Skip to main content
  • Research Note
  • Open access
  • Published:

Naringenin-loaded nanoparticles modulate HIF-driven oxygen-sensing pathways in lung adenocarcinoma cells

Abstract

Background

Hypoxia is a common symptom of lung cancer. Proliferation and neovascularization mediated by hypoxia-inducible factors (HIF) influence several adaptations. It has recently been established that naringenin (NAR) and its nanoparticles are chemo-preventive flavonoids in lung cancer.

Aim

Adjust HIF activity by reviving oxygen-sensing enzyme activity while considering possible therapeutic targets.

Method

The bindings of NAR to target proteins were examined using computational modeling techniques. Additionally, NAR nanoparticles (NARNPs) were synthesized and characterized. Normal fibroblast cells and A549 cells were used to determine cytotoxicity. Colorimetric analysis of α-ketoglutarate detection for hydroxylases.

Results

According to molecular modeling, NAR and target proteins have a high affinity. The PHD and FIH activities in A549 are significantly stimulated.

Conclusion

NAR and NARNPs diminish hypoxia in lung cancer by stimulating oxygen-sensing hydroxylases.

Peer Review reports

Introduction

Hypoxia, or the lack of oxygen, is a characteristic of lung tumors that encourage genomic instability, increased aggressiveness, and metastasis. One of these causes is the disruption of the normal oxygen-sensing mechanisms, which causes cells to respond as though there is insufficient oxygen [1]. Hypoxia-inducible factors (HIFs), transcription factors whose levels are (typically) extremely sensitive to changes in O2 tension, are important cellular regulators of chronic O2 homeostasis [2]. HIF is a heterodimeric transcription factor, and in human cells, the amount of the HIF-1α domain is modulated by enzymes called HIF hydroxylases [3]. The oxygen-dependent degradation domains (ODDs) at the N- and C-termini of HIF-1, Pro 402, and Pro 564 are respectively hydroxylated by the prolyl hydroxylase (PHDs) under normal oxygen circumstances. Due to these changes, the von Hippel-Lindau tumor suppressor protein (VHL) interacts more favorably with other proteins for ubiquitination and proteasome-mediated destruction [4]. On the other hand, Asn803 in the CAD (C-terminal activation domain) of HIF-1 undergoes asparagine hydroxylation, which is catalyzed by factor inhibiting hypoxia (FIH), preventing interaction between HIF- and the co-transcriptional activator, p300 [5]. HIF hydroxylase activity decreases, HIF-α levels rise, and it can dimerize with HIF-β, to increase the activity of genes implicated in the response to hypoxia. Consequently, the identification of a novel drug that specifically targets the HIF-1-regulated pathway represents a promising chemotherapeutic approach for human lung cancer treatment. Naringenin (NAR), a common flavonoid, has been proposed as an antioxidant as well as having anti-cancer characteristics [6]. It has been demonstrated to have an anti-carcinogenic impact on lung cancer cells by altering the processes of cell proliferation, apoptosis, and metastasis because of its antioxidant activity [7, 8]. However, the hydrophobic and crystalline structures are primarily responsible for its instability, limited oral bioavailability, and water solubility. Additionally, it lacks targeting and is susceptible to rapid breakdown in an acidic environment. These disadvantages make it difficult to use it medicinally effectively [7]. Nevertheless, encapsulation NAR helps to shield them from metabolic activity, to encourage controlled and prolonged drug release, and to improve their bioavailability. Chitosan (CS) is utilized to encapsulate the flavonoid NAR due to its unique properties [9]. A naturally occurring polymer CS is produced when chitin undergoes partial N-deacetylation [10]. Because of its special qualities such as hydrophilicity, non-toxicity, biodegradability, biocompatibility, and affordability, it is employed as a nanocarrier. It can interact with different epithelia and the potential for cohesion. Additionally, the CS-NPs shield the drugs from outside influences like pH and enzymes [10]. Because they have increased surface area to absorb, bind, and transport the medicines, nano encapsulations of CS polymers are efficient drug delivery vehicles. By encouraging the ionic interaction of CS amino groups with their anionic groups, tripolyphosphate (TPP) can be used as a cross-linker and raise the solution’s ionic strength [11]. Ionic cross-linking with TPP resulted in CS-NPs that demonstrated improved drug loading efficiency and a longer drug release duration. Consequently, our studies aim to target and investigate the effect of HIF-1-regulated hydroxylases by loading NAR on CSNPs as drug carriers as it’s a promising chemotherapeutic approach for the treatment of hypoxia in lung cancer.

Methods

In silico studies & Drug likeness/pharmacophore and ADMET profile analysis

The interaction of NAR with proteins involved in the HIF-regulated pathway proteins was studied by molecular docking through the methodology proposed in [12].

Nanoparticles preparation

CS-NPs were prepared by the ionic gelation technique. Briefly, a concentration of 0.5 mg/mL of NAR was added to CS to TPP ratios of 5:1 (w/w), which were then agitated to achieve complete dissolution at pH 4.8 [7].

Characterization ofs NARNP

A UV-Vis 2550 spectrometer (Shimadzu) [13], Fourier transform infrared (FTIR) spectroscopy (JASCO, FT/IR-6100) [14], XRD analysis (GNR, APD2000PRO, Italy) [15]. A transmission electronic microscope (TEM) (JEOL-2100) [16], SEM connected to a JEOL JSM-6510 LV energy dispersive spectroscopy (EDS) device [17]. Zetasizer Nano Series instrument (Horiba Nano partica SZ-100, Japan), the Zeta potential (ζ) of the mixture was determined [18]. The release study of NAR from NARNPs was made using formula at 37 °C and in 1X PBS pH 7 [7].

Cell viability and cell proliferation assays

Briefly, on 96-well plates, 2 × 103 human pulmonary adenocarcinoma A549 cells (# ATCC CCL-185) and normal lung fibroblast cells WI-38 (# ATCC WI-38)/samples were planted, and after 48 h, they were exposed to varying doses of NAR, NARNPs, and 5-FLU. As described [19].

Prolyl hydroxylase (PHD) & asparagine hydroxylase (FIH) activity assays

Briefly, five mature male Swiss albino mice weighing between 28 and 30 g were acquired from the Faculty of Pharmacy, Alexandria University, in Egypt. The mice were kept in an appropriate temperature range with a 12-hour light/dark cycle. Following intraperitoneal (i.p.) administration of sodium pentobarbital anesthesia (300 mg/kg) [20]. The mice were cervically dislocated to finish the experiment. From the peritoneal cavity, the liver was extracted, and the gallbladder was then found and extracted using a scalpel. Moreover, the cytosol fraction was isolated from 60 × 106 for A549 and normal cells in accordance using the previously mentioned procedure [14].The Guidelines for the Care and Use of Laboratory Animals published by the National Institutes of Health were adhered to in the care of the experimental animals. The Egyptian Ethical Committee of Tanta University, Faculty of Science (#IACUC-SCI-TU-0287) oversaw the animal studies. Time-course assays were carried out in the absence/presence of IC50 of NAR, NARNPs, and 5-FLU. Initially, 100µL of 5mM α-ketoglutarate, 100µL (500 Mm) HEPES, 100µL (20 mM) Dithiothreitol, 100µL 1.5 Mm FeSO4, 100µL (5mM) ascorbic acid, 100µL (5mM) proline for PHD or 5mM Aspartic acid for FIH activities were added to prepare the in vitro hydroxylation assay [19].

Results

Molecular docking

Auto Dock Vina was believed to be the primary virtual screening platform. As shown in Fig. 2, the binding affinity of NAR with the targets PHD, FIH, and VHL was − 10.1 kcal/mol, − 8.1 kcal/mol, and − 8.3 kcal/mol, in that order (Table S4, Fig. 2). Furthermore, NAR was shown to have good drug-likeness qualities, target precision, and safety for human use [21].

Fig. 1
figure 1

2D representations of the NAR and active pocket interaction (A) PHD (B) FIH (C) VHL-like protein’s active site at left and right sides respectively

Naringenin-loaded nanoparticles characterization results (NARNPs)

Over time, the NAR release profile from NARNPs in vitro was evaluated. As demonstrated, 80% of NAR released from NARNPs in 20 h in Fig. 2A. Positive zeta potential is frequently seen in chitosan nanoparticles. The total positive zeta potential of our formulations varied from + 30 mV to + 50 mV as in Fig. 2B. A characteristic absorption band that appears between 3,310 and 3,120 cm− 1 in the case of NAR alone Fig. 2C. NAR demonstrated the crystalline nature of the material in an X-ray diffractogram. The amorphous nature of chitosan nanoparticles was revealed in Fig. 2D. The form and size of nanoparticles varied between 19 and 40 nm, with a spherical shape and a smooth surface Fig. 2E. SEM analysis was used to evaluate the size, surface morphology, and homogeneity of NARNPs as demonstrated in Fig. 2F.

Fig. 2
figure 2

Illustrates nanoparticles characterization (A) Fourier transform - infrared spectroscopy of NARNPs, (B) The XRD pattern of NARNPs, NAR, and CS nanoparticles, in that sequence (C) The in vitro release profile of NAR from the NARNPs, (D) The zeta potential of NAR nanoparticles, (E) TEM micrograph displaying chitosan nanoparticles (CNPs) on the left side with a size range of 30–40 nm and NARNPs on the right side with a size range of 19–40 nm, (F) The surface structure of NAR-chitosan nanoparticles

Cell viability

The MTT method was used to measure cell growth. Next, the results were contrasted with those attained with 5-FLU acting as a positive control. There was a substantial (p ≤ 0.01) drop in the viability cells as the treatment concentration increased. The IC50 values of NAR, NARNPs, CNPs, and 5-FLU in A549 cells were 61, 35.2, 394, and 5.5 µg/mL respectively, whereas the IC50 values of these compounds were 461, 71, 311, and 8.07 µg/mL respectively in normal fibroblasts.(Table S1).

PHD & FIH activities

NAR, NARNPs, and 5-FLU significantly stimulate PHD and FIH activities in A549 cells with sequence (NARNPs > NAR > 5-FLU) as shown in (Table S2, Fig. 3A). On the other hand, there is a slight decrease in both activities in normal cells and mice as in (Table S3, Fig. 3B, C).

Fig. 3
figure 3

The influences of NAR, NARNPs, and 5-FLU on PHD and FIH activities (A) Relative activities are upsurged in sequence (NARNPs > NAR > 5-FLU) in A549 for both PHD and FIH. (B) Normal, there was significant fluctuation for both PHD and FIH (5-FLU > NAR > NARNPs) (NAR > NARNPs > 5- FLU), respectively (C) Normal mice, (NAR = 5-FLU > NARNPs) towards activities. Where ****p ≤ 0.0001, ***P ≤ 0.001, **P ≤ 0.01, *P ≤ 0.05, # for non- significant against control and each other

Discussion

Hypoxia is the main hallmark of lung cancer which is the primary cause of cancer-related deaths globally and is still a difficult public health concern [22]. Due to its pivotal function in the adaptive reaction to hypoxia, the oxygen-sensing pathway has garnered significant attention as a potential therapeutic target for lung cancer. We can forecast the binding behavior and affinity between NAR and our target proteins PDH, FIH, and VHL that may occur during the non-covalent binding process by molecular docking, Moreover, the NAR-PDH, FIH, and VHL proteins respective Gibbs free energy values of -10.1, -8.1, and − 8.3 kcal/mol were determined, confirming their high affinity and spontaneous complex formation [23]. It has been demonstrated that NAR possesses good drug-likeness properties, target accuracy, and human safety [21]. To prepare CS-NPs, TPP was cross-linked ionically at a ratio of 1 to 2.5. While Kumar, Birundha, et al. (2015) showed that a CS: TPP ratio at 5:1 in 0.5 mg/ml of NAR concentration displayed a significantly higher percentage of encapsulation efficiency (~ 70%), our formulation with a 1:2.5 CS: TPP ratio at 0.5 mg/ml by ~ 96%. The surface charge of the particles, as measured by zeta potential, our formulation agreed with other previous studies which increased from + 22.73 mV to + 58.43 mV [7]. High positive charge particles can penetrate tissues and adhere to mucous membranes, making them more stable. In terms of drug release our formulation showed 80% NAR release from NARNPs in 20 h. In prior studies, depending on the mass ratio, the release of entrapped NAR from CS-NPs was found to be between 45 and 55% [24, 25]. The molecular interactions of the CS-NPs with NAR were obtained from FT-IR suggesting that hydrogen bond interactions linked the NAR to the CS-NPs [7, 26]. We confirmed the structure by using an X-ray diffractogram, and NAR showed that the material was crystalline. It was discovered that chitosan nanoparticles are amorphous. It showed a prominent peak at 22°. Our nanoparticles had a smooth surface, spherical shape, and varied in size from 19 to 40 nm. The size of NARNPs, surface shape, and homogeneity were assessed by SEM analysis with a ratio of 1:2.5. This result was contrary to previous studies were found that the average diameter of 5:1 ratio CS-NPs was 53.2 nm with an average diameter of 407.47 nm, the encapsulated NAR in CS-NPs (CS-NPs/NAR) revealed spherical-shaped NPs [7]. It has been observed that when NAR is encapsulated, its characteristics are preserved. The cytotoxicity impact in A549 cells indicated that free and nano formulations of NAR had cytotoxic effects on cancer cells while normal had no cytotoxicity this study agreed with the prior study [27]. PDH and FIH activities in A549 cells are considerably stimulated by NAR, NARNPs, and 5-FLU in the order (NARNPs > NAR > 5-FLU), respectively. The quantity of 2-OG that is still present in the reaction mixture has a direct relationship with the intensity [19]. These results were supported by previous studies that indicated flavonoids decreased the stability of HIF-1α, with enhanced prolyl hydroxylation, which causes it to be more susceptible to ubiquitination and destruction [8, 28]. In contrast to the inhibition of HIF prolyl hydroxylases (PHDs), there has been less research on the activators of FIH [29]. For the first time, our data support theoretically strong affinity for FIH active site with binding affinity − 8.1, and practically the relative activity was 18.2%, 13%, and 12.4% for NARNPs, NAR, and 5-FLU, respectively in A549.

Limitations

Scientific studies suggest that the likelihood of serious side effects occurring is low and that its profile should be safer than that of other chemotherapy drugs. We predicted that NAR-CSNPs will be a promising activator to lessen hypoxia in lung cancers for the foreseeable future. Additionally, chitosan nanoparticles coated with NAR are opening the door for the development of innovative hypoxic lung cancer therapies. However, further studies have needed for genetic and epigenetic investigations in vivo and in vitro are required to be comprehended in regulated HIF pathways.

Conclusion

Lung cancer was estimated as a hypoxic state which obligates the cell to tumorigenesis. Therefore, Oxygen-sensing prolyl hydroxylase (PHD) is responsible for proteasomal degradation, and factor inhibiting hypoxia (FIH) for transcription activity could be a therapeutic target for hypoxia in lung cancer. Our recent studies were done on the effect of NAR and its nanoparticles theoretically and practically on PHD, FIH. Our molecular modeling showed a strong affinity between NAR and target proteins. Further enzymatic activity assays showed an upsurge in PHD and FIH hydroxylase enzymes. We conclude that NAR and NARNPs efficiently alleviate oxygen-sensing HIF hydroxylases through the hypoxic pathway in lung cancer.

Data availability

All data generated or analysed during this study are included in this published article [and its supplementary information files].

Abbreviations

HIF1:

Hypoxia-Inducible Factor 1

ODD:

Oxygen-dependent degradation domains

CAD:

C-terminal activation domain

PHDs:

Prolyl hydroxylase

VHL:

Von Hippel-Lindau tumor suppressor protein

FIH:

Factor Inhibiting Hypoxia

NAR:

Naringenin

NARNPs:

Naringenin Nanoparticles

5-FLU:

5-Fluorouracil

References

  1. Saggese P, et al. Glucose deprivation promotes Pseudohypoxia and Dedifferentiation in Lung Adenocarcinoma. Cancer Res. 2024;84(2):305–27.

    Article  PubMed  CAS  Google Scholar 

  2. Rani S et al. Regulation of transactivation at C-tad domain of HIF-1α by factor-inhibiting HIF-1α (FIH-1): A potential target for therapeutic intervention in cancer. Oxidative Medicine and Cellular Longevity, 2022. 2022.

  3. Lippl K et al. Born to sense: biophysical analyses of the oxygen sensing prolyl hydroxylase from the simplest animal Trichoplax adhaerens. Hypoxia, 2018: pp. 57–71.

  4. Ortmann BM. Hypoxia-inducible factor in cancer: from pathway regulation to therapeutic opportunity. BMJ Oncol, 2024. 3(1).

  5. Ma Q, et al. Acetyltransferase P300 regulates glucose metabolic reprogramming through catalyzing Succinylation in Lung Cancer. Int J Mol Sci. 2024;25(2):1057.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Azeredo PdS, et al. Potential of plant-derived compounds in preventing and reversing Organ Fibrosis and the underlying mechanisms. Cells. 2024;13(5):421.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Kumar SP, et al. Antioxidant studies of chitosan nanoparticles containing naringenin and their cytotoxicity effects in lung cancer cells. Int J Biol Macromol. 2015;78:87–95.

    Article  PubMed  CAS  Google Scholar 

  8. Ragab EM, et al. Comprehensive overview of how to fade into succinate dehydrogenase dysregulation in cancer cells by naringenin-loaded chitosan nanoparticles. Genes Nutr. 2024;19(1):10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Divya K, Jisha M. Chitosan nanoparticles preparation and applications. Environ Chem Lett. 2018;16:101–12.

    Article  CAS  Google Scholar 

  10. Mamah SC, et al. The utilization of chitin and chitosan as green modifiers in nanocomposite membrane for water treatment. J Water Process Eng. 2024;62:105394.

    Article  Google Scholar 

  11. Hamedinasab H et al. Formulation, optimization, and characterization of naringeninloaded halloysite nanotube to achieve enhanced antioxidant and anticancer properties. Nanomed J, 2025. 12(1).

  12. Geng PX. Characterizing the Binding Mechanism of Sparsentan to the Type-2 Angiotensin II Receptor Using AutoDock Vina. bioRxiv, 2024: p. 2024.03. 22.586129.

  13. Vemuri SK, et al. Novel biosynthesized gold nanoparticles as anti-cancer agents against breast cancer: synthesis, biological evaluation, molecular modelling studies. Mater Sci Engineering: C. 2019;99:417–29.

    Article  CAS  Google Scholar 

  14. Ragab EM, et al. Impairment of electron transport chain and induction of apoptosis by chrysin nanoparticles targeting succinate-ubiquinone oxidoreductase in pancreatic and lung cancer cells. Volume 18. Genes & Nutrition; 2023. pp. 1–15. 1.

  15. Zhao W, et al. Green synthesis of gold nanoparticles from Dendrobium officinale and its anticancer effect on liver cancer. Drug Delivery. 2021;28(1):985–94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Nisha UM, et al. Assessment of solar light sensitive Chitosan integrated CeO2-CuO ternary composites for the efficient degradation of Malachite Green, Acid Blue 113 dyes and microbial studies. Inorg Chem Commun. 2024;160:111942.

    Article  Google Scholar 

  17. Sargazi, S., et al., Application of Green Gold Nanoparticles in Cancer Therapy and Diagnosis. Nanomaterials 2022, 12, 1102. 2022, s Note: MDPI stays neutral with regard to jurisdictional claims in published&#8230.

  18. Voci S, et al. Ascorbic acid-loaded gliadin nanoparticles as a novel nutraceutical formulation. Food Res Int. 2022;161:111869.

    Article  PubMed  CAS  Google Scholar 

  19. Wong SJ et al. Development of a colorimetric α-ketoglutarate detection assay for prolyl hydroxylase domain (PHD) proteins. J Biol Chem, 2021. 296.

  20. Albasher G, et al. Protective effects of Artemisia Judaica extract compared to metformin against hepatorenal injury in high-fat diet/streptozotocine-induced diabetic rats. Environ Sci Pollut Res. 2020;27:40525–36.

    Article  CAS  Google Scholar 

  21. Supian S, et al. Molecular docking and in silico drug-likeness studies of Garcinia Prainiana King bioactive compounds as pancreatic lipase inhibitors. Food Res. 2022;6(2):83–90.

    Article  Google Scholar 

  22. Cords L et al. Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell, 2024.

  23. Pan J, et al. Naringenin protects against septic cardiomyopathy in mice by targeting HIF-1α. Biochem Biophys Res Commun. 2024;149613:p.

    Google Scholar 

  24. Hu B, et al. Optimization of fabrication parameters to produce chitosan– tripolyphosphate nanoparticles for delivery of tea catechins. J Agric Food Chem. 2008;56(16):7451–8.

    Article  PubMed  CAS  Google Scholar 

  25. Winarti L, Sari LORK, Nugroho AE. Naringenin-loaded chitosan nanoparticles formulation, and its in vitro evaluation against T47D breast cancer cell line. 2015.

  26. Li H, et al. Reaction mechanism of nanomedicine based on porphyrin skeleton and its application prospects. Photodiagn Photodyn Ther. 2023;41:103236.

    Article  CAS  Google Scholar 

  27. Ragab EM, et al. Therapeutic potential of chrysin nanoparticle-mediation inhibition of succinate dehydrogenase and ubiquinone oxidoreductase in pancreatic and lung adenocarcinoma. Eur J Med Res. 2022;27(1):172.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Deng X, et al. Anticancer activity of natural flavonoids: inhibition of HIF-1α signaling pathway. Curr Org Chem. 2019;23(26):2945–59.

    Article  CAS  Google Scholar 

  29. Wu Y, et al. Inhibition of the oxygen-sensing asparaginyl hydroxylase factor inhibiting hypoxia-inducible factor: a potential hypoxia response modulating strategy. J Med Chem. 2021;64(11):7189–209.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors of this study thank all members of the Faculty of Science and Faculty of Medicine, at Tanta University, Egypt.

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

RE: methodology, writing, and interpreting the data of the original draft. ED: conceptualization and formal analysis. MT: supervision investigation and review. KA: analyzed and interpreted the data and edited it. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Eman M. Ragab.

Ethics declarations

Ethics approval and consent to participate

The Guidelines for the Care and Use of Laboratory Animals published by the National Institutes of Health were adhered to in the care of the experimental animals. The experiments will be monitored by the Egyptian Ethical Committee of Tanta University’s Faculty of Science (#IACUC-SCI-TU-0287).

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.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ragab, E.M., El Gamal, D.M., Mohamed, T.M. et al. Naringenin-loaded nanoparticles modulate HIF-driven oxygen-sensing pathways in lung adenocarcinoma cells. BMC Res Notes 18, 64 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07133-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07133-2

Keywords