Academic Editor

Article Metrics

  • Fig. 1.

    View in Article
    Full Image
  • Fig. 2.

    View in Article
    Full Image
  • Fig. 3.

    View in Article
    Full Image
  • Fig. 4.

    View in Article
    Full Image
  • Fig. 5.

    View in Article
    Full Image
  • Information

  • Download

  • Contents

Abstract

Background:

LncRNA taurine-upregulated gene 1 (TUG1) can regulate vascular endothelial cell injury, a critical mechanism in treating hemorrhagic shock and fluid resuscitation (HS/R). Therefore, this study explored the influence of TUG1 in HS/R.

Methods:

An in vivo rat model of ischemia-reperfusion (I/R) injury post-HS/R and an in vitro model of oxidative stress injury in rat cardiomyocyte cell line (H9C2) were constructed. In vivo, we silenced TUG1 and quantified its expression along with inflammatory factors through quantitative reverse transcription polymerase chain reaction (qRT-PCR), mean arterial pressure (MAP) detection and blood gas analysis. Myocardial functional impairment was assessed via Triphenyl-2H-Tetrazolium Chloride (TTC), Hematoxylin and eosin, and Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) stainings. Oxidative stress level in rat serum was measured. In vitro, we examined the changes of cell viability, apoptosis, oxidative stress levels, inflammatory factor secretion and nuclear factor-κB (NF-κB)/p65 expression by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, Enzyme-linked immunosorbent assay (ELISA) and Western blot.

Results:

TUG1 level was elevated in rats of I/R model caused by HS/R. TUG1 silencing ameliorated the decline in MAP, acid-base imbalance and myocardial tissue damage, and suppressed oxidative stress and inflammatory factor levels in model rat. TUG1 silencing enhanced viability, impeded apoptosis, and reduced oxidative stress, inflammatory factor contents and NF-κB/p65 expression in H2O2 treated H9C2 cells.

Conclusion:

TUG1 participates in regulating oxidative stress damage and inflammation induced by HS/R.

1. Introduction

Hemorrhagic shock (HS) refers to the pathophysiological process of acute blood or serum loss caused by various reasons, resulting in reduction of effective circulating volume and cardiac output, insufficient tissue perfusion, metabolic acidosis and functional impairment, and systemic inflammation [1, 2, 3]. Despite the major advances in resuscitation treatment and intensive care, HS still threatens the life of trauma patients worldwide [4]. Fluid resuscitation can effectively mitigate severe HS by ensuring the blood perfusion of important organs in a short time [5]. However, recent clinical study has found without effective hemostasis, large-dose fluid resuscitation can produce many adverse reactions, such as weakened tissue oxygen supply resulting from reduced hemoglobin concentration, coagulation dysfunction, reperfusion injury, and immune dysfunction [6]. Notably, hemorrhagic shock and fluid resuscitation (HS/R) may trigger oxidative stress damage [7, 8]. Moreover, reactive oxygen species (ROS) activates activating nuclear factor-κB (NF-κB), and the elevation of NF-κB causes systemic inflammation by increasing the expression of cytokines [9]; hence, alleviating the oxidative stress and inflammatory response can help improve the effectiveness of fluid resuscitation.

Reportedly, long non-coding RNA (lncRNA) greatly impacts the process of ischemia/reperfusion injury [10]. By high-throughput RNA sequencing analysis, Lin et al. [11] discovered 851 significantly up-regulated lncRNAs and 1533 obviously down-regulated lncRNAs in rat heart tissues with severe HS-induced ischemia-reperfusion (I/R) injury. LncRNA taurine-upregulated gene 1 (TUG1) is located on chromosome 22q12.2, and has many biological functions. Sun et al. [12] reported that TUG1 can alleviate renal ischemia/reperfusion injury by regulating ferroptosis. Wang et al. [13] revealed that TUG1 can be mediated by hypoxia inducible factor 1 subunit alpha (HIF-1α) and promote the occurrence of myocardial infarction.

Since TUG1 acts a part in HS/R with an obscure mechanism, this research illustrated the influence of TUG1 on HS/R.

2. Materials and Methods
2.1 Experimental Animals

36 Sprague–Dawley rats (male, 8–9 weeks old, 300–360 g; Hangzhou Medical College, Hangzhou, China) were housed under specific pathogen-free conditions at animal laboratory (20 ± 2 °C, 12-h circadian rhythm) with unlimited supply of food and sterile tap water. Additionally, research ratification was obtained from Zhejiang Baiyue Biotech Co., Ltd.’s Ethics Committee for Experimental Animals Welfare (ZJBYLA-IACUC-20220427).

2.2 Establishment of I/R Model Caused by HS/R

Establishment of severe HS rat model referred to an existing study [11]. After being fasted for 12 h, the rats were anesthetized by intraperitoneal injection of 45 mg/kg sodium pentobarbital (P-010, Merck, Darmstadt, Germany), the femoral artery of which was separated and cannulated with polyethylene catheter. Subsequently, their left femoral artery was connected to a multi-channel physiological signal acquisition and processing system (BIOPAC, Goleta, CA, USA) to detect blood pressure (mean arterial pressure, MAP) and heart rate, while the right femoral artery was employed to control hemorrhage. The left jugular vein was cannulated with a PE-50 tube for fluid resuscitation. The total blood loss rate was controlled at 45%, and the bleeding time was controlled within 1 h. The blood flowing out of the left femoral artery was then perfused back into the rats within 40 min.

Based on previous description [14, 15], rats were given subcutaneous injection of phosphate buffer saline (PBS) (A610100, Sangon, Shanghai, China) containing small interfering RNA targeting TUG1 (siTUG1, 5-GCAGUAAUUGGAGUGGAUATT-3, siBDM0001, Ribobio, Guangzhou, China) or its negative control (siNC, 5-UUCUCCGAACGUGUCACGUTT-3, siN0000001-4-200, Ribobio, Guangzhou, China) 2 h before surgery. After 6 h of resuscitation, the collected blood samples were centrifuged (3000 g, 15 min), followed by serum storage (–80 °C). The rats were euthanized by an overdose of pentobarbital sodium (200 mg/kg, iv) for heart tissue collection.

Six groups were designed based on experimental animals (Sham, Model, Sham + siNC, Sham + siTUG1, Model + siNC and Model + siTUG1) (6 mice/group). The rats in Sham group experienced anesthesia and surgery, but without bloodletting.

2.3 Blood Gas Analysis

Anticoagulated whole blood samples were collected 2 hours after resuscitation, in which bicarbonate (HCO3) and lactate content were determined employing a Hemogas analyzer (ABL800 FLEX, Radiometer, Copenhagen, Denmark).

2.4 Triphenyl-2H-Tetrazolium Chloride (TTC) Staining

The heart was quickly taken out, and repeatedly rinsed with normal saline. Heart tissue was immediately frozen in a –20 °C refrigerator for 20 min, and then cut into 4–5 sections (thickness of 2 mm). Sections were stained with 1.5% TTC (15 min; A610558, Sangon, Shanghai, China), and fixed with 10% neutral formaldehyde solution (E672001, Sangon, Shanghai, China). After the stained sections were imaged under a fluorescence microscope, their infarct area (white area) and non-infarcted region (red area) were analyzed and calculated through AlphaEaseFC image processing software (version 4.0, Alpha Innotech, San Leandro, CA, USA). Infarct size (%) = infarct area/total area × 100%.

2.5 Hematoxylin and Eosin (H&E) Staining

Heart tissues experienced fixation (10% neutral formalin solution, BL-G001, Senbeijia, Nanjing, China), paraffin embedding and sectioning into 5-µm-thick slices. The deparaffinized and hydrated slices underwent color development exploiting H&E kit (BP-DL001, Senbeijia, China), followed by dehydration and transparentization. After sealing with mounting medium (SBJ-0700, Senbeijia, China), the degree of damage to the rat myocardial tissue was observed (microscope, AE2000, Motic, Xiamen, China, 100×).

2.6 Terminal Deoxynucleotidyl Transferase-Mediated Deoxyuridine Triphosphate Nick End Labeling (TUNEL) Staining

TUNEL Apoptosis Detection Kit (C1086, Beyotime, Shanghai, China) was used for TUNEL staining of heart tissue. After being deparaffinized, hydrated, treated with proteinase K and washed, the section was incubated with TUNEL solution (60 min), and reacted with 4,6-Diamidino-2-phenylindole (DAPI) solution (C1002, Beyotime, China) for nuclear staining. Following dehydration, transparentization and mounting, TUNEL-positive cells were finally observed (fluorescence microscope, 200×, DM2500, Leica, Wetzlar, Germany). Relative positive cell rate (%) = positive TUNEL staining/total cell × 100%. Data were normalized to Sham + siNC group.

2.7 Detection of Malondialdehyde (MDA), Glutathione (GSH) and Superoxide Dismutase (SOD)

MDA content in rat serum was determined by MDA detection kit (SBJ-R0007, Senbeijia, China). Briefly, the standard solution was diluted into different concentrations in preparation for the subsequent standard curve plotting. Afterwards, the diluted serum was added to an antibody-coated 96-well plate, and cultured with different reagents. Following absorbance measurement (450 nm, microplate reader, CMaxPlus, Molecular Devices, San Jose, CA, USA), MDA concentration detection was carried out based on the standard curve.

By means of GSH detection kit (BC1175, Solarbio, Beijing, China), the absorbance (412 nm) of the sample or reagent was detected, and GSH content in rat serum was calculated according to the standard curve.

Using SOD detection kit (BC0170, Solarbio, China), the absorbance (560 nm) of the sample or reagent was quantified, followed by measurement of SOD content in rat serum or cell supernatant.

2.8 Cell Culture

Rat cardiomyocyte cell line (H9C2, CL-0089, Procell, Wuhan, China) were soaked (37 °C, 5% CO2) in Dulbecco’s Modified Eagle’s Medium (30-2002, ATCC, Manassas, VA, USA) with 10% fetal bovine serum, 0.1 mg/mL streptomycin and 100 U/mL penicillin. H9C2 cells were tested negative for mycoplasma. H9C2 cells were validated by short tandem repeat (STR) profiling.

2.9 Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR)

Post total RNA extraction (total RNA extraction kit, EZB-RN4, HiFunBio, Shanghai, China) from rat heart tissues or H9C2 cells, reverse transcription and qPCR were completed by One-Step qRT-PCR kit (AE341, TransGen Biotech, Beijing, China) using Real-Time PCR Detection system (CFX Connect, Bio-rad, Hercules, CA, USA). Data were dissected by 2-Δ⁢Δ⁢Ct method [16]. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH, for lncRNA and mRNA) functioned as an internal control. The details of primers are listed in Table 1.

Table 1. Primers used in this study.
List of oligonucleotide sequences 5 3
Primers for PCR
R-TUG1 F CTGCTGAAGTTGTTTGCCTGCTTAC
R-TUG1 R AATTGGGCACGAGAGGCTGAAAG
R-GAPDH F TGCCACTCAGAAGACTGTGG
R-GAPDH R TTCAGCTCTGGGATGACCTT
R-TNF-α F GTCGTAGCAAACCACCAAG
R-TNF-α R GTCGCCTCACAGAGCAAT
R-IL-6 F GAGTTGTGCAATGGCAATTC
R-IL-6 R ACTCCAGAAGACCAGAGCAG

Abbreviation: F, Forward; R, Reverse; PCR, polymerase chain reaction; TUG1, taurine-upregulated gene 1; GAPDH, Glyceraldehyde-3-phosphate dehydrogenase; TNF-α, Tumor necrosis factor-α; IL-6, Interleukin-6.

2.10 Transfection and Treatment

SiTUG1 (siG151012045709-1-5, 5-GCAGUAAUUGGAGUGGAUATT-3) and siNC (siN0000002-1-5, 5-UUCUCCGAACGUGUCACGUTT-3) were purchased from Ribobio (China). The oligonucleotide sequences above were transfected into the cells in a 6-well plate (4 × 106/well) with the help of transfection reagent (STF02, SinoBiological, Beijing, China). After 24 h, the transfection results were determined by qRT-PCR.

In line with a previous description [17], an oxidative stress injury cell model was set up based on 500 µM H2O2 treated H9C2 (24 h). H9C2 were assigned into 4 groups: Control (normal culture), H2O2, H2O2 + siNC and H2O2 + siTUG1 groups (4-h treatment with 400 µM H2O2 after siNC/siTUG1 transfection or not).

2.11 Cell Viability

With 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay kit (SBJ-0191, Senbeijia, China), treated cells (3 × 103 per well) were first reacted with 10 µL MTT solution (4 h), and crystal dissolution in each well was realized by 110 µL Formazan solution. Absorbance (490 nm) determination was conducted using multiple detection readers to calculate the relative cell viability. Relative cell viability (%) = [optical density (OD) (experiment) – OD (blank)]/[OD (control) – OD (blank)] × 100%. Data were normalized to Control group.

2.12 Cell Apoptosis

Using an Annexin V-FITC Apoptosis Detection Kit (CA1020, Solarbio, China), treated cells (1 × 106) were first re-suspended with binding buffer, followed by incubation of suspension with 5 µL Annexin V-fluorescein isothiocyanate (room temperature, 10 min, darkness) and 5 µL propidium iodide under the same conditions (5 min). The volume of cell suspension was supplemented to 500 µL with PBS, and then the apoptosis was detected by a flow cytometer (Accuri C6, BD Biosciences, San Jose, CA, USA). Apoptosis rate (%) = Early apoptotic cells (%) + advanced apoptotic cells (%).

2.13 Measurement of Lactate Dehydrogenase (LDH) Release

LDH release in different groups was evaluated by LDH Cytotoxicity Assay Kit (C0017, Beyotime, China). After different cell treatments, cells in each well were cultivated with 60 µL LDH detection working solution (30 min). Absorbance (490 nm) was detected with a multiple detection reader for the calculation of LDH content. LDH (U/mL) = (sample hole absorbance – background blank control hole absorbance)/(standard tube absorbance – standard blank tube absorbance) × standard product concentration (U/mL).

2.14 Enzyme-Linked Immunosorbent Assay (ELISA)

Tumor necrosis factor (TNF)-α and Interleukin (IL)-6 in cell supernatant were measured by TNF-α and IL-6 ELISA kits (SBJ-H0038, SBJ-H0465, Senbeijia, China). More precisely, cell supernatant was diluted, and standard dilutions were prepared at the same time. The diluted sample or standard was incubated (90 min) in a 96-well plate, followed by addition of different reagents. Absorbance (450 nm) measurement was performed employing a microplate reader. According to the abscissa (standard concentration) and the ordinate (OD value), corresponding concentration was measured from the standard curve in line with the OD value of the sample and then multiplied by the dilution multiple to obtain the actual concentration of the sample.

2.15 Western Blot

Total protein extracted (ExKine™ Total Protein Extraction Kit, KTP3006, Abbkine, Wuhan, China) [18] experienced quantification, separation, electrophoresis, transference to membrane and blocking (5% bovine serum albumin, BSA). The membrane was probed with the primary antibodies (GAPDH as an internal control), rinsed off, and reacted with secondary antibodies. The details of antibodies are exhibited in Table 2. Protein band visualization was conducted by enhanced chemiluminescence (ECL) Western Blotting Substrate (W028-2-1, Nanjing Jiancheng Bioengineering Institute, Nanjing, China) on a gel imaging system (610020-9q, QinXiang, Shanghai, China), after which the band intensity was calculated by ImageJ2x V2.1.4.7 (Rawak Software, Stuttgart, Germany). Relative protein expression levels = (gray value of protein band)/(gray value of GAPDH band). Data were normalized to Control group.

Table 2. Antibodies used in this study.
Antibody Catalog Molecular weight (kDa) Dilution Manufacturer (UK)
NF-κB p65 (phospho S536) ab239882 65 1/1000 abcam
NF-κB p65 ab16502 64 1/2000 abcam
GAPDH ab8245 37 1/5000 abcam
Goat anti rabbit ab205718 1/10,000 abcam
Goat anti mouse ab6789 1/10,000 abcam

NF-κB, nuclear factor-κB.

2.16 Statistical Analyses

Data represented as mean ± standard deviation were dissected by Graph Prism v8.0 (GraphPad software, San Diego, CA, USA). TUG1 expression between Sham and model groups was analyzed by independent sample t test. One-way analysis of variance (ANOVA) was leveraged for multi-group comparison. p < 0.05 was indicative of statistical noteworthy.

3. Results
3.1 TUG1 Silencing Improved the Decrease of MAP and Acid-Base Imbalance of HS/R Model Rat

TUG1 expression level in rat heart tissue was much higher in model group than Sham group (Fig. 1A, p < 0.001). qRT-PCR results showed TUG1 downregulation after transfection with siTUG1, indicating that the transfection was successful (Fig. 1B, p < 0.001). As shown in Fig. 1C, the MAP of I/R model rat was decreased significantly at maximum bleed out (MBO), start of resuscitation, and 1 and 2 h after the end of resuscitation, which was improved at the end of resuscitation, 1 and 2 hours after the end of resuscitation following siTUG1 transfection (Fig. 1C, p < 0.05). In addition, the decrease of blood actual bicarbonate (HCO3) level and the increase of lactate level in rats after HS/R induction were restored to normal levels by siTUG1 treatment (Fig. 1D,E, p < 0.001), denoting that siTUG1 improved the HS/R-caused acid-base imbalance.

Fig. 1.

Effects of TUG1 silencing on MAP and metabolic acidosis in rat model with HS/R. Construction of 6 groups based on experimental animals (6 mice/group): Sham (receiving anesthesia and surgery without bloodletting); Model (receiving anesthesia, surgery and bloodletting); Sham + siNC (receiving subcutaneous injection of siNC 2 h before anesthesia and surgery without bloodletting); Sham + siTUG1 (receiving subcutaneous injection of siTUG1 2 h before anesthesia and surgery without bloodletting); Model + siNC (receiving subcutaneous injection of siNC 2 h before anesthesia, surgery and bloodletting); Model + siTUG1groups (receiving subcutaneous injection of siTUG1 2 h before surgery anesthesia, surgery and bloodletting). (A) TUG1 expression in myocardial tissue of rats in Sham and Model group (n = 6) (qRT-PCR, GAPDH as internal control). (B) TUG1 expression in myocardial tissue after transfected with siTUG1 (qRT-PCR, GAPDH as internal control). (C) MAP at MBO, at the start of resuscitation, at the end of resuscitation, and 1 and 2 h post resuscitation. (D,E) Actual bicarbonate (HCO3) and lactate levels in rat blood samples (commercially available assay kits). *p < 0.05, **p < 0.01, *⁣**p < 0.001. The experiments were independently repeated at least thrice. n = 3. siTUG1, small interfering RNA targeting TUG1; HS, hemorrhagic shock; HS/R, hemorrhagic shock and fluid resuscitation; qRT-PCR, quantitative reverse transcription polymerase chain reaction; GAPDH, Glyceraldehyde-3-phosphate dehydrogenase; NC, negative control; MAP, mean arterial pressure; MBO, maximum bleed out.

3.2 TUG1 Silencing Attenuated HS/R-Induced Heart Damage, Cell Apoptosis, Oxidative Stress and Inflammation in Rats

TTC staining results suggested the area of myocardial infarction in the Model + siNC group was increased relative to Sham + siNC group, which however was then reduced by silencing TUG1 (Fig. 2A,B, p < 0.001). Meanwhile, H&E staining data were presented at Fig. 2C, based on which we could observe that cardiomyocytes were irregularly arranged, the myocardial gap was widened, the muscle fibers were broken in Model + siNC group, as compared with Sham + siNC group. Besides, TUG1 silencing improved the myocardial injury of the model group, cardiomyocytes were irregularly arranged, however, the space between cardiomyocytes was narrowed. As shown in Fig. 3A,B, TUNEL-positive cell rate in Model + siNC group was significantly increased, which was reversed by siTUG1 (Fig. 3A,B, p < 0.001). Afterwards, we assessed MDA, GSH and SOD levels in rat serum, and discovered that MDA level was higher but GSH and SOD levels were lower in Model + siNC group than Sham + siNC group (Fig. 3C,D, p < 0.001). However, TUG1 silencing offset the effect of modeling on MDA, GSH and SOD levels (Fig. 3C,D, p < 0.01). Similarly, the upregulation in inflammatory factors TNF-α and IL-6 in model rats was also suppressed by siTUG1 (Fig. 3E, p < 0.001).

Fig. 2.

Effect of TUG1 silencing on myocardial injury in rat model with HS/R. Construction of 4 groups based on experimental animals (6 mice/group) (Sham + siNC, Sham + siTUG1, Model + siNC and Model + siTUG1). The rats were given subcutaneous injection of phosphate buffer saline containing siTUG1 or siNC 2 h before surgery and then underwent Sham or HS/R operation. (A,B) Myocardial infarction size of rats (TTC staining). (C) The myocardial injury of rats (H&E staining, 100× magnification). Scale bars, 50 µm. Arrow in Model + siNC group: cardiomyocytes were irregularly arranged, the myocardial gap was widened, the muscle fibers were broken; arrow in Model + siTUG1 group: cardiomyocytes were irregularly arranged, the space between cardiomyocytes was narrowed. *⁣**p < 0.001. The experiments were independently repeated at least thrice. n = 3. TTC, Triphenyl-2H-Tetrazolium Chloride; H&E, Hematoxylin and eosin.

Fig. 3.

Effects of TUG1 silencing on apoptosis, oxidative stress and inflammatory factor expressions of myocardial tissue in rat model with HS/R. Construction of 4 groups based on experimental animals (6 mice/group) (Sham + siNC, Sham + siTUG1, Model + siNC and Model + siTUG1). (A,B) The apoptosis of myocardial tissue (TUNEL staining) (magnification, 200×). Scale bars, 50 µm. (C,D) MDA, GSH and SOD levels in rat serum (corresponding kits). (E) TNF-α and IL-6 levels in rat myocardial tissue (qRT-PCR). **p < 0.01, *⁣**p < 0.001. The experiments were independently repeated at least thrice. n = 3. TUNEL, Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling; MDA, malondialdehyde; GSH, glutathione; SOD, superoxide dismutase; TNF-α, Tumor necrosis factor-α; IL-6, Interleukin-6.

3.3 TUG1 Silencing Regulated the Cell Viability, Apoptosis, Oxidative Stress, Inflammation and NF-κB/p65 Expression of H2O2-Treated H9C2 Cells

To explore whether TUG1 mediated oxidative stress of H9C2, we transfected siTUG1 into H9C2 cells and determined the transfection efficiency by qRT-PCR (Fig. 4A, p < 0.001). In H2O2-treated H9C2, TUG1 level was increased greatly (Fig. 4B, p < 0.001). Interestingly, the H2O2-induced upregulation of TUG1 can be abrogated by siTUG1 (Fig. 4B, p < 0.001).

Fig. 4.

TUG1 silencing counteracted the influences of H2O2 on viability and apoptosis of H9C2 cells. H9C2 cells were assigned into 6 groups: Control (normal culture); siNC (transfection with siNC); siTUG1 (transfection with siTUG1); H2O2 (4-h 400 µM H2O2 treatment); H2O2 + siNC (transfection with siNC and 4-h 400 µM H2O2 treatment); H2O2 + siTUG1 (transfection with siTUG1 and 4-h 400 µM H2O2 treatment). (A) TUG1 expression in H9C2 cells transfected with siTUG1 or siNC (qRT-PCR, GAPDH as internal control). *⁣**p < 0.001. (B) TUG1expression in H9C2 cells treated with H2O2 and transfected with siTUG1/siNC (qRT-PCR, GAPDH as internal control). (C) viability of H9C2 cells treated with H2O2 and transfected with siTUG1/siNC (MTT). (D,E) Apoptosis rates of H9C2 cells treated with H2O2 and transfected with siTUG1/siNC (flow cytometry). **p < 0.01, *⁣**p < 0.001. The experiments were independently repeated at least thrice. n = 3. MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.

H2O2 treatment attenuated cell viability (Fig. 4C, p <0.001) and facilitated apoptosis (Fig. 4D,E, p < 0.001). Further, H2O2treatment promoted the secretion of LDH (Fig. 5A, p < 0.001), decreased SOD level (Fig. 5B, p < 0.001), and elevated TNF-α and IL-6 levels (Fig. 5C,D, p < 0.001). However, these effects were reversed by siTUG1 (Fig. 4C–E and Fig. 5A–D, p < 0.01). We also found that H2O2 can induce NF-κB/p65 expression in H9C2 cells, which can be counteracted by TUG1 silencing (Fig. 5E,F, p < 0.001). The same results also were observed in p-NF-κB/NF-κB ratio (Fig. 5G, p < 0.001).

Fig. 5.

TUG1 silencing reversed the influences of H2O2 on oxidative stress, inflammation and NF-κB pathway of H9C2 cells. H9C2 cells were divided into 4 groups (Control; H2O2; H2O2 + siNC; H2O2 + siTUG1). (A,B) LDH and SOD levels in cell supernatant (corresponding kits). (C,D) TNF-α and IL-6 contents in cell supernatant (ELISA). (E–G) p-NF-κB/p65 and NF-κB/p65 protein levels in H9C2 cells treated with H2O2 and transfected with siTUG1/siNC (Western blot, GAPDH as internal control). **p < 0.01, *⁣**p < 0.001. The experiments were independently repeated at least thrice. n = 3. LDH, lactate dehydrogenase.

4. Discussion

Severe HS/R can produce I/R injury in the ischemic tissue, manifested as a large number of free radicals and excessive inflammatory factors. In addition to restoring tissue perfusion and improving oxygen supply, the mass fluid resuscitation also can reduce oxidative stress and inflammation [19]. Our study clarified that TUG1 participated in the regulation of oxidative stress and inflammation caused by HS/R, and is of great significance for potentiating the effectiveness of fluid resuscitation.

We first constructed I/R rat model by HS/R, and found more TUG1 in model group than Sham group, hinting that TUG1 may impact HS. TUG1 silencing mitigated MAP reduction after resuscitation. The proper pH value of various body fluids in organisms is one of the important conditions for maintaining normal physiological activities. After treatment with siTUG1, the acid-base imbalance in the blood of model rat was improved. HS may cause damage to myocardial function due to the reduction of effective circulating blood volume and insufficient tissue perfusion [20]. In our study, we observed that following TUG1 silencing, the area of myocardial infarction, the degree of myocardial tissue damage and the level of apoptosis were all effectively alleviated of model rats. SOD, a vital antioxidant enzyme in the organism, can scavenge oxygen free radicals and repress oxidative stress [21]. MDA is the end product of lipid oxidation, the level of which can be upregulated by the rising free radicals [22]. GSH is a common antioxidant [23]. TNF-α and IL-6 are common inflammatory factors, and inhibiting their secretion can prevent myocardial I/R injury [24]. Moreover, Xu et al. [25] reported that TUG1 silencing can improve the inflammation and apoptosis of I/R-induced renal tubular epithelial cells. TUG1 mediating oxidative stress has also been demonstrated. Knocking down TUG1 prevents oxidative stress-induced damage in lens epithelial cells [26]. In this study, we found that TUG1 silencing can suppress MDA, TNF-α and IL-6 levels and elevate GSH and SOD levels, implying that TUG1 silencing attenuated the oxidative stress-induced damage and inflammation caused by HS.

NF-κB signaling pathway is a classic inflammatory signal transduction pathway [27], inhibition of which can prevent HS-induced heart injury [28]. In addition, Yan et al. [29] revealed that NF-κB inhibitors can dampen inflammatory reaction at different stages of HS/R, thus alleviating tissue damage. Previous studies also reported the regulatory effect of TUG1 on NF-κB. Jia et al. [30] found TUG1 downregulation can inhibit NF-κB pathway, thus alleviating the inflammatory injury resulting from spinal cord I/R. Yue et al. [31] confirmed that TUG1 downregulation can inhibit NF-κB pathway to block the development of multiple sclerosis. Herein, H2O2-induced p-NF-κB/p65 increase was abrogated by TUG1 silencing, indicating that TUG1 silencing might inhibit NF-κB signal transduction pathway to alleviate tissue damage.

Taken together, TUG1 acts as an ideal and novel target for diagnosis and treatment of hemorrhagic shock and HS/R. However, the absence of experiments (NF-κB inhibitory genes) to verify the NF-κB pathway activation is one of the defects of this study. In addition, clinical studies need to be designed to re-validate the diagnostic potential of TUG1 as biomarkers.

5. Conclusion

To sum up, we corroborate TUG1 participates in mediating impaired cardiac function, oxidative stress damage and inflammatory response caused by HS/R, providing a new target for clinical treatment of HS/R.

Availability of Data and Materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Author Contributions

Substantial contributions to conception and design: WL, HYC. Data acquisition, data analysis and interpretation: XLZ, MRL. Drafting the article or critically revising it for important intellectual content: All authors. Final approval of the version to be published: All authors. Agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of the work are appropriately investigated and resolved: All authors.

Ethics Approval and Consent to Participate

Our research was approved by Zhejiang Baiyue Biotech Co., Ltd.’s Ethics Committee for Experimental Animals Welfare (ZJBYLA-IACUC-20220427).

Acknowledgment

Not applicable.

Funding

This work was supported by the Special Project of Fujian National Clinical Research Base of Traditional Chinese Medicine (JDZX201902).

Conflict of Interest

The authors declare no conflict of interest.

References

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.