemic injuries remain unclear and the effect of some lncRNAs remains controversial (126,127).

Based on review of the entire literature, it can be concluded that MALAT1 plays a critical role not only in cerebral ischemic injury, but also in the ischemic injury of the heart, kidneys and lungs. Why is the lncRNA MALAT1 of such vital importance to ischemic injury? MALAT1 is a highly conserved nuclear retained lncRNA and plays a role in the transcriptional and post-transcriptional regulation of gene expression in an environment-dependent manner (137,138). Furthermore, MALAT1 is involved in a variety of physiological processes, including alternative splicing, epigenetic modification of gene expression, synapse formation and myogenesis (138). In recent years, it has been found that an abnormal MALAT1 expression is closely related to cancer. MALAT1 regulates cancer progression by interacting with molecules, such as proteins, RNA and DNA to alter different signaling pathways (23). In addition to lung cancer, recent studies have shown that MALAT1 is involved in other types of cancer, such as breast, pancreatic, prostate cancer, glioma and leukemia (139). MALAT1 is therefore considered as a potential biomarker for the diagnosis and prediction of cancer, as well as a therapeutic target for specific tumors.

The present review article demonstrates that MALAT1 plays a key role in ischemic injury, and is involved in the heart, brain, lungs and kidneys (Fig. 1). Similarly, studies have shown that in most cases, MALAT1 expression or overexpression can aggravate ischemic injury in these organs. Moreover, MALAT1 is involved in the regulation of ischemic injury mainly through the miRNA-mediated regulation of inflammatory factors, apoptosis, autophagy and other pathways. MALAT1 promotes the development of stroke by regulating MDM2 and subsequently activating the p53 signaling pathway. Further study of the MALAT1/MDM2/p53 signaling pathway may provide a more effective clinical treatment strategy for patients with ischemic stroke (Fig. 1). Accordingly, MALAT1 may be a novel target for the treatment of cerebral ischemic stroke. In heart ischemic injury, MALAT1 can function as a ceRNA for miR-20b-5p to upregulate expression of Beclin1 and enhance autophagy-mediated cardiomyocyte injury (109). MALAT1 can also sponge miR-133 and miRNA-320 to enhance inflammation in ischemia-reperfusion injury and apoptosis in acute myocardial infarction (110,111). The Wnt/β-catenin signal pathway has been shown to exert a protective effect in cerebral, hepatic and myocardial I/R injury (140-142). The knockdown of MALAT1 attenuates I-R induced myocardial infarction and downregulates β-catenin expression (112). The mechanisms underlying the regulation of β-catenin expression by MALAT1 require further investigation. MALTA1 also exacerbates I/R injury following lung transplantation by upregulating IL-8 expression (12). However, MALAT1 has no effect or, in fact, the opposite effect on renal I/R injury (111,127). The underlying mechanisms of the functional difference of MALAT1 between organs warrant further investigation.

The function of lncRNAs in ischemic injury appears to be complex. An single lncRNA may have multiple miRNA targets and is associated with different signaling pathways. For example, in brain ischemic injury, lncRNA TUG1 can sponge miR-9, miR-29b-1-5p, miR-145, miR-410, miR-410-3p and miR-493-3p, can regulate expression of downstream targeting genes, markedly induce apoptosis and inflammation, and aggravate brain ischemic injury (Fig. 2) (44,84,143-145); in heart ischemic injury, lncRNA TUG1 can bind to miR-9, miR-132-3p, miR-142-3p and miR-340, subsequently regulating the expression of targeting genes, enhancing oxidative damage, autophagy and apoptosis, and promoting heart ischemic injury (Fig. 2) (104-106,146); in kidney ischemic injury, the reported effects of lncRNA TUG1 on ischemic injury are not consistent. A previous study demonstrated that lncRNA TUG1 sponged miR-494-3p and disinhibited E-cadherin, reducing I/R induced acute kidney injury (147). However, another study reported that lncRNA TUG1 interacted with miR-449b-5p and induced apoptosis and inflammation, aggravating I/R induced kidney injury (Fig. 2) (130). It is interesting that lncRNA TUG1 sponges the same miRNA (miR-9), but that the downstream target is different (FOXO3 in the brain and RLF5 in the heart) (Fig. 2). The complex multiple targets and associated signaling pathways of lncRNA renders the development of treatments more challenging.

Figure 2

Individual lncRNAs have multiple miRNA targets and involve different signaling pathways. In brain ischemic injury, for example, lncRNA TUG1 can bind miR-9, upregulate FOXO3 expression and increase neuronal apoptosis in mice with middle cerebral artery occlusion (143); lncRNA TUG1 binds miR-29b-1-5p, activates the NF-κB/IL-β signaling pathway, and induces inflammatory damage in rats with spinal cord I/R injury (144); lncRNA TUG1 can directly interact with miR-145 and function as a competing endogenous RNA of miR-145, regulate AQP4 expression and induce cell damage in cerebral I/R injury (84); lncRNA TUG1 binds miR-410, regulates FOXO3 expression and induces apoptosis and inflammation in cerebral I/R injury (145); lncRNA TUG1 binds miR-410-3p and miR-493-3p, activates the JNK and p38 MAPK signaling pathways, and induces inflammation and oxidative damage in cerebral I/R injury (44). In heart ischemic injury, lncRNA TUG1 binds miR-9, regulates KLF5 expression and induces apoptosis in myocardial I/R injury (106); lncRNA TUG1 activates HDAC3 by sponging miR-132-3p, stimulates intracellular ROS accumulation and aggravates myocardial ischemic injury (104); lncRNA TUG1 binds miR-142-3p, regulates expression of HMGB1 and Rac1, and induces the apoptosis and autophagy of ischemic/hypoxia cardiomyocytes (104); lncRNA TUG1 binds miR-340, regulates HDAC4 expression, mediates β-catenin/GLUT1 and induces apoptosis in myocardial I/R injury (146). In kidney ischemic injury, lncRNA TUG1 interacts with miR-449b-5p, and then regulates expression of HMGB1 and MMP2, inducing apoptosis and inflammation in I/R injury (130). However, lncRNA TUG1 binds miR-494-3p, regulates E-cadherin expression and inhibits apoptosis, alleviating I/R induced acute kidney injury (147). lncRNA, long non-coding RNA; TUG1, taurine up-regulated gene 1; I/R, ischemia/reperfusion; KLF5, Kruppel like factor 5; HMGB1, high mobility group box 1; HDAC3, histone deacetylase 3.

8. Conclusion and future perspectives

Ischemic injuries cause severe trauma to the body and are associated with high morbidity and mortality rates. Ischemic stroke, myocardial infarction and renal transplantation failures have attracted extensive clinical attention. Numerous studies (cited in the present review) have demonstrated that lncRNAs play an essential role in ischemic injuries and that their interaction with miRNA is the main mode of action. The downregulation of the majority of lncRNAs reduces inflammation, apoptosis, autophagy, or other cell damage, thus alleviating ischemic injuries. Since the plethora of data are derived from in vitro and pre-clinical studies, further investigations are urgently required to develop novel therapeutic strategies for patients with ischemic injury.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

LS and BY contributed to the conception of the study. YC, JL, QL, KH and NJ analyzed the data from the literature for inclusion in the review and prepared the manuscript. JR and XS revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by grants from the Outstanding Youth Project of the Hunan Provincial Department of Education (no. 19B508), the Jiangxi Provincial Natural Science Foundation (no. 20202BABL206061), the Cultivation Scientific Research Fund for the Junior Teachers of Medicine in NanChang University (PY201826), and the Lotus Scholarship Program of Hunan Province (2019-23).

References

1 

Akbari G: Role of zinc supplementation on ischemia/reperfusion injury in various organs. Biol Trace Elem Res. 196:1–9. 2020. View Article : Google Scholar

2 

Akella A, Bhattarai S and Dharap A: Long noncoding RNAs in the pathophysiology of ischemic stroke. Neuromolecular Med. 21:474–483. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Haddad G, Kölling M, Wegmann UA, Dettling A, Seeger H, Schmitt R, Soerensen-Zender I, Haller H, Kistler AD, Dueck A, et al: Renal AAV2-mediated overexpression of long non-coding RNA H19 attenuates ischemic acute kidney injury through sponging of microRNA-30a-5p. J Am Soc Nephrol. 32:323–341. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Frangogiannis NG: Pathophysiology of myocardial infarction. Compr Physiol. 5:1841–1875. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Zhang L and Wang H: Long non-coding RNA in CNS injuries: A new target for therapeutic intervention. Mol Ther Nucleic Acids. 17:754–766. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Kumar MM and Goyal R: LncRNA as a therapeutic target for angiogenesis. Curr Top Med Chem. 17:1750–1757. 2017. View Article : Google Scholar :

7 

Das A, Samidurai A and Salloum FN: Deciphering non-coding RNAs in cardiovascular health and disease. Front Cardiovasc Med. 5:732018. View Article : Google Scholar : PubMed/NCBI

8 

Zhou J, Chen H and Fan Y: Systematic analysis of the expression profile of non-coding RNAs involved in ischemia/reperfusion-induced acute kidney injury in mice using RNA sequencing. Oncotarget. 8:100196–100215. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Li J, Hao M, Yang B, Shi T, Zhang Y, Feng J and Chen J: Long non-coding RNAs expression profile and functional analysis of acute ischemic stroke. Medicine (Baltimore). 99:e229642020. View Article : Google Scholar

10 

Wang Y, Pan WY, Ge JS, Wang XD, Chen W, Luo X and Wang YL: A review of the relationship between long noncoding RNA and post-stroke injury repair. J Int Med Res. 47:4619–4624. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Su M, Hu X, Lin J, Zhang L, Sun W, Zhang J, Tian Y and Qiu W: Identification of candidate genes involved in renal ischemia/reperfusion injury. DNA Cell Biol. 38:256–262. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Wei L, Li J, Han Z, Chen Z and Zhang Q: Silencing of lncRNA MALAT1 prevents inflammatory injury after lung transplant ischemia-reperfusion by downregulation of IL-8 via p300. Mol Ther Nucleic Acids. 18:285–297. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Ali T and Grote P: Beyond the RNA-dependent function of LncRNA genes. Elife. 9:e605832020. View Article : Google Scholar : PubMed/NCBI

14 

Uszczynska-Ratajczak B, Lagarde J, Frankish A, Guigó R and Johnson R: Towards a complete map of the human long non-coding RNA transcriptome. Nat Rev Genet. 19:535–548. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Fang S, Zhang L, Guo J, Niu Y, Wu Y, Li H, Zhao L, Li X, Teng X, Sun X, et al: NONCODEV5: A comprehensive annotation database for long non-coding RNAs. Nucleic Acids Res. 46(D1): D308–D314. 2018. View Article : Google Scholar :

16 

Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar

17 

Bridges MC, Daulagala AC and Kourtidis A: LNCcation: lncRNA localization and function. J Cell Biol. 220:e2020090452021. View Article : Google Scholar : PubMed/NCBI

18 

Statello L, Guo CJ, Chen LL and Huarte M: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 22:96–118. 2021. View Article : Google Scholar

19 

Charles Richard JL and Eichhorn PJA: Platforms for investigating LncRNA functions. SLAS Technol. 23:493–506. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Wang SW, Liu Z and Shi ZS: Non-coding RNA in acute ischemic stroke: Mechanisms, biomarkers and therapeutic targets. Cell Transplant. 27:1763–1777. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Yang J, Chen M, Cao RY, Li Q and Zhu F: The role of circular RNAs in cerebral ischemic diseases: Ischemic stroke and cerebral ischemia/reperfusion injury. Adv Exp Med Biol. 1087:309–325. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Li ZX, Zhu QN, Zhang HB, Hu Y, Wang G and Zhu YS: MALAT1: A potential biomarker in cancer. Cancer Manag Res. 10:6757–6768. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Liu S, Yan G, Zhang J and Yu L: Knockdown of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) inhibits proliferation, migration, and invasion and promotes apoptosis by targeting miR-124 in retinoblastoma. Oncol Res. 26:581–591. 2018. View Article : Google Scholar

24 

Lelli A, Nolan KA, Santambrogio S, Gonçalves AF, Schönenberger MJ, Guinot A, Frew IJ, Marti HH, Hoogewijs D and Wenger RH: Induction of long noncoding RNA MALAT1 in hypoxic mice. Hypoxia (Auckl). 3:45–52. 2015.

25 

Zhang X, Tang X, Liu K, Hamblin MH and Yin KJ: Long noncoding RNA Malat1 regulates cerebrovascular pathologies in ischemic stroke. J Neurosci. 37:1797–1806. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Zhang T, Wang H, Li Q, Fu J, Huang J and Zhao Y: MALAT1 activates the P53 signaling pathway by regulating MDM2 to promote ischemic stroke. Cell Physiol Biochem. 50:2216–2228. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Guo D, Ma J, Yan L, Li T, Li Z, Han X and Shui S: Down-regulation of lncrna MALAT1 attenuates neuronal cell death through suppressing beclin1-dependent autophagy by regulating Mir-30a in cerebral ischemic stroke. Cell Physiol Biochem. 43:182–194. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Wang H, Zheng X, Jin J, Zheng L, Guan T, Huo Y, Xie S, Wu Y and Chen W: LncRNA MALAT1 silencing protects against cerebral ischemia-reperfusion injury through miR-145 to regulate AQP4. J Biomed Sci. 27:402020. View Article : Google Scholar : PubMed/NCBI

29 

Cao DW, Liu MM, Duan R, Tao YF, Zhou JS, Fang WR, Zhu JR, Niu L and Sun JG: The lncRNA Malat1 functions as a ceRNA to contribute to berberine-mediated inhibition of HMGB1 by sponging miR-181c-5p in poststroke inflammation. Acta Pharmacol Sin. 41:22–33. 2020. View Article : Google Scholar :

30 

Zhang L, Yang H, Li WJ and Liu YH: LncRNA MALAT1 promotes OGD-induced apoptosis of brain microvascular endothelial cells by sponging miR-126 to repress PI3K/Akt signaling pathway. Neurochem Res. 45:2091–2099. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Ruan W, Li J, Xu Y, Wang Y, Zhao F, Yang X, Jiang H, Zhang L, Saavedra JM, Shi L and Pang T: MALAT1 Up-regulator polydatin protects brain microvascular integrity and ameliorates stroke through C/EBPβ/MALAT1/CREB/PGC-1α/PPARγ pathway. Cell Mol Neurobiol. 39:265–286. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Li Z, Li J and Tang N: Long noncoding RNA Malat1 is a potent autophagy inducer protecting brain microvascular endothelial cells against oxygen-glucose deprivation/reoxygenation-induced injury by sponging miR-26b and upregulating ULK2 expression. Neuroscience. 354:1–10. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Al-Rugeebah A, Alanazi M and Parine NR: MEG3: An oncogenic long non-coding RNA in different cancers. Pathol Oncol Res. 25:859–874. 2019. View Article : Google Scholar : PubMed/NCBI

34 

You D and You H: Repression of long non-coding RNA MEG3 restores nerve growth and alleviates neurological impairment after cerebral ischemia-reperfusion injury in a rat model. Biomed Pharmacother. 111:1447–1457. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Liang J, Wang Q, Li JQ, Guo T and Yu D: Long non-coding RNA MEG3 promotes cerebral ischemia-reperfusion injury through increasing pyroptosis by targeting miR-485/AIM2 axis. Exp Neurol. 325:1131392020. View Article : Google Scholar

36 

Han L, Dong Z, Liu N, Xie F and Wang N: Maternally expressed gene 3 (MEG3) enhances PC12 cell hypoxia injury by targeting MiR-147. Cell Physiol Biochem. 43:2457–2469. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Deng D and Liang H: Silencing MEG3 protects PC12 cells from hypoxic injury by targeting miR-21. Artif Cells Nanomed Biotechnol. 48:610–619. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Zhou XM, Liu J, Wang Y and Zhang MH: Silencing of long noncoding RNA MEG3 enhances cerebral protection of dexmedetomidine against hypoxic-ischemic brain damage in neonatal mice by binding to miR-129-5p. J Cell Biochem: Nov. 28:2018.Epub ahead of print.

39 

Zhong L, Liu P, Fan J and Luo Y: Long non-coding RNA H19: Physiological functions and involvements in central nervous system disorders. Neurochem Int. 148:1050722021. View Article : Google Scholar : PubMed/NCBI

40 

Zhu H, Wang L, Chen J, Shen H and Chen Z: Mechanisms underlying abnormal expression of lncRNA H19 in neonatal hypoxic-ischemic encephalopathy. Am J Perinatol. Oct 27–2020.Epub ahead of print.

41 

Fang H, Li HF, Pan Q, Yang M, Zhang FX, Wang RR, Wang QY and Zhang JP: Long noncoding RNA H19 overexpression protects against hypoxic-ischemic brain damage by inhibiting miR-107 and up-regulating vascular endothelial growth factor. Am J Pathol. 191:503–514. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Hu S, Zheng J, Du Z and Wu G: Knock down of lncRNA H19 promotes axon sprouting and functional recovery after cerebral ischemic stroke. Brain Res. 1732:1466812020. View Article : Google Scholar : PubMed/NCBI

43 

Xiao Z, Qiu Y, Lin Y, Medina R, Zhuang S, Rosenblum JS, Cui J, Li Z, Zhang X and Guo L: Blocking lncRNA H19-miR-19a-Id2 axis attenuates hypoxia/ischemia induced neuronal injury. Aging (Albany NY). 11:3585–3600. 2019. View Article : Google Scholar

44 

Du J, Li W and Wang B: Long non-coding RNA TUG1 aggravates cerebral ischemia and reperfusion injury by sponging miR-493-3p/miR-410-3p. Open Med (Wars). 16:919–930. 2021. View Article : Google Scholar

45 

Yin M, Chen WP, Yin XP, Tu JL, Hu N and Li ZY: LncRNA TUG1 demethylated by TET2 promotes NLRP3 expression, contributes to cerebral ischemia/reperfusion inflammatory injury. ASN Neuro. 13:175909142110032472021. View Article : Google Scholar : PubMed/NCBI

46 

Cai J, Shangguan S, Li G, Cai Y, Chen Y, Ma G, Miao Z, Liu L and Deng Y: Knockdown of lncRNA Gm11974 protect against cerebral ischemic reperfusion through miR-766-3p/NR3C2 axis. Artif Cells Nanomed Biotechnol. 47:3847–3853. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Gai HY, Wu C, Zhang Y and Wang D: Long non-coding RNA CHRF modulates the progression of cerebral ischemia/reperfusion injury via miR-126/SOX6 signaling pathway. Biochem Biophys Res Commun. 514:550–557. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Li EY, Zhao PJ, Jian J, Yin BQ, Sun ZY, Xu CX, Tang YC and Wu H: LncRNA MIAT overexpression reduced neuron apoptosis in a neonatal rat model of hypoxic-ischemic injury through miR-211/GDNF. Cell Cycle. 18:156–166. 2019. View Article : Google Scholar :

49 

Jing H, Liu L, Jia Y, Yao H and Ma F: Overexpression of the long non-coding RNA Oprm1 alleviates apoptosis from cerebral ischemia-reperfusion injury through the Oprm1/miR-155/GATA3 axis. Artif Cells Nanomed Biotechnol. 47:2431–2439. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Yao P, Li YL, Chen Y, Shen W, Wu KY and Xu WH: Overexpression of long non-coding RNA Rian attenuates cell apoptosis from cerebral ischemia-reperfusion injury via Rian/miR-144-3p/GATA3 signaling. Gene. 737:1444112020. View Article : Google Scholar : PubMed/NCBI

51 

Yu S, Yu M, He X, Wen L, Bu Z and Feng J: KCNQ1OT1 promotes autophagy by regulating miR-200a/FOXO3/ATG7 pathway in cerebral ischemic stroke. Aging Cell. 18:e129402019. View Article : Google Scholar : PubMed/NCBI

52 

Tan X, Guo W, Peng Z, Gu C, Xiang P, Tu Y, Fei H, Liu X, Lu Y, Li M, et al: LncRNA-Malat1 down-regulates miR-211-5p expression to promote neuronal damage from cerebral ischemia reperfusion injury. Biochem Pharmacol. 192:1146942021. View Article : Google Scholar : PubMed/NCBI

53 

Meng S, Wang B and Li W: LncRNA MALAT1 improves cerebral ischemia-reperfusion injury and cognitive dysfunction by regulating miR-142-3p/SIRT1 axis. Int J Neurosci. 1:192021.Epub ahead of print.

54 

Zhang G, Wang Q, Su D and Xie Y: Long non-coding RNAMALAT1 knockdown alleviates cerebral ischemia/reperfusion injury of rats through regulating the miR-375/PDE4D axis. Front Neurol. 11:5787652020. View Article : Google Scholar

55 

Jia Y, Yi L, Li Q, Liu T and Yang S: LncRNA MALAT1 aggravates oxygen-glucose deprivation/reoxygenation-induced neuronal endoplasmic reticulum stress and apoptosis via the miR-195a-5p/HMGA1 axis. Biol Res. 54:82021. View Article : Google Scholar : PubMed/NCBI

56 

Hu Y, Ye C, Cheng S and Chen J: Propofol downregulates lncRNA MALAT1 to alleviate cerebral ischemia-reperfusion injury. Inflammation. 44:2580–2591. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Shi YL, Wang Q and Wei JC: Influence of lncRNA-MALAT1 on neuronal apoptosis in rats with cerebral infarction through regulating the ERK/MAPK signaling pathway. Eur Rev Med Pharmacol Sci. 23:8039–8048. 2019.PubMed/NCBI

58 

Wang LQ and Zhou HJ: LncRNA MALAT1 promotes high glucose-induced inflammatory response of microglial cells via provoking MyD88/IRAK1/TRAF6 signaling. Sci Rep. 8:83462018. View Article : Google Scholar : PubMed/NCBI

59 

Jin J, Wang H, Zheng X, Xie S, Zheng L and Zhan R: Inhibition of LncRNA MALAT1 attenuates cerebral ischemic reperfusion injury via regulating AQP4 expression. Eur Neurol. 83:581–590. 2020. View Article : Google Scholar : PubMed/NCBI

60 

Yang L, Wang L, Wang J and Liu P: Long non-coding RNA Gm11974 aggravates oxygen-glucose deprivation-induced injury via miR-122-5p/SEMA3A axis in ischaemic stroke. Metab Brain Dis. 36:2059–2069. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Luo HC, Yi TZ, Huang FG, Wei Y, Luo XP and Luo QS: Role of long noncoding RNA MEG3/miR-378/GRB2 axis in neuronal autophagy and neurological functional impairment in ischemic stroke. J Biol Chem. 295:14125–14139. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Li TH, Sun HW, Song LJ, Yang B, Zhang P, Yan DM, Liu XZ and Luo YR: Long non-coding RNA MEG3 regulates autophagy after cerebral ischemia/reperfusion injury. Neural Regen Res. 17:824–831. 2022. View Article : Google Scholar

63 

Zhang F, Wang Z, Sun B, Huang Y, Chen C, Hu J, Li L, Xia P and Ye Z: Propofol rescued astrocytes from LPS-induced inflammatory response via blocking LncRNA-MEG3/NF-κB axis. Curr Neurovasc Res. Mar 16–2022.Epub ahead of print. View Article : Google Scholar

64 

Chen C, Huang Y, Xia P, Zhang F, Li L, Wang E, Guo Q and Ye Z: Long noncoding RNA Meg3 mediates ferroptosis induced by oxygen and glucose deprivation combined with hyperglycemia in rat brain microvascular endothelial cells, through modulating the p53/GPX4 axis. Eur J Histochem. 65:32242021. View Article : Google Scholar :

65 

Li H, Tang C and Wang D: LncRNA H19 promotes inflammatory response induced by cerebral ischemia-reperfusion injury through regulating the miR-138-5p-p65 axis. Biochem Cell Biol. 98:525–536. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Xu J, Wang C, Meng F and Xu P: Long non-coding RNA H19 inhibition ameliorates oxygen-glucose deprivation-induced cell apoptosis and inflammatory cytokine expression by regulating the microRNA-29b/SIRT1/PGC-1α axis. Mol Med Rep. 23:1312021. View Article : Google Scholar

67 

Huang Y, Deng L, Zeng L, Bao S, Ye K, Li C, Hou X, Yao Y, Li D and Xiong Z: Silencing of H19 alleviates oxygen-glucose deprivation/reoxygenation-triggered injury through the regulation of the miR-1306-5p/BCL2L13 axis. Metab Brain Dis. 36:2461–2472. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Gao N, Tang H, Gao L, Tu GL, Luo H and Xia Y: LncRNA H19 aggravates cerebral ischemia/reperfusion injury by functioning as a ceRNA for miR-19a-3p to target PTEN. Neuroscience. 437:117–129. 2020. View Article : Google Scholar : PubMed/NCBI

69 

Wang J, Cao B, Han D, Sun M and Feng J: Long non-coding RNA H19 induces cerebral ischemia reperfusion injury via activation of autophagy. Aging Dis. 8:71–84. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Chen H and Li X: LncRNA ROR is involved in cerebral hypoxia/reoxygenation-induced injury in PC12 cells via regulating miR-135a-5p/ROCK1/2. Am J Transl Res. 11:6145–6158. 2019.PubMed/NCBI

71 

Zhou Q, An Y and Tang Y: Long noncoding RNA-regulator of reprogramming alleviates hypoxia-induced cerebral injury in mice model and human via modulating apoptosis complexes. J Integr Neurosci. 18:431–437. 2019. View Article : Google Scholar

72 

Yin WL, Yin WG, Huang BS and Wu LX: LncRNA SNHG12 inhibits miR-199a to upregulate SIRT1 to attenuate cerebral ischemia/reperfusion injury through activating AMPK signaling pathway. Neurosci Lett. 690:188–195. 2019. View Article : Google Scholar

73 

Deng Y, Chen D, Wang L, Gao F, Jin B, Lv H, Zhang G, Sun X, Liu L, Mo D, et al: Silencing of long noncoding RNA nespas aggravates microglial cell death and neuroinflammation in ischemic stroke. Stroke. 50:1850–1858. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Yao X, Yao R, Huang F and Yi J: LncRNA SNHG12 as a potent autophagy inducer exerts neuroprotective effects against cerebral ischemia/reperfusion injury. Biochem Biophys Res Commun. 514:490–496. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Wu Y, Huang Y, Cai J, Zhang D, Liu S and Pang B: LncRNA SNHG12 improves cerebral ischemic-reperfusion injury by activating SIRT1/FOXO3a pathway through I nhibition of autophagy and oxidative stress. Curr Neurovasc Res. 17:394–401. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Zhong Y, Yu C and Qin W: LncRNA SNHG14 promotes inflammatory response induced by cerebral ischemia/reperfusion injury through regulating miR-136-5p/ROCK1. Cancer Gene Ther. 26:234–247. 2019. View Article : Google Scholar

77 

Deng Z, Ou H, Ren F, Guan Y, Huan Y, Cai H and Sun B: LncRNA SNHG14 promotes OGD/R-induced neuron injury by inducing excessive mitophagy via miR-182-5p/BINP3 axis in HT22 mouse hippocampal neuronal cells. Biol Res. 53:382020. View Article : Google Scholar : PubMed/NCBI

78 

Bu X, Zhao Y, Chang M and Ge X: Downregulation of lncRNA SNHG14 alleviates neurons injury by modulating the miR-181c-5p/BMF axis in ischemic stroke. Brain Res Bull. 174:379–388. 2021. View Article : Google Scholar : PubMed/NCBI

79 

Zhang G, Li T, Chang X and Xing J: Long noncoding RNA SNHG14-promotes ischemic brain injury via regulating miR-199b/AQP4 axis. Neurochem Res. 46:1280–1290. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Deng W, Fan C, Shen R, Wu Y, Du R and Teng J: Long noncoding MIAT acting as a ceRNA to sponge microRNA-204-5p to participate in cerebral microvascular endothelial cell injury after cerebral ischemia through regulating HMGB1. J Cell Physiol. 235:4571–4586. 2020. View Article : Google Scholar

81 

Guo X, Wang Y, Zheng D, Cheng X and Sun Y: LncRNA-MIAT promotes neural cell autophagy and apoptosis in ischemic stroke by up-regulating REDD1. Brain Res. 1763:1474362021. View Article : Google Scholar : PubMed/NCBI

82 

Wang H, Liao S, Li H, Chen Y and Yu J: Long non-coding RNA TUG1 sponges Mir-145a-5p to regulate microglial polarization after oxygen-glucose deprivation. Front Mol Neurosci. 12:2152019. View Article : Google Scholar : PubMed/NCBI

83 

Xiang P, Hu J, Wang H, Luo Y, Gu C, Tan X, Tu Y, Guo W, Chen L, Gao L, et al: miR-204-5p is sponged by TUG1 to aggravate neuron damage induced by focal cerebral ischemia and reperfusion injury through upregulating COX2. Cell Death Discov. 8:892022. View Article : Google Scholar : PubMed/NCBI

84 

Shan W, Chen W, Zhao X, Pei A, Chen M, Yu Y, Zheng Y and Zhu S: Long noncoding RNA TUG1 contributes to cerebral ischaemia/reperfusion injury by sponging mir-145 to up-regulate AQP4 expression. J Cell Mol Med. 24:250–259. 2020. View Article : Google Scholar

85 

Li L, Zhang Q, Wang Y, Yin S, Chi S, Han F and Wang W: Knockdown of lncRNA TUG1 attenuates cerebral ischemia/reperfusion injury through regulating miR-142-3p. Biofactors. 47:819–827. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Qiao P, Yan H and Wang J: EGb761 protects brain microvascular endothelial cells against oxygen-glucose deprivation-induced injury through lncRNA Rmst/miR-150 axis. Neurochem Res. 45:2398–2408. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Hou XX and Cheng H: Long non-coding RNA RMST silencing protects against middle cerebral artery occlusion (MCAO)-induced ischemic stroke. Biochem Biophys Res Commun. 495:2602–2608. 2018. View Article : Google Scholar

88 

Wu Z, Wu P, Zuo X, Yu N, Qin Y, Xu Q, He S, Cen B, Liao W and Ji A: LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation. Mol Neurobiol. 54:7670–7685. 2017. View Article : Google Scholar

89 

Lejay A, Fang F, John R, Van JA, Barr M, Thaveau F, Chakfe N, Geny B and Scholey JW: Ischemia reperfusion injury, ischemic conditioning and diabetes mellitus. J Mol Cell Cardiol. 91:11–22. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Yu SY, Dong B, Fang ZF, Hu XQ, Tang L and Zhou SH: Knockdown of lncRNA AK139328 alleviates myocardial ischaemia/reperfusion injury in diabetic mice via modulating miR-204-3p and inhibiting autophagy. J Cell Mol Med. 22:4886–4898. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Cai B, Ma W, Ding F, Zhang L, Huang Q, Wang X, Hua B, Xu J, Li J, Bi C, et al: The long noncoding RNA CAREL controls cardiac regeneration. J Am Coll Cardiol. 72:534–550. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Zhao J, Chen F, Ma W and Zhang P: Suppression of long noncoding RNA NEAT1 attenuates hypoxia-induced cardiomyocytes injury by targeting miR-378a-3p. Gene. 731:1443242020. View Article : Google Scholar : PubMed/NCBI

93 

Rui PF, Wang JH and Xu J: Long non-coding NEAT1 weakens the protective role of sevoflurane on myocardial ischemia/reperfusion injury by mediating the microRNA-140/RhoA axis. J Biol Regul Homeost Agents. 35:933–944. 2021.PubMed/NCBI

94 

Wei Q, Zhou HY, Shi XD, Cao HY and Qin L: Long noncoding RNA NEAT1 promotes myocardiocyte apoptosis and suppresses proliferation through regulation of miR-129-5p. J Cardiovasc Pharmacol. 74:535–541. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Ruan Z, Wang S, Yu W and Deng F: LncRNA NEAT1 aggravates diabetic myocardial ischemia-reperfusion injury through regulating PINK1 by targeting miR-27b. Int J Cardiol. 286:1362019. View Article : Google Scholar : PubMed/NCBI

96 

Wang SM, Liu GQ, Xian HB, Si JL, Qi SX and Yu YP: LncRNA NEAT1 alleviates sepsis-induced myocardial injury by regulating the TLR2/NF-κB signaling pathway. Eur Rev Med Pharmacol Sci. 23:4898–4907. 2019.PubMed/NCBI

97 

Gidlöf O, Bader K, Celik S, Grossi M, Nakagawa S, Hirose T, Metzler B, Olde B and Erlinge D: Inhibition of the long non-coding RNA NEAT1-protects cardiomyocytes from hypoxia in vitro via decreased pri-miRNA processing. Cell Death Dis. 11:6772020. View Article : Google Scholar

98 

Zhang BF, Chen J and Jiang H: LncRNA H19 ameliorates myocardial ischemia-reperfusion injury by targeting miR-22-3P. Int J Cardiol. 278:2242019. View Article : Google Scholar : PubMed/NCBI

99 

Zhang BF, Jiang H, Chen J, Hu Q, Yang S, Liu XP and Liu G: LncRNA H19 ameliorates myocardial infarction-induced myocardial injury and maladaptive cardiac remodelling by regulating KDM3A. J Cell Mol Med. 24:1099–1115. 2020. View Article : Google Scholar

100 

Luo H, Wang J, Liu D, Zang S, Ma N, Zhao L, Zhang L, Zhang X and Qiao C: The lncRNA H19/miR-675 axis regulates myocardial ischemic and reperfusion injury by targeting PPARα. Mol Immunol. 105:46–54. 2019. View Article : Google Scholar

101 

Zhang X, Cheng L, Xu L, Zhang Y, Yang Y, Fu Q, Mi W and Li H: The lncRNA, H19 mediates the protective effect of hypoxia postconditioning against hypoxia-reoxygenation injury to senescent cardiomyocytes by targeting microRNA-29b-3p. Shock. 52:249–256. 2019. View Article : Google Scholar

102 

Choong OK, Chen CY, Zhang J, Lin JH, Lin PJ, Ruan SC, Kamp TJ and Hsieh PCH: Hypoxia-induced H19/YB-1 cascade modulates cardiac remodeling after infarction. Theranostics. 9:6550–6567. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Chen C, Liu M, Tang Y, Sun H, Lin X, Liang P and Jiang B: LncRNA H19 is involved in myocardial ischemic preconditioning via increasing the stability of nucleolin protein. J Cell Physiol. 235:5985–5994. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Su Q, Liu Y, Lv XW, Dai RX, Yang XH and Kong BH: LncRNA TUG1 mediates ischemic myocardial injury by targeting miR-132-3p/HDAC3 axis. Am J Physiol Heart Circ Physiol. 318:H332–H344. 2020. View Article : Google Scholar

105 

Su Q, Liu Y, Lv XW, Ye ZL, Sun YH, Kong BH and Qin ZB: Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J Mol Cell Cardiol. 133:12–25. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Yang D, Yu J, Liu HB, Yan XQ, Hu J, Yu Y, Guo J, Yuan Y and Du ZM: The long non-coding RNA TUG1-miR-9a-5p axis contributes to ischemic injuries by promoting cardiomyocyte apoptosis via targeting KLF5. Cell Death Dis. 10:9082019. View Article : Google Scholar : PubMed/NCBI

107 

Song T, Wang P and Xin L: LncRNA TUG1 Contributes to hypoxia-induced myocardial cell injury through downregulating miR-29a-3p in AC16 cells. J Cardiovasc Pharmacol. 76:533–539. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Cai X, Wang S, Hong L, Yu S, Li B, Zeng H, Yang X, Zhang P and Shao L: Long noncoding RNA taurine-upregulated gene 1 knockdown protects cardiomyocytes against hypoxia/reoxygenation-induced injury through regulating miR-532-5p/Sox8 axis. J Cardiovasc Pharmacol. 76:556–563. 2020. View Article : Google Scholar : PubMed/NCBI

109 

Wang S, Yao T, Deng F, Yu W, Song Y, Chen J and Ruan Z: LncRNA MALAT1 promotes oxygen-glucose deprivation and reoxygenation induced cardiomyocytes injury through sponging miR-20b to enhance beclin1-mediated autophagy. Cardiovasc Drugs Ther. 33:675–686. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Yu SY, Dong B, Tang L and Zhou SH: LncRNA MALAT1 sponges miR-133 to promote NLRP3 inflammasome expression in ischemia-reperfusion injured heart. Int J Cardiol. 254:502018. View Article : Google Scholar : PubMed/NCBI

111 

Tian H, Wu M, Zhou P, Huang C, Ye C and Wang L: The long non-coding RNA MALAT1 is increased in renal ischemia-reperfusion injury and inhibits hypoxia-induced inflammation. Ren Fail. 40:527–533. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Xu XZ, Luo B, Xiao Y and Zheng WQ: Effects of lncRNA MALAT1-mediated β-catenin signaling pathway on myocardial cell apoptosis in rats with myocardial ischemia/reperfusion injury. Eur Rev Med Pharmacol Sci. 23:9557–9565. 2019.PubMed/NCBI

113 

Shu L, Zhang W, Huang C, Huang G, Su G and Xu J: lncRNA ANRIL protects H9c2 cells against hypoxia-induced injury through targeting the miR-7-5p/SIRT1 axis. J Cell Physiol. 235:1175–1183. 2020. View Article : Google Scholar

114 

Li L, Zhang M, Chen W, Wang R, Ye Z, Wang Y, Li X and Cai C: LncRNA-HOTAIR inhibition aggravates oxidative stress-induced H9c2 cells injury through suppression of MMP2 by miR-125. Acta Biochim Biophys Sin (Shanghai). 50:996–1006. 2018. View Article : Google Scholar

115 

Du J, Yang ST, Liu J, Zhang KX and Leng JY: Silence of LncRNA GAS5 protects cardiomyocytes H9c2 against hypoxic injury via sponging miR-142-5p. Mol Cells. 42:397–405. 2019.PubMed/NCBI

116 

Liu CY, Zhang YH, Li RB, Zhou LY, An T, Zhang RC, Zhai M, Huang Y, Yan KW, Dong YH, et al: LncRNA CAIF inhibits autophagy and attenuates myocardial infarction by blocking p53-mediated myocardin transcription. Nat Commun. 9:292018. View Article : Google Scholar : PubMed/NCBI

117 

Li Z, Zhang Y, Ding N, Zhao Y, Ye Z, Shen L, Yi H and Zhu Y: Inhibition of lncRNA XIST improves myocardial I/R injury by targeting miR-133a through inhibition of autophagy and regulation of SOCS2. Mol Ther Nucleic Acids. 18:764–773. 2019. View Article : Google Scholar : PubMed/NCBI

118 

Li Y, Li J, Zhang P, Jiang X, Pan Z, Zheng W and Lin H: LncRNA-LET relieves hypoxia-induced injury in H9c2 cells through regulation of miR-138. J Cell Biochem. 121:259–268. 2020. View Article : Google Scholar

119 

Li T, Tian H, Li J, Zuo A, Chen J, Xu D, Guo Y and Gao H: Overexpression of lncRNA Gm2691 attenuates apoptosis and inflammatory response after myocardial infarction through PI3K/Akt signaling pathway. IUBMB Life. 71:1561–1570. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Chen L, Zhang D, Yu L and Dong H: Targeting MIAT reduces apoptosis of cardiomyocytes after ischemia/reperfusion injury. Bioengineered. 10:121–132. 2019. View Article : Google Scholar : PubMed/NCBI

121 

Kong F, Jin J, Lv X, Han Y, Liang X, Gao Y and Duan X: RETRACTED: Long noncoding RNA RMRP upregulation aggravates myocardial ischemia-reperfusion injury by sponging miR-206 to target ATG3 expression. Biomed Pharmacother. 109:716–725. 2019. View Article : Google Scholar

122 

Ong SB, Katwadi K, Kwek XY, Ismail NI, Chinda K, Ong SG and Hausenloy DJ: Non-coding RNAs as therapeutic targets for preventing myocardial ischemia-reperfusion injury. Expert Opin Ther Targets. 22:247–261. 2018. View Article : Google Scholar : PubMed/NCBI

123 

Basile DP, Donohoe D, Roethe K and Osborn JL: Renal ischemic injury results in permanent damage to peritubular capillaries and influences long-term function. Am J Physiol Renal Physiol. 281:F887–F899. 2001. View Article : Google Scholar : PubMed/NCBI

124 

Tao Q, Tianyu W, Jiangqiao Z, Zhongbao C, Xiaoxiong M, Long Z and Jilin Z: Expression analysis of long non-coding RNAs in a renal ischemia-reperfusion injury model. Acta Cir Bras. 34:e2019004032019. View Article : Google Scholar : PubMed/NCBI

125 

Liu F, Yang Y, Liu T, Deng J, Zhang H, Luo D and Lou YL: Analysis of differentially expressed long noncoding RNA in renal ischemia-reperfusion injury. Kidney Blood Press Res. 45:686–701. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Kölling M, Genschel C, Kaucsar T, Hübner A, Rong S, Schmitt R, Sörensen-Zender I, Haddad G, Kistler A, Seeger H, et al: Hypoxia-induced long non-coding RNA Malat1 is dispensable for renal ischemia/reperfusion-injury. Sci Rep. 8:34382018. View Article : Google Scholar : PubMed/NCBI

127 

Puthanveetil P, Gutschner T and Lorenzen J: MALAT1: A therapeutic candidate for a broad spectrum of vascular and cardiorenal complications. Hypertens Res. 43:372–379. 2020. View Article : Google Scholar

128 

Jiang X, Li D, Shen W, Shen X and Liu Y: LncRNA NEAT1 promotes hypoxia-induced renal tubular epithelial apoptosis through downregulating miR-27a-3p. J Cell Biochem. 120:16273–16282. 2019. View Article : Google Scholar : PubMed/NCBI

129 

Geng X, Song N, Zhao S, Xu J, Liu Y, Fang Y, Liang M, Xu X and Ding X: LncRNA GAS5 promotes apoptosis as a competing endogenous RNA for miR-21 via thrombospondin 1 in ischemic AKI. Cell Death Discov. 6:192020. View Article : Google Scholar : PubMed/NCBI

130 

Xu Y, Niu Y, Li H and Pan G: Downregulation of lncRNA TUG1 attenuates inflammation and apoptosis of renal tubular epithelial cell induced by ischemia-reperfusion by sponging miR-449b-5p via targeting HMGB1 and MMP2. Inflammation. 43:1362–1374. 2020. View Article : Google Scholar : PubMed/NCBI

131 

Tian X, Ji Y, Liang Y, Zhang J, Guan L and Wang C: LINC00520 targeting miR-27b-3p regulates OSMR expression level to promote acute kidney injury development through the PI3K/AKT signaling pathway. J Cell Physiol. 234:14221–14233. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Lu J, Miao J and Sun J: LncRNA np_5318 promotes renal ischemia-reperfusion injury through the TGF-β/Smad signaling pathway. Exp Ther Med. 19:2833–2840. 2020.PubMed/NCBI

133 

Zhou X, Li Y, Wu C, Yu W and Cheng F: Novel lncRNA XLOC_032768 protects against renal tubular epithelial cells apoptosis in renal ischemia-reperfusion injury by regulating FNDC3B/TGF-β1. Ren Fail. 42:994–1003. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Lorenzen JM, Schauerte C, Kielstein JT, Hübner A, Martino F, Fiedler J, Gupta SK, Faulhaber-Walter R, Kumarswamy R, Hafer C, et al: Circulating long noncoding RNATapSaki is a predictor of mortality in critically ill patients with acute kidney injury. Clin Chem. 61:191–201. 2015. View Article : Google Scholar

135 

Shah RJ and Diamond JM: Primary graft dysfunction (PGD) following lung transplantation. Semin Respir Crit Care Med. 39:148–154. 2018. View Article : Google Scholar : PubMed/NCBI

136 

Li J, Wei L, Han Z, Chen Z and Zhang Q: Long non-coding RNA X-inactive specific transcript silencing ameliorates primary graft dysfunction following lung transplantation through microRNA-21-dependent mechanism. EBioMedicine. 52:1026002020. View Article : Google Scholar : PubMed/NCBI

137 

Arun G, Aggarwal D and Spector DL: MALAT1 long non-coding RNA: Functional implications. Noncoding RNA. 6:222020.

138 

Zhang X, Hamblin MH and Yin KJ: The long noncoding RNA Malat1: Its physiological and pathophysiological functions. RNA Biol. 14:1705–1714. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Sun Y and Ma L: New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers (Basel). 11:2162019. View Article : Google Scholar

140 

Li P, Zhang Y and Liu H: The role of Wnt/β-catenin pathway in the protection process by dexmedetomidine against cerebral ischemia/reperfusion injury in rats. Life Sci. 236:1169212019. View Article : Google Scholar

141 

Lehwald N, Tao GZ, Jang KY, Sorkin M, Knoefel WT and Sylvester KG: Wnt-β-catenin signaling protects against hepatic ischemia and reperfusion injury in mice. Gastroenterology. 141:707–718. 718.e1–e5. 2011. View Article : Google Scholar

142 

Ban Q, Qiao L, Xia H, Xie B, Liu J, Ma Y, Zhang L, Zhang M, Liu LG, Jiao W, et al: β-catenin regulates myocardial ischemia/reperfusion injury following heterotopic heart transplantation in mice by modulating PTEN pathways. Am J Transl Res. 12:4757–4771. 2020.

143 

Xiong ZJ, Zhang Q, Wang DX and Hu L: Overexpression of TUG1 promotes neuronal death after cerebral infarction by regulating microRNA-9. Eur Rev Med Pharmacol Sci. 22:7393–7400. 2018.PubMed/NCBI

144 

Jia H, Li Z, Chang Y, Fang B, Zhou Y and Ma H: Downregulation of long noncoding RNA TUG1 attenuates MTDH-mediated inflammatory damage via targeting miR-29b1-5p after spinal cord ischemia reperfusion. J Neuropathol Exp Neurol. 80:254–264. 2021. View Article : Google Scholar

145 

He Z, Zhao Y, Zhu Y, Wang W, Liu X and Lu F: Interfering TUG1 attenuates cerebrovascular endothelial apoptosis and inflammatory injury after cerebral ischemia/reperfusion via TUG1/miR-410/FOXO3 ceRNA axis. Neurotox Res. 40:1–13. 2022. View Article : Google Scholar

146 

Wu X, Liu Y, Mo S, Wei W, Ye Z and Su Q: LncRNA TUG1 competitively binds to miR-340 to accelerate myocardial ischemia-reperfusion injury. FASEB J. 35:e211632021.

147 

Chen L, Xu JY and Tan HB: LncRNA TUG1 regulates the development of ischemia-reperfusion mediated acute kidney injury through miR-494-3p/E-cadherin axis. J Inflamm (Lond). 18:122021. View Article : Google Scholar

Related Articles

Journal Cover

July-2022
Volume 50 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cao Y, Liu J, Lu Q, Huang K, Yang B, Reilly J, Jiang N, Shu X and Shang L: An update on the functional roles of long non‑coding RNAs in ischemic injury (Review). Int J Mol Med 50: 91, 2022
APA
Cao, Y., Liu, J., Lu, Q., Huang, K., Yang, B., Reilly, J. ... Shang, L. (2022). An update on the functional roles of long non‑coding RNAs in ischemic injury (Review). International Journal of Molecular Medicine, 50, 91. https://doi.org/10.3892/ijmm.2022.5147
MLA
Cao, Y., Liu, J., Lu, Q., Huang, K., Yang, B., Reilly, J., Jiang, N., Shu, X., Shang, L."An update on the functional roles of long non‑coding RNAs in ischemic injury (Review)". International Journal of Molecular Medicine 50.1 (2022): 91.
Chicago
Cao, Y., Liu, J., Lu, Q., Huang, K., Yang, B., Reilly, J., Jiang, N., Shu, X., Shang, L."An update on the functional roles of long non‑coding RNAs in ischemic injury (Review)". International Journal of Molecular Medicine 50, no. 1 (2022): 91. https://doi.org/10.3892/ijmm.2022.5147