1887

Abstract

Fungal infections have increased in recent decades, with being the fourth most common aetiological agent of nosocomial infections. Disaccharide trehalose has been proposed as a target for the development of new antifungals. In we have examined the susceptibility shown by two mutants deficient in trehalose biosynthesis, namely and , to amphotericin B (AmB) and micafungin (MF).

Minimum inhibitory concentrations (MICs) were calculated according to the Clinical and Laboratory Standards Institute (CLSI) criteria. Cell viability was assessed by cell counting. Intracellular reactive oxygen species (ROS) and the mitochondrial membrane potential were measured by flow cytometry, while the trehalose content and biofilm formation were determined by enzymatic assays.

While the mutant was highly sensitive to AmB exposure, its resistance to MF was similar to that of the wild-type. Notably, the opposite phenotype was recorded in the mutant. In turn, MF induced a significant level of endogenous ROS production in the parental SC5314 and cells, whereas the ROS formation in cells was virtually undetectable. The level of endogenous ROS correlated positively with the rise in mitochondrial activity. Only AmB was able to promote intracellular synthesis of trehalose in the parental strain; it was absent from cells and showed low levels in , confirming the unspecific dephosphorylation of trehalose-6P in . Furthermore, the capacity of both and mutants to form biofilms was drastically reduced after AmB exposure, whereas it increased in cells treated with MF.

Our data lend weight to the idea of using trehalose biosynthesis as a potential target for antifungal therapy.

Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001053
2019-08-05
2024-05-13
Loading full text...

Full text loading...

/deliver/fulltext/jmm/68/10/1479.html?itemId=/content/journal/jmm/10.1099/jmm.0.001053&mimeType=html&fmt=ahah

References

  1. Pfaller MA, Castanheira M. Nosocomial candidiasis: antifungal stewardship and the importance of rapid diagnosis. Med Mycol 2015; 2014:myv076 [View Article]
    [Google Scholar]
  2. Wiederhold NP. Echinocandin Resistance in Candida species: a review of recent developments. Curr Infect Dis Rep 2016; 18:42 [View Article]
    [Google Scholar]
  3. Campoy S, Adrio JL. Antifungals. Biochem Pharmacol 2017; 133:86–96 [View Article]
    [Google Scholar]
  4. Alvarez-Peral FJ, Zaragoza O, Pedreno Y, Argüelles JC. Protective role of trehalose during severe oxidative stress caused by hydrogen peroxide and the adaptive oxidative stress response in Candida albicans . Microbiology 2002; 148:2599–2606 [View Article]
    [Google Scholar]
  5. Eleutherio E, Brasil AdeA, França MB, de Almeida DSG, Rona GB et al. Oxidative stress and aging: learning from yeast lessons. Fungal Biol 2018; 122:514–525 [View Article]
    [Google Scholar]
  6. Argüelles J-C. Why can't vertebrates synthesize trehalose?. J Mol Evol 2014; 79:111–116 [View Article]
    [Google Scholar]
  7. Argüelles J-C. Trehalose as antifungal target: the picture is still incomplete. Virulence 2017; 8:237–238 [View Article]
    [Google Scholar]
  8. Perfect JR, Tenor JL, Miao Y, Brennan RG. Trehalose pathway as an antifungal target. Virulence 2017; 8:143–149 [View Article]
    [Google Scholar]
  9. Maidan MM, De Rop L, Relloso M, Diez-Orejas R, Thevelein JM et al. Combined inactivation of the Candida albicans GPR1 and TPS2 genes results in avirulence in a mouse model for systemic infection. Infect Immun 2008; 76:1686–1694 [View Article]
    [Google Scholar]
  10. Martínez-Esparza M, Martínez-Vicente E, González-Párraga P, Ros JM, García-Peñarrubia P et al. Role of trehalose-6P phosphatase (TPS2) in stress tolerance and resistance to macrophage killing in Candida albicans . Int J Med Microbiol 2009; 299:453–464 [View Article]
    [Google Scholar]
  11. Puttikamonkul S, Willger SD, Grahl N, Perfect JR, Movahed N et al. Trehalose 6-phosphate phosphatase is required for cell wall integrity and fungal virulence but not trehalose biosynthesis in the human fungal pathogen Aspergillus fumigatus . Mol Microbiol 2010; 77:
    [Google Scholar]
  12. Foster AJ, Jenkinson JM, Talbot NJ. Trehalose synthesis and metabolism are required at different stages of plant infection by Magnaporthe grisea . Embo J 2003; 22:225–235 [View Article]
    [Google Scholar]
  13. Petzold EW, Himmelreich U, Mylonakis E, Rude T, Toffaletti D et al. Characterization and regulation of the trehalose synthesis pathway and its importance in the pathogenicity of Cryptococcus neoformans . Infect Immun 2006; 74:5877–5887 [View Article]
    [Google Scholar]
  14. Richards AB, Krakowka S, Dexter LB, Schmid H, Wolterbeek APM et al. Trehalose: a review of properties, history of use and human tolerance, and results of multiple safety studies. Food Chem Toxicol 2002; 40:871–898 [View Article]
    [Google Scholar]
  15. Maicas S, Guirao-Abad JP, Argüelles J-C. Yeast trehalases: two enzymes, one catalytic mission. Biochim Biophys Acta - Gen Subj 1860; 2016:2249–2254
    [Google Scholar]
  16. Martínez-Esparza M, Aguinaga A, González-Párraga P, García-Peñarrubia P, Jouault T et al. Role of trehalose in resistance to macrophage killing: study with a tps1/tps1 trehalose-deficient mutant of Candida albicans . Clin Microbiol Infect 2007; 13:384–394 [View Article]
    [Google Scholar]
  17. Zaragoza O, Blazquez MA, Gancedo C. Disruption of the Candida albicans TPS1 gene encoding trehalose-6-phosphate synthase impairs formation of hyphae and decreases infectivity. J Bacteriol 1998; 180:3809–3815
    [Google Scholar]
  18. Van Dijck P, De Rop L, Szlufcik K, Van Ael E, Thevelein JM. Disruption of the Candida albicans TPS2 gene encoding trehalose-6-phosphate phosphatase decreases infectivity without affecting hypha formation. Infect Immun 2002; 70:1772–1782 [View Article]
    [Google Scholar]
  19. Miao Y, Tenor JL, Toffaletti DL, Maskarinec SA, Liu J et al. Structural and In vivo studies on trehalose-6-phosphate synthase from pathogenic fungi provide insights into its catalytic mechanism, biological necessity, and potential for novel antifungal drug design. MBio 2017; 8:e00643-17 [View Article]
    [Google Scholar]
  20. Zaragoza O, de Virgilio C, Pontón J, Gancedo C. Disruption in Candida albicans of the TPS2 gene encoding trehalose-6-phosphate phosphatase affects cell integrity and decreases infectivity. Microbiology 2002; 148:1281–1290 [View Article]
    [Google Scholar]
  21. González-Párraga P, Hernández JA, Argüelles JC. Role of antioxidant enzymatic defences against oxidative stress H(2)O(2) and the acquisition of oxidative tolerance in Candida albicans . Yeast 2003; 20:1161–1169 [View Article]
    [Google Scholar]
  22. Sangalli-Leite F, Scorzoni L, Mesa-Arango AC, Casas C, Herrero E et al. Amphotericin B mediates killing in Cryptococcus neoformans through the induction of a strong oxidative burst. Microbes Infect 2011; 13:457–467 [View Article]
    [Google Scholar]
  23. Sánchez-Fresneda R, Guirao-Abad JP, Martinez-Esparza M, Maicas S, Valentín E et al. Homozygous deletion of ATC1 and NTC1 genes in Candida parapsilosis abolishes trehalase activity and affects cell growth, sugar metabolism, stress resistance, infectivity and biofilm formation. Fungal Genetics and Biology 2015; 85:45–57 [View Article]
    [Google Scholar]
  24. Kubota H, Senda S, Nomura N, Tokuda H, Uchiyama H. Biofilm formation by lactic acid bacteria and resistance to environmental stress. J Biosci Bioeng 2008; 106:381–386 [View Article]
    [Google Scholar]
  25. O'Toole GA. Microtiter dish biofilm formation assay. JoVE 2011 [View Article]
    [Google Scholar]
  26. Pierce CG, Uppuluri P, Tummala S, Lopez-Ribot JL. A 96 well microtiter plate-based method for monitoring formation and antifungal susceptibility testing of Candida albicans biofilms. J Vis Exp 20102287 [View Article]
    [Google Scholar]
  27. González-Párraga P, Sánchez-Fresneda R, Zaragoza O, Argüelles J-C. Amphotericin B induces trehalose synthesis and simultaneously activates an antioxidant enzymatic response in Candida albicans . Biochim Biophys Acta 1810; 2011:777–783
    [Google Scholar]
  28. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem 1951; 193:265–275
    [Google Scholar]
  29. Guirao-Abad JP, Sánchez-Fresneda R, Alburquerque B, Hernández JA, Argüelles J-C. ROS formation is a differential contributory factor to the fungicidal action of amphotericin B and micafungin in Candida albicans . Int J Med Microbiol 2017; 307:241–248 [View Article]
    [Google Scholar]
  30. Guirao-Abad JP, Sánchez-Fresneda R, Machado F, Argüelles JC, Martínez-Esparza M. Micafungin Enhances the Human Macrophage Response to Candida albicans through β-Glucan Exposure. Antimicrob Agents Chemother 2018; 62: [View Article]
    [Google Scholar]
  31. Mesa-Arango AC, Trevijano-Contador N, Román E, Sánchez-Fresneda R, Casas C et al. The production of reactive oxygen species is a universal action mechanism of amphotericin B against pathogenic yeasts and contributes to the fungicidal effect of this drug. Antimicrob Agents Chemother 2014; 58:6627–6638 [View Article]
    [Google Scholar]
  32. Alonso-Monge R, Navarro-García F, Román E, Negredo AI, Eisman B et al. The Hog1 mitogen-activated protein kinase is essential in the oxidative stress response and chlamydospore formation in Candida albicans . Eukaryot Cell 2003; 2:351–361 [View Article]
    [Google Scholar]
  33. Sánchez-Fresneda R, Martínez-Esparza M, Maicas S, Argüelles J-C, Valentín E. In Candida parapsilosis the ATC1 Gene encodes for an acid trehalase involved in trehalose hydrolysis, stress resistance and virulence. PLoS One 2014; 9:e99113 [View Article]
    [Google Scholar]
  34. Raut JS, Shinde RB, Chauhan NM, Karuppayil SM. Phenylpropanoids of plant origin as inhibitors of biofilm formation by Candida albicans . J Microbiol Biotechnol 2014; 24:1216–1225 [View Article]
    [Google Scholar]
  35. Mukherjee PK, Chandra J. Candida biofilms: development, architecture, and resistance. Microbiol Spectr 2015; 3: [View Article]
    [Google Scholar]
  36. Ellis D. Amphotericin B: spectrum and resistance. J Antimicrob Chemother 2002; 49:7–10 [View Article]
    [Google Scholar]
  37. de Oliveira Santos GC, Vasconcelos CC, Lopes AJO, de Sousa Cartágenes MdoS, Filho AKDB et al. Candida Infections and therapeutic strategies: mechanisms of action for traditional and alternative agents. Front Microbiol 2018; 9:1351 [View Article]
    [Google Scholar]
  38. Vincent BM, Lancaster AK, Scherz-Shouval R, Whitesell L, Lindquist S. Fitness trade-offs restrict the evolution of resistance to amphotericin B. PLoS Biol 2013; 11:e1001692 [View Article]
    [Google Scholar]
  39. Salci TP, Negri M, Abadio AKR, Svidzinski TIE, Kioshima Érika S. Targeting Candida spp. to develop antifungal agents. Drug Discov Today 2018; 23:802–814 [View Article]
    [Google Scholar]
  40. Noubhani A, Bunoust O, Bonini BM, Thevelein JM, Devin A et al. The trehalose pathway regulates mitochondrial respiratory chain content through hexokinase 2 and cAMP in Saccharomyces cerevisiae . J Biol Chem 2009; 284:27229–27234 [View Article]
    [Google Scholar]
  41. Mailloux RJ, Dumouchel T, Aguer C, deKemp R, Beanlands R et al. Hexokinase II acts through UCP3 to suppress mitochondrial reactive oxygen species production and maintain aerobic respiration. Biochem J 2011; 437:301–311 [View Article]
    [Google Scholar]
  42. Gancedo C, Flores C. The importance of a functional trehalose biosynthetic pathway for the life of yeasts and fungi. FEMS Yeast Res 2004; 4:351–359 [View Article]
    [Google Scholar]
  43. Belenky P, Camacho D, Collins JJ. Fungicidal drugs induce a common oxidative-damage cellular death pathway. Cell Rep 2013; 3:350–358 [View Article]
    [Google Scholar]
  44. Cuéllar-Cruz M, López-Romero E, Villagómez-Castro JC, Ruiz-Baca E. Candida species: new insights into biofilm formation. Future Microbiol 2012; 7:755–771 [View Article]
    [Google Scholar]
  45. Marcos-Zambrano LJ, Escribano P, Bouza E, Guinea J. Production of biofilm by Candida and non- Candida spp. isolates causing fungemia: comparison of biomass production and metabolic activity and development of cut-off points. Int J Med Microbiol 2014; 304:1192–1198 [View Article]
    [Google Scholar]
  46. Nett J, Lincoln L, Marchillo K, Andes D. β‐1,3 glucan as a test for central venous catheter biofilm infection. J Infect Dis 2007; 195:1705–1712 [View Article]
    [Google Scholar]
  47. Taff HT, Mitchell KF, Edward JA, Andes DR. Mechanisms of Candida biofilm drug resistance. Future Microbiol 2013; 8:1325–1337 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001053
Loading
/content/journal/jmm/10.1099/jmm.0.001053
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error