Skip to main content

Afzelin induces immunogenic cell death against lung cancer by targeting NQO2

Abstract

Background

Lung cancer is one of the most common malignant cancers worldwide. Previous studies have shown that Afzelin, a flavonoid, possesses anticancer activity. The aim of this study was to explore Afzelin’s effect on lung cancer cells and delineate potential anti-cancer mechanism.

Methods

The effect of Afzelin on cell viability, proliferation, and apoptosis of lung cancer cells i.e., A549 and H1299 cells, was studied. The targets for Afzelin in lung cancer were predicted using SwissTargetPrediction, Next, the GO analysis and pathway enrichment were analyzed using String. For in vitro studies, the overexpression plasmid of NQO2, the identified target of Afzelin, was transfected into Afzelin-treated cells to verify the regulatory role of Afzelin on its target and signaling pathway.

Results

In in vitro studies, Afzelin markedly inhibited cell viability, proliferation, and raised apoptotic rate of A549 and H1299 cells. In addition, Afzelin activated endoplasmic reticulum (ER) stress and increased ATP, HMGB1, and CRT levels in lung cancer cells, indicating that Afzelin induced immunogenic cell death (ICD). SwissTargetPrediction identified NQO2 as a target of Afzelin. Further, Afzelin markedly inhibited NQO2 protein expression and in turn, overexpression of NQO2 attenuated the effect of Afzelin on A549 and H1299 cells.

Conclusion

Afzelin inhibits lung cancer progression by targeting NQO2, in turn, activating ER stress and inducing ICD.

Highlight

1. Afzelin suppresses lung cancer cell proliferation and accelerates their apoptosis

2. NQO2 is identified as a potential target for Afzelin in lung cancer cells

3. Afzelin induces endoplasmic reticulum stress and immunogenic cell death via NQO2

Peer Review reports

Background

Lung cancer is the most common cancer in the world and the leading cause of death due to cancer. Worldwide, there are more than 2.2 million new cases and 1.8 million deaths from lung cancer [1]. Pathologically, lung cancer is divided into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) [2]. Among lung cancer patients, NSCLC accounts for about 85% of all cases [3]. Five-year survival rate for advanced NSCLC patients is less than 20% due to difficulties in early diagnosis, drug resistance, and frequent recurrence [4]. Therefore, it is important to find effective drugs against lung cancer.

Afzelin (kaempferol 3-O-rhamnoside, Fig. 1A) is a flavonoid glycoside found in Houttuynia cordata Thunb [5]. In addition, Afzelin is found in a variety of edible plants such as annonoya, peppercorns, dried ginger, lotus, and ginkgo biloba [6]. The anti-inflammatory and antioxidant effects of Afzelin have been found useful in fulminant hepatic failure [7], ultraviolet B-induced skin damage [8], and cognitive function in Alzheimer’s disease [9]. More importantly, Afzelin inhibits breast cancer by stimulating apoptosis [10]. In addition, Afzelin has an anticancer effect on prostate cancer cells [11]. In particular, Afzelin was found to be cytotoxic to lung cancer cells, but the mechanism of cytotoxicity is yet to be established [12].

Fig. 1
figure 1

Afzelin inhibits proliferation and promotes apoptosis of lung cancer cells. A The chemical structure of Afzelin, B viability, C proliferation, D apoptosis, and E expression of Bax, Bcl-2, and cleaved caspase-3 in A549 and H1299 cells. *P < 0.05, **P < 0.01, ***P < 0.001

Immunogenic cell death (ICD) is a unique form of tumor cell death where the cells progress from non-immunogenicity to immunogenicity, triggering tumor immune effects in vivo and leading to cell death [13, 14]. ICD-induced dying cells release DAMPs, including ATP, HMGB1, calreticulin (CRT), and heat shock proteins (such as HSP70 and HSP90), which have been previously identified as specific molecular markers for ICD [15,16,17]. ICD can be triggered in response to various antitumor therapies, including targeted agents, radiation therapy, and chemotherapy [18]. Therefore, we hypothesized that Afzelin may inhibit the development of lung cancer via the induction of ICD. Several studies have investigated the effects of Afzelin on cellular processes and signaling pathways. For example, Jung et al. has shown that Afzelin positively regulates melanogenesis via the p38 MAPK pathway [19]. Jung E et al. also demonstrated that Afzelin protects melanocytes by activating the Nrf2- ARE pathway and inhibiting GSK-3β [20]. In addition, Shin et al. found that Afzelin exerts an antagonistic effect against ultraviolet B-induced cell damage [8]. In addition, Rachmi et al. identified potential targets of Afzelin in inhibiting the migration of triple-negative breast cancer using reverse docking [21]. Although these studies do not directly address ICD, they provide important information about the role of Afzelin in cellular processes and pathway regulation. Further studies could provide a more comprehensive understanding of the possible relationship between Afzelin and ICD.

Therefore, the primary objective of this study was to investigate the potential role of Afzelin in ICD and unveil its underlying molecular mechanisms involved in immune regulation. To achieve this, we conducted a series of experiments to examine the effects of Afzelin on the proliferation and apoptosis of lung cancer cells in vitro. Additionally, we utilized bioinformatic analysis to identify the targets of Afzelin and explored possible mechanisms associated with its action. The findings of our study present novel prospects for the development of promising therapeutic agents targeting lung cancer.

Methods

Cells and treatment

Lung cancer cell lines, A549 and H1299 (Procell), were cultured at 37 °C in RPMI-1640 medium supplemented with 10% FBS. Afzelin (MedChemExpress) was dissolved in ethanol and adjusted to 40, 20, 10, and 0 μM final concentration. Cells were treated with these different concentrations of Afzelin for 48 h at 37 °C.

Cell transfection

The NQO2-overexpressing plasmid (NQO2) and the interfering plasmid (Vector) were transfected into cells using Lipofectamine 3000 (Invitrogen). After culturing for 24 h, cells were treated with 40 μM Afzelin and cultured for an additional 48 h. The overexpression plasmid and vector were obtained from GenePharma.

CCK-8 assay

Cells were seeded in 96-well plates (5 × 103 cells/well) and treated with various concentrations of Afzelin. CCK-8 solution (Beyotime) was added to each well and incubated for 2 h. Fluorescence was measured using a microplate reader at 450 nm.

Colony formation assay

Proliferation of A549 and H1299 cells was determined using a colony formation assay. Cells were treated with Afzelin for 48 h or transfected with NQO2 and then seeded in 6-well plates (103 cells/well). After 10–14 days, cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. Colony number was calculated using a light microscope. In the image analysis process, we utilized ImagineJ software. After converting images to 8-bit format, we adjusted the threshold to isolate cell nuclei. Overlapping nuclei were separated using the “Process-Binary-Watershed” function. We automated particle analysis with the “Analyze-Analyze Particles” tool. To minimize errors, we cross-referenced results with color images, ensuring all visible cells were counted.

Flow cytometry analysis

Cells were treated with Afzelin or transfected with NQO2 for 48 h and collected in a centrifuge tube. The cells were washed with PBS and digested with trypsin cell digestion solution. After centrifugation at 1000 g for 5 min, cells were collected, resuspended and counted to separate 5 × 104 cells. The 5 × 104 cells cells were centrifuged at 1000 g for 5 min and resuspended in Annexin V-FITC binding solution (Beyotime). Next, the cells were mixed with Annexin V-FITC and propidium iodide staining solution and incubated for 20 min at room temperature in the dark. After incubation, the apoptosis rate was measured using the BD C6 Plus flow cytometer (Becton, Dickinson and Company). The BD C6 Plus is a flow cytometry system that allows the measurement and analysis of various cellular parameters, including fluorescence signals emitted by labeled cells. Data acquisition and analysis was performed using BD Accuri C6 PLUS software.

Western blot analysis

The cells were either treated with Afzelin for 48 h or transfected with NQO2. Subsequently, the total protein of A549 and H1299 cells was isolated using RIPA lysis buffer (Beyotime), and the total protein concentration was determined using the BCA kit (Beyotime). Total protein, equivalent to 30 μg, was separated with 10% SDS-PAGE and transferred to PVDF membrane. The membrane was blocked with 5% skin milk and incubated with primary antibody (Bcl-2: ab182858, Abcam; Bax: ab32503, Abcam; cleaved caspase-3: ab32042, Abcam; p- PERK: #3179, CST; p-eIF2α: #3398, CST; GRP78: 11587–1- AP, Proteintech; CHOP: #5554, CST; CRT: #12238, CST; HMGB1: ab18256, Abcam; NQO2: ab137612, Abcam; GAPDH: ab181603, Abcam) overnight at 4 °C. Subsequently, the membrane was incubated with Goat Anti-Rabbit IgG H&L (HRP) (ab205718, Abcam) for 2 h. GAPDH, a housekeeping protein, was used as an internal control. Bands were visualized using the ECL kit (Beyotime) and analyzed using Image-Pro Plus software.

Detection of ATP, HMGB1, and CRT

After treatment with Afzelin for 48 h or transfection with NQO2, the supernatant was centrifuged for assessment of ATP, HMGB1, and CRT levels using the ATP Assay Kit (Beyotime), the ELISA Kit for HMGB1 (Wuhan USCN Business Co., Ltd.), and the Human Calreticulin ELISA Kit (Invitrogen), respectively.

qRT-PCR analysis

Total RNA, from A549 and H1299 cells, was extracted with Trizol reagent (Beyotime) and transcribed into cDNA using the PrimeScript RT kit (Takara). qPCR was performed using BeyoFastTM SYBR Green qPCR Mix (Beyotime). GAPDH, a housekeeping gene, was used as an internal reference. Relative NQO2 mRNA expression was calculated using the 2−ΔΔCT method. The primer sequences were as follows: NQO2 forward primer: 5'-GCTGGTCGGAAGATTGCTGG-3' and reverse primer: 5'-CCTGCCTGCTCAGTTCATCT-3'; GAPDH forward primer: 5'- GCTCTCTGCTCCTCCTGTTC-3' and reverse primer: 5'- GCAGGAGGCATTGCTGATGA-3'.

Immunofluorescence

Cells were treated with Afzelin for 48 h or transfected with NQO2 and then seeded onto slides and incubated overnight. After washing with PBS, cells were fixed with 4% paraformaldehyde and permeabilized using 0.1% Triton X-100 followed by blocking of non-specific sites with 1% BSA. Cells were then incubated overnight with anti-CRT antibody (#12,238, CST) at 4 °C and Goat Anti-Rabbit secondary antibody H&L (ab150077, Abcam) for 2 h at 25 °C. Fluorescence was analyzed using a fluorescence microscope. In the image analysis process, we utilized ImagineJ software. CRT and DAPI cell counts were obtained separately, and the merged image count was derived by calculating the CRT-to-DAPI ratio multiplied by 100.

Differential Scanning Calorimetry (DSC)

The DSC experiment was used to confirm the change in thermal transition point (Tm) after the combination of protein and small molecule i.e., Afzelin. Briefly, the thermal behavior was studied using a differential scanning calorimeter (PerkinElmer) at a heating rate of 10 °C/min. Measurements were made in the heating range of 20–250 °C in N2 [22, 23].

Bioinformatic analysis

To elucidate the potential targets of Afzelin, we employed the SwissTargetPrediction tools (http://swisstargetprediction.ch/) to analyze its target genes. Subsequently, we conducted Gene Ontology (GO) analysis and pathway enrichment using the String database (https://string-db.org), utilizing the enriched genes identified from the SwissTargetPrediction analysis results. To visualize and present the results of the GO analysis and pathway enrichment, we utilized the R package ggplot2. Molecular docking between Afzelin and NQO2 was performed using Molecular Operating Environment software (MOE).

Statistical analysis

Data is presented as mean ± SD and has been analyzed with GraphPad Prism 7.0. The difference between any two groups was analyzed with Student’s t test. Differences between multiple groups were analyzed with one-way ANOVA. A P value of less than 0.05 was considered statistically significant.

Results

Afzelin inhibits cell proliferation and promotes apoptosis of lung cancer cells

CCK-8 assay showed that Afzelin treatment significantly reduced cell viability of A549 and H1299 cells at concentrations ranging from 10 to 40 μM after 48 h (Fig. 1B). In A549 cells, the IC50 values decreased from 68.41 μM at 24 h to 35.47 μM at 48 h and further to 14.07 μM at 72 h. Similarly, in H1299 cells, the IC50 values decreased progressively from 133.44 μM at 24 h to 46.92 μM at 48 h and ultimately to 16.30 μM at 72 h. The CCK-8 experiment was employed to investigate the impact of Afzelin on normal lung epithelial cells. Notably, the results demonstrated that Afzelin did not significantly inhibit the cell viability of 16HBE cells (Figure S1A). Simultaneously, Afzelin demonstrated a lack of toxicity to 16HBE cells at both 24 h (IC50: 211.0042 μM) and 48 h (IC50: 152.2603 μM). Only at 72 h (IC50: 131.3185 μM) did a mild cytotoxic effect become apparent. Colony formation analysis showed that Afzelin treatment drastically reduced the number of colonies (Fig. 1C). In parallel experiments, treatment with Afzelin at concentrations of 10–40 μM significantly accelerated apoptosis (Fig. 1D). In addition, Afzelin treatment significantly decreased Bcl-2 expression while increase in Bax and cleaved caspase-3 levels were observed in A549 and H1299 cells (Fig. 1E).

Afzelin induces ER stress and ICD in lung cancer cells

SwissTargetPrediction (http://swisstargetprediction.ch/) was employed to predict the target gene of Afzelin. Subsequently, the web tool String (https://string-db.org) was used for GO analysis and pathway enrichment based on these target genes (Fig. 2A). Afzelin treatment significantly increased the levels of p-PERK, p-eIF-2α, GRP78, and CHOP in A549 and H1299 cells (Fig. 2B). In addition, Afzelin treatment significantly increased the protein levels of the ICD markers, ATP, CRT and HMGB1 (Fig. 2C). Afzelin treatment markedly increased the concentrations of ATP, CRT, and HMGB1 in the supernatant, indicating that Afzelin induces the release of ATP, CRT, and HMGB1 (Fig. 2D-F). Immunofluorescence experiments suggested that Afzelin promotes the transfer of CRT to the cell membrane (Fig. 2G).

Fig. 2
figure 2

Afzelin induces ER stress and ICD in lung cancer cells. A The biological process analysis for afzelin using String, B expression of ER stress-related proteins, and C ICD markers in A549 and H1299 cells. The levels of (D) ATP, (E) CTR, and (F) HMGB1. G CTR expression was assessed by immunofluorescence. *P < 0.05, **P < 0.01

NQO2 is identified as a potential target of Afzelin in lung cancer cells

NQO2, ranked first in SwissTargetPrediction as a target of Afzelin, and, was selected for further analysis and molecular docking between Afzelin and NQO2 was performed using MOE (Fig. 3A-B). The distance between Afzelin and predicted interacting amino acids was analyzed (Fig. 3C) and the change in the mean melting temperature (Tm) of NQO2 was investigated using DSC. The Tm was significantly lower for NQO2 bound to Afzelin (Fig. 3D). In addition, NQO2 protein expression after Afzelin treatment showed a significant decrease (Fig. 3E).

Fig. 3
figure 3

NQO2 is identified as a potential target of Afzelin in lung cancer cells. A The target gene of Afzelin was predicted using SwissTargetPrediction. B Molecular docking between Afzelin and NQO2 was performed using MOE software. C The distance between Afzelin and the predicted interacting amino acids is shown. D The Tm for NQO2 was determined using DSC. E The NQO2 protein expression in A549 and H1299 cells is shown

Afzelin regulates cell proliferation and apoptosis of lung cancer cells via NQO2

NQO2 overexpression plasmid was transfected into Afzelin-treated A549 and H1299 cells. Western blot and qRT-PCR showed that NQO2 overexpression significantly increased NQO2 levels (Fig. 4A and B). Evidently, Afzelin significantly decreased the NQO2 protein level in comparison to the control group. However, the decreased level of NQO2 protein was significantly restored by NQO2 overexpression (Fig. 4C). The colony formation analysis showed that Afzelin significantly inhibited cell proliferation. Overexpression of NQO2 markedly promoted proliferation, previously inhibited by Afzelin (Fig. 4D). Flow cytometry showed that Afzelin accelerated apoptosis. NQO2 overexpression reduced apoptosis in Afzelin enhanced apoptotic cells (Fig. 4E). These data suggest that overexpression of NQO2 can reverse the effect of Afzelin on proliferation and apoptosis of A549 and H1299 cells.

Fig. 4
figure 4

Afzelin inhibits proliferation and promotes apoptosis of lung cancer cells via NQO2. A NQO2 protein and B its mRNA expression are shown. The NQO2 protein expression (C), proliferation (D), and apoptosis (E) of A549 and H1299 cells is visible. *P < 0.05, **P < 0.01, ***P < 0.001 vs. control group. #P < 0.05, ##P < 0.01 vs. Afzelin plus Vector group

Afzelin induces ER stress and ICD via NQO2

As shown in Fig. 5A, Afzelin treatment significantly increased the levels of ER stress-related proteins in A549 and H1299 cells. NQO2 overexpression decreased Afzelin induced ER stress protein levels. Similarly, levels of CRT and the ICD markers, ATP and HMGB1, were significantly increased after Afzelin treatment and reversed by NQO2 overexpression (Fig. 5B-F). A schematic model of NQO2 function in lung cancer was shown in Fig. 5G.

Fig. 5
figure 5

Afzelin induces ER stress and ICD via NQO2. A ER stress-related proteins, B CRT and HMGB1, C ATP, D CTR, and E HMGB1 expression in A549 and H1299 cells is shown. F The expression of CTR in A549 and H1299 cells was examined by immunofluorescence. G A schematic model of NQO2 function in lung cancer. *P < 0.05 vs control group. #P < 0.05 vs. Afzelin plus Vector group

Discussion

Lung cancer has the highest mortality rate, posing a major threat to people’s health and a huge economic burden on society [1]. Although new treatments have been developed in the past decades, the survival rate of lung cancer patients remains low due to metastases, recurrences or complications [24, 25]. The current study reports that Afzelin inhibits cancer cell proliferation in vitro and induces apoptosis. Further investigation of the mechanism of Afzelin-mediated apoptosis showed that Afzelin specifically inhibits NQO2 expression, thereby activating ER stress and ICD, and ultimately triggering apoptosis.

ICD is a newly reported form of regulated cell death that triggers an adaptive immune response to cancer cells [26]. CRT exposure, HMGB1 and ATP release are known as ICD markers [27, 28]. HMGB1 is passively released from necrotic or damaged cells and transmits a signal for cell death [29]. The extracellularly secreted ATP binds to the receptor on the target cell and attracts macrophages [30]. CRT is a soluble protein in the ER cavity that reaches the cell surface at the pre-death stage and activates phagocytic cells to effectively phagocytose dead cells [31]. In this study, the levels of ATP, CRT, and HMGB1 were significantly increased after Afzelin treatment. Immunofluorescence studies displayed that CRT translocated from intracellular space to membrane after Afzelin treatment. The above results suggest that Afzelin treatment induced ICD in lung cancer cells. Induction of ICD in cancer cells can activate the immune system, thereby inhibiting breast cancer progression [32]. This induction has been reported to effectively inhibit metastasis and tumor growth in gastric cancer [33]. In this study, Afzelin promoted apoptosis of lung cancer cells by inducing ICD, which is also consistent with previous reports.

Furthermore, ICD is divided into two modes according to its induction mechanism: I ICD i.e., an indirect signal and II ICD i.e., direct signal inducing ER stress [34]. Phosphorylation of eIF2α by activated PERK (p-PERK) is the hallmark of ER stress [35,36,37]. Under ER stress, p-PERK phosphorylates EIF2α to p-eIF2α, leading to high expression of the pro-apoptotic transcription factor, CHOP [38]. The chaperone protein, GRP78, is the major regulator of ER homeostasis [39]. In this study, bioinformatic analysis revealed that Afzelin is involved in ER stress. Afzelin promoted the phosphorylation of PERK and EIF2 α and upregulated the protein levels of GRP78 and CHOP. Studies have shown that triggering ER stress can induce lung cancer cell death [40]. Chen P et al. have showed that the induction of ER stress can trigger the death of lung cancer cells induced by Lathyrol [41]. The current results are consistent with the above study and suggest that Afzelin promotes apoptosis of lung cancer cells via activation of ER stress.

In addition, bioinformatic analysis revealed that NQO2 was the target of Afzelin. NQO2, a flavin adenine mononucleotide-dependent quinone oxidoreductase, is ubiquitously found in various tissues such as liver, kidney, brain, heart, and lung [42]. Earlier studies have shown that NQO2 may accelerate the inactivation of cancer-causing orthoquinones and thus, contribute to breast cancer progression [43]. In addition, NQO2 knockout mouse significantly inhibits prostate cancer cell growth [44]. More importantly, elimination of NQO2 enhances apoptosis of lung cancer cells [45]. In this study, Afzelin significantly inhibited the expression of NQO2 in lung cancer cells. Meanwhile, our results also showed that overexpression of NQO2 promoted the proliferation of A549 and H1299 cells, while inhibiting apoptosis, ER stress, and ICD in A549 and H1299 cells (Figure S1B-G). This is also consistent with previous research [45]. Further study found that overexpression of NQO2 apparently attenuated the role of Afzelin in apoptosis of lung cancer cells. In addition, the overexpression of NQO2 counteracts the ER stress induced by Afzelin, resulting in reduced levels of p-PERK and p-eIF2α. This, in turn, affects the protein levels of GRP78 and CHOP, as well as the induction of ICD markers including CRT, HMGB1, and ATP caused by Afzelin. These findings provide compelling evidence that Afzelin activates ER stress and induces ICD through the selective inhibition of NQO2.

Overall, our study highlights the involvement of NQO2 in the effects of Afzelin on lung cancer cells. By targeting NQO2, Afzelin promotes ER stress activation, induces ICD, and inhibits lung cancer proliferation. These findings provide valuable insights into the potential therapeutic implications of Afzelin in lung cancer treatment. Further investigations are warranted to explore the underlying mechanisms and clinical applications of Afzelin-induced inhibition of NQO2 in lung cancer therapy.

Availability of data and materials

The data used to support the findings of this study are available from the corresponding author upon request.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Pan YQ, Xiao Y, Li Z, Tao L, Chen G, Zhu JF, Lv L, Liu JC, Qi JQ, Shao A. Comprehensive analysis of the significance of METTL7A gene in the prognosis of lung adenocarcinoma. Front Oncol. 2022;12:1071100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553(7689):446–54.

    Article  CAS  PubMed  Google Scholar 

  4. Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, Paz-Ares L. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311.

    Article  CAS  PubMed  Google Scholar 

  5. Satthakarn S, Hladik F, Promsong A, Nittayananta W. Vaginal innate immune mediators are modulated by a water extract of Houttuynia cordata Thunb. BMC Complement Altern Med. 2015;15(183):015–0701.

    Google Scholar 

  6. Afendi FM, Okada T, Yamazaki M, Hirai-Morita A, Nakamura Y, Nakamura K, Ikeda S, Takahashi H, Altaf-Ul-Amin M, Darusman LK, et al. KNApSAcK family databases: integrated metabolite-plant species databases for multifaceted plant research. Plant Cell Physiol. 2012;53(2):28.

    Article  Google Scholar 

  7. Lee SB, Kang JW, Kim SJ, Ahn J, Kim J, Lee SM. Afzelin ameliorates D-galactosamine and lipopolysaccharide-induced fulminant hepatic failure by modulating mitochondrial quality control and dynamics. Br J Pharmacol. 2017;174(2):195–209.

    Article  CAS  PubMed  Google Scholar 

  8. Shin SW, Jung E, Kim S, Kim JH, Kim EG, Lee J, Park D. Antagonizing effects and mechanisms of afzelin against UVB-induced cell damage. PLoS One. 2013;8(4):e61971.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Oh SY, Jang MJ, Choi YH, Hwang H, Rhim H, Lee B, Choi CW, Kim MS. Central administration of afzelin extracted from Ribes fasciculatum improves cognitive and memory function in a mouse model of dementia. Sci Rep. 2021;11(1):021–88463.

    Google Scholar 

  10. Diantini A, Subarnas A, Lestari K, Halimah E, Susilawati Y, Supriyatna, Julaeha E, Achmad TH, Suradji EW, Yamazaki C, et al. Kaempferol-3-O-rhamnoside isolated from the leaves of Schima wallichii Korth. inhibits MCF-7 breast cancer cell proliferation through activation of the caspase cascade pathway. Oncol Lett. 2012;3(5):1069–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhu KC, Sun JM, Shen JG, Jin JZ, Liu F, Xu XL, Chen L, Liu LT, Lv JJ. Afzelin exhibits anti-cancer activity against androgen-sensitive LNCaP and androgen-independent PC-3 prostate cancer cells through the inhibition of LIM domain kinase 1. Oncol Lett. 2015;10(4):2359–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fouseki MM, Damianakos H, Karikas GA, Roussakis C, Gupta MP, Chinou I. Chemical constituents from Cordia alliodora and C. colloccoca (Boraginaceae) and their biological activities. Fitoterapia. 2016;115:9–14.

    Article  CAS  PubMed  Google Scholar 

  13. Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202(12):1691–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tesniere A, Panaretakis T, Kepp O, Apetoh L, Ghiringhelli F, Zitvogel L, Kroemer G. Molecular characteristics of immunogenic cancer cell death. Cell Death Differ. 2008;15(1):3–12.

    Article  CAS  PubMed  Google Scholar 

  15. Wittwer C, Boeck S, Heinemann V, Haas M, Stieber P, Nagel D, Holdenrieder S. Circulating nucleosomes and immunogenic cell death markers HMGB1, sRAGE and DNAse in patients with advanced pancreatic cancer undergoing chemotherapy. Int J Cancer. 2013;133(11):2619–30.

    CAS  PubMed  Google Scholar 

  16. Qin J, Kunda N, Qiao G, Calata JF, Pardiwala K, Prabhakar BS, Maker AV. Colon cancer cell treatment with rose bengal generates a protective immune response via immunogenic cell death. Cell Death Dis. 2017;8(2):473.

    Article  Google Scholar 

  17. Huang FY, Dai SZ, Xu WT, Xiong W, Sun Y, Huang YH, Wang JY, Lin YY, Chen H, Tan GH, et al. 3’-epi-12β-hydroxyfroside-mediated autophagy degradation of RIPK1/RIPK3 necrosomes leads to anergy of immunogenic cell death in triple-negative breast cancer cells. Pharmacol Res. 2022;187:106613.

    Article  PubMed  Google Scholar 

  18. Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, Zhao L, Spisek R, Kroemer G, Galluzzi L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):020–03221.

    Article  Google Scholar 

  19. Jung E, Kim JH, Kim MO, Jang S, Kang M, Oh SW, Nho YH, Kang SH, Kim MH, Park SH, et al. Afzelin positively regulates melanogenesis through the p38 MAPK pathway. Chem Biol Interact. 2016;254:167–72.

    Article  CAS  PubMed  Google Scholar 

  20. Jung E, Kim JH, Kim MO, Lee SY, Lee J. Melanocyte-protective effect of afzelin is mediated by the Nrf2-ARE signalling pathway via GSK-3β inactivation. Exp Dermatol. 2017;26(9):764–70.

    Article  CAS  PubMed  Google Scholar 

  21. Rachmi E, Purnomo BB, Endharti AT, Fitri LE. Identification of afzelin potential targets in inhibiting triple-negative breast cancer cell migration using reverse docking. Porto Biomed J. 2020;5(6):e095.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mahajan NM, Malghade AD, Dumore NG, Thenge RR. Design and development of crystallo-co-agglomerates of Ritonavir for the improvement of physicochemical properties. Turk J Pharm Sci. 2018;15(3):248–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lund BA, Thomassen AM, Carlsen TJO, Leiros HKS. Structure, activity and thermostability investigations of OXA-163, OXA-181 and OXA-245 using biochemical analysis, crystal structures and differential scanning calorimetry analysis. Acta Crystallogr F Struct Biol Commun. 2017;73(Pt 10):579–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bai Y, Liu X, Qi X, Peng F, Li H, Fu H, Pei S, Chen L, Chi X, Zhang L, et al. PDIA6 modulates apoptosis and autophagy of non-small cell lung cancer cells via the MAP4K1/JNK signaling pathway. EBioMedicine. 2019;42:311–25.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Bade BC, Dela Cruz CS. Lung cancer 2020: epidemiology, etiology, and prevention. Clin Chest Med. 2020;41(1):1–24.

    Article  PubMed  Google Scholar 

  26. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486–541.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17(2):97–111.

    Article  CAS  PubMed  Google Scholar 

  28. Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, Chan TA, Coukos G, Demaria S, Deutsch E, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 2020;8(1):2019–000337.

    Article  Google Scholar 

  29. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191–5.

    Article  CAS  PubMed  Google Scholar 

  30. Kronlage M, Song J, Sorokin L, Isfort K, Schwerdtle T, Leipziger J, Robaye B, Conley PB, Kim HC, Sargin S, et al. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci Signal. 2010;3(132):2000588.

    Article  Google Scholar 

  31. Ahmed A, Tait SWG. Targeting immunogenic cell death in cancer. Mol Oncol. 2020;14(12):2994–3006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Calvillo-Rodríguez KM, Mendoza-Reveles R, Gómez-Morales L, Uscanga-Palomeque AC, Karoyan P, Martínez-Torres AC, Rodríguez-Padilla C. PKHB1, a thrombospondin-1 peptide mimic, induces anti-tumor effect through immunogenic cell death induction in breast cancer cells. Oncoimmunology. 2022;11(1):2054305.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Xiao Y, Yao W, Lin M, Huang W, Li B, Peng B, Ma Q, Zhou X, Liang M. Icaritin-loaded PLGA nanoparticles activate immunogenic cell death and facilitate tumor recruitment in mice with gastric cancer. Drug Deliv. 2022;29(1):1712–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12(12):860–75.

    Article  CAS  PubMed  Google Scholar 

  35. Tabas I, Ron D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol. 2011;13(3):184–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rufo N, Garg AD, Agostinis P. The unfolded protein response in immunogenic cell death and cancer immunotherapy. Trends Cancer. 2017;3(9):643–58.

    Article  CAS  PubMed  Google Scholar 

  37. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2000;2(6):326–32.

    Article  CAS  PubMed  Google Scholar 

  38. Xu J, Zhou Q, Xu W, Cai L. Endoplasmic reticulum stress and diabetic cardiomyopathy. Exp Diabetes Res. 2012;827971(10):24.

    Google Scholar 

  39. Lee AS. Glucose-regulated proteins in cancer: molecular mechanisms and therapeutic potential. Nat Rev Cancer. 2014;14(4):263–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang J, Du T, Lu Y, Lv Y, Du Y, Wu J, Ma R, Xu C, Feng J. VLX1570 regulates the proliferation and apoptosis of human lung cancer cells through modulating ER stress and the AKT pathway. J Cell Mol Med. 2022;26(1):108–22.

    Article  CAS  PubMed  Google Scholar 

  41. Chen P, Li Y, Zhou Z, Pan C, Zeng L. Lathyrol promotes ER stress-induced apoptosis and proliferation inhibition in lung cancer cells by targeting SERCA2. Biomed Pharmacother. 2022;158:114123.

    Article  PubMed  Google Scholar 

  42. Long DJ 2nd, Jaiswal AK. NRH:quinone oxidoreductase2 (NQO2). Chem Biol Interact. 2000;129(1–2):99–112.

    Article  CAS  PubMed  Google Scholar 

  43. Gaikwad NW, Yang L, Rogan EG, Cavalieri EL. Evidence for NQO2-mediated reduction of the carcinogenic estrogen ortho-quinones. Free Radic Biol Med. 2009;46(2):253–62.

    Article  CAS  PubMed  Google Scholar 

  44. Hsieh TC, Yang CJ, Lin CY, Lee YS, Wu JM. Control of stability of cyclin D1 by quinone reductase 2 in CWR22Rv1 prostate cancer cells. Carcinogenesis. 2012;33(3):670–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang J, Zhou Y, Li N, Liu WT, Liang JZ, Sun Y, Zhang WX, Fang RD, Huang SL, Sun ZH, et al. Curcumol overcomes TRAIL resistance of non-small cell lung cancer by targeting NRH: quinone oxidoreductase 2 (NQO2). Adv Sci. 2020;7(22):2002306.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

L X and F C are responsible for experimental implementation; XQ X is responsible for collating data; MJ L and XY Z are responsible for data compilation and statistics; L X and F C are responsible for experimental design and technical guidance.

Corresponding author

Correspondence to Fang Cao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

Impact of Afzelin on 16HBE cells and NQO2 on Proliferation, Apoptosis, ER Stress, and ICD in A549 and H1299 cells. (A) The CCK-8 assay was employed to assess cell viability in normal lung epithelial cells. (B) Colony assay was performed to evaluate the formation of colonies. (C) The detection of apoptosis in A549 and H1299 cells. (D) ER stress-related proteins were examined by western blot. (E-G) ELISA kits were used to measure the levels of ATP (E), CTR (F), and HMGB1 (G) in A549 and H1299 cells. *P < 0.05 vs vector group.

Additional file 2.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xia, L., Xu, X., Li, M. et al. Afzelin induces immunogenic cell death against lung cancer by targeting NQO2. BMC Complement Med Ther 23, 381 (2023). https://doi.org/10.1186/s12906-023-04221-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-023-04221-3

Keywords