HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury

  • Authors:
    • Jun‑Jun Huang
    • Jie Xia
    • Li‑Li Huang
    • Ya‑Chun Li
  • View Affiliations

  • Published online on: August 8, 2019     https://doi.org/10.3892/mmr.2019.10575
  • Pages: 3424-3432
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The inflammatory response is one of the most important factors in the occurrence and development of acute lung injury (ALI). Hypoxia‑inducible factor‑1α (HIF‑1α) and the NOD‑like receptor 3 (NLRP3) inflammasome have been demonstrated to serve an important role in the pathogenesis of ALI. The objective of the present study was to investigate whether HIF‑1α could regulate activation of the NLRP3 inflammasome and its potential function and specific mechanism in bleomycin (BLM)‑induced ALI. Activation of the NLRP3 inflammasome and secretion of IL‑1β were detected following silencing of HIF‑1α or NF‑κB, respectively, in BLM‑treated A549 and RLE‑6TN cells. The results demonstrated that the NLRP3 inflammasome could be activated after BLM treatment. HIF‑1α and NF‑κB expression significantly increased in the BLM group. The levels of NF‑κB‑ and NLRP3 inflammasome‑associated proteins, including NLRP3, apoptosis‑associated speck‑like protein containing CARD and caspase‑1, markedly decreased after treating A549 and RLE‑6TN cells with HIF‑1α small interfering RNA. Activation of the NLRP3 inflammasome was also inhibited after silencing NF‑κB. Furthermore, the levels of IL‑1β markedly decreased in the cellular culture supernatants following inhibition of HIF‑1α and NF‑κB. Therefore, the present study indicated that HIF‑1α could modulate the activation of the NLRP3 inflammasome and the secretion of IL‑1β through NF‑κB signaling in BLM‑induced ALI. The current results improve understanding of the mechanism of ALI and may provide new ideas for identifying therapeutic targets of ALI.

Introduction

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are caused by multiple factors, which characterize persistent hypoxemia, neutrophil infiltration, disseminated alveolar damage, acute inflammatory syndrome and impaired blood gas barrier (1,2). Numerous studies have focused on the mechanism and therapy of ALI (35); however, there are still no effective therapeutic methods in the clinic. In previous years, inflammation has been considered to serve a key role in the development of ALI. However, specific mechanisms have not yet been fully elucidated. Inflammatory factors, including nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β, could increase the damage to endothelial and epithelial cells (6,7). Therefore, it is important to further study the mechanism of inflammation in ALI.

The inflammasome is a protein complex that includes the receptor protein apoptosis-associated speck-like protein containing CARD (ASC) and effector molecules, including pre-caspase-1 and pro-caspase-1. The inflammasome can promote the maturation of pro-IL-1β by activating caspase-1. Subsequently, IL-1β is released to the extracellular environment to participate in inflammation, injury and other processes (8,9). Receptor proteins include NOD like receptors (NLRs), including NLRP1 and NLRP3, and the interferon-inducible p200-proteins, including absent in melanoma 2. Different receptor proteins can be activated by different endogenous or exogenous stimuli. The NLRP3 inflammasome is the most studied inflammasome and has been demonstrated to be activated by numerous factors, including Listeria, Aeromonas, ATP and insoluble crystals, including uric acid crystal, silica and asbestos (10,11). It has been reported that in ALI, pulmonary fibrosis, chronic obstructive pulmonary disease, asthma and other lung diseases, the NLRP3 inflammasome serves a key role in inflammation (12). Studies have demonstrated that the NLRP3 inflammasome can be activated through the toll-like receptor 4 signaling pathway and participates in inflammatory injury in ventilator-induced lung injury; the ventilator induced lung injury was markedly alleviated after NLRP3-knockout (13,14). However, to the best of the authors' knowledge, the mechanism of NLRP3 inflammasome activation remains unknown in ALI.

Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that is widely expressed in the body. Under normal conditions, HIF-1α can be degraded via the ubiquitin-proteasome pathway. However, during hypoxia the degradation of HIF-1α is inhibited; therefore, it accumulates and is transferred to the nucleus to promote expression of its target genes (15,16). The lung tissue is in a state of persistent hypoxia and the expression of HIF-1α is significantly increased (1719). It has been demonstrated that HIF-1α participates in inflammation and promotes the expression of inflammatory factors, including TNF-α, IL-6 and IL-1β, in ALI (20,21). In single-stranded RNA viruses-induced inflammatory reactions, HIF-1α has been reported to activate the NLRP3 inflammasome, induce the activation of caspase-1 and then convert inactive pro-IL-1β to IL-1β in human THP-1 myeloid macrophages (22). However, whether HIF-1α can regulate the activation of the NLRP3 inflammasome and the potential function of HIF-1α in ALI remain unknown.

Therefore, the aim of the present study was to investigate whether HIF-1α can regulate the activation of the NLRP3 inflammasome and its potential mechanism in bleomycin (BLM)-induced ALI.

Materials and methods

Main reagents

BLM was purchased from Selleck Chemicals. Anti-HIF-1α (1:500; cat. no. BS3514) primary rabbit monoclonal antibody was obtained from BioWorld Technology, Inc. NF-κB p65 (1:1,000; cat. no. sc-8008), ASC (1:500; cat. no. sc-22514) and caspase-1 (1:500; cat. no. sc-56036) primary mouse monoclonal antibodies were purchased from Santa Cruz Biotechnology, Inc. Anti-NLRP3 (1:1,000; cat. no. ab210491) rabbit monoclonal antibody and anti-β-actin (1:2,000; cat. no. ab179467) primary mouse monoclonal antibody were purchased from Abcam. RIPA buffer, BCA protein assay kit, SDS-PAGE gel preparation kit, 4.0% paraformaldehyde, Triton X-100, DAPI and fluorescein isothiocyanate (FITC)-conjugated goat anti-rabbit IgG (1:200; cat. no. A0562) were obtained from Beyotime Institute of Biotechnology.

Cell culture and transfection

A549 cells and rat type II alveolar cells (RLE-6TN) were purchased from the American Type Culture Collection. A549 cells were cultured in F-12K medium (Genom Biotech Pvt., Ltd.) with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin). RLE-6TN cells were grown in DMEM/F-12 (HyClone; GE Healthcare) with 10% FBS and 1% antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin). Both cell lines were grown at 37°C in a 5% carbon dioxide incubator. HIF-1α small interfering RNA (siRNA) and NF-κB siRNA were designed and generated by Shanghai Genepharma Co., Ltd. The nucleotides sequences of the HIF-1α, NF-κB p65 (abbreviated as NF-κB) and siControl siRNAs are presented in Table I. Both siRNAs were transfected into A549 and RLE-6TN cells using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. After 1 h, A549 cells were treated with BLM at a concentration of 120 µM and RLE-6TN cells were treated with BLM at a concentration of 40 µM for 24 h, according to a previous study (23).

Table I.

The sequences of HIF-1α and NF-κB siRNAs.

Table I.

The sequences of HIF-1α and NF-κB siRNAs.

SpeciesGeneSenseAntisense
Homo sapiensHIF-1α 5′GCCGAGGAAGAACUAUGAATT3′ 5′UUCAUAGUUCUUCCUCGGCTT3′
NF-κB 5′GGACAUAUGAGACCUUCAA3′ 5′UUGAAGGUCUCAUAUGUCC3′
siControl 5′UUCUCCGAACGUGUCACGU3′ 5′ACGUGACACGUUCGGAGAA3′
Rattus NorvegicusHIF-1α 5′GGGCCGUUCAAUUUAUGAATT3′ 5′UUCAUAAAUUGAACGGCCCTT3′
NF-κB 5′AAGAAGCACAGAUACCACCAA3′ 5′UUGGUGGUAUCUGUGCUUCUU3′
siControl 5′UUCUCCGAACGUGUCACGUTT3′ 5′ACGUGACACGUUCGGAGAATT3′

[i] HIF-1α, hypoxia-inducible factor-1 α; NF-κB, nuclear factor-κB; siRNA, small interfering RNA.

Western blotting

Total proteins were extracted from cells with RIPA buffer. Concentrations of the total proteins were determined using a BCA protein assay kit. The total protein samples (4 µg/µl) were separated via 10% SDS-PAGE g, transferred to PVDF membranes, blocked with 5% non-fat milk in TBS with 0.1% Tween 20 (TBST) at 37°C for 1.5 h and incubated with relevant primary antibodies overnight at 4°C. The secondary antibodies were then incubated with the PVDF membrane for 60 min at room temperature. After the membranes were washed with TBST, the bands were visualized with an ECL detection system (Immobilon Western Chemiluminescent HRP substrate; EMD Millipore), according to the manufacturer's protocol.

Immunofluorescence

A549 (8×104/well) and RLE-6TN (1×105/well) cells were seeded in confocal dishes 24 h prior to treatment. The cells were the treated with BLM (120 µM for A549 cells and 40 µM for RLE-6TN cells) for 24 h. Subsequently, the cells were washed with PBS three times, fixed with 4.0% paraformaldehyde for 10 min at room temperature and permeabilized with 0.5% Triton X-100 for 10 min at room temperature. Subsequently, the cells were incubated with primary antibody (NLRP3, 1:400; Caspase-1, 1:200; ASC, 1:200) overnight at 4°C and then incubated with FITC-conjugated goat anti-rabbit IgG for 90 min at room temperature. Nuclei were stained with DAPI for 3 min at 37°C. The cells were observed under a laser confocal microscope (Leica TCS SP8; Leica Microsystems GmbH).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from A549 and RLE-6TN cells with RNAiso Plus reagent (Takara Biotechnology, Co., Ltd.) and the concentration of total RNA in each group was detected with an ultraviolet spectrophotometer. RT (37°C for 15 min and 85°C for 5 sec, then stored at 4°C until further use) was performed using a HiScriptII Q RT SuperMix for qPCR (Vazyme), according to the manufacturer's protocol. qPCR (95°C for 2 min, followed by 40 cycles of 95°C for 10 sec, 60°C for 30 sec and 72°C for 30 sec) was conducted with a ChamQ™ SYBR qPCR Master mix (Vazyme) using an ABI ViiA™ 7 system. The specific primers for HIF-1α, NF-κB and β-actin were designed and generated by BioTNT. The primer sequences of HIF-1α, NF-κB and β-actin are presented in Table II. All samples were read in triplicate and β-actin served as a loading control. The 2−∆∆Cq method was used to determine fold changes (24).

Table II.

The primer sequences of HIF-1α, NF-κB and β-actin used in reverse transcription-quantitative PCR.

Table II.

The primer sequences of HIF-1α, NF-κB and β-actin used in reverse transcription-quantitative PCR.

SpeciesGenePrimer sequence (Forward primer, 5′-3′; Reverse primer, 5′-3′)
Homo sapiensHIF-1α GTCTGAGGGGACAGGAGGAT; CTCCTCAGGTGGCTTGTCAG
NF-κB GAGACATCCTTCCGCAAACT; TCCTTCCTGCCCATAATCA
β-actin ATGATGATATCGCCGCGCTC; CCACCATCACGCCCTGG
Rattus NorvegicusHIF-1α AAGTCTAGGGATGCAGCACG; AGATGGGAGCTCACGTTGTG
NF-κB AGCTCCTGTCCCAGTTCTAGC; ACTCCTGGGTCTGTGTTGTTG
β-actin TCCTTCCTGGGTATGGAATC; GCACTGTGTTGGCATAGAGG

[i] HIF-1α, hypoxia-inducible factor-1 α; NF-κB, nuclear factor-κB; siRNA, small interfering RNA.

Enzyme linked immunosorbent assay (ELISA)

The human IL-1β ELISA kit (cat. no. EL10028) and the rat IL-1β ELISA kit (cat. no. A1010A0301b) were purchased from Anogen-Yes Biotech Laboratories Ltd., and BioTNT, respectively. The expression levels of IL-1β in the A549 and RLE-6TN cell culture supernatants were determined, according to the manufacturer's protocol. All experiments were performed in triplicate.

Statistical analysis

All data are presented as the mean ± standard error of mean of at least three experimental repeats. Mean values data showed a Gaussian distribution. Comparisons between two groups were performed using a t test. Multigroup comparisons of the means were carried out using one-way analysis of variance test. The Bonferroni correction was applied for post hoc analysis. All the statistics were analyzed GraphPad Prism 7 (GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

NLRP3 inflammasome is activated after BLM-treatment

Activation of the NLRP3 inflammasome and the protein expression of HIF-1α and NF-κB in A549 and RLE-6TN cells were evaluated following treatment with BLM 24 h. The levels of proteins associated with the NLRP3 inflammasome, including NLRP3, ASC and caspase-1, were analyzed by immunofluorescence. In the BLM-treated groups, the expression levels of NLRP3, ASC and caspase-1 markedly increased in A549 and RLE-6TN cells (Fig. 1A). In addition, the results demonstrated that HIF-1α and NF-κB expression markedly increased in both cell lines (Fig. 1B and C). These data confirmed that the NLRP3 inflammasome is activated in BLM-treated alveolar epithelial cells.

HIF-1α regulates BLM-induced activation of the NLRP3 inflammasome and the expression of NF-κB

Next, the present study investigated the role of HIF-1α in BLM-induced activation of the NLRP3 inflammasome by transfecting A549 and RLE-6TN cells with HIF-1α siRNA. The expression of HIF-1α significantly increased when both cell lines were treated with BLM for 24 h (P<0.05). Furthermore, HIF-1α siRNA significantly reduced the expression of HIF-1α mRNA in the siHIF-1α group and the siHIF-1α + BLM group (P<0.05; Fig. 2A and B). The protein level of HIF-1α was also inhibited in both cell lines following transfection with HIF-1α siRNA. The levels of proteins associated with the NLRP3 inflammasome, including NLRP3, ASC and caspase-1, and NF-κB were notably decreased in the siHIF-1α + BLM group compared with the BLM group (Fig. 2C and D). The immunofluorescence results also demonstrated that the expression of NLRP3, ASC and caspase-1 deceased after transfection with HIF-1α siRNA (Fig. 2E). These data indicate that HIF-1α could regulate the activation of the NLRP3 inflammasome in BLM-induced ALI.

Figure 2.

Expression of NF-κB and NLRP3 inflammasome-associated proteins after silencing HIF-1α. (A) RT-qPCR for detecting the mRNA expression level of HIF-1α in A549 cells after transfection with HIF-1α siRNA for 48 h and treatment with BLM for 24 h. β-actin served as a loading control. These data are presented as the mean ± standard error of the mean. *P<0.05 [siHIF-1α vs. sicontrol; P=0.0206 (Bonferroni correction, n=2); sicontrol + BLM vs. sicontrol; P=0.0234 (Bonferroni correction, n=2), $P<0.05 (control + BLM vs. control; P=0.0158 (ANOVA), #P<0.05 (siHIF-1α + BLM vs. sicontrol + BLM; P=0.0232 (Bonferroni correction, n=2)]. (B) RT-qPCR for detecting the mRNA expression level of HIF-1α in RLE-6TN cells after transfection with HIF-1α siRNA for 48 h and treatment with BLM for 24 h. β-actin served as a loading control. These data are presented as the mean ± standard error of the mean. $P<0.05 [control + BLM vs. control, P=0.0141 (ANOVA)]. **P<0.01 (siHIF-1α vs. sicontrol, P=0.0046 (Bonferroni correction, n=2); sicontrol + BLM vs. sicontrol, P=0.0038 (Bonferroni correction, n=2), ##P<0.01 (siHIF-1α + BLM vs. sicontrol + BLM, P=0.0046 (Bonferroni correction, n=2). Western blotting for detecting the expression levels of HIF-1α, NF-κB and NLRP3 inflammasome-associated proteins in (C) A549 and (D) RLE-6TN cell lines after transfection with HIF-1α siRNA and treatment with BLM. β-actin served as a loading control. (E) Immunofluorescence for evaluating changes in the expression levels of proteins associated with the NLRP3 inflammasome (NLRP3, ASC and caspase-1) in A549 cells after inhibition of HIF-1α. Scale bar, 50 µm. NLRP3, NOD-like receptor 3; HIF-1α, hypoxia-inducible factor-1α; BLM, bleomycin; ASC, apoptosis-associated speck-like protein containing CARD; RT-qPCR, reverse transcription-quantitative PCR; siRNA, small interfering RNA; NF-κB, nuclear factor-κB; ANOVA, analysis of variance.

BLM-induced activation of the NLRP3 inflammasome is modulated by NF-κB

Subsequently, the current study aimed to confirm the role of NF-κB in BLM-induced activation of the NLRP3 inflammasome by transfecting A549 and RLE-6TN cells with NF-κB siRNA. The level of NF-κB significantly increased when both cell lines were treated with BLM for 24 h (P<0.01). In addition, NF-κB siRNA significantly decreased the level of NF-κB mRNA in the siNF-κB group and the siNF-κB + BLM group (P<0.05; Fig. 3A and B). The protein level of NF-κB was also inhibited in both cell lines after transfection with NF-κB siRNA. Proteins associated with the NLRP3 inflammasome, including NLRP3, ASC and caspase-1, decreased in the siNF-κB + BLM group (Fig. 3C and D). The immunofluorescence results also demonstrated that the expression levels of NLRP3, ASC and caspase-1 deceased following transfection with NF-κB siRNA (Fig. 3E). These data indicate that NF-κB may also participate in modulating activation of the NLRP3 inflammasome in BLM-induced ALI.

Figure 3.

Expression of NLRP3 inflammasome-associated proteins after silencing NF-κB. (A) RT-qPCR for detecting the mRNA expression level of NF-κB in A549 cells after transfection with NF-κB siRNA for 48 h and treatment with BLM for 24 h. β-actin served as a loading control. These data are presented as the mean ± standard error of the mean. *P<0.05 [siNF-κB vs. sicontrol; P=0.0246 (Bonferroni correction, n=2)], ***P<0.001 [sicontrol + BLM vs. sicontrol; P=0.0008 (Bonferroni correction, n=2)], $$P<0.01 [control + BLM vs. control; P=0.0041 (ANOVA)], ##P<0.01 [siNF-κB + BLM vs. sicontrol + BLM; P=0.002 (Bonferroni correction, n=2)]. (B) RT-qPCR for detecting the mRNA expression level of NF-κB in RLE-6TN cells after transfection with NF-κB siRNA for 48 h and treatment with BLM for 24 h. β-actin served as a loading control. These data are presented as the mean ± standard error of the mean. *P<0.05 [siNF-κB vs. sicontrol; P=0.023 (Bonferroni correction, n=2); sicontrol + BLM vs. sicontrol; P=0.021 (Bonferroni correction, n=2)], $P<0.05 [control + BLM vs. control; P=0.0168 (ANOVA)], ##P<0.01 [siNF-κB + BLM vs. sicontrol + BLM; P=0.0046 (Bonferroni correction, n=2)]. Western blotting for detecting the expression levels of NF-κB and NLRP3 inflammasome-associated proteins in (C) A549 and (D) RLE-6TN cell lines after transfection with NF-κB siRNA and treatment with BLM. β-actin served as a loading control. (E) Immunofluorescence for evaluating changes in the expression levels of proteins associated with the NLRP3 inflammasome (NLRP3, ASC and caspase-1) in A549 cells after inhibition of NF-κB. Scale bar, 50 µm. NLRP3, NOD-like receptor 3; BLM, bleomycin; ASC, apoptosis-associated speck-like protein containing CARD; RT-qPCR, reverse transcription-quantitative PCR; siRNA, small interfering RNA; siRNA, small interfering RNA; NF-κB, nuclear factor-κB.

IL-1β level is regulated by HIF-1α and NF-κB

It has been reported that IL-1β expression significantly increases after activation of the NLRP3 inflammasome (25,26). Therefore, the present study detected the levels of IL-1β in the cellular culture supernatants of A549 following inhibition of HIF-1α or NF-κB. The results demonstrated that IL-1β expression significantly increased in the BLM-treatment group (P<0.05) and significantly decreased in the siHIF-1α + BLM group and the siNF-κB + BLM group (P<0.05; Fig. 4A and B). The similar results were observed in RLE-6TN cells (Fig. 4C and D). This result indicates that BLM-induced IL-1β expression may also be regulated by HIF-1α and NF-κB.

Figure 4.

Levels of IL-1β in cellular culture supernatants after silencing HIF-1α or NF-κB. (A) The cellular culture supernatants of A549 cells were collected after transfection with HIF-1α siRNA. The change of IL-1β in the cellular culture supernatants was detected by ELISA. *P<0.05 [sicontrol + BLM vs. sicontrol; P=0.0354 (Bonferroni correction, n=2)], #P<0.05 [siHIF-1α + BLM vs. sicontrol + BLM; P=0.0403 (Bonferroni correction, n=2)]. (B) The cellular culture supernatants of A549 cells were collected after transfection with NF-κB siRNA. The change of IL-1β in the cellular culture supernatants was detected by ELISA. *P<0.05 [sicontrol + BLM vs. sicontrol; P=0.022 (Bonferroni correction, n=2)], #P<0.05 [siNF-κB + BLM vs. sicontrol + BLM; P=0.0432 (Bonferroni correction, n=2)]. (C) The cellular culture supernatants of RLE-6TN cells were collected after transfection with HIF-1α siRNA. The change of IL-1β in the cellular culture supernatants was detected by ELISA. *P<0.05 [siHIF-1α + BLM vs. sicontrol + BLM; P=0.0256 (Bonferroni correction, n=2)], #P<0.05 [siHIF-1α + BLM vs. sicontrol + BLM; P=0.0406 (Bonferroni correction, n=2)]. (D) The cellular culture supernatants of RLE-6TN cells were collected after transfection with NF-κB siRNA. The change of IL-1β in the cellular culture supernatants was detected by ELISA. *P<0.05 [sicontrol + BLM vs. sicontrol; P=0.029 (Bonferroni correction, n=2)], #P<0.05 [siNF-κB + BLM vs. sicontrol + BLM; P=0.0366 (Bonferroni correction, n=2)]. SiRNA, small interfering RNA; BLM, bleomycin; HIF-1α, hypoxia-inducible factor-1α; IL, interleukin.

Discussion

ALI and ARDS, which can be caused by multiple factors, are common clinical syndromes. It has been confirmed that over-regulation of the inflammatory response is one of the most important factors leading to the occurrence and development of ALI (27,28). IL-1β is the main pro-inflammatory cytokine that may be a potential molecular biomarker for predicting morbidity and mortality (4). The present study identified that HIF-1α may regulate activation of the NLRP3 inflammasome via NF-κB and could promote the expression of IL-1β in BLM-induced ALI.

The NLRP3 inflammasome is currently the most studied and most widely activated inflammasome. It can be activated by bacteria, fungus, virus and damage-associated molecular patterns, including uric acid crystals and silicon dioxide (2933). It has been reported that the NLRP3 inflammasome is activated in transfusion-associated acute lung injury, ventilator-induced lung injury, asthma, chronic obstructive pulmonary disease and other pulmonary diseases. The NLRP3 inflammasome has been demonstrated to increase the release of IL-1β to participate in inflammation and the immune reaction (12). Tian et al (23) demonstrated that the NLRP3 inflammasome could also regulate the epithelial-mesenchymal transition in BLM-induced pulmonary fibrosis. The present study identified that the expression of proteins associated with the NLRP3 inflammasome, including NLRP3, ASC and caspase-1, were significantly increased in the BLM-treatment group. Therefore, it was confirmed that the NLRP3 inflammasome could be activated in BLM-induced ALI. However, the mechanism of the activation of the NLRP3 inflammasome requires further investigation.

It is understood that the body is in a state of hypoxia when ALI occurs. Numerous studies have confirmed that HIF-1α is an important regulatory factor under hypoxic conditions (3436). In addition, a recent study demonstrated that HIF-1α could also increase under normoxic conditions (37). HIF-1α could promote the expression of inflammatory cytokines, including TNF-α, IL-6 and IL-1β, in septic lymph treated A549 cells and human pulmonary microvascular endothelial cells (18). Ouyang et al (38) reported that HIF-1α sustains inflammasome activity via the cAMP/PKA/CREB/HIF-1α pathway in adenosine-stimulated murine peritoneal macrophages. However, whether HIF-1α can regulate the activation of the NLRP3 inflammasome in ALI has not been reported. The current study identified that HIF-1α was increased in BLM-treated A549 and RLE-6TN cells. In addition, activation of the NLRP3 inflammasome was inhibited when the expression of HIF-1α was silenced in BLM-induced ALI. These results indicate that BLM-induced activation of the NLRP3 inflammasome could be regulated by HIF-1α.

NF-κB is a transcription factor that serves an important role in regulating the transcription of multiple inflammatory factors and cytokines. A number of studies have reported that NF-κB can increase in response to multiple factors induced in ALI (39,40). In a lipopolysaccharide-treated alveolar macrophages cell line, myeloid differentiation protein 2 could regulate the activation of the NLRP3 inflammasome and IL-1β expression via the MyD88/NF-κB pathway (41). Under hypoxic conditions, NF-κB is activated by HIF-1α to participate in regulation of inflammation, cell death and angiogenesis (42). In the present study, activation of the NLRP3 inflammasome was inhibited when HIF-1α and NF-κB were silenced in BLM-treated alveolar epithelial cells. Additionally, the levels of IL-1β were markedly decreased in the cellular culture supernatants after inhibition of HIF-1α and NF-κB. Thus, these data indicate that HIF-1α may modulate the activation of the NLRP3 inflammasome and the secretion of IL-1β via NF-κB signaling.

In conclusion, it was confirmed that the NLRP3 inflammasome is activated in BLM-induced ALI. Furthermore, HIF-1α was demonstrated to modulate activation of the NLRP3 inflammasome through NF-κB and subsequently promote the expression of IL-1β. The present results promoted understanding regarding the mechanism of ALI and may provide new ideas in identifying therapeutic targets of ALI. However, the current study is limited to clinical guidance as it was primarily performed in vitro. In the future, further research will be performed in in vivo experiments and clinical research.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

JH and YL conceived and designed the study, and analyzed and interpreted the results. JH performed the experiments and wrote the manuscript. JX and LH conducted the experiments. All authors 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.

Glossary

Abbreviations

Abbreviations:

ALI

acute lung injury

ARDS

acute respiratory distress syndrome

ASC

apoptosis-associated speck-like protein containing CARD

BLM

bleomycin

HIF-1α

hypoxia-inducible factor-1 α

IL-1β

interleukin-1β

NF-κB

nuclear factor-κB

NLRP3

NOD-like receptor 3

TNF-α

tumor necrosis factor-α

References

1 

ARDS Definition Task Force, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L and Slutsky AS: Acute respiratory distress syndrome: The Berlin Definition. JAMA. 307:2526–2533. 2012.PubMed/NCBI

2 

Matthay MA and Zemans RL: The acute respiratory distress syndrome: Pathogenesis and treatment. Annu Rev Pathol. 6:147–163. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Spragg RG, Bernard GR, Checkley W, Curtis JR, Gajic O, Guyatt G, Hall J, Israel E, Jain M, Needham DM, et al: Beyond mortality: Future clinical research in acute lung injury. Am J Respir Crit Care Med. 181:1121–1127. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Butt Y, Kurdowska A and Allen TC: Acute lung injury: A clinical and molecular review. Arch Pathol Lab Med. 140:345–350. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Qu L, Chen C, Chen Y, Li Y, Tang F, Huang H, He W, Zhang R and Shen L: High-Mobility group box 1 (HMGB1) and autophagy in acute lung injury (ALI): A review. Med Sci Monit. 25:1828–1837. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Bouwmeester T, Bauch A, Ruffner H, Angrand PO, Bergamini G, Croughton K, Cruciat C, Eberhard D, Gagneur J, Ghidelli S, et al: A physical and functional map of the human TNF-alpha/NF-kappa B signal transduction pathway. Nat Cell Biol. 6:97–105. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Bhatia M and Moochhala S: Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol. 202:145–156. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Schroder K and Tschopp J: The inflammasomes. Cell. 140:821–832. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Strowig T, Henao-Mejia J, Elinav E and Flavell R: Inflammasomes in health and disease. Nature. 481:278–286. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Bauernfeind F, Ablasser A, Bartok E, Kim S, Schmid-Burgk J, Cavlar T and Hornung V: Inflammasomes: Current understanding and open questions. Cell Mol Life Sci. 68:765–783. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Eisenbarth SC and Flavell RA: Innate instruction of adaptive immunity revisited: The inflammasome. EMBO Mol Med. 1:92–98. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Hosseinian N, Cho Y, Lockey RF and Kolliputi N: The role of the NLRP3 inflammasome in pulmonary diseases. Ther Adv Respir Dis. 9:188–197. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Kuipers MT, Aslami H, Janczy JR, van der Sluijs KF, Vlaar AP, Wolthuis EK, Choi G, Roelofs JJ, Flavell RA, Sutterwala FS, et al: Ventilator-induced lung injury is mediated by the NLRP3 inflammasome. Anesthesiology. 116:1104–1115. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Dai H, Pan L, Lin F, Ge W, Li W and He S: Mechanical ventilation modulates Toll-like receptors 2, 4, and 9 on alveolar macrophages in a ventilator-induced lung injury model. J Thorac Dis. 7:616–624. 2015.PubMed/NCBI

15 

Vriend J and Reiter RJ: Melatonin and the von Hippel- Lindau/HIF-1 oxygen sensing mechanism: A review. Biochim Biophys Acta. 1865:176–183. 2016.PubMed/NCBI

16 

Rhim T, Lee DY and Lee M: Hypoxia as a target for tissue specific gene therapy. J Control Release. 172:484–494. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Tang M, Tian Y, Li D, Lv J, Li Q, Kuang C, Hu P, Wang Y, Wang J, Su K and Wei L: TNF-α mediated increase of HIF-1α inhibits VASP expression, which reduces alveolar-capillary barrier function during acute lung injury (ALI). PLoS One. 9:e1029672014. View Article : Google Scholar : PubMed/NCBI

18 

Jiang H, Hu R, Sun L, Chai D, Cao Z and Li Q: Critical role of toll-like receptor 4 in hypoxia-inducible factor 1α activation during trauma/hemorrhagic shocky induced acute lung injury after lymph infusion in mice. Shock. 42:271–278. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Suresh MV, Ramakrishnan SK, Thomas B, Machado-Aranda D, Bi Y, Talarico N, Anderson E, Yatrik SM and Raghavendran K: Activation of hypoxia-inducible factor-1α in type 2 alveolar epithelial cell is a major driver of acute inflammation following lung contusion. Crit Care Med. 42:e642–e653. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Sun HD, Liu YJ, Chen J, Chen MY, Ouyang B and Guan XD: The pivotal role of HIF-1α in lung inflammatory injury induced by septic mesenteric lymph. Biomed Pharmacother. 91:476–484. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Vohwinkel CU, Hoegl S and Eltzschig HK: Hypoxia signaling during acute lung injury. J Appl Physiol (1985). 119:1157–1163. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Nicholas SA, Bubnov VV, Yasinska IM and Sumbayev VV: Involvement of xanthine oxidase and hypoxia-inducible factor 1 in Toll-like receptor 7/8-mediated activation of caspase 1 and interleukin-1β. Cell Mol Life Sci. 68:151–158. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Tian R, Zhu Y, Yao J, Meng X, Wang J, Xie H and Wang R: NLRP3 participates in the regulation of EMT in bleomycin-induced pulmonary fibrosis. Exp Cell Res. 357:328–334. 2017. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Grebe A, Hoss F and Latz E: NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ Res. 122:1722–1740. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Sarvestani ST and McAuley JL: The role of the NLRP3 inflammasome in regulation of antiviral responses to influenza A virus infection. Antiviral Res. 148:32–42. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Sciuto AM, Clapp DL, Hess ZA and Moran TS: The temporal profile of cytokines in the bronchoalveolar lavage fluid in mice exposed to the industrial gas phosgene. Inhal Toxicol. 15:687–700. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Wang P, Ye XL, Liu R, Chen HL, Liang X, Li WL, Zhang XD, Qin XJ, Bai H, Zhang W, et al: Mechanism of acute lung injury due to phosgene exposition and its protection by cafeic acid phenethyl ester in the rat. Exp Toxicol Pathol. 65:311–318. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Toma C, Higa N, Koizumi Y, Nakasone N, Ogura Y, McCoy AJ, Franchi L, Uematsu S, Sagara J, Taniguchi S, et al: Pathogenic Vibrio activate NLRP3 inflammasome via cytotoxins and TLR/nucleotide-binding oligomerization domain-mediated NF-kappa B signaling. J Immunol. 184:5287–5297. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, Hartmann G, Tardivel A, Schweighoffer E, Tybulewicz V, et al: Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature. 459:433–436. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Thomas PG, Dash P, Aldridge JR Jr, Ellebedy AH, Reynolds C, Funk AJ, Martin WJ, Lamkanfi M, Webby RJ, Boyd KL, et al: The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 30:566–575. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Martinon F, Petrilli V, Mayor A, Tardivel A and Tschopp J: Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 440:237–241. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA and Latz E: Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 9:847–856. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Semenza GL: Hypoxia-inducible factors in physiology and medicine. Cell. 148:399–408. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Palazon A, Goldrath AW, Nizet V and Johnson RS: HIF transcription factors, inflammation, and immunity. Immunity. 41:518–528. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Rohwer N, Zasada C, Kempa S and Cramer T: The growing complexity of HIF-1α's role in tumorigenesis: DNA repair and beyond. Oncogene. 32:3569–3576. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Koyasu S, Kobayashi M, Goto Y, Hiraoka M and Harada H: Regulatory mechanisms of hypoxia-inducible factor 1 activity: Two decades of knowledge. Cancer Sci. 109:560–571. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Ouyang X, Ghani A, Malik A, Wilder T, Colegio OR, Flavell RA, Cronstein BN and Mehal WZ: Adenosine is required for sustained inflammasome activation via the A2A receptor and the HIF-1α pathway. Nat Commun. 4:29092013. View Article : Google Scholar : PubMed/NCBI

39 

Li YC, Yeh CH, Yang ML and Kuan YH: Luteolin suppresses inflammatory mediator expression by blocking the Akt/NFκB pathway in acute lung injury induced by lipopolysaccharide in mice. Evid Based Complement Alternat Med. 2012:3836082012. View Article : Google Scholar : PubMed/NCBI

40 

Niu X, Hu H, Li W, Li Y, Huang H, Mu Q, Yao H and Li H: Protective effect of total alkaloids on lipopolysaccharide-induced acute lung injury. J Surg Res. 189:126–134. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Luo M, Hu L, Li D, Wang Y, He Y, Zhu L and Ren W: MD-2 regulates LPS-induced NLRP3 inflammasome activation and IL-1beta secretion by a MyD88/NF-κB-dependent pathway in alveolar macrophages cell line. Mol Immunol. 90:1–10. 2017. View Article : Google Scholar : PubMed/NCBI

42 

D'Ignazio L and Rocha S: Hypoxia Induced NF-κB. Cells. 5(pii): E102016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

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
Huang JJ, Xia J, Huang LL and Li YC: HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury. Mol Med Rep 20: 3424-3432, 2019
APA
Huang, J., Xia, J., Huang, L., & Li, Y. (2019). HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury. Molecular Medicine Reports, 20, 3424-3432. https://doi.org/10.3892/mmr.2019.10575
MLA
Huang, J., Xia, J., Huang, L., Li, Y."HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury". Molecular Medicine Reports 20.4 (2019): 3424-3432.
Chicago
Huang, J., Xia, J., Huang, L., Li, Y."HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury". Molecular Medicine Reports 20, no. 4 (2019): 3424-3432. https://doi.org/10.3892/mmr.2019.10575