Open Access

lncRNA GMDS‑AS1 upregulates IL‑6, TNF‑α and IL‑1β, and induces apoptosis in human monocytic THP‑1 cells via miR‑96‑5p/caspase 2 signaling

  • Authors:
    • Lei Jiang
    • Jinghui Li
  • View Affiliations

  • Published online on: January 3, 2022     https://doi.org/10.3892/mmr.2022.12583
  • Article Number: 67
  • Copyright: © Jiang 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

Long non‑coding RNA (lncRNA) is considered a crucial modulator of the initiation and progression of several diseases. However, the roles of lncRNA in sepsis have yet to be fully elucidated. Thus, the aim of the present study was to investigate the effects of the lncRNA GDP‑mannose 4,6‑dehydratase antisense 1 (GMDS‑AS1) and its target in order to understand its role in the pathogenesis of sepsis. An in vitro sepsis model was established by lipopolysaccharide (LPS) induction. Reverse transcription‑quantitative PCR analysis was applied to detect the expression of inflammatory cytokines and the levels of GMDS‑AS1, microRNA (miR)‑96‑5p and caspase‑2 (CASP2). Flow cytometry was used to quantify the rate of apoptosis. In addition, the interaction between miR‑96‑5p and CASP2 was verified using a luciferase reporter assay. Western blot analysis was performed to assess the protein levels of CASP2 following alterations in GMDS‑AS1 and miR‑96‑5p expression using transfection. The levels of interleukin (IL)‑6, tumor necrosis factor‑α and IL‑1β were increased by LPS treatment in THP‑1 cells, whereas miR‑96‑5p expression was downregulated. miR‑96‑5p overexpression inhibited LPS‑induced inflammatory responses and apoptosis. In addition, GMDS‑AS1 expression increased, and upregulation of GMDS‑AS1 inhibited, the expression of miR‑96‑5p in the in vitro sepsis model. Moreover, CASP2 was confirmed to be a direct target of miR‑96‑5p. Therefore, the lncRNA GMDS‑AS1 regulated inflammatory responses and apoptosis by modulating CASP2 and sponging miR‑96‑5p in LPS‑induced THP‑1 cells. In summary, the findings of the present study demonstrated that lncRNA GMDS‑AS1 could promote the development of sepsis by targeting miR‑96‑5p/CASP2, indicating that the GMDS‑AS1/miR‑96‑5p/CASP2 axis may be a new therapeutic target and potential research direction for sepsis therapy.

Introduction

Sepsis is a life-threatening systemic inflammatory response syndrome, which may be accompanied by multiple organ failure and septic shock in severe cases (1,2). Sepsis is a dominant cause of mortality worldwide, particularly in intensive care units, with a higher mortality rate compared with that of breast and lung cancer (3). In recent years, research had demonstrated that dysregulation of gene expression and dysfunction of the immune system are closely associated with the pathogenesis and pathophysiology of sepsis, in addition to pathogens and endotoxins (46). Furthermore, sepsis induces the excessive release of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α, as well as immunosuppression and tissue injury, thereby promoting susceptibility to secondary infections, these inflammatory cytokines contribute to aggressive immunopathology, including sepsis (7,8).

Long non-coding RNA (lncRNA) is a large class of non-protein-coding transcripts that are >200 nucleotides in length (9). It has been demonstrated that lncRNA plays various roles in numerous biological processes, such as cell proliferation, apoptosis, inflammatory and immune responses (10). Aberrant expression of lncRNA has been implicated in several inflammatory and immune diseases, including sepsis (1113). For example, lncRNA H19 functions as a competitive endogenous (ce)RNA of microRNA (miRNA/miR)-874 to regulate the progression of sepsis, both in septic patients and in animal models of sepsis (14). The lncRNA HOX transcript antisense RNA accelerates the secretion of TNF-α in mice with lipopolysaccharide (LPS)-induced sepsis (15). The lncRNA GDP-mannose 4,6-dehydratase antisense 1 (GMDS-AS1) is a novel functional lncRNA that has only been studied in lung adenocarcinoma, in which it was found to inhibit cell proliferation and induce apoptosis by targeting the miR-96-5p/CYLD axis (16). However, few studies have investigated the role of GMDS-AS1 and its mechanism of action in the progression of sepsis.

In the present study, the expression of GMDS-AS1 was examined in LPS-induced THP-1 cells. The effects of GMDS-AS1 on the production of inflammatory factors and cell apoptosis were investigated. The study explored whether GMDS-AS1 functioned as a ceRNA to regulate the expression of caspase-2 (CASP2) by sponging miR-96-5p in LPS-induced THP-1 cells. The findings may contribute to the diagnosis and treatment of sepsis in the clinical setting.

Materials and methods

Cell culture and treatment

The human monocytic leukemia THP-1 cell line was obtained from the American Type Culture Collection and cultured in RPMI 1640 medium (MilliporeSigma) supplemented with 10% FBS (HyClone; Cytiva) at 37°C in a humidified atmosphere containing 5% CO2. To mimic sepsis in vitro, THP-1 cells were stimulated with l µg/mlLPS (Sigma-Aldrich; Merck KGaA) for 24 h at 37°C.

Cell transfection

For the overexpression of GMDS-AS1 and miR-96-5p, the pcDNA3.1 vector containing full-length GMDS-AS1 (pcDNA-GMDS-AS1) and empty vector (pcDNA-NC), miR-96-5p mimics (5′-UUUGGCACAGCACAUUUUUGCUCAAAAAUGUGCUAGUGCCAAAUU-3′) and miR-NC (cat. no. 4464061) were all designed and synthesized by Thermo Fisher Scientific, Inc. In addition, the cells that were not transfected with the plasmid were used as the control group. Following stimulation with l µg/ml LPS for 24 h at 37°C, THP-1 cells were transfected with pcDNA-GMDS-AS1 or/and miR-96-5p mimic using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at a concentration of 50 ng/ml. Following transfection for 48 h at 37°C, the transfection efficiency was detected by reverse transcription-quantitative PCR (RT-qPCR).

RT-qPCR analysis

Total RNA was extracted from cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The quality of the RNA was assessed using a NanoDrop 2000 spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.) at 260 and 280 nm, according to the manufacturer's protocol. RT was performed using PrimeScript RT Master Mix (Takara Bio, Inc.) at 50°C for 45 min. qPCR was then performed with SYBR Premix EX Taq™ II (Takara Bio, Inc.) on an ABI PRISM 7300 detection system (Thermo Fisher Scientific, Inc.) with the following thermocycling conditions: Initial denaturation at 85°C for 30 sec, followed by 22 cycles at 55°C for 30 sec and 72°C for 30 sec. The results are presented by using the 2−ΔΔCq method (17). U6 was used as an internal control of miR-96-5p and GAPDH served as the internal reference of IL-6, TNF-α, IL-1β, GMDS-AS1 and CASP2. The following primer sequences were used: IL-6, forward 5′-GGAGACTTGCCTGGTGAAA-3′ and reverse, 5′-CTGGCTTGTTCCTCACTACTC-3′ and TNF-α, forward, 5′-AGCCGATGGGTTGTACCT-3′ and reverse, 5′-TGAGTTGGTCCCCCTTCT-3′; and IL-1β, forward 5′-TGTGGCAGCTACCTATGTCT-3′ and reverse, 5′-GGGAACATCACACACTAGCA′; and GMDS-AS1, forward 5′-AATGCTTTGAGGCCAAGCTA-3′ and reverse, 5′-TGGGTTCATAAGGGTTGCAT-3′; and CASP2, forward 5′-GCAAACCTCAGGGAAACATTC′ and reverse, 5′-TGTCGGCATACTGTTTCAGCA-3′; and GAPDH, forward 5′-CTGGGCTACACTGAGCACC-3′ and reverse, 5′-AAGTGGTCGTTGAGGGCAATG-3′; and U6, forward 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′.

Apoptosis analysis

Apoptosis was evaluated by flow cytometry using the FITC Annexin V/propidium iodide (PI) Apoptosis Detection Kit I (Guangzhou RiboBio Co., Ltd.). After transfection, the cells were harvested and re-suspended in binding buffer, then incubated with Annexin V-FITC and PI (10 mg/ml) for 20 min at 37°C in the dark. The samples were then placed in an ice bath and data were obtained by flow cytometer (FACSCalibur; BD Biosciences). FlowJo software (BD Biosciences; Version 7.6) was used to analyze the double-stained cells Q1, Q2 and Q3 regions represented early apoptosis rate, late apoptosis rate and dead cell rate, respectively; cell apoptosis (%)=Q1+ Q2 + Q3.

Luciferase reporter assay

The wild-type and mutant CASP2 3′-untranslated region (UTR) fragments, including putative miR-96-5p-binding sites, were synthesized and cloned into the pGL3 vector (Promega Corporation). THP-1 cells were co-transfected with the wild-type and mutant constructs and miRNA (miR-NC; cat. no. 4464061 or miR-96-5p mimic (5′-UUUGGCACAGCACAUUUUUGCUCAAAAAUGUGCUAGUGCCAAAUU-3; Thermo Fisher Scientific, Inc.) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). The relative luciferase activities were measured 48 h after transfection using a Dual-Luciferase Reporter Assay System (Promega Corporation) Renilla luciferase activity served as the internal reference.

Western blot analysis

Cells were lysed using RIPA lysis buffer (Beyotime Institute of Biotechnology), and the concentration of the proteins extracted from the cells was detected using a BCA Protein Assay kit (Beijing Dingguo Changsheng Biotechnology Co., Ltd.). A total of 20 µg protein samples were separated on 10% gels using SDS-PAGE and transferred onto PVDF membranes. After being blocked using 5% non-fat milk at room temperature for 1 h, the membranes were incubated with anti-caspase-2 primary antibody (1:500; cat. no. ab32021; Abcam) and anti-GAPDH primary antibody (1:2,500; cat. no. ab9485; Abcam) at 4°C overnight. The membranes were then incubated with a goat anti-rabbit secondary antibody (1:5,000; cat. no. ab6721; Abcam) for 1 h at room temperature. Proteins bands were visualized using an ECL reagent (Invitrogen; Thermo Fisher Scientific, Inc.). and analyzed with Image J software (version 3.0; National Institutes of Health).

Bioinformatics analysis

The Targetscan DataBase (version 7.2; targetscan.org/vert_72/) was used to screen the target genes of miR-96-5p. PCT (probability of preferentially conserved targeting) was used to evaluate the conservative targeting probability of all highly conservative miRNA families.

Statistical analysis

SPSS 23.0 software (IBM Corp.) and GraphPad Prism 6 (GraphPad Software, Inc.) were used for statistical analysis. The data are presented as the mean ± SD. Two-tailed Student's t-tests (unpaired) were used to compare the differences between two groups. Comparisons among multiple groups were performed with one-way ANOVA followed by Tukey's or Dunnett's post hoc test. P<0.05 was considered to indicate a statistically significant difference. All experiments were performed in triplicate.

Results

miR-96-5p is downregulated in an in vitro model of LPS-induced sepsis

An in vitro sepsis model was first established using LPS treatment of THP-1 cells, and the expression levels of IL-1β, IL-6 and TNF-α were detected. As shown in Fig. 1A, a significant increase in the secretion of IL-1β, IL-6 and TNF-α was observed following LPS stimulation. Additionally, miR-96-5p expression in THP-1 cells significantly decreased following induction with LPS (Fig. 1B). These results indicated that the septic cell model was successfully constructed and that miR-96-5p may be associated with the pathophysiology of sepsis.

miR-96-5p overexpression decreases LPS-induced inflammatory cytokine production and apoptosis

To explore the biological role of miR-96-5p in sepsis, a miR-96-5p mimic was transfected into THP-1 cells to upregulate miR-96-5p expression, and transfection efficiency was verified by RT-qPCR. The expression levels of miR-96-5p were significantly increased in the miR-96-5P mimic group compared with the control and miR-NC groups, demonstrating efficient transfection (Fig. 1C).

THP-1 cells were stimulated by LPS and transfected with miR-96-5p mimic or miR-NC (Fig. 2A), and the effects of miR-96-5p overexpression on the inflammatory responses and apoptosis of THP-1 cells were determined. As shown in Fig. 2B, LPS treatment significantly increased the mRNA expression of IL-6, TNF-α and IL-1β, whereas miR-96-5p mimic decreased the levels of these inflammatory cytokines. Moreover, flow cytometry revealed a significantly increased rate of apoptosis in LPS-stimulated cells, whereas the opposite results were observed in miR-96-5p-overexpressing cells (Fig. 2C). These results suggested a protective role for miR-96-5p in LPS-induced THP-1 cells.

GMDS-AS1 is highly expressed and regulates miR-96-5p expression in LPS-induced THP-1 cells

RT-qPCR showed that the expression levels of GMDS-AS1 and mir-96-5p were significantly increased after co-transfection (Fig. 3A-B). The GMDS-AS1 level was then examined in THP-1 cells following LPS stimulation. As shown in Fig. 3C, the expression of GMDS-AS1 significantly increased following LPS stimulation. A GMDS-AS1 overexpression vector was transfected into THP-1 cells, and the transfection efficiency was verified by RT-qPCR. The expression levels of GMDS-AS1 were significantly increased in the pcDNA-GMDS-AS1 group compared with those in the control and pcDNA-NC groups, suggesting that the transfection was efficient (Fig. 3D). After transfection with pcDNA-GMDS-AS1 in THP-1 cells, miR-96-5p expression was significantly downregulated compared with that in the control and pcDNA-NC groups (Fig. 3E). These data indicated that GMDS-AS1 exerted a regulatory effect on miR-96-5p expression in LPS-induced THP-1 cells.

CASP2 is the target gene of miR-96-5p

To investigate the target gene of miR-96-5p in LPS-induced THP-1 cells, bioinformatics analysis was performed using TargetScan (www.targetscan.com). CASP2 was identified as one of the targets of miR-96-5p and the binding sequence is shown in Fig. 4A. Luciferase reporter assays demonstrated that the luciferase activity of the wild-type CASP2 construct was significantly inhibited by the miR-96-5p mimic, whereas that of the mutated CASP2 was not (Fig. 4B). In addition, the mRNA expression level of CASP2 significantly decreased after overexpression of miR-96-5p (Fig. 4C). Furthermore, LPS exposure significantly increased the expression of CASP2 (Fig. 4D). These findings confirmed the interaction between CASP2 and miR-96-5p in LPS-stimulated THP-1 cells.

GMDS-AS1/miR-96-5p affects inflammatory responses and apoptosis by modulating CASP2 expression

The subsequent experiments investigated how the GMDS-AS1/miR-96-5p axis might regulate inflammatory responses and apoptosis by CASP2. CASP2 and miR-96-5p mimic were transfected into THP-1 cells, and transfection efficiency was verified by RT-qPCR. The expression of CASP2 was significantly increased in the pcDNA-CASP2 group compared with that in the control and pcDNA-NC groups, demonstrating that the transfection was efficient (Fig. 5A). Moreover, the expression of CASP2 and miR-96-5p was maintained following co-transfection with miR-96-5p mimic and pcDNA-CASP2 (Fig. 5B).

THP-1 cells were stimulated by LPS, then co-transfected with pcDNA-GMDS-AS1 or pcDNA-NC and miR-96-5p or miR-NC. As shown in Fig. 5C, western blotting results revealed that following LPS stimulation, GMDS-AS1 overexpression significantly increased the protein levels of CASP2 compared with the control and pcDNA-NC groups; however, miR-96-5p mimic transfection significantly inhibited the CASP2 levels compared with pcDNA-GMDS-AS1 + miR-NC. Moreover, GMDS-AS1 increased, whereas miR-96-5p decreased, the levels of IL-6, TNF-α and IL-1β, indicating that CASP2 overexpression accelerated the production of inflammatory factors while downregulation of CASP2 exerted the opposite effects (Fig. 5D). In addition, the apoptosis rate significantly increased following GMDS-AS1 overexpression. However, this increase was inhibited by transfection with the miR-96-5p mimic (Fig. 5E). Thus, it may be concluded that the GMDS-AS1/miR-96-5p/CASP2 axis regulates the levels of inflammatory cytokines and apoptosis in THP-1 cells following LPS exposure.

Discussion

Severe sepsis is as a life-threatening medical emergency (18). Although a standardized approach and new strategies for sepsis treatment have gradually developed, the pathogenesis of sepsis has yet to be fully elucidated (19,20). In the present study, the expression level of miR-96-5p and effects of miR-96-5p overexpression on inflammatory cytokine production and apoptosis were investigated in LPS-induced THP-1 cells. Moreover, the mechanism through which GMDS-AS1 regulates miR-96-5p and CASP2 to affect inflammatory responses and apoptosis.

miRNA participates in a variety of cellular processes, such as cell proliferation, metastasis and apoptosis (21,22). The dysregulation of miRNA contributes to the occurrence and development of multiple diseases, including sepsis (2325). miR-96-5p has been reported to be involved in several cancer types. For example, Ress et al (26) reported that miR-96-5p affected the proliferation of colorectal cancer (CRC) cells and was associated with poor survival of patients with CRC. Furthermore, miR-96-5p accelerated ovarian cancer cell proliferation and migration by targeting Caveolae1 (27). In addition, a previous study revealed that miR-96-5p expression was significantly downregulated in clinical samples from patients with sepsis (28). In the present study, the expression level of miR-96-5p was decrased in an LPS-induced THP-1 cell model, which is consistent with previous reports (29,30). Moreover, upregulation of miR-96-5p inhibited the secretion of inflammatory factors, including IL-6, TNF-α and IL-1β, as well as apoptosis.

A previous study have reported that lncRNAs may act as ceRNAs that bind to sites similar to the 3′-UTR region of mRNA to regulate biological processes (31). In this regard, in the present study, the expression of GMDS-AS1 was detected in THP-1 cells exposed to LPS, revealing higher expression of GMDS-AS1 compared with that in untreated THP-1 cells. Subsequent experiments revealed that GMDS-AS1 overexpression inhibited miR-96-5p expression, highlighting the regulatory effect of GMDS-AS1 on miR-96-5p. In addition, the target gene of miR-96-5p was also examined in order to further elucidate the mechanisms underlying sepsis. Through bioinformatics analysis, CASP2 was predicted as the target gene of miR-96-5p in sepsis, and a luciferase reporter assay verified the association between miR-96-5p and CASP2. The subsequent experiments also demonstrated that CASP2 expression was altered in LPS-treated cells and was negatively modulated by miR-96-5p.

Excessive inflammatory responses and cell apoptosis are two major characteristics of sepsis (32,33). Anti-inflammatory and anti-apoptosis strategies have been considered as effective approaches for relieving or treating sepsis (34,35). Previous studies demonstrated that certain lncRNAs and miRNAs play regulatory roles in inflammatory responses and apoptosis to slow down the development of sepsis (36,37). Yong et al (38) demonstrated that the lncRNA metastasis-associated lung adenocarcinoma transcript 1 decreased the expression of breast cancer susceptibility gene 1 and recruited zeste homolog 2 to promote skeletal muscle cell apoptosis and inflammatory response in sepsis. Lu et al (39) reported that sepsis-induced kidney injury associated transcript 1 (SIKIAT1) was highly expressed both in sepsis patients and an LPS-treated sepsis cell model. SIKIAT1 silencing repressed cell apoptosis, whereas its overexpression promoted apoptosis by regulating the miR-96/forkhead box A1 axis (39). In the present study, upregulation of GMDS-AS1 increased the CASP2 level and this effect was reversed by miR-96-5p overexpression. The protein levels of IL-6, TNF-α and IL-1β and cell apoptosis were increased after GMDS-AS1 overexpression and decreased after treatment with miR-96-5p mimic, suggesting that GMDS-AS1 and miR-96-5p jointly regulate the inflammatory response and cell apoptosis by targeting CASP2.

In summary, the present study uncovered the significance of the lncRNA GMDS-AS1 in sepsis. GMDS-AS1 was demonstrated to facilitate inflammatory responses and cell apoptosis by targeting miR-96-5p/CASP2 in LPS-induced THP-1 cells. This ceRNA mechanism may provide novel evidence and a new research direction for the clinical treatment of sepsis.

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

LJ and JL designed the experiments. LJ was involved in the collection, interpretation and analysis of the data and wrote the manuscript. JL designed the study and was involved in data collection, analysis and interpretation, as well as preparation of manuscript. LJ and JL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chaudhry H, Zhou J, Zhong Y, Ali MM, McGuire F, Nagarkatti PS and Nagarkatti M: Role of cytokines as a double-edged sword in sepsis. In vivo. 27:669–684. 2013.PubMed/NCBI

2 

Parrillo JE, Parker MM, Natanson C, Suffredini AF, Danner RL, Cunnion RE and Ognibene FP: Septic shock in humans. Advances in the understanding of pathogenesis, cardiovascular dysfunction, and therapy. Ann Intern Med. 113:227–242. 1990. View Article : Google Scholar : PubMed/NCBI

3 

Becker KL, Snider R and Nylen ES: Procalcitonin in sepsis and systemic inflammation: A harmful biomarker and a therapeutic target. Br J Pharmacol. 159:253–264. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Bickler SW and De Maio A: Dysfunction of the innate immune system during sepsis: A call for research. Crit Care Med. 41:364–365. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Delano MJ and Ward PA: The immune system's role in sepsis progression, resolution, and long-term outcome. Immunol Rev. 274:330–353. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Maslove DM and Wong HR: Gene expression profiling in sepsis: Timing, tissue, and translational considerations. Trends Mol Med. 20:204–213. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Martin GS: Sepsis, severe sepsis and septic shock: Changes in incidence, pathogens and outcomes. Expert Rev Anti Infect Ther. 10:701–706. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Wang J, Wang H, Zhu R, Liu Q, Fei J and Wang S: Anti-inflammatory activity of curcumin-loaded solid lipid nanoparticles in IL-1β transgenic mice subjected to the lipopolysaccharide-induced sepsis. Biomaterials. 53:475–483. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Li Y, Egranov SD, Yang L and Lin C: Molecular mechanisms of long noncoding RNAs-mediated cancer metastasis. Genes Chromosomes Cancer. 58:200–207. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Fang Y and Fullwood MJ: Roles, functions, and mechanisms of long Non-coding RNAs in cancer. Genomics Proteomics Bioinformatics. 14:42–54. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Dai Y, Liang Z, Li Y, Li C and Chen L: Circulating Long Noncoding RNAs as potential biomarkers of sepsis: A preliminary study. Genet Test Mol Biomarkers. 21:649–657. 2017. View Article : Google Scholar : PubMed/NCBI

12 

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. View Article : Google Scholar : PubMed/NCBI

13 

Sun L, Li L and Yan J: Progress in relationship of the long non-coding RNA and sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 29:181–183. 2017.(In Chinese). PubMed/NCBI

14 

Fang Y, Hu J, Wang Z, Zong H, Zhang L, Zhang R and Sun L: LncRNA H19 functions as an Aquaporin 1 competitive endogenous RNA to regulate microRNA-874 expression in LPS sepsis. Biomed Pharmacother. 105:1183–1191. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Zhang HJ, Wei QF, Wang SJ, Zhang HJ, Zhang XY, Geng Q, Cui YH and Wang XH: LncRNA HOTAIR alleviates rheumatoid arthritis by targeting miR-138 and inactivating NF-kappaB pathway. Int Immunopharmacol. 50:283–290. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Zhao M, Xin XF, Zhang JY, Dai W, Lv TF and Song Y: LncRNA GMDS-AS1 inhibits lung adenocarcinoma development by regulating miR-96-5p/CYLD signaling. Cancer Med. 9:1196–1208. 2020. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Jain S: Sepsis: An update on current practices in diagnosis and management. Am J Med Sci. 356:277–286. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Rello J, Valenzuela-Sanchez F, Ruiz-Rodriguez M and Moyano S: Sepsis: A review of advances in management. Adv Ther. 34:2393–2411. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Hamers L, Kox M and Pickkers P: Sepsis-induced immunoparalysis: Mechanisms, markers, and treatment options. Minerva Anestesiol. 81:426–439. 2015.PubMed/NCBI

21 

Liz J and Esteller M: lncRNAs and microRNAs with a role in cancer development. Biochim Biophys Acta. 1859:169–176. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Varshney J and Subramanian S: MicroRNAs as potential target in human bone and soft tissue sarcoma therapeutics. Front Mol Biosci. 2:312015. View Article : Google Scholar : PubMed/NCBI

23 

Fu D, Dong J, Li P, Tang C, Cheng W, Xu Z, Zhou W, Ge J, Xia C and Zhang Z: MiRNA-21 has effects to protect kidney injury induced by sepsis. Biomed Pharmacother. 94:1138–1144. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Ge C, Liu J and Dong S: MiRNA-214 protects sepsis-induced myocardial injury. Shock. 50:112–118. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Wang Z, Ruan Z, Mao Y, Dong W, Zhang Y, Yin N and Jiang L: MiR-27a is up regulated and promotes inflammatory response in sepsis. Cell Immunol. 290:190–195. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Ress AL, Stiegelbauer V, Winter E, Schwarzenbacher D, Kiesslich T, Lax S, Jahn S, Deutsch A, Bauernhofer T, Ling H, et al: MiR-96-5p influences cellular growth and is associated with poor survival in colorectal cancer patients. Mol Carcinog. 54:1442–1450. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Liu B, Zhang J and Yang D: MiR-96-5p promotes the proliferation and migration of ovarian cancer cells by suppressing Caveolae1. J Ovarian Res. 12:572019. View Article : Google Scholar : PubMed/NCBI

28 

Chen J, Jiang S, Cao Y and Yang Y: Altered miRNAs expression profiles and modulation of immune response genes and proteins during neonatal sepsis. J Clin Immunol. 34:340–348. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Cheng Q, Tang L and Wang Y: Regulatory role of miRNA-26a in neonatal sepsis. Exp Ther Med. 16:4836–4842. 2018.PubMed/NCBI

30 

How CK, Hou SK, Shih HC, Huang MS, Chiou SH, Lee CH and Juan CC: Expression profile of MicroRNAs in gram-negative bacterial sepsis. Shock. 43:121–127. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Hotchkiss RS and Nicholson DW: Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol. 6:813–822. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Gotts JE and Matthay MA: Sepsis: Pathophysiology and clinical management. BMJ. 353:i15852016. View Article : Google Scholar : PubMed/NCBI

34 

Matsuda A, Jacob A, Wu R, Aziz M, Yang WL, Matsutani T, Suzuki H, Furukawa K, Uchida E and Wang P: Novel therapeutic targets for sepsis: Regulation of exaggerated inflammatory responses. J Nippon Med Sch. 79:4–18. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Oberholzer C, Oberholzer A, Clare-Salzler M and Moldawer LL: Apoptosis in sepsis: A new target for therapeutic exploration. FASEB J. 15:879–892. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Jia Y, Li Z, Cai W, Xiao D, Han S, Han F, Bai X, Wang K, Liu Y, Li X, et al: SIRT1 regulates inflammation response of macrophages in sepsis mediated by long noncoding RNA. Biochim Biophys Acta Mol Basis Dis. 1864:784–792. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Zheng D, Yu Y, Li M, Wang G, Chen R, Fan GC, Martin C, Xiong S and Peng T: Inhibition of MicroRNA 195 prevents apoptosis and multiple-organ injury in mouse models of sepsis. J Infect Dis. 213:1661–1670. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Yong H, Wu G, Chen J, Liu X, Bai Y, Tang N, Liu L and Wei J: lncRNA MALAT1 accelerates skeletal muscle cell apoptosis and inflammatory response in sepsis by decreasing BRCA1 expression by recruiting EZH2. Mol Ther Nucleic Acids. 19:97–108. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Lu S, Wu H, Xu J, He Z, Li H and Ning C: SIKIAT1/miR-96/FOXA1 axis regulates sepsis-induced kidney injury through induction of apoptosis. Inflamm Res. 69:645–656. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2022
Volume 25 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang L and Jiang L: lncRNA GMDS‑AS1 upregulates IL‑6, TNF‑α and IL‑1β, and induces apoptosis in human monocytic THP‑1 cells via miR‑96‑5p/caspase 2 signaling. Mol Med Rep 25: 67, 2022
APA
Jiang, L., & Jiang, L. (2022). lncRNA GMDS‑AS1 upregulates IL‑6, TNF‑α and IL‑1β, and induces apoptosis in human monocytic THP‑1 cells via miR‑96‑5p/caspase 2 signaling. Molecular Medicine Reports, 25, 67. https://doi.org/10.3892/mmr.2022.12583
MLA
Jiang, L., Li, J."lncRNA GMDS‑AS1 upregulates IL‑6, TNF‑α and IL‑1β, and induces apoptosis in human monocytic THP‑1 cells via miR‑96‑5p/caspase 2 signaling". Molecular Medicine Reports 25.2 (2022): 67.
Chicago
Jiang, L., Li, J."lncRNA GMDS‑AS1 upregulates IL‑6, TNF‑α and IL‑1β, and induces apoptosis in human monocytic THP‑1 cells via miR‑96‑5p/caspase 2 signaling". Molecular Medicine Reports 25, no. 2 (2022): 67. https://doi.org/10.3892/mmr.2022.12583