Open Access

MicroRNA‑23a‑3p targeting of HMGB1 inhibits LPS‑induced inflammation in murine macrophages in vitro

  • Authors:
    • Qi Sun
    • Bing Wang
    • Mengqiu Li
  • View Affiliations

  • Published online on: March 10, 2022     https://doi.org/10.3892/etm.2022.11251
  • Article Number: 322
  • Copyright: © Sun 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

Inflammatory cytokines, including high mobility group box 1 (HMGB1), play a key role in sepsis via various mechanisms, some of which remain unknown. Sepsis is a common cause of death in patients admitted to the intensive care unit. MicroRNAs (miRs) serve an important role in the inflammatory response. The present study aimed to investigate the role of miR‑23a‑3p in macrophage inflammation and the targeted regulation of HMGB1 expression. The murine macrophage cell line RAW264.7 was subjected to lipopolysaccharide (LPS) treatment to mimic the inflammation involved in sepsis in vitro. Reverse transcription‑quantitative PCR was performed to measure miR‑23a‑3p expression and mRNA expression. Protein levels were determined using ELISA and western blotting. The target binding relationship between miR‑23a‑3p and the HMGB1 3'untranslated region was predicted and validated with a dual luciferase reporter assay. HMGB1 expression was increased and miR‑23a‑3p expression significantly reduced in patients with sepsis and in LPS‑treated RAW264.7 cells in comparison with controls. Overexpression of miR‑23a‑3p reduced interleukin (IL)‑6 and tumor necrosis factor (TNF)‑α expression in RAW264.7 cells under LPS stimulation, while silencing of miR‑23a‑3p elevated the expression of IL‑6 and TNF‑α in comparison with controls. The inhibitory effect of miR‑23a‑3p on LPS‑induced inflammation could be abolished by HMGB1 upregulation in RAW264.7 cells. HMGB1 was targeted by miR‑23a‑3p. miR‑23a‑3p is expressed at reduced levels during inflammation in sepsis, and overexpression of miR‑23a‑3p inhibits LPS‑induced inflammation in murine macrophages in vitro by directly downregulating HMGB1. The results of the present study provided a novel insight into the molecular mechanism underlying HMGB1 expression at the post‑transcriptional level in sepsis.

Introduction

Sepsis is a systemic inflammatory response that is driven by a variety of pathogenic microorganisms including bacteria and their derived products such as endotoxins (1). Sepsis-mediated inflammation may lead to organ dysfunction or circulatory disorders (2). Globally, it's estimated that there are 31.5 million sepsis patients and potentially 5.3 million mortalities every year (3); 51.1% of sepsis patients receive intensive care and ~10% of all intensive care unit patients have severe sepsis in the United States (4,5). Several biomarkers of sepsis have been identified, including phase proteins and inflammatory cytokines such as interleukin (IL)-1, 6 and 10, as well as tumor necrosis factor (TNF)-α (6). Unfortunately, the outcome of patients with sepsis following treatment is often unsatisfactory, due to inflammatory cytokine secretion during the progression of sepsis (7,8). Therefore, it is essential to figure out novel biomarkers for an early diagnosis of sepsis, and thus to develop pharmacological therapy for sepsis by blocking the inflammatory cascade.

High-mobility group box 1 (HMGB1) is a highly conserved non-histone DNA-binding protein (9). It has been documented that HMGB, an extracellularly released mediator, can regulate the inflammatory response (10). Typically, the production of pro-inflammatory cytokines such as TNF-α and IL-1β is induced immediately once inflammation spreads into the blood stream, which then triggers HMGB1 expression (11). HMGB1 is known as a late mediator of inflammation (11). Existing evidence indicates that extracellular HMGB1 may exhibit pro-inflammatory activity in the pathogenesis of various inflammatory diseases (12). HMGB1 is secreted by injured cells and innate immune cells including macrophages (10,13). Moreover, in response to LPS stimulation, HMGB1 can translocate from the nuclei to the cytoplasm in macrophages (14). In the clinic, plasma HMGB1 levels are suggested to be positively correlated with organ dysfunction and death in patients with sepsis (15). Therefore, HMGB1 has been regarded as a promising therapeutic target in inflammatory diseases, including sepsis (16). However, the precise mechanism of HMGB1 involvement in sepsis remains unknown.

MicroRNAs (miRNAs/miRs) are endogenous non-coding transcripts of ~22 nucleotides in length that hinder protein translation through direct base pairing to a broad range of biological systems in animal cells (17). miRNAs serve as essential regulators in inflammatory cytokine release and immune responses (18). In sepsis, it has been highlighted that numerous miRNAs are differentially expressed (19). Moreover, several miRNAs have been identified to be diagnostic or prognostic biomarkers in sepsis, including miR-25 and miR-150 (20-22). miR-23a-3p has been reported to contribute to pathology in cancer, cardiac hypertrophy and muscular atrophy (23). miR-23a-3p is observed to be upregulated in brain tissue, leukocytes and blood plasma during focal cerebral ischemia (24). Additionally, it has been reported that the downregulation of miR-23a-3p in inflammation is associated with TNF-α-induced insulin resistance, LPS-induced immune activation of rat testis and sepsis-induced acute kidney injury (25-27). Furthermore, circulating plasma miRNAs including miR-23a-3p can differentiate human sepsis and systemic inflammatory response syndrome (SIRS) (1). Macrophage activation is involved in the host immune response and inflammatory response to sepsis (28), and, thus, the role of miR-23a-3p in macrophage inflammation needs to be elucidated.

Although the role of HMGB1 in sepsis is well-known, the underlying regulatory mechanism has yet to be fully uncovered (29). The present study aimed to provide a novel insight into the targeted regulation of HMGB1 expression in sepsis by studying the role of miR-23a-3p in LPS-activated RAW264.7 macrophage cells and investigating the regulatory relationship between miR-23a-3p and HMGB1.

Materials and methods

Clinical specimens

A total of 20 patients (male:female, 12:8; age, 37-60 years) with severe sepsis were recruited from Jingzhou Central Hospital of Hubei Province (Jingzhou, China) during July 2018-December 2018 in the present study, together with 12 healthy controls (male:female, 7:5; age, 33-60 years). Peripheral venous blood (4 ml) was collected from all participants. Sepsis was defined according to the Sepsis-3 criteria (30). The excluding criteria for healthy controls: Pregnant women, bone marrow or organ transplant recipients, patients with cancer, individuals suffering with neutropenia, leukopenia or acquired immune deficiency syndrome. The present study was approved by the Ethics Committee of Jingzhou Central Hospital of Hubei, and informed written consent was obtained from all participants prior to study commencement.

Isolation of serum and peripheral blood mononuclear cells (PBMCs)

Blood freshly collected in sodium citrate was used for isolation. For serum preparation, 2 ml blood was kept at -4˚C overnight, then centrifuged at 800 x g for 10 min at 4˚C. The supernatant was collected as serum samples and stored at -80˚C until further use. For PBMC isolation, 2 ml blood was centrifuged at 400 x g for 30 min at 18˚C on Ficoll-Paque PREMIUM (GE Healthcare; Cytiva) according to the manufacturer's instructions. The PBMC layer was collected and washed, and the cell pellet was re-suspended in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin; Invitrogen; Thermo Fisher Scientific, Inc.).

Cell culture and lipopolysaccharide (LPS) treatment

The murine macrophage cell line RAW264.7 was acquired from the American Type Culture Collection and cultured in DMEM supplemented with 10% FBS at 37˚C in 5% CO2. To induce inflammation in vitro, LPS (cat. no. L4516) was purchased from Sigma-Aldrich (Merck KGaA). RAW264.7 cells were exposed to 100 ng/ml LPS at 37˚C for 48 h prior to collection of cells and supernatants and RAW264.7 cells without LPS treatment served as the control.

Cell transfection

RAW264.7 cells were transferred to a six-well plate (Corning, Inc.) and incubated overnight. The pcDNA3.1+ (Invitrogen; Thermo Fisher Scientific, Inc.) was used to overexpress HMGB1 via molecular cloning technology. Oligonucleotides (30 nM) and pcDNA-HMGB1 plasmids (2 µg) were transfected into RAW264.7 cells using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. GMR-miR miR-23a-3p mimic (miR-23a-3p; 5'-AUCACAUUGCCAGGGAUUU-3'), miR-23a-3p inhibitor (in-miR-23a-3p; 5'-AAAUCCCUGGCAAUGUGAU-3') and the indicated negative controls (miR-NC; 5'-CAGUACUUUUGUGUAGUACAA-3') and in-miR-NC (5'-UUGUACUACACAAAAGUACUG-3') were acquired from Shanghai GenePharma Co., Ltd. Transfected cells were cultured for an additional 48 h prior to further studies, among which transfected cells were treated LPS (100 ng/ml at 37˚C for 48 h).

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). For examination of the mRNAs (including IL-6, TNF-α and HMGB1), first-strand cDNA was synthesized using All-In-One 5X RT MasterMix (Abcam), and the reaction thermal profile was 37˚C for 15 min, 60˚C for 10 min and 95˚C for 3 min. For examination of the miRs, a miScript Reverse Transcription kit (Qiagen GmbH) was used, and the reaction thermal profile was 37˚C for 60 min, and 95˚C for 5 min. To determine the levels of mRNAs and mature miR-23a-3p, qPCR was performed using SYBR Green qPCR Mix (Abcam) and miScript SYBR Green PCR Kit (Qiagen GmbH), respectively on an ABI PRISM 7500 Real-time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycling conditions were 95˚C for 15 min, and 35 cycles of 94˚C for 15 sec, 55˚C for 30 sec and 70˚C for 30 sec. GAPDH and U6 small nuclear RNA were used as internal controls for mRNA and miRNA, respectively. We chose 2-ΔΔCq method for evaluation of data (31). The reactions were performed in triplicate for each sample. The primers involved were listed as follows: IL-6 forward (F), 5'-ACGGCCTTCCCTACTTC-3', and reverse (R), 5'-GCTGGACTGTTTCTAATGC-3'; TNF-α F, 5'-GGGTGTTCATCCATTCTC-3', and R, 5'-GGAAAGCCCATTTGAGT-3'; HMGB1 F, 5'-GGAGTGGCTTTTGTCCCTCAT-3', and R, 5'-TGCCTCTCGGCTTTTTAGGA-3'; GAPDH F, 5'-GGTTGTCTCCTGCGACTTCA-3', and R, 5'-GGTGGTCCAGGGTTTCTTACT-3'; U6 F, 5'-CTCGCTTCGGCAGCACA-3', and R, 5'-AACGCTTCACGAATTTGCGT-3'. These primer sequences were supported by a previous literature (32). Primers for hsa-miR-23a-3p and mmu-miR-23a-3p were purchased from Exiqon A/S (Qiagen AB).

Western blotting

Total protein was extracted in RIPA lysis buffer (Beyotime Institute of Biotechnology). After determining the protein concentration using a Bradford protein assay (Bio-Rad Laboratories, Inc.), 20 µg samples were loaded for the standard procedures of western blotting. In brief, 15% sodium dodecyl sulfate polyacrylamide gel electrophoresis gel and polyvinylidene fluoride membrane (MilliporeSigma) were used, and the blocking was performed in 5% skim milk for 1 h at 25˚C. β-actin served as a loading control to normalize protein levels. Primary antibodies including HMGB1 (cat. no. ab92310; 1:2,000; Abcam) and β-actin (cat. no. ab179467; 1:10,000; Abcam) were incubated overnight at 4˚C, and secondary antibody goat anti-rabbit IgG H&L (HRP-conjugated; cat. no. ab6721; 1:20,000; Abcam) was incubated at 25˚C for 1 h. Then, protein blot signals were detected by enhanced chemiluminescence (MilliporeSigma) and quantified using Image Lab™ v3.0 Software (Bio-Rad Laboratories, Inc.).

ELISA

The protein levels of IL-6 and TNF-α in the culture supernatants or serum were measured using mouse IL-6 ELISA kit (cat. no. EM004-48; Genetimes Technology, Inc.) and mouse TNF-α ELISA kit (cat. no. EM008-96; Genetimes Technology, Inc.) according to the instructions of the manufacturer. The serum supernatant was obtained from whole bloods after clot formation and culture supernatant was collected from cell culture media after LPS treatment by centrifuging at 800 x g for 10 min at 4˚C. The reactions were repeated for three times for each sample.

Dual luciferase reporter assay

DianaTools (Diana Lab, University of Thessaly, Thessaly, Greece) with microT-CDS algorithm (33) determined that there was a potential target binding site for miR-23a-3p in the 3'untranslated region (3'UTR) of HMGB1. For determination, the fragment of the HMGB1 3'UTR containing the putative sequence (positions 649-669) was mutated by replacing the AAU…GC…UGUGAU of the complementary sequence. Then, the wild type and mutant of HMGB1 3'UTR (HMGB1 WT/MUT) were cloned into a pGL3 vector (Invitrogen; Thermo Fisher Scientific, Inc.). RAW264.7 cells were co-transfected with HMGB1 WT/MUT (2 µg) and miR-23a-3p/NC mimic (30 nM), or co-transfected with HMGB1 WT/MUT (2 µg) and in-miR-23a-3p/NC (30 nM) using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). pRL-TK plasmids were used as an internal control and co-transfected with those at a dose of 50 ng. Every transfection group was carried out in triplicate. After 48 h, transfected cells were collected to measure Firefly and Renilla luciferase activity using a dual-luciferase reporter assay system (Promega Corporation), and relative luciferase activity was the ratio of Firefly and Renilla.

Statistical analysis

Data are presented as the mean ± standard error of mean. Statistical analyses were performed using GraphPad Prism 5.0 (GraphPad Software, Inc.). Differences between groups were evaluated using one-way analysis of variance followed by Tukey's post hoc test, and P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of HMGB1 is increased and miR-23a-3p is downregulated in patients with sepsis

Whole blood from healthy volunteers (control; n=12) and patients with sepsis (sepsis; n=20) was collected, and serum and PBMCs were subsequently isolated. Expression levels of miR-23a-3p and HMGB1 were measured, as were the levels of pro-inflammatory cytokines. RT-qPCR analysis demonstrated that mRNA expression of two critical pro-inflammatory cytokines, IL-6 and TNF-α, was significantly elevated in sepsis group PBMCs compared with the control group (Fig. 1A and B). In addition, ELISA data showed that the secretion of IL-6 and TNF-α was significantly higher in serum and PBMCs from the sepsis group compared with the control group (Fig. 1C and D). In patients with sepsis, expression of miR-23a-3p was significantly downregulated (~0.22-fold) and the level of HMGB1 protein was significantly upregulated (~2.15-fold) in the serum and PBMCs (Fig. 1E and F) compared with levels in controls. These results suggested an inhibition of miR-23a-3p, and a promotion of HMGB1, IL-6 and TNF-α expression in patients with sepsis, indicating a potential role for miR-23a-3p in the inflammatory response during sepsis.

miR-23a-3p and HMGB1 are differentially expressed following LPS treatment in murine macrophage cells

To identify the biological function of miR-23a-3p and HMGB1 in innate immunity, a cell model of sepsis was constructed using RAW264.7 cells stimulated by LPS. The levels of pro-inflammatory cytokines released during LPS treatment were determined compared with control cells without LPS treatment. RT-qPCR data showed that LPS induced the expression of IL-6 and TNF-α at the mRNA level compared with the control (Fig. 2A and B), accompanied with increased secretion of these cytokines into the culture supernatants (Fig. 2C and D). These findings indicated the successful construction of a LPS-induced inflammation model in murine macrophages in vitro. RT-qPCR and western blotting determined that the expression of miR-23a-3p was significantly reduced, and that of HMGB1 protein was increased, in LPS-stimulated RAW264.7 cells compared with control cells without LPS treatment (Fig. 2E and F). These results suggested that LPS stimulation induced an inflammatory response comparable to sepsis in RAW264.7 cells, during which miR-23a-3p was downregulated and HMGB1 was upregulated.

miR-23a-3p modulates LPS-induced inflammation in murine macrophage cells in vitro

RAW264.7 cells were transfected with miR-23a-3p mimic to overexpress miR-23a-3p, and they were transfected with in-miR-23a-3p to silence miR-23a-3p. Efficiency of transfection was evaluated via RT-qPCR. miR-23a-3p expression was significantly increased following treatment with miR-23a-3p mimic, whereas it was inhibited by transfection of in-miR-23a-3p, compared with the corresponding NCs (Fig. 3A). RT-qPCR and ELISA were used to analyze LPS-induced TNF-α and IL-6 expression. As indicated by Fig. 3B and C, overexpression of miR-23a-3p reduced IL-6 and TNF-α mRNA expression levels in RAW264.7 cells under LPS stimulation compared with control cells, whereas silencing of miR-23a-3p had the opposite effect. Moreover, the release of IL-6 and TNF-α into culture supernatants was decreased by miR-23a-3p overexpression, but promoted by miR-23a-3p inhibition (Fig. 3D and E). These data suggested miR-23a-3p may inhibit LPS-induced inflammation in murine macrophages in vitro.

miR-23a-3p physically targets HMGB1 via complementary binding

DianaTools in silico data predicted that there were complementary binding sites of miR-23a-3p in HMGB1 3'UTR at position 649-669. As presented in Fig. 4A, a HMGB1 MUT was constructed. A dual luciferase reporter assay showed that miR-23a-3p mimic transfection induced a significant decrease in the luciferase activity of HMGB1 WT in RAW264.7 cells, whereas in-miR-23a-3p transfection caused a significant increase (Fig. 4B and C). These results suggested a direct targeting relationship between HMGB1 and miR-23a-3p. RT-qPCR and western blotting data demonstrated that expression of HMGB1 at both the mRNA and the protein level was significantly inhibited in RAW264.7 cells transfected with miR-23a-3p, but it was promoted following in-miR-23a-3p transfection (Fig. 4D and E). These findings indicated that miR-23a-3p modulated LPS-induced inflammation in murine macrophages by targeting HMGB1.

Overexpression of HMGB1 attenuates the effects of miR-23a-3p overexpression in LPS-induced inflammation in vitro

RAW264.7 cells were divided into four transfection groups: miR-NC, miR-23a-3p, miR-23a-3p + pcDNA and miR-23a-3p + pcDNA-HMGB1. The expression of HMGB1 in RAW264.7 cells was evaluated via western blotting, and HMGB1 protein expression was significantly increased following pcDNA-HMGB1 transfection compared with the empty vector control (Fig. 5A). Furthermore, miR-23a-3p transfection inhibited HMGB1 in RAW264.7 cells, and this inhibition was attenuated following pcDNA-HMGB1 transfection (Fig. 5B). As presented in Fig. 5C and D, overexpression of miR-23a-3p reduced IL-6 and TNF-α mRNA expression levels in RAW264.7 cells under LPS stimulation compared with the NC, but overexpression of HMGB1 reversed this effect. Moreover, the release of IL-6 and TNF-α into the culture supernatant was suppressed by miR-23a-3p, which was subsequently reversed by HMGB1 restoration (Fig. 5E and F). These results suggested that the suppressive effect of miR-23a-3p overexpression on LPS-induced inflammation in murine macrophages was partially mediated via downregulation of HMGB1.

Discussion

Several preclinical studies in lethal sepsis have suggested that HMGB1 may be a promising target for improving therapeutic outcomes (34,35). In the present study, an increase of HMGB1 in patients with sepsis and LPS-challenged macrophages was observed alongside decreased expression of miR-23a-3p. In addition, overexpression of miR-23a-3p reduced, whereas silencing of miR-23a-3p elevated, pro-inflammatory cytokine expression (IL-6 and TNF-α) in RAW264.7 cells under LPS stimulation, and the inhibitory effect of miR-23a-3p overexpression on LPS-induced inflammation was attenuated following HMGB1 upregulation. Of note, HMGB1 was targeted by miR-23a-3p. These findings indicated a novel mechanism of HMGB1 in sepsis regulated by miR-23a-3p.

HMGB1 is a member of the HMGB family, which contributes to the regulation of gene expression (14,32). However, in response to LPS stimulation, HMGB1 is translocated from the nuclei to the cytoplasm of macrophages (14). Moreover, superfluous HMGB1 has been demonstrated to be secreted by macrophages and monocytes, thus participating in the occurrence of sepsis (36,37). In sepsis studies, Wang et al (9) indicated that HMGB1 and LPS in harmful concentrations were synergistically toxic or lethal. Additionally, it was reported that HMGB1 has a role in LPS-induced cytotoxicity for reasons that have not been fully elucidated (16,38). Deng et al (39) discovered that hepatocyte-released HMGB1 could bind and target LPS into the lysosomes of macrophages and endothelial cells. Therefore, HMGB1 enabled LPS to reach cytosolic caspase-11, thus forming multiple critical inflammatory mediators (40,41). In the present study, miR-23a-3p mimic-mediated upregulation of HMGB1 attenuated the expression of IL-6 and TNF-α in LPS-stimulated murine macrophages. Overexpression of HMGB1 promoted the production of IL-6 and TNF-α. Collectively, HMGB1 could be important for LPS to express its cytotoxicity.

A growing number of studies have indicated that multiple miRNAs are involved in the inflammatory response of macrophages by regulating HMGB1 expression. For example, miR-212-3p has been claimed to inhibit inflammatory responses in LPS-treated RAW264.7 cells by targeting HMGB1(32). Zhou et al (29) demonstrated that HMGB1 was regulated by a handful of miRNAs, and that miR-205-5b expression was negatively associated with HMGB1 expression. Peroxisome proliferator-activated receptor γ suppresses inflammatory gene expression and pro-inflammatory transcription-factor signaling pathways in various cell types, and its agonist troglitazone mediates HMGB1 inhibition, which is associated with the upregulation of miR-142-3p in inflammatory responses in vitro and in vivo (42). In the present study, it was observed that miR-23a-3p was downregulated and inversely expressed with HMGB1 in LPS-induced RAW264.7 cells; overexpression of miR-23a-3p reduced the expression of IL-6, TNF-α and HMGB1 both at the mRNA and protein level by targeting HMGB1. These data indicated the protective effect of miR-23a-3p in sepsis, and suggested that miR-23a-3p may act as a novel negative regulator of macrophage inflammation. Unfortunately, the role of miR-23a-3p in inflammation-related signal pathways, such as the MAPK (32), JAK/STAT1(43) and NF-κB (44) pathways, was not further investigated in the present study. Animal experiments with miR-23a-3p should be performed for further investigation of the expression of IL-6, TNF-α and HMGB1 in serum and organs, including the liver, lung and kidney (29). In addition, miR-23a-3p together miRNAs were significantly decreased in both sepsis-induced acute kidney injury (AKI) and other AKI groups, and potential target genes of miR-23a-3p were predicted, including IL-6(27). In the present study, it was suggested that IL-6 could be directly and indirectly regulated by miR-23a-3p in sepsis cell models. However, the target relationship between miR-23a-3p and IL-6 in LPS-induced RAW264.7 cells still requires further investigation.

miR-23a-3p functions are extensive. Accumulating evidence indicates that miR-23a-3p is involved in multiple diseases, including cancer, ischemia injury and inflammation. For example, in renal cell carcinoma (RCC), miR-23a-3p targeted proline-rich nuclear receptor coactivator 2 to act as an oncogene by enhancing cell proliferation and cell mobility, and inhibiting apoptosis in ACHN and 786-O cells, thus being a potential prognostic biomarker for RCC (45). miR-23a-3p, together with 8 other miRNAs, was observed to exhibit increased expression in brain tissue, leukocytes and blood plasma 48 h after onset of photochemically-induced focal cerebral ischemia (24). Moreover, it was also reported that oxidative stress injury was alleviated in a mouse model of focal cerebral ischemia-reperfusion (46). Several studies indicated the downregulation of miR-23a-3p under inflammation. For example, inflammation response affected the miRNA profile of the male reproductive tract; five miRNAs, including miR-23a-3p, let-7f-5p, miR-200c-3p, miR-23b-3p and miR-98-5p, exhibited >2-fold downregulation after intraperitoneal injection of LPS in rats for 3 h (26). Serum miR-23a-3p was lower in sepsis-induced human AKI, as well as miR-4456, miR-142-5p, miR-22-3p and miR-191-5p (27). Furthermore, 20 circulating inflammation-associated miRNAs were downregulated in sepsis compared with SIRS, and miR-23a-3p was one of the top 6 most differentially expressed miRNAs in severe sepsis (1). However, there remained a lack of detailed information upon the dysregulation of miR-23a-3p and its molecular regulatory mechanism. Therefore, the expression of miR-23a-3p in sepsis was investigated. The results showed miR-23a-3p was downregulated to ~0.22-fold in patients with sepsis and an LPS-induced cell model of sepsis. Functionally, upregulation of miR-23a-3p resulted in the inhibitory influence on inflammation with decreased expression of IL-6 and TNF-α via targeting HMGB1.

The present study provided novel insight into regulation of HMGB1 by miR-23a-3p, and the miR-23a-3p/HMGB1 axis may represent a clinically relevant potential pharmacological target for effective therapeutic intervention in sepsis. Nevertheless, it should also be noted that targeted therapies, including those involving monoclonal antibodies or antagonists, could be limited, due to redundancy in the inflammatory response (47).

Collectively, it was demonstrated that miR-23a-3p negatively regulated LPS-induced inflammatory cytokine secretion in murine macrophages in vitro. Additionally, a novel mechanism for HMGB1 in sepsis was uncovered that was mediated by miR-23a-3p. Functional experiments suggested an inhibitory effect of miR-23a-3p on inflammatory cytokine expression via direct downregulation of HMGB1.

Acknowledgements

Not applicable.

Funding

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 reasonable request.

Authors' contributions

QS and BW analyzed and interpreted the patient data, performed all experiments and wrote the first draft. ML designed the experiments, agreed to be accountable for all aspects of the work, revised this manuscript critically and gave the final approval of the version to be published. All authors read and approved the final manuscript. QS and BW confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Ethical approval was granted by the Ethics Committee of Jingzhou Central Hospital of Hubei. Participants provided their written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Caserta S, Kern F, Cohen J, Drage S, Newbury SF and Llewelyn MJ: Circulating plasma microRNAs can differentiate human sepsis and systemic inflammatory response syndrome (SIRS). Sci Rep. 6(28006)2016.PubMed/NCBI View Article : Google Scholar

2 

Tracey KJ: The inflammatory reflex. Nature. 420:853–859. 2002.PubMed/NCBI View Article : Google Scholar

3 

Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Tsaganos T, Schlattmann P, Angus DC and Reinhart K: International Forum of Acute Care Trialists. Assessment of global incidence and mortality of Hospital-treated sepsis. Am J Respir Crit Care Mede. 193:259–272. 2016.PubMed/NCBI View Article : Google Scholar

4 

Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J and Pinsky MR: Epidemiology of severe sepsis in the United States: Analysis of incidence, outcome, and associated costs of care. Crit Care Med. 29:1303–1310. 2001.PubMed/NCBI View Article : Google Scholar

5 

Cho W, Koo JY, Park Y, Oh K, Lee S, Song JS, Bae MA, Lim D, Lee DS and Park SB: Treatment of sepsis pathogenesis with high mobility group box protein 1-regulating anti-inflammatory agents. J Med Chem. 60:170–179. 2017.PubMed/NCBI View Article : Google Scholar

6 

Adib-Conquy M and Cavaillon JM: Stress molecules in sepsis and systemic inflammatory response syndrome. FEBS Lett. 581:3723–3733. 2007.PubMed/NCBI View Article : Google Scholar

7 

Lyle NH, Pena OM, Boyd JH and Hancock RE: Barriers to the effective treatment of sepsis: Antimicrobial agents, sepsis definitions, and host-directed therapies. Ann N Y Acad Sci. 1323:101–114. 2014.PubMed/NCBI View Article : Google Scholar

8 

Silman NJ: Rapid diagnosis of sepsis using biomarker signatures. Crit Care. 17(1020)2013.PubMed/NCBI View Article : Google Scholar

9 

Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, et al: HMG-1 as a late mediator of endotoxin lethality in mice. Science. 285:248–251. 1999.PubMed/NCBI View Article : Google Scholar

10 

Gonelevue S, Bandyopadhyay A, Bhagat S, Alam MI and Khan GA: Sterile inflammatory role of high mobility group Box 1 protein: Biological functions and involvement in disease. J Vasc Res. 55:244–254. 2018.PubMed/NCBI View Article : Google Scholar

11 

Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, et al: HMGB1 in health and disease. Mol Aspects Med. 40:1–116. 2014.PubMed/NCBI View Article : Google Scholar

12 

Magna M and Pisetsky DS: The role of HMGB1 in the pathogenesis of inflammatory and autoimmune diseases. Mol Med. 20:138–146. 2014.PubMed/NCBI View Article : Google Scholar

13 

Ulloa L and Tracey KJ: The ʻCytokine Profileʼ: A code for sepsis. Trends Mol Med. 11:56–63. 2005.PubMed/NCBI View Article : Google Scholar

14 

Wang H, Yang H and Tracey KJ: Extracellular role of HMGB1 in inflammation and sepsis. J Intern Med. 255:320–331. 2004.PubMed/NCBI View Article : Google Scholar

15 

Gibot S, Massin F, Cravoisy A, Barraud D, Nace L, Levy B and Bollaert PE: High-mobility group box 1 protein plasma concentrations during septic shock. Intensive Care Med. 33:1347–1353. 2007.PubMed/NCBI View Article : Google Scholar

16 

Andersson U and Tracey KJ: HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev Immunol. 29:139–162. 2011.PubMed/NCBI View Article : Google Scholar

17 

Kingsley SMK and Bhat BV: Role of microRNAs in sepsis. Inflamm Res. 66:553–569. 2017.PubMed/NCBI View Article : Google Scholar

18 

Bulun SE and Nezhat C: Aromatase, microRNA, and inflammation: A complex relationship. Fertil Steril. 106:552–553. 2016.PubMed/NCBI View Article : Google Scholar

19 

Essandoh K and Fan GC: Role of extracellular and intracellular microRNAs in sepsis. Biochim Biophys Acta. 1842:2155–2162. 2014.PubMed/NCBI View Article : Google Scholar

20 

Yao Y, Sun F and Lei M: miR-25 inhibits sepsis-induced cardiomyocyte apoptosis by targetting PTEN. Biosci Rep. 38(BSR20171511)2018.PubMed/NCBI View Article : Google Scholar

21 

Ma Y, Liu Y, Hou H, Yao Y and Meng H: MiR-150 predicts survival in patients with sepsis and inhibits LPS-induced inflammatory factors and apoptosis by targeting NF-κB1 in human umbilical vein endothelial cells. Biochem Biophys Res Commun. 500:828–837. 2018.PubMed/NCBI View Article : Google Scholar

22 

Benz F, Roy S, Trautwein C, Roderburg C and Luedde T: Circulating MicroRNAs as biomarkers for sepsis. Int J Mol Sci. 17(78)2016.PubMed/NCBI View Article : Google Scholar

23 

Chhabra R, Dubey R and Saini N: Cooperative and individualistic functions of the microRNAs in the miR-23a~27a~24-2 cluster and its implication in human diseases. Mol Cancer. 9(232)2010.PubMed/NCBI View Article : Google Scholar

24 

Gusar VA, Timofeeva AV, Zhanin IS, Shram SI and Pinelis VG: Estimation of Time-dependent microRNA expression patterns in brain tissue, leukocytes, and blood plasma of rats under photochemically induced focal cerebral ischemia. Mol Biol (Mosk). 51:683–695. 2017.PubMed/NCBI View Article : Google Scholar : (In Russian).

25 

Lozano-Bartolomé J, Llauradó G, Portero-Otin M, Altuna-Coy A, Rojo-Martinez G, Vendrell J, Jorba R, Rodríguez-Gallego E and Chacón MR: Altered expression of miR-181a-5p and miR-23a-3p is associated with obesity and TNFα-induced insulin resistance. J Clin Endocrinol Metab. 103:1447–1458. 2018.PubMed/NCBI View Article : Google Scholar

26 

Parker MI and Palladino MA: MicroRNAs downregulated following immune activation of rat testis. Am J Reprod Immunol: 77, 2017 doi: 10.1111/aji.12673.

27 

Ge QM, Huang CM, Zhu XY, Bian F and Pan SM: Differentially expressed miRNAs in sepsis-induced acute kidney injury target oxidative stress and mitochondrial dysfunction pathways. PLoS One. 12(e0173292)2017.PubMed/NCBI View Article : Google Scholar

28 

Chen X, Liu Y, Gao Y, Shou S and Chai Y: The roles of macrophage polarization in the host immune response to sepsis. Int Immunopharmacoly. 96(107791)2021.PubMed/NCBI View Article : Google Scholar

29 

Zhou W, Wang J, Li Z, Li J and Sang M: MicroRNA-2055b inhibits HMGB1 expression in LPS-induced sepsis. Int J Mol Med. 38:312–318. 2016.PubMed/NCBI View Article : Google Scholar

30 

Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al: The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 315:801–810. 2016.PubMed/NCBI View Article : Google Scholar

31 

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.PubMed/NCBI View Article : Google Scholar

32 

Chen W, Ma X, Zhang P, Li Q, Liang X and Liu J: miR-212-3p inhibits LPS-induced inflammatory response through targeting HMGB1 in murine macrophages. Exp Cell Res. 350:318–326. 2017.PubMed/NCBI View Article : Google Scholar

33 

Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T, Reczko M, Filippidis C, Dalamagas T and Hatzigeorgiou AG: DIANA-microT web server v5.0: Service integration into miRNA functional analysis workflows. Nucleic Acids Res. 41:W169–W173. 2013.PubMed/NCBI View Article : Google Scholar

34 

Stevens NE, Chapman MJ, Fraser CK, Kuchel TR, Hayball JD and Diener KR: Therapeutic targeting of HMGB1 during experimental sepsis modulates the inflammatory cytokine profile to one associated with improved clinical outcomes. Sci Rep. 7(5850)2017.PubMed/NCBI View Article : Google Scholar

35 

Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, Susarla SM, Ulloa L, Wang H, DiRaimo R, et al: Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci USA. 101:296–301. 2004.PubMed/NCBI View Article : Google Scholar

36 

Charoensup J, Sermswan RW, Paeyao A, Promakhejohn S, Punasee S, Chularari C, Krabkraikaew S, Lertanekawattana S and Wongratanacheewin S: High HMGB1 level is associated with poor outcome of septicemic melioidosis. Int J Infect Dis. 28:111–116. 2014.PubMed/NCBI View Article : Google Scholar

37 

Guo ZS, Liu Z, Bartlett DL, Tang D and Lotze MT: Life after death: Targeting high mobility group box 1 in emergent cancer therapies. Am J Cancer Res. 3:1–20. 2013.PubMed/NCBI

38 

Andersson U, Yang H and Harris H: Extracellular HMGB1 as a therapeutic target in inflammatory diseases. Expert Opin Ther Targets. 22:263–277. 2018.PubMed/NCBI View Article : Google Scholar

39 

Deng M, Tang Y, Li W, Wang X, Zhang R, Zhang X, Zhao X, Liu J, Tang C, Liu Z, et al: The endotoxin delivery protein HMGB1 mediates Caspase-11-dependent lethality in sepsis. Immunity. 49:740–753.e7. 2018.PubMed/NCBI View Article : Google Scholar

40 

Kim HM and Kim YM: HMGB1: LPS delivery vehicle for caspase-11-mediated pyroptosis. Immunity. 49:582–584. 2018.PubMed/NCBI View Article : Google Scholar

41 

Wu D, Pan P, Su X, Zhang L, Qin Q, Tan H, Huang L and Li Y: Interferon regulatory factor-1 mediates alveolar macrophage pyroptosis during LPS-induced acute lung injury in mice. Shock. 46:329–338. 2016.PubMed/NCBI View Article : Google Scholar

42 

Yuan Z, Luo G, Li X, Chen J, Wu J and Peng Y: PPARγ inhibits HMGB1 expression through upregulation of miR-142-3p in vitro and in vivo. Cell Signal. 28:158–164. 2016.PubMed/NCBI View Article : Google Scholar

43 

Park EJ, Kim YM, Kim HJ and Chang KC: Degradation of histone deacetylase 4 via the TLR4/JAK/STAT1 signaling pathway promotes the acetylation of high mobility group box 1 (HMGB1) in lipopolysaccharide-activated macrophages. FEBS Open Bio. 8:1119–1126. 2018.PubMed/NCBI View Article : Google Scholar

44 

Lee W, Ku SK and Bae JS: Zingerone reduces HMGB1-mediated septic responses and improves survival in septic mice. Toxicol Appl Pharmacol. 329:202–211. 2017.PubMed/NCBI View Article : Google Scholar

45 

Quan J, Pan X, Li Y, Hu Y, Tao L, Li Z, Zhao L, Wang J, Li H, Lai Y, et al: miR-23a-3p acts as an oncogene and potential prognostic biomarker by targeting PNRC2 in RCC. Biomed Pharmacother. 110:656–666. 2018.PubMed/NCBI View Article : Google Scholar

46 

Zhao H, Tao Z, Wang R, Liu P, Yan F, Li J, Zhang C, Ji X and Luo Y: MicroRNA-23a-3p attenuates oxidative stress injury in a mouse model of focal cerebral ischemia-reperfusion. Brain Res. 1592:65–72. 2014.PubMed/NCBI View Article : Google Scholar

47 

Minnich DJ and Moldawer LL: Anti-cytokine and anti-inflammatory therapies for the treatment of severe sepsis: Progress and pitfalls. Proc Nutr Soc. 63:437–441. 2004.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2022
Volume 23 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
Sun Q, Wang B and Li M: MicroRNA‑23a‑3p targeting of HMGB1 inhibits LPS‑induced inflammation in murine macrophages <em>in vitro</em>. Exp Ther Med 23: 322, 2022
APA
Sun, Q., Wang, B., & Li, M. (2022). MicroRNA‑23a‑3p targeting of HMGB1 inhibits LPS‑induced inflammation in murine macrophages <em>in vitro</em>. Experimental and Therapeutic Medicine, 23, 322. https://doi.org/10.3892/etm.2022.11251
MLA
Sun, Q., Wang, B., Li, M."MicroRNA‑23a‑3p targeting of HMGB1 inhibits LPS‑induced inflammation in murine macrophages <em>in vitro</em>". Experimental and Therapeutic Medicine 23.5 (2022): 322.
Chicago
Sun, Q., Wang, B., Li, M."MicroRNA‑23a‑3p targeting of HMGB1 inhibits LPS‑induced inflammation in murine macrophages <em>in vitro</em>". Experimental and Therapeutic Medicine 23, no. 5 (2022): 322. https://doi.org/10.3892/etm.2022.11251