Download

ORIGINAL ARTICLE

Dihydrokaempferol attenuates LPS-induced inflammation and apoptosis in WI-38 cells

Qiao Wang*, Liwen Zhang, Ping Pang

Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China

*Corresponding author: Qiao Wang, Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 83, Feishan Street, Guiyang City, Guizhou Province 550003, China. Email address: [email protected]

Abstract

Background: Globally, pneumonia has been associated as a primary cause of mortality in children aged less than 5 years. Dihydrokaempferol (DHK) has been proposed for being correlated with the process of various diseases. Nevertheless, whether DHK has a role in the progression of infantile pneumonia remains unclear. This study aimed at exploring whether DHK was involved in the progression of infantile pneumonia.

Methods: Human fibroblast cells WI-38 were treated with lipopolysaccharide (LPS). The viability of WI-38 cells was measured via Cell counting kit-8. Reverse transcription-quantitative polymerase chain reaction was used to evaluate the levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α). Western blot analysis revealed the protein levels of IL-1β, IL-6, TNF-α, Bax, and cleaved-caspase 3. Flow cytometry was applied for exploring the apoptosis of WI-38 cells. The concentrations of IL-1β, IL-6, and TNF-α were assessed via enzyme-linked-immunosorbent serologic assay.

Results: DHK modulated the viability of WI-38 cells in infantile pneumonia. Furthermore, we identified that DHK treatment inversely changed LPS induction-mediated elevation on the levels of inflammation biomarkers. Besides, DHK counteracted LPS-induced production of reactive oxygen species (ROS) in WI-38 cells. DHK also decreased LPS-induced elevation of WI-38 cells apoptosis and mediated the levels of apoptosis-associated indexes. Moreover, modulating sirtuin-1 (SIRT1) protein level was lowered by the induction of LPS, and was reversed by DHK treatment. In addition, DHK counteracted LPS induction-mediated elevation of p-p65 and phosphorylated inhibitor of nuclear factor kappa-B kinase subunit alpha (p-IκBα) protein levels.

Conclusion: DHK alleviated LPS-induced WI-38 cells inflammation injury in infantile pneumonia through SIRT1/NF-κB pathway. The results shed light on the implications of DHK on the prevention and treatment of infantile pneumonia.

Key words: dihydrokaempferol, infantile pneumonia, SIRT1, inflammation injury, lipopolysaccharide

Received 7 August 2023; Accepted 23 August 2023; Available online 1 November 2023

DOI: 10.15586/aei.v51i6.971

Copyright: Wang Q, et al.
License: This open access article is licensed under Creative Commons Attribution 4.0 International (CC BY 4.0). http://creativecommons.org/licenses/by/4.0/

Introduction

Globally, pneumonia has been associated as a primary cause of mortality among children aged <5 years, with a mortality rate of 14%.1,2 Although the pneumonia-related mortality in China has decreased, it remains a significant contributor to morbidity among children.3,4 According to the World Health Organization (WHO) data, pneumonia is responsible for more than 900,000 child deaths globally.5 Infantile pneumonia is still a significant clinical and public health problem.6 It is still imperative to identify reliable biomarkers associated with infantile pneumonia to decrease the incidence and mortality of this disease.

As a natural compound from Chinese herbal plant Bauhinia championii (Benth), dihydrokaempferol (DHK) (CAS Number: 480-20-6) is a type of flavonoid with a variety of effects, such as anti-inflammatory and antioxidant stress.79 DHK is considered as one of the essential groups of flavonoids extracted from plants of the Rutaceae family. DHK is found to have significant potential as a therapeutic and pharmaceutical agent, making it an important candidate for the future medicine.10

Previously, DHK was proposed for being correlated with the process of various diseases. For instance, DHK mediates Kelch-like ECH-associated protein 1–nuclear factor erythroid 2-related factor 2 (Keap1/Nrf2) pathway to ameliorate the progression of severe acute pancreatitis.11 CCl4-induced hepatic fibrosis is weakened by DHK by suppressing poly(ADP-ribose) polymerase 1 (PARP-1) and its downstream cytokines and pathways.12 DHK exerts a protective function on liver injury induced by acetaminophen-via modulating sirtuin-1 (SIRT1).13 Nevertheless, whether DHK has a role in the progression of infantile pneumonia remains unclear.

The aim of this work was to appraise the regulatory impacts of DHK on lipopolysaccharide (LPS)-triggered WI-38 cells inflammation injury in infantile pneumonia. In this study, DHK’s participation in the development of infantile pneumonia was analyzed. The results depicted that DHK alleviated lipopolysaccharide (LPS)-induced WI-38 cells inflammation injury in infantile pneumonia through modulating SIRT1/NF-κB pathway. The findings underlined the potential importance of DHK in infantile pneumonia.

Methods

Cell culture and treatment

Added to fetal bovine serum (FBS) (10%; Gibco, Grand Island, NE, USA), gentamycin (50 μg/mL; Invitrogen, Carlsbad, NM, USA), antibiotic/antimycotic solution (100 units; Invitrogen), and Na pyruvate (1 mM), Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, MO, USA) was employed for culture of normal human fibroblast cells WI-38 (ATCC, Manassas, VA, USA). WI-38 is a diploid human cell line composed of fibroblasts derived from female fetal lung tissues during the third trimester of pregnancy. WI-38 is a fibroblast that produces collagen. The cells were incubated in a humidified atmosphere containing 5% CO2 at 37°C. Seeded in six-well plates and incubated overnight, LPS (10 μg/mL) was supplemented to mimic cell model for infantile pneumonia, while cells without LPS treatment served as a negative control. After 24 h, cell suspension was harvested.

DHK (purity > 98.5%) was bought from Meilune (Dalian, China). Different concentrations of DHK (5, 10, and 20 μM) were utilized for treating WI-38 cells.

Cell counting kit-8 (CCK-8) assay

WI-38 cells were seeded in 96-well plates and cultured for 24 h at 37°C in 5% CO2. Subsequently, each well was treated with 10 µL of CCK-8 reagent (Beyotime, China) and incubated for 1 h. The absorbance at 450 nm was evaluated via a microplate reader.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

TRIzol reagent (Invitrogen) was used for extracting RNA from cells. TIANScript RT kit (Tiangen Biotech, Beijing, China) was used for reverse transcription of complementary DNA (cDNA), followed by RT-qPCR using the SYBR® Premix Dimmer Eraser kit (Takara, Dalian, China) on the CFX96 Touch™ RT-PCR detection system (Bio-Rad Laboratories, Hercules, USA). The 2-ΔΔCt method was applied for calculating the relative expression of interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as control, as follows:

IL-1β

F: 5'-AATCTCACAGCAGCATCTCGACAAG-3'

R: 5'-TCCACGGGCAAGACATAGGTAGC-3'

IL-6

F: 5'-AGTTGCCTTCTTGGGACTGATGTTG-3'

R: 5'-GGTATCCTCTGTGAAGTCTCCTCTCC-3'

TNF-α

F: 5'-CCACGCTCTTCTGTCTACTGAACTTC-3'

R: 5'-TGGGCTACGGGCTTGTCACTC-3'

GAPDH

F: 5'-AGGTCGGTGTGAACGGATTTG-3'

R: 5'-GGGGTCGTTGATGGCAACA-3'

Enzyme-linked immunosorbent serological assay (ELISA)

Concentrations of IL-1β, IL-6, and TNF-α were assessed via the following ELISA kits (MultiSciences; Biotech Co., Hangzhou, China): IL-6 (Cat. No. EK206/3-96), IL-1β (Cat. No. EK201B/3-96), and TNF-α (Cat. No. EK282/3-96).

Flow cytometry

Fluorescein isothiocyanate (FITC) Annexin V apoptosis detection kit (BD Biosciences, Franklin Lakes, NJ, USA) was applied for investigating the apoptosis of WI-38 cells. Digested by ethylenediaminetetraacetic acid (EDTA)-free trypsin, WI-38 cells were rinsed in PBS at 4°C. Dyed with FITC Annexin V (5 μL) and propidium iodide (5 μL PI) for 15 min at indoor temperature without light, WI-38 cells were observed using the FACS Calibur flow cytometer (BD Biosciences, San Jose, CA, USA). The results were analyzed via FlowjoV 1.8.1 (Becton, Dickinson, Franklin Lakes, NJ, USA).

Assessment of oxidative stress

The 2',7'-DCF diacetate (DCF-DA; Sigma-Aldrich) fluorescence was applied for assessing oxidative stress. Cultured with 2'-7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) at 37°C for 15 min without light, WI-38 cells were rinsed in a fresh medium and re-suspended using PBS (20 mM, pH 7.0). Fluorescence microscopy (Leica Microsystems GmbH, Wetzlar, Germany) was utilized for visualization, with fluorescence levels detected through a fluorescence microplate reader at 488-nm excitation and 525-nm emission wavelengths.

Western blot analysis

Bicinchoninic acid (BCA) protein kit was used for determining protein concentrations. Proteins were loaded and separated using 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), followed by transferring to polyvinylidene fluoride (PVDF) membranes. Post-sealed by bovine serum albumin (BSA, 5%) at 25°C for 1 h, primary antibodies against IL-1β (1:5000; ab254360), IL-6 (1:5000; ab303458), TNF-α (1:5000; ab183218), Bax (0.477 µg/mL; ab270742), cleaved-caspase 3 (1:500; ab2302), p-p65 (1:1000; ab76302), p65 (1:1000; ab32536), phosphorylated inhibitor of nuclear factor kappa-B kinase subunit alpha (p-IκBα, 1:10,000; ab133462), IκBα (1:1000; ab32518), and β-actin (1:5000; ab8226) (Abcam, Shanghai, China) were supplemented to the membranes overnight at 4°C. Membranes were washed with phosphate-buffered saline solution (PBST), and horseradish peroxidase (HRP)-conjugated affinipure goat anti-rabbit Immunoglobulin G (IgG) secondary antibody was supplemented on the next day. Images of the band were observed using the enhanced chemiluminescence (ECL) detection kit and a chemidoc XRS Imaging system (Bio-Rad Laboratories).

Statistical Analysis

GraphPad Prism 8 was used for data analysis. The data were represented as mean ± standard deviation (SD), with the Student’s t-test used for comparing differences between groups. The data were analyzed attributing to the normal distribution and homogeneity of variance. Differences between multiple groups were analyzed by one-way Analysis of Variance (ANOVA) and Tukey’s multiple comparisons post-test; P < 0.05 was considered statistically significant.

Results

DHK modulated cell viability in infantile pneumonia

The chemical structure of DHK is displayed in Figure 1A. The viability of WI-38 cells was assessed via CCK-8 assay. The data showed that 5-, 10-, and 20-μM DHK treatment had no obvious effect on the viability of WI-38 cells, while 40-μM DHK treatment markedly reduced the viability of WI-38 cells (from 99% to 70%; P < 0.001; Figure 1B). Furthermore, the viability of WI-38 cells was evidently lowered due to LPS treatment (from 99% to 43%, P < 0.001), and 10- and 20-μM DHK treatment reversed the effect of LPS on the viability of WI-38 cells (from 43% to 74% and 85%; P < 0.001; Figure 1C). To sum up, DHK modulated the viability of WI-38 cells in infantile pneumonia.

Figure 1 DHK modulated cell viability in infantile pneumonia. (A) The chemical structure of DHK. (B) CCK-8 assay assessed the viability of WI-38 cells. ***P < 0.001 relative to 0-μM DHK. (C) WI-38 cells viability was measured by CCK-8 assay. ***P < 0.001 relative to control.###P < 0.001 relative to LPS.

DHK attenuated inflammation in WI-38 cells induced by LPS

The effect of DHK on inflammation in infantile pneumonia was evaluated. The messenger RNA (mRNA) levels of IL-1β, IL-6, and TNF-α were elevated because of LPS induction (P < 0.001), and the effects were inversely changed by 5-, 10-, and 20-μM DHK treatment (P < 0.05; Figure 2A). Similarly, the increased concentrations of IL-1β, IL-6, and TNF-α because of the induction of LPS was offset by 5-, 10-, and 20-μM DHK treatment (P < 0.01; Figure 2B). Altogether, DHK attenuated inflammation in WI-38 cells induced by LPS.

Figure 2 DHK attenuated inflammation in WI-38 cells induced by LPS. (A) RT-qPCR evaluated the levels of IL-1β, IL-6, and TNF-α. ***P < 0.001 relative to control.#P < 0.05, ##P < 0.01, and###P < 0.001 relative to LPS. (B) ELISA evaluated the concentrations of IL-1β, IL-6, and TNF-α. ***P < 0.001 relative to control.##P < 0.01 and###P < 0.001 relative to LPS.

DHK inhibited reactive oxygen species (ROS) induced by LPS

The treatment of 5-, 10-, and 20-μM DHK counteracted LPS-induced production of ROS in WI-38 cells (from 1.05 to 5.83; P < 0.001), and there was a tendency to increase amelioration with the increased dose of DHK (from 5.83 to 4.16, 3.19, and 2.15; P < 0.05; Figure 3). Hence, DHK inhibited ROS induced by LPS.

Figure 3 DHK inhibited ROS induced by LPS. DCF-DA fluorescence was applied for the assessment of oxidative stress.

DHK restrained cell apoptosis induced by LPS

Next, the apoptosis of WI-38 cells was detected. The results of flow cytometry revealed that DHK (5 μM, 10 μM, and 20 μM) treatment reversed LPS induction that caused increase in WI-38 cells apoptosis (from 32.98% to 26.46%, 20.56%, and 14.86%; P < 0.01; Figure 4A). In addition, LPS treatment elevated the protein levels of Bax and cleaved-caspase 3 (P < 0.001), while 5-, 10-, and 20-μM DHK treatment decreased Bax and cleaved-caspase 3 protein levels (P < 0.01; Figure 4B). Taken together, DHK restrained LPS-induced cell apoptosis.

Figure 4 DHK restrained cell apoptosis induced by LPS. (A) Apoptosis of WI-38 cells was detected by flow cytometry. ***P < 0.001 relative to control.##P < 0.01 and###P < 0.001 relative to LPS. (B) Protein levels of Bax and cleaved-caspase 3 were evaluated via Western blot analysis. ***P < 0.001 relative to control.##P < 0.01 and###P < 0.001 relative to LPS.

DHK modulated infantile pneumonia via SIRT1/NF-κB pathway

Finally, the potential mechanism associated between DHK and infantile pneumonia was evaluated. SIRT1 protein level was lowered with the induction of LPS (P < 0.001), and was reversed by 5-, 10-, and 20-μM DHK treatment (P < 0.001; Figure 5A). The treatment of 5-, 10-, and 20-μM DHK counteracted LPS induction-mediated elevation of p-p65 and p-IκBα protein levels (P < 0.01; Figure 5B). To conclude, DHK modulated infantile pneumonia via SIRT1/NF-κB pathway.

Figure 5 DHK modulated infantile pneumonia via SIRT1/NF-κB pathway. (A) Protein levels of SIRT1 were analyzed by Western blot analysis. ***P < 0.001 relative to control.###P < 0.001 relative to LPS. (B) Western blot analysis presented the protein levels of p-p65, p65, p-IκBα, and IκBα. ***P < 0.001 relative to control.##P < 0.01 and###P < 0.001 relative to LPS.

Discussion

Pneumonia is a globally prevalent infection causing mortality in children.14,15 Some other rehabilitation strategies, such as exercise regime, can diminish pneumonia;16 such a scheme has anti-inflammatory effects and could be a good strategy to treat pneumonia.17,18 Severe infantile pneumonia can lead to heart failure, encephalitis, and other complications, eventually causing death.1921 Currently, it is important to find more reliable biomarkers linked to infantile pneumonia. DHK was identified as having an important function in severe acute pancreatitis,11 hepatic fibrosis,12 and liver injury,13 but the potential effect of DHK on infantile pneumonia was unknown. The present research examined the regulatory functions of DHK on cell viability, information, ROS, and cell apoptosis in LPS-triggered WI-38 cells. It further evaluated the role of DHK in infantile pneumonia. The data showed that 40-μM DHK treatment markedly reduced the viability of WI-38 cells, and DHK modulated the viability of WI-38 cells in infantile pneumonia. Furthermore, it was identified that DHK treatment inversely changed LPS-induced elevation in the levels of inflammation biomarkers, which suggested that DHK attenuated inflammation in WI-38 cells induced by LPS. DHK counteracted LPS-induced production of ROS in WI-38 cells. DHK also decreased LPS-induced elevation of WI-38 cells apoptosis and mediated the levels of apoptosis-associated indexes, indicating DHK restrained cell apoptosis induced by LPS. On the whole, DHK relieved LPS-induced WI-38 cells inflammation injury in infantile pneumonia.

SIRT1 is a widely expressed and extensively explored member of sirtuin family. SIRT1 is an NAD+-dependent deacetylase that acts as an intracellular regulator of transcriptional activity and various protein functions.22 The protective effect of SIRT1 has been demonstrated in a variety of pathological conditions, including atherosclerosis,23 neurodegenerative diseases,24 and cerebral ischemia.25 Past studies showed that SIRT1 exacerbated pressure overload-induced hypertrophic heart failure by modulating the metabolism of energy.26 Treatment of pinocembrin (flavanone) regulates early brain injury after subarachnoid hemorrhage by modulation of SIRT1.27 Interestingly, SIRT1 is found to mediate several important key targets via deacetylation, including p53 and NF-κB.28 SIRT1 represses inflammatory response through deacetylation of p65 subunit and inhibiting NF-kB activity.29 SIRT1 participates in herpes simplex 1 (HSV-1)-mediated microglial inflammation via NF-κB signaling.30 Notably, SIRT1 is reported for being correlated with infections. For instance, SIRT1 inhibition impairs glycolysis in infectious challenge to potentiate endothelial dysfunction.31 SIRT1 has a role on regulatory mechanisms of bacterial, viral, and parasitic infections.32 However, whether SIRT1 is implicated in infantile pneumonia needs consideration. Herein, SIRT1 protein level was lowered with the induction of LPS, but was reversed by DHK treatment. What’s more, DHK counteracted LPS induction-mediated elevation of p-p65 and p-IκBα protein levels. In summary, DHK modulated infantile pneumonia via SIRT1/NF-κB pathway.

Conclusion

This study initially validated that DHK lessened inflammation injury in LPS-induced WI-38 cells by modulating SIRT1/NF-κB pathway. The results could shed light on the implication of DHK for preventing and treating infantile pneumonia.

Funding

No funding was used in this study.

Competing interests

The authors stated that there was no conflict of interest to declare.

Ethical approval

This article did not require ethical approval, as it contained no research involving human participants or animals performed by any of the authors.

Data availability

The authors declare that all data supporting the findings of this study are available within the paper and any raw data can be obtained from the corresponding author upon request.

Author Contributions

Qiao Wang designed and conducted the study. Qiao Wang, Liwen Zhang, and Ping Pang supervised data collection, analysis, and interpretation. All the authors prepared the manuscript for publication and reviewed its draft. Finally, all the authors read and approved the final manuscript.

REFERENCES

1. World Health Organization (WHO). Pneumonia [Internet]. [cited 2021]. Available from: https://www.who.int/news-room/fact-sheets/detail/pneumonia. Accessed 11 May 2021.

2. Yang A, Chen C, Hu Y, Zheng G, Chen P, Xie Z et al. Application of metagenomic next-generation sequencing (mNGS) using bronchoalveolar lavage fluid (BALF) in diagnosing pneumonia of children. Microbiol Spectr. 2022;10(5):e0148822. 10.1128/spectrum.01488-22

3. Chinese Preventive Medicine Association; Vaccine and Immunology Branch of the Chinese Preventive Medicine Association. Expert consensus on immunoprophylaxis of pneumococcal disease (2020 version). Zhonghua Liu Xing Bing Xue Za Zhi. 2020;54(12):1315–63. 10.3760/cma.j.cn112338-20201111-01322

4. Haesuk Jung, Youngho Seo, Seung Baik Han, Ji Hye Kim, Areum Durey. Risk factors of methicillin-resistant Staphylococcus aureus bacteremic pneumonia in the emergency department. Signa Vitae. 2022;18(5):103–9. https://www.signavitae.com/articles/10.22514/sv.2022.003

5. Wang J, Xu ZH, Lu J. Hospitalization costs for children with pneumonia in Shanghai, China from 2019 to 2020. Hum Vaccin Immunother 2022;18(5):2081459. 10.1080/21645515.2022.2081459

6. Yadav KK Awasthi S. Childhood pneumonia: What's unchanged, and what's new? Indian J Pediatr. 2023;90(7):693–9. 10.1007/s12098-023-04628-3

7. Braune A, Gütschow M, Blaut M. An NADH-dependent reductase from eubacterium ramulus catalyzes the stereospecific heteroring cleavage of flavanones and flavanonols. Appl Environ Microbiol. 2019;85(19):e01233–19. 10.1128/AEM.01233-19

8. Lee JW, Kim NH, Kim JY, Park JH, Shin SY, Kwon YS et al. Aromadendrin inhibits lipopolysaccharide-induced nuclear translocation of NF-κB and phosphorylation of JNK in RAW 264.7 macrophage cells. Biomol Ther (Seoul). 2013;21(3):216–21. 10.4062/biomolther.2013.023

9. Cui S, Cui Y, Li Y, Zhang Y, Wang H, Qin W et al. Inhibition of cardiac hypertrophy by aromadendrin through down-regulating NFAT and MAPKs pathways. Biochem Biophys Res Commun. 2018;506(4):805–11. 10.1016/j.bbrc.2018.10.143

10. Alam F, Mohammadin K, Shafique Z, Amjad ST, Asad MHHB. Citrus flavonoids as potential therapeutic agents: A review. Phytother Res. 2022;36(4):1417–41. 10.1002/ptr.7261

11. Liang X, Hu C, Liu C, Yu K, Zhang J, Jia Y. Dihydrokaempferol (DHK) ameliorates severe acute pancreatitis (SAP) via Keap1/Nrf2 pathway. Life Sci. 2020;261:118340. 10.1016/j.lfs.2020.118340

12. Huang H, Wei S, Wu X, Zhang M, Zhou B, Huang D et al. Dihydrokaempferol attenuates CCl(4)-induced hepatic fibrosis by inhibiting PARP-1 to affect multiple downstream pathways and cytokines. Toxicol Appl Pharmacol. 2023;464:116438. 10.1016/j.taap.2023.116438

13. Zhang J, Hu C, Li X, Liang L, Zhang M, Chen B et al. Protective effect of dihydrokaempferol on acetaminophen-induced liver injury by activating the SIRT1 pathway. Am J Chin Med. 2021;49(3):705–18. 10.1142/S0192415X21500324

14. B K RK, Shrestha S, Adhikari S, Maharjan S et al. Pneumonia among children admitted to the department of medicine in a tertiary care centre: A descriptive cross-sectional study. JNMA J Nepal Med Assoc. 2022;60(253):785–8. 10.31729/jnma.7859

15. Laimoud M, Abdel-moaty M, Elshobary M, Mohamed T, Ahmed W. Predictors of non-invasive ventilation failure in adult patients with cardiac dysfunction presenting with community-acquired pneumonia: An Egyptian multicenter prospective study. Signa Vitae. 2023;19(3):121–31. https://www.signavitae.com/articles/10.22514/sv.2022.078

16. Yang M, Yan Y, Yin X, Wang BY, Dong BR. Chest physiotherapy for pneumonia in adults. Coch Database Syst Rev. 2013;2(2):Cd006338. 10.1002/14651858.CD006338.pub3

17. Bogusławski S, Strzelak A, Gajko K, Peradzyńska J, Popielska J, Marczyńska M et al. The outcomes of COVID-19 pneumonia in children-clinical, radiographic, and pulmonary function assessment. Pediatr Pulmonol. 2023;58(4):1042–50. 10.1002/ppul.26291

18. Udina C, Ars J, Morandi A, Vilaró J, Cáceres C, Inzitari M. Rehabilitation in adult post-COVID-19 patients in post-acute care with therapeutic exercise. J Frailty Aging. 2021;10(3):297–300. 10.14283/jfa.2021.1

19. Zhang J, Wang CH, Liu XJ, Cheng SF, Han LH, Lv CL. Efficacy and safety analysis of dopamine combined with creatine phosphate sodium in the treatment of infantile pneumonia combined with heart failure. J Biol Regul Homeost Agents. 2020;34(6):2103–8. 10.23812/20-300-L

20. Bai D, Han A, Cong S. The effect of down-regulation of CCL5 on lipopolysaccharide-induced WI-38 fibroblast injury: A potential role for infantile pneumonia. Iran J Basic Med Sci. 2018;21(5):449–54. 10.22038/IJBMS.2018.27165.6640

21. Yao YQ, Wang ZW, Ding YX, Yu Y, Jiang WX, Liu XH et al. Effect of Zhifei mixture combined Western drugs on symptoms and signs of children with mycoplasma pneumonia. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2014;34(5):522–5. PMid: 24941836.

22. Wu M, Zhang J, Gu R, Dai F, Yang D, Zheng Y et al. The role of Sirtuin 1 in the pathophysiology of polycystic ovary syndrome. Eur J Med Res. 2022;27(1):158. 10.1186/s40001-022-00746-4

23. Karnewar S, Pulipaka S, Katta S, Panuganti D, Neeli PK, Thennati R et al. Mitochondria-targeted esculetin mitigates atherosclerosis in the setting of aging via the modulation of SIRT1-mediated vascular cell senescence and mitochondrial function in Apoe(-/-) mice. Atherosclerosis. 2022;356:28–40. 10.1016/j.atherosclerosis.2022.07.012

24. Liu H, Xu S, Wang C, Deng Y, Xu B, Yang T et al. The beneficial role of sirtuin 1 in preventive or therapeutic options of neurodegenerative diseases. Neuroscience. 2022;504:79–92. 10.1016/j.neuroscience.2022.09.021

25. Fangma Y, Wan H, Shao C, Jin L, He Y. Research progress on the role of sirtuin 1 in cerebral ischemia. Cell Mol Neurobiol. 2023;43(5):1769–83. 10.1007/s10571-022-01288-3

26. Van Le TN, Zoungrana LI, Wang H, Fatmi MK, Ren D, Krause-Hauch M et al. Sirtuin 1 aggravates hypertrophic heart failure caused by pressure overload via shifting energy metabolism. Biochem Biophys Res Commun. 2022;637:170–80. 10.1016/j.bbrc.2022.11.014

27. Zeng Y, Fang Z, Lai J, Wu Z, Lin W, Yao H et al. Activation of sirtuin-1 by pinocembrin treatment contributes to reduced early brain injury after subarachnoid hemorrhage. Oxid Med Cell Longev. 2022;2022:2242833. 10.1155/2022/2242833

28. Fujita Y, Yamashita T. Sirtuins in neuroendocrine regulation and neurological diseases. Front Neurosci. 2018;12:778. 10.3389/fnins.2018.00778

29. Qian Y, Xin Z, Lv Y, Wang Z, Zuo L, Huang X et al. Asiatic acid suppresses neuroinflammation in BV2 microglia via modulation of the Sirt1/NF-κB signaling pathway. Food Funct. 2018;9(2):1048–57. 10.1039/C7FO01442B

30. Song X, Cao W, Wang Z, Li F, Xiao J, Zeng Q et al. Nicotinamide n-oxide attenuates HSV-1-induced microglial inflammation through sirtuin-1/NF-κB signaling. Int J Mol Sci. 2022;23(24):16085. 10.3390/ijms232416085

31. Stark RJ, Koch SR, Stothers CL, Pourquoi A, Lamb CK, Miller MR et al. Loss of sirtuin 1 (SIRT1) potentiates endothelial dysfunction via impaired glycolysis during infectious challenge. Clin Transl Med. 2022;12(9):e1054. 10.1002/ctm2.1054

32. Kim JK, Silwal P, Jo EK. Sirtuin 1 in host defense during Infection. Cells. 2022;11(18):2921. 10.3390/cells11182921