EC Pharmacology and Toxicology

Research Article Volume 11 Issue 3 - 2023

Insilico and Invitro Optimization of Naringin and Rutin Molecules Targeting DNA Damage in Breast Cancer Cells

Badhe Pravin1,2,3* and Badhe Ashwini1,2

1Swalife Biotech Ltd Unit 3D North Point House, North Point Business Park, Ireland
2Swalife Foundation, India
3Department of Pharmacology, Sinhgad College of Pharmacy, Pune, India

*Corresponding Author: Badhe Pravin, Swalife Biotech Ltd Unit 3D North Point House, North Point Business Park, Ireland and Swalife Foundation and Department of Pharmacology, Sinhgad College of Pharmacy, Pune, India.
Received: December 29, 2022; Published: February 24, 2023



Discovering the molecular mechanisms of DNA damage response pathways has led to new therapeutic approaches in oncology. Our study optimized DNA damage- targeting molecules naringin and rutin in breast cancer cells.

Our study involved MTT assays for detection of its toxicity and proliferative activity in breast cancer cells and normal cancer cells. Our studies determined the molecules' antioxidant properties using the DPPH assay. The role in reducing free radicals has been evaluated using a variety of free radical scavenging activity assays.

Further evaluation of the molecules was carried out by high alkaline comet assay (pH > 13) to test for genotoxicity. Human Dermal Fibroblast cells (2DD) (1 x 105 cells/ml) and breast cancer cells (MDA-MB-231) were pre-incubated with Naringin and Rutin (10 µM) for one hour.

In normal cells, rutin and naringin molecules do not cause genotoxicity, but they cause DNA damage in breast cancer cells when they are diluted to 10 µM. The results from our study indicate that both molecules cause 60 - 70% DNA damage in breast cancer cells.

Keyword: DNA Damage; Anti-Oxidant; Naringin; Rutin; Free Radicals; Insilico; Invitro

  1. Srinivas US., et al. “ROS and the DNA damage response in cancer”. Redox Biology, Redox Regulation of Cell State and Fate 25 (2019): 101084.
  2. Parplys AC., et al. “DNA damage by X-rays and their impact on replication processes”. Radiotherapy and Oncology 102 (2012): 466-471.
  3. Salar RK and Purewal SS. “Improvement of DNA damage protection and antioxidant activity of biotransformed pearl millet (Pennisetum glaucum) cultivar PUSA-415 using Aspergillus oryzae MTCC 3107”. Biocatalysis and Agricultural Biotechnology 8 (2016): 221-227.
  4. Cadet J., et al. “Oxidatively generated base damage to cellular DNA”. Free Radical Biology and Medicine 49 (2010): 9-21.
  5. Sedelnikova OA., et al. “Role of oxidatively induced DNA lesions in human pathogenesis”. MUT External Residencies 704 (2010): 152-159.
  6. Hwang ES and Bowen PE. “DNA damage, a biomarker of carcinogenesis: its measurement and modulation by diet and environment”. Critical Reviews in Food Science and Nutrition 47 (2007): 27-50.
  7. Jackson SP and Bartek J. “The DNA-damage response in human biology and disease”. Nature 461 (2009): 1071-1078.
  8. Pérez-Coyotl, I., et al. “DNA damage and cytotoxicity induced on common carp by pollutants in water from an urban reservoir. Madín reservoir, a case study”. Chemosphere 185 (2017): 789-797.
  9. Kaur R., et al. “Effect of incorporation of flaxseed to wheat rusks: Antioxidant, nutritional, sensory characteristics, and in vitro DNA damage protection activity”. Journal of Food Processing and Preservation (2018).
  10. Salar RK and Purewal SS. “Phenolic content, antioxidant potential and DNA damage protection of pearl millet (Pennisetum glaucum) cultivars of North Indian region”. Journal of Food Measurement and Characterization 11 (2017): 126-133.
  11. Dexheimer TS. “DNA repair pathways and mechanisms', in DNA repair of cancer stem cells”. Springer (2013): 19-32.
  12. Lindahl T and Barnes DE. “Repair of endogenous DNA damage”. Cold Spring Harbor Symposia on Quantitative Biology 65 (2000): 127-133.
  13. Hoeijmakers JH. “DNA damage, aging, and cancer”. New England Journal of Medicine 15 (2009): 1475-1485.
  14. O’Driscoll M., et al. “An overview of three new disorders associated with genetic instability: LIG4 syndrome, RS-SCID and ATR-Seckel syndrome”. DNA Repair 8 (2004): 1227-1235.
  15. Altieri F., et al. “DNA damage and repair: from molecular mechanisms to health implications”. Antioxidants and Redox Signaling 5 (2008): 891-938.
  16. Molinaro C., et al. “Proteins from the DNA Damage Response: Regulation, Dysfunction, and Anticancer Strategies”. Cancers 13 (2021): 3819.
  17. Huang RX and Zhou PK. “DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer”. Signal Transduction and Targeted Therapy 5 (2020): 60.
  18. Zou L and Elledge SJ. “Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes”. Science 300 (2003): 1542-1548.
  19. Weber AM and Ryan AJ. “ATM and ATR as therapeutic targets in cancer”. Pharmacology and Therapeutics 149 (2015): 124-138.
  20. Kai M. “Roles of RNA-binding proteins in DNA damage response”. International Journal of Molecular Sciences 17 (2016): 310.
  21. Wang Y., et al. “Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells”. International Journal of Oncology 53 (2018): 1667-1680.
  22. Qiu L., et al. “Wee1 and Cdc25 control morphogenesis, virulence and multistress tolerance of Beauveria bassiana by balancing cell cycle-required cyclin-dependent kinase 1 activity”. Environmental Microbiology 17 (2015): 1119-1133.
  23. Lee YY., et al. “Anti-tumor effects of Wee1 kinase inhibitor with radiotherapy in human cervical cancer”. Scientific Reports 9 (2019): 15394.
  24. Sancar A., et al. “Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints”. Annual Review of Biochemistry 73 (2004): 39-85.
  25. Gonzalez Besteiro MA and Gottifredi V. “The fork and the kinase: a DNA replication tale from a CHK1 perspective”. Mutation Research - Reviews in Mutation Research 763 (2015): 168-180.
  26. Fakhri S., et al. “The effects of anticancer medicinal herbs on vascular endothelial growth factor based on pharmacological aspects: A review study”. Nutrition and Cancer 73 (2019): 1-15.
  27. Ochwang’I DO., et al. “Medicinal plants used in treatment and management of cancer in Kakamega County, Kenya”. Journal of Ethnopharmacology 151 (2014): 1040-1055.
  28. Pandey P., et al. “Rutin (Bioflavonoid) as Cell Signaling Pathway Modulator: Prospects in Treatment and Chemoprevention”. Pharmaceuticals 14 (2021): 1069.
  29. Nadaf K and Badhe P. In-silico Study of Oyster Mushroom (Pleurotus ostreatus) Targeting PARP Protein (4UND) (2021).
  30. Gosavi HD and Badhe P. In Silico Study of Silybum Marianum Targeting Parp Protein (4UND Protein) (2021).
  31. Shelke S and Badhe P. In-silico study of Agaricus bisporus on DNA damaging protein (2021): 3.
  32. Caparica R., et al. “Anticancer Activity of Rutin and Its Combination with Ionic Liquids on Renal Cells”. Biomolecules 10 (2020): 233.
  33. Rui Chen., et al. “Therapeutic potential of naringin: an overview”. Pharmaceutical Biology 12 (2016): 3203-3210.
  34. Salehi B., et al. “The Therapeutic Potential of Naringenin: A Review of Clinical Trials”. Pharmaceuticals (2019).
  35. Dallakyan S and Olson AJ. “Small-molecule library screening by docking with PyRx”. In Chemical biology Humana Press, New York, NY (2015): 243-250.
  36. Jacob Reed B. "Dockomatic: An Emerging Resource to Manage Molecular Docking". Boise State University Theses and Dissertations (2012): 297.
  37. Daina A., et al. “SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules”. Scientific Reports 7 (2017): 42717.
  38. Pires DE., et al. “pkCSM: predicting small- molecule pharmacokinetic and toxicity properties using graph-based signatures”. Journal of Medicinal Chemistry 9 (2015): 4066-4072.
  39. Saad B., et al. “Evaluation of Medicinal Plant Hepatotoxicity in Co-cultures of Hepatocytes and Monocytes”. Evidence-Based Complementary and Alternative Medicine 3 (2006): 93-98.
  40. Badhe P. “Characterisation of fractions from Andrographis paniculata and Silybum marianum plant extracts that protect human cells against DNA damage (Doctoral dissertation, Brunel University London) (2016).
  41. Patlolla AK., et al. “Potassium dichromate induced cytotoxicity, genotoxicity and oxidative stress in human liver carcinoma (HepG2) cells”. International Journal of Environmental Research and Public Health 2 (2009): 643-653.
  42. Prakash A., et al. “Antioxidant activity”. Medallion Laboratories Analytical Progress 2 (2001): 1-4.
  43. Kalim MD., et al. “Oxidative DNA damage preventive activity and antioxidant potential of plants used in Unani system of medicine”. BMC Complementary and Alternative Medicine 1 (2010): 1-11.
  44. Fontana M., et al. “Interaction of enkephalins with oxyradicals”. Biochemical Pharmacology 10 (2001): 1253-1257.
  45. Hazra B., et al. “Antioxidant and free radical scavenging activity of Spondias pinnata”. BMC complementary and Alternative Medicine 1 (2008): 1.
  46. O’Connor MJ. “Targeting the DNA damage response in cancer”. Molecular Cell 4 (2015): 547-560.
  47. Bryant HE., et al. “Specific killing of BRCA2-deficient tumours with inhibitors of poly (ADP-ribose) polymerase”. Nature 7035 (2005): 913-917.
  48. Farmer H., et al. “Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy”. Nature 7035 (2005): 917-921.

Badhe Pravin and Badhe Ashwini. Insilico and Invitro Optimization of Naringin and Rutin Molecules Targeting DNA Damage in Breast Cancer Cells. EC Pharmacology and Toxicology 11.3 (2023): 59-79.