TY - JOUR AU - Rónavári, Andrea AU - Igaz, Nóra AU - Gopisetty, Mohana Krishna AU - Szerencsés, Bettina AU - Kovács, Dávid AU - Papp, Csaba Gergő AU - Vágvölgyi, Csaba AU - Boros, Imre Miklós AU - Kónya, Zoltán AU - Csontné Kiricsi, Mónika AU - Pfeiffer, Ilona TI - Biosynthesized silver and gold nanoparticles are potent antimycotics against opportunistic pathogenic yeasts and dermatophytes JF - INTERNATIONAL JOURNAL OF NANOMEDICINE J2 - INT J NANOMED VL - 13 PY - 2018 SP - 695 EP - 703 PG - 9 SN - 1176-9114 DO - 10.2147/IJN.S152010 UR - https://m2.mtmt.hu/api/publication/3335848 ID - 3335848 N1 - Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary MTA-SZTE Reaction Kinetics and Surface Chemistry Research Group, Szeged, Hungary Cited By :27 Export Date: 31 May 2021 Correspondence Address: Pfeiffer, I.; Department of Microbiology, Közép fasor 52, Hungary; email: pfeiffer@bio.u-szeged.hu AB - Background: Epidemiologic observations indicate that the number of systemic fungal infections has increased significantly during the past decades, however in human mycosis, mainly cutaneous infections predominate, generating major public health concerns and providing much of the impetus for current attempts to develop novel and efficient agents against cutaneous mycosis causing species. Innovative, environmentally benign and economic nanotechnology-based approaches have recently emerged utilizing principally biological sources to produce nano-sized structures with unique antimicrobial properties. In line with this, our aim was to generate silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs) by biological synthesis and to study the effect of the obtained nanoparticles on cutaneous mycosis causing fungi and on human keratinocytes. Methods: Cell-free extract of the red yeast Phaffia rhodozyma proved to be suitable for nanoparticle preparation and the generated AgNPs and AuNPs were characterized by transmission electron microscopy, dynamic light scattering and X-ray powder diffraction. Results: Antifungal studies demonstrated that the biosynthesized silver particles were able to inhibit the growth of several opportunistic Candida or Cryptococcus species and were highly potent against filamentous Microsporum and Trichophyton dermatophytes. Among the tested species only Cryptococcus neoformans was susceptible to both AgNPs and AuNPs. Neither AgNPs nor AuNPs exerted toxicity on human keratinocytes. Conclusion: Our results emphasize the therapeutic potential of such biosynthesized nanoparticles, since their biocompatibility to skin cells and their outstanding antifungal performance can be exploited for topical treatment and prophylaxis of superficial cutaneous mycosis. © 2018 Rónavári et al. LA - English DB - MTMT ER - TY - JOUR AU - Németh, Tibor Mihály AU - Tóth, Adél AU - Szenzenstein, Judit AU - Horváth, Péter Ferenc AU - Nosanchuk, JD AU - Grózer, Zsuzsanna Barbara AU - Tóth, Renáta AU - Papp, Csaba Gergő AU - Kozma-Bognárné Hamari, Zsuzsanna AU - Vágvölgyi, Csaba AU - Gácser, Attila TI - Characterization of Virulence Properties in the C. parapsilosis Sensu Lato Species. JF - PLOS ONE J2 - PLOS ONE VL - 8 PY - 2013 IS - 7 PG - 10 SN - 1932-6203 DO - 10.1371/journal.pone.0068704 UR - https://m2.mtmt.hu/api/publication/2367028 ID - 2367028 AB - The C. parapsilosis sensu lato group involves three closely related species, C. parapsilosis sensu stricto, C. orthopsilosis and C. metapsilosis. Although their overall clinical importance is dramatically increasing, there are few studies regarding the virulence properties of the species of the psilosis complex. In this study, we tested 63 C. parapsilosis sensu stricto, 12 C. metapsilosis and 18 C. orthopsilosis isolates for the ability to produce extracellular proteases, secrete lipases and form pseudohyphae. Significant differences were noted between species, with the C. metapsilosis strains failing to secrete lipase or to produce pseudohyphae. Nine different clinical isolates each of C. parapsilosis sensu stricto, C. orthopsilosis and C. metapsilosis were co-cultured with immortalized murine or primary human macrophages. C. parapsilosis sensu stricto isolates showed a significantly higher resistance to killing by primary human macrophages compared to C. orthopsilosis and C. metapsilosis isolates. In contrast, the killing of isolates by J774.2 mouse macrophages did not differ significantly between species. However, C. parapsilosis sensu stricto isolates induced the most damage to murine and human macrophages, and C. metapsilosis strains were the least toxic. Furthermore, strains that produced lipase or pseudohyphae were most resistant to macrophage-mediated killing and produced the most cellular damage. Finally, we used 9 isolates of each of the C. parapsilosis sensus lato species to examine their impact on the survival of Galleriamellonella larvae. The mortality rate of G. mellonella larvae infected with C. metapsilosis isolates was significantly lower than those infected with C. parapsilosis sensu stricto or C. orthopsilosis strains. Taken together, our findings demonstrate that C. metapsilosis is indeed the least virulent member of the psilosis group, and also highlight the importance of pseudohyphae and secreted lipases during fungal-host interactions. LA - English DB - MTMT ER - TY - JOUR AU - Papp, Csaba Gergő AU - Kocsis, Katica AU - Tóth, Renáta AU - Bodai, László AU - Willis, Jesse R. AU - Ksiezopolska, Ewa AU - Lozoya-Pérez, Nancy E. AU - Vágvölgyi, Csaba AU - Mora Montes, Hector AU - Gabaldón, Toni AU - Nosanchuk, Joshua D. AU - Gácser, Attila TI - Echinocandin-Induced Microevolution of Candida parapsilosis Influences Virulence and Abiotic Stress Tolerance JF - mSPHERE J2 - MSPHERE VL - 3 PY - 2018 IS - 6 SN - 2379-5042 DO - 10.1128/mSphere.00547-18 UR - https://m2.mtmt.hu/api/publication/30376956 ID - 30376956 N1 - Funding Agency and Grant Number: Spanish Ministry of Economy, Industry, and Competitiveness (MEIC) [BFU2015-67107]; Catalan Research Agency (AGAUR) [SGR857]; European Union's Horizon 2020 research and innovation program [ERC-2016-724173]; European Union's Horizon 2020 research and innovation program under Marie Sklodowska-Curie grant [H2020-MSCA-ITN-2014-642095]; Consejo Nacional de Ciencia y TecnologiaConsejo Nacional de Ciencia y Tecnologia (CONACyT) [PDCPN2014-247109, FC 2015-02-834]; Universidad de Guanajuato [1025/2016, CIIC 95/2018]; NKFIH grant [K112294]; [GINOP-2.3.2-15-2016-00035NKFIH K123952] Funding text: This research was supported by GINOP-2.3.2-15-2016-00035 and NKFIH K123952. T.G.'s research group acknowledges support from the Spanish Ministry of Economy, Industry, and Competitiveness (MEIC) for grant BFU2015-67107 cofounded by European Regional Development Fund (ERDF), from the Catalan Research Agency (AGAUR) SGR857, and a grant from the European Union's Horizon 2020 research and innovation program under grant agreement ERC-2016-724173, the Marie Sklodowska-Curie grant agreement H2020-MSCA-ITN-2014-642095. H.M.M. and N.E.L.-P. are supported by Consejo Nacional de Ciencia y Tecnologia (reference PDCPN2014-247109, and FC 2015-02-834), and Universidad de Guanajuato (reference 1025/2016; CIIC 95/2018). L.B. is supported by NKFIH grant K112294. Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Guanajuato, Mexico Universitat Pompeu Fabra, Barcelona, Spain ICREA, Barcelona, Spain Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States MTA-SZTE Lendület Mycobiome Research Group, University of Szeged, Szeged, Hungary Cited By :2 Export Date: 14 July 2020 Correspondence Address: Gácser, A.; Department of Microbiology, Faculty of Science and Informatics, University of SzegedHungary; email: gacsera@gmail.com Chemicals/CAS: echinocandin, 80619-41-6; glucosyltransferase, 9031-48-5; Antifungal Agents; Echinocandins; Glucosyltransferases LA - English DB - MTMT ER - TY - JOUR AU - Szerencsés, Bettina AU - Igaz, Nóra AU - Tóbiás, Ákos AU - Prucsi, Zsombor AU - Rónavári, Andrea AU - Bélteky, Péter AU - Madarász, Dániel AU - Papp, Csaba Gergő AU - Makra, Ildikó AU - Vágvölgyi, Csaba AU - Kónya, Zoltán AU - Pfeiffer, Ilona AU - Csontné Kiricsi, Mónika TI - Size-dependent activity of silver nanoparticles on the morphological switch and biofilm formation of opportunistic pathogenic yeasts JF - BMC MICROBIOLOGY J2 - BMC MICROBIOL VL - 20 PY - 2020 IS - 1 PG - 13 SN - 1471-2180 DO - 10.1186/s12866-020-01858-9 UR - https://m2.mtmt.hu/api/publication/31353819 ID - 31353819 N1 - Department of Microbiology, University of Szeged, Szeged, Hungary Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, Szeged, H-6726, Hungary Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary HAS-USZ Reaction Kinetics and Surface Chemistry Research Group, Szeged, Hungary Cited By :2 Export Date: 31 May 2021 Correspondence Address: Kiricsi, M.; Department of Biochemistry and Molecular Biology, Közép fasor 52, Hungary; email: kiricsim@gmail.com LA - English DB - MTMT ER - TY - JOUR AU - Kovács, Renátó AU - Holzknecht, J AU - Hargitai, Z AU - Papp, Csaba Gergő AU - Farkas, Attila AU - Borics, Attila AU - Tóth, Liliána AU - Váradi, Györgyi AU - Tóth, Gábor AU - Kovács, Ilona AU - Dubrac, S AU - Majoros, László AU - Marx, F AU - Galgóczi, László Norbert TI - In vivo applicability of Neosartorya fischeri antifungal protein 2 (NFAP2) in treatment of vulvovaginal candidiasis JF - ANTIMICROBIAL AGENTS AND CHEMOTHERAPY J2 - ANTIMICROB AGENTS CH VL - 63 PY - 2019 IS - 2 PG - 12 SN - 0066-4804 DO - 10.1128/AAC.01777-18 UR - https://m2.mtmt.hu/api/publication/30332415 ID - 30332415 N1 - Funding Agency and Grant Number: Postdoctoral Excellence Programme of the Hungarian National Research, Development, and Innovation Office (NKFI Office) [PD 120808]; bilateral Austrian-Hungarian Joint Research Project of the Hungarian National Research, Development, and Innovation Office (NKFI Office) [ANN 122833]; Austrian Science Fund [I3132-B21]; European Union; European Regional Development Fund [GINOP-2.3.2-15-2016-00014]; Janos Bolyai Research Scholarship of the Hungarian Academy of Sciences [UNKP-18-4]; New National Excellence Program of the Ministry of Human Capacities [UNKP-18-4] Funding text: L.G. is financed by the Postdoctoral Excellence Programme (PD 120808) and the bilateral Austrian-Hungarian Joint Research Project (ANN 122833) of the Hungarian National Research, Development, and Innovation Office (NKFI Office). This work was supported by a grant from the Austrian Science Fund (I3132-B21) to F.M. The research was also supported by the European Union and cofinanced by the European Regional Development Fund under project GINOP-2.3.2-15-2016-00014 (to G.K.T.). Research of A. B. and L.G. has been supported by the Janos Bolyai Research Scholarship of the Hungarian Academy of Sciences. The present work of L. G. was supported by the UNKP-18-4 New National Excellence Program of the Ministry of Human Capacities. Funding Agency and Grant Number: Postdoctoral Excellence Programme of the Hungarian National Research, Development, and Innovation Office (NKFI Office) [PD 120808]; bilateral Austrian-Hungarian Joint Research Project of the Hungarian National Research, Development, and Innovation Office (NKFI Office) [ANN 122833]; Austrian Science FundAustrian Science Fund (FWF) [I3132-B21]; European UnionEuropean Union (EU); European Regional Development FundEuropean Union (EU) [GINOP-2.3.2-15-2016-00014]; Janos Bolyai Research Scholarship of the Hungarian Academy of SciencesHungarian Academy of Sciences [UNKP-18-4]; New National Excellence Program of the Ministry of Human Capacities [UNKP-18-4] Funding text: L.G. is financed by the Postdoctoral Excellence Programme (PD 120808) and the bilateral Austrian-Hungarian Joint Research Project (ANN 122833) of the Hungarian National Research, Development, and Innovation Office (NKFI Office). This work was supported by a grant from the Austrian Science Fund (I3132-B21) to F.M. The research was also supported by the European Union and cofinanced by the European Regional Development Fund under project GINOP-2.3.2-15-2016-00014 (to G.K.T.). Research of A. B. and L.G. has been supported by the Janos Bolyai Research Scholarship of the Hungarian Academy of Sciences. The present work of L. G. was supported by the UNKP-18-4 New National Excellence Program of the Ministry of Human Capacities. Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria Department of Pathology, Kenézy Gyula Hospital, University of Debrecen, Debrecen, Hungary Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary Department of Dermatology, Venerology and Allergy, Medical University of Innsbruck, Innsbruck, Austria Cited By :4 Export Date: 14 February 2020 CODEN: AMACC Correspondence Address: Galgóczy, L.; Institute of Plant Biology, Biological Research Centre, Hungarian Academy of SciencesHungary; email: galgoczi.laszlo@brc.mta.hu Chemicals/CAS: fluconazole, 86386-73-4; Antifungal Agents Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria Department of Pathology, Kenézy Gyula Hospital, University of Debrecen, Debrecen, Hungary Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary Department of Dermatology, Venerology and Allergy, Medical University of Innsbruck, Innsbruck, Austria Cited By :7 Export Date: 16 July 2020 CODEN: AMACC Correspondence Address: Galgóczy, L.; Institute of Plant Biology, Biological Research Centre, Hungarian Academy of SciencesHungary; email: galgoczi.laszlo@brc.mta.hu Chemicals/CAS: fluconazole, 86386-73-4; Antifungal Agents Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria Department of Pathology, Kenézy Gyula Hospital, University of Debrecen, Debrecen, Hungary Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary Department of Dermatology, Venerology and Allergy, Medical University of Innsbruck, Innsbruck, Austria Cited By :15 Export Date: 23 August 2021 CODEN: AMACC Correspondence Address: Galgóczy, L.; Institute of Plant Biology, Hungary; email: galgoczi.laszlo@brc.mta.hu Chemicals/CAS: fluconazole, 86386-73-4; Antifungal Agents AB - As a consequence of emerging numbers of vulvovaginitis cases caused by azole-resistant and biofilm-forming Candida species, fast and efficient treatment of this infection has become challenging. The problem is further exacerbated by the severe side effects of azoles as long-term-use medications in the recurrent form. There is therefore an increasing demand for novel and safely applicable effective antifungal therapeutic strategies. The small, cysteine-rich, and cationic antifungal proteins from filamentous ascomycetes are potential candidates, as they inhibit the growth of several Candida spp. in vitro; however, no information is available about their in vivo antifungal potency against yeasts. In the present study, we investigated the possible therapeutic application of one of their representatives in the treatment of vulvovaginal candidiasis, Neosartorya fischeri antifungal protein 2 (NFAP2). NFAP2 inhibited the growth of a fluconazole (FLC)-resistant Candida albicans strain isolated from a vulvovaginal infection, and it was effective against both planktonic cells and biofilm in vitro. We observed that the fungal cell-killing activity of NFAP2 is connected to its pore-forming ability in the cell membrane. NFAP2 did not exert cytotoxic effects on primary human keratinocytes and dermal fibroblasts at the MIC in vitro. In vivo murine vulvovaginitis model experiments showed that NFAP2 significantly decreases the number of FLC-resistant C. albicans cells, and combined application with FLC enhances the efficacy. These results suggest that NFAP2 provides a feasible base for the development of a fundamental new, safely applicable mono- or polytherapeutic topical agent for the treatment of superficial candidiasis. LA - English DB - MTMT ER - TY - JOUR AU - Tóth, Renáta AU - Tóth, Adél AU - Papp, Csaba Gergő AU - Jankovics, Ferenc AU - Vágvölgyi, Csaba AU - Alonso, MF AU - Bain, JM AU - Erwig, LP AU - Gácser, Attila TI - Kinetic studies of Candida parapsilosis phagocytosis by macrophages and detection of intracellular survival mechanisms. JF - FRONTIERS IN MICROBIOLOGY J2 - FRONT MICROBIOL VL - 5 PY - 2014 PG - 12 SN - 1664-302X DO - 10.3389/fmicb.2014.00633 UR - https://m2.mtmt.hu/api/publication/2802304 ID - 2802304 N1 - PMC PMC4238376 Department of Microbiology, University of Szeged, Szeged, Hungary Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom Cited By :16 Export Date: 13 February 2020 Correspondence Address: Gácser, A.; Department of Microbiology, University of Szeged, Közép fasor 52, Hungary; email: gacsera@gmail.com Department of Microbiology, University of Szeged, Szeged, Hungary Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom Cited By :16 Export Date: 18 February 2020 Correspondence Address: Gácser, A.; Department of Microbiology, University of Szeged, Közép fasor 52, Hungary; email: gacsera@gmail.com AB - Even though the number of Candida infections due to non-albicans species like C. parapsilosis has been increasing, little is known about their pathomechanisms. Certain aspects of C. parapsilosis and host interactions have already been investigated; however we lack information about the innate cellular responses toward this species. The aim of our project was to dissect and compare the phagocytosis of C. parapsilosis to C. albicans and to another Candida species C. glabrata by murine and human macrophages by live cell video microscopy. We broke down the phagocytic process into three stages: macrophage migration, engulfment of fungal cells and host cell killing after the uptake. Our results showed increased macrophage migration toward C. parapsilosis and we observed differences during the engulfment processes when comparing the three species. The engulfment time of C. parapsilosis was comparable to that of C. albicans regardless of the pseudohypha length and spatial orientation relative to phagocytes, while the rate of host cell killing and the overall uptake regarding C. parapsilosis showed similarities mainly with C. glabrata. Furthermore, we observed difference between human and murine phagocytes in the uptake of C. parapsilosis. UV-treatment of fungal cells had varied effects on phagocytosis dependent upon which Candida strain was used. Besides statistical analysis, live cell imaging videos showed that this species similarly to the other two also has the ability to survive in host cells via the following mechanisms: yeast replication, and pseudohypha growth inside of phagocytes, exocytosis of fungal cells and also abortion of host cell mitosis following the uptake. According to our knowledge this is the first study that provides a thorough examination of C. parapsilosis phagocytosis and reports intracellular survival mechanisms associated with this species. LA - English DB - MTMT ER -