We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Selective estrogen receptor modulators against Gram-positive and Gram-negative bacteria: an experimental study

    Aakriti Garg

    Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India

    ,
    Arti Singh

    Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India

    &
    Anoop Kumar

    *Author for correspondence: Tel.: +91 011 2955 2039;

    E-mail Address: abitmesra@gmail.com

    Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India

    Department of Pharmacology & Clinical Research, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi, 110017, India

    Published Online:https://doi.org/10.2217/fmb-2020-0310

    Aim: This study was conducted to explore the antibacterial potential of selective estrogen receptor modulators (SERMs). Materials & methods: The percentage growth retardation, bacterial growth kinetics, biofilm, checkerboard and bacterial burden assays were conducted to check antibacterial potential of SERMs. Finally, docking study was also conducted to predict possible antibacterial mechanism of SERMs. Results:In vitro and in vivo studies have shown the antibacterial activity of SERMs against different tested strains of bacteria. The synergistic activity of SERMs in combination with standard antibacterial agents was also observed and tested further under in vivo conditions. In vivo results have shown decreased bacterial bioburden. Docking studies have predicted the multimodal antibacterial mechanism of SERMs. Conclusion: SERMs can be considered as promising broad-spectrum antibacterial agents.

    References

    • 1. Page last reviewed: april 2, 2013 Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Division of Healthcare Quality Promotion (DHQP) (April 2, 2013). https://www.cdc.gov/hai/organisms/pseudomonas.html
    • 2. Gupta M, Kumar A. Comparison of minimum inhibitory concentration (MIC) value of statin drugs: a systematic review. Anti-Infect. Agents 17(1), 4–19 (2019).
    • 3. Rana R, Sharma R, Kumar A. Repurposing of existing statin drugs for treatment of microbial infections: how much promising? Infect. Disord. Drug Targets 19(3), 224–237 (2018).
    • 4. Sethi K, Singh A, Kumar A. Exploring the therapeutic potential of atorvastatin against Gram-positive and Gram-negative bacteria: in silico, in vitro and in vivo evidence. Infect. Disord. Drug Targets 20(6), 798–815 (2020).
    • 5. Kapoor Y, Sharma R, Kumar A. Repurposing of existing drugs for the bacterial infections: an in silico and in vitro study. Infect. Disord. Drug Targets 20(2), 182–197 (2020).
    • 6. Gupta M, Sharma R, Kumar A. Comparative potential of simvastatin, rosuvastatin and fluvastatin against bacterial infection: an in silico and in vitro study. Orient. Pharm. Exp. Med. 19(3), 259–275 (2019).
    • 7. Rana R, Sharma R, Kumar A. Repurposing of Fluvastatin against Candida albicans CYP450 lanosterol 14 α-demethylase, a target enzyme for antifungal therapy: an in silico and in vitro study. Curr. Mol. Med. 19(7), 506–524 (2019).
    • 8. Peng J, Sengupta S, Jordan VC. Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer. Anticancer Agents Med. Chem. 9(5), 481–499 (2009).
    • 9. Haskell SG. Selective estrogen receptor modulators (featured CME topic). South Med. J. 96(5), 469–477 (2003).
    • 10. Singh MM. Centchroman, a selective estrogen receptor modulator, as a contraceptive and for the management of hormone-related clinical disorders. Med. Res. Rev. 21(4), 302–347 (2001).
    • 11. Eder SE. Ospemifene: a novel selective estrogen receptor modulator for treatment of dyspareunia. Women Health 10(5), 499–503 (2014).
    • 12. Gennari L, Merlotti D, Nuti R. Selective estrogen receptor modulator (SERM) for the treatment of osteoporosis in postmenopausal women: focus on lasofoxifene. Clin. Inter. Aging. 5, 19–29 (2010).
    • 13. Deroo BJ, Korach KS. Estrogen receptors and human disease. J. Clin. Invest. 116(3), 561–570 (2006).
    • 14. Xu B, Lovre D, Mauvais-Jarvis F. The effect of selective estrogen receptor modulators on type 2 diabetes onset in women: basic and clinical insights. J. Diabetes Complications 31(4), 773–779 (2017).
    • 15. Barreto GE, Santos-Galindo M, Garcia-Segura LM. Selective estrogen receptor modulators regulate reactive microglia after penetrating brain injury. Front. Aging Neurosci. 6, 132 (2014).
    • 16. Christodoulakos GE, Lambrinoudaki IV, Botsis DC. The cardiovascular effects of selective estrogen receptor modulators. Ann. NY Acad. Sci. 1092(1), 374–384 (2006).
    • 17. Hussein MH, Schneider EK, Elliott AG et al. From breast cancer to antimicrobial: combating extremely resistant Gram-negative “superbugs” using novel combinations of polymyxin B with selective estrogen receptor modulators. Microb. Drug Resist. 23(5), 640–650 (2017).
    • 18. Sui SJ, Lo R, Fernandes AR et al. Raloxifene attenuates Pseudomonas aeruginosa pyocyanin production and virulence. Int. J. Antimicrob. Agents 40(3), 246–251 (2012).
    • 19. Wiseman H, Cannon M, Arnstein HR. Observation and significance of growth inhibition of Saccharomyces cerevisiae (A224A) by the antioestrogen drug tamoxifen. Biochem. Soc. Trans. 17, 1038–1039 (1989).
    • 20. Doroodgar M, Delavari M, Doroodgar M, Abbasi A, Taherian AA, Doroodgar A. Tamoxifen induces apoptosis of Leishmania major promastigotes in vitro. Korean J. Parasitol. 54(1), 9–14 (2016).
    • 21. Reimão JQ, Miguel DC, Taniwaki NN, Trinconi CT, Yokoyama-Yasunaka JK, Uliana SR. Antileishmanial activity of the estrogen receptor modulator raloxifene. PLoS Negl. Trop. Dis. 8(5), e2842 (2014).
    • 22. Miguel DC, Yokoyama-Yasunaka JK, Andreoli WK, Mortara RA, Uliana SR. Tamoxifen is effective against Leishmania and induces a rapid alkalinization of parasitophorous vacuoles harbouring Leishmania (Leishmania) amazonensis amastigotes. J. Antimicrob. Chemother. 60(3), 526–534 (2007).
    • 23. Trinconi CT, Reimão JQ, Coelho AC, Uliana SR. Efficacy of tamoxifen and miltefosine combined therapy for cutaneous leishmaniasis in the murine model of infection with Leishmania amazonensis. J. Antimicrob. Chemother. 71(5), 1314–1322 (2016).
    • 24. Dittmar AJ, Drozda AA, Blader IJ. Drug repurposing screening identifies novel compounds that effectively inhibit Toxoplasma gondii growth. mSphere 1(2), e00042-15 (2016).
    • 25. Escobedo G, Palacios-Arreola MI, Olivos A, López-Griego L, Morales-Montor J. Tamoxifen treatment in hamsters induces protection during taeniosis by Taenia solium. Bio. Med. Res. Int. 2013, 280496 (2013).
    • 26. Cong Y, Hart BJ, Gross R et al. MERS-CoV pathogenesis and antiviral efficacy of licensed drugs in human monocyte-derived antigen-presenting cells. PLoS ONE 13(3), e0194868 (2018).
    • 27. Sun W, He S, Martínez-Romero C et al. Synergistic drug combination effectively blocks Ebola virus infection. Antiviral Res. 137, 165–172 (2017).
    • 28. Zhao Y, Ren J, Harlos K et al. Toremifene interacts with and destabilizes the Ebola virus glycoprotein. Nature 535(7610), 169–172 (2016).
    • 29. Butts A, Koselny K, Chabrier-Roselló Y et al. Estrogen receptor antagonists are anti-cryptococcal agents that directly bind EF hand proteins and synergize with fluconazole in vivo. MBio 5(1), e00765-13 (2014).
    • 30. Jang WS, Kim S, Podder B et al. Anti-mycobacterial activity of tamoxifen against drug-resistant and intra-macrophage Mycobacterium tuberculosis. J. Microbiol. Biotechnol. 25(6), 946–950 (2015).
    • 31. Theophel K, Schacht VJ, Schlüter M et al. The importance of growth kinetic analysis in determining bacterial susceptibility against antibiotics and silver nanoparticles. Front. Microbiol. 5, 544 (2014).
    • 32. Graziano TS, Cuzzullin MC, Franco GC et al. Statins and antimicrobial effects: simvastatin as a potential drug against Staphylococcus aureus biofilm. PLoS ONE 10(5), e0128098 (2015).
    • 33. Yang B, Lei Z, Zhao Y et al. Combination susceptibility testing of common antimicrobials in vitro and the effects of sub-MIC of antimicrobials on Staphylococcus aureus biofilm formation. Front. Microb. 8, 2125 (2017).
    • 34. Neely MN, Pfeifer JD, Caparon M. Streptococcus-zebrafish model of bacterial pathogenesis. Infect. Immun. 70(7), 3904–3914 (2002).
    • 35. Sundaramoorthy NS, Mohan HM, Subramaniam S et al. Ursolic acid inhibits colistin efflux and curtails colistin resistant Enterobacteriaceae. AMB Express 9(1), 27 (2019).
    • 36. Farha MA, Czarny TL, Myers CL et al. Antagonism screen for inhibitors of bacterial cell wall biogenesis uncovers an inhibitor of undecaprenyl diphosphate synthase. Proc. Natl Acad Sci. USA 112(35), 11048–11053 (2015).
    • 37. Jacobs AC, DiDone L, Jobson J, Sofia MK, Krysan D, Dunman PM. Adenylate kinase release as a high-throughput-screening-compatible reporter of bacterial lysis for identification of antibacterial agents. Antimicrob. Agents Chemother. 57(1), 26–36 (2013).
    • 38. Dufour D, Leung V, Lévesque CM. Bacterial biofilm: structure, function, and antimicrobial resistance. Endod. Topics 22(1), 2–16 (2010).
    • 39. Algburi A, Comito N, Kashtanov D, Dicks LM, Chikindas ML. Control of biofilm formation: antibiotics and beyond. Appl. Environ. Microbiol. 83(3), e02508-16 (2017).
    • 40. Yasir M, Willcox M, Dutta D. Action of antimicrobial peptides against bacterial biofilms. Materials 11(12), e2468 (2018).
    • 41. Torres NS, Montelongo-Jauregui D, Abercrombie JJ et al. Antimicrobial and antibiofilm activity of synergistic combinations of a commercially available small compound library with colistin against Pseudomonas aeruginosa. Front. Microbiol. 9, 2541 (2018).
    • 42. Cheesman MJ, Ilanko A, Blonk B, Cock IE. Developing new antimicrobial therapies: are synergistic combinations of plant extracts/compounds with conventional antibiotics the solution? Pharmacogn. Rev. 11(22), 57–72 (2017).
    • 43. Delattin N, De Brucker K, Vandamme K et al. Repurposing as a means to increase the activity of amphotericin B and caspofungin against Candida albicans biofilms. J. Antimicrob. Chemother. 69(4), 1035–1044 (2013).
    • 44. Trinconi CT, Reimão JQ, Yokoyama-Yasunaka JK, Miguel DC, Uliana SR. Combination therapy with tamoxifen and amphotericin B in experimental cutaneous leishmaniasis. Antimicrob. Agents Chemother. 58(5), 2608–2613 (2014).
    • 45. Sullivan C, Matty MA, Jurczyszak D et al. Infectious disease models in zebrafish. Methods Cell Biol. 138, 101–136 (2017).
    • 46. Gupta M, Sharma R, Singh M, Kumar A. Docking techniques in pharmacology: how much promising? Comput. Biol. Chem. 76, 210–217 (2018).
    • 47. Garg A, Singh B, Sharma R, Singh A, Kumar A. Selective estrogen receptor modulators (SERMs): mechanistic insights against microbial infections. Curr. Mol. Med. 20(2), 102–115 (2019).
    • 48. Oefner C, Bandera M, Haldimann A et al. Increased hydrophobic interactions of iclaprim with Staphylococcus aureus dihydrofolate reductase are responsible for the increase in affinity and antibacterial activity. J. Antimicrob. Chemother. 63(4), 687–698 (2009).
    • 49. Bush K, Bradford PA. β-Lactams and β-lactamase inhibitors: an overview. Cold Spring Harb. Perspect. Med. 6(8), a025247 (2016).