Open Access

SIRT1 regulates cigarette smoke extract‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway

  • Authors:
    • Fang Yang
    • Huiping Qin
    • Chaoqun Qin
    • Bing Huang
    • Feng Gao
    • Yi Liao
    • Yanping Tang
    • Yanju Mo
    • Qianjie Yang
    • Changming Wang
  • View Affiliations

  • Published online on: November 29, 2024     https://doi.org/10.3892/mmr.2024.13408
  • Article Number: 43
  • Copyright: © Yang 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

M1 macrophages activated by cigarette smoke extract (CSE) serve a pro‑inflammatory role in chronic obstructive pulmonary disease (COPD). The expression of silent information regulator 1 (SIRT1) is decreased in the alveolar macrophages of patients with COPD. However, whether SIRT1 is involved in COPD by regulating macrophage polarization remains unknown. Rat Alveolar Macrophage NR8383 cells were exposed to CSE. Cell Counting Kit‑8 assay, western blot assay and ELISA showed that with increasing concentration of CSE, the activity of NR8383 cells and expression of SIRT1 gradually decreased, while the release of inflammatory cytokines TNFα, IL‑1β and IL‑6 increased. As shown in western blot or Immunofluorescence assays, exposure to CSE also increased expression levels of the M1 markers inducible nitric oxide synthase and CD86, whereas it downregulated expression of the M2 markers arginase 1 and CD206. In addition, CSE increased expression of TNF receptor associated factor 6 (TRAF6), NOD‑like receptor thermal protein domain associated protein 3 (NLRP3) and cleaved caspase‑1 protein in NR8383 cells. Overexpression plasmids of SIRT1 and TRAF6 significantly reversed the aforementioned changes induced by CSE. Moreover, immunoprecipitation demonstrated that TRAF6 could bind to NLRP3. The overexpression of TRAF6 notably attenuated the regulatory effects of overexpression of SIRT1 on polarization and inflammation in NR8383 cells. Conversely, overexpression of SIRT1 inhibited the TRAF6/NLRP3 signaling pathway, thereby suppressing CSE‑induced M1 polarization and release of inflammatory factors in NR8383 cells. The present study demonstrates that SIRT1 regulates CSE‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway.

Introduction

Chronic obstructive pulmonary disease (COPD) is a prevalent chronic respiratory condition associated with high morbidity, mortality and disability rates, which result in death of approximately 3.2 million individuals annually (1,2). By 2030, COPD is projected to become the third leading cause of mortality worldwide (3). Currently, the clinical treatment for COPD aims to alleviate the clinical symptoms, typically through the use of bronchodilators and oxygen therapy (4). However, COPD faces issues, such as under-diagnosis and misdiagnosis, and there is currently no treatment method to halt the progression of COPD (5). Therefore, it is imperative to study potential mechanisms responsible for the occurrence of COPD and develop novel predictive and therapeutic targets.

Inflammation is the key mechanism in the development of COPD (6). Alveolar macrophages (AMs) are the first line of defense in the lung and play a key role in lung inflammation (7). Studies have shown that in COPD, there is an increase in number of lung macrophages and a decrease in antigen presentation ability (8,9). Smoking is a common risk factor for the development of COPD (10); M1 polarization of lung macrophages is increased in smokers and patients with COPD (11). Therefore, inhibiting cigarette smoke (CS)-induced M1 polarization of AMs may offer a novel approach for anti-inflammatory treatment of COPD.

Silent information regulator 1 (SIRT1) is the mammalian homolog of the yeast silent information regulator 2 protein and is expressed in low levels in the lung of patients with COPD; SIRT1 is associated with a decline in lung function, indicating it is a potential biological marker for the severity of COPD (12). Studies have shown that SIRT1 serves a key role in COPD by regulating oxidative stress, inflammatory responses, autophagy and apoptosis (1315). The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) signal is a classic inflammatory signaling pathway and NLRP3 expression is increased in CS-induced AMs (16). SIRT1 can inhibit the expression of TNF receptor associated factor 6 (TRAF6) (17), which is involved in promoting polarization of M1 macrophages; its expression is also increased in serum of patients with COPD (18). Notably, TRAF6 can bind to NLRP3 and activate its signaling (19,20).

The present study aimed to explore whether SIRT1 can inhibit CS extract (CSE)-induced AM damage via the TRAF6/NLRP3 signaling pathway, thus contributing to its protective role in COPD. The aim of the present study was to elucidate the role and mechanisms of action of SIRT1 in the occurrence and development of COPD and to provide an experimental basis for understanding the pathogenesis COPD. The present results may aid in the development of novel drug and clinical treatments for COPD.

Materials and methods

Preparation of CSE

CSE was prepared by burning three cigarettes [11.0 tar, 1.1 nicotine, 17.0 mg carbon monoxide; Chongqing Hongsheng Industrial (Group) Co., Ltd.]. The smoke was dissolved in serum-free F-12K medium (20 ml; cat. no. 21127022, Gibco; Thermo Fisher Scientific, Inc) and filtered through a 0.22-µm filter. The resulting 100% CSE was used within 1 h of preparation. F-12K medium was used to adjust the CSE working concentration.

Cell culture and treatment

The rat alveolar macrophage cell line NR8383 (American Type Culture Collection), was cultured in F-12K medium (cat. no. 21127022, Gibco; Thermo Fisher Scientific, Inc) with 15% FBS (Thermo Fisher Scientific, Inc) and 1% penicillin-streptomycin at 37°C in a humidified atmosphere of 5% CO2. The cells were treated as aforementioned (21) with CSE (5, 10 and 20%) for 24, 48 and 72 h at 37°C, respectively.

Cell transfection

The overexpression plasmids of SIRT1 [Ov-SIRT1; vector, pEX-3(pGCMV/MCS/Neo)], TRAF6 [(Ov-TRAF6; vector, pEX-3(pGCMV/MCS/Neo)] and negative control (NC) plasmid pEX-3(pGCMV/MCS/Neo) were purchased from Genepharm Biotech Corp. The cells were inoculated in a 6-well plate at a density of 1×105 cells/well. Following 24 h culture and upon reaching 75% confluency, the cells were transfected using 2 µg overexpression plasmid and Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 48 h. After 48 h, western blot analysis was performed to detect transfection efficiency.

Cell Counting Kit (CCK)-8 assay

CCK-8 (BIOSS, cat. no. BA00208) was employed to assess cell viability. NR8383 cells were inoculated into 96-well plates at a density of 3×104/l. Following stimulation with CSE, 10 µl CCK8 solution was added to each well and incubated at 37°C for 2 h. The optical density at 450 nm was determined by a microplate reader (Bio-Rad Laboratories, Inc.).

ELISA

The concentrations of TNFα (cat. no. JL13202-96T), IL-1β (cat. no. JL20884-96T) and IL-6 (cat. no. JL20896-96T; all JONLNBIO) in the cell supernatant were measured using ELISA kits according to the manufacturer's instructions.

Western blot analysis

NR8383 cells were lysed in RIPA buffer with protease inhibitor cocktail (both Beyotime Institute of Biotechnology). After measuring the protein concentration by BCA assay, 20 µg/lane protein was electrophoresed on 10% SDS-PAGE and transferred onto PVDF membranes (Amersham Biosciences). The membrane was blocked in 5% BSA (Biofroxx; neoFroxx) at room temperature for 2 h and incubated with anti-SIRT1 (1:1,000; cat. no. ab110304, Abcam), anti-inducible nitric oxide synthase (iNOS; 1:1,500; cat. no. AF0199, Affinity Biosciences), anti-CD86 (1:1,500; cat. no. DF6332, Affinity Biosciences), anti-arginase 1 (Arg-1; 1:1,000; cat. no. DF6657, Affinity Biosciences), anti-CD206 (1:1,500; cat. no. DF4149, Affinity Biosciences), anti-TRAF6 (1:1,000; cat. no. ab40675, Abcam), anti-NLRP3 (1:1,500; cat. no. DF7438, Affinity Biosciences), anti-cleaved caspase-1 (1:1,000; cat. no. AF4022, Affinity Biosciences), anti-pro-caspase-1 (1:1,000; cat. no. DF6148, Affinity Biosciences) and anti-GAPDH (1:2,500; cat. no. ab9485, Abcam) at 4°C overnight. The membrane was incubated with secondary antibody (goat anti-rabbit IgG-HRP; 1:5,000, cat. no. S0001, Affinity Biosciences) at 37°C for 2 h. The protein signals were exposed using ECL reagent (Beyotime Institute of Biotechnology) and analyzed using ImageJ software version 1.50 (National Institutes of Health).

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted from the NR8383 cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). RT was performed using the RevertAid RT kit (cat. no. MR101-02, Vazyme Biotech Co., Ltd.) according to the manufacturer's instructions. qPCR was performed at 95°C for 120 sec for initial denaturation, followed by denaturation at 95°C for 15 sec and annealing and extension at 60°C for 30 sec (40 cycles). Gene expression was normalized to GAPDH and relative mRNA expression levels were determined using the 2−ΔΔCq method (22). The primer sequences were obtained from PrimerBank (pga.mgh.harvard.edu/primerbank) (Table I).

Table I.

Primer sequences used for reverse transcription-quantitative PCR.

Table I.

Primer sequences used for reverse transcription-quantitative PCR.

GenePrimerSequence, 5′→3′
GAPDHForward GGCAAGTTCAACGGCACAGTC
Reverse TCGCTCCTGGAAGATGGTGATG
TNFαForward GCCCAGACCCTCACACTCAG
Reverse CCGCTTGGTGGTTTGCTACG
IL-1βForward GACTTCACCATGGAACCCGT
Reverse GGAGACTGCCCATTCTCGAC
IL-6Forward CACTTCACAAGTCGGAGGCT
Reverse TCTGACAGTGCATCATCGCT
TRAF6Forward AATCACTTGGCACGGCACTTG
Reverse GGAGAGGAGGCATCGCATGG

[i] TNF, tumor necrosis factor; TRAF, TNF receptor-associated factor.

Immunofluorescence assay

Transfected NR8383 cells were inoculated into six-well plates and when the cell fusion reached 80%, the cells were fixed with 4% neutral formaldehyde for 30 min at 4°C. Sealing solution was added to each well followed by incubation at 37°C for 30 min. The primary antibodies [iNOS (1:200; cat. no. AF0199, Affinity Biosciences) and Arg-1 (1:200; cat. no. DF6332, Affinity Biosciences)] were incubated overnight at 4°C. The diluted secondary antibody Goat Anti-Rabbit IgG (H+L) Fluor488-conjugated (1:200; cat. no. S0018, Affinity Biosciences) was incubated at room temperature without light for 1 h. After washing the cells with PBS, nuclei of the cells were re-stained with DAPI solution (Beyotime Institute of Biotechnology) at room temperature for 10 min and observed under a fluorescence microscope (200×) (Nikon Corporation).

Co-immunoprecipitation (Co-IP) assay

NR8383 cell lysates were prepared by IP lysate (Beyotech Institute of Biotechnology; cat. no. P0013) (150 µl/well) and incubated with anti-TRAF6 (1 µg) (1:100; cat. no. ab137452, Abcam) overnight at 4°C followed by addition of 30 µl protein G Agarose beads (Cytiva) at 4°C for 4 h. After being washed three times with cold wash and once with lysis buffer (both New Cell and Molecular Biotech Co., Ltd.), the complexes were isolated by centrifuging at 1,000 × g at 4°C for 3 min. The immunoprecipitate was resuspended in 30 µl loading buffer. The expression of NLRP3 was detected using western blot analysis as aforementioned.

Statistical analysis

All analyses were performed using GraphPad Prism 9 (Dotmatics) and data are expressed as the mean ± SD of at least three independent experiments. All data were analyzed for normality distribution using the Shapiro-Wilk test. Data were analyzed by one-way ANOVA followed by Tukey's multiple comparisons post hoc tests. P<0.05 was considered to indicate a statistically significant difference.

Results

SIRT1 expression is decreased in CSE-induced AMs

With increasing concentrations of CSE, cell viability gradually decreased at 24 and 48 h, but there was no significant difference between 48 and 72 h (Fig. 1A). Therefore, CSE was used to induce cells for 48 h for subsequent experiments. The secretion of inflammatory cytokines (TNFα, IL-β, and IL-6) was elevated (Fig. 1B), whereas SIRT1 expression exhibited a dose-dependent decrease (Fig. 1C). The greatest effects were observed at 20% CSE, therefore 20% CSE was used in further experiments.

SIRT1 overexpression inhibits CSE-induced AM M1-type polarization and inflammatory factor release

There was a significant increase in SIRT1 expression in cells transfected with Ov-SIRT1 compared with both the control and Ov-NC groups (Fig. 2A). The detection of macrophage polarization markers indicated a significant elevation in both mRNA (Fig. 2B) and protein expression levels (Fig. 2C) of the M1 marker iNOS following exposure to 20% CSE, along with an enhanced fluorescence intensity of iNOS (Fig. 2D) and augmented expression of CD86 (Fig. 2C). Following exposure to 20% CSE, there was a significant decrease in both mRNA (Fig. 2B) and protein expression levels (Fig. 2C) of the M2 marker Arg-1 accompanied by a reduced fluorescence intensity of Arg-1 (Fig. 2E) and a diminished expression of CD206 (Fig. 2C). The iNOS/Arg-1 ratio exhibited a significant decrease (Fig. 2B). Moreover, exposure to 20% CSE increased expression of TNFα, IL-β and IL-6 in the cells (Fig. 2F and G). SIRT1 overexpression significantly reversed these changes induced by CSE.

SIRT1 overexpression inhibits TRAF6/NLRP3 signaling activation in CSE-induced macrophages

The present study assessed the TRAF6/NLRP3 signaling pathway in macrophages stimulated by CSE. There was a significant increase in the levels of TRAF6, NLRP3 and cleaved caspase-1 proteins in NR8383 cells following exposure to 20% CSE. Overexpression of SIRT1 significantly reversed this trend (Fig. 3A). IP demonstrated the ability of TRAF6 to interact with NLRP3 (Fig. 3B). Western blot analysis (Fig. 3C) and RT-qPCR (Fig. 3D) revealed a significant increase in both the protein and mRNA levels of TRAF6 in NR8383 cells following transfection with Ov-TRAF6. Moreover, TRAF6 overexpression attenuated the effects of Ov-SIRT1 on expression of TRAF6, NLRP3 and cleaved caspase-1 proteins in CSE-induced macrophages (Fig. 3E).

Overexpression of SIRT1 inhibits CSE-induced AM M1-type polarization and inflammatory factor release by suppressing TRAF6/NLRP3 signaling

Finally, the present study used the TRAF6 overexpression vector to examine whether TRAF6/NLRP3 signaling mediates the role of SIRT1. The results revealed marked suppression of the effects of Ov-SIRT1 on CSE-induced M1 polarization and inflammatory release in NR8383 cells, supported by the upregulated protein expression of the M1 markers iNOS and CD86 and the downregulated protein expression of the M2 markers Arg-1 and CD206 following TRAF6 overexpression (Fig. 4A). Following TRAF6 overexpression, there was an increase in iNOS mRNA levels (Fig. 4B) and fluorescence intensity (Fig. 4C), whereas Arg-1 mRNA levels (Fig. 4B) and fluorescence intensity decreased (Fig. 4D). The release and mRNA levels of inflammatory cytokines (TNFα, IL-β, and IL-6) also exhibited an increase (Fig. 4E and F).

Discussion

The present study investigated the involvement of SIRT1 in AMs induced by CSE. The results revealed a decrease in SIRT1 expression in NR8383 cells induced by CSE. SIRT1 overexpression suppressed M1 polarization, inflammatory factor release and TRAF6/NLRP3 signaling activation in NR8383 cells exposed to CSE. However, enhancing TRAF6 expression markedly diminished the protective effects of SIRT1 overexpression on NR8383 cells.

Inflammation is a key pathological feature of COPD (23). Smoking is a contributing factor for COPD. CS stimulates release of inflammatory mediators and activates pathways associated with inflammation, leading to pulmonary inflammation (24). As the concentration of CSE increased, TNFα, IL-β, and IL-6 release in NR8383 cells increased, accompanied by a gradual decrease in cellular viability.

SIRT1 is involved in oxidative stress and chronic inflammatory responses, known for its anti-aging properties and associated with the development and progression of COPD (25). Previous research has revealed a decrease in SIRT1 levels in pulmonary macrophages of patients with COPD (26), suggesting its potential involvement in pathogenesis of COPD through the modulation of these macrophages. AMs are key components of innate immune responses (27). Depending on environmental stimuli, they are activated classically (also known as macrophage M1 polarization) or alternatively (also known as macrophage M2 polarization). The two polarization directions of AMs suggest two different inflammatory states. M1-type macrophages primarily release inflammatory factors (TNFα, IL-β and IL-6) and promote development of inflammation, and their markers mainly include iNOS and CD86. M2-type macrophages primarily release anti-inflammatory factors (TGF-β and IL-10) and inhibit progression of inflammation, and their markers mainly include CD206 and Arg-1 (28). Dysregulated macrophage function exacerbates pulmonary inflammation in COPD by affecting initiation of inflammation, disrupting alveolar architecture and remodeling the airways (29). Modulating the balance of M1/M2 macrophage polarization is effective in alleviating COPD (28). Macrophage polarization is one of the pathogenic mechanisms of COPD. Feng and Zheng (10) found that M1 polarization of AMs is inhibited in smokers and patients with COPD and that CS may inhibit LPS-induced M1 polarization of AMs by suppressing NLRP3. Both macrophages from individuals with COPD and (30), and CSE-induced macrophages tend to be M1-type (29). Mu et al (31) showed that CSE promotes macrophage polarization towards the M1 type and high-mobility group box-1 is involved in regulation of macrophage polarization. Li et al (32) demonstrated that Fritillaria cirrhosa D. Don inhibits CSE treatment-induced macrophage M1 polarization, thereby attenuating inflammatory responses. It has been reported that salidroside serves a protective role in COPD by inhibiting JNK/c-Jun to reduce the M1 polarization of AMs induced by CS (7). BML-111 (lipoxin receptor agonist) treatment decreases iNOS levels and increases Arg-1 expression, which has the potential to convert macrophages from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype, thereby preventing COPD (33). Rosiglitazone, an exogenous ligand of PPARγ, has been shown to inhibit CS-induced M1 macrophage polarization and decrease M1/M2 ratio, thereby attenuating emphysema induced by CS exposure and inflammatory responses (34). The present study demonstrated a significant decrease in inflammatory factor release in NR8383 cells following the overexpression of SIRT1. This reduction was accompanied by a decrease in expression of M1 polarization markers, such as iNOS and CD86, induced by CSE in NR8383 cells, along with an increase in expression of M2 markers, including Arg-1 and CD206. Numerous studies have investigated the role of SIRT1 in COPD: For example, SIRT1 improves COPD by regulating CS-induced autophagy (35), endoplasmic reticulum stress (36), airway remodeling and epithelial-mesenchymal transition (37). To the best of our knowledge, the present study is the first to reveal that SIRT1 has the capability to inhibit the M1 polarization of AMs induced by CSE. This provides novel insight into the role of SIRT1 in COPD.

SIRT1 exerts a protective effect against COPD by influencing downstream signaling molecules. For example, SIRT1 activates the proliferator-activated receptor-γ coactivator-1α/NF-κB signaling axis, mitigating oxidative stress induced by CS in mice with COPD (38). SIRT1 has also been shown to inhibit inflammation in vivo via the Nrf2/p65 NF-κB pathway, thereby reversing oxidative stress and inflammation induced by CS (39). Therefore, the present study explored the mechanisms underlying SIRT1-mediated regulation of AM polarization. SIRT1 mediates the toll-like receptor 4 (TLR4)/NF-κB signaling pathway to regulate the polarization of microglial cells (40). Tetramethylpyrazine improves acute lung injury by inhibiting the TLR4/TRAF6/NF-κB/NLRP3/caspase-1 signaling pathway (41). This suggests a potential association between SIRT1 and TLR4/TRAF6 and their downstream signaling. Previous research has suggested that TRAF6 can interact with NLRP3, thereby activating the NLRP3 signaling pathway (19). IP experiments demonstrated the ability of TRAF6 to interact with NLRP3. The present study revealed overexpression of SIRT1 significantly reversed the increased expression of TRAF6, NLRP3 and cleaved caspase-1 induced by exposure to 20% CSE treatment. Conversely, overexpression of TRAF6 attenuated the effects of SIRT1 overexpression on NR8383 cells, indicating that the regulatory role of SIRT1 in CSE-induced polarization and inflammation of NR8383 cells was mediated by the TRAF6/NLRP3 signaling pathway. However, further experimental validation is required to determine whether this process involves other up- and downstream components of the TRAF6/NLRP3 pathway. Additionally, the present study primarily examined the role of SIRT1 in the polarization of AMs induced by CSE at the cellular level. However, in vitro experiments may not completely replicate the complexity of the in vivo environment. Thus, further studies are warranted to validate the findings of the present study by establishing an animal model of COPD.

In conclusion, the present study conducted a preliminary exploration of the role of SIRT1 in AMs. The overexpression of SIRT1 reduced CSE-induced M1 polarization and inflammatory release in NR8383 cells by inhibiting the TRAF6/NLRP3 signaling pathway.

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation Committee, Regional Science Fund Project (grant no. 82160002) and Guangxi Natural Science Foundation, Youth Science Project (grant no. 2021GXNSFBA220071).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

FY conceptualized the study and wrote the manuscript. HQ was responsible for conceptualization. CQ and BH designed and performed experiments. FG, YL, YT, YM and QY analyzed data and constructed figures. CW designed experiments and edited the manuscript. All authors have read and approved the final manuscript. FY and CW confirm the authenticity of all the raw data.

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 

Celli B, Fabbri L, Criner G, Martinez FJ, Mannino D, Vogelmeier C, Montes de Oca M, Papi A, Sin DD, Han MK and Agusti A: Definition and nomenclature of chronic obstructive pulmonary disease: Time for its revision. Am J Respir Crit Care Med. 206:1317–1325. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Cunha AS, Raposo B, Dias F, Henriques S, Martinho H and Pedro AR: Management of chronic obstructive pulmonary disease: Constraints in patient pathway and mitigation strategies. Port J Public Health. 42:93–100. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Gai X, Allwood B and Sun Y: Post-tuberculosis lung disease and chronic obstructive pulmonary disease. Chin Med J (Engl). 136:1923–1928. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Dorababu A and Maraswami M: Recent advances (2015–2020) in drug discovery for attenuation of pulmonary fibrosis and COPD. Molecules. 28:36742023. View Article : Google Scholar : PubMed/NCBI

5 

Han MK: From conundrum to cures, pioneering breakthroughs in chronic obstructive pulmonary disease research: Introduction to an AJRCCM special issue. Am J Respir Crit Care Med. 208:339–340. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Barnes PJ: Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 138:16–27. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Feng H, Zhang D, Yin Y, Kang J and Zheng R: Salidroside ameliorated the pulmonary inflammation induced by cigarette smoke via mitigating M1 macrophage polarization by JNK/c-Jun. Phytother Res. 37:4251–4264. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Baßler K, Fujii W, Kapellos TS, Dudkin E, Reusch N, Horne A, Reiz B, Luecken MD, Osei-Sarpong C, Warnat-Herresthal S, et al: Alveolar macrophages in early stage COPD show functional deviations with properties of impaired immune activation. Front Immunol. 13:9172322022. View Article : Google Scholar : PubMed/NCBI

9 

Akata K and van Eeden SF: Lung macrophage functional properties in chronic obstructive pulmonary disease. Int J Mol Sci. 21:8532020. View Article : Google Scholar : PubMed/NCBI

10 

Brake SJ, Lu W, Chia C, Haug G, Larby J, Hardikar A, Singhera GK, Hackett TL, Eapen MS and Sohal SS: Transforming growth factor-β1 and SMAD signalling pathway in the small airways of smokers and patients with COPD: Potential role in driving fibrotic type-2 epithelial mesenchymal transition. Front Immunol. 14:12165062023. View Article : Google Scholar : PubMed/NCBI

11 

Feng H and Zheng R: Cigarette smoke prevents M1 polarization of alveolar macrophages by suppressing NLRP3. Life Sci. 327:1218542023. View Article : Google Scholar : PubMed/NCBI

12 

Kato R, Mizuno S, Kadowaki M, Shiozaki K, Akai M, Nakagawa K, Oikawa T, Iguchi M, Osanai K, Ishizaki T, et al: Sirt1 expression is associated with CD31 expression in blood cells from patients with chronic obstructive pulmonary disease. Respir Res. 17:1392016. View Article : Google Scholar : PubMed/NCBI

13 

Li S, Huang Q and He B: SIRT1 as a potential therapeutic target for chronic obstructive pulmonary disease. Lung. 201:201–215. 2023. View Article : Google Scholar : PubMed/NCBI

14 

He B, Zhang W, Qiao J, Peng Z and Chai X: Melatonin protects against COPD by attenuating apoptosis and endoplasmic reticulum stress via upregulating SIRT1 expression in rats. Can J Physiol Pharmacol. 97:386–391. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Li BS, Zhu RZ, Lim SH, Seo JH and Choi BM: Apigenin alleviates oxidative stress-induced cellular senescence via modulation of the SIRT1-NAD[Formula: See text]-CD38 Axis. Am J Chin Med. 49:1235–1250. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Sul OJ, Choi HW, Oh J and Ra SW: GSPE attenuates CSE-induced lung inflammation and emphysema by regulating autophagy via the reactive oxygen species/TFEB signaling pathway. Food Chem Toxicol. 177:1137952023. View Article : Google Scholar : PubMed/NCBI

17 

Rajendrasozhan S, Yang SR, Kinnula VL and Rahman I: SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 177:861–870. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Ren W, Xi G, Li X, Zhao L, Yang K, Fan X, Gao L, Xu H and Guo J: Long non-coding RNA HCG18 promotes M1 macrophage polarization through regulating the miR-146a/TRAF6 axis, facilitating the progression of diabetic peripheral neuropathy. Mol Cell Biochem. 476:471–482. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Wu X, Zhang Y, Zhang Y, Xia L, Yang Y, Wang P, Xu Y, Ren Z and Liu H: MST4 attenuates NLRP3 inflammasome-mediated neuroinflammation and affects the prognosis after intracerebral hemorrhage in mice. Brain Res Bull. 177:31–38. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Wan SY, Li GS, Tu C, Chen WL, Wang XW, Wang YN, Peng LB and Tan F: MicroNAR-194-5p hinders the activation of NLRP3 inflammasomes and alleviates neuroinflammation during intracerebral hemorrhage by blocking the interaction between TRAF6 and NLRP3. Brain Res. 1752:1472282021. View Article : Google Scholar : PubMed/NCBI

21 

Xu SW, Zhang YJ, Liu WM, Zhang XF, Wang Y, Xiang SY, Su JC and Liu ZB: Cigarette smoke extract-induced inflammatory response via inhibition of the TFEB-mediated autophagy in NR8383 cells. Exp Lung Res. 49:39–48. 2023. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Uwagboe I, Adcock IM, Lo Bello F, Caramori G and Mumby S: New drugs under development for COPD. Minerva Med. 113:471–496. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Cheng Y, Wang D, Wang B, Li H, Xiong J, Xu S, Chen Q, Tao K, Yang X, Zhu Y and He S: HMGB1 translocation and release mediate cigarette smoke-induced pulmonary inflammation in mice through a TLR4/MyD88-dependent signaling pathway. Mol Biol Cell. 28:201–209. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Conti V, Corbi G, Manzo V, Pelaia G, Filippelli A and Vatrella A: Sirtuin 1 and aging theory for chronic obstructive pulmonary disease. Anal Cell Pathol (Amst). 2015:8973272015.PubMed/NCBI

26 

Yanagisawa S, Papaioannou AI, Papaporfyriou A, Baker JR, Vuppusetty C, Loukides S, Barnes PJ and Ito K: Decreased Serum Sirtuin-1 in COPD. Chest. 152:343–352. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Cruz T, López-Giraldo A, Noell G, Casas-Recasens S, Garcia T, Molins L, Juan M, Fernandez MA, Agustí A and Faner R: Multi-level immune response network in mild-moderate chronic obstructive pulmonary disease (COPD). Respir Res. 20:1522019. View Article : Google Scholar : PubMed/NCBI

28 

Deng YS, Fan YP, Zheng YQ, Xi LH, Li WY, Liang TW, Huang H and Lin J: Traditional Chinese medicine and active ingredients regulate M1/M2 macrophage polarization balance to treat chronic obstructive pulmonary disease: A review. Zhongguo Zhong Yao Za Zhi. 49:4298–4312. 2024.(In Chinese). PubMed/NCBI

29 

Sun X, Liu Y, Feng X, Li C, Li S and Zhao Z: The key role of macrophage depolarization in the treatment of COPD with ergosterol both in vitro and in vivo. Int Immunopharmacol. 79:1060862020. View Article : Google Scholar : PubMed/NCBI

30 

Le Y, Cao W, Zhou L, Fan X, Liu Q, Liu F, Gai X, Chang C, Xiong J, Rao Y, et al: Infection of Mycobacterium tuberculosis Promotes Both M1/M2 Polarization and MMP production in cigarette smoke-exposed macrophages. Front Immunol. 11:19022020. View Article : Google Scholar : PubMed/NCBI

31 

Mu Q, Wang Q, Yang Y, Wei G, Wang H, Liao J, Yang X and Wang F: HMGB1 promotes M1 polarization of macrophages and induces COPD inflammation. Cell Biol Int. Oct 4–2024.(Epub ahead of print). doi: 10.1002/cbin.12252. View Article : Google Scholar : PubMed/NCBI

32 

Li L, Sun J, Li Q, Sun K and Jiang J: Fritillaria cirrhosa D. Don Alleviates Inflammatory Progression and Suppresses M1 Polarization of Macrophages in Chronic Obstructive Pulmonary Disease. Int Arch Allergy Immunol. 16:1–9. 2024. View Article : Google Scholar

33 

Cao E, Xu J, Gong Y, Yuan J, Chen A, Liu J, Fan Y, Fan X and Kuang X: Effect of the Lipoxin Receptor Agonist BML-111 on cigarette smoke extract-induced macrophage polarization and inflammation in RAW264.7 Cells. Int J Chron Obstruct Pulmon Dis. 18:919–932. 2023. View Article : Google Scholar : PubMed/NCBI

34 

Feng H, Yin Y, Zheng R and Kang J: Rosiglitazone ameliorated airway inflammation induced by cigarette smoke via inhibiting the M1 macrophage polarization by activating PPARγ and RXRα. Int Immunopharmacol. 97:1078092021. View Article : Google Scholar : PubMed/NCBI

35 

Iqbal IK, Bajeli S, Sahu S, Bhat SA and Kumar A: Hydrogen sulfide-induced GAPDH sulfhydration disrupts the CCAR2-SIRT1 interaction to initiate autophagy. Autophagy. 17:3511–3529. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Tang F and Ling C: Curcumin ameliorates chronic obstructive pulmonary disease by modulating autophagy and endoplasmic reticulum stress through regulation of SIRT1 in a rat model. J Int Med Res. 47:4764–4774. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Guan R, Wang J, Cai Z, Li Z, Wang L, Li Y, Xu J, Li D, Yao H, Liu W, et al: Hydrogen sulfide attenuates cigarette smoke-induced airway remodeling by upregulating SIRT1 signaling pathway. Redox Biol. 28:1013562020. View Article : Google Scholar : PubMed/NCBI

38 

Wang S, He N, Xing H, Sun Y, Ding J and Liu L: Function of hesperidin alleviating inflammation and oxidative stress responses in COPD mice might be related to SIRT1/PGC-1α/NF-κB signaling axis. J Recept Signal Transduct Res. 40:388–394. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Deng M, Tong R, Bian Y and Hou G: Astaxanthin attenuates cigarette smoking-induced oxidative stress and inflammation in a sirtuin 1-dependent manner. Biomed Pharmacother. 159:1142302023. View Article : Google Scholar : PubMed/NCBI

40 

Wu J, Hao Z, Wang Y, Yan D, Meng J and Ma H: Melatonin alleviates BDE-209-induced cognitive impairment and hippocampal neuroinflammation by modulating microglia polarization via SIRT1-mediated HMGB1/TLR4/NF-κB pathway. Food Chem Toxicol. 172:1135612023. View Article : Google Scholar : PubMed/NCBI

41 

Jiang R, Xu J, Zhang Y, Zhu X, Liu J and Tan Y: Ligustrazine Alleviate acute lung injury through suppressing pyroptosis and apoptosis of alveolar macrophages. Front Pharmacol. 12:6805122021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2025
Volume 31 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
Yang F, Qin H, Qin C, Huang B, Gao F, Liao Y, Tang Y, Mo Y, Yang Q, Wang C, Wang C, et al: SIRT1 regulates cigarette smoke extract‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway. Mol Med Rep 31: 43, 2025.
APA
Yang, F., Qin, H., Qin, C., Huang, B., Gao, F., Liao, Y. ... Wang, C. (2025). SIRT1 regulates cigarette smoke extract‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway. Molecular Medicine Reports, 31, 43. https://doi.org/10.3892/mmr.2024.13408
MLA
Yang, F., Qin, H., Qin, C., Huang, B., Gao, F., Liao, Y., Tang, Y., Mo, Y., Yang, Q., Wang, C."SIRT1 regulates cigarette smoke extract‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway". Molecular Medicine Reports 31.2 (2025): 43.
Chicago
Yang, F., Qin, H., Qin, C., Huang, B., Gao, F., Liao, Y., Tang, Y., Mo, Y., Yang, Q., Wang, C."SIRT1 regulates cigarette smoke extract‑induced alveolar macrophage polarization and inflammation by inhibiting the TRAF6/NLRP3 signaling pathway". Molecular Medicine Reports 31, no. 2 (2025): 43. https://doi.org/10.3892/mmr.2024.13408