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

3-phenacylideneoxindoles as a new class of antifungal compounds against Paracoccidioides spp.

    Lívia C Silva

    **Author for correspondence:

    E-mail Address: liviacarmo.bio@gmail.com

    Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil

    ,
    Raimundo F dos Santos Filho

    Chemistry institute, Federal University of Bahia, Salvador, Bahia, 40170-115, Brazil

    ,
    Amanda A de Oliveira

    Institute of Tropical Pathology & Public Health, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil

    ,
    Felipe T Martins

    Chemistry institute, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil

    ,
    Silvio Cunha

    Chemistry institute, Federal University of Bahia, Salvador, Bahia, 40170-115, Brazil

    ,
    Célia M de Almeida Soares

    Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil

    &
    Maristela Pereira

    *Author for correspondence: Tel.: +55 623 521 1110;

    E-mail Address: maristelaufg@gmail.com

    Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil

    Published Online:https://doi.org/10.2217/fmb-2022-0133

    Aims: Considering the need to identify new compounds with antifungal action, the activity of five 3-phenacylideneoxindoles compounds was evaluated. Materials & methods: The compounds were synthesized, and their antifungal activity was elucidated through minimum inhibitory concentration tests and interaction assay with other antifungals. Potential targets of compounds were predicted in silico. Results: 3-phenacylideneoxindoles compounds inhibited fungal growth with minimum inhibitory concentration and minimum fungicidal concentration ranging from 3.05 to 12.26 μM. The compounds demonstrated high selectivity index and presented a synergistic effect with itraconazole. In silico prediction revealed the pentafunctional AROM polypeptide, enolase, superoxide dismutase, catalase and kinases as proteins targets of the compound 4a. Conclusion: The results demonstrate that 3-phenacylideneoxindoles is a potential new class of antifungal compounds for paracoccidioidomycosis treatment.

    Plain language summary

    Patients affected by paracoccidioidomycosis (PCM) require long-term treatment, which commonly influences their adherence. In addition, only three drugs are in clinical use, which indicates the relevance of research in identifying new drugs for treating PCM. Thus, five drugs were tested in the laboratory to verify whether they could prevent the growth of the fungus without being toxic to humans. In addition, whether these compounds in combination with drugs used to treat PCM could be even more potent was evaluated. All compounds tested efficiently inhibited the growth of Paracoccidioides, the fungus that causes PCM. One drug was identified that, combined with itraconazole, decreased the required dose of both the discovered compound and itraconazole needed to inhibit fungal growth. Using computational tools, this work suggests how the new drug could act against the fungus. The results demonstrate a potential new treatment option, but more studies are needed to confirm the safety of these drugs.

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

    References

    • 1. Martinez R. New trends in paracoccidioidomycosis epidemiology. J. Fungi 3(1), 1 (2017).
    • 2. Hotez PJ, Bottazzi ME, Franco-Paredes C, Ault SK, Periago MR. The neglected tropical diseases of Latin America and the Caribbean: a review of disease burden and distribution and a roadmap for control and elimination. PLOS Negl. Trop. Dis. 2(9), e300 (2008).
    • 3. Shikanai-Yasuda MA, Mendes RP, Colombo AL et al. Brazilian guidelines for the clinical management of paracoccidioidomycosis. Rev. Soc. Bras. Med. Trop. 50(5), 715–740 (2017). •• Presents the consensus on paracoccidioidomycosis (PCM) that establish the guidelines of the clinical, diagnostic and treatment approach.
    • 4. Bocca AL, Amaral AC, Teixeira MM, Sato PK et al. Paracoccidioidomycosis: eco-epidemiology, taxonomy and clinical and therapeutic issues. Future Microbiol. 8(9), 1177–1191 (2013).
    • 5. Martinez R. Epidemiology of paracoccidioidomycosis. Rev. Inst. Med. Trop. São Paulo 57(Suppl. 19), 11–20 (2015).
    • 6. Shikanai-Yasuda MA. Paracoccicidoidomycosis treatment. Rev. Inst. Med. Trop. São Paulo 57(Suppl. 19), 31–37 (2015).
    • 7. do Carmo Silva L, de Oliveira AA, de Souza DR et al. Overview of antifungal drugs against paracoccidioidomycosis: how do we start, where are we, and where are we going? J. Fungi 6(4), 300 (2020). • Presents the history of development of the current antifungals used in the treatment of PCM as it presents the compounds already tested against the fungus.
    • 8. da Silva LS, Barbosa UR, Silva L do C et al. Identification of a new antifungal compound against isocitrate lyase of Paracoccidioides brasiliensis. Future Microbiol. 14(18), 1589–1606 (2019).
    • 9. Silva LC, Neves BJ, Gomes MN et al. Computer-aided identification of novel anti-paracoccidioidomycosis compounds. Future Microbiol. 13(13), 1523–1535 (2018).
    • 10. Wang J, Yun D, Yao J et al. Design, synthesis and QSAR study of novel isatin analogues inspired Michael acceptor as potential anticancer compounds. Eur. J. Med. Chem. 144, 493–503 (2018).
    • 11. Klöck C, Jin X, Choi K et al. Acylideneoxoindoles: a new class of reversible inhibitors of human transglutaminase 2. Bioorg. Med. Chem. Lett. 21(9), 2692–2696 (2011).
    • 12. Woodard CL, Li Z, Kathcart AK et al. Oxindole-based compounds are selective inhibitors of Plasmodium falciparum cyclin dependent protein kinases. J. Med. Chem. 46(18), 3877–3882 (2003).
    • 13. Gupta A, Kalpana S, Malik J. Synthesis and In vitro antioxidant activity of new 3-substituted-2-oxindole derivatives. Indian J. Pharm. Sci. 74(5), 481 (2012).
    • 14. Pajouhesh H, Parson R, Popp FD. Potential anticonvulsants VI: condensation of isatins with cyclohexanone and other cyclic ketones. J. Pharm. Sci. 72(3), 318–321 (1983).
    • 15. Kaur J, Utreja D, Ekta et al. Recent developments in the synthesis and antimicrobial activity of indole and its derivatives. COS 16(1), 17–37 (2019). • Presents the advances in the synthesis of indole derivatives and antimicrobial properties exhibited by them.
    • 16. Özdemir A, Altintop MD, Kaplancıklı ZA et al. Synthesis and biological evaluation of pyrazoline derivatives bearing an indole moiety as new antimicrobial agents. Arch. Pharm. Pharm. Med. Chem. 346(6), 463–469 (2013).
    • 17. Zhong F, Jiang C, Yao W et al. Molecular sieve mediated decarboxylative Mannich and aldol reactions of β-ketoacids. Tetrahedron Lett. 54(32), 4333–4336 (2013).
    • 18. Bai M, You Y, Chen Y-Z et al. An unprecedented protocol for the synthesis of 3-hydroxy-3-phenacyloxindole derivatives with indolin-2-ones and α-substituted ketones. Org. Biomol. Chem. 14(4), 1395–1401 (2016).
    • 19. Thanikachalam PV, Maurya RK, Garg V, Monga V. An insight into the medicinal perspective of synthetic analogs of indole: a review. Eur. J. Med. Chem. 180, 562–612 (2019).
    • 20. Bruker. Bruker. APEX2, SAINT e SADABS. Bruker AXS Inc., Madison., Bruker AXS Inc., Madison (2009).
    • 21. Sheldrick GM. A short history of SHELX. Acta Crystallog A. 64(1), 112–122 (2007).
    • 22. CLSI. Clinical and Laboratory Standards Institute. Reference method for broth dilution antifungal susceptibility testing of yeasts, approved standard. Document M27 (2008). https://clsi.org/standards/products/microbiology/documents/m27/
    • 23. Rocha OB, do Carmo Silva L, de Carvalho Júnior MAB et al. In vitro and in silico analysis reveals antifungal activity and potential targets of curcumin on Paracoccidioides spp. Braz. J. Microbiol. 52(4), 1897–1911 (2021).
    • 24. Jaccard P. Distribution de la flore alpine dans le Bassin des Dranses et dans quelques regiões voisines. Bulletin de la Société Vaudoise des Sciences Naturelles. La socit Vaudoise Des Sxi. Nat. 37, 241–272 (1901).
    • 25. Bajusz D, Rácz A, Héberger K. Why is Tanimoto index an appropriate choice for fingerprint-based similarity calculations? J. Chem. 7(1), 1–13 (2015).
    • 26. Anderson S. Graphical representation of molecules and substructure-search queries in MACCStm. J. Mol. Graph. 2(3), 83–90 (1984).
    • 27. Berthold MR, Cebron N, Dill F et al. KNIME: the Konstanz Information Miner. In: Data Analysis, Machine Learning and Applications. Preisach CBurkhardt HSchmidt-Thieme LDecker R (Eds). Springer Berlin Heidelberg, Berlin, Heidelberg, 319–326 (2008).
    • 28. Beisken S, Meinl T, Wiswedel B et al. KNIME-CDK: Workflow-driven cheminformatics. BMC Bioinform. 14(1), 257 (2013).
    • 29. Keiser MJ, Roth BL, Armbruster BN et al. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 25(2), 197–206 (2007). •• Demonstrates the technique that quantitatively groups and relates proteins based on the chemical similarity of their ligands and proposes the PharmMapper server.
    • 30. Waterhouse A, Bertoni M, Bienert S et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46(W1), W296–W303 (2018).
    • 31. Williams CJ, Headd JJ, Moriarty NW et al. MolProbity: more and better reference data for improved all-atom structure validation. Protein Sci. 27(1), 293–315 (2018).
    • 32. Bhattacharya D, Nowotny J, Cao R, Cheng J. 3D refine: an interactive web server for efficient protein structure refinement. Nucleic acids Res. 44(W1), W406–W409 (2016).
    • 33. Fourches D, Muratov E, Tropsha A. Trust, but verify II: A practical guide to chemogenomics data curation. J. Chem. Inf. Model. 56(7), 1243–1252 (2016).
    • 34. OpenEye Scientific. OMEGA v.3.0.0.1: OpenEye Scientific Software, Santa Fe, NM. http://www.eyesopen.com
    • 35. Jakalian A, Jack DB, Bayly CI. Fast, efficient generation of high-quality atomic charges. AM1-BCC model: II. Parameterization and validation. J. Comput. Chem. 23(16), 1623–1641 (2002).
    • 36. OpenEye Scientific. QUACPAC v.1.7.0.2: OpenEye Scientific Software, Santa Fe, NM. http://www.eyesopen.com
    • 37. OpenEye Scientific. OEDocking v.3.2.0: OpenEye Scientific Software, Santa Fe, NM, USA. http://www.eyesopen.com
    • 38. McGann M. FRED and HYBRID docking performance on standardized datasets. J. Comput. Aided Mol. Des. 26(8), 897–906 (2012).
    • 39. Edeson SJ, Jiang J, Swanson S et al. Studies on the stereochemical assignment of 3-acylidene 2-oxindoles. Org. Biomol. Chem. 12(20), 3201 (2014).
    • 40. Jain AK, Sharma S, Vaidya A, Ravichandran V, Agrawal RK. 1,3,4-Thiadiazole and its derivatives: a review on recent progress in biological activities. Chem. Biol. Drug Des. 81(5), 557–576 (2013).
    • 41. Khetmalis YM, Shivani M, Murugesan S, Chandra Sekhar KVG. Oxindole and its derivatives: a review on recent progress in biological activities. Biomed. Pharmacother. 141, 111842 (2021).
    • 42. Restrepo A, Tabares C de BAM. In vitro susceptibility of Paracoccidioides brasiliensis yeast form to antifungal agents. Rev. Inst. Med. Trop. São Paulo 26(6), 322–328 (1984).
    • 43. Bueno PSA, Rodrigues FAV, Santos JL et al. New inhibitors of homoserine dehydrogenase from Paracoccidioides brasiliensis presenting antifungal activity. J. Mol. Model. 25(11), 325 (2019).
    • 44. Silva L do C, Tauhata SBF, Baeza LC et al. Argentilactone molecular targets in Paracoccidioides brasiliensis identified by chemoproteomics. Antimicrob. Agents Chemother. 62(11), e00737–18 (2018).
    • 45. do Carmo Silva L, Miranda MACM, de Freitas JV et al. Antifungal activity of Copaíba resin oil in solution and nanoemulsion against Paracoccidioides spp. Braz. J. Microbiol. 51(1), 125–134 (2020).
    • 46. Liu J, Jung JH, Liu Y. Antimicrobial compounds from marine invertebrates-derived microorganisms. Curr. Med. Chem. 23(25), 2892–2905 (2016).
    • 47. Comess KM, McLoughlin SM, Oyer JA et al. Emerging approaches for the identification of protein targets of small molecules – a practitioners' perspective. J. Med. Chem. 61(19), 8504–8535 (2018).
    • 48. Wang Y-H, Dong H-H, Zhao F et al. The synthesis and synergistic antifungal effects of chalcones against drug resistant Candida albicans. Bioorganic Med. Chem. Lett. 26(13), 3098–3102 (2016).
    • 49. Bentley R, Haslam E. The Shikimate pathway – a metabolic tree with many branche. Crit. Rev. Biochem. Mol. Biol. 25(5), 307–384 (1990).
    • 50. Nogueira SV, Fonseca FL, Rodrigues ML et al. Paracoccidioides brasiliensis enolase is a surface protein that binds plasminogen and mediates interaction of yeast forms with host cells. Infect. Immun. 78(9), 4040–4050 (2010).
    • 51. Marcos CM, de Fátima da Silva J, de Oliveira HC, Moraes da Silva RA, Mendes-Giannini MJS, Fusco-Almeida AM. Surface-expressed enolase contributes to the adhesion of Paracoccidioides brasiliensis to host cells. FEMS Yeast Res. 12(5), 557–570 (2012).
    • 52. Tamayo D, Muñoz JF, Lopez Á et al. Identification and analysis of the role of superoxide dismutases isoforms in the pathogenesis of Paracoccidioides spp. PLOS Negl. Trop. Dis. 10(3), e0004481 (2016).
    • 53. Tamayo D, Muñoz JF, Almeida AJ et al. Paracoccidioides spp. catalases and their role in antioxidant defense against host defense responses. Fungal Genet. Biol. 100, 22–32 (2017).
    • 54. Caplan T, Lorente-Macías Á, Stogios PJ et al. Overcoming fungal echinocandin resistance through inhibition of the non-essential stress kinase Yck2. Cell Chem. Biol. 27(3), 269–282.e5 (2020).
    • 55. Fuchs BB, Mylonakis E. Our paths might cross: the role of the fungal cell wall integrity pathway in stress response and cross talk with other stress response pathways. Eukaryot. Cell 8(11), 1616–1625 (2009).
    • 56. Levin DE, Bartlett-Heubusch E. Mutants in the S. cerevisiae PKC1 gene display a cell cycle-specific osmotic stability defect. J. Cell Biol. 116(5), 1221–1229 (1992).
    • 57. Jung K-W, Lee Y, Huh EY et al. Rad53- and Chk1-dependent DNA damage response pathways cooperatively promote fungal pathogenesis and modulate antifungal drug susceptibility. mBio 10(1), e01726-18 (2019).