Open Access

Long non‑coding RNA‑HOTAIR promotes the progression of sepsis by acting as a sponge of miR‑211 to induce IL‑6R expression

  • Authors:
    • Jianan Chen
    • Xingsheng Gu
    • Li Zhou
    • Shuguang Wang
    • Limei Zhu
    • Yangneng Huang
    • Feng Cao
  • View Affiliations

  • Published online on: September 27, 2019     https://doi.org/10.3892/etm.2019.8063
  • Pages: 3959-3967
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Sepsis remains the primary cause of death in intensive care units and multiple long non‑coding RNAs (lncRNAs) have been demonstrated to be dysregulated in samples of patients with sepsis. However, whether lncRNA‑HOTAIR is involved in the etiology of sepsis remains unclear. The aim of the present study was to investigate the role of HOTAIR in sepsis and to reveal the associated mechanisms. A bioinformatics analysis and dual‑luciferase reporter assay was performed to evaluate the interaction between HOTAIR and miR‑211, as well as miR‑211 and IL‑6R. An animal model of sepsis was established in mice via cecal ligation and puncture. Interferon (IFN)‑γ, interleukin (IL)‑6, IL‑17, tumor necrosis factor (TNF)‑α, IL‑1β, IL‑6 receptor (R), microRNA (miR)‑211 and HOTAIR expression was measured using reverse transcription‑quantitative PCR. Cellular proliferation and apoptosis of monocytes were assessed using cell counting kit‑8 assay and flow cytometry, respectively. miR‑211 was revealed to be targeted by HOTAIR and IL‑6R. The expression of IFN‑γ, IL‑6, IL‑17, TNF‑α, IL‑1β, IL‑6R and HOTAIR was significantly upregulated in the septic mice, whereas miR‑211 expression was downregulated. The overexpression of hox transcript antisense RNA (HOTAIR) and knockdown of miR‑211 were associated with an increased expression of IFN‑γ, IL‑6, IL‑17, TNF‑α, IL‑1β and IL‑6R in monocytes, while the overexpression of miR‑211 exhibited the opposite effect. HOTAIR overexpression and miR‑211 knockdown significantly inhibited cellular proliferation and promoted monocyte apoptosis, whereas the overexpression of miR‑211 exhibited the opposite effects in monocytes. Therefore, HOTAIR may promote the progression of sepsis by indirectly regulating the expression of IL‑6R via miR‑211.

Introduction

Sepsis is a serious clinical condition caused by multiple agents, including bacterial, viral and fungal infections, which subsequently initiates the inflammatory response, leading to organ failure in the affected host (1). Currently, sepsis remains the primary cause of death in intensive care units (ICU), despite recent advancements in medical technology (2,3). It has been estimated that in 2017, the percentage of admissions to the ICU caused by sepsis is ~25% and is associated with a mortality rate >50% worldwide (2,4). Although the precise pathophysiology of sepsis remains unclear, increasing evidence has indicated that it may move from an early hyper-inflammatory phase characterized by systemic inflammation induced by the excessive release of pro-inflammatory factors, followed by a late immuno-suppressive phase, characterized by the apoptosis of immune cells (including monocytes and lymphocytes) (5,6). Furthermore, apoptosis that occurs in the cells of tissues in the site of primary infection during sepsis can result in microvascular dysfunction, which subsequently leads to organ failure (7,8). Therefore, inhibiting sepsis-induced apoptosis may be a potential therapeutic approach for patients with sepsis.

Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) are two important ncRNAs, which are characterized by a lack of protein encoding ability (912). These ncRNAs have been demonstrated to be involved in multiple biological processes, including apoptosis, proliferation and differentiation (1315). miRNAs are a type of short RNA molecule consisting of ~20–22 nucleotides that negatively regulate gene expression by binding to the 3′-untranslated regions (3′-UTR) of the mRNA of a target gene (16). Aberrant miRNA expression has been observed in a number of human diseases, including cancer, neurodegenerative disorders and inflammatory-associated disorders (1719). LncRNAs also participate in the progression of various human diseases, including cancer (20,21), cardiovascular disease (22) and rheumatic diseases (23) by acting as a miRNA sponge (24). LncRNA-hox transcript antisense RNA (HOTAIR) has been previously reported to function as an oncogenic molecule in a number of human malignancies including lung (25), prostate (26), gastric (27) and colorectal cancer (28), etc.. Recently, HOTAIR was observed to be upregulated in mice and cardiomyocytes following lipopolysaccharide (LPS)-induced sepsis, in which silencing HOTAIR protected the cardiac function of septic mice by downregulating tumor necrosis factor-α (TNF-α) via the NF-κB signaling pathway (29).

It has been well documented that sepsis may be mediated by multiple inflammatory cytokines, including TNF-α, interleukin-6 (IL)-6 and IL-1β (30,31). In addition, emerging studies have revealed an association between plasma inflammatory cytokine concentrations and mortality in patients with sepsis (30,32). In particular, the upregulation of IL-6 and its receptor, IL-6R, has been frequently observed in patients with sepsis and the production of IL-6 was demonstrated to be a good prognostic agent in the early phase of sepsis (33,34). These results indicate that IL-6 and its receptor may function as two potential therapeutic targets for patients with sepsis.

In the present study, HOTAIR and IL-6R were revealed to be targeted by miR-211; however, since it remains unknown how the interaction between HOTAIR, IL-6R and miR-211 contribute to the etiology of sepsis, the aim of the present study was to investigate the effects of the HOTAIR/miR-211/IL-6R axis on the pathogenesis of sepsis.

Materials and methods

Establishment of an animal model of sepsis

C57BL/6 mice (age, 8 weeks; mean weight, 23.4±0.92 g; weight range, 22–25 g) were purchased from the Animal Experiment Center of the Institute of Radiation Medicine of the Chinese Academy of Medical Sciences and all of the animal protocols used in the present study were approved by the Institute of Radiation Medicine of the Chinese Academy of Medical Sciences. All the animals were raised for seven days to adapt to the environment prior to experimentation. Animals were raised with sufficient water and feed, at a temperature of 20–24°C, humidity of 50–60% with a 12-h light/dark cycle. A total of 8 male C57BL/6 mice (8 weeks old) were used to induce sepsis via cecal ligation and puncture (CLP). After anesthetizing the mice with 2% pentobarbital sodium (50 mg/kg, intraperitoneally), a small incision was made in the abdomen. The cecum of the mice was then exposed and a sterile 21-gauge needle was used to puncture the cecum twice to extrude fecal matter. Subsequently, the cecum was returned into the abdominal cavity and the incision was closed in two layers. Control mice (n=8) were treated the same as the experimental animals, but without CLP.

Monocytes isolation

Following the establishment of the mouse model 48 h following CPL, C57BL/6 mice in the sepsis and control groups were decapitated and the spleen was subsequently removed. After washing with PBS, the spleen was broken by collagenase (cat. no. 17104019; Gibco; Thermo Fisher Scientific, Inc.) for 5 mins at 37°C, filtered through a 74 µm pore size strainer (BD Biosciences) to create a single-cell suspension. Cell concentration was determined using a hemocytometer (Hausser Scientific) and adjusted to 1×108 cells/ml. After which the mouse monocytes were purified using CD11b MicroBeads (cat. no. 130-049-001; Miltenyi Biotec GmbH) according to the manufacturer's protocol.

Cell culture

Monocytes were isolated from the spleens as aforementioned and maintained in DMEM medium (Sigma-Aldrich; Merck KGaA) containing 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc.) with 1% penicillin and streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified incubator with 5% CO2.

Transfections of miRNA mimics and inhibitors

Negative control (NC or scramble for mimics and inhibitors; 5′-UUCUCCGAACGUGUCACGUTT-3′), miR-211 mimics (5′-UUCCCUUUGUCAUCCUUUGCCU-3′) and miR-211 inhibitors (5′-AGGCAAAGGATGACAAAGGGAA-3′) were synthesized by Shanghai GenePharma Co., Ltd. Monocytes (5×104 cells/well) were seeded in 6-well plates and transfected with NC (50 nM), miR-211 mimics (50 nM) and miR-211 inhibitors (50 nM) using Lipofectamine® 2000 Reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to manufacturer's protocol. Subsequent experiments were performed 48 h following transfection.

Vector construction and transfection

DNA was extracted from the 293T cells using TIANamp Genomic DNA Kit (Tiangen Biotech Co., Ltd.) according to manufacturer's protocol. HOTAIR was amplified using Taq PCR Master Mix Kit (Qiagen, Inc.) with XhoI and BamHI restriction sites. The temperature protocol for the PCR consisted of 94°C for 3 min; followed by 30 cycles of 94°C for 30 sec, 55°C for 30 sec and 72°C 1 min; and final extension at 72°C for 5 min. The primers for amplification were forward, 5′-CCGCTCGAGACATTCTGCCCTGATTTCCGGAACC-3′ and reverse, 5′-CGCGGATCCCCACCACACACACACAACCTACAC-3′. HOTAIR DNAs were inserted into the pcDNA3.0 vector (Invitrogen; Thermo Fisher Scientific, Inc.) according to previous studies (35,36). A total of 1×105 monocytes were seeded into 6-well plates and transfected with the HOTAIR-expression vector or empty vector (control) using Lipofectamine® 2000 Reagent (Invitrogen; Thermo Fisher Scientific, Inc.).

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR) assay

The total RNA isolated from the splenic tissues of septic and control mice, monocytes transfected with HOTAIR, miR-211 mimics, miR-211 inhibitor and corresponding controls were all prepared using the TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. cDNA was subsequently synthesized using PrimeScript™ RT Master Mix (Takara Biotechnology Co., Ltd.) using 50 ng total RNA with the temperature protocol consisting of 95°C for 30 sec and 60°C for 30 mins. The amplification of interferon (IFN)-γ, IL-6, IL-17, TNF-α, IL-1β, IL-6R, miR-211 and HOTAIR was performed using a Bestar® SYBR Green qPCR master mix (DBI Bioscience; Shanghai Xinghan Biotechnology Co., Ltd.) kit using an ABI PRISM 7500 system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycling conditions were: 95°C for 2 min; followed by 30 cycles of 95°C 10 sec, and 60°C 34 sec. The primer sequences used were listed in Table I. The expressions of IFN-γ, IL-6, IL-17, TNF-α, IL-1β, IL-6R and HOTAIR were normalized to the level of GAPDH, whereas miR-211 expression was normalized to the level of U6. The relative expression levels were analyzed using 2−ΔΔCq method (37).

Table I.

Primer sequences for RT-qPCR assay used in this study.

Table I.

Primer sequences for RT-qPCR assay used in this study.

GeneSequence
GAPDH-F 5′-TGTTCGTCATGGGTGTGAAC-3′
GAPDH-R 5′-ATGGCATGGACTGTGGTCAT-3′
U6-F 5′-CTCGCTTCGGCAGCACA-3′
U6-R 5′-AACGCTTCACGAATTTGCGT-3′
IFN-γ-F 5′-AGCGGATAATGGAACTCTTTTCTTAG-3′
IFN-γ-R 5′-AAGTTTGAAGTAAAAGAAGACAATTTGG-3′
IL-6-F 5′-AGTTGCCTTCTTGGGACTGA-3′
IL-6-R 5′-CAGAATTGCCATTGCACAAC-3′
IL-17-F 5′-CCGGACTGTGATGGTCAA-3′
IL-17-R 5′-CTCATTGCGGTGGAGATT-3′
TNF-α-F 5′-CGGGCAGGTCTACTTTGGAG-3′
TNF-α-R 5′-CAGGTCACTGTCCCAGCATC-3′
IL-1β-F 5′-CTTCTTCGACACATGGGATAAC-3′
IL-1β-R 5′-TTTGGGATCTACACTCTCCAGC-3′
IL-6R-F 5′-TGAGCTCAGATATCGGGCTGAAC-3′
IL-6R-R 5′-CGTCGTGGATGACACAGTGATG-3′
miR-211-F 5′-TTGTGGGCTTCCCTTTGTCATCCT-3′
miR-211-R 5′-TGCTGTGGGAAGTGACAACTGA-3′
HOTAIR-F 5′-CAGTGGGGAACTCTGACTCG-3′
HOTAIR-R 5′-GTGCCTGGTGCTCTCTTACC-3′

[i] IFN-γ, interferon-γ; IL, interleukin; TNF-α, tumor-necrosis factor-α; miR, microRNA; HOTAIR, hox transcript antisense RNA.

Western blot assay

The proteins were isolated from the spleens of septic and control mice, monocytes transfected with HOTAIR, miR-211 mimics οr miR-211 inhibitor and corresponding control using RIPA Lysis Buffer System (Santa Cruz Biotechnology, Inc.) supplemented with 1.5 mM PMSF (Sigma-Aldrich; Merck KGaA). The lysates were then subjected to centrifugation at 12,000 × g for 15 min at 4°C, after which the supernatants were collected. The protein concentration was determined using a bicinchoninic acid kit (Pierce; Thermo Fisher Scientific, Inc.). A total of 30 µg proteins per lane were isolated using 10% SDS-PAGE and transferred onto nitrocellulose membranes (EMD Millipore; Merck KGaA) and incubated with 5% skimmed milk at room temperature for 2 h. The membranes were then incubated with primary rabbit antibodies against IFN-γ (1:1,000; cat. no. ab77246), IL-6 (1:2,000; cat. no. ab6672), IL-17 (1:500; cat. no. ab136668), TNF-α (1:500; cat. no. ab6671), IL-1β (1:1,000; cat. no. ab200478), and IL-6R (1:200; cat. no ab128008; all Abcam) at 4°C for 8 h. The membranes were incubated with horseradish peroxidase-conjugated donkey anti-rabbit secondary antibodies (1:2,000; cat. no. ab7083; Abcam) at room temperature for 2 h. Finally, the signals were detected using enhanced chemiluminescent (ECL) kit (Pierce; Thermo Fisher Scientific, Inc.). The grayscale values of the membranes were counted using an ImageJ software (ver. 1.51d; National institutes of Health).

Bioinformatics analysis

TargetScan (http://targetscan.org/) was applied to analyze the possible binding sites between HOTAIR and miR-211 and between miR-211 and IL-6; TargetScan (http://targetscan.org/) (38), StarBase v2.0 (http://starbase.sysu.edu.cn/) (39) and miRDB (http://mirdb.org/miRDB/) (40) databases were utilized to analyze the possible binding site of IL-6 as the downstream target of miR-211.

Dual-luciferase reporter assay

The interaction between miR-211 and HOTAIR, as well as miR-211 and IL-6R were verified with a dual-luciferase reporter assay. Wild-type (WT) HOTAIR, mutant type (Mut) HOTAIR, WT IL-6R and Mut IL-6R were purchased from Hanbio Co., Ltd. (Hanbio Biotechnology Co., Ltd.). Briefly, total DNA was extracted from 293T cells using TIANamp Genomic DNA Kit (Tiangen Biotech Co., Ltd.), and the 3′-untranslated regions (3′-UTR) of the wild type (WT) HOTAIR containing the miR-211 binding sites were amplified using Taq PCR Master Mix Kit (Qiagen GmbH; cat. no. 201443). The temperature protocol for the PCR consisted of 94°C for 3 min; followed by 30 cycles of 94°C for 30 sec, 55°C for 30 sec and 72°C 1 min; and final extension at 72°C for 5 min. The mutant (Mut) 3′-UTR of HOTAIR was generated by changing the sequence from ‘AAAGGGAA’ to ‘UUUCCCUU’. The DNA products were sub-cloned into the luciferase vector, psi-CHECK2 (Promega Corporation) to form a recombinant reporter plasmid. The WT IL-6R (WT-IL-6R) and Mut IL-6R (MUT-IL-6R) were constructed in the same manner as WT-HOTAIR and Mut-HOTAIR. For the miR-211 and HOTAIR dual-luciferase reporter assay, 293T cells were seeded into 24-well plates at a density of 1×104 cells/well. After culturing overnight at 37°C, 293T cells were co-transfected with WT-HOTAIR or Mut-HOTAIR combined with miR-211 mimics or its negative control using a Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). The collected 293T cells were seeded into 24-well plates at a concentration of 2×105 cells/well, and cultured at 37°C overnight. Based on manufacturer's instructions, the luciferase activities were measured with a Dual Luciferase Assay System (Promega Corporation) to verify that the interaction between miR-211 and IL-6R was the same as miR-211 and HOTAIR. All luciferase activities were normalized to Renilla luciferase activity.

Cell proliferation analysis

Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA) was used to evaluate the effects of HOTAIR and miR-211 on monocyte proliferation. The transfected monocytes (1×103 cell/well) were seeded into 96-well plates and transfected with HOTAIR, miR-211 mimics or miR-211 inhibitors for 48 h at 37°C in a humidified incubator with 5% CO2. The optical density (OD) was then measured at 450 nm using a microtiter plate reader (SpectraMax; Molecular Devices, LCC).

Cell apoptosis analysis

Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double staining and flow cytometry were performed to determine the effects of HOTAIR and miR-211 on monocyte apoptosis. After culturing in DMEM for 48 h at 37°C, monocytes transfected with either HOTAIR, miR-211 mimic or miR-211 inhibitor were harvested by centrifugation (1,000 × g for 5 min) and washed twice with PBS. The monocytes were fixed in 70% ethanol for 2 h at room temperature and then incubated with annexin V-FITC and PI (Keygentec) for 10 min in the dark. Finally, the apoptotic cells were evaluated using flow cytometry (BD Biosciences), and analyzed using BD CellQuest software (Version 3.3; BD Biosciences).

Statistical analysis

SPSS software (version 22.0; IBM, Corp.) was used for all statistical analyses. Each experiment was repeated at least three times and the data were expressed as the mean ± standard deviation (SD). A Student's t-test was used for the statistical analyses between two groups and the statistical differences between more than two groups were analyzed using a one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

HOTAIR is directly targeted by miR-211

The interaction between miR-211 and HOTAIR was evaluated using online bioinformatics analysis and a dual-luciferase reporter assay in 293T cells. The bioinformatics analysis revealed that there were putative binding sites for miR-211 in HOTAIR (Fig. 1). Further analysis confirmed that luciferase activity was driven by WT-HOTAIR as it was significantly attenuated by the miR-211 mimics. However, no significant difference was observed with MUT-HOTAIR (P<0.05; Fig. 1).

The 3′-UTR of IL-6R is targeted by miR-211

The interaction between miR-211 and IL-6 using an online bioinformatics analysis tool and dual-luciferase reporter assay. The bioinformatics analysis indicated that there were putative binding sites in the 3′-UTR of IL-6 for miR-211 (Fig. 2). Subsequently, a dual-luciferase reporter assay was performed to verify the interaction between miR-211 and IL-6 and the results revealed that the luciferase activity driven by WT-IL-6R was significantly reduced by the miR-211 mimics; however, there was no significant difference in luciferase activity with MUT-IL-6R following treatment with the miR-211 mimics (P<0.05; Fig. 2).

miR-211 and HOTAIR expression is significantly upregulated in the spleens of mice with CLP-induced sepsis

To explore whether miR-211 and HOTAIR were involved in the pathogenesis of sepsis, a CLP-induced mouse model of sepsis was established. The animal model of sepsis was initially verified by detecting the levels of various inflammatory factors in the spleens via RT-qPCR. The results indicated that the levels of IFN-γ, IL-6, IL-17, TNF-α and IL-1β expression were significantly upregulated in septic mice compared with control mice (Fig. 3A). In addition, there was a significant upregulation in IL-6R expression in the septic mice compared with control mice (P<0.05; Fig. 3A). The level of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R expression was further examined via western blotting. The results demonstrated a significant increase in IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R in the spleens of septic mice compared with control mice (P<0.05; Fig. 4). The expression of miR-211 and HOTAIR in the septic mice was subsequently examined using RT-qPCR. The results indicated that the relative levels of miR-211 and HOTIAR were significantly increased in the splenic tissues from the septic mice compared with control mice (P<0.05; Fig. 3B).

Overexpression of HOTIAR and knockdown of miR-211 promotes the inflammatory response in monocytes

To further explore the biological function of miR-211 and HOTAIR in sepsis, RT-qPCR was performed to determine the levels of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R in the monocytes transfected with HOTAIR, the miR-211 mimics and miR-211 inhibitors. The relative level of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R mRNA and protein expression was demonstrated to be significantly upregulated in the HOTAIR and miR-211 inhibitor-treated groups compared with that in the negative control group (P<0.05; Figs. 5A and 6). However, there was a significant downregulation in the level of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R expression in monocytes treated with the miR-211 mimics compared with the negative control (P<0.05; Figs. 5A and 6). In addition, miR-211 expression was significantly decreased in the HOTAIR overexpressed group, and HOTAIR expression was significantly increased in the miR-211-silenced group. These results indicated that there was a negative association between the expression of miR-211 and HOTAIR in monocytes (P<0.05; Fig. 5B).

Overexpression of HOTAIR and knockdown of miR-211 inhibits proliferation and promotes apoptosis in monocytes

A CCK-8 assay and flow cytometry were performed to investigate the effects of HOTAIR and miR-211 on cellular proliferation and apoptosis, respectively, in monocytes. The results revealed that the rate of cellular monocyte proliferation transfected with HOTAIR and miR-211 inhibitors was significantly reduced, whereas the cells transfected the miR-211 mimics exhibited a significant increase in proliferation compared with the negative control groups, respectively (P<0.05; Fig. 7A). Furthermore, monocytes transfected with the HOTAIR and miR-211 inhibitors demonstrated a significant increase in apoptosis, whereas those transfected with the miR-211 mimics showed a significant decrease in apoptosis, compared with the negative control group (P<0.05, Fig. 7Ba and Bb).

Discussion

HOTAIR is transcribed by the antisense strand of the HOXC gene located on chromosome 12 and is an important lncRNA that was first identified by Rinn et al (41) in 2007, using a microarray assay. In addition, evidence has indicated that HOTAIR regulates chromatin dynamics and induces gene silencing by interacting with histone methylase and histone demethylase (42). Additionally, HOTAIR has been reported to be involved in the etiology of multiple types of human cancer, including hepatocellular, breast and lung cancer (4345). HOTAIR has also been demonstrated to regulate the expression of miRNAs by acting as a competitive endogenous RNA (ceRNA) (42). For example, HOTAIR was revealed to possess the binding sites for miR-130a, which were demonstrated to be critical for the modulation of miR-130a by HOTAIR (46). In the present study, HOTAIR was identified to function as a ceRNA of miR-211, and the expression of HOTAIR and miR-211 were negatively associated in monocytes.

As a major public health issue, sepsis is frequently accompanied by microbial infection, systemic inflammation and cellular dysfunction, which can ultimately result in tissue damage, organ failure and even death (47). There are currently three main hypotheses used to explain the pathogenesis of sepsis: i) Pro-inflammatory response; ii) impaired compensatory anti-inflammatory responses; and iii) immune-paralysis, all of which involve the excessive release of inflammatory mediators responsible for the initiation and development of systemic inflammation (1,48). TNF-α was considered to be a central regulator of the immune response and involved in the pathophysiological alterations associated with sepsis (49,50). In addition, TNF-α was demonstrated to promote the release of inflammatory mediators, including IL-6, IL-8, IL-17 and IL-1β, which initiate the host inflammatory response (51,52). IL-6 is primarily released by activated monocytes and has been demonstrated to be negatively associated with the prognosis of patients with sepsis (53). In the present study, since miR-211 was observed to bind to the 3′-UTR of IL-6R, it was hypothesized whether HOTAIR was involved in the pathogenesis of sepsis by indirectly regulating the expression of IL-6R through miR-211.

Currently, various animal models including zebrafish (54), rat (55) and mice (56) have been established to investigate the etiology of sepsis, including toxin treatments such as LPS, zymosan or endotoxins, viable pathogens (including bacteria), as well as altering the endogenous protective barrier in animals (including the induction of colonic permeability leading to bacterial translocation). In addition, CLP is the most frequently applied model in rodents (57,58), which can be used to create sepsis-inducing animal models (5961). Research has also demonstrated that CLP-induced murine sepsis does not cause lung injury (62), therefore CLP was used to induce murine sepsis in the present study.

Recently, the abnormal expression of lncRNAs has been found in a number of animal models of sepsis, including mice (29) and rat (63), indicating that lncRNAs may be involved in the pathogenesis of sepsis (64,65). In the present study, an animal model of sepsis was established using CLP in mice, which was verified by detecting the increase in mRNA and protein levels of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R. In addition, HOTAIR expression was significantly upregulated, whereas the expression of miR-211 was substantially downregulated in the spleens of septic mice. Furthermore, both HOTAIR overexpression and miR-211 knockdown upregulated the expression of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R in monocytes. Treatment with the miR-211 mimics exhibited the opposite effect in monocytes.

Immune suppression caused by corresponding apoptosis (in monocytes and lymphocytes), has been reported to be associated with the pathogenesis of sepsis (5). Therefore, the abrogation of immune cell apoptosis is considered to be a potential therapeutic measure for patients with sepsis. In the present study, HOTAIR overexpression and miR-211 knockdown were revealed to inhibit cellular proliferation and promote apoptosis in monocytes, whereas miR-211 overexpression was demonstrated to induce the opposite effect in monocytes.

In conclusion, the results of the current study indicated that HOTAIR promoted the progression of sepsis indirectly by regulating IL-6R expression via miR-211. Therefore, HOTAIR may be a potential therapeutic target for patients with sepsis.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on a reasonable request.

Authors' contributions

JC, FC, XG and LZ conceived, designed and coordinated the study and prepared the draft of the manuscript. SW, LZ and YH performed literature research, collected the data, participated in the design of the study and performed the statistical analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All of the animal protocols in the present study were approved by Institute of Radiation Medicine of the Chinese Academy of Medical Sciences.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sagy M, Al-Qaqaa Y and Kim P: Definitions and pathophysiology of sepsis. Curr Probl Pediatr Adolesc Health Care. 43:260–263. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Mohamed AKS, Mehta AA and James P: Predictors of mortality of severe sepsis among adult patients in the medical Intensive Care Unit. Lung India. 34:330–335. 2017. View Article : Google Scholar : PubMed/NCBI

3 

van Vught LA, Wiewel MA, Hoogendijk AJ, Frencken JF, Scicluna BP, Klein Klouwenberg PMC, Zwinderman AH, Lutter R, Horn J, Schultz MJ, et al: The host response in sepsis patients developing intensive care unit-acquired secondary Infections. Am J Respir Crit Care Med. 196:458–470. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Sánchez B, Ferrer R, Suarez D, Romay E, Piacentini E, Gomà G, Martínez ML and Artigas A; Edusepsis Study Group, : Declining mortality due to severe sepsis and septic shock in Spanish intensive care units: A two-cohort study in 2005 and 2011. Med Intensiva. 41:28–37. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Hotchkiss RS, Coopersmith CM, McDunn JE and Ferguson TA: The sepsis seesaw: Tilting toward immunosuppression. Nat Med. 15:496–497. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Jiang W, Zhong W, Deng Y, Chen C, Wang Q, Zhou M, Li X, Sun C and Zeng H: Evaluation of a combination ‘lymphocyte apoptosis model’ to predict survival of sepsis patients in an intensive care unit. BMC Anesthesiol. 18:892018. View Article : Google Scholar : PubMed/NCBI

7 

Gill SE, Rohan M and Mehta S: Role of pulmonary microvascular endothelial cell apoptosis in murine sepsis-induced lung injury in vivo. Respir Res. 16:1092015. View Article : Google Scholar : PubMed/NCBI

8 

Pool R, Gomez H and Kellum JA: Mechanisms of organ dysfunction in sepsis. Crit Care Clin. 34:63–80. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Halimulati M, Duman B, Nijiati J and Aizezi A: Long noncoding RNA TCONS_00024652 regulates vascular endothelial cell proliferation and angiogenesis via microRNA 21. Exp Ther Med. 16:3309–3316. 2018.PubMed/NCBI

10 

Li TT, He RQ, Ma J, Li ZY, Hu XH and Chen G: Long non-coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review). Oncol Rep. 40:1831–1842. 2018.PubMed/NCBI

11 

Ling Z, Liu D, Zhang G, Liang Q, Xiang P, Xu Y, Han C and Tao T: miR-361-5p modulates metabolism and autophagy via the Sp1-mediated regulation of PKM2 in prostate cancer. Oncol Rep. 38:1621–1628. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Ni X, Liao Y, Li L, Zhang X and Wu Z: Therapeutic role of long non-coding RNA TCONS_00019174 in depressive disorders is dependent on Wnt/β-catenin signaling pathway. J Integr Neurosci. 17:125–132. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Zhang TN, Li D, Xia J, Wu QJ, Wen R, Yang N and Liu CF: Non-coding RNA: A potential biomarker and therapeutic target for sepsis. Oncotarget. 8:91765–91778. 2017.PubMed/NCBI

14 

Diamantopoulos MA, Tsiakanikas P and Scorilas A: Non-coding RNAs: The riddle of the transcriptome and their perspectives in cancer. Ann Transl Med. 6:2412018. View Article : Google Scholar : PubMed/NCBI

15 

Gibbons A, Udawela M and Dean B: Non-coding RNA as novel players in the pathophysiology of schizophrenia. Noncoding RNA. 4(pii): E112018.PubMed/NCBI

16 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

17 

O'Connell RM, Rao DS and Baltimore D: microRNA regulation of inflammatory responses. Annu Rev Immunol. 30:295–312. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Schoof CR, Botelho EL, Izzotti A and Vasques Ldos R: MicroRNAs in cancer treatment and prognosis. Am J Cancer Res. 2:414–433. 2012.PubMed/NCBI

19 

Qiu L, Tan EK and Zeng L: microRNAs and neurodegenerative diseases. Adv Exp Med Biol. 888:85–105. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Kondo Y, Shinjo K and Katsushima K: Long non-coding RNAs as an epigenetic regulator in human cancers. Cancer Sci. 108:1927–1933. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Uchida S and Dimmeler S: Long noncoding RNAs in cardiovascular diseases. Circ Res. 116:737–750. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Tang Y, Zhou T, Yu X, Xue Z and Shen N: The role of long non-coding RNAs in rheumatic diseases. Nat Rev Rheumatol. 13:657–669. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Rong D, Sun H, Li Z, Liu S, Dong C, Fu K, Tang W and Cao H: An emerging function of circRNA-miRNAs-mRNA axis in human diseases. Oncotarget. 8:73271–73281. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Loewen G, Jayawickramarajah J, Zhuo Y and Shan B: Functions of lncRNA HOTAIR in lung cancer. J Hematol Oncol. 7:902014. View Article : Google Scholar : PubMed/NCBI

26 

Ling Z, Wang X, Tao T, Zhang L, Guan H, You Z, Lu K, Zhang G, Chen S, Wu J, et al: Involvement of aberrantly activated HOTAIR/EZH2/miR-193a feedback loop in progression of prostate cancer. J Exp Clin Cancer Res. 36:1592017. View Article : Google Scholar : PubMed/NCBI

27 

Liu XH, Sun M, Nie FQ, Ge YB, Zhang EB, Yin DD, Kong R, Xia R, Lu KH, Li JH, et al: Lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331-3p in gastric cancer. Mol Cancer. 13:922014. View Article : Google Scholar : PubMed/NCBI

28 

Lu X, Liu Z, Ning X, Huang L and Jiang B: The long noncoding RNA HOTAIR promotes colorectal cancer progression by sponging miR-197. Oncol Res. 26:473–481. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Wu H, Liu J, Li W, Liu G and Li Z: LncRNA-HOTAIR promotes TNF-α production in cardiomyocytes of LPS-induced sepsis mice by activating NF-κB pathway. Biochem Biophys Res Commun. 471:240–246. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Bosmann M and Ward PA: The inflammatory response in sepsis. Trends Immunol. 34:129–136. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Walborn A, Hoppensteadt D, Syed D, Mosier M and Fareed J: Biomarker profile of sepsis-associated coagulopathy using biochip assay for inflammatory cytokines. Clin Appl Thromb Hemost. 24:625–632. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Schulte W, Bernhagen J and Bucala R: Cytokines in sepsis: Potent immunoregulators and potential therapeutic targets-an updated view. Mediators Inflam. 2013:1659742013. View Article : Google Scholar

33 

Shao WX, Yu DJ, Zhang WY and Wang XJ: Clinical significance of IL-6 in the diagnosis of sepsis and discriminating sepsis induced by gram-negative bacteria. Pediatr Infect Dis J. 37:801–805. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Zhang W and He J: Interleukin-6 is a key factor for immunoglobulin-like transcript-4-mediated immune injury in sepsis. J Intensive Care. 6:222018. View Article : Google Scholar : PubMed/NCBI

35 

Chen Y, Lin MC, Yao H, Wang H, Zhang AQ, Yu J, Hui CK, Lau GK, He ML, Sung J and Kung HF: Lentivirus-mediated RNA interference targeting enhancer of zeste homolog 2 inhibits hepatocellular carcinoma growth through down-regulation of stathmin. Hepatology. 46:200–208. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Jiang L, Lai YK, Zhang J, Wang H, Lin MC, He ML and Kung HF: Targeting S100P inhibits colon cancer growth and metastasis by Lentivirus-mediated RNA interference and proteomic analysis. Mol Med. 17:709–716. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Liu S, Li JH, Wu J, Zhou KR, Zhou H, Yang JH and Qu LH: StarScan: A web server for scanning small RNA targets from degradome sequencing data. Nucleic Acids Res. 43:W480–W486. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Yang JH, Li JH, Shao P, Zhou H, Chen YQ and Qu LH: starBase: A database for exploring microRNA-mRNA interaction maps from Argonaute CLIP-Seq and Degradome-Seq data. Nucleic Acids Res 39 (Database Issue). D202–D209. 2011. View Article : Google Scholar

40 

Wong N and Wang X: miRDB: An online resource for microRNA target prediction and functional annotations. Nucleic Acids Res 43 (Database Issue). D146–D152. 2015. View Article : Google Scholar

41 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Bhan A and Mandal SS: LncRNA HOTAIR: A master regulator of chromatin dynamics and cancer. Biochim Biophys Acta. 1856:151–164. 2015.PubMed/NCBI

43 

Hu ML, Wang XY and Chen WM: TGF-β1 upregulates the expression of lncRNA UCA1 and its downstream HXK2 to promote the growth of hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 22:4846–4854. 2018.PubMed/NCBI

44 

Perrot-Applanat M, Kolf-Clauw M, Michel C and Beausoleil C: Alteration of mammary gland development by bisphenol a and evidence of a mode of action mediated through endocrine disruption. Mol Cell Endocrinol. 475:29–53. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Han L, Zhang HC, Li L, Li CX, Di X and Qu X: Downregulation of long noncoding RNA HOTAIR and EZH2 induces apoptosis and inhibits proliferation, invasion, and migration of human breast cancer cells. Cancer Biother Radiopharm. 33:241–251. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Huang J, Ke P, Guo L, Wang W, Tan H, Liang Y and Yao S: Lentivirus-mediated RNA interference targeting the long noncoding RNA HOTAIR inhibits proliferation and invasion of endometrial carcinoma cells in vitro and in vivo. Int J Gynecol Cancer. 24:635–642. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Yuki K and Murakami N: Sepsis pathophysiology and anesthetic consideration. Cardiovasc Hematol Disord Drug Targets. 15:57–69. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Bone RC, Grodzin CJ and Balk RA: Sepsis: A new hypothesis for pathogenesis of the disease process. Chest. 112:235–243. 1997. View Article : Google Scholar : PubMed/NCBI

49 

Ge L, Hu Q, Chen J, Shi M, Yang H and Zhu G: Inhibition of TNF-α sepsis of lipopolysaccharide induction using nano cerium oxide system. Mater Sci Eng C Mater Biol Appl. 77:405–410. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Y, Cui X, Ning L and Wei D: The effects of tumor necrosis factor-α (TNF-α) rs1800629 and rs361525 polymorphisms on sepsis risk. Oncotarget. 8:111456–111469. 2017.PubMed/NCBI

51 

Gao H, Liu L, Zhao Y, Hara H, Chen P, Xu J, Tang J, Wei L, Li Z, Cooper DKC, et al: Human IL-6, IL-17, IL-1β, and TNF-α differently regulate the expression of pro-inflammatory related genes, tissue factor and swine leukocyte antigen class I in porcine aortic endothelial cells. Xenotransplantation. 24:2017. View Article : Google Scholar

52 

Feng S, Yu H, Yu Y, Geng Y, Li D, Yang C, Lv Q, Lu L, Liu T, Li G and Yuan L: Levels of inflammatory cytokines IL-1β, IL-6, IL-8, IL-17A, and TNF-α in aqueous humour of patients with diabetic retinopathy. J Diabetes Res. 2018:85464232018. View Article : Google Scholar : PubMed/NCBI

53 

McGuire TR, Reardon NT, Bogard K, Plumb TJ, Bultsma CJ, Nissen SW, Fuller PD and Olsen KM: IL6 plasma concentrations in patients with sepsis receiving SLED and antibiotics: A predictor for survival. In Vivo. 28:1131–1134. 2014.PubMed/NCBI

54 

Philip AM, Wang Y, Mauro A, El-Rass S, Marshall JC, Lee WL, Slutsky AS, dosSantos CC and Wen XY: Development of a zebrafish sepsis model for high-throughput drug discovery. Mol Med. 23:134–148. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Shen L, Sun Z, Zhao F, Wang W, Zhang W and Zhu H: Expression of c-FLIP in a rat model of sepsis and its effects on endothelial apoptosis. Mol Med Rep. 16:231–237. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Buras JA, Holzmann B and Sitkovsky M: Animal models of sepsis: Setting the stage. 4:854–865. 2005.PubMed/NCBI

57 

Rittirsch D, Hoesel LM and Ward PA: The disconnect between animal models of sepsis and human sepsis. J Leukoc Biol. 81:137–143. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Chang R, Holcomb JB, Johansson PI, Pati S, Schreiber MA and Wade CE: Plasma resuscitation improved survival in a cecal ligation and puncture rat model of sepsis. Shock. 49:53–61. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Oberbaum M, Spira RM, Lukasiewicz E, Armon Y, Samuels N, Singer SR, Barak V, Izbicki G, Einav S and Hersch M: Effect of Traumeel S on cytokine profile in a cecal ligation and puncture (CLP) sepsis model in rats. J Altern Complement Med. 17:909–913. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Zhang J, Bi J, Liu S, Pang Q, Zhang R, Wang S and Liu C: 5-HT Drives Mortality in Sepsis Induced by Cecal Ligation and Puncture in Mice. Mediators Inflamm. 2017:63742832017. View Article : Google Scholar : PubMed/NCBI

61 

Sakai M, Suzuki T, Tomita K, Yamashita S, Palikhe S, Hattori K, Yoshimura N, Matsuda N and Hattori Y: Diminished responsiveness to dobutamine as an inotrope in mice with cecal ligation and puncture-induced sepsis: Attribution to phosphodiesterase 4 upregulation. Am J Physiol Heart Circ Physiol. 312:H1224–H1237. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Iskander KN, Craciun FL, Stepien DM, Duffy ER, Kim J, Moitra R, Vaickus LJ, Osuchowski MF and Remick DG: Cecal ligation and puncture-induced murine sepsis does not cause lung injury. Crit Care Med. 41:159–170. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Jiang ZJ, Zhang MY, Fan ZW, Sun WL and Tang Y: Influence of lncRNA HOTAIR on acute kidney injury in sepsis rats through regulating miR-34a/Bcl-2 pathway. Eur Rev Med Pharmacol Sci. 23:3512–3519. 2019.PubMed/NCBI

64 

Chen H, Wang X, Yan X, Cheng X, He X and Zheng W: LncRNA MALAT1 regulates sepsis-induced cardiac inflammation and dysfunction via interaction with miR-125b and p38 MAPK/NFκB. Int Immunopharmacol. 55:69–76. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Huang W, Lan X, Li X, Wang D, Sun Y, Wang Q, Gao H and Yu K: Long non-coding RNA PVT1 promote LPS-induced septic acute kidney injury by regulating TNFα and JNK/NF-κB pathways in HK-2 cells. Int Immunopharmacol. 47:134–140. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen J, Gu X, Zhou L, Wang S, Zhu L, Huang Y and Cao F: Long non‑coding RNA‑HOTAIR promotes the progression of sepsis by acting as a sponge of miR‑211 to induce IL‑6R expression. Exp Ther Med 18: 3959-3967, 2019
APA
Chen, J., Gu, X., Zhou, L., Wang, S., Zhu, L., Huang, Y., & Cao, F. (2019). Long non‑coding RNA‑HOTAIR promotes the progression of sepsis by acting as a sponge of miR‑211 to induce IL‑6R expression. Experimental and Therapeutic Medicine, 18, 3959-3967. https://doi.org/10.3892/etm.2019.8063
MLA
Chen, J., Gu, X., Zhou, L., Wang, S., Zhu, L., Huang, Y., Cao, F."Long non‑coding RNA‑HOTAIR promotes the progression of sepsis by acting as a sponge of miR‑211 to induce IL‑6R expression". Experimental and Therapeutic Medicine 18.5 (2019): 3959-3967.
Chicago
Chen, J., Gu, X., Zhou, L., Wang, S., Zhu, L., Huang, Y., Cao, F."Long non‑coding RNA‑HOTAIR promotes the progression of sepsis by acting as a sponge of miR‑211 to induce IL‑6R expression". Experimental and Therapeutic Medicine 18, no. 5 (2019): 3959-3967. https://doi.org/10.3892/etm.2019.8063