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
Preliminary Communication

Effect of fluoxetine on planktonic and biofilm growth and the antimicrobial susceptibility of Burkholderia pseudomallei

    Gláucia MM Guedes‡

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Laboratory of Emerging & Reemerging Pathogens, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ‡Both Authors contributed equally

    Search for more papers by this author

    ,
    Emanuela S Araújo‡

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ‡Both Authors contributed equally

    Search for more papers by this author

    ,
    Késia VC Ribeiro

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Vinícius C Pereira

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Ana CCF Soares

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Alyne S Freitas

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Bruno R Amando

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Rossana A Cordeiro

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Laboratory of Emerging & Reemerging Pathogens, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    ,
    Marcos FG Rocha

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Laboratory of Emerging & Reemerging Pathogens, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    College of Veterinary, State University of Ceara. Av. Dr Silas Munguba, 1700, Campus do Itaperi – CEP 60714–903, Fortaleza, Ceará, Brazil

    ,
    José JC Sidrim

    *Author for correspondence:

    E-mail Address: sidrim@ufc.br

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Laboratory of Emerging & Reemerging Pathogens, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    &
    Débora SCM Castelo-Branco

    **Author for correspondence:

    E-mail Address: deb_castelobranco@yahoo.com

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Laboratory of Emerging & Reemerging Pathogens, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    Department of Pathology & Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceara, Rua Cel, Nunes de Melo, 1315 – Rodolfo Teófilo – CEP 60430–275, Fortaleza, Ceará, Brazil

    Published Online:https://doi.org/10.2217/fmb-2022-0272

    Aim: This study evaluated the effect of fluoxetine (FLU) on planktonic and biofilm growth and the antimicrobial susceptibility of Burkholderia pseudomallei. Materials & methods: The minimum inhibitory concentrations (MICs) for FLU were determined by broth microdilution. Its effect on growing and mature biofilms and its interaction with antibacterial drugs were evaluated by assessing biofilm metabolic activity, biomass and structure through confocal microscopy. Results: The FLU MIC range was 19.53–312.5 μg/ml. FLU eradicated growing and mature biofilms of B. pseudomallei at 19.53–312.5 μg/ml and 1250–2500 μg/ml, respectively, with no structural alterations and enhanced the antibiofilm activity of antimicrobial drugs. Conclusion: These results bring perspectives for the use of FLU in the treatment of melioidosis, requiring further studies to evaluate its applicability.

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

    References

    • 1. Limmathurotsakul D, Golding N, Dance DAB et al. Predicted global distribution of Burkholderia pseudomallei and burden of melioidosis. Nat. Microbiol. 1(1), 15008 (2016).
    • 2. Benoit TJ, Blaney DD, Doker TJ et al. A review of melioidosis cases in the Americas. Am. J. Trop. Med. Hyg. 93(6), 1134–1139 (2015).
    • 3. Brilhante RSN, Bandeira TJPG, Cordeiro RA et al. Clinical–epidemilogical features of 13 cases of melioidosis in Brazil. J. Clin. Microbiol. 50(10), 3349–3352 (2012a).
    • 4. Dance D, Davong V, Soeng S et al. Trimethoprim/sulfamethoxazole resistance in Burkholderia pseudomallei. Int. J. Antimicrob. Agents 44(4), 368–369 (2014).
    • 5. Brilhante RSN, Valente LGA, Rocha MFG et al. Sesquiterpene farnesol contributes to increased susceptibility to β-lactams in strains of Burkholderia pseudomallei. Antimicrob. Agents Chemother. 56(4), 2198–2200 (2012b).
    • 6. Castelo-Branco DS, Riello GB, Vasconcelos DC et al. Farnesol increases the susceptibility of Burkholderia pseudomallei biofilm to antimicrobials used to treat melioidosis. J. Appl. Microbiol. 120(3), 600–606 (2016).
    • 7. Limmathurotsakul D, Paeyao A, Wongratanacheewin S et al. Role of Burkholderia pseudomallei biofilm formation and lipopolysaccharide in relapse of melioidosis. Clin. Microbiol. Infect. 20(11), 854–856 (2014). • Shows the importance of biofilm production for bacterial virulence and resitance.
    • 8. Munoz-Bellido JL, Munoz-Criado S, García-Rodríguez JA. Antimicrobial activity of psychotropic drugs selective serotonin reuptake inhibitors. Int. J. Antimicrob. Agents 14(3), 177–80 (2000).
    • 9. Kaatz GW, Moudgal VV, Seo SM, Kristiansen JE. Phenothiazines and thioxanthenes inhibit multidrug efflux pump activity in Staphylococcus aureus. Antimicrob. Agents Chemother. 47(2), 719–726 (2003).
    • 10. Kvist M, Hancock V, Klemm P. Inactivation of efflux pumps abolishes bacterial biofilm formation. Appl. Environ. Microbiol. 74(23), 7376–7382 (2008).
    • 11. Bohnert JA, Szymaniak-Vits M, Schuster S, Kern WV. Efflux inhibition by selective serotonin reuptake inhibitors in Escherichia coli. J. Antimicrob. Chemother. 66(9), 2057–2060 (2011).
    • 12. Nzakizwanayo J, Scavone P, Jamshidi S, Hawthorne JA et al. Fluoxetine and thioridazine inhibit efflux and attenuate crystalline biofilm formation by Proteus mirabilis. Sci. Rep. 7(12222), 1–14 (2017). •• Shows that fluoxetine is a promising strategy for the disassembly of bacterial biofilms.
    • 13. Silva RA, Silva CR, Neto JB et al. In vitro anti-Candida activity of selective serotonina reuptake inhibitors against fluconazole-resistant strains and their activity against biofilm-forming isolates. Microb. Pathog. 107, 341–348 (2017).
    • 14. Oliveira AS, Martinez-de-Oliveira J, Donders GG, Palmeira-de-Oliveira R, Palmeira-de-Oliveira A. Anti-Candida activity of antidepressants sertraline and fluoxetine: effect upon pre-formed biofilms. Med. Microbiol. Immunol. 207(3-4), 195–200 (2018).
    • 15. Jiang L, Zheng L, Sun KA et al. In vitro and in vivo evaluation of the antifungal activity of fluoxetine combined with antifungals against Candida albicans biofilms and oral candidiasis. Biofouling 36(5), 537–548 (2020).
    • 16. Pereira TC, Menezes RT, Oliveira HC, Oliveira LD, Scorzoni L. In vitro synergistic effects of fluoxetine and paroxetine in combination with amphothericin B against Cryptococcus neoformans. Pathog. Dis. 79(2), ftab001 (2021).
    • 17. Lyte M, Brown DR. Evidence for PMAT- and OCT-like biogenic amine transporters in a probiotic strain of Lactobacillus: implications for interkingdom communication within the microbiota-gut-brain axis. PLOS ONE 13(1), e0191037 (2018).
    • 18. Josino MA, Silva CR, Neto JB et al. Development and in vitro evaluation of microparticles of fluoxetine in galactomannan against biofilms of S. aureus methicilin resistant. Carbohydr. Polym. 252, 117184 (2021).
    • 19. Bandeira TJPG, Castelo-Branco DSCM, Rocha MFG et al. Clinical and environmental isolates of Burkholderia pseudomallei from Brazil: genotyping and detection of virulence gene. Asian. Pac. J. Trop. Med. 10, 945–951 (2017).
    • 20. CLSI. Methods for dilution antimicrobial susceptibility tests f or bacteria that grow aerobically; approved standard – Ninth Edition. CLSI document M07-A9. Clinical and Laboratory Standards Institute, PA, USA (2012).
    • 21. CLSI. Methods for antimicrobial dilution and disk susceptibility testing of infrequently isolated or fastidious bacteria; approved guideline – Second Edition. CLSI document M45-A3. Clinical and Laboratory Standards Institute, PA, USA (2015).
    • 22. BrCast. Tabelas de pontos de corte para interpretação de CIMs e diâmetros de halos. Brazilian Committee on Antimicrobial Susceptibility Testing: 2023. https://brcast.org.br/documentos/documentos-3/
    • 23. Sidrim JJC, Ocadaque CJ, Amando BR et al. Rhamnolipid enhances Burkholderia pseudomallei biofilm susceptibility, disassembly and production of virulence factors. Future Microbiol. 15, 1109–1121 (2020).
    • 24. Sidrim JJC, Vasconcelos DC, Riello GB et al. Promethazine improves antibiotic efficacy and disrupts biofilms of Burkholderia pseudomallei. Biofouling 33(1), 88–97 (2017).
    • 25. Muhammad MH, Idris AL, Fan X et al. Beyond risk: bacterial biofilms and their regulating approaches. Front. Microbiol. 11(928), 1–20 (2020).
    • 26. Yin W, Wang Y, Liu L, He J. Biofilms: the microbial “protective clothing” in extreme environments. Int. J. Mol. Sci. 20(14), 3423 (2019).
    • 27. Hadera M, Mehari S, Basha NS, Amha ND, Berhane Y. Study on antimicrobial potential of selected non-antibiotics and its interaction with conventional antibiotics. J. Pharm. Biosci. 6(1), 1–7 (2018).
    • 28. Das S, Basak R, Roy S. Screening of antimicrobial effect of some non-antibiotics (anti-hypertensive) against some selected microorganisms. Ind. Res. J. Pharm. Sci. 4(3), 1068–1079 (2017).
    • 29. Sousa AK, Rocha JE, Souza TG et al. New roles of fluoxetine in pharmacology: antibacterial effect and modulation of antibiotic activity. Microb. Pathog. 123, 368–371 (2018). •• Shows that fluoxetine is a promising strategy for modulation of antibiotic activity.
    • 30. Neto JB, Josino MA, Silva CR et al. A mechanistic approach to the in-vitro resistance modulating effects of fluoxetine against meticillin resistant Staphylococcus aureus strains. Microb. Pathog. 127, 335–340 (2019).
    • 31. Schweizer HP. Understanding efflux in Gram-negative bacteria: opportunities for drug discovery. Expert Opin. Drug. Discov. 7(7), 633–642 (2012).
    • 32. Oliveira AS, Gaspar CA, Palmeira-de-Oliveira R, Martinez-de-Oliveira J, Palmeira-de-Oliveira A. Anti-Candida activity of fluoxetine alone and combined with fluconazole: a synergistic action against fluconazole-resistant strains. Antimicrob. Agents Chemother. 58(7), 4224–4226 (2014).
    • 33. Foletto VS, Serafin MB, Bottega A et al. Repositioning of fluoxetine and paroxetine: study of potential antibacterial activity and its combination with ciprofloxacin. Med. Chem. Res. 29, 556–563 (2020). •• Shows that fluoxetine is a promising strategy for modulation of antibiotic activity.
    • 34. Jin M, Lu J, Chen Z et al. Antidepressant fluoxetine induces multiple antibiotics resistance in Escherichia coli via ROS-mediated mutagenesis. Environ. Int. 120, 421–430 (2018).
    • 35. Alav I, Sutton JM, Rahman KM. Role of bacterial efflux pumps in biofilm formation. J. Antimicrob. Chemother. 73(8), 2003–2020 (2018).
    • 36. Dusman E, Almeida IV, Mariucii RG, Mantovani MS, Vicentini VE. Cytotoxicity and mutagenicity of fluoxetine hydrochloride (Prozac), with or without vitamins A and C, in plant and animal model systems. Genet. Mol. Res. 13(1), 578–589 (2014).
    • 37. Rosetti M, Frasnelli M, Tesei A, Zoli W, Conti M. Cytotoxicity of different selective serotonin reuptake inhibitors (SSRIs) against cancer cells. J. Exp. Ther. Oncol. 6(1), 23–29 (2006).
    • 38. Tekintas Y, Temel A, Ates A et al. Antifungal and antibiofilm activities of selective serotonin reuptake inhibitors alone and in combination with fluconazole. Turk. J. Pharm. Sci. 17(6), 667–672 (2020).
    • 39. Gu W, Guo D, Zhang L, Xu D, Sun S. The synergistic effect of azoles and fluoxetine against resistant Candida albicans strains is attributed to attenuating fungal virulence. Antimicrob. Agents Chemother. 60(10), 6179–6188 (2016).