1
|
Mohamed MA, Awadalla MKA, Mohamed MS, Elsaman T, Eltayib EM. Repurposing FDA-Approved Drugs for Eumycetoma Treatment: Homology Modeling and Computational Screening of CYP51 Inhibitors. Int J Mol Sci 2025; 26:315. [PMID: 39796172 PMCID: PMC11720416 DOI: 10.3390/ijms26010315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Eumycetoma, a chronic fungal infection caused by Madurella mycetomatis, is a neglected tropical disease characterized by tumor-like growths that can lead to permanent disability and deformities if untreated. Predominantly affecting regions in Africa, South America, and Asia, it imposes significant physical, social, and economic burdens. Current treatments, including antifungal drugs like itraconazole, often show variable efficacy, with severe cases necessitating surgical intervention or amputation. Drug discovery for eumycetoma faces challenges due to limited understanding of the disease's molecular mechanisms and the lack of 3D structures for key targets such as Madurella mycetomatis CYP51, a well-known target for azoles' antifungal agents. To address these challenges, this study employed computational approaches, including homology modeling, virtual screening, free energy calculations, and molecular dynamics simulations, to repurpose FDA-approved drugs as potential treatments for eumycetoma targeting Madurella mycetomatis CYP51. To this end, a library of 2619 FDA-approved drugs was screened, identifying three promising candidates: montelukast, vilanterol, and lidoflazine. These compounds demonstrated favorable binding affinities, strong interactions with critical residues of the homology model of Madurella mycetomatis CYP51, and stability in molecular dynamics simulations, offering potential for further investigation as effective therapeutic options for eumycetoma.
Collapse
Affiliation(s)
- Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | | | - Malik Suliman Mohamed
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia; (M.S.M.); (E.M.E.)
| | - Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Eyman Mohamed Eltayib
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia; (M.S.M.); (E.M.E.)
| |
Collapse
|
2
|
Wu Y, Xi Z, Liu F, Hu W, Feng H, Zhang Q. A deep semantic network-based image segmentation of soybean rust pathogens. FRONTIERS IN PLANT SCIENCE 2024; 15:1340584. [PMID: 38601300 PMCID: PMC11004377 DOI: 10.3389/fpls.2024.1340584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Introduction Asian soybean rust is a highly aggressive leaf-based disease triggered by the obligate biotrophic fungus Phakopsora pachyrhizi which can cause up to 80% yield loss in soybean. The precise image segmentation of fungus can characterize fungal phenotype transitions during growth and help to discover new medicines and agricultural biocides using large-scale phenotypic screens. Methods The improved Mask R-CNN method is proposed to accomplish the segmentation of densely distributed, overlapping and intersecting microimages. First, Res2net is utilized to layer the residual connections in a single residual block to replace the backbone of the original Mask R-CNN, which is then combined with FPG to enhance the feature extraction capability of the network model. Secondly, the loss function is optimized and the CIoU loss function is adopted as the loss function for boundary box regression prediction, which accelerates the convergence speed of the model and meets the accurate classification of high-density spore images. Results The experimental results show that the mAP for detection and segmentation, accuracy of the improved algorithm is improved by 6.4%, 12.3% and 2.2% respectively over the original Mask R-CNN algorithm. Discussion This method is more suitable for the segmentation of fungi images and provide an effective tool for large-scale phenotypic screens of plant fungal pathogens.
Collapse
Affiliation(s)
- Yalin Wu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Zhuobin Xi
- Mechanical Electrical Engineering School, Beijing Information Science & Technology University, Beijing, China
| | - Fen Liu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Weiming Hu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Hongjuan Feng
- Zhongzhen Kejian (ShenZhen) Holdings Co., Ltd, Shenzhen, China
| | - Qinjian Zhang
- Mechanical Electrical Engineering School, Beijing Information Science & Technology University, Beijing, China
| |
Collapse
|
3
|
de Oliveira AA, Carmo Silva LD, Neves BJ, Fiaia Costa VA, Muratov EN, Andrade CH, de Almeida Soares CM, Alves VM, Pereira M. Cheminformatics-driven discovery of hit compounds against Paracoccidioides spp. Future Med Chem 2023; 15:1553-1567. [PMID: 37727967 DOI: 10.4155/fmc-2022-0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Aims: The development of safe and effective therapies for treating paracoccidioidomycosis using computational strategies were employed to discover anti-Paracoccidioides compounds. Materials & methods: We 1) collected, curated and integrated the largest library of compounds tested against Paracoccidioides spp.; 2) employed a similarity search to virtually screen the ChemBridge database and select nine compounds for experimental evaluation; 3) performed an experimental evaluation to determine the minimum inhibitory concentration and minimum fungicidal concentration as well as cytotoxicity; and 4) employed computational tools to identify potential targets for the most active compounds. Seven compounds presented activity against Paracoccidioides spp. Conclusion: These compounds are new hits with a predicted mechanisms of action, making them potentially attractive to develop new compounds.
Collapse
Affiliation(s)
- Amanda Alves de Oliveira
- Institute of Tropical Pathology & Public Health, Federal University of Goiás, Goiânia, 74690-900, Brazil
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| | - Lívia do Carmo Silva
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| | - Bruno Junior Neves
- Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | | | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology & Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, 58051-900, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling & Design, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | | | - Vinicius M Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology & Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Laboratory for Molecular Modeling & Design, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | - Maristela Pereira
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| |
Collapse
|
4
|
Awada B, Chahine DA, Derbaj G, Khalek PA, Awad MK, Fayad AA. Antimicrobial Natural Products Derived from Microorganisms Inhabiting the MENA Region. Nat Prod Commun 2023. [DOI: 10.1177/1934578x231154989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Objective/Background Natural products (NPs) derived from microorganisms are the basis of a plethora of clinically utilized medications, namely, antimicrobial remedies. Although these secondary metabolites have been extensively explored all over the planet, they remain understudied in the Middle East and North Africa (MENA) region. Methods A literature search was conducted to first find NPs that were isolated from environmental fungi and bacteria that inhabit the soils and seawater of the MENA region. Then, purified molecules with biological activity against pathogenic bacteria, biofilms, fungi, and parasites were described in terms of structure, function, and location. Moreover, the methods that could be used to ameliorate the discovery of novel NPs from this region were investigated. Results A multitude of antimicrobial molecules from various chemical classes were found to be derived from the environmental microbes of MENA. Although many were rediscovered, some represented novel structural scaffolds for novel families of antimicrobial agents. Additionally, the geographical distribution showed a high number of these NPs were unraveled in a restricted area leaving much of MENA untapped. Furthermore, as relatively traditional and low-efficiency methods were typically used in the discovery process, advanced high-throughput techniques were suggested to enhance this practice at the regional level. Conclusion MENA represents a fairly unexploited region where antimicrobial drug discovery could be performed comprehensively through the concomitant exploration of untouched geographical locations and advanced molecular techniques.
Collapse
Affiliation(s)
- Bassel Awada
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Drug Discovery, American University of Beirut, Beirut, Lebanon
| | - Dany Abi Chahine
- Center for Drug Discovery, American University of Beirut, Beirut, Lebanon
- Laboratory of Biodiversity and Functional Genomics, UR EGP, Faculty of Science, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
| | - Ghada Derbaj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Drug Discovery, American University of Beirut, Beirut, Lebanon
| | - Pascal Abdel Khalek
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Drug Discovery, American University of Beirut, Beirut, Lebanon
| | - Mireille Kallassy Awad
- Laboratory of Biodiversity and Functional Genomics, UR EGP, Faculty of Science, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
| | - Antoine Abou Fayad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Drug Discovery, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
5
|
Aaghaz S, Sharma K, Maurya IK, Rudramurthy SM, Singh S, Kumar V, Tikoo K, Jain R. Anticryptococcal activity and mechanistic studies of short amphipathic peptides. Arch Pharm (Weinheim) 2023; 356:e2200576. [PMID: 36592413 DOI: 10.1002/ardp.202200576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 01/03/2023]
Abstract
Cryptococcus neoformans, an opportunistic fungal pathogen, causes cryptococcosis in immunocompromised persons. A series of modified L-histidines-containing peptides are synthesized that exhibit promising activity against C. neoformans. Analog 11d [L-His(2-adamantyl)-L-Trp-L-His(2-phenyl)-OMe] produced potency with an IC50 of 3.02 µg/ml (MIC = 5.49 µg/ml). This peptide is noncytotoxic and nonhaemolytic at the MIC and displays synergistic effects with amphotericin B at subinhibitory concentration. Mechanistic investigation of 11d using microscopic tools indicates cell wall and membrane disruption of C. neoformans, while flow cytometric analysis confirms cell death by apoptosis. This study indicates that 11d exhibits antifungal potential and acts via the rapid onset of action.
Collapse
Affiliation(s)
- Shams Aaghaz
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Komal Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Indresh K Maurya
- Center for Infectious Diseases, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, India
| | - Shreya Singh
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, India
| | - Vinod Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India.,Center for Infectious Diseases, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| |
Collapse
|
6
|
Macias-Paz IU, Pérez-Hernández S, Tavera-Tapia A, Luna-Arias JP, Guerra-Cárdenas JE, Reyna-Beltrán E. Candida albicans the main opportunistic pathogenic fungus in humans. Rev Argent Microbiol 2022:S0325-7541(22)00084-0. [PMID: 36411138 DOI: 10.1016/j.ram.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/03/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
|
7
|
Xie F, Hao Y, Liu J, Bao J, Ni T, Liu Y, Chi X, Wang T, Yu S, Jin Y, Li L, Zhang D, Yan L. Discovery of Novel Thiosemicarbazides Containing 1,3,5-Triazines Derivatives as Potential Synergists against Fluconazole-Resistant Candida albicans. Pharmaceutics 2022; 14:2334. [PMID: 36365153 PMCID: PMC9693882 DOI: 10.3390/pharmaceutics14112334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2023] Open
Abstract
The clinical prevalence of antifungal drug resistance has been increasing over recent years, resulting in the failure of treatments. In an attempt to overcome this critical problem, we sought novel synergistic enhancers to restore the effectiveness of fluconazole against resistant Candida albicans. Based on the structural optimization of hit compound 8 from our in-house library, a series of novel 1,3,5-triazines derivatives was designed, synthesized, and biologically evaluated for synergistic activity in combination with fluconazole. Among them, compounds 10a-o, which contain thiosemicarbazides side chains, exhibited excellent in vitro synergistic antifungal potency (MIC80 = 0.125-2.0 μg/mL, FICI range from 0.127 to 0.25). Interestingly, compound 10l exhibited moderate C. albicans activity as monotherapy with an MIC80 value of 4.0 μg/mL, and also on several Cryptococcus strains (MIC80 ranging from ≤ 0.125-0.5 μg/mL) and C. glabrata (MIC80 ≤ 0.125 μg/mL). These effects were fungal-selective, with much lower levels of cytotoxicity towards human umbilical vein endothelial cells. Here, we report a series of thiosemicarbazides containing 1,3,5-triazines derivatives as potent synergists with fluconazole, and have preliminarily validated compound 10l as a promising antifungal lead for further investigation.
Collapse
Affiliation(s)
- Fei Xie
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Jiacun Liu
- Center of New Drug Research, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Junhe Bao
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Yu Liu
- Center of New Drug Research, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Xiaochen Chi
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang 110016, China
| | - Ting Wang
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Shichong Yu
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Yongsheng Jin
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Liping Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Dazhi Zhang
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Lan Yan
- Center of New Drug Research, School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
8
|
Hunting for Novel Routes in Anticancer Drug Discovery: Peptides against Sam-Sam Interactions. Int J Mol Sci 2022; 23:ijms231810397. [PMID: 36142306 PMCID: PMC9499636 DOI: 10.3390/ijms231810397] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 01/10/2023] Open
Abstract
Among the diverse protein binding modules, Sam (Sterile alpha motif) domains attract attention due to their versatility. They are present in different organisms and play many functions in physiological and pathological processes by binding multiple partners. The EphA2 receptor contains a Sam domain at the C-terminus (EphA2-Sam) that is able to engage protein regulators of receptor stability (including the lipid phosphatase Ship2 and the adaptor Odin). Ship2 and Odin are recruited by EphA2-Sam through heterotypic Sam-Sam interactions. Ship2 decreases EphA2 endocytosis and consequent degradation, producing chiefly pro-oncogenic outcomes in a cellular milieu. Odin, through its Sam domains, contributes to receptor stability by possibly interfering with ubiquitination. As EphA2 is upregulated in many types of tumors, peptide inhibitors of Sam-Sam interactions by hindering receptor stability could function as anticancer therapeutics. This review describes EphA2-Sam and its interactome from a structural and functional perspective. The diverse design strategies that have thus far been employed to obtain peptides targeting EphA2-mediated Sam-Sam interactions are summarized as well. The generated peptides represent good initial lead compounds, but surely many efforts need to be devoted in the close future to improve interaction affinities towards Sam domains and consequently validate their anticancer properties.
Collapse
|
9
|
Ben Hammouda M, Ahmad I, Hamdi A, Dbeibia A, Patel H, Bouali N, Sabri Hamadou W, Hosni K, Ghannay S, Alminderej F, Noumi E, Snoussi M, Aouadi K, Kadri A. Design, synthesis, biological evaluation and in silico studies of novel 1,2,3-triazole linked benzoxazine-2,4-dione conjugates as potent antimicrobial, antioxidant and anti-inflammatory agents. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
10
|
Li L, Wu H, Zhu S, Ji Z, Chi X, Xie F, Hao Y, Lu H, Yang F, Yan L, Zhang D, Jiang Y, Ni T. Discovery of Novel 7-Hydroxy-5-oxo-4,5-dihydrothieno[3,2- b]pyridine-6-carboxamide Derivatives with Potent and Selective Antifungal Activity against Cryptococcus Species. J Med Chem 2022; 65:11257-11269. [PMID: 35922963 DOI: 10.1021/acs.jmedchem.2c00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cryptococcus neoformans and Cryptococcus gattii can cause fatal invasive infections, especially in immunocompromised patients. However, few antifungal drugs are available to help treat cryptococcosis. In this study, by compound library screening, we presented the first report of hit compound P163-0892, which had potent in vitro and in vivo antifungal activity against Cryptococcus spp. In vitro tests showed that P163-0892 was not cytotoxic and had highly selective and strong antifungal activities against Cryptococcus spp. with MIC values less than 1 μg/mL. Synergism of P163-0892 and fluconazole was also observed in vitro. The in vivo antifungal efficacy of P163-0892 was assessed in a wax moth larval fungal infection model, and treatment with 10 mg/kg P163-0892 caused a significant reduction in fungal burden and significant extension of the survival time. Taken together, our data indicate that the hit compound P163-0892 warrants further investigation as a novel anti-Cryptococcus agent.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Hao Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Shuo Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaochen Chi
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Xie
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Lan Yan
- Center for New Drug Research, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
11
|
Jabli S, Hrichi S, Chaabane-Banaoues R, Molton F, Loiseau F, Roisnel T, Turowska-Tyrk I, Babba H, Nasri H. Study on the synthesis, physicochemical, electrochemical properties, molecular structure and antifungal activities of the 4-pyrrolidinopyridine Mg(II) meso-tetratolylporphyrin complex. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
12
|
Moghimi S, Shafiei M, Foroumadi A. Drug design strategies for the treatment azole-resistant candidiasis. Expert Opin Drug Discov 2022; 17:879-895. [PMID: 35793245 DOI: 10.1080/17460441.2022.2098949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite the availability of novel antifungals and therapeutic strategies, the rate of global mortality linked to invasive fungal diseases from fungal infection remains high. Candida albicans account for the most invasive mycosis produced by yeast. Thus, the current arsenal of medicinal chemists is focused on finding new effective agents with lower toxicity and broad-spectrum activity. In this review article, recent efforts to find effective agents against azole-resistant candidiasis, a common fungal infection, are covered. AREAS COVERED Herein, the authors outlined all azole-based compounds, dual target, and new scaffolds (non-azole-based compounds) which were effective against azole-resistant candidiasis. In addition, the mechanism of action and SAR studies were also discussed, if the data were available. EXPERT OPINION The current status of fungal infections and the drawbacks of existing drugs have encouraged scientists to find novel scaffolds based on different methods like virtual screening and fragment-based drug discovery. Machine learning and in-silico methods have found their role in this field and experts are hopeful to find novel scaffolds/compounds by using these methods.
Collapse
Affiliation(s)
- Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shafiei
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Rocha da Silva C, Sá LGDAV, Dos Santos EV, Ferreira TL, Coutinho TDNP, Moreira LEA, de Sousa Campos R, de Andrade CR, Barbosa da Silva WM, de Sá Carneiro I, Silva J, Dos Santos HS, Marinho ES, Cavalcanti BC, de Moraes MO, Júnior HVN, Andrade Neto JB. Evaluation of the antifungal effect of chlorogenic acid against strains of Candida spp. resistant to fluconazole: apoptosis induction and in silico analysis of the possible mechanisms of action. J Med Microbiol 2022; 71. [PMID: 35575783 DOI: 10.1099/jmm.0.001526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Introduction. Candida spp. are commensal fungal pathogens of humans, but when there is an imbalance in the microbiota, or weak host immunity, these yeasts can become pathogenic, generating high medical costs.Gap Statement. With the increase in resistance to conventional antifungals, the development of new therapeutic strategies is necessary. This study evaluated the in vitro antifungal activity of chlorogenic acid against fluconazole-resistant strains of Candida spp. Mechanism of action through flow cytometry and in silico analyses, as well as molecular docking assays with ALS3 and SAP5, important proteins in the pathogenesis of Candida albicans associated with the adhesion process and biofilm formation.Results. The chlorogenic acid showed in vitro antifungal activity against the strains tested, causing reduced cell viability, increased potential for mitochondrial depolarization and production of reactive oxygen species, DNA fragmentation and phosphatidylserine externalization, indicating an apoptotic process. Concerning the analysis through docking, the complexes formed between chlorogenic acid and the targets Thymidylate Kinase, CYP51, 1Yeast Cytochrome BC1 Complex e Exo-B-(1,3)-glucanase demonstrated more favourable binding energy. In addition, chlorogenic acid presented significant interactions with the ALS3 active site residues of C. albicans, important in the adhesion process and resistance to fluconazole. Regarding molecular docking with SAP5, no significant interactions were found between chlorogenic acid and the active site of the enzyme.Conclusion. We concluded that chlorogenic acid has potential use as an adjuvant in antifungal therapies, due to its anti-Candida activity and ability to interact with important drug targets.
Collapse
Affiliation(s)
- Cecília Rocha da Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lívia Gurgel do Amaral Valente Sá
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | | | | | | | - Lara Elloyse Almeida Moreira
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Rosana de Sousa Campos
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | | | | | - Igor de Sá Carneiro
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jacilene Silva
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Hélcio Silva Dos Santos
- Science and Technology Centre, Course of Chemistry, State University Vale do Acaraú, Sobral, CE, Brazil
| | - Emmanuel Silva Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Bruno Coelho Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico de Moraes
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio Vitoriano Nobre Júnior
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João Batista Andrade Neto
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| |
Collapse
|
14
|
Barroso FD, da Silva LJ, Sá LG, da Silva CR, Neto JB, do Nascimento FB, Queiroz HA, Leitão AC, Cabral VP, Rodrigues DS, Barbosa AD, Cavalcanti BC, Morais MO, Júnior HV. Synergistic activity of dobutamine combined with azoles and its mechanism of action against strains of Candida glabrata. Future Microbiol 2022; 17:437-448. [PMID: 35285249 DOI: 10.2217/fmb-2020-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To evaluate the antifungal effect of dobutamine against Candida glabrata as well as its synergism with azoles and its action on biofilm. Methods: The M27-A3 protocol and flow cytometry were used for elucidation of the possible mechanism of action. Results: The tested isolates presented MICs ranging from 2 to 32 μg/ml for dobutamine, with fungistatic effect. A total of 82% of the strains showed synergism with fluconazole, with 90% showing synergism with itraconazole. The effect on biofilm formation was nonsignificant. Cytometry tests showed that dobutamine induced mitochondrial depolarization. Conclusion: Dobutamine has an antifungal effect on strains of C. glabrata and synergistic activity with azoles. This effect is probably mediated by increased oxidative damage to the membrane.
Collapse
Affiliation(s)
- Fatima Dd Barroso
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Lisandra J da Silva
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Lívia Gav Sá
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil.,Faculty of Biomedicine, Unichristus University Center, Fortaleza, Brazil
| | - Cecília R da Silva
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - João Ba Neto
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil.,Faculty of Biomedicine, Unichristus University Center, Fortaleza, Brazil
| | - Francisca Ba do Nascimento
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Helaine A Queiroz
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Amanda C Leitão
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Vitória Pf Cabral
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Daniel S Rodrigues
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Amanda D Barbosa
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Bruno C Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 60430-276, Brazil
| | - Manoel O Morais
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 60430-276, Brazil
| | - Hélio Vn Júnior
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
15
|
Ding Y, Zhang K, Yin Y, Wu J. D319 induced antifungal effects through ROS-mediated apoptosis and inhibited isocitrate lyase in Candida albicans. Biochim Biophys Acta Gen Subj 2022; 1866:130050. [PMID: 34800580 DOI: 10.1016/j.bbagen.2021.130050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Candida albicans (C. albicans) is an opportunistic pathogen that can cause superficial and life-threatening systemic infections in immunocompromised patients. However, the available clinically antifungals are limited. Therefore, the development of effective antifungal agents and therapies is urgently needed. Quinoline type of compounds were reported to possess potent anti-fungal effect. A series of quinoline derivatives were synthesized. Moreover their inhibitory activities and mechanisms on C. albicans were evaluated in this study. METHODS The structure of D319 was identified by extensive spectroscopic analysis. The antifungal activity of D319 on C. albicans was evaluated using conventional methods, including the inhibition zone diameters with filter paper, Clinical Laboratory Standard Institute (CLSI) broth microdilution method in vitro, and in a murine model in vivo. Flow cytometry, fluorescence microscopy, western blot, knockout mutant and revertant strain techniques, and molecular modeling were applied to explore the mechanism of action of D319 in anti-Candida. RESULTS D319 exhibited potent anti-Candida activity with Minimum Inhibitory Concentration value of 2.5 μg/mL in vitro. D319 significantly improved survival rate and reduced fungal burden compared to vehicle control in a murine model in vivo. The treatment of C. albicans with D319 resulted in fungal apoptosis through reactive oxygen species (ROS) accumulation in C. albicans. Furthermore, D319 inhibited the glyoxylate enzyme isocitrate lyase (ICL) of C. albicans, which was also confirmed by docking analysis. CONCLUSIONS Quinoline compound D319 exhibited strong anti-Candida activities in vitro and in vivo models through inhibiting ICL activity and ROS accumulation in C. albicans. GENERAL SIGNIFICANCE This study showed that compound D319 as a novel isocitrate lyase inhibitor, would be a promising anti-Candida lead compound, which provided a potential application of this type of compounds in fighting clinical fungal infections. Furthermore, this study also supported ICL as a potential target for anti-Candida drug discovery.
Collapse
Affiliation(s)
- Yanjiao Ding
- Department of Pharmacy, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250022, Shandong, PR China.
| | - Kai Zhang
- Department of Ophthalmology, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250022, Shandong, PR China
| | - Yiqiang Yin
- Department of Pathology, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250022, Shandong, PR China
| | - Jiyong Wu
- Department of Pharmacy, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan 250022, Shandong, PR China.
| |
Collapse
|
16
|
A Recent Overview of 1,2,3-Triazole-Containing Hybrids as Novel Antifungal Agents: Focusing on Synthesis, Mechanism of Action, and Structure-Activity Relationship (SAR). J CHEM-NY 2022. [DOI: 10.1155/2022/7884316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A pharmacophore system has been found as 1,2,3-triazole, a five-membered heterocycle ring with nitrogen heteroatoms. These heterocyclic compounds can be produced using azide-alkyne cycloaddition processes catalyzed by ruthenium or copper. The bioactive compounds demonstrated antitubercular, antibacterial, anti-inflammatory, anticancer, antioxidant, antiviral, and antidiabetic properties. This heterocycle molecule, in particular, with one or more 1,2,3-triazole cores has been found to have the most powerful antifungal effects. The goal of this review is to highlight recent developments in the synthesis and structure-activity relationship (SAR) investigation of this prospective fungicidal chemical. Also there have been explained drugs and mechanism of action of a triazole compound with antifungal activity. This review will be useful in a variety of fields, including medicinal chemistry, organic chemistry, mycology, and pharmacology.
Collapse
|
17
|
Li D, She X, Calderone R. Assessing Complex I (CI) Mitochondrial Subunit Protein Functions in Candida albicans. Methods Mol Biol 2022; 2542:151-160. [PMID: 36008663 DOI: 10.1007/978-1-0716-2549-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mitochondria of Candida species play critical roles in cell metabolism and pathogenesis. Greater emphasis in specific mitochondria activities of this fungus have been revealed through studies that defined fungal or Candida-specific subunit proteins of ETC Complexes I, III, and IV (CI, CIII, and CIV). Functional activities of these subunits have been characterized through the construction of single-gene null mutants. Activities common to mitochondria of most eukaryotes include their importance in metabolism, ATP synthesis, oxidative phosphorylation, oxygen consumption, and redox potential. An important difference among specific subunits compared to eukaryotic species is the role of CI fungal-specific subunit proteins in activities specific to Candida albicans, such as cell wall synthesis, especially cell wall mannan and β-glucan synthesis. We have associated cell wall synthesis with a signal transduction pathway that includes a Chk1p fungal-specific pathway. Recently, based upon the specificity of CI subunit specificities, a suggestion is the development of novel antifungals that target mitochondrial activity.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Xiaodong She
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
- Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS), Jiangsu Key Laboratory of Molecular Biology for Skin Disease and STIs, Nanjing, China
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
18
|
Recent Molecular Tools for the Genetic Manipulation of Highly Industrially Important Mucoromycota Fungi. J Fungi (Basel) 2021; 7:jof7121061. [PMID: 34947043 PMCID: PMC8705501 DOI: 10.3390/jof7121061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/27/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Mucorales is the largest and most well-studied order of the phylum Mucormycota and is known for its rapid growth rate and various industrial applications. The Mucorales fungi are a fascinating group of filamentous organisms with many uses in research and the industrial and medical fields. They are widely used biotechnological producers of various secondary metabolites and other value-added products. Certain members of Mucorales are extensively used as model organisms for genetic and molecular investigation and have extended our understanding of the metabolisms of other members of this order as well. Compared with other fungal species, our understanding of Mucoralean fungi is still in its infancy, which could be linked to their lack of effective genetic tools. However, recent advancements in molecular tools and approaches, such as the construction of recyclable markers, silencing vectors, and the CRISPR-Cas9-based gene-editing system, have helped us to modify the genomes of these model organisms. Multiple genetic modifications have been shown to generate valuable products on a large scale and helped us to understand the morphogenesis, basic biology, pathogenesis, and host–pathogen interactions of Mucoralean fungi. In this review, we discuss various conventional and modern genetic tools and approaches used for efficient gene modification in industrially important members of Mucorales.
Collapse
|
19
|
dos Reis TF, Horta MAC, Colabardini AC, Fernandes CM, Silva LP, Bastos RW, Fonseca MVDL, Wang F, Martins C, Rodrigues ML, Silva Pereira C, Del Poeta M, Wong KH, Goldman GH. Screening of Chemical Libraries for New Antifungal Drugs against Aspergillus fumigatus Reveals Sphingolipids Are Involved in the Mechanism of Action of Miltefosine. mBio 2021; 12:e0145821. [PMID: 34372704 PMCID: PMC8406317 DOI: 10.1128/mbio.01458-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Aspergillus fumigatus is an important fungal pathogen and the main etiological agent of aspergillosis, a disease characterized by a noninvasive process that can evolve to a more severe clinical manifestation, called invasive pulmonary aspergillosis (IPA), in immunocompromised patients. The antifungal arsenal to threat aspergillosis is very restricted. Azoles are the main therapeutic approach to control IPA, but the emergence of azole-resistant A. fumigatus isolates has significantly increased over recent decades. Therefore, new strategies are necessary to combat aspergillosis, and drug repurposing has emerged as an efficient and alternative approach for identifying new antifungal drugs. Here, we used a screening approach to analyze A. fumigatus in vitro susceptibility to 1,127 compounds. A. fumigatus was susceptible to 10 compounds, including miltefosine, a drug that displayed fungicidal activity against A. fumigatus. By screening an A. fumigatus transcription factor null library, we identified a single mutant, which has the smiA (sensitive to miltefosine) gene deleted, conferring a phenotype of susceptibility to miltefosine. The transcriptional profiling (RNA-seq) of the wild-type and ΔsmiA strains and chromatin immunoprecipitation coupled to next-generation sequencing (ChIP-Seq) of an SmiA-tagged strain exposed to miltefosine revealed genes of the sphingolipid pathway that are directly or indirectly regulated by SmiA. Sphingolipid analysis demonstrated that the mutant has overall decreased levels of sphingolipids when growing in the presence of miltefosine. The identification of SmiA represents the first genetic element described and characterized that plays a direct role in miltefosine response in fungi. IMPORTANCE The filamentous fungus Aspergillus fumigatus causes a group of diseases named aspergillosis, and their development occurs after the inhalation of conidia dispersed in the environment. Very few classes of antifungal drugs are available for aspergillosis treatment, e.g., azoles, but the emergence of global resistance to azoles in A. fumigatus clinical isolates has increased over recent decades. Repositioning or repurposing drugs already available on the market is an interesting and faster opportunity for the identification of novel antifungal agents. By using a repurposing strategy, we identified 10 different compounds that impact A. fumigatus survival. One of these compounds, miltefosine, demonstrated fungicidal activity against A. fumigatus. The mechanism of action of miltefosine is unknown, and, aiming to get more insights about it, we identified a transcription factor, SmiA (sensitive to miltefosine), important for miltefosine resistance. Our results suggest that miltefosine displays antifungal activity against A. fumigatus, interfering in sphingolipid biosynthesis.
Collapse
Affiliation(s)
- Thaila Fernanda dos Reis
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- MicroControl Innovation Ltd., Ribeirão Preto, São Paulo, Brazil
| | | | - Ana Cristina Colabardini
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Caroline Mota Fernandes
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Lilian Pereira Silva
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Rafael Wesley Bastos
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Fang Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, China
| | - Celso Martins
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Márcio L. Rodrigues
- Instituto Carlos Chagas (ICC), Fundação Oswaldo Cruz–Fiocruz, Curitiba, Brazil
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Cristina Silva Pereira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Veteran Administration Medical Center, Northport, New York, USA
- MicroRid Technologies Inc., Dix Hills, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, New York, USA
| | - Koon Ho Wong
- Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, SAR, China
| | - Gustavo H. Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
20
|
Dangi M, Khichi A, Jakhar R, Chhillar AK. Growing Preferences towards Analog-based Drug Discovery. Curr Pharm Biotechnol 2021; 22:1030-1045. [PMID: 32900347 DOI: 10.2174/1389201021666200908121409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/29/2020] [Accepted: 08/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The major concern of today's time is the developing resistance in most of the clinically derived pathogenic micro-organisms for available drugs through several mechanisms. Therefore, there is a dire need to develop novel molecules with drug-like properties that can be effective against the otherwise resistant micro-organisms. METHODS New drugs can be developed using several methods like structure-based drug design, ligandbased drug design, or by developing analogs of the available drugs to further improve their effects. However, the smartness is to opt for the techniques that have comparatively less expenditure, lower failure rates, and faster discovery rates. RESULTS Analog-Based Drug Design (ABDD) is one such technique that researchers worldwide are opting to develop new drug-like molecules with comparatively lower market values. They start by first designing the analogs sharing structural and pharmacological similarities to the existing drugs. This method embarks on scaffold structures of available drugs already approved by the clinical trials, but are left ineffective because of resistance developed by the pathogens. CONCLUSION In this review, we have discussed some recent examples of anti-fungal and anti-bacterial (antimicrobial) drugs that were designed based on the ABDD technique. Also, we have tried to focus on the in silico tools and techniques that can contribute to the designing and computational screening of the analogs, so that these can be further considered for in vitro screening to validate their better biological activities against the pathogens with comparatively reduced rates of failure.
Collapse
Affiliation(s)
- Mehak Dangi
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Alka Khichi
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Ritu Jakhar
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Anil K Chhillar
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| |
Collapse
|
21
|
Recent Advances in Genome Editing Tools in Medical Mycology Research. J Fungi (Basel) 2021; 7:jof7040257. [PMID: 33808382 PMCID: PMC8067129 DOI: 10.3390/jof7040257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Manipulating fungal genomes is an important tool to understand the function of target genes, pathobiology of fungal infections, virulence potential, and pathogenicity of medically important fungi, and to develop novel diagnostics and therapeutic targets. Here, we provide an overview of recent advances in genetic manipulation techniques used in the field of medical mycology. Fungi use several strategies to cope with stress and adapt themselves against environmental effectors. For instance, mutations in the 14 alpha-demethylase gene may result in azole resistance in Aspergillusfumigatus strains and shield them against fungicide's effects. Over the past few decades, several genome editing methods have been introduced for genetic manipulations in pathogenic fungi. Application of restriction enzymes to target and cut a double-stranded DNA in a pre-defined sequence was the first technique used for cloning in Aspergillus and Candida. Genome editing technologies, including zinc-finger nucleases (ZFNs) and transcriptional activator-like effector nucleases (TALENs), have been also used to engineer a double-stranded DNA molecule. As a result, TALENs were considered more practical to identify single nucleotide polymorphisms. Recently, Class 2 type II Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 technology has emerged as a more useful tool for genome manipulation in fungal research.
Collapse
|
22
|
Zhang M, Lu J, Duan X, Chen J, Jin X, Lin Z, Pang Y, Wang X, Lou H, Chang W. Rimonabant potentiates the antifungal activity of amphotericin B by increasing cellular oxidative stress and cell membrane permeability. FEMS Yeast Res 2021; 21:6168383. [PMID: 33705544 DOI: 10.1093/femsyr/foab016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/02/2021] [Indexed: 12/23/2022] Open
Abstract
Amphotericin B (AmB) is a very effective antifungal agent, and resistance in clinical isolates is rare. However, clinical treatment with AmB is often associated with severe side effects. Reducing the administration dose of AmB by combining it with other agents is a promising strategy to minimize this toxicity. In this study, we screened a small compound library and observed that the anti-obesity drug rimonabant exhibited synergistic antifungal action with AmB against Candida species and Cryptococcus neoformans. Moreover, the combination of AmB and rimonabant exhibited synergistic or additive effects against Candida albicans biofilm formation and cell viability in preformed biofilms. The effects of this combination were further confirmed in vivo using a murine systemic infection model. Exploration of the mechanism of synergy revealed that rimonabant enhances the fungicidal activity of AmB by increasing cellular oxidative stress and cell membrane permeability. These findings provide a foundation for the possible development of AmB-rimonabant polytherapies for fungal infections.
Collapse
Affiliation(s)
- Ming Zhang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jinghui Lu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ximeng Duan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jinyao Chen
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xueyang Jin
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhaomin Lin
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xuexiang Wang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Wenqiang Chang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
23
|
Zhang Y, Fang W, Raimi OG, Lockhart DEA, Ferenbach AT, Lu L, van Aalten DMF. Genetic and structural validation of phosphomannomutase as a cell wall target in Aspergillus fumigatus. Mol Microbiol 2021; 116:245-259. [PMID: 33629421 DOI: 10.1111/mmi.14706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 11/29/2022]
Abstract
Aspergillus fumigatus is an opportunistic mold responsible for severe life-threatening fungal infections in immunocompromised patients. The cell wall, an essential structure composed of glucan, chitin, and galactomannan, is considered to be a target for the development of antifungal drugs. The nucleotide sugar donor GDP-mannose (GDP-Man) is required for the biosynthesis of galactomannan, glycosylphosphatidylinositol (GPI) anchors, glycolipid, and protein glycosylation. Starting from fructose-6-phosphate, GDP-Man is produced by the sequential action of the enzymes phosphomannose isomerase, phosphomannomutase (Pmm), and GDP-mannose pyrophosphorylase. Here, using heterokaryon rescue and gene knockdown approaches we demonstrate that the phosphomannomutase encoding gene in A. fumigatus (pmmA) is essential for survival. Reduced expression of pmmA is associated with significant morphological defects including retarded germination, growth, reduced conidiation, and abnormal polarity. Moreover, the knockdown strain exhibited an altered cell wall organization and sensitivity toward cell wall perturbing agents. By solving the first crystal structure of A. fumigatus phosphomannomutase (AfPmmA) we identified non-conservative substitutions near the active site when compared to the human orthologues. Taken together, this work provides a genetic and structural foundation for the exploitation of AfPmmA as a potential antifungal target.
Collapse
Affiliation(s)
- Yuanwei Zhang
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,School of Life Sciences, University of Dundee, Dundee, UK
| | - Wenxia Fang
- School of Life Sciences, University of Dundee, Dundee, UK.,National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China
| | | | | | | | - Ling Lu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | | |
Collapse
|
24
|
Abd Algaffar SO, Verbon A, van de Sande WWJ, Khalid SA. Development and Validation of an In Vitro Resazurin-Based Susceptibility Assay against Madurella mycetomatis. Antimicrob Agents Chemother 2021; 65:e01338-20. [PMID: 33318015 PMCID: PMC8092548 DOI: 10.1128/aac.01338-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023] Open
Abstract
We present an in vitro susceptibility assay for Madurella mycetomatis hyphae using resazurin for endpoint reading. Using this assay, reproducible MICs were obtained for amphotericin B, itraconazole, voriconazole, posaconazole, terbinafine, and micafungin. Results were comparable with those of a 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide salt (XTT)-based susceptibility assay. The lowest MICs were obtained for itraconazole and posaconazole (MIC50, 0.016 µg/ml) followed by voriconazole (MIC50, 0.063 µg/ml). Amphotericin B, micafungin, and terbinafine appeared much less effective.
Collapse
Affiliation(s)
- S O Abd Algaffar
- Faculty of Pharmacy, University of Science & Technology, Omdurman, Sudan
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - A Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - W W J van de Sande
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - S A Khalid
- Faculty of Pharmacy, University of Science & Technology, Omdurman, Sudan
| |
Collapse
|
25
|
The Future of Antifungal Drug Therapy: Novel Compounds and Targets. Antimicrob Agents Chemother 2021; 65:AAC.01719-20. [PMID: 33229427 DOI: 10.1128/aac.01719-20] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fungal infections are a universal problem and are routinely associated with high morbidity and mortality rates in immunocompromised patients. Existing therapies comprise five different classes of antifungal agents, four of which target the synthesis of ergosterol and cell wall glucans. However, the currently available antifungals have many limitations, including poor oral bioavailability, narrow therapeutic indices, and emerging drug resistance resulting from their use, thus making it essential to investigate the development of novel drugs which can overcome these limitations and add to the antifungal armamentarium. Advances have been made in antifungal drug discovery research and development over the past few years as evidenced by the presence of several new compounds currently in various stages of development. In the following minireview, we provide a comprehensive summary of compounds aimed at one or more novel molecular targets. We also briefly describe potential pathways relevant for fungal pathogenesis that can be considered for drug development in the near future.
Collapse
|
26
|
History of the development of antifungal azoles: A review on structures, SAR, and mechanism of action. Bioorg Chem 2020; 104:104240. [DOI: 10.1016/j.bioorg.2020.104240] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/17/2020] [Accepted: 08/11/2020] [Indexed: 01/12/2023]
|
27
|
Hahn F, Guth FM. The ambruticins and jerangolids - chemistry, biology and chemoenzymatic synthesis of potent antifungal drug candidates. Nat Prod Rep 2020; 37:1300-1315. [PMID: 32420573 DOI: 10.1039/d0np00012d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: 1977 to 2020The ambruticins and jerangolids are myxobacterial reduced polyketides, which are produced via highly unusual biosynthetic pathways containing a plethora of non-canonical enzymatic transformations. Since the discovery of the first congeners in the late 1970s, they have been in the focus of drug development due to their good antifungal activity and low toxicity in mammals, which result from interaction with an unusual innercellular target in fungi. Despite significant efforts, which have led to the development of various total syntheses, their structural complexity has yet avoided full exploitation of their pharmacological potential. This article summarises biological, total and semisynthetic as well as biosynthetic studies on both compounds. An outlook on the biosynthesis-based approaches to them and their derivatives is presented. Due to the structural and biosynthetic characteristics of the ambruticins and jerangolids, chemoenzymatic processes that make use of their biosynthetic pathway enzymes are particularly promising to gain efficient access to derivative libraries for structure activity relationship studies.
Collapse
Affiliation(s)
- Frank Hahn
- Department of Chemistry, University of Bayreuth, 51427 Bayreuth, Germany.
| | | |
Collapse
|
28
|
Yuan-Biao Q, Lan-Fang Z, Qi Q, Jia-Hui N, Ze-Mei R, Hai-Mei Y, Chen-Chen Z, Hong-Ju P, Nan-Nan D, Qing-Shan L. Antifungal resistance-modifying multiplexing action of Momordica charantia protein and phosphorylated derivatives on the basis of growth-dependent gene coregulation in Candida albicans. Med Mycol 2020; 59:myaa070. [PMID: 32871589 DOI: 10.1093/mmy/myaa070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/26/2020] [Indexed: 01/08/2023] Open
Abstract
Fungal growth-dependent gene coregulation is strongly implicated in alteration of gene-encoding target proteases ruling with an antifungal resistance niche and biology of resistant mutants. On the basis of multi-alterative processes in this platform, the resistance-modifying strategy is designed in ketoconazole resistant Candida albicans and evaluated with less selective Momordica charantia protein and allosterically phosphorylated derivatives at the Thr102, Thr24 and Thr255 sites, respectively. We demonstrate absolutely chemo-sensitizing efficacy regarding stepwise-modifying resistance in sensitivity, by a load of only 26.23-40.00 μg/l agents in Sabouraud's dextrose broth. Five successive modifying-steps realize the decreasing of ketoconazole E-test MIC50 from 11.10 to a lower level than 0.10 mg/l. With the ketoconazole resistance-modifying, colony undergoes a high-frequency morphological switch between high ploidy (opaque) and small budding haploid (white). A cellular event in the first modifying-step associates with relatively slow exponential growth (ie, a 4-h delay)-dependent action, mediated by agents adsorption. Moreover, multiple molecular roles are coupled with intracellularly and extracellularly binding to ATP-dependent RNA helicase dbp6; the 0.08-2.45 fold upregulation of TATA-box-binding protein, rRNA-processing protein and translation initiation factor 5A; and the 7.52-55.33% decrease of cytochrome P450 lanosterol 14α-demethylase, glucan 1, 3-β glucosidase, candidapepsin-1 and 1-acylglycerol-3-phosphate O-acyltransferase. Spatial and temporal gene coregulation, in the transcription and translation initiation stages with rRNA-processing, is a new coprocessing platform enabling target protease attenuations for resistance-impairing. An updated resistance-modifying measure of these agents in the low-dose antifungal strategic design may provide opportunities to a virtually safe therapy that is in high dose-dependency. LAY SUMMARY A new platform to modify resistance is fungal growth-dependent gene coregulation. MAP30 and phosphorylated derivatives are candidate resistance-modifying agents. Low-dose stepwise treatment absolutely modifies azole resistance in model fungus.
Collapse
Affiliation(s)
- Qiao Yuan-Biao
- Shanxi Key Laboratory of Innovative Drugs for the Treatment of Serious Diseases Basing on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, P. R. China
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Zhang Lan-Fang
- Shanxi Key Laboratory of Innovative Drugs for the Treatment of Serious Diseases Basing on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, P. R. China
| | - Qiao Qi
- Department of Medical Biochemistry and Microbiology, Uppsala Biomedical Center, Uppsala University, Husargatan 3, Box 582, SE-751 23 Uppsala, Sweden
| | - Niu Jia-Hui
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Ren Ze-Mei
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Yang Hai-Mei
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Zhu Chen-Chen
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Pan Hong-Ju
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Duan Nan-Nan
- Graduate Institute of Pharmaceutical Chemistry, Luliang University, Luliang, Shanxi 033001, P. R. China
| | - Li Qing-Shan
- Shanxi Key Laboratory of Innovative Drugs for the Treatment of Serious Diseases Basing on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, P. R. China
| |
Collapse
|
29
|
Xue A, Robbins N, Cowen LE. Advances in fungal chemical genomics for the discovery of new antifungal agents. Ann N Y Acad Sci 2020; 1496:5-22. [PMID: 32860238 DOI: 10.1111/nyas.14484] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/09/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022]
Abstract
Invasive fungal infections have escalated from a rare curiosity to a major cause of human mortality around the globe. This is in part due to a scarcity in the number of antifungal drugs available to combat mycotic disease, making the discovery of novel bioactive compounds and determining their mode of action of utmost importance. The development and application of chemical genomic assays using the model yeast Saccharomyces cerevisiae has provided powerful methods to identify the mechanism of action of diverse molecules in a living cell. Furthermore, complementary assays are continually being developed in fungal pathogens, most notably Candida albicans and Cryptococcus neoformans, to elucidate compound mechanism of action directly in the pathogen of interest. Collectively, the suite of chemical genetic assays that have been developed in multiple fungal species enables the identification of candidate drug target genes, as well as genes involved in buffering drug target pathways, and genes involved in general cellular responses to small molecules. In this review, we examine current yeast chemical genomic assays and highlight how such resources provide powerful tools that can be utilized to bolster the antifungal pipeline.
Collapse
Affiliation(s)
- Alice Xue
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
LeBlanc EV, Polvi EJ, Veri AO, Privé GG, Cowen LE. Structure-guided approaches to targeting stress responses in human fungal pathogens. J Biol Chem 2020; 295:14458-14472. [PMID: 32796038 DOI: 10.1074/jbc.rev120.013731] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/11/2020] [Indexed: 11/06/2022] Open
Abstract
Fungi inhabit extraordinarily diverse ecological niches, including the human body. Invasive fungal infections have a devastating impact on human health worldwide, killing ∼1.5 million individuals annually. The majority of these deaths are attributable to species of Candida, Cryptococcus, and Aspergillus Treating fungal infections is challenging, in part due to the emergence of resistance to our limited arsenal of antifungal agents, necessitating the development of novel therapeutic options. Whereas conventional antifungal strategies target proteins or cellular components essential for fungal growth, an attractive alternative strategy involves targeting proteins that regulate fungal virulence or antifungal drug resistance, such as regulators of fungal stress responses. Stress response networks enable fungi to adapt, grow, and cause disease in humans and include regulators that are highly conserved across eukaryotes as well as those that are fungal-specific. This review highlights recent developments in elucidating crystal structures of fungal stress response regulators and emphasizes how this knowledge can guide the design of fungal-selective inhibitors. We focus on the progress that has been made with highly conserved regulators, including the molecular chaperone Hsp90, the protein phosphatase calcineurin, and the small GTPase Ras1, as well as with divergent stress response regulators, including the cell wall kinase Yck2 and trehalose synthases. Exploring structures of these important fungal stress regulators will accelerate the design of selective antifungals that can be deployed to combat life-threatening fungal diseases.
Collapse
Affiliation(s)
- Emmanuelle V LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G Privé
- Departments of Medical Biophysics and Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Li D, She X, Calderone R. The antifungal pipeline: the need is established. Are there new compounds? FEMS Yeast Res 2020; 20:5827531. [DOI: 10.1093/femsyr/foaa023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
Our review summarizes and compares the temporal development (eras) of antifungal drug discovery as well as antibacterial ventures. The innovation gap that occurred in antibacterial discovery from 1960 to 2000 was likely due to tailoring of existing compounds to have better activity than predecessors. Antifungal discovery also faced innovation gaps. The semi-synthetic antibiotic era was followed closely by the resistance era and the heightened need for new compounds and targets. With the immense contribution of comparative genomics, antifungal targets became part of the discovery focus. These targets by definition are absolutely required to be fungal- or even lineage (clade) specific. Importantly, targets need to be essential for growth and/or have important roles in disease and pathogenesis. Two types of antifungals are discussed that are mostly in the FDA phase I–III clinical trials. New antifungals are either modified to increase bioavailability and stability for instance, or are new compounds that inhibit new targets. One of the important developments in incentivizing new antifungal discovery has been the prolific number of publications of global and country-specific incidence. International efforts that champion global antimicrobial drug discovery are discussed. Still, interventions are needed. The current pipeline of antifungals and alternatives to antifungals are discussed including vaccines.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| | - Xiaodong She
- Jiangsu Key laboratory of Molecular Biology for Skin Disease and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS), Nanjing 210029, China
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| |
Collapse
|
32
|
Pippi B, Joaquim A, Lopes W, Machado G, Bergamo V, Giuliani L, Abegg M, Cruz L, Vainstein M, Fuentefria A, Andrade S. 8‐Hydroxyquinoline‐5‐sulfonamides are promising antifungal candidates for the topical treatment of dermatomycosis. J Appl Microbiol 2019; 128:1038-1049. [DOI: 10.1111/jam.14545] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/08/2019] [Accepted: 11/30/2019] [Indexed: 12/15/2022]
Affiliation(s)
- B. Pippi
- Programa de Pós‐Graduação em Microbiologia Agrícola e do Ambiente Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - A.R. Joaquim
- Programa de Pós‐Graduação em Ciências Farmacêuticas Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - W. Lopes
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - G.R.M. Machado
- Programa de Pós‐Graduação em Microbiologia Agrícola e do Ambiente Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - V.Z. Bergamo
- Programa de Pós‐Graduação em Microbiologia Agrícola e do Ambiente Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - L.M. Giuliani
- Programa de Pós‐graduação em Ciências Farmacêuticas Universidade Federal de Santa Maria Santa Maria RS Brazil
| | - M.A. Abegg
- Instituto de Ciências Exatas e Tecnologia Universidade Federal do Amazonas Itacoatiara AM Brazil
| | - L. Cruz
- Programa de Pós‐graduação em Ciências Farmacêuticas Universidade Federal de Santa Maria Santa Maria RS Brazil
| | - M.H. Vainstein
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - A.M. Fuentefria
- Programa de Pós‐Graduação em Microbiologia Agrícola e do Ambiente Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - S.F. Andrade
- Programa de Pós‐Graduação em Microbiologia Agrícola e do Ambiente Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| |
Collapse
|
33
|
Jamzivar F, Shams-Ghahfarokhi M, Khoramizadeh M, Yousefi N, Gholami-Shabani M, Razzaghi-Abyaneh M. Unraveling the importance of molecules of natural origin in antifungal drug development through targeting ergosterol biosynthesis pathway. IRANIAN JOURNAL OF MICROBIOLOGY 2019; 11:448-459. [PMID: 32148676 PMCID: PMC7048963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Over the past decades, the incidence of life-threatening fungal infections has increased dramatically in particular among patients with hampered immune function. Fungal infections cause around 1.5 million deaths annually, superior to malaria and tuberculosis. With respect to high toxicity, narrow spectrum of activity and drug resistance to current antifungals, there is an urgent need to discover novel leads from molecules of natural origin especially those derived from plants and microorganisms for antifungal drug discovery. Among antifungal drugs introduced into the clinic, those affecting ergosterol biosynthesis are still superior to other classes and the vital role of ergosterol in fungal growth and development. This review highlights current knowledge about available antifungal agents and further issues on antifungal drug discovery from compounds of natural origin which affect ergosterol biosynthesis. Special attention is made to the fungal sterol C24-methyltransferase (SMT), a crucial enzyme in ergosterol biosynthesis pathway as a novel target for rational drug design.
Collapse
Affiliation(s)
| | | | - Mansoor Khoramizadeh
- School of Iranian Traditional Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Mehdi Razzaghi-Abyaneh
- Department of Mycology, Pasteur Institute of Iran, Tehran, Iran,Corresponding author: Mehdi Razzaghi-Abyaneh, PhD, Department of Mycology, Pasteur Institute of Iran, Tehran, Iran., Tel: +98 21 64112804, Fax: +98 21 64112804, &
| |
Collapse
|
34
|
C16-Fengycin A affect the growth of Candida albicans by destroying its cell wall and accumulating reactive oxygen species. Appl Microbiol Biotechnol 2019; 103:8963-8975. [PMID: 31630240 DOI: 10.1007/s00253-019-10117-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Abstract
Candida albicans is the most common clinical pathogenic fungus, which is highly susceptible to immunodeficiency. Development of novel antifungal agents has become a growing trend in the treatment of Candida infections. C16-Fengycin A, a lipopeptide isolated from Bacillus amyloliquefaciens fmb60 showed significant fungicidal activity against C. albicans. In the study, we explored the possible antifungal mode of C16-Fengycin A. It was predicted that C16-Fengycin A had the ability to disrupt the cell wall due to its alterations of cell ultrastructure, and reduction of cell wall hydrophobicity. This was further confirmed by the changes in the exposure of the cell wall components and down-regulation of the genes related in the cell wall synthesis. Meanwhile, with the treatment of C16-Fengycin A, the levels of reactive oxygen species (ROS) increased, resulting in mitochondrial dysfunction in the cells. We hypothesized that the antifungal mechanism of C16-Fengycin A might be via the destruction of the cell wall and the accumulation of ROS, which could activate the High-Osmolarity Glycerol Mitogen-Activated Protein Kinase (HOG-MAPK) pathway. Our findings indicated that C16-Fengycin A could be a potential antifungal agent that could be used to treat candida infections.
Collapse
|
35
|
Li L, Zhang T, Xu J, Wu J, Wang Y, Qiu X, Zhang Y, Hou W, Yan L, An M, Jiang Y. The Synergism of the Small Molecule ENOblock and Fluconazole Against Fluconazole-Resistant Candida albicans. Front Microbiol 2019; 10:2071. [PMID: 31555252 PMCID: PMC6742966 DOI: 10.3389/fmicb.2019.02071] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
Candida albicans is the most common opportunistic fungal pathogen which can cause life-threatening bloodstream infections known as candidaemia. It is very important to discover new drugs and targets for the treatment of candidaemia. In this study, we first investigated the combination antifungal effects of the small molecule ENOblock and fluconazole (FLC) against FLC-resistant C. albicans. A checkerboard microdilution assay showed that ENOblock has a significant synergistic effect in combination with FLC against FLC-resistant C. albicans. The time-kill curve further confirmed the synergism of this compound with FLC against FLC-resistant C. albicans. Moreover, we demonstrated the significant inhibitory effects of ENOblock alone and in combination with FLC against C. albicans hypha and biofilm formation. Furthermore, the XTT assay showed that ENOblock has relatively low toxicity to human umbilical vein endothelial cells. The in vivo antifungal efficacy of ENOblock was further assessed in a murine model of systemic C. albicans infection. Although ENOblock alone was not sufficient to treat C. albicans infection, the combination of FLC and ENOblock showed significant in vivo activity against FLC-resistant C. albicans. Finally, using surface plasmon resonance analysis as well as an inhibition assay, we determined that ENOblock directly interacted with CaEno1 and significantly inhibited the transglutaminase activity of this enzyme, which is involved in the growth and morphogenesis of C. albicans. In summary, these results demonstrate the synergistic effects of FLC and ENOblock against FLC-resistant C. albicans, and indicate that inhibition of the transglutaminase activity of CaEno1 by ENOblock might confer an advantage for the synergism of FLC and ENOblock, suggesting the potential of ENOblock as a new antifungal candidate.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Teng Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yida Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiran Qiu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weitong Hou
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Yan
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Maomao An
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Abstract
The enigmatic yeast Candida auris has emerged over the last decade and rapidly penetrated our consciousness. The global threat from this multidrug-resistant yeast has generated a call to arms from within the medical mycology community. Over the past decade, our understanding of how this yeast has spread globally, its clinical importance, and how it tolerates and resists antifungal agents has expanded. This review highlights the clinical importance of antifungal resistance in C. auris and explores our current understanding of the mechanisms associated with azole, polyene, and echinocandin resistance. We also discuss the impact of phenotypic tolerance, with particular emphasis on biofilm-mediated resistance, and present new pipelines of antifungal drugs that promise new hope in the management of C. auris infection.
Collapse
Affiliation(s)
- Ryan Kean
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Gordon Ramage
- Oral Sciences Research Group, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
37
|
Kadry AA, El-Ganiny AM, Mosbah RA, Kaminskyj SGW. Deletion of Aspergillus nidulans GDP-mannose transporters affects hyphal morphometry, cell wall architecture, spore surface character, cell adhesion, and biofilm formation. Med Mycol 2018; 56:621-630. [PMID: 29420778 DOI: 10.1093/mmy/myx082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
Systemic human fungal infections are increasingly common. Aspergillus species cause most of the airborne fungal infections. Life-threatening invasive aspergillosis was formerly found only in immune-suppressed patients, but recently some strains of A. fumigatus have become primary pathogens. Many fungal cell wall components are absent from mammalian systems, so they are potential drug targets. Cell-wall-targeting drugs such as echinocandins are used clinically, although echinocandin-resistant strains were discovered shortly after their introduction. Currently there are no fully effective anti-fungal drugs. Fungal cell wall glycoconjugates modulate human immune responses, as well as fungal cell adhesion, biofilm formation, and drug resistance. Guanosine diphosphate (GDP) mannose transporters (GMTs) transfer GDP-mannose from the cytosol to the Golgi lumen prior to mannosylation. Aspergillus nidulans GMTs are encoded by gmtA and gmtB. Here we elucidate the roles of A. nidulans GMTs. Strains engineered to lack either or both GMTs were assessed for hyphal and colonial morphology, cell wall ultrastructure, antifungal susceptibility, spore hydrophobicity, adherence and biofilm formation. The gmt-deleted strains had smaller colonies with reduced sporulation and with thicker hyphal walls. The gmtA deficient spores had reduced hydrophobicity and were less adherent and less able to form biofilms in vitro. Thus, gmtA not only participates in maintaining the cell wall integrity but also plays an important role in biofilm establishment and adherence of A. nidulans. These findings suggested that GMTs have roles in A. nidulans growth and cell-cell interaction and could be a potential target for new antifungals that target virulence determinants.
Collapse
Affiliation(s)
- Ashraf A Kadry
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Amira M El-Ganiny
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Rasha A Mosbah
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.,Zagazig University Hospitals, Zagazig, Egypt
| | | |
Collapse
|
38
|
Khandelwal NK, Sarkar P, Gaur NA, Chattopadhyay A, Prasad R. Phosphatidylserine decarboxylase governs plasma membrane fluidity and impacts drug susceptibilities of Candida albicans cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2308-2319. [DOI: 10.1016/j.bbamem.2018.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/01/2018] [Accepted: 05/27/2018] [Indexed: 12/28/2022]
|
39
|
Mayer FL, Sánchez-León E, Kronstad JW. A chemical genetic screen reveals a role for proteostasis in capsule and biofilm formation by Cryptococcus neoformans. MICROBIAL CELL 2018; 5:495-510. [PMID: 30483521 PMCID: PMC6244295 DOI: 10.15698/mic2018.11.656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pathogenic microorganisms employ specialized virulence factors to cause disease. Biofilm formation and the production of a polysaccharide capsule are two important virulence factors in Cryptococcus neoformans, the fungal pathogen that causes meningoencephalitis. Here, we show that the bipolar disorder drug lithium inhibits formation of both virulence factors by a mechanism involving dysregulation of the ubiquitin/proteasome system. By using a chemical genetics approach and bioinformatic analyses, we describe the cellular landscape affected by lithium treatment. We demonstrate that lithium affects many different pathways in C. neoformans, including the cAMP/protein kinase A, inositol biosynthesis, and ubiquitin/proteasome pathways. By analyzing mutants with defects in the ubiquitin/proteasome system, we uncover a role for proteostasis in both capsule and biofilm formation. Moreover, we demonstrate an additive influence of lithium and the proteasome inhibitor bortezomib in inhibiting capsule production, thus establishing a link between lithium activity and the proteasome system. Finally, we show that the lithium-mimetic drug ebselen potently blocks capsule and biofilm formation, and has additive activity with lithium or bortezomib. In summary, our results illuminate the impact of lithium on C. neoformans, and link dysregulation of the proteasome to capsule and biofilm inhibition in this important fungal pathogen.
Collapse
Affiliation(s)
- François L Mayer
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eddy Sánchez-León
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - James W Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
40
|
Vallières C, Raulo R, Dickinson M, Avery SV. Novel Combinations of Agents Targeting Translation That Synergistically Inhibit Fungal Pathogens. Front Microbiol 2018; 9:2355. [PMID: 30349511 PMCID: PMC6186996 DOI: 10.3389/fmicb.2018.02355] [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: 07/30/2018] [Accepted: 09/14/2018] [Indexed: 12/29/2022] Open
Abstract
A range of fungicides or antifungals are currently deployed to control fungi in agriculture or medicine, but resistance to current agents is growing so new approaches and molecular targets are urgently needed. Recently, different aminoglycoside antibiotics combined with particular transport inhibitors were found to produce strong, synergistic growth-inhibition of fungi, by synergistically increasing the error rate of mRNA translation. Here, focusing on translation fidelity as a novel target for combinatorial antifungal treatment, we tested the hypothesis that alternative combinations of agents known to affect the availability of functional amino acids would synergistically inhibit growth of major fungal pathogens. We screened 172 novel combinations against three phytopathogens (Rhizoctonia solani, Zymoseptoria tritici, and Botrytis cinerea) and three human pathogens (Cryptococcus neoformans, Candida albicans, and Aspergillus fumigatus), showing that 48 combinations inhibited strongly the growth of the pathogens; the growth inhibition effect was significantly greater with the agents combined than by a simple product of their individual effects at the same doses. Of these, 23 combinations were effective against more than one pathogen, including combinations comprising food-and-drug approved compounds, e.g., quinine with bicarbonate, and quinine with hygromycin. These combinations [fractional inhibitory combination (FIC) index ≤0.5] gave up to 100% reduction of fungal growth yield at concentrations of agents which, individually, had negligible effect. No synergy was evident against bacterial, plant or mammalian cells, indicating specificity for fungi. Mode-of-action analyses for quinine + hygromycin indicated that synergistic mistranslation was the antifungal mechanism. That mechanism was not universal as bicarbonate exacerbated quinine action by increasing drug uptake. The study unveils chemical combinations and a target process with potential for control of diverse fungal pathogens, and suggests repurposing possibilities for several current therapeutics.
Collapse
Affiliation(s)
- Cindy Vallières
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| | - Roxane Raulo
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| | - Matthew Dickinson
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Simon V Avery
- School of Life Sciences, University of Nottingham, University Park Campus, Nottingham, United Kingdom
| |
Collapse
|
41
|
Futuro DO, Ferreira PG, Nicoletti CD, Borba-Santos LP, Silva FCDA, Rozental S, Ferreira VF. The Antifungal Activity of Naphthoquinones: An Integrative Review. AN ACAD BRAS CIENC 2018; 90:1187-1214. [PMID: 29873671 DOI: 10.1590/0001-3765201820170815] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/08/2017] [Indexed: 01/05/2023] Open
Abstract
Naphthoquinones are the most commonly occurring type of quinones in nature. They are a diverse family of secondary metabolites that occur naturally in plants, lichens and various microorganisms. This subgroup is constantly being expanded through the discovery of new natural products and by the synthesis of new compounds via innovative techniques. Interest in quinones and the search for new biological activities within the members of this class have intensified in recent years, as evidenced by the evaluation of the potential antimicrobial activities of quinones. Among fungi of medical interest, yeasts of the genus Candida are of extreme importance due to their high frequency of colonization and infection in humans. The objective of this review is to describe the development of naphthoquinones as antifungals for the treatment of Candida species and to note the most promising compounds. By using certain criteria for selection of publications, 68 reports involving both synthetic and natural naphthoquinones are discussed. The activities of a large number of substances were evaluated against Candida albicans as well as against 7 other species of the genus Candida. The results discussed in this review allowed the identification of 30 naphthoquinones with higher antifungal activities than those of the currently used drugs.
Collapse
Affiliation(s)
- Débora O Futuro
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Patricia G Ferreira
- PPGCAPS, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Caroline D Nicoletti
- PPGCAPS, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Luana P Borba-Santos
- Centro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernando C DA Silva
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Sonia Rozental
- Centro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Vitor Francisco Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| |
Collapse
|
42
|
Rational Discovery of (+) (S) Abscisic Acid as a Potential Antifungal Agent: a Repurposing Approach. Sci Rep 2018; 8:8565. [PMID: 29867091 PMCID: PMC5986790 DOI: 10.1038/s41598-018-26998-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Fungal infections are spreading widely worldwide, and the types of treatment are limited due to the lack of diverse therapeutic agents and their associated side effects and toxicity. The discovery of new antifungal classes is vital and critical. We discovered the antifungal activity of abscisic acid through a rational drug design methodology that included the building of homology models for fungal chorismate mutases and a pharmacophore model derived from a transition state inhibitor. Ligand-based virtual screening resulted in some hits that were filtered using molecular docking and molecular dynamic simulations studies. Both in silico methods and in vitro antifungal assays were used as tools to select and validate the abscisic acid repurposing. Abscisic acid inhibition assays confirmed the inhibitory effect of abscisic acid on chorismate mutase through the inhibition of phenylpyruvate production. The repositioning of abscisic acid, the well-known and naturally occurring plant growth regulator, as a potential antifungal agent because of its suggested action as an inhibitor to several fungal chorismate mutases was the main result of this work.
Collapse
|
43
|
Action mechanism of naphthofuranquinones against fluconazole-resistant Candida tropicalis strains evidenced by proteomic analysis: The role of increased endogenous ROS. Microb Pathog 2018; 117:32-42. [DOI: 10.1016/j.micpath.2017.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 10/09/2017] [Accepted: 12/05/2017] [Indexed: 01/12/2023]
|
44
|
Reyna-Beltrán E, Iranzo M, Calderón-González KG, Mondragón-Flores R, Labra-Barrios ML, Mormeneo S, Luna-Arias JP. The Candida albicans ENO1 gene encodes a transglutaminase involved in growth, cell division, morphogenesis, and osmotic protection. J Biol Chem 2018; 293:4304-4323. [PMID: 29386353 PMCID: PMC5868267 DOI: 10.1074/jbc.m117.810440] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
Candida albicans is an opportunistic fungus that is part of the normal microflora commonly found in the human digestive tract and the normal mucosa or skin of healthy individuals. However, in immunocompromised individuals, it becomes a serious health concern and a threat to their lives and is ranked as the leading fungal infection in humans worldwide. As existing treatments for this infection are non-specific or under threat of developing resistance, there is a dire necessity to find new targets for designing specific drugs to defeat this fungus. Some authors reported the presence of the transglutaminase activity in Candida and Saccharomyces, but its identity remains unknown. We report here the phenotypic effects produced by the inhibition of transglutaminase enzymatic activity with cystamine, including growth inhibition of yeast cells, induction of autophagy in response to damage caused by cystamine, alteration of the normal yeast division pattern, changes in cell wall, and inhibition of the yeast-to-mycelium transition. The latter phenomenon was also observed in the C. albicans ATCC 26555 strain. Growth inhibition by cystamine was also determined in other Candida strains, demonstrating the importance of transglutaminase in these species. Finally, we identified enolase 1 as the cell wall protein responsible for TGase activity. After studying the inhibition of enzymatic activities with anti-CaEno1 antibodies and through bioinformatics studies, we suggest that the enolase and transglutaminase catalytic sites are localized in different domains of the protein. The aforementioned data indicate that TGase/Eno1 is a putative target for designing new drugs to control C. albicans infection.
Collapse
Affiliation(s)
| | - María Iranzo
- the Department de Microbiologia i Ecologia, Facultad de Farmacia, Unidad de Microbiología, Universitat de València, 46100 Valencia, España
| | | | - Ricardo Mondragón-Flores
- Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), C.P. 07360, Ciudad de México, México and
| | | | - Salvador Mormeneo
- the Department de Microbiologia i Ecologia, Facultad de Farmacia, Unidad de Microbiología, Universitat de València, 46100 Valencia, España
| | | |
Collapse
|
45
|
Lazić J, Ajdačić V, Vojnovic S, Zlatović M, Pekmezovic M, Mogavero S, Opsenica I, Nikodinovic-Runic J. Bis-guanylhydrazones as efficient anti-Candida compounds through DNA interaction. Appl Microbiol Biotechnol 2018; 102:1889-1901. [PMID: 29330691 DOI: 10.1007/s00253-018-8749-3] [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: 08/25/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
Candida spp. are leading causes of opportunistic mycoses, including life-threatening hospital-borne infections, and novel antifungals, preferably aiming targets that have not been used before, are constantly needed. Hydrazone- and guanidine-containing molecules have shown a wide range of biological activities, including recently described excellent antifungal properties. In this study, four bis-guanylhydrazone derivatives (BG1-4) were generated following a previously developed synthetic route. Anti-Candida (two C. albicans, C. glabrata, and C. parapsilosis) minimal inhibitory concentrations (MICs) of bis-guanylhydrazones were between 2 and 15.6 μg/mL. They were also effective against preformed 48-h-old C. albicans biofilms. In vitro DNA interaction, circular dichroism, and molecular docking analysis showed the great ability of these compounds to bind fungal DNA. Competition with DNA-binding stain, exposure of phosphatidylserine at the outer layer of the cytoplasmic membrane, and activation of metacaspases were shown for BG3. This pro-apoptotic effect of BG3 was only partially due to the accumulation of reactive oxygen species in C. albicans, as only twofold MIC and higher concentrations of BG3 caused depolarization of mitochondrial membrane which was accompanied by the decrease of the activity of fungal mitochondrial dehydrogenases, while the activity of oxidative stress response enzymes glutathione reductase and catalase was not significantly affected. BG3 showed synergistic activity with amphotericin B with a fractional inhibitory concentration index of 0.5. It also exerted low cytotoxicity and the ability to inhibit epithelial cell (TR146) invasion and damage by virulent C. albicans SC5314. With further developments, BG3 may further progress in the antifungal pipeline as a DNA-targeting agent.
Collapse
Affiliation(s)
- Jelena Lazić
- Faculty of Chemistry, University of Belgrade, Studentski trg 16, P.O. Box 51, Belgrade, 11158, Serbia.,Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade, 11000, Serbia
| | - Vladimir Ajdačić
- Faculty of Chemistry, University of Belgrade, Studentski trg 16, P.O. Box 51, Belgrade, 11158, Serbia
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade, 11000, Serbia
| | - Mario Zlatović
- Faculty of Chemistry, University of Belgrade, Studentski trg 16, P.O. Box 51, Belgrade, 11158, Serbia
| | - Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, Jena, Germany
| | - Igor Opsenica
- Faculty of Chemistry, University of Belgrade, Studentski trg 16, P.O. Box 51, Belgrade, 11158, Serbia.
| | - Jasmina Nikodinovic-Runic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade, 11000, Serbia.
| |
Collapse
|
46
|
Discovery of simplified sampangine derivatives as novel fungal biofilm inhibitors. Eur J Med Chem 2018; 143:1510-1523. [DOI: 10.1016/j.ejmech.2017.10.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 09/28/2017] [Accepted: 10/15/2017] [Indexed: 01/15/2023]
|
47
|
FUTURO DÉBORAO, FERREIRA PATRICIAG, NICOLETTI CAROLINED, BORBA-SANTOS LUANAP, SILVA FERNANDOCDA, ROZENTAL SONIA, FERREIRA VITORFRANCISCO. The Antifungal Activity of Naphthoquinones: An Integrative Review. AN ACAD BRAS CIENC 2018. [DOI: 10.1590/0001-3765201820170815 pmid: 29873671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
48
|
Sun N, Li D, Zhang Y, Killeen K, Groutas W, Calderone R. Repurposing an inhibitor of ribosomal biogenesis with broad anti-fungal activity. Sci Rep 2017; 7:17014. [PMID: 29209049 PMCID: PMC5717060 DOI: 10.1038/s41598-017-17147-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/22/2017] [Indexed: 12/23/2022] Open
Abstract
The lack of new antifungal compounds with unique mechanisms of action is a concern for therapeutic management of patients. To identify inhibitors against human pathogenic fungi, we screened ~3000 compounds provided by the Developmental Therapeutics Program of NIH/NCI against a panel of pathogenic fungi including Candida species, Aspergillus fumigatus, and Cryptococcus neoformans. NSC319726 (a thiosemicarbazone) had broad antifungal activity in the range of 0.1–2.0 µg/ml and was also inhibitory to fluconazole-resistant isolates of Candida species. Synergy was demonstrated with NSC319726 and azoles, as well as caspofungin. The inhibitory concentration 50% (IC50) of NSC319726 was 35–800-fold higher than the Minimum Inhibitory Concentration 50% (MIC50 values), which indicates low compound toxicity to human cells in vitro. Transcriptome analysis of treated and untreated C. albicans using Gene Ontology (GO) revealed a large cluster of down regulated genes that encode translational proteins, especially those with ribosome biogenesis functions. As NSC319726 was first shown to have anti-cancer activity, its affects against human pathogenic fungi establish NSC319726 as a repurposed, off-patent compound that has potential antifungal activity. The minimal in vitro toxicity of lead optimized NSC319726 and its reasonable inhibitory activity against pathogens suggest advancing this compound to in vivo toxicity testing and protection studies against candidiasis.
Collapse
Affiliation(s)
- Nuo Sun
- Georgetown University Medical Center, Washington DC, 20057, USA
| | - Dongmei Li
- Georgetown University Medical Center, Washington DC, 20057, USA
| | - Yuhan Zhang
- Georgetown University Medical Center, Washington DC, 20057, USA
| | - Kyle Killeen
- Georgetown University Medical Center, Washington DC, 20057, USA
| | | | | |
Collapse
|
49
|
Jain T, Muktapuram PR, Pochampalli S, Sharma K, Pant G, Mitra K, Bathula SR, Banerjee D. Chain-length-specific anti-Candida activity of cationic lipo-oxazoles: a new class of quaternary ammonium compounds. J Med Microbiol 2017; 66:1706-1714. [DOI: 10.1099/jmm.0.000610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Tushar Jain
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow-226 031, U.P., India
- Academy of Scientific & Innovative Research, Chennai-600113, India
| | - Pratap Reddy Muktapuram
- Division of Natural Products Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad -500007, India
| | - Satyanarayana Pochampalli
- Academy of Scientific & Innovative Research, Chennai-600113, India
- Division of Natural Products Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad -500007, India
| | - Komal Sharma
- Division of Natural Products Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad -500007, India
| | - Garima Pant
- Electron Microscopy Unit, Sophisticated and Analytical Instruments Facility, CSIR-Central Drug Research Institute, Lucknow-226 031, U.P., India
| | - Kalyan Mitra
- Electron Microscopy Unit, Sophisticated and Analytical Instruments Facility, CSIR-Central Drug Research Institute, Lucknow-226 031, U.P., India
| | - Surendar Reddy Bathula
- Division of Natural Products Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad -500007, India
- Academy of Scientific & Innovative Research, Chennai-600113, India
| | - Dibyendu Banerjee
- Academy of Scientific & Innovative Research, Chennai-600113, India
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow-226 031, U.P., India
| |
Collapse
|
50
|
Clausen JD, Kjellerup L, Cohrt KO, Hansen JB, Dalby-Brown W, Winther AML. Elucidation of antimicrobial activity and mechanism of action by N-substituted carbazole derivatives. Bioorg Med Chem Lett 2017; 27:4564-4570. [PMID: 28893470 PMCID: PMC5609566 DOI: 10.1016/j.bmcl.2017.08.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/20/2017] [Accepted: 08/25/2017] [Indexed: 11/15/2022]
Abstract
Compounds belonging to a carbazole series have been identified as potent fungal plasma membrane proton adenosine triphophatase (H+-ATPase) inhibitors with a broad spectrum of antifungal activity. The carbazole compounds inhibit the adenosine triphosphate (ATP) hydrolysis activity of the essential fungal H+-ATPase, thereby functionally inhibiting the extrusion of protons and extracellular acidification, processes that are responsible for maintaining high plasma membrane potential. The compound class binds to and inhibits the H+-ATPase within minutes, leading to fungal death after 1-3h of compound exposure in vitro. The tested compounds are not selective for the fungal H+-ATPase, exhibiting an overlap of inhibitory activity with the mammalian protein family of P-type ATPases; the sarco(endo)plasmic reticulum calcium ATPase (Ca2+-ATPase) and the sodium potassium ATPase (Na+,K+-ATPase). The ion transport in the P-type ATPases is energized by the conversion of ATP to adenosine diphosphate (ADP) and phosphate and a general inhibitory mechanism mediated by the carbazole derivative could therefore be blocking of the active site. However, biochemical studies show that increased concentrations of ATP do not change the inhibitory activity of the carbazoles suggesting they act as allosteric inhibitors. Furthermore decreased levels of intracellular ATP would suggest that the compounds inhibit the H+-ATPase indirectly, but Candida albicans cells exposed to potent H+-ATPase-inhibitory carbazoles result in increased levels of intracellular ATP, indicating direct inhibition of H+-ATPase.
Collapse
Affiliation(s)
| | - Lasse Kjellerup
- Pcovery ApS, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark; Department of Plant and Environmental Sciences, University of Copenhagen, DK-1871 Frederiksberg, Denmark
| | | | | | | | | |
Collapse
|