We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Application of a fluorescent H2S probe based on excited-state intramolecular proton transfer for detecting latent mechanism of H2S-induced MCF-7 apoptosis

    Jiao Liu‡

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Xiaoxuan Wang‡

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yuan Cheng‡

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yinghua Yu‡

    Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Maofang Zhao

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ,
    Jie Huang

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ,
    Peiquan Yu

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ,
    Qian Fu

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    ,
    Yilin Song

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    &
    Yi Liu

    *Author for correspondence: Tel.: +86 137 7589 5636;

    E-mail Address: cbpeliuyinew@163.com

    Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

    Published Online:https://doi.org/10.4155/fmc-2021-0309

    Background: H2S is the third gas transmitter affecting the growth, reproduction and survival of cancer cells. However, the H2S anticancer and antitumor mechanism still needs to be further studied. Methods: Here, FHS-1 was synthesized utilizing excited-state intramolecular proton transfer to detect H2S in MCF-7 cells, and investigated the effects of varying concentrations NaHS on apoptosis. Results: The study found that FHS-1 detects H2S levels with high selectivity and pH stability and that H2S may regulate apoptosis in MCF-7 cells through the p53/mTOR/STAT3 pathway. Conclusion: Researching the influence of H2S on apoptosis can serve as a theoretical foundation for future research into H2S-related anticancer medicines, and the H2S probe can be used as an effective cancer screening tool.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Sestak I. Preventative therapies for healthy women at high risk of breast cancer. Cancer Manag. Res. 6, 423–430 (2014).
    • 2. Lehrer S, Rosenzweig KE. Wilms tumor and breast cancer. Cancer 121(12), 2099 (2015).
    • 3. Ma JH, Qin L, Li X. Role of STAT3 signaling pathway in breast cancer. Cell Commun. Signal. 18, 33 (2020).
    • 4. Yang L, Li DX, Cao BQ et al. Exercise training ameliorates early diabetic kidney injury by regulating the H2S/SIRT1/p53 pathway. FASEB J. 35(9), e21823 (2021).
    • 5. Ngowi EE, Afzal A, Sarfraz M et al. Role of hydrogen sulfide donors in cancer development and progression. Int. J. Biol. Sci. 17(1), 73–88 (2021).
    • 6. Szabo C. Gasotransmitters in cancer: from pathophysiology to experimental therapy. Nat. Rev. Drug Discov. 15(3), 185–203 (2016). •• Discusses the role of each gasotransmitter in cancer and the effects of pharmacological agents (some of which are in early-stage clinical studies) that modulate levels of each gasotransmitter.
    • 7. Lee ZW, Teo XY, Tay EY et al. Utilizing hydrogen sulfide as a novel anti-cancer agent by targeting cancer glycolysis and pH imbalance. Br. J. Pharmacol. 171(18), 4322–4336 (2014).
    • 8. Chattopadhyay M, Kodela R, Santiago G et al. NOSH-aspirin (NBS-1120) inhibits pancreatic cancer cell growth in a xenograft mouse model: modulation of FoxM1, p53, NF-κB, iNOS, caspase-3 and ROS. Biochem. Pharmacol. 176, 113857 (2020).
    • 9. Liang Y, Zhang H, Song X et al. Metastatic heterogeneity of breast cancer: molecular mechanism and potential therapeutic targets. Semin. Cancer Biol. 60, 14–27 (2020).
    • 10. Cai F, Xu H, Cao N et al. ADT-OH, a hydrogen sulfide-releasing donor, induces apoptosis and inhibits the development of melanoma in vivo by upregulating FADD. Cell Death Dis. 11(1), 33 (2020).
    • 11. Zhao L, Wang Y, Yan Q et al. Exogenous hydrogen sulfide exhibits anti-cancer effects though p38/MAPK signaling pathway in C6 glioma cells. Biol. Chem. 396(11), 1247–1253 (2015).
    • 12. Wang RH, Chu YH, Lin KT. The hidden role of hydrogen sulfide metabolism in cancer. Int. J. Mol. Sci. 22(12), 6562 (2021).
    • 13. Wu D, Li J, Zhang Q et al. Exogenous hydrogen sulfide regulates the growth of human thyroid carcinoma cells. Oxid. Med. Cell. Longev. 2019, 6927298 (2019).
    • 14. Yang C, He L, He P et al. Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/Bcl-2 signaling pathway. Med. Oncol. 32(2), 352 (2015).
    • 15. Wang X, Qiu W, Zhang G et al. MicroRNA-204 targets JAK2 in breast cancer and induces cell apoptosis through the STAT3/Bcl-2/survivin pathway. Int. J. Clin. Exp. Pathol. 8(5), 5017–5025 (2015).
    • 16. Zhang L, Meng WQ, Lu L et al. Selective detection of endogenous H2S in living cells and the mouse hippocampus using a ratiometric fluorescent probe. Sci. Rep. 4, 5870 (2014).
    • 17. Zhang X, Jin X, Zhang C et al. A fluorescence turn-on probe for hydrogen sulfide and biothiols based on PET & TICT and its imaging in HeLa cells. Spectrochim. Acta A Mol. Biomol. Spectrosc. 244, 118839 (2021).
    • 18. Wang KP, Zhang QL, Wang X et al. A fluorescent probe based on tetrahydro[5]helicene derivative with large Stokes shift for rapid and highly selective recognition of hydrogen sulfide. Spectrochim. Acta A Mol. Biomol. Spectrosc. 214, 487–495 (2019).
    • 19. Shaydyuk YO, Bashmakova NV, Dmytruk AM et al. Nature of fast relaxation processes and spectroscopy of a membrane-active peptide modified with fluorescent amino acid exhibiting excited state intramolecular proton transfer and efficient stimulated emission. ACS Omega 6(15), 10119–10128 (2021).
    • 20. Lin CC, Chen CL, Chung MW et al. Effects of multibranching on 3-hydroxyflavone-based chromophores and the excited-state intramolecular proton transfer dynamics. J. Phys. Chem. A 114(38), 10412–10420 (2010).
    • 21. Hou P, Li HM, Chen S. A highly selective and sensitive 3-hydroxyflavone-based colorimetric and fluorescent probe for hydrogen sulfide with a large Stokes shift. Tetrahedron 72(24), 3531–3534 (2016).
    • 22. Chen Y, Piao Y, Feng X et al. Excited state intramolecular proton transfer (ESIPT) luminescence mechanism for 4-N,N-diethylamino-3-hydroxyflavone in propylene carbonate, acetonitrile and the mixed solvents. Spectrochim. Acta A Mol. Biomol. Spectrosc. 224, 117416 (2020).
    • 23. Dereka B, Svechkarev D, Rosspeintner A et al. Solvent tuning of photochemistry upon excited-state symmetry breaking. Nat. Commun. 11(1), 1925 (2020).
    • 24. Sedgwick AC, Wu L, Han HH et al. Excited-state intramolecular proton-transfer (ESIPT) based fluorescence sensors and imaging agents. Chem. Soc. Rev. 47(23), 8842–8880 (2018).
    • 25. Annenkov VV, Verkhozina ON, Shishlyannikova TA et al. Application of 4-chloro-7-nitrobenzo-2-oxa-1,3-diazole in analysis: fluorescent dyes and unexpected reaction with tertiary amines. Anal. Biochem. 486, 5–13 (2015).
    • 26. Ismail I, Chen Z, Ji X et al. A fast-response red shifted fluorescent probe for detection of H2S in living cells. Molecules 25(3), 437 (2020).
    • 27. Ibrahim H, Serag A, Farag MA. Emerging analytical tools for the detection of the third gasotransmitter H2S, a comprehensive review. J. Adv. Res. 27, 137–153 (2020).
    • 28. Gunduz S, Goren AC, Ozturk T. Facile syntheses of 3-hydroxyflavones. Org. Lett. 14(6), 1576–1579 (2012).
    • 29. Zhang L, Zheng XE, Zou F et al. A highly selective and sensitive near-infrared fluorescent probe for imaging of hydrogen sulphide in living cells and mice. Sci. Rep. 6, 18868 (2016).
    • 30. Long Y, Liu J, Tian D et al. Cooperation of ESIPT and ICT processes in the designed 2-(2′-hydroxyphenyl) benzothiazole derivative: a near-infrared two-photon fluorescent probe with a large Stokes shift for the detection of cysteine and its application in biological environments. Anal. Chem. 92(20), 14236–14243 (2020).
    • 31. Li X, Yu P, Yu Y et al. Hydrogen sulfide ameliorates high glucose-induced pro-inflammation factors in HT-22 cells: involvement of SIRT1-mTOR/NF-κB signaling pathway. Int. Immunopharmacol. 95, 107545 (2021). • Previous studies have shown that exogenous H2S treatment or enhancement of endogenous H2S synthesis can prevent neuronal inflammation under high glucose exposure.
    • 32. Fitzmaurice C, Dicker D, Pain A et al. The global burden of cancer 2013. JAMA Oncol. 1(4), 505–527 (2015).
    • 33. Filipovic MR, Zivanovic J, Alvarez B, Banerjee R. Chemical biology of H2S signaling through persulfidation. Chem. Rev. 118(3), 1253–1337 (2018).
    • 34. Click RE. Potential alteration of tumor microenvironments by β-mercaptoethanol. Future Oncol. 17(3), 315–331 (2021).
    • 35. Li Z, Geng ZR, Zhang C, Wang XB, Wang ZL. BODIPY-based azamacrocyclic ensemble for selective fluorescence detection and quantification of homocysteine in biological applications. Biosens. Bioelectron. 72, 1–9 (2015).
    • 36. Wang Z, Han DM, Jia WP, Zhou QZ, Deng WP. Reaction-based fluorescent probe for selective discrimination of thiophenols over aliphaticthiols and its application in water samples. Anal. Chem. 84(11), 4915–4920 (2012).
    • 37. Lv M, Li Y, Ji MH, Zhuang M, Tang JH. Inhibition of invasion and epithelial-mesenchymal transition of human breast cancer cells by hydrogen sulfide through decreased phospho-p38 expression. Mol. Med. Rep. 10(1), 341–346 (2014).
    • 38. Hellmich MR, Szabo C. Hydrogen sulfide and cancer. Handb. Exp. Pharmacol. 230, 233–241 (2015).
    • 39. Ye M, Yu M, Yang D et al. Exogenous hydrogen sulfide donor NaHS alleviates nickel-induced epithelial-mesenchymal transition and the migration of A549 cells by regulating TGF-β1/Smad2/Smad3 signaling. Ecotoxicol. Environ. Saf. 195, 110464 (2020).
    • 40. Li H, Xu F, Gao G et al. Hydrogen sulfide and its donors: novel antitumor and antimetastatic therapies for triple-negative breast cancer. Redox Biol. 34, 101564 (2020).
    • 41. Hellmich MR, Coletta C, Chao C, Szabo C. The therapeutic potential of cystathionine β-synthetase/hydrogen sulfide inhibition in cancer. Antioxid. Redox Signal. 22(5), 424–448 (2015).
    • 42. Chakraborty PK, Xiong X, Mustafi SB et al. Role of cystathionine beta synthase in lipid metabolism in ovarian cancer. Oncotarget 6(35), 37367–37384 (2015).
    • 43. Cai FF, Xu HR, Yu SH et al. ADT-OH inhibits malignant melanoma metastasis in mice via suppressing CSE/CBS and FAK/paxillin signaling pathway. Acta Pharmacol. Sin. doi:10.1038/s41401-021-00799-x (2021) (Epub ahead of print).
    • 44. Hu X, Xiao Y, Sun J et al. New possible silver lining for pancreatic cancer therapy: hydrogen sulfide and its donors. Acta Pharm. Sin. B 11(5), 1148–1157 (2021).
    • 45. Szabo C, Papapetropoulos A. International Union of Basic and Clinical Pharmacology. CII: pharmacological modulation of H2S levels: H2S donors and H2S biosynthesis inhibitors. Pharmacol. Rev. 69(4), 497–564 (2017).
    • 46. Youness RA, Gad AZ, Sanber K et al. Targeting hydrogen sulphide signaling in breast cancer. J. Adv. Res. 27, 177–190 (2020).
    • 47. Bakshi HA, Hakkim FL, Sam S. Molecular mechanism of crocin induced caspase mediated MCF-7 cell death: in vivo toxicity profiling and ex vivo macrophage activation. Asian Pac. J. Cancer Prev. 17(3), 1499–1506 (2016).
    • 48. Kanamori Y, Via LD, Macone A et al. Aged garlic extract and its constituent, S-allyl-L-cysteine, induce the apoptosis of neuroblastoma cancer cells due to mitochondrial membrane depolarization. Exp. Ther. Med. 19(2), 1511–1521 (2020).
    • 49. Zhang J, Li C, Zhang L et al. Andrographolide induces noxa-dependent apoptosis by transactivating ATF4 in human lung adenocarcinoma cells. Front. Pharmacol. 12, 680589 (2021).
    • 50. Zilfou JT, Spector MS, Lowe SW. Slugging it out: fine tuning the p53-PUMA death connection. Cell 123(4), 545–548 (2005).
    • 51. Wu WS, Heinrichs S, Xu D et al. Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 123(4), 641–653 (2005).
    • 52. Xie ZZ, Li MM, Deng PF et al. Paris saponin-induced autophagy promotes breast cancer cell apoptosis via the Akt/mTOR signaling pathway. Chem. Biol. Interact. 264, 1–9 (2017).
    • 53. Chang CT, Korivi M, Huang HC et al. Inhibition of ROS production, autophagy or apoptosis signaling reversed the anticancer properties of Antrodia salmonea in triple-negative breast cancer (MDA-MB-231) cells. Food Chem. Toxicol. 103, 1–17 (2017). • Evaluated the combined effect of ADT-OH treatment and FADD overexpression on melanoma cell death in vivo using a mouse xenograft model and found that tumor-specific delivery of FADD through a recombinant Salmonella strain (VNP-FADD) combined with low-dose ADT-OH treatment significantly inhibited tumor growth and induced cancer cell apoptosis.
    • 54. Miricescu D, Totan A, Stanescu-Spinu II et al. PI3K/AKT/mTOR signaling pathway in breast cancer: from molecular landscape to clinical aspects. Int. J. Mol. Sci. 22(1), 173 (2020).
    • 55. Lee C, Dhillon J, Wang MY et al. Targeting YB-1 in HER-2 overexpressing breast cancer cells induces apoptosis via the mTOR/STAT3 pathway and suppresses tumor growth in mice. Cancer Res. 68, 8661–8666 (2008).