51
|
Beom JY, Jung JA, Lee KT, Hwangbo A, Song MC, Lee Y, Lee SJ, Oh JH, Ha SJ, Nam SJ, Cheong E, Bahn YS, Yoon YJ. Biosynthesis of Nonimmunosuppressive FK506 Analogues with Antifungal Activity. JOURNAL OF NATURAL PRODUCTS 2019; 82:2078-2086. [PMID: 31321978 DOI: 10.1021/acs.jnatprod.9b00144] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A reduction in the strong immunosuppressive activity of FK506 (1) is essential for developing this compound as an antifungal agent. Seven new FK506 analogues modified at both the FK506-binding protein 12- and the calcineurin-binding regions were biosynthesized. 9-DeoxoFK520 (7) exhibited a >900-fold reduction in the in vitro immunosuppressive activity but maintained significant antifungal activity, indicating that the C-9 and C-21 positions are critical for separation of immunosuppressive and antifungal activities. 7 exhibited robust synergistic antifungal activity with fluconazole. FK506 (1) is a 23-membered macrolide produced by several Streptomyces species and is used as an immunosuppressive drug to prevent the rejection of transplanted organs. FK506 has also exhibited antifungal, neuroprotective, and neuroregenerative activities. In humans, FK506 binds to FK506-binding protein (FKBP) 12, and the resulting FKBP12-FK506 complex interacts with a Ca2+-calmodulin-dependent phosphatase, calcineurin (CaN). Inactivation of CaN by forming the FKBP12-FK506-CaN ternary complex prevents the activation of nuclear factor of activated T cells (NF-AT), inhibiting the production of interleukin-2 and subsequent T-cell proliferation. This CaN signaling pathway also plays a critical role in the growth and pathogenesis of major fungal pathogens such as Cryptococcus neoformans, Candida albicans, and Aspergillus fumigatus. Therefore, the synthesis of FK506 analogues that can discriminate human FKBP12/CaN from its fungal counterparts may separate antifungal activity from the immunosuppressive activity, thereby allowing the development of a novel antifungal agent.
Collapse
Affiliation(s)
- Ji Yoon Beom
- Department of Chemistry and Nanoscience , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Jin A Jung
- Department of Chemistry and Nanoscience , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Kyung-Tae Lee
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Areum Hwangbo
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Myoung Chong Song
- Department of Chemistry and Nanoscience , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Yeonseon Lee
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Soo Jung Lee
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Ji Hoon Oh
- Department of Biochemistry, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology , Yonsei University , Seoul 03722 , Republic of Korea
| | - Yeo Joon Yoon
- Department of Chemistry and Nanoscience , Ewha Womans University , Seoul 03760 , Republic of Korea
| |
Collapse
|
52
|
Liu Y, Wang W, Yan H, Wang D, Zhang M, Sun S. Anti- Candida activity of existing antibiotics and their derivatives when used alone or in combination with antifungals. Future Microbiol 2019; 14:899-915. [PMID: 31394935 DOI: 10.2217/fmb-2019-0076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Fungal infections are a growing challenge in immunocompromised patients, especially candidiasis. The prolonged use of traditional antifungals to treat Candida infection has caused the emergence of drug resistance, especially fluconazole. Therefore, new therapeutic strategies for Candida infection are warranted. Recently, attention has been paid to the anti-Candida activity of antibiotics and their derivatives. Studies revealed that a series of antibiotics/derivatives displayed potential anti-Candida activity and some of them could significantly increase the susceptibility of antifungals. Interestingly, the derivatives of aminoglycosides were even more active than fluconazole/itraconazole/posaconazole. This article reviews the anti-Candida activities and mechanisms of antibiotics/derivatives used alone or in combination with antifungals. This review will helpfully provide novel insights for overcoming Candida resistance and discovering new antifungals.
Collapse
Affiliation(s)
- Yaxin Liu
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, People's Republic of China
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Weixin Wang
- Department of Pharmacy, Taishan hospital of Shandong Province, Taian, Shandong Province, People's Republic of China
| | - Haiying Yan
- Department of Pharmacy, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan 250014, People's Republic of China
| | - Decai Wang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, People's Republic of China
| | - Min Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, People's Republic of China
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Shujuan Sun
- Department of Pharmacy, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan 250014, People's Republic of China
| |
Collapse
|
53
|
Sonthalia S, Goldust M. Innovative Physical Approaches for Onychomycosis: Peeling, Lasers and Beyond. Skin Appendage Disord 2019; 5:197-200. [PMID: 31367597 DOI: 10.1159/000497044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/16/2019] [Indexed: 01/19/2023] Open
Affiliation(s)
- Sidharth Sonthalia
- Department of Dermatology and Dermatosurgery, Skinnocence: The Skin Clinic and Research Center, Gurugram, India
| | | |
Collapse
|
54
|
Lee JH, Kim YG, Khadke SK, Yamano A, Watanabe A, Lee J. Inhibition of Biofilm Formation by Candida albicans and Polymicrobial Microorganisms by Nepodin via Hyphal-Growth Suppression. ACS Infect Dis 2019; 5:1177-1187. [PMID: 31055910 DOI: 10.1021/acsinfecdis.9b00033] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Candida albicans is an opportunistic pathogenic yeast and is responsible for candidiasis. It readily colonizes host tissues and implant devices, and forms biofilms, which play an important role in pathogenesis and drug resistance. In this study, the antibiofilm, antihyphal, and antivirulence activities of nepodin, isolated from Rumex japonicus roots, were investigated against a fluconazole-resistant C. albicans strain and against polymicrobial-microorganism-biofilm formation. Nepodin effectively inhibited C. albicans biofilm formation without affecting its planktonic cell growth. Also, Rumex-root extract and nepodin both inhibited hyphal growth and cell aggregation of C. albicans. Interestingly, nepodin also showed antibiofilm activities against Candida glabrata, Candida parapsilosis, Staphylococcus aureus, and Acinetobacter baumannii strains and against dual biofilms of C. albicans and S. aureus or A. baumannii but not against Pseudomonas aeruginosa. Transcriptomic analysis performed by RNA-seq and qRT-PCR showed nepodin repressed the expression of several hypha- and biofilm-related genes (ECE1, HGT10, HWP1, and UME6) and increased the expression of several transport genes (CDR4, CDR11, and TPO2), which supported phenotypic changes. Moreover, nepodin reduced C. albicans virulence in a nematode-infection model and exhibited minimal cytotoxicity against the nematode and an animal cell line. These results demonstrate that nepodin and Rumex-root extract might be useful for controlling C. albicans infections and multispecies biofilms.
Collapse
Affiliation(s)
- Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Sagar Kiran Khadke
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Aki Yamano
- Okinawa Research Center Company, Ltd., 12-75 Ulumasi, Okinawa 904-2234, Japan
| | - Akio Watanabe
- Research Institute for Biological Functions, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
55
|
Xu J, Liu R, Sun F, An L, Shang Z, Kong L, Yang M. Eucalyptal D Enhances the Antifungal Effect of Fluconazole on Fluconazole-Resistant Candida albicans by Competitively Inhibiting Efflux Pump. Front Cell Infect Microbiol 2019; 9:211. [PMID: 31281800 PMCID: PMC6595430 DOI: 10.3389/fcimb.2019.00211] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
The frequent emergence of azole-resistant strains has increasingly led azoles to fail in treating candidiasis. Combination with other drugs is a good option to effectively reduce or retard its incidence of resistance. Natural products are a promising synergist source to assist azoles in treating resistant candidiasis. Eucalyptal D (ED), a formyl-phloroglucinol meroterpenoid, is one of the natural synergists, which could significantly enhance the anticandidal activity of fluconazole (FLC) in treating FLC resistant C. albicans. The checkerboard microdilution assay showed their synergistic effect. The agar disk diffusion test illustrated the key role of ED in synergy. The rhodamine 6G (R6G) efflux assay reflected ED could reduce drug efflux, but quantitative reverse transcription PCR analysis revealed the upregulation of CDR1 and CDR2 genes in ED treating group. Efflux pump-deficient strains were hyper-susceptible to ED, thus ED was speculated to be the substrate of efflux pump Cdr1p and Cdr2p to competitively inhibit the excretion of FLC or R6G, which mainly contributed to its synergistic effect.
Collapse
Affiliation(s)
- Jiali Xu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Ruihuan Liu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Fujuan Sun
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lin An
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Zhichun Shang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Minghua Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
56
|
Kumar A, Alam A, Grover S, Pandey S, Tripathi D, Kumari M, Rani M, Singh A, Akhter Y, Ehtesham NZ, Hasnain SE. Peptidyl-prolyl isomerase-B is involved in Mycobacterium tuberculosis biofilm formation and a generic target for drug repurposing-based intervention. NPJ Biofilms Microbiomes 2019; 5:3. [PMID: 30675370 PMCID: PMC6333787 DOI: 10.1038/s41522-018-0075-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/15/2018] [Indexed: 01/03/2023] Open
Abstract
Tuberculosis (TB), a disease caused by Mycobacterium tuberculosis (M.tb), takes one human life every 15 s globally. Disease relapse occurs due to incomplete clearance of the pathogen and reactivation of the antibiotic tolerant bacilli. M.tb, like other bacterial pathogens, creates an ecosystem of biofilm formed by several proteins including the cyclophilins. We show that the M.tb cyclophilin peptidyl-prolyl isomerase (PpiB), an essential gene, is involved in biofilm formation and tolerance to anti-mycobacterial drugs. We predicted interaction between PpiB and US FDA approved drugs (cyclosporine-A and acarbose) by in-silico docking studies and this was confirmed by surface plasmon resonance (SPR) spectroscopy. While all these drugs inhibited growth of Mycobacterium smegmatis (M.smegmatis) when cultured in vitro, acarbose and cyclosporine-A showed bacteriostatic effect while gallium nanoparticle (GaNP) exhibited bactericidal effect. Cyclosporine-A and GaNP additionally disrupted M.tb H37Rv biofilm formation. Co-culturing M.tb in their presence resulted in significant (2–4 fold) decrease in dosage of anti-tubercular drugs- isoniazid and ethambutol. Comparison of the cyclosporine-A and acarbose binding sites in PpiB homologues of other biofilm forming infectious pathogens revealed that these have largely remained unaltered across bacterial species. Targeting bacterial biofilms could be a generic strategy for intervention against bacterial pathogens. Tuberculosis, caused by Mycobacterium tuberculosis, is the leading cause of death due to a single infectious agent. New therapeutic options are needed, and repurposing clinically approved drugs to destroy biofilms is an attractive approach, as these microbial communities are often less susceptible to antibiotics. A team lead by Seyed Hasnain at the Indian Institute of Technology Delhi identified an enzyme, PpiB, from M. tuberculosis that promoted biofilm formation and showed that PpiB interacts with several drugs that are currently used to treat diabetes, immunological diseases and cancer. These drugs destabilise M. tuberculosis biofilms in culture and enhanced the potency of two current anti-tuberculosis antibiotics. Future work is needed to test these medications against tuberculosis in humans, but given PpiB is found in different bacteria, there may be broader promise of using these repurposed drugs to combat other infections.
Collapse
Affiliation(s)
- Ashutosh Kumar
- 1JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,Present Address: Department of Microbiology, Tripura Central University, Suryamaninagar, Agartala, Tripura India
| | - Anwar Alam
- 1JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,2Molecular Infection and Functional Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, New Delhi, India
| | - Sonam Grover
- 1JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Saurabh Pandey
- 3National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.,11Present Address: Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Deeksha Tripathi
- 2Molecular Infection and Functional Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, New Delhi, India.,4Department of Microbiology, Central University of Rajasthan, Ajmer, Rajasthan India
| | - Monika Kumari
- 5Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Himachal Pradesh, India
| | - Mamta Rani
- 6Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, India
| | - Aditi Singh
- 7School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Yusuf Akhter
- 8Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Nasreen Z Ehtesham
- 3National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Seyed E Hasnain
- 1JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,2Molecular Infection and Functional Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, New Delhi, India.,9Dr Reddy's Institute of Life Sciences, University of Hyderabad Campus, Prof CR Rao Road, Hyderabad, India
| |
Collapse
|
57
|
In Vitro Interactions of Amphotericin B Combined with Non-antifungal Agents Against Rhodotorula mucilaginosa Strains. Mycopathologia 2019; 184:35-43. [DOI: 10.1007/s11046-019-0317-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 01/02/2019] [Indexed: 10/27/2022]
|
58
|
Zhang M, Yang X, Wang D, Yu C, Sun S. Antifungal activity of immunosuppressants used alone or in combination with fluconazole. J Appl Microbiol 2018; 126:1304-1317. [PMID: 30307675 DOI: 10.1111/jam.14126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/07/2018] [Accepted: 09/29/2018] [Indexed: 01/26/2023]
Abstract
Fungal infections remain a challenge to clinicians due to the limited available antifungals. With the increasing use of antifungals in clinical practice, drug resistance has been emerging continuously, especially to fluconazole (FLC). Thus, a search for new antifungals and approaches to overcome antifungal resistance is needed. However, the development of new antifungals is usually costly and time consuming; discovering the antifungal activity of non-antifungal agents is one way to address these problems. Interestingly, some researchers have demonstrated that several classes of immunosuppressants (calcineurin inhibitors, glucocorticoids, etc) also displayed potent antifungal activity when used alone or in combination with antifungals, especially with FLC. Some of them could increase FLC's susceptibility against resistant Candida albicans significantly reversing fungal resistance to FLC. This article reviews the antifungal activities of immunosuppressants used alone or in combination with antifungals and their potential antifungal mechanisms that have been discovered so far. Although immunosuppressive agents have been identified as risk factors for fungal infection, we believe these findings are very important for overcoming drug resistance and developing new antifungals.
Collapse
Affiliation(s)
- M Zhang
- School of Pharmaceutical Sciences, Taishan Medical University, Taian, Shandong Province, China
| | - X Yang
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - D Wang
- School of Pharmaceutical Sciences, Taishan Medical University, Taian, Shandong Province, China
| | - C Yu
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - S Sun
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
59
|
Abstract
The prevalence of fungal infections has seen a rise in the past decades due to advances in modern medicine leading to an expanding population of device-associated and immunocompromised patients. Furthermore, the spectrum of pathogenic fungi has changed, with the emergence of multidrug-resistant strains such as C. auris. High mortality related to fungal infections points to major limitations of current antifungal therapy and an unmet need for new antifungal drugs. We screened a library of repurposed FDA-approved inhibitors to identify compounds with activities against a diverse range of fungi in varied phases of growth. The assays identified alexidine dihydrochloride (AXD) to have pronounced antifungal activity, including against preformed biofilms, at concentrations lower than mammalian cell toxicity. AXD potentiated the activity of fluconazole and amphotericin B against Candida biofilms in vitro and prevented biofilm growth in vivo. Thus, AXD has the potential to be developed as a pan-antifungal, antibiofilm drug. Invasive fungal infections due to Candida albicans, Aspergillus fumigatus, and Cryptococcus neoformans constitute a substantial threat to hospitalized immunocompromised patients. Further, the presence of drug-recalcitrant biofilms on medical devices and emergence of drug-resistant fungi, such as Candida auris, introduce treatment challenges with current antifungal drugs. Worse, currently there is no approved drug capable of obviating preformed biofilms, which increase the chance of infection relapses. Here, we screened a small-molecule New Prestwick Chemical Library, consisting of 1,200 FDA-approved off-patent drugs against C. albicans, C. auris, and A. fumigatus, to identify those that inhibit growth of all three pathogens. Inhibitors were further prioritized for their potency against other fungal pathogens and their ability to kill preformed biofilms. Our studies identified the bis-biguanide alexidine dihydrochloride (AXD) as a drug with the highest antifungal and antibiofilm activity against a diverse range of fungal pathogens. Finally, AXD significantly potentiated the efficacy of fluconazole against biofilms, displayed low mammalian cell toxicity, and eradicated biofilms growing in mouse central venous catheters in vivo, highlighting its potential as a pan-antifungal drug. IMPORTANCE The prevalence of fungal infections has seen a rise in the past decades due to advances in modern medicine leading to an expanding population of device-associated and immunocompromised patients. Furthermore, the spectrum of pathogenic fungi has changed, with the emergence of multidrug-resistant strains such as C. auris. High mortality related to fungal infections points to major limitations of current antifungal therapy and an unmet need for new antifungal drugs. We screened a library of repurposed FDA-approved inhibitors to identify compounds with activities against a diverse range of fungi in varied phases of growth. The assays identified alexidine dihydrochloride (AXD) to have pronounced antifungal activity, including against preformed biofilms, at concentrations lower than mammalian cell toxicity. AXD potentiated the activity of fluconazole and amphotericin B against Candida biofilms in vitro and prevented biofilm growth in vivo. Thus, AXD has the potential to be developed as a pan-antifungal, antibiofilm drug.
Collapse
|
60
|
In Vitro and In Vivo Assessment of FK506 Analogs as Novel Antifungal Drug Candidates. Antimicrob Agents Chemother 2018; 62:AAC.01627-18. [PMID: 30181374 DOI: 10.1128/aac.01627-18] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/26/2018] [Indexed: 12/17/2022] Open
Abstract
FK506 (tacrolimus) is an FDA-approved immunosuppressant indicated for the prevention of allograft rejections in patients undergoing organ transplants. In mammals, FK506 inhibits the calcineurin-nuclear factor of activated T cells (NFAT) pathway to prevent T-cell proliferation by forming a ternary complex with its binding protein, FKBP12, and calcineurin. FK506 also exerts antifungal activity by inhibiting calcineurin, which is essential for the virulence of human-pathogenic fungi. Nevertheless, FK506 cannot be used directly as an antifungal drug due to its immunosuppressive action. In this study, we analyzed the cytotoxicity, immunosuppressive activity, and antifungal activity of four FK506 analogs, 31-O-demethyl-FK506, 9-deoxo-FK506, 9-deoxo-31-O-demethyl-FK506, and 9-deoxo-prolyl-FK506, in comparison with that of FK506. The four FK506 analogs generally possessed lower cytotoxicity and immunosuppressive activity than FK506. The FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against Cryptococcus neoformans and Candida albicans, which are two major invasive pathogenic yeasts, due to the inhibition of the calcineurin pathway. Furthermore, the FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against the invasive filamentous fungus Aspergillus fumigatus Notably, 9-deoxo-31-O-demethyl-FK506 and 31-O-demethyl-FK506 exhibited robust synergistic antifungal activity with fluconazole, similar to FK506. Considering the antifungal efficacy, cytotoxicity, immunosuppressive activity, and synergistic effect with commercial antifungal drugs, we selected 9-deoxo-31-O-demethyl-FK506 for further evaluation of its in vivo antifungal efficacy in a murine model of systemic cryptococcosis. Although 9-deoxo-31-O-demethyl-FK506 alone was not sufficient to treat the cryptococcal infection, when it was used in combination with fluconazole, it significantly extended the survival of C. neoformans-infected mice, confirming the synergistic in vivo antifungal efficacy between these two agents.
Collapse
|
61
|
Make azoles active again: chalcones as potent reversal agents of transporters-mediated resistance in Candida albicans. Future Med Chem 2018; 10:2177-2186. [PMID: 30043631 DOI: 10.4155/fmc-2018-0081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Resistance against antifungals used for Candida albicans (Ca) treatment is mediated by two multidrug transporters, Mdr1p and Cdr1p, which are of enormous interest to the development of modulators combined with antifungals. EXPERIMENTAL A set of chalcones was synthesized by condensation reactions in laboratory and was then subject to biological assays to evaluate the effects on different yeast strains. Results: The obtained chalcones were screened using the checkerboard liquid chemosensitization assays. Compounds 4, 10, 12 and 18, when combined with fluconazole, triggered strong sensitization on yeast strains overexpressing CaMdr1p and CaCdr1p, whereas displaying no cytotoxicity by themselves towards control strains and transporter-expressing yeast cells. In the Nile Red transport assay, the two most active compounds, 12 and 18 showed moderate-to-high accumulation of Nile Red with different behaviors towards the two transporters. CONCLUSION Chalcones are promising drug candidates for further development to make azole antifungals active again.
Collapse
|
62
|
Antifungal tolerance is a subpopulation effect distinct from resistance and is associated with persistent candidemia. Nat Commun 2018; 9:2470. [PMID: 29941885 PMCID: PMC6018213 DOI: 10.1038/s41467-018-04926-x] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/05/2018] [Indexed: 11/23/2022] Open
Abstract
Tolerance to antifungal drug concentrations above the minimal inhibitory concentration (MIC) is rarely quantified, and current clinical recommendations suggest it should be ignored. Here, we quantify antifungal tolerance in Candida albicans isolates as the fraction of growth above the MIC, and find that it is distinct from susceptibility/resistance. Instead, tolerance is due to the slow growth of subpopulations of cells that overcome drug stress more efficiently than the rest of the population, and correlates inversely with intracellular drug accumulation. Many adjuvant drugs used in combination with fluconazole, a widely used fungistatic drug, reduce tolerance without affecting resistance. Accordingly, in an invertebrate infection model, adjuvant combination therapy is more effective than fluconazole in treating infections with highly tolerant isolates and does not affect infections with low tolerance isolates. Furthermore, isolates recovered from immunocompetent patients with persistent candidemia display higher tolerance than isolates readily cleared by fluconazole. Thus, tolerance correlates with, and may help predict, patient responses to fluconazole therapy. The authors show that antifungal tolerance, defined as the fraction of growth of a fungal pathogen above the minimal inhibitory concentration, is due to the slow growth of subpopulations of cells that overcome drug stress, and that high tolerance is often associated with persistent infections.
Collapse
|
63
|
Van Dijck P, Sjollema J, Cammue BPA, Lagrou K, Berman J, d’Enfert C, Andes DR, Arendrup MC, Brakhage AA, Calderone R, Cantón E, Coenye T, Cos P, Cowen LE, Edgerton M, Espinel-Ingroff A, Filler SG, Ghannoum M, Gow NA, Haas H, Jabra-Rizk MA, Johnson EM, Lockhart SR, Lopez-Ribot JL, Maertens J, Munro CA, Nett JE, Nobile CJ, Pfaller MA, Ramage G, Sanglard D, Sanguinetti M, Spriet I, Verweij PE, Warris A, Wauters J, Yeaman MR, Zaat SA, Thevissen K. Methodologies for in vitro and in vivo evaluation of efficacy of antifungal and antibiofilm agents and surface coatings against fungal biofilms. MICROBIAL CELL (GRAZ, AUSTRIA) 2018; 5:300-326. [PMID: 29992128 PMCID: PMC6035839 DOI: 10.15698/mic2018.07.638] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
Unlike superficial fungal infections of the skin and nails, which are the most common fungal diseases in humans, invasive fungal infections carry high morbidity and mortality, particularly those associated with biofilm formation on indwelling medical devices. Therapeutic management of these complex diseases is often complicated by the rise in resistance to the commonly used antifungal agents. Therefore, the availability of accurate susceptibility testing methods for determining antifungal resistance, as well as discovery of novel antifungal and antibiofilm agents, are key priorities in medical mycology research. To direct advancements in this field, here we present an overview of the methods currently available for determining (i) the susceptibility or resistance of fungal isolates or biofilms to antifungal or antibiofilm compounds and compound combinations; (ii) the in vivo efficacy of antifungal and antibiofilm compounds and compound combinations; and (iii) the in vitro and in vivo performance of anti-infective coatings and materials to prevent fungal biofilm-based infections.
Collapse
Affiliation(s)
- Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- KU Leuven Laboratory of Molecular Cell Biology, Leuven, Belgium
| | - Jelmer Sjollema
- University of Groningen, University Medical Center Groningen, Department of BioMedical Engineering, Groningen, The Netherlands
| | - Bruno P. A. Cammue
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Department of Plant Systems Biology, VIB, Ghent, Belgium
| | - Katrien Lagrou
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Clinical Department of Laboratory Medicine and National Reference Center for Mycosis, UZ Leuven, Belgium
| | - Judith Berman
- School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Christophe d’Enfert
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - David R. Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Maiken C. Arendrup
- Unit of Mycology, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Axel A. Brakhage
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute (HKI), Dept. Microbiology and Molecular Biology, Friedrich Schiller University Jena, Institute of Microbiology, Jena, Germany
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington DC, USA
| | - Emilia Cantón
- Severe Infection Research Group: Medical Research Institute La Fe (IISLaFe), Valencia, Spain
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- ESCMID Study Group for Biofilms, Switzerland
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Belgium
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mira Edgerton
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY USA
| | | | - Scott G. Filler
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mahmoud Ghannoum
- Center for Medical Mycology, Department of Dermatology, University Hospitals Cleveland Medical Center and Case Western Re-serve University, Cleveland, OH, USA
| | - Neil A.R. Gow
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Hubertus Haas
- Biocenter - Division of Molecular Biology, Medical University Innsbruck, Innsbruck, Austria
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry; Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, USA
| | - Elizabeth M. Johnson
- National Infection Service, Public Health England, Mycology Reference Laboratory, Bristol, UK
| | | | | | - Johan Maertens
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium and Clinical Department of Haematology, UZ Leuven, Leuven, Belgium
| | - Carol A. Munro
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jeniel E. Nett
- University of Wisconsin-Madison, Departments of Medicine and Medical Microbiology & Immunology, Madison, WI, USA
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, USA
| | - Michael A. Pfaller
- Departments of Pathology and Epidemiology, University of Iowa, Iowa, USA
- JMI Laboratories, North Liberty, Iowa, USA
| | - Gordon Ramage
- ESCMID Study Group for Biofilms, Switzerland
- College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Dominique Sanglard
- Institute of Microbiology, University of Lausanne and University Hospital, CH-1011 Lausanne
| | - Maurizio Sanguinetti
- Institute of Microbiology, Università Cattolica del Sacro Cuore, IRCCS-Fondazione Policlinico "Agostino Gemelli", Rome, Italy
| | - Isabel Spriet
- Pharmacy Dpt, University Hospitals Leuven and Clinical Pharmacology and Pharmacotherapy, Dpt. of Pharmaceutical and Pharma-cological Sciences, KU Leuven, Belgium
| | - Paul E. Verweij
- Center of Expertise in Mycology Radboudumc/CWZ, Radboud University Medical Center, Nijmegen, the Netherlands (omit "Nijmegen" in Radboud University Medical Center)
| | - Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Joost Wauters
- KU Leuven-University of Leuven, University Hospitals Leuven, Department of General Internal Medicine, Herestraat 49, B-3000 Leuven, Belgium
| | - Michael R. Yeaman
- Geffen School of Medicine at the University of California, Los Angeles, Divisions of Molecular Medicine & Infectious Diseases, Har-bor-UCLA Medical Center, LABioMed at Harbor-UCLA Medical Center
| | - Sebastian A.J. Zaat
- Department of Medical Microbiology, Amsterdam Infection and Immunity Institute, Academic Medical Center, University of Am-sterdam, Netherlands
| | - Karin Thevissen
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
64
|
Huang MC, Shen M, Huang YJ, Lin HC, Chen CT. Photodynamic Inactivation Potentiates the Susceptibility of Antifungal Agents against the Planktonic and Biofilm Cells of Candida albicans. Int J Mol Sci 2018; 19:ijms19020434. [PMID: 29389883 PMCID: PMC5855656 DOI: 10.3390/ijms19020434] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 11/16/2022] Open
Abstract
Photodynamic inactivation (PDI) has been shown to be a potential treatment modality against Candida infection. However, limited light penetration might leave some cells alive and undergoing regrowth. In this study, we explored the possibility of combining PDI and antifungal agents to enhance the therapeutic efficacy of Candida albicans and drug-resistant clinical isolates. We found that planktonic cells that had survived toluidine blue O (TBO)-mediated PDI were significantly susceptible to fluconazole within the first 2 h post PDI. Following PDI, the killing efficacy of antifungal agents relates to the PDI dose in wild-type and drug-resistant clinical isolates. However, only a 3-log reduction was found in the biofilm cells, suggesting limited therapeutic efficacy under the combined treatment of PDI and azole antifungal drugs. Using confocal microscopic analysis, we showed that TBO-mediated PDI could partially remove the extracellular polymeric substance (EPS) of biofilm. Finally, we showed that a combination of PDI with caspofungin could result in the complete killing of biofilms compared to those treated with caspofungin or PDI alone. These results clearly indicate that the combination of PDI and antifungal agents could be a promising treatment against C. albicans infections.
Collapse
Affiliation(s)
- Mu-Ching Huang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Mandy Shen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Yi-Jhen Huang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Hsiao-Chi Lin
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Chin-Tin Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
65
|
Liu X, Ma Z, Zhang J, Yang L. Antifungal Compounds against Candida Infections from Traditional Chinese Medicine. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4614183. [PMID: 29445739 PMCID: PMC5763084 DOI: 10.1155/2017/4614183] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/25/2017] [Accepted: 12/06/2017] [Indexed: 12/22/2022]
Abstract
Infections caused by Candida albicans, often refractory and with high morbidity and mortality, cause a heavy burden on the public health while the current antifungal drugs are limited and are associated with toxicity and resistance. Many plant-derived molecules including compounds isolated from traditional Chinese medicine (TCM) are reported to have antifungal activity through different targets such as cell membrane, cell wall, mitochondria, and virulence factors. Here, we review the recent progress in the anti-Candida compounds from TCM, as well as their antifungal mechanisms. Considering the diverse targets and structures, compounds from TCM might be a potential library for antifungal drug development.
Collapse
Affiliation(s)
- Xin Liu
- Eye Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jingxiao Zhang
- Department of Emergency, The Second Hospital of Jilin University, Changchun 130041, China
| | - Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
66
|
Antileishmanial Efficacy and Pharmacokinetics of DB766-Azole Combinations. Antimicrob Agents Chemother 2017; 62:AAC.01129-17. [PMID: 29061761 DOI: 10.1128/aac.01129-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/18/2017] [Indexed: 01/04/2023] Open
Abstract
Given the limitations of current antileishmanial drugs and the utility of oral combination therapy for other infections, developing an oral combination against visceral leishmaniasis should be a high priority. In vitro combination studies with DB766 and antifungal azoles against intracellular Leishmania donovani showed that posaconazole and ketoconazole, but not fluconazole, enhanced DB766 potency. Pharmacokinetic analysis of DB766-azole combinations in uninfected Swiss Webster mice revealed that DB766 exposure was increased by higher posaconazole and ketoconazole doses, while DB766 decreased ketoconazole exposure. In L. donovani-infected BALB/c mice, DB766-posaconazole combinations given orally for 5 days were more effective than DB766 or posaconazole alone. For example, 81% ± 1% (means ± standard errors) inhibition of liver parasite burden was observed for 37.5 mg/kg of body weight DB766 plus 15 mg/kg posaconazole, while 37.5 mg/kg DB766 and 15 mg/kg posaconazole administered as monotherapy gave 40% ± 5% and 21% ± 3% inhibition, respectively. Combination index (CI) analysis indicated that synergy or moderate synergy was observed in six of nine combined dose groups, while the other three were nearly additive. Liver concentrations of DB766 and posaconazole increased in almost all combination groups compared to monotherapy groups, although many increases were not statistically significant. For DB766-ketoconazole combinations evaluated in this model, two were antagonistic, one displayed synergy, and one was nearly additive. These data indicate that the efficacy of DB766-posaconazole and DB766-ketoconazole combinations in vivo is influenced in part by the pharmacokinetics of the combination, and that the former combination deserves further consideration in developing new treatment strategies against visceral leishmaniasis.
Collapse
|
67
|
Gong Y, Li T, Yu C, Sun S. Candida albicans Heat Shock Proteins and Hsps-Associated Signaling Pathways as Potential Antifungal Targets. Front Cell Infect Microbiol 2017; 7:520. [PMID: 29312897 PMCID: PMC5742142 DOI: 10.3389/fcimb.2017.00520] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/07/2017] [Indexed: 11/28/2022] Open
Abstract
In recent decades, the incidence of invasive fungal infections has increased notably. Candida albicans (C. albicans), a common opportunistic fungal pathogen that dwells on human mucosal surfaces, can cause fungal infections, especially in immunocompromised and high-risk surgical patients. In addition, the wide use of antifungal agents has likely contributed to resistance of C. albicans to traditional antifungal drugs, increasing the difficulty of treatment. Thus, it is urgent to identify novel antifungal drugs to cope with C. albicans infections. Heat shock proteins (Hsps) exist in most organisms and are expressed in response to thermal stress. In C. albicans, Hsps control basic physiological activities or virulence via interaction with a variety of diverse regulators of cellular signaling pathways. Moreover, it has been demonstrated that Hsps confer drug resistance to C. albicans. Many studies have shown that disrupting the normal functions of C. albicans Hsps inhibits fungal growth or reverses the tolerance of C. albicans to traditional antifungal drugs. Here, we review known functions of the diverse Hsp family, Hsp-associated intracellular signaling pathways and potential antifungal targets based on these pathways in C. albicans. We hope this review will aid in revealing potential new roles of C. albicans Hsps in addition to canonical heat stress adaptions and provide more insight into identifying potential novel antifungal targets.
Collapse
Affiliation(s)
- Ying Gong
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Tao Li
- Intensive Care Unit, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| | - Cuixiang Yu
- Respiration Medicine, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| | - Shujuan Sun
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
68
|
Romo JA, Pierce CG, Chaturvedi AK, Lazzell AL, McHardy SF, Saville SP, Lopez-Ribot JL. Development of Anti-Virulence Approaches for Candidiasis via a Novel Series of Small-Molecule Inhibitors of Candida albicans Filamentation. mBio 2017; 8:e01991-17. [PMID: 29208749 PMCID: PMC5717394 DOI: 10.1128/mbio.01991-17] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
Candida albicans remains the main etiologic agent of candidiasis, the most common fungal infection and now the third most frequent infection in U.S. hospitals. The scarcity of antifungal agents and their limited efficacy contribute to the unacceptably high morbidity and mortality rates associated with these infections. The yeast-to-hypha transition represents the main virulence factor associated with the pathogenesis of C. albicans infections. In addition, filamentation is pivotal for robust biofilm development, which represents another major virulence factor for candidiasis and further complicates treatment. Targeting pathogenic mechanisms rather than growth represents an attractive yet clinically unexploited approach in the development of novel antifungal agents. Here, we performed large-scale phenotypic screening assays with 30,000 drug-like small-molecule compounds within ChemBridge's DIVERSet chemical library in order to identify small-molecule inhibitors of C. albicans filamentation, and our efforts led to the identification of a novel series of bioactive compounds with a common biaryl amide core structure. The leading compound of this series, N-[3-(allyloxy)-phenyl]-4-methoxybenzamide, was able to prevent filamentation under all liquid and solid medium conditions tested, suggesting that it impacts a common core component of the cellular machinery that mediates hypha formation under different environmental conditions. In addition to filamentation, this compound also inhibited C. albicans biofilm formation. This leading compound also demonstrated in vivo activity in clinically relevant murine models of invasive and oral candidiasis. Overall, our results indicate that compounds within this series represent promising candidates for the development of novel anti-virulence approaches to combat C. albicans infections.IMPORTANCE Since fungi are eukaryotes, there is a limited number of fungus-specific targets and, as a result, the antifungal arsenal is exceedingly small. Furthermore, the efficacy of antifungal treatment is compromised by toxicity and development of resistance. As a consequence, fungal infections carry high morbidity and mortality rates, and there is an urgent but unmet need for novel antifungal agents. One appealing strategy for antifungal drug development is to target pathogenetic mechanisms associated with infection. In Candida albicans, one of the most common pathogenic fungi, morphogenetic transitions between yeast cells and filamentous hyphae represent a key virulence factor associated with the ability of fungal cells to invade tissues, cause damage, and form biofilms. Here, we describe and characterize a novel small-molecule compound capable of inhibiting C. albicans filamentation both in vitro and in vivo; as such, this compound represents a leading candidate for the development of anti-virulence therapies against candidiasis.
Collapse
Affiliation(s)
- Jesus A Romo
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Christopher G Pierce
- Department of Biology, University of the Incarnate Word, San Antonio, Texas, USA
| | - Ashok K Chaturvedi
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Anna L Lazzell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Stanton F McHardy
- Department of Chemistry and Center for Innovation in Drug Discovery, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Stephen P Saville
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Jose L Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
69
|
Clinical Implications of Candida Biofilms. CURRENT FUNGAL INFECTION REPORTS 2017. [DOI: 10.1007/s12281-017-0302-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
70
|
Synergistic Effects of Tacrolimus and Azoles against Exophiala dermatitidis. Antimicrob Agents Chemother 2017; 61:AAC.00948-17. [PMID: 28923863 DOI: 10.1128/aac.00948-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/10/2017] [Indexed: 11/20/2022] Open
Abstract
In vitro interactions of tacrolimus, a calcineurin inhibitor, and azoles, including itraconazole, voriconazole, and posaconazole, against planktonic cells and biofilms of Exophiala dermatitidis were assessed via a broth microdilution checkerboard technique. A total of 16 clinical isolates were studied. The results revealed favorable synergistic inhibitory activity between tacrolimus and itraconazole, voriconazole, or posaconazole against 68.8%, 87.5%, and 100% of tested strains of planktonic E. dermatitidis, respectively.However, limited synergism was observed against biofilms of E. dermatitidis No antagonism was observed in all combinations.
Collapse
|
71
|
Borba-Santos LP, Reis de Sá LF, Ramos JA, Rodrigues AM, de Camargo ZP, Rozental S, Ferreira-Pereira A. Tacrolimus Increases the Effectiveness of Itraconazole and Fluconazole against Sporothrix spp. Front Microbiol 2017; 8:1759. [PMID: 28966608 PMCID: PMC5605639 DOI: 10.3389/fmicb.2017.01759] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/30/2017] [Indexed: 12/17/2022] Open
Abstract
Calcineurin inhibitors – such as the clinically used drug tacrolimus – are active against important fungal pathogens, particularly when combined with azoles. However, tacrolimus has not been tested against sporotrichosis, an endemic subcutaneous mycosis with worldwide distribution. Here, we evaluated the activity of tacrolimus and cyclosporine A in vitro – as monotherapy and in combination with itraconazole or fluconazole – against yeasts of Sporothrix brasiliensis and S. schenckii, the main sporotrichosis agents in Brazil. We also analyzed the effect of tacrolimus treatment on intracellular neutral lipid levels, which typically increase after azole treatment. Tacrolimus inhibited the growth of yeasts from S. brasiliensis and S. schenckii reference isolates, with minimum inhibitory concentration (MIC) values (required for ≥50% growth inhibition) of 1 and 2 mg/L, respectively. Importantly, the combination of tacrolimus and azoles exhibited high synergy toward reference Sporothrix isolates. Tacrolimus combined with itraconazole significantly increased neutral lipid accumulation in S. brasiliensis, but not in S. schenckii. Clinical isolates of S. brasiliensis and S. schenckii were more sensitive to tacrolimus as monotherapy than feline-borne isolates, however, synergy between tacrolimus and azoles was only observed for feline-borne isolates. Cyclosporine A was effective against S. brasiliensis and S. schenckii as monotherapy (MIC = 1 mg/L), but exhibited no synergy with itraconazole and fluconazole. We conclude that tacrolimus has promising antifungal activity against sporotrichosis agents, and also increases the activity of the current anti-sporotrichosis therapy (itraconazole and fluconazole) in combination assays against S. brasiliensis feline-borne isolates.
Collapse
Affiliation(s)
- Luana P Borba-Santos
- Laboratório de Biologia Celular de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Leandro F Reis de Sá
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Juliene A Ramos
- Instituto Federal de Educação, Ciência e TecnologiaRio de Janeiro, Brazil
| | - Anderson M Rodrigues
- Divisão de Biologia Celular, Departamento de Microbiologia, Universidade de São PauloSão Paulo, Brazil
| | - Zoilo P de Camargo
- Divisão de Biologia Celular, Departamento de Microbiologia, Universidade de São PauloSão Paulo, Brazil
| | - Sonia Rozental
- Laboratório de Biologia Celular de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Antonio Ferreira-Pereira
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
72
|
Cai Z, Ding Z, Hao Y, Ni T, Xie F, Zhao J, Li R, Yu S, Wang T, Chai X, Jin Y, Gao Y, Zhang D, Jiang Y. Design, synthesis, and SAR study of 3-(benzo[d][1,3]dioxol-5-yl)-N-benzylpropanamide as novel potent synergists against fluconazole-resistant Candida albicans. Bioorg Med Chem Lett 2017; 27:4571-4575. [PMID: 28874321 DOI: 10.1016/j.bmcl.2017.08.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 11/28/2022]
Abstract
Based on our previous discovery and SAR study on the lead compounds 7d, 5 and berberine which can significantly enhance the susceptibility of fluconazole against fluconazole-resistant Candida albicans, a series of 3-(benzo[d][1,3]dioxol-5-yl)-N-(substituted benzyl)propanamides were designed, synthesized, and evaluated for their in vitro synergistic activity in combination with fluconazole. The series 2a-f were designed by replacing the amide moiety of the lead compound 7d with retro-amide moiety, and compounds 2a and 2b showed more activity than the lead 7d. Furthermore, introducing biphenyl moiety into series 2d-f afforded series 3a-r, most of which exhibited significantly superior activity to the series 2d-f. Especially, compound 3e, at a concentration of 1.0µg/ml, can enhance the susceptibility of fluconazole against fluconazole-resistant Candida albicans from 128.0µg/ml to 0.125-0.25µg/ml. A clear SAR of the compounds is discussed.
Collapse
Affiliation(s)
- Zhan Cai
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zichao Ding
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China; School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou 350112, China
| | - Tingjunhong Ni
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Fei Xie
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jing Zhao
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou 350112, China
| | - Ran Li
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Shichong Yu
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Ting Wang
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xiaoyun Chai
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yongsheng Jin
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yue Gao
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Dazhi Zhang
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China; School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou 350112, China.
| | - Yuanying Jiang
- Center of New Drug Research, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.
| |
Collapse
|
73
|
Mood AD, Premachandra IDUA, Hiew S, Wang F, Scott KA, Oldenhuis NJ, Liu H, Van Vranken DL. Potent Antifungal Synergy of Phthalazinone and Isoquinolones with Azoles Against Candida albicans. ACS Med Chem Lett 2017; 8:168-173. [PMID: 28197306 DOI: 10.1021/acsmedchemlett.6b00355] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/11/2017] [Indexed: 11/29/2022] Open
Abstract
Four phthalazinones (CIDs 22334057, 22333974, 22334032, 22334012) and one isoquinolone (CID 5224943) were previously shown to be potent enhancers of antifungal activity of fluconazole against Candida albicans. Several even more potent analogues of these compounds were identified, some with EC50 as low as 1 nM, against C. albicans. The compounds exhibited pharmacological synergy (FIC < 0.5) with fluconazole. The compounds were also shown to enhance the antifungal activity of isavuconazole, a recently FDA approved azole antifungal. Isoquinolone 15 and phthalazinone 24 were shown to be active against several resistant clinical isolates of C. albicans.
Collapse
Affiliation(s)
- Aaron D. Mood
- Department
of Chemistry, University of California, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | | | - Stanley Hiew
- Department
of Chemistry, University of California, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Fuqiang Wang
- Department
of Biological Chemistry, University of California, Irvine, California 92697-1700, United States
| | - Kevin A. Scott
- Department
of Chemistry, University of California, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Nathan J. Oldenhuis
- Department
of Chemistry, University of California, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Haoping Liu
- Department
of Biological Chemistry, University of California, Irvine, California 92697-1700, United States
| | - David L. Van Vranken
- Department
of Chemistry, University of California, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| |
Collapse
|
74
|
Abstract
The high incidence and mortality of invasive fungal infections and serious drug resistance have become a global public health issue. The ability of fungal cells to form biofilms is an important reason for the emergence of severe resistance to most clinically available antifungal agents. Targeting fungal biofilm formation by small molecules represents a promising new strategy for the development of novel antifungal agents. This perspective will provide a comprehensive review of fungal biofilm inhibitors. In particular, discovery strategies, chemical structures, antibiofilm/antifungal activities, and structure-activity relationship studies will be discussed. Development of inhibitors to treat biofilm-related resistant fungal infections is a new yet clinically unexploited paradigm, and there is still a long way to go to clinical application. Better understanding of fungal biofilms in combination with systematic drug discovery efforts will pave the way for potential clinical applications.
Collapse
Affiliation(s)
- Shanchao Wu
- School of Pharmacy, Second Military Medical University , 325 Guohe Road, Shanghai 200433, People's Republic of China
| | - Yan Wang
- School of Pharmacy, Second Military Medical University , 325 Guohe Road, Shanghai 200433, People's Republic of China
| | - Na Liu
- School of Pharmacy, Second Military Medical University , 325 Guohe Road, Shanghai 200433, People's Republic of China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University , 325 Guohe Road, Shanghai 200433, People's Republic of China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University , 325 Guohe Road, Shanghai 200433, People's Republic of China
| |
Collapse
|
75
|
Bandara HMHN, Matsubara VH, Samaranayake LP. Future therapies targeted towards eliminating Candida biofilms and associated infections. Expert Rev Anti Infect Ther 2016; 15:299-318. [PMID: 27927053 DOI: 10.1080/14787210.2017.1268530] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Candida species are common human commensals and cause either superficial or invasive opportunistic infections. The biofilm form of candida as opposed to its suspended, planktonic form, is predominantly associated with these infections. Alternative or adjunctive therapies are urgently needed to manage Candida infections as the currently available short arsenal of antifungal drugs has been compromised due to their systemic toxicity, cross-reactivity with other drugs, and above all, by the emergence of drug-resistant Candida species due to irrational drug use. Areas covered: Combination anti-Candida therapies, antifungal lock therapy, denture cleansers, and mouth rinses have all been proposed as alternatives for disrupting candidal biofilms on different substrates. Other suggested approaches for the management of candidiasis include the use of natural compounds, such as probiotics, plants extracts and oils, antifungal quorum sensing molecules, anti-Candida antibodies and vaccines, cytokine therapy, transfer of primed immune cells, photodynamic therapy, and nanoparticles. Expert commentary: The sparsity of currently available antifungals and the plethora of proposed anti-candidal therapies is a distinct indication of the urgent necessity to develop efficacious therapies for candidal infections. Alternative drug delivery approaches, such as probiotics, reviewed here is likely to be a reality in clinical settings in the not too distant future.
Collapse
Affiliation(s)
- H M H N Bandara
- a School of Dentistry , The University of Queensland , Herston , QLD , Australia
| | - V H Matsubara
- b School of Dentistry , University of São Paulo , São Paulo , SP , Brazil.,c Department of Microbiology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , SP , Brazil
| | - L P Samaranayake
- a School of Dentistry , The University of Queensland , Herston , QLD , Australia.,d Faculty of Dentistry , University of Kuwait , Kuwait
| |
Collapse
|
76
|
Beauvericin Potentiates Azole Activity via Inhibition of Multidrug Efflux, Blocks Candida albicans Morphogenesis, and Is Effluxed via Yor1 and Circuitry Controlled by Zcf29. Antimicrob Agents Chemother 2016; 60:7468-7480. [PMID: 27736764 DOI: 10.1128/aac.01959-16] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/04/2016] [Indexed: 01/09/2023] Open
Abstract
Invasive fungal infections are a leading cause of human mortality. Effective treatment is hindered by the rapid emergence of resistance to the limited number of antifungal drugs, demanding new strategies to treat life-threatening fungal infections. Here, we explore a powerful strategy to enhance antifungal efficacy against leading human fungal pathogens by using the natural product beauvericin. We found that beauvericin potentiates the activity of azole antifungals against azole-resistant Candida isolates via inhibition of multidrug efflux and that beauvericin itself is effluxed via Yor1. As observed in Saccharomyces cerevisiae, we determined that beauvericin inhibits TOR signaling in Candida albicans To further characterize beauvericin activity in C. albicans, we leveraged genome sequencing of beauvericin-resistant mutants. Resistance was conferred by mutations in transcription factor genes TAC1, a key regulator of multidrug efflux, and ZCF29, which was uncharacterized. Transcriptional profiling and chromatin immunoprecipitation coupled to microarray analyses revealed that Zcf29 binds to and regulates the expression of multidrug transporter genes. Beyond drug resistance, we also discovered that beauvericin blocks the C. albicans morphogenetic transition from yeast to filamentous growth in response to diverse cues. We found that beauvericin represses the expression of many filament-specific genes, including the transcription factor BRG1 Thus, we illuminate novel circuitry regulating multidrug efflux and establish that simultaneously targeting drug resistance and morphogenesis provides a promising strategy to combat life-threatening fungal infections.
Collapse
|
77
|
Abstract
ABSTRACT
Invasive fungal infections are becoming an increasingly important cause of human mortality and morbidity, particularly for immunocompromised populations. The fungal pathogens
Candida albicans
,
Cryptococcus neoformans
, and
Aspergillus fumigatus
collectively contribute to over 1 million human deaths annually. Hence, the importance of safe and effective antifungal therapeutics for the practice of modern medicine has never been greater. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for drug development remains limited. Only three classes of molecules are currently approved for the treatment of invasive mycoses. The efficacy of these agents is compromised by host toxicity, fungistatic activity, or the emergence of drug resistance in pathogen populations. Here we describe our current arsenal of antifungals and highlight current strategies that are being employed to improve the therapeutic safety and efficacy of these drugs. We discuss state-of-the-art approaches to discover novel chemical matter with antifungal activity and highlight some of the most promising new targets for antifungal drug development. We feature the benefits of combination therapy as a strategy to expand our current repertoire of antifungals and discuss the antifungal combinations that have shown the greatest potential for clinical development. Despite the paucity of new classes of antifungals that have come to market in recent years, it is clear that by leveraging innovative approaches to drug discovery and cultivating collaborations between academia and industry, there is great potential to bolster the antifungal armamentarium.
Collapse
|
78
|
Gu X, Xue W, Yin Y, Liu H, Li S, Sun X. The Hsp90 Co-chaperones Sti1, Aha1, and P23 Regulate Adaptive Responses to Antifungal Azoles. Front Microbiol 2016; 7:1571. [PMID: 27761133 PMCID: PMC5050212 DOI: 10.3389/fmicb.2016.01571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/20/2016] [Indexed: 12/26/2022] Open
Abstract
Heat Shock Protein 90 (Hsp90) is essential for tumor progression in humans and drug resistance in fungi. However, the roles of its many co-chaperones in antifungal resistance are unknown. In this study, by susceptibility test of Neurospora crassa mutants lacking each of 18 Hsp90/Calcineurin system member genes (including 8 Hsp90 co-chaperone genes) to antifungal drugs and other stresses, we demonstrate that the Hsp90 co-chaperones Sti1 (Hop1 in yeast), Aha1, and P23 (Sba1 in yeast) were required for the basal resistance to antifungal azoles and heat stress. Deletion of any of them resulted in hypersensitivity to azoles and heat. Liquid chromatography-mass spectrometry (LC-MS) analysis showed that the toxic sterols eburicol and 14α-methyl-3,6-diol were significantly accumulated in the sti1 and p23 deletion mutants after ketoconazole treatment, which has been shown before to led to cell membrane stress. At the transcriptional level, Aha1, Sti1, and P23 positively regulate responses to ketoconazole stress by erg11 and erg6, key genes in the ergosterol biosynthetic pathway. Aha1, Sti1, and P23 are highly conserved in fungi, and sti1 and p23 deletion also increased the susceptibility to azoles in Fusarium verticillioides. These results indicate that Hsp90-cochaperones Aha1, Sti1, and P23 are critical for the basal azole resistance and could be potential targets for developing new antifungal agents.
Collapse
Affiliation(s)
- Xiaokui Gu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; College of Life Sciences, University of Chinese Academy of SciencesBeijing, China
| | - Wei Xue
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; College of Life Sciences, University of Chinese Academy of SciencesBeijing, China
| | - Yajing Yin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; College of Life Sciences, University of Chinese Academy of SciencesBeijing, China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences Beijing, China
| | - Shaojie Li
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences Beijing, China
| | - Xianyun Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences Beijing, China
| |
Collapse
|
79
|
Kubiça TF, Denardi LB, Azevedo MI, Oliveira V, Severo LC, Santurio JM, Alves SH. Antifungal activities of tacrolimus in combination with antifungal agents against fluconazole-susceptible and fluconazole-resistant Trichosporon asahii isolates. Braz J Infect Dis 2016; 20:539-545. [PMID: 27697432 PMCID: PMC9427583 DOI: 10.1016/j.bjid.2016.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 12/04/2022] Open
Abstract
The antifungal activity of tacrolimus in combination with antifungal agents against different fungal species has been previously reported. Here we report the in vitro interactions between tacrolimus and amphotericin B, fluconazole, itraconazole, and caspofungin against 30 clinical isolates of both fluconazole-susceptible and fluconazole-resistant Trichosporon asahii. For these analyses, we used the broth microdilution method based on the M27-A3 technique and checkerboard microdilution method. Tacrolimus showed no activity against T. asahii strains (minimal inhibitory concentrations, MICs > 64.0 μg mL−1). However, a larger synergistic interaction was observed by the combinations tacrolimus + amphotericin B (96.67%) and tacrolimus + caspofungin (73.33%) against fluconazole-susceptible isolates. Combinations with azole antifungal agents resulted in low rates of synergism for this group (fluconazole + tacrolimus = 40% and itraconazole + tacrolimus = 10%). Antagonistic interactions were not observed. For the fluconazole-resistant T. asahii group, all tested combinations showed indifferent interactions. The synergism showed against fluconazole-susceptible T. asahii isolates suggests that the potential antifungal activity of tacrolimus deserves in vivo experimental investigation, notably, the combination of tacrolimus with amphotericin B or caspofungin.
Collapse
Affiliation(s)
- Thaís Felli Kubiça
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brazil; Universidade Regional Integrada do Alto Uruguai e das Missões (URI), Santiago, RS, Brazil
| | - Laura Bedin Denardi
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brazil
| | - Maria Isabel Azevedo
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Santa Maria, RS, Brazil
| | - Vanessa Oliveira
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Santa Maria, RS, Brazil
| | | | - Janio Morais Santurio
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Santa Maria, RS, Brazil; Universidade Federal de Santa Maria (UFSM), Departamento de Microbiologia e Parasitologia, Santa Maria, RS, Brazil
| | - Sydney Hartz Alves
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brazil; Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Santa Maria, RS, Brazil; Universidade Federal de Santa Maria (UFSM), Departamento de Microbiologia e Parasitologia, Santa Maria, RS, Brazil.
| |
Collapse
|
80
|
Abstract
Candida albicans, the most pervasive fungal pathogen that colonizes humans, forms biofilms that are architecturally complex. They consist of a basal yeast cell polylayer and an upper region of hyphae encapsulated in extracellular matrix. However, biofilms formed in vitro vary as a result of the different conditions employed in models, the methods used to assess biofilm formation, strain differences, and, in a most dramatic fashion, the configuration of the mating type locus (MTL). Therefore, integrating data from different studies can lead to problems of interpretation if such variability is not taken into account. Here we review the conditions and factors that cause biofilm variation, with the goal of engendering awareness that more attention must be paid to the strains employed, the methods used to assess biofilm development, every aspect of the model employed, and the configuration of the MTL locus. We end by posing a set of questions that may be asked in comparing the results of different studies and developing protocols for new ones. This review should engender the notion that not all biofilms are created equal.
Collapse
Affiliation(s)
- David R Soll
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| | - Karla J Daniels
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
81
|
Molecular Insights into the Fungus-Specific Serine/Threonine Protein Phosphatase Z1 in Candida albicans. mBio 2016; 7:mBio.00872-16. [PMID: 27578752 PMCID: PMC4999541 DOI: 10.1128/mbio.00872-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The opportunistic pathogen Candida is one of the most common causes of nosocomial bloodstream infections. Because candidemia is associated with high mortality rates and because the incidences of multidrug-resistant Candida are increasing, efforts to identify novel targets for the development of potent antifungals are warranted. Here, we describe the structure and function of the first member of a family of protein phosphatases that is specific to fungi, protein phosphatase Z1 (PPZ1) from Candida albicans We show that PPZ1 not only is active but also is as susceptible to inhibition by the cyclic peptide inhibitor microcystin-LR as its most similar human homolog, protein phosphatase 1α (PP1α [GLC7 in the yeast Saccharomyces cerevisiae]). Unexpectedly, we also discovered that, despite its 66% sequence identity to PP1α, the catalytic domain of PPZ1 contains novel structural elements that are not present in PP1α. We then used activity and pulldown assays to show that these structural differences block a large subset of PP1/GLC7 regulatory proteins from effectively binding PPZ1, demonstrating that PPZ1 does not compete with GLC7 for its regulatory proteins. Equally important, these unique structural elements provide new pockets suitable for the development of PPZ1-specific inhibitors. Together, these studies not only reveal why PPZ1 does not negatively impact GLC7 activity in vivo but also demonstrate that the family of fungus-specific phosphatases-especially PPZ1 from C. albicans-are highly suitable targets for the development of novel drugs that specifically target C. albicans without cross-reacting with human phosphatases. IMPORTANCE Candida albicans is a medically important human pathogen that is the most common cause of fungal infections in humans. In particular, approximately 46,000 cases of health care-associated candidiasis occur each year in the United States. Because these infections are associated with high mortality rates and because multiple species of Candida are becoming increasingly resistant to antifungals, there are increasing efforts to identify novel targets that are essential for C. albicans virulence. Here we use structural and biochemical approaches to elucidate how a member of a fungus-specific family of enzymes, serine/threonine phosphatase PPZ1, functions in C. albicans We discovered multiple unique features of PPZ1 that explain why it does not cross-react with, and in turn compete for, PP1-specific regulators, a long-standing question in the field. Most importantly, however, these unique features identified PPZ1 as a potential target for the development of novel antifungal therapeutics that will provide new, safe, and potent treatments for candidiasis in humans.
Collapse
|
82
|
How promising are combinatorial drug strategies in combating Candida albicans biofilms? Future Med Chem 2016; 8:1383-5. [PMID: 27463947 DOI: 10.4155/fmc-2016-0127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
83
|
Butts A, Palmer GE, Rogers PD. Antifungal adjuvants: Preserving and extending the antifungal arsenal. Virulence 2016; 8:198-210. [PMID: 27459018 DOI: 10.1080/21505594.2016.1216283] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
As the rates of systemic fungal infections continue to rise and antifungal drug resistance becomes more prevalent, there is an urgent need for new therapeutic options. This issue is exacerbated by the limited number of systemic antifungal drug classes. However, the discovery, development, and approval of novel antifungals is an extensive process that often takes decades. For this reason, there is growing interest and research into the possibility of combining existing therapies with various adjuvants that either enhance activity or overcome existing mechanisms of resistance. Reports of antifungal adjuvants range from plant extracts to repurposed compounds, to synthetic peptides. This approach would potentially prolong the utility of currently approved antifungals and mitigate the ongoing development of resistance.
Collapse
Affiliation(s)
- Arielle Butts
- a Department of Clinical Pharmacy , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Glen E Palmer
- a Department of Clinical Pharmacy , University of Tennessee Health Science Center , Memphis , TN , USA
| | - P David Rogers
- a Department of Clinical Pharmacy , University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
84
|
Jia W, Zhang H, Li C, Li G, Liu X, Wei J. The calcineruin inhibitor cyclosporine a synergistically enhances the susceptibility of Candida albicans biofilms to fluconazole by multiple mechanisms. BMC Microbiol 2016; 16:113. [PMID: 27316338 PMCID: PMC4912705 DOI: 10.1186/s12866-016-0728-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Biofilms produced by Candida albicans (C. albicans) are intrinsically resistant to fungicidal agents, which are a main cause of the pathogenesis of catheter infections. Several lines of evidence have demonstrated that calcineurin inhibitor FK506 or cyclosporine A (CsA) can remarkably enhance the antifungal activity of fluconazole (FLC) against biofilm-producing C. albicans strain infections. The aim of present study is thus to interrogate the mechanism underpinning the synergistic effect of FLC and calcineurin inhibitors. RESULTS Twenty four clinical C. albicans strains isolated from bloodstream showed a distinct capacity of biofilm formation. A combination of calcineurin inhibitor CsA and FLC exhibited a dose-dependent synergistic antifungal effect on the growth and biofilm formation of C. albicans isolates as determined by a XTT assay and fluorescent microscopy assay. The synergistic effect was accompanied with a significantly down-regulated expression of adhesion-related genes ALS3, hypha-related genes HWP1, ABC transporter drug-resistant genes CDR1 and MDR1, and FLC targeting gene, encoding sterol 14alpha-demethylase (ERG11) in clinical C. albicans isolates. Furthermore, an addition of CsA significantly reduced the cellular surface hydrophobicity but increased intracellular calcium concentration as determined by a flow cytometry assay (p < 0.05). CONCLUSION The results presented in this report demonstrated that the synergistic effect of CsA and FLC on inhibited C. albicans biofilm formation and enhanced susceptibility to FLC was in part through a mechanism involved in suppressing the expression of biofilm related and drug-resistant genes, and reducing cellular surface hydrophobicity, as well as evoking intracellular calcium concentration.
Collapse
Affiliation(s)
- Wei Jia
- Ningxia Key laboratory of Clinical and Pathogenic Microbiology, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.,Center of Laboratory Medicine, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Haiyun Zhang
- The First People's Hospital of Mudanjiang City, Mudanjiang, Helongjiang, 157011, China
| | - Caiyun Li
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Gang Li
- Ningxia Key laboratory of Clinical and Pathogenic Microbiology, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.,Center of Laboratory Medicine, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Xiaoming Liu
- Ningxia Key laboratory of Clinical and Pathogenic Microbiology, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| | - Jun Wei
- Ningxia Key laboratory of Clinical and Pathogenic Microbiology, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China. .,Center of Laboratory Medicine, the General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
85
|
Spitzer M, Robbins N, Wright GD. Combinatorial strategies for combating invasive fungal infections. Virulence 2016; 8:169-185. [PMID: 27268286 DOI: 10.1080/21505594.2016.1196300] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Invasive fungal infections are an important cause of human mortality and morbidity, particularly for immunocompromised populations. However, there remains a paucity of antifungal drug treatments available to combat these fungal pathogens. Further, antifungal compounds are plagued with problems such as host toxicity, fungistatic activity, and the emergence of drug resistance in pathogen populations. A promising therapeutic strategy to increase drug effectiveness and mitigate the emergence of drug resistance is through the use of combination drug therapy. In this review we describe the current arsenal of antifungals in medicine and elaborate on the benefits of combination therapy to expand our current antifungal drug repertoire. We examine those antifungal combinations that have shown potential against fungal pathogens and discuss strategies being employed to discover novel combination therapeutics, in particular combining antifungal agents with non-antifungal bioactive compounds. The findings summarized in this review highlight the promise of combinatorial strategies in combatting invasive mycoses.
Collapse
Affiliation(s)
- Michaela Spitzer
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| | - Nicole Robbins
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| | - Gerard D Wright
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| |
Collapse
|
86
|
Abstract
Fungal biofilms have become an increasingly important clinical problem. The widespread use of antibiotics, frequent use of indwelling medical devices, and a trend toward increased patient immunosuppression have resulted in a creation of opportunity for clinically important yeasts and molds to form biofilms. This review will discuss the diversity and importance of fungal biofilms in the context of clinical medicine, provide novel insights into the clinical management of fungal biofilm infection, present evidence why these structures are recalcitrant to antifungal therapy, and discuss how our knowledge and understanding may lead to novel therapeutic intervention.
Collapse
|
87
|
Abstract
Fungal infections constitute a major threat to an escalating number of critically ill patients. Fungi are eukaryotic organisms and, as such, there is a limited armamentarium of antifungal drugs, which leads to high mortality rates. Moreover, fungal infections are often associated with the formation of biofilms, which contribute to virulence and further complicate treatment due to the high level of antifungal drug resistance displayed by sessile cells within these microbial communities. Thus, the treatment of fungal infections associated with a biofilm etiology represents a formidable and unmet clinical challenge. The increasing importance and awareness of fungal biofilms is reflected by the fact that this is now an area of very active research. Studies in the last decade have provided important insights into fungal biofilm biology, physiology, and pathology, as well as into the molecular basis of biofilm resistance. Here we discuss how this accumulated knowledge may inform the development of new antibiofilm strategies and therapeutics that are urgently needed.
Collapse
|
88
|
Abstract
During infection, fungi frequently transition to a biofilm lifestyle, proliferating as communities of surface-adherent aggregates of cells. Phenotypically, cells in a biofilm are distinct from free-floating cells. Their high tolerance of antifungals and ability to withstand host defenses are two characteristics that foster resilience. Biofilm infections are particularly difficult to eradicate, and most available antifungals have minimal activity. Therefore, the discovery of novel compounds and innovative strategies to treat fungal biofilms is of great interest. Although many fungi have been observed to form biofilms, the most well-studied is Candida albicans. Animal models have been developed to simulate common Candida device-associated infections, including those involving vascular catheters, dentures, urinary catheters, and subcutaneous implants. Models have also reproduced the most common mucosal biofilm infections: oropharyngeal and vaginal candidiasis. These models incorporate the anatomical site, immune components, and fluid dynamics of clinical niches and have been instrumental in the study of drug resistance and investigation of novel therapies. This chapter describes the significance of fungal biofilm infections, the animal models developed for biofilm study, and how these models have contributed to the development of new strategies for the eradication of fungal biofilm infections.
Collapse
|
89
|
Haque F, Alfatah M, Ganesan K, Bhattacharyya MS. Inhibitory Effect of Sophorolipid on Candida albicans Biofilm Formation and Hyphal Growth. Sci Rep 2016; 6:23575. [PMID: 27030404 PMCID: PMC4876995 DOI: 10.1038/srep23575] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 03/09/2016] [Indexed: 12/18/2022] Open
Abstract
Candida albicans causes superficial and life-threatening systemic infections.
These are difficult to treat often due to drug resistance, particularly because
C. albicans biofilms are inherently resistant to most antifungals.
Sophorolipid (SL), a glycolipid biosurfactant, has been shown to have antimicrobial
and anticancer properties. In this study, we investigated the effect of SL on C.
albicans biofilm formation and preformed biofilms. SL was found to inhibit
C. albicans biofilm formation as well as reduce the viability of
preformed biofilms. Moreover, SL, when used along with amphotericin B (AmB) or
fluconazole (FLZ), was found to act synergistically against biofilm formation and
preformed biofilms. Effect of SL on C. albicans biofilm formation was further
visualized by scanning electron microscopy (SEM) and confocal laser scanning
microscopy (CLSM), which revealed absence of hyphae, typical biofilm architecture
and alteration in the morphology of biofilm cells. We also found that SL
downregulates the expression of hypha specific genes HWP1, ALS1,
ALS3, ECE1 and SAP4, which possibly explains the inhibitory
effect of SL on hyphae and biofilm formation.
Collapse
Affiliation(s)
- Farazul Haque
- Biocatalysis and Fermentation Science Laboratory, Biochemical Engineering Research &Process Development Center (BERPDC), CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh-160 036, India
| | - Md Alfatah
- Yeast Molecular Biology Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh-160 036, India
| | - K Ganesan
- Yeast Molecular Biology Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh-160 036, India
| | - Mani Shankar Bhattacharyya
- Biocatalysis and Fermentation Science Laboratory, Biochemical Engineering Research &Process Development Center (BERPDC), CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh-160 036, India
| |
Collapse
|
90
|
Cui J, Ren B, Tong Y, Dai H, Zhang L. Synergistic combinations of antifungals and anti-virulence agents to fight against Candida albicans. Virulence 2016; 6:362-71. [PMID: 26048362 DOI: 10.1080/21505594.2015.1039885] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Candida albicans, one of the pathogenic Candida species, causes high mortality rate in immunocompromised and high-risk surgical patients. In the last decade, only one new class of antifungal drug echinocandin was applied. The increased therapy failures, such as the one caused by multi-drug resistance, demand innovative strategies for new effective antifungal drugs. Synergistic combinations of antifungals and anti-virulence agents highlight the pragmatic strategy to reduce the development of drug resistant and potentially repurpose known antifungals, which bypass the costly and time-consuming pipeline of new drug development. Anti-virulence and synergistic combination provide new options for antifungal drug discovery by counteracting the difficulty or failure of traditional therapy for fungal infections.
Collapse
Affiliation(s)
- Jinhui Cui
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology; Institute of Microbiology; Chinese Academy of Sciences ; Beijing , China
| | | | | | | | | |
Collapse
|
91
|
Prasad R, Shah AH, Rawal MK. Antifungals: Mechanism of Action and Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 892:327-349. [PMID: 26721281 DOI: 10.1007/978-3-319-25304-6_14] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There are currently few antifungals in use which show efficacy against fungal diseases. These antifungals mostly target specific components of fungal plasma membrane or its biosynthetic pathways. However, more recent class of antifungals in use is echinocandins which target the fungal cell wall components. The availability of mostly fungistatic antifungals in clinical use, often led to the development of tolerance to these very drugs by the pathogenic fungal species. Thus, the development of clinical multidrug resistance (MDR) leads to higher tolerance to drugs and its emergence is helped by multiple mechanisms. MDR is indeed a multifactorial phenomenon wherein a resistant organism possesses several mechanisms which contribute to display reduced susceptibility to not only single drug in use but also show collateral resistance to several drugs. Considering the limited availability of antifungals in use and the emergence of MDR in fungal infections, there is a continuous need for the development of novel broad spectrum antifungal drugs with better efficacy. Here, we briefly present an overview of the current understanding of the antifungal drugs in use, their mechanism of action and the emerging possible novel antifungal drugs with great promise.
Collapse
Affiliation(s)
- Rajendra Prasad
- Membrane Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India. .,AMITY Institute of Integrative Sciences and Health (AIISH), Amity University Haryana, Manesar, Gurgaon, Haryana, India.
| | - Abdul Haseeb Shah
- Membrane Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Manpreet Kaur Rawal
- Membrane Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
92
|
Albataineh MT, Kadosh D. Regulatory roles of phosphorylation in model and pathogenic fungi. Med Mycol 2015; 54:333-52. [PMID: 26705834 PMCID: PMC4818690 DOI: 10.1093/mmy/myv098] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/01/2015] [Indexed: 12/25/2022] Open
Abstract
Over the past 20 years, considerable advances have been made toward our understanding
of how post-translational modifications affect a wide variety of biological
processes, including morphology and virulence, in medically important fungi.
Phosphorylation stands out as a key molecular switch and regulatory modification that
plays a critical role in controlling these processes. In this article, we first
provide a comprehensive and up-to-date overview of the regulatory roles that both
Ser/Thr and non-Ser/Thr kinases and phosphatases play in model and pathogenic fungi.
Next, we discuss the impact of current global approaches that are being used to
define the complete set of phosphorylation targets (phosphoproteome) in medically
important fungi. Finally, we provide new insights and perspectives into the potential
use of key regulatory kinases and phosphatases as targets for the development of
novel and more effective antifungal strategies.
Collapse
Affiliation(s)
- Mohammad T Albataineh
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - David Kadosh
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| |
Collapse
|
93
|
Soberón JR, Lizarraga EF, Sgariglia MA, Carrasco Juárez MB, Sampietro DA, Ben Altabef A, Catalán CAN, Vattuone MA. Antifungal activity of 4-hydroxy-3-(3-methyl-2-butenyl)acetophenone against Candida albicans: evidence for the antifungal mode of action. Antonie van Leeuwenhoek 2015; 108:1047-57. [PMID: 26342699 DOI: 10.1007/s10482-015-0559-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/13/2015] [Indexed: 10/23/2022]
Abstract
The main secondary metabolite of Senecio nutans is 4-hydroxy-3-(3-methyl-2-butenyl)acetophenone (4HMBA). The antifungal activity of this compound and three derivatives was assessed using Candida albicans. 4HMBA exhibited the highest antifungal activity among the assayed compounds. The Fractional Inhibitory Concentration (FIC = 0.133) indicated a synergistic fungicidal effect of 4HMBA (5 mg L(-1)) and fluconazole (FLU) (0.5 mg L(-1)) against the C. albicans reference strain (ATCC 10231). Microscopy showed that 4HMBA inhibits filamentation and reduces cell wall thickness. Our findings suggest that 4HMBA is an interesting compound to diminish resistance to commercial fungistatic drugs such as fluconazole.
Collapse
Affiliation(s)
- José R Soberón
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina. .,Laboratorio de Biología de Agentes Bioactivos y Fitopatógenos (LABIFITO), Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina.
| | - Emilio F Lizarraga
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina.,Instituto de Química del Noroeste Argentino (INQUINOA), CONICET, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina
| | - Melina A Sgariglia
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Laboratorio de Biología de Agentes Bioactivos y Fitopatógenos (LABIFITO), Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina
| | - María B Carrasco Juárez
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Laboratorio de Biología de Agentes Bioactivos y Fitopatógenos (LABIFITO), Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina
| | - Diego A Sampietro
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Laboratorio de Biología de Agentes Bioactivos y Fitopatógenos (LABIFITO), Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina
| | - Aida Ben Altabef
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina.,Instituto de Química del Noroeste Argentino (INQUINOA), CONICET, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina
| | - César A N Catalán
- Universidad Nacional de Tucumán, Ayacucho 491 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina.,Instituto de Química del Noroeste Argentino (INQUINOA), CONICET, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina
| | - Marta A Vattuone
- Laboratorio de Biología de Agentes Bioactivos y Fitopatógenos (LABIFITO), Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471 (T4000INI), San Miguel De Tucumán, Tucumán, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Rivadavia 1917 (C1033AAJ), Ciudad Autónoma De Buenos Aires, Argentina
| |
Collapse
|
94
|
In vitro interactions of antifungal agents and tacrolimus against Aspergillus biofilms. Antimicrob Agents Chemother 2015; 59:7097-9. [PMID: 26303797 DOI: 10.1128/aac.01510-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/16/2015] [Indexed: 12/16/2022] Open
Abstract
Aspergillus biofilms were prepared from Aspergillus fumigatus, Aspergillus flavus, and Aspergillus terreus via a 96-well plate-based method, and the combined antifungal activity of tacrolimus with azoles or amphotericin B against Aspergillus biofilms was investigated via a broth microdilution checkerboard technique system. Our results suggest that combinations of tacrolimus with voriconazole or amphotericin B have synergistic inhibitory activity against Aspergillus biofilms. However, combinations of tacrolimus with itraconazole or posaconazole exhibit no synergistic or antagonistic effects.
Collapse
|
95
|
Pierce CG, Chaturvedi AK, Lazzell AL, Powell AT, Saville SP, McHardy SF, Lopez-Ribot JL. A Novel Small Molecule Inhibitor of Candida albicans Biofilm Formation, Filamentation and Virulence with Low Potential for the Development of Resistance. NPJ Biofilms Microbiomes 2015; 1. [PMID: 26691764 PMCID: PMC4681527 DOI: 10.1038/npjbiofilms.2015.12] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND/OBJECTIVES Candida albicans is the principal causative agent of candidiasis, the most common fungal infection in humans. Candidiasis represents the third-to-fourth most frequent nosocomial infection worldwide, as this normal commensal of humans causes opportunistic infections in an expanding population of immune- and medically-compromised patients. These infections are frequently associated with biofilm formation, which complicates treatment and contributes to unacceptably high mortality rates. METHODS To address the pressing need for new antifungals we have performed a high content screen of 20,000 small molecules in a chemical library (NOVACore™) to identify compounds that inhibit C. albicans biofilm formation, and conducted a series of follow-up studies to examine the in vitro and in vivo activity of the identified compounds. RESULTS The screen identified a novel series of diazaspiro-decane structural analogs which were largely represented among the bioactive compounds. Characterization of the leading compound from this series indicated that it inhibits processes associated with C. albicans virulence, most notably biofilm formation and filamentation, without having an effect on overall growth or eliciting resistance. This compound demonstrated in vivo activity in clinically-relevant murine models of both invasive and oral candidiasis and as such represents a promising lead for antifungal drug development. Furthermore, these results provide proof of concept for the implementation of anti-virulence approaches against C. albicans and other fungal infections that would be less likely to foster the emergence of resistance.
Collapse
Affiliation(s)
- Christopher G Pierce
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Ashok K Chaturvedi
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Anna L Lazzell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Alexander T Powell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Stephen P Saville
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Stanton F McHardy
- Department of Chemistry and Center for Innovation in Drug Discovery, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Jose L Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| |
Collapse
|
96
|
Anju S, Kumar NS, Krishnakumar B, Kumar BSD. Synergistic combination of violacein and azoles that leads to enhanced killing of major human pathogenic dermatophytic fungi Trichophyton rubrum. Front Cell Infect Microbiol 2015; 5:57. [PMID: 26322275 PMCID: PMC4531294 DOI: 10.3389/fcimb.2015.00057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/21/2015] [Indexed: 11/15/2022] Open
Abstract
Superficial mycoses caused by dermatophytic fungi such as Trichophyton rubrum represent the most common type of worldwide human infection affecting various keratinized tissues in our body such as the skin, hair, and nails, etc. The dermatophytic infection is a significant public health threat due to its persistent nature and high recurrence rates, and thus alternative treatments to cure this fungal infection are urgently required. The present study mainly focused on the synergistic activity of violacein with four azole drugs (ketoconazole, fluconazole, clotrimazole, and itraconazole) against T. rubrum. The synergistic antifungal activities of violacein and azoles were measured by checkerboard microdilution and time-kill assays. In our study, combinations of violacein and azoles predominantly recorded synergistic effect (FIC index < 0.5). Significant synergistic value was recorded by the combination of violacein and clotrimazole. Time-kill assay by the combination of MIC concentration of violacein and azoles recorded that the growth of the T. rubrum was significantly arrested after 4–12 h of treatment. The combination of violacein and azoles leads to the enhanced inhibition of mycelial growth and conidial germination. Moreover combination enhanced the rate of release of intracellular materials. Morphological changes by SEM analysis were also prominent with the combination. A normal human cell line [Foreskin (FS) normal fibroblast] was used to check the cytotoxicity of violacein. Interestingly violacein recorded no cytotoxicity up to 100 μg/ml. The in vitro synergistic effect of violacein and azoles against clinically relevant fungi, T. rubrum, is reported here for the first time. Finally, our findings support the potential use of the violacein as an antifungal agent especially against dermatophytic fungi T. rubrum.
Collapse
Affiliation(s)
- S Anju
- Environmental Technology, CSIR-National Institute for Interdisciplinary Science and Technology Thiruvananthapuram, India
| | - Nishanth S Kumar
- Agroprocessing and Natural Products Division, CSIR-National Institute for Interdisciplinary Science and Technology Thiruvananthapuram, India
| | - B Krishnakumar
- Environmental Technology, CSIR-National Institute for Interdisciplinary Science and Technology Thiruvananthapuram, India
| | - B S Dileep Kumar
- Agroprocessing and Natural Products Division, CSIR-National Institute for Interdisciplinary Science and Technology Thiruvananthapuram, India
| |
Collapse
|
97
|
Potential Targets for Antifungal Drug Discovery Based on Growth and Virulence in Candida albicans. Antimicrob Agents Chemother 2015. [PMID: 26195510 DOI: 10.1128/aac.00726-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fungal infections, especially infections caused by Candida albicans, remain a challenging problem in clinical settings. Despite the development of more-effective antifungal drugs, their application is limited for various reasons. Thus, alternative treatments with drugs aimed at novel targets in C. albicans are needed. Knowledge of growth and virulence in fungal cells is essential not only to understand their pathogenic mechanisms but also to identify potential antifungal targets. This article reviews the current knowledge of the mechanisms of growth and virulence in C. albicans and examines potential targets for the development of new antifungal drugs.
Collapse
|
98
|
De Cremer K, Staes I, Delattin N, Cammue BPA, Thevissen K, De Brucker K. Combinatorial drug approaches to tackleCandida albicansbiofilms. Expert Rev Anti Infect Ther 2015; 13:973-84. [DOI: 10.1586/14787210.2015.1056162] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
99
|
Samber N, Khan A, Varma A, Manzoor N. Synergistic anti-candidal activity and mode of action of Mentha piperita essential oil and its major components. PHARMACEUTICAL BIOLOGY 2015; 53:1496-1504. [PMID: 25853964 DOI: 10.3109/13880209.2014.989623] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Mentha piperita L. (Lamiaceae) has been used in folk medicine since antiquity. Its essential oil (mint EO) and major bioactive components have antimicrobial properties but their mechanism of action is still not clear. OBJECTIVE The present work aims to elucidate M. piperita's anti-Candida activity and mode of action. MATERIALS AND METHODS Chemical constituents of mint EO were identified by GC-MS by injecting 0.1 ml sample in a splitless mode. MIC was determined by the broth dilution method. Synergy with fluconazole (FLC) was evaluated by checkerboard assay and FICI. Mid log phase cells harvested from YPD media were used for proton extrusion measurement and the rate of glucose-induced H(+) efflux gives PM-ATPase activity. Cell membrane integrity was estimated by total ergosterol content and scanning microscopy at respective MIC and sub-MIC values. In vitro hemolytic activity was performed to rule out possible cytotoxicity of the test compounds. RESULTS The MIC value of mint EO, carvone, menthol, and menthone was 225, 248, 500, and 4200 µg/ml, respectively. At their respective MICs, these compounds showed 47, 42, 35, and 29% decrease in PM-ATPase activity besides showing synergy with FLC. In case of FLC-resistant strains, the decrease in H(+) efflux was by 52, 48, 32, and 30%, a trend similar to the susceptible cases. Exposed Candida cells showed a 100% decrease in the ergosterol content, cell membrane breakage, and alterations in morphology. DISCUSSION AND CONCLUSION Our studies suggest that mint EO and its lead compounds exert antifungal activity by reducing ergosterol levels, inhibiting PM-ATPase leading to intracellular acidification, and ultimately cell death. Our results suggest that mint EO and its constituents are potential antifungal agents and need to be further investigated.
Collapse
Affiliation(s)
- Neha Samber
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia , New Delhi , India and
| | | | | | | |
Collapse
|
100
|
Cordeiro RDA, Teixeira CE, Brilhante RS, Castelo-Branco DS, Alencar LP, de Oliveira JS, Monteiro AJ, Bandeira TJ, Sidrim JJ, Moreira JLB, Rocha MF. Exogenous tyrosol inhibits planktonic cells and biofilms of Candida species and enhances their susceptibility to antifungals. FEMS Yeast Res 2015; 15:fov012. [DOI: 10.1093/femsyr/fov012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2015] [Indexed: 12/29/2022] Open
|