1
|
Rai M, Ingle AP, Törős G, Prokisch J. Assessing the efficacy of carbon nanodots derived from curcumin on infectious diseases. Expert Rev Anti Infect Ther 2024:1-15. [PMID: 39317385 DOI: 10.1080/14787210.2024.2409401] [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: 05/01/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION The threat of new, emerging, and multidrug-resistant microbes is increasing which has created the necessity for new antimicrobials. In this regard, nanotechnology can be an alternative for the treatment of infectious microbes. Curcumin has been used since ancient times as antimicrobials; however, it has limitations due to its less aqueous solubility, bioavailability, and biocompatibility. This problem can be solved by curcumin-derived carbon nanodots, which are emerging antimicrobials of <10 nm size, water-soluble, biocompatible, less toxic, and fluorescent. AREAS COVERED The review discusses the application of curcumin-derived carbon nanodots against various pathogenic microbes including bacteria and dreaded viruses like SARS-CoV-2. In addition, the role of curcumin carbon nanodots in biolabelling of pathogenic microbes, mechanism of action, bioimaging, and therapy has been critically examined. EXPERT OPINION Carbon nanodots play an important role in combating pathogenic microbes by early diagnosis, bioimaging, nanocarrier for antimicrobial drugs, and therapy of infectious diseases. Curcumin carbon nanodots have already demonstrated their benefits of being water soluble, bioavailable, and biocompatible. However, more thorough research is needed to understand the efficacy and safety of curcumin carbon nanodots. In the future, curcumin-derived carbon nanodots can be used as alternative antimicrobial agents to fight microbial infections including multidrug-resistant microbes.
Collapse
Affiliation(s)
- Mahendra Rai
- Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, India
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
| | - Avinash P Ingle
- Biotechnology Centre, Department of Agricultural Botany, Dr. PDKV, Akola, India
| | - Gréta Törős
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
- School of Animal Husbandry, University of Debrecen, Debrecen, Hungary
| | - József Prokisch
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Williams CC, Gregory JB, Usher J. Understanding the clinical and environmental drivers of antifungal resistance in the One Health context. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001512. [PMID: 39475703 PMCID: PMC11524418 DOI: 10.1099/mic.0.001512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/12/2024] [Indexed: 11/02/2024]
Abstract
Antifungal drugs have had a tremendous impact on human health and the yields of crops. However, in recent years, due to usage both in a health setting and in agriculture, there has been a rapid emergence of antifungal drug resistance that has outpaced novel compound discovery. It is now globally recognized that new strategies to tackle fungal infection are urgently needed, with such approaches requiring the cooperation of both sectors and the development of robust antifungal stewardship rationales. In this review, we examine the current antifungal regimes in clinical and agricultural settings, focusing on two pathogens of importance, Candida auris and Aspergillus fumigatus, examining their drivers of antifungal resistance, the impact of dual-use azoles and the impact agricultural practices have on driving the emergence of resistance. Finally, we postulate that a One Health approach could offer a viable alternative to prolonging the efficacy of current antifungal agents.
Collapse
Affiliation(s)
- Catrin C. Williams
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Jack B. Gregory
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
3
|
Peng X, Kong Q, Wei Q, Guo S, Chen Q, Peng M, An B, Wang X, Zhang C, Sang H. Verapamil enhances the activity of Caspofungin against Cryptococcus neoformans, coinciding with inhibited Ca 2+/CN pathway and damage to cell wall integrity. Int J Antimicrob Agents 2024; 64:107303. [PMID: 39151646 DOI: 10.1016/j.ijantimicag.2024.107303] [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] [Received: 03/08/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVES Given the challenges posed by toxicity and drug resistance in the treatment of cryptococcal infections, we sought to explore the antifungal potential of verapamil (VER), a calcium channel blocker, against Cryptococcus neoformans (C. neoformans), and its potential synergy with antifungals, specifically caspofungin (CAS). MATERIALS AND METHODS In vitro and in vivo (Galleria mellonella) models were employed to assess VER's antifungal activity and its interaction with CAS. Mechanisms underlying the synergism were explored through analysis of cell wall integrity, membrane permeability, and gene expression related to the calcineurin pathway. Additionally, the influence of Ca2+ on chitin deacetylase activity was investigated. RESULTS VER exhibited a pronounced antifungal effect on C. neoformans and synergized with CAS, enhancing antifungal efficacy in Galleria mellonella. VER reduced chitosan content and disrupted cell wall integrity, evidenced by melanin leakage and fluorescence staining. VER+CAS modified membrane permeability, triggering intracellular ROS accumulation and mitochondrial membrane potential alterations. VER mitigated CAS-induced calcium fluctuations and downregulated calcineurin pathway genes. Furthermore, it was found that the enzyme activity of chitin deacetylase of C. neoformans is significantly influenced by the presence of Ca2+, suggesting that the use of VER may affect this activity. CONCLUSIONS The synergistic antifungal effect of VER and CAS represents a promising therapeutic strategy for cryptococcal infections. The multifaceted mechanisms, including disruption of cell wall integrity and modulation of membrane permeability, and regulation of intracellular calcium signaling pathways, offer new insights into antifungal drug development.
Collapse
Affiliation(s)
- Xinyuan Peng
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingtao Kong
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qian Wei
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shilin Guo
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiying Chen
- Department of Dermatology, the First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Min Peng
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Binyi An
- Department of Dermatology, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Wang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chen Zhang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Sang
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Liu F, Chen Y, Huang Y, Jin Q, Ji J. Nanomaterial-based therapeutics for enhanced antifungal therapy. J Mater Chem B 2024; 12:9173-9198. [PMID: 39192670 DOI: 10.1039/d4tb01484g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The application of nanotechnology in antifungal therapy is gaining increasing attention. Current antifungal drugs have significant limitations, such as severe side effects, low bioavailability, and the rapid development of resistance. Nanotechnology offers an innovative solution to address these issues. This review discusses three key strategies of nanotechnology to enhance antifungal efficacy. Firstly, nanomaterials can enhance their interaction with fungal cells via ingenious surface tailoring of nanomaterials. Effective adhesion of nanoparticles to fungal cells can be achieved by electrostatic interaction or specific targeting to the fungal cell wall and cell membrane. Secondly, stimuli-responsive nanomaterials are developed to realize smart release of drugs in the specific microenvironment of pathological tissues, such as the fungal biofilm microenvironment and inflammatory microenvironment. Thirdly, nanomaterials can be designed to cross different physiological barriers, effectively addressing challenges posed by skin, corneal, and blood-brain barriers. Additionally, some new nanomaterial-based strategies in treating fungal infections are discussed, including the development of fungal vaccines, modulation of macrophage activity, phage therapy, the application of high-throughput screening in drug discovery, and so on. Despite the challenges faced in applying nanotechnology to antifungal therapy, its significant potential and innovation open new possibilities for future clinical antifungal applications.
Collapse
Affiliation(s)
- Fang Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China
| |
Collapse
|
5
|
Wang J, Su L, Li Y, Liu Y, Xie L. Nanoscale fluconazole-constructed metal-organic frameworks with smart drug release for eradication of Candida biofilms in vulvovaginitis infection. Colloids Surf B Biointerfaces 2024; 245:114238. [PMID: 39270401 DOI: 10.1016/j.colsurfb.2024.114238] [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: 06/10/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Fungal infections associated with oral, gynecological, and skin ailments pose significant clinical challenges. The presence of biofilms often hampers the efficacy of conventional antifungal drugs owing to the complex microenvironment they create. In this study, the widely used antifungal medication fluconazole is utilized as a foundational component to be incorporated into zinc 2-methylimidazolate frameworks, resulting in the synthesis of nanoscale fluconazole-constructed metal-organic frameworks (F-ZIF). The F-ZIF is constructed through coordination interactions between zinc and fluconazole, retaining the structure and pH-responsiveness of the zinc 2-methylimidazolate framework. The pH-responsiveness F-ZIF makes sure the fluconazole can be released in acidic biofilm, which prevents the undesired release in healthy tissue, resulting in good biocompatibility both in vitro and in vivo. The in vitro studies demonstrated that F-ZIF exhibits enhanced efficacy in eradicating fungal pathogens in their biofilm growth state compared with the free fluconazole. Furthermore, in vivo experiments reveal the better effectiveness of F-ZIF in treating Candida albicans-induced vulvovaginal candidiasis, and less infection-related inflammation was observed. Hence, the one-port synthetic F-ZIF presents a promising solution for addressing fungal biofilm-related infections.
Collapse
Affiliation(s)
- Jinhui Wang
- The People's Hospital of Yuhuan, Yuhuan, Zhejiang 317600, China
| | - Linzhu Su
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Yuanfeng Li
- Translational Medicine Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Zhejiang 325001, China.
| | - Lingping Xie
- The People's Hospital of Yuhuan, Yuhuan, Zhejiang 317600, China.
| |
Collapse
|
6
|
Gavandi T, Patil S, Basrani S, Yankanchi S, Chougule S, Karuppayil SM, Jadhav A. MIG1, TUP1 and NRG1 mediated yeast to hyphal morphogenesis inhibition in Candida albicans by ganciclovir. Braz J Microbiol 2024; 55:2047-2056. [PMID: 38789908 PMCID: PMC11405576 DOI: 10.1007/s42770-024-01344-8] [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: 11/06/2023] [Accepted: 04/09/2024] [Indexed: 05/26/2024] Open
Abstract
Candida albicans is a polymorphic human fungal pathogen and the prime etiological agent responsible for candidiasis. The main two aspects of C. albicans virulence that have been suggested are yeast-to-hyphal (Y-H) morphological transitions and biofilm development. Anti-fungal agents targeting these virulence attributes enhances the antifungal drug development process. Repositioning with other non-fungal drugs offered a one of the new strategies and a potential alternative option to counter the urgent need for antifungal drug development. In the current study, an antiviral drug ganciclovir was screened as an antifungal agent against ATCC 90028, 10231 and clinical isolate (C1). Ganciclovir at 0.5 mg/ml concentration reduced 50% hyphal development on a silicon-based urinary catheter and was visualized using scanning electron microscopy. Ganciclovir reduced ergosterol biosynthesis in both strains and C1 isolate of C. albicans in a concentration-dependent manner. Additionally, a gene expression profile study showed that ganciclovir treatment resulted in upregulation of hyphal-specific repressors MIG1, TUP1, and NRG1 in C. albicans. Additionally, an in vivo study on the Bombyx mori silkworm model further evidenced the virulence inhibitory ability of ganciclovir (0.5 mg/ml) against C. albicans. This is the first report that explore the novel anti-morphogenic activities of ganciclovir against the pathogenic C. albicans strains, along with clinical isolates. Further, ganciclovir may be considered for therapeutic purpose after combinations with standard antifungal agents.
Collapse
Affiliation(s)
- Tanjila Gavandi
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India
| | - Shivani Patil
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India
| | - Sargun Basrani
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India
| | - Shivanand Yankanchi
- Department of Zoology, Shivaji University, Vidya Nagar, 416004, Kolhapur, Maharashtra, India
| | - Sayali Chougule
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India
| | - S Mohan Karuppayil
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India.
| | - Ashwini Jadhav
- Department of Stem cell and Regenerative Medicine, Medical Biotechnology, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), 416006, Kolhapur, Maharashtra, India.
| |
Collapse
|
7
|
Sarkar R, Adhikary K, Banerjee A, Ganguly K, Sarkar R, Mohanty S, Dhua R, Bhattacharya K, Ahuja D, Pal S, Maiti R. Novel targets and improved immunotherapeutic techniques with an emphasis on antimycosal drug resistance for the treatment and management of mycosis. Heliyon 2024; 10:e35835. [PMID: 39224344 PMCID: PMC11367498 DOI: 10.1016/j.heliyon.2024.e35835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Infections due to pathogenic fungi are endemic in particular area with increased morbidity and mortality. More than a thousand people are infected per year and the way of treatment is of high demand having a significant impact on the population health. Medical practitioners confront various troublesome analytic and therapeutical challenges in the administration of immunosuppressed sufferer at high danger of expanding fungal infections. An upgraded antimycosal treatment is fundamental for a fruitful result while treating intrusive mycoses. A collection of antimycosal drugs keeps on developing with their specific antifungal targets including cell membrane, mitochondria, cell wall, and deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) or protein biosynthesis. Some fundamental classes of ordinarily directed medications are the polyenes, amphotericin B, syringomycin, allylamines, honokiol, azoles, flucytosine, echinocandins etc. However, few immunotherapy processes and vaccinations are being developed to mark this need, although one presently can't seem to arrive at the conclusion. In this review article, there has been a trial to give details upgradation about the current immune therapeutic techniques and vaccination strategies against prevention or treatment of mycosis as well as the difficulties related with their turn of events. There has been also a visualization in the mentioned review paper about the various assorted drugs and their specific target analysis along with therapeutic interventions.
Collapse
Affiliation(s)
- Riya Sarkar
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Adhikary
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Arundhati Banerjee
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Ganguly
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Riya Sarkar
- Department of Medical Laboratory Technology, Dr. B. C. Roy Academy of Professional Courses, Durgapur, West Bengal, 713206, India
| | - Satyajit Mohanty
- Department of Advanced Pharmacology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rumpa Dhua
- Department of Nutrition, Bankura Sammilani College, Kenduadihi, Bankura, West Bengal, 722102, India
| | - Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Deepika Ahuja
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Suchandra Pal
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, 713209, India
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, 722101, India
| |
Collapse
|
8
|
Honorato L, Bonilla JJA, Valdez AF, Frases S, Araújo GRDS, Sabino ALRDN, da Silva NM, Ribeiro L, Ferreira MDS, Kornetz J, Rodrigues ML, Cunningham I, Gow NAR, Gacser A, Guimarães AJ, Dutra FF, Nimrichter L. Toll-like receptor 4 (TLR4) is the major pattern recognition receptor triggering the protective effect of a Candida albicans extracellular vesicle-based vaccine prototype in murine systemic candidiasis. mSphere 2024; 9:e0046724. [PMID: 39037263 PMCID: PMC11351041 DOI: 10.1128/msphere.00467-24] [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: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic candidiasis remains a significant public health concern worldwide, with high mortality rates despite available antifungal drugs. Drug-resistant strains add to the urgency for alternative therapies. In this context, vaccination has reemerged as a prominent immune-based strategy. Extracellular vesicles (EVs), nanosized lipid bilayer particles, carry a diverse array of native fungal antigens, including proteins, nucleic acids, lipids, and glycans. Previous studies from our laboratory demonstrated that Candida albicans EVs triggered the innate immune response, activating bone marrow-derived dendritic cells (BMDCs) and potentially acting as a bridge between innate and adaptive immunity. Vaccination with C. albicans EVs induced the production of specific antibodies, modulated cytokine production, and provided protection in immunosuppressed mice infected with lethal C. albicans inoculum. To elucidate the mechanisms underlying EV-induced immune activation, our study investigated pathogen-associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) involved in EVs-phagocyte engagement. EVs from wild-type and mutant C. albicans strains with truncated mannoproteins were compared for their ability to stimulate BMDCs. Our findings revealed that EV decoration with O- and N-linked mannans and the presence of β-1,3-glucans and chitin oligomers may modulate the activation of specific PRRs, in particular Toll-like receptor 4 (TLR4) and dectin-1. The protective effect of vaccination with wild-type EVs was found to be dependent on TLR4. These results suggest that fungal EVs can be harnessed in vaccine formulations to selectively activate PRRs in phagocytes, offering potential avenues for combating or preventing candidiasis.IMPORTANCESystemic candidiasis is a serious global health concern with high mortality rates and growing drug resistance. Vaccination offers a promising solution. A unique approach involves using tiny lipid-coated particles called extracellular vesicles (EVs), which carry various fungal components. Previous studies found that Candida albicans EVs activate the immune response and may bridge the gap between innate and adaptive immunity. To understand this better, we investigated how these EVs activate immune cells. We demonstrated that specific components on EV surfaces, such as mannans and glucans, interact with receptors on immune cells, including Toll-like receptor 4 (TLR4) and dectin-1. Moreover, vaccinating with these EVs led to strong immune responses and full protection in mice infected with Candida. This work shows how harnessing fungal EVs might lead to effective vaccines against candidiasis.
Collapse
Affiliation(s)
- Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jhon J. Artunduaga Bonilla
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F. Valdez
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Glauber Ribeiro de Sousa Araújo
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natalia Martins da Silva
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Ribeiro
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Julio Kornetz
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio L. Rodrigues
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Carlos Chagas (ICC), Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
| | - Iain Cunningham
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Attila Gacser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Allan J. Guimarães
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Fabianno F. Dutra
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Elfadil A, Ibrahem K. Antifungal Activity of 3-Hydrazinoquinoxaline-2-Thiol, a Novel Quinoxaline Derivative against Candida Species. MYCOBIOLOGY 2024; 52:191-200. [PMID: 38948451 PMCID: PMC11210417 DOI: 10.1080/12298093.2024.2362497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
Candida ranks as among the most frequently encountered fungal infections that associated with high morbidity and mortality. Quinoxaline derivatives are a group of small molecules that showed a promising antimicrobial activity. This study aimed to investigate the fungicidal effects of 3-hydrazinoquinoxaline-2-thiol against Candida in comparison with Amphotericin B in vitro as a reference. Also, we aim to assess the efficacy of 3-hydrazinoquinoxaline-2-thiol in vivo using mice oral candidiasis model. Fifty-six Candida isolates were subjected to susceptibility testing by broth microdilution method for 3-hydrazinoquinoxaline-2-thiol and Amphotericin B. Therefore, Minimal inhibitory concentrations (MIC) were assessed and compared. The oral candidiasis mice model was used to evaluate the activity of 3-hydrazinoquinoxaline-2-thiol in vivo. Microbiological evaluation of progression and ELISA were used in this study. 3-hydrazinoquinoxaline-2-thiol was more effective than Amphotericin B against most clinical isolates of Candida albicans. Higher effectiveness was seen against Candida glabrata and Candida parapsilosis isolates. However, the efficiency against Candida tropicalis isolates varies. 3-hydrazinoquinoxaline-2-thiol was also effective against Pichia kudriavzevii and Clavispora lusitaniae. 3-hydrazinoquinoxaline-2-thiol showed a good efficacy in mice model against C. albicans cells ATCC 10231. 3-hydrazinoquinoxaline-2-thiol has shown promising antifungal and anti-inflammatory activity against different Candida species. More tests and experiments are needed.
Collapse
Affiliation(s)
- Abdelbagi Elfadil
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Karem Ibrahem
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Bazuhair MA, Alsieni M, Abdullah H, Mokhtar JA, Attallah D, Abujamel TS, Alkuwaity KK, Niyazi HA, Niyazi HA, AbdulMajed H, Juma N, Al-Rabia MW, Alfadil A, Ibrahem K. The Combination of 3-Hydrazinoquinoxaline-2-Thiol with Thymoquinone Demonstrates Synergistic Activity Against Different Candida Strains. Infect Drug Resist 2024; 17:2289-2298. [PMID: 38860227 PMCID: PMC11164204 DOI: 10.2147/idr.s464287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024] Open
Abstract
Introduction Candida is the primary cause of invasive fungal disease, candidiasis, especially in developed nations. The increasing resistance observed in multiple antibiotics, coupled with the prolonged process of creating new antibiotics from the ground up, emphasizes the urgent requirement for innovative methods and new compounds to combat Candida infections. Employing a treatment strategy that combines antibiotics can improve efficacy, broaden the spectrum of targeted fungal, and reduce the chances of resistance emergence. This approach shows potential in tackling the escalating problem of antibiotic resistance. The objective of this research is to explore the potential synergistic effects of combining 3-hydrazinoquinoxaline-2-thiol and thymoquinone against a variety of Candida isolates. This investigation aims to offer an understanding of the collective antimicrobial action of these compounds. Methods Broth microdilution was utilized to assess the Minimum Inhibitory Concentrations (MICs) of 3-hydrazinoquinoxaline-2-thiol and thymoquinone for 22 clinical Candida isolates. Following this, a checkerboard assay was employed to analyze the interaction between 3-hydrazinoquinoxaline-2-thiol and thymoquinone, with a specific focus on the Fractional Inhibitory Concentration Index (FICI). Results The MICs of thymoquinone and 3-hydrazinoquinoxaline-2-thiol were determined for 22 clinical Candida strains, with thymoquinone exhibiting MICs ranging from 64 to 8 µg/mL, and 3-hydrazinoquinoxaline-2-thiol displaying MICs varying from 64 to 8 µg/mL. Notably, the combination of 3-hydrazinoquinoxaline-2-thiol and thymoquinone resulted in a synergistic effect, leading to a significant reduction in MICs, with reductions of up to 64-fold with FICI below 0.5 against tested strains. Conclusion The prospect of using 3-hydrazinoquinoxaline-2-thiol in combination with thymoquinone as an effective solution against Candida looks encouraging. Nevertheless, to validate its practical applicability, additional comprehensive testing and experiments are imperative.
Collapse
Affiliation(s)
- Mohammed A Bazuhair
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Centre of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Alsieni
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hani Abdullah
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jawahir A Mokhtar
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology Laboratory, King Abdulaziz University Hospital, Jeddah, 21589, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Dalya Attallah
- Department of Clinical Microbiology Laboratory, King Abdulaziz University Hospital, Jeddah, 21589, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Khalil K Alkuwaity
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hanouf A Niyazi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hatoon A Niyazi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hind AbdulMajed
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noha Juma
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed W Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelbagi Alfadil
- Centre of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Karem Ibrahem
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Balaes T, Marandis CG, Mangalagiu V, Glod M, Mangalagiu II. New insides into chimeric and hybrid azines derivatives with antifungal activity. Future Med Chem 2024; 16:1163-1180. [PMID: 38916566 PMCID: PMC11216630 DOI: 10.1080/17568919.2024.2351288] [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] [Received: 02/22/2024] [Accepted: 04/25/2024] [Indexed: 06/26/2024] Open
Abstract
During the last decades, five or six member rings azaheterocycles compounds appear to be an extremely valuable source of antifungal agents. Their use seems to be a very attractive solution in antifungal therapy and to overcome antifungal resistance in agriculture. The present review highlights the main results obtained in the field of hybrid and chimeric azine (especially pyridine, quinoline, phenanthroline, bypyridine, naphthyridine and their fused derivatives) derivatives presented in scientific literature from the last 10 years, with emphasis on antifungal activity of the mentioned compounds. A special attention was played to hybrid and chimeric azole-azine class, having in view the high antifungal potential of azoles.
Collapse
Affiliation(s)
- Tiberius Balaes
- Alexandru Ioan Cuza University of Iasi, Department of Biology, Faculty of Biology, 20A Carol 1st Bvd, Iasi, 700505, Romania
| | | | - Violeta Mangalagiu
- Alexandru Ioan Cuza University of Iasi, Institute of Interdisciplinary Research-CERNESIM center, 11 Carol I, Iasi700506, Romania
- Stefan Cel Mare University of Suceava, Faculty of Food Engineering, 13 Universitatii Str., Suceava720229, Romania
| | - Mihai Glod
- Grigore T. Popa University of Medicine & Pharmacy, Clinical Hospital CF Iasi, Universității 16 Str., Iasi700115, Romania
| | - Ionel I Mangalagiu
- Alexandru Ioan Cuza University of Iasi, Faculty of Chemistry, 11 Carol 1st Bvd, Iasi700506, Romania
| |
Collapse
|
12
|
Ahlawat V, Sura K, Singh B, Dangi M, Chhillar AK. Bioinformatics Approaches in the Development of Antifungal Therapeutics and Vaccines. Curr Genomics 2024; 25:323-333. [PMID: 39323620 PMCID: PMC11420568 DOI: 10.2174/0113892029281602240422052210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 09/27/2024] Open
Abstract
Fungal infections are considered a great threat to human life and are associated with high mortality and morbidity, especially in immunocompromised individuals. Fungal pathogens employ various defense mechanisms to evade the host immune system, which causes severe infections. The available repertoire of drugs for the treatment of fungal infections includes azoles, allylamines, polyenes, echinocandins, and antimetabolites. However, the development of multidrug and pandrug resistance to available antimycotic drugs increases the need to develop better treatment approaches. In this new era of -omics, bioinformatics has expanded options for treating fungal infections. This review emphasizes how bioinformatics complements the emerging strategies, including advancements in drug delivery systems, combination therapies, drug repurposing, epitope-based vaccine design, RNA-based therapeutics, and the role of gut-microbiome interactions to combat anti-fungal resistance. In particular, we focused on computational methods that can be useful to obtain potent hits, and that too in a short period.
Collapse
Affiliation(s)
- Vaishali Ahlawat
- Centre for Biotechnology, M.D. University, Rohtak, Haryana, India
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Kiran Sura
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Bharat Singh
- Department of Biotechnology and Central Research Cell, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana-133207, India
| | - Mehak Dangi
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | | |
Collapse
|
13
|
Silva NBS, Menezes RP, Gonçalves DS, Santiago MB, Conejo NC, Souza SL, Santos ALO, da Silva RS, Ramos SB, Ferro EAV, Martins CHG. Exploring the antifungal, antibiofilm and antienzymatic potential of Rottlerin in an in vitro and in vivo approach. Sci Rep 2024; 14:11132. [PMID: 38750088 PMCID: PMC11096346 DOI: 10.1038/s41598-024-61179-z] [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: 01/05/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Candida species have been responsible for a high number of invasive infections worldwide. In this sense, Rottlerin has demonstrated a wide range of pharmacological activities. Therefore, this study aimed to evaluate the antifungal, antibiofilm and antivirulence activity of Rottlerin in vitro against Candida spp. and its toxicity and antifungal activity in vivo. Rottlerin showed antifungal activity against all yeasts evaluated, presenting Minimum Inhibitory and Fungicidal Concentration (MIC and MFC) values of 7.81 to > 1000 µg/mL. Futhermore, it was able to significantly inhibit biofilm production, presenting Biofilm Inhibitory Concentration (MICB50) values that ranged from 15.62 to 250 µg/mL and inhibition of the cell viability of the biofilm by 50% (IC50) from 2.24 to 12.76 µg/mL. There was a considerable reduction in all hydrolytic enzymes evaluated, with emphasis on hemolysin where Rottlerin showed a reduction of up to 20%. In the scanning electron microscopy (SEM) analysis, Rottlerin was able to completely inhibit filamentation by C. albicans. Regarding in vivo tests, Rottlerin did not demonstrate toxicity at the therapeutic concentrations demonstrated here and was able to increase the survival of C. elegans larvae infected. The results herein presented are innovative and pioneering in terms of Rottlerin's multipotentiality against these fungal infections.
Collapse
Affiliation(s)
- Nagela Bernadelli Sousa Silva
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Ralciane Paula Menezes
- Technical School of Health (ESTES), Federal University of Uberlândia (UFU), Uberlândia, Brazil
| | - Daniela Silva Gonçalves
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Mariana Brentini Santiago
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Noemi Chagas Conejo
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Sara Lemes Souza
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Anna Lívia Oliveira Santos
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil
| | - Robinson Sabino da Silva
- Innovation Center in Salivary Diagnostic and Nanotheranostics, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlandia (UFU), Uberlândia, Brazil
| | - Salvador Boccaletti Ramos
- Department of Engineering and Exact Sciences, Faculty of Agricultural and Veterinary Sciences - Jaboticabal (FCAV), São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlandia, Uberlândia, Brazil
| | - Carlos Henrique Gomes Martins
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia (UFU), Av. Pará, 1720 - Umuarama, Uberlândia, 38405-320, Brazil.
| |
Collapse
|
14
|
Grayton QE, Conlon IL, Broberg CA, Schoenfisch MH. Impact of Nitric Oxide-Release Kinetics on Antifungal Activity. J Fungi (Basel) 2024; 10:308. [PMID: 38786663 PMCID: PMC11121837 DOI: 10.3390/jof10050308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Pathogenic fungi are an increasing health threat due to the rise in drug resistance. The limited number of antifungals currently available and growing incidence of multi-drug-resistant fungi has caused rising healthcare costs and a decreased quality of life for patients with fungal infections. Nitric oxide (NO) has previously been shown to act as an antimicrobial agent, albeit with a limited understanding of the effects of the NO-release kinetics against pathogenic fungi. Herein, the antifungal effects of four nitric oxide-releasing small molecules were studied against the pathogenic fungi Candida albicans, Candida auris, Cryptococcus neoformans, and Aspergillus fumigatus, to demonstrate the broad-spectrum antifungal activity of NO. A bolus dose of NO was found to eradicate fungi after 24 h, where nitric oxide donors with shorter half-lives achieved antifungal activity at lower concentrations and thus had wider selectivity indexes. Each NO donor was found to cause a severe surface destruction of fungi, and all NO donors exhibited compatibility with currently prescribed antifungals against several different fungi species.
Collapse
Affiliation(s)
- Quincy E. Grayton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Ivie L. Conlon
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Christopher A. Broberg
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Q.E.G.); (C.A.B.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
15
|
Zhou M, Peng J, Ren K, Yu Y, Li D, She X, Liu W. Divergent mitochondrial responses and metabolic signal pathways secure the azole resistance in Crabtree-positive and negative Candida species. Microbiol Spectr 2024; 12:e0404223. [PMID: 38442003 PMCID: PMC10986575 DOI: 10.1128/spectrum.04042-23] [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: 11/27/2023] [Accepted: 02/07/2024] [Indexed: 03/07/2024] Open
Abstract
Azole drugs are the main therapeutic drugs for invasive fungal infections. However, azole-resistant strains appear repeatedly in the environment, posing a major threat to human health. Several reports have shown that mitochondria are associated with the virulence of pathogenic fungi. However, there are few studies on the mechanisms of mitochondria-mediated azoles resistance. Here, we first performed mitochondrial proteomic analysis on multiple Candida species (Candida albicans, Nakaseomyces glabrata, Pichia kudriavzevii, and Candida auris) and analyzed the differentially expressed mitochondrial proteins (DEMPs) between azole-sensitive and azole-resistant Candida species. Subsequently, we performed Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, gene ontology analysis, and protein-protein interaction network analysis of DEMPs. Our results showed that a total of 417, 165, and 25 DEMPs were identified in resistant C. albicans, N. glabrata, and C. auris, respectively. These DEMPs were enriched in ribosomal biogenesis at cytosol and mitochondria, tricarboxylic acid cycle, glycolysis, transporters, ergosterol, and cell wall mannan biosynthesis. The high activations of these cellular activities, found in C. albicans and C. auris (at low scale), were mostly opposite to those observed in two fermenter species-N. glabrata and P. kudriavzevii. Several transcription factors including Rtg3 were highly produced in resistant C. albicans that experienced a complex I activation of mitochondrial electron transport chain (ETC). The reduction of mitochondrial-related activities and complex IV/V of ETC in N. glabrata and P. kudriavzevii was companying with the reduced proteins of Tor1, Hog1, and Snf1/Snf4.IMPORTANCECandida spp. are common organisms that cause a variety of invasive diseases. However, Candida spp. are resistant to azoles, which hinders antifungal therapy. Exploring the drug-resistance mechanism of pathogenic Candida spp. will help improve the prevention and control strategy and discover new targets. Mitochondria, as an important organelle in eukaryotic cells, are closely related to a variety of cellular activities. However, the role of mitochondrial proteins in mediating azole resistance in Candida spp. has not been elucidated. Here, we analyzed the mitochondrial proteins and signaling pathways that mediate azole resistance in Candida spp. to provide ideas and references for solving the problem of azole resistance. Our work may offer new insights into the connection between mitochondria and azoles resistance in pathogenic fungi and highlight the potential clinical value of mitochondrial proteins in the treatment of invasive fungal infections.
Collapse
Affiliation(s)
- Meng Zhou
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Jingwen Peng
- Department of Critical Care Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medicine School, Nanjing University, Nanjing, China
| | - Kun Ren
- Centers for pharmaceutical preparations, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yu Yu
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Dongmei Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Xiaodong She
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Weida Liu
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Ji L, Tan L, Shang Z, Li W, Mo X, Yang S, Yu G. Discovery of New Antimicrobial Metabolites in the Coculture of Medicinal Mushrooms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5247-5257. [PMID: 38425052 DOI: 10.1021/acs.jafc.3c09476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Bioactivity screening revealed that the antifungal activities of EtOAc extracts from coculture broths of Trametes versicolor SY630 with either Vanderbylia robiniophila SY341 or Ganoderma gibbosum SY1001 were significantly improved compared to that of monocultures. Activity-guided isolation led to the discovery of five aromatic compounds (1-5) from the coculture broth of T. versicolor SY630 and V. robiniophila SY341 and two sphingolipids (6 and 7) from the coculture broth of T. versicolor SY630 and G. gibbosum SY1001. Tramevandins A-C (1-3) and 17-ene-1-deoxyPS (6) are new compounds, while 1-deoxyPS (7) is a new natural product. Notably, compound 2 represents a novel scaffold, wherein the highly modified p-terphenyl bears a benzyl substituent. The absolute configurations of those new compounds were elucidated by X-ray diffraction, ECD calculations, and analysis of physicochemical constants. Compounds 1, 2, and 5-7 exhibited different degrees of antimicrobial activity, and the antifungal activities of compounds 6 and 7 against Candida albicans and Cryptococcus neoformans are comparable to those of fluconazole, nystatin, and sphingosine, respectively. Transcriptome analysis, propidium iodide staining, ergosterol quantification, and feeding assays showed that the isolated sphingolipids can extensively downregulate the late biosynthetic pathway of ergosterol in C. albicans, representing a promising mechanism to combat antibiotic-resistant fungi.
Collapse
Affiliation(s)
- Linwei Ji
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Lingling Tan
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Zhaomeng Shang
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Wanting Li
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Xuhua Mo
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Song Yang
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| | - Guihong Yu
- School of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, and Qingdao International Center on Microbes Utilizing Biogas, Qingdao Agricultural University, Qingdao 266109, Shandong Province, People's Republic of China
| |
Collapse
|
17
|
Váradi G, Bende G, Borics A, Dán K, Rákhely G, Tóth GK, Galgóczy L. Rational Design of Antifungal Peptides Based on the γ-Core Motif of a Neosartorya ( Aspergillus) fischeri Antifungal Protein to Improve Structural Integrity, Efficacy, and Spectrum. ACS OMEGA 2024; 9:7206-7214. [PMID: 38371770 PMCID: PMC10870298 DOI: 10.1021/acsomega.3c09377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/20/2024]
Abstract
Antifungal peptides offer promising alternative compounds for the treatment of fungal infections, for which new antifungal compounds are urgently needed. Constant and broad antifungal spectra of these peptides play essential roles in their reliable therapeutic application. It has been observed that rationally designed peptides using the evolutionarily conserved γ-core region (GXC-X3-9-C) of an antifungal protein from Neosartorya (Aspergillus) fischeri highly inhibit the growth of fungi. The cysteines in these peptides have free sulfhydryl groups, which allow cyclization and dimerization under oxidative conditions, thereby impairing antifungal efficacy. To overcome this problem, one or two cysteine residues were substituted by serines or S-tert-butyl was applied as a cysteine-protecting group. Furthermore, structural integrity and antifungal efficacy investigations before and after oxidative exposure revealed that substituting both cysteines with serines and S-tert-butylation helped maintain the structural integrity. However, it slightly decreased the antifungal efficacy against a yeast, Candida albicans. Interestingly, S-tert-butylation maintained the efficacy and could extend the antifungal activity to a mold, Aspergillus fumigatus. Usually, cyclization and dimerization did not influence the antifungal efficacy of most peptides. Additionally, hemolysis tests and Galleria mellonella toxicity model experiments indicated that none of the applied modifications made the peptides harmful to animals.
Collapse
Affiliation(s)
- Györgyi Váradi
- Department
of Medical Chemistry, University of Szeged, Szeged 6720, Hungary
| | - Gábor Bende
- Department
of Biotechnology, University of Szeged, Szeged 6726, Hungary
- Doctoral
School of Biology, University of Szeged, Szeged 6720, Hungary
| | - Attila Borics
- Institute
of Biochemistry, HUN-REN Biological Research
Centre, Szeged 6726, Hungary
| | - Kinga Dán
- Department
of Biotechnology, University of Szeged, Szeged 6726, Hungary
- Doctoral
School of Biology, University of Szeged, Szeged 6720, Hungary
| | - Gábor Rákhely
- Department
of Biotechnology, University of Szeged, Szeged 6726, Hungary
- Institute
of Biophysics, HUN-REN Biological Research
Centre, Szeged 6726, Hungary
| | - Gábor K. Tóth
- Department
of Medical Chemistry, University of Szeged, Szeged 6720, Hungary
- MTA-SZTE
Biomimetic Systems Research Group, University
of Szeged, Szeged 6720, Hungary
| | - László Galgóczy
- Department
of Biotechnology, University of Szeged, Szeged 6726, Hungary
- Institute
of Biochemistry, HUN-REN Biological Research
Centre, Szeged 6726, Hungary
| |
Collapse
|
18
|
Pintye A, Bacsó R, Kovács GM. Trans-kingdom fungal pathogens infecting both plants and humans, and the problem of azole fungicide resistance. Front Microbiol 2024; 15:1354757. [PMID: 38410389 PMCID: PMC10896089 DOI: 10.3389/fmicb.2024.1354757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024] Open
Abstract
Azole antifungals are abundantly used in the environment and play an important role in managing fungal diseases in clinics. Due to the widespread use, azole resistance is an emerging global problem for all applications in several fungal species, including trans-kingdom pathogens, capable of infecting plants and humans. Azoles used in agriculture and clinics share the mode of action and facilitating cross-resistance development. The extensive use of azoles in the environment, e.g., for plant protection and wood preservation, contributes to the spread of resistant populations and challenges using these antifungals in medical treatments. The target of azoles is the cytochrome p450 lanosterol 14-α demethylase encoded by the CYP51 (called also as ERG11 in the case of yeasts) gene. Resistance mechanisms involve mainly the mutations in the coding region in the CYP51 gene, resulting in the inadequate binding of azoles to the encoded Cyp51 protein, or mutations in the promoter region causing overexpression of the protein. The World Health Organization (WHO) has issued the first fungal priority pathogens list (FPPL) to raise awareness of the risk of fungal infections and the increasingly rapid spread of antifungal resistance. Here, we review the main issues about the azole antifungal resistance of trans-kingdom pathogenic fungi with the ability to cause serious human infections and included in the WHO FPPL. Methods for the identification of these species and detection of resistance are summarized, highlighting the importance of these issues to apply the proper treatment.
Collapse
Affiliation(s)
- Alexandra Pintye
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Renáta Bacsó
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
| | - Gábor M. Kovács
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
19
|
Shinohara T, Wada A, Abe M, Nakayama N, Nagi M, Miyazaki Y. A novel zinc-chelating compound has antifungal activity against a wide range of Candida species, including multidrug-resistant Candida auris. JAC Antimicrob Resist 2024; 6:dlad155. [PMID: 38384742 PMCID: PMC10880073 DOI: 10.1093/jacamr/dlad155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024] Open
Abstract
Objectives In recent years, the incidence of invasive fungal infections has increased, resulting in considerable morbidity and mortality, particularly among immunocompromised individuals. Potential challenges in treating these infections with the few existing antifungal agents highlight the urgency of developing new ones. Here, we evaluated six alkyl polyamine compounds (APCs), not previously reported as antifungal drugs to our knowledge, that could deprive fungi of essential transition metals. Methods The APC with confirmed antifungal activity against Candida spp. was analysed by using transcriptomics, followed by metal-addition experiments, mass spectrometric analyses and intracellular zinc quantification with a fluorescent probe. Results A cyclic APC with three pyridylmethyl groups, APC6, had high antifungal activity against a wide range of Candida species, including MDR Candida auris. We conclusively demonstrated that APC6 was able to capture zinc within fungal cells. APC6 not only exhibited activity against C. auris as a single agent but also enhanced the efficacy of an azole antifungal agent, voriconazole, in vitro and in vivo. APC6 disrupted the biofilms formed by Candida species. Conclusions This zinc-chelating compound has potential as an antifungal agent, and the control of zinc levels in Candida species could be a powerful approach to treating drug-resistant candidiasis.
Collapse
Affiliation(s)
- Takayuki Shinohara
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Akira Wada
- Center for Biosystems Dynamics Research, RIKEN, Kanagawa, Japan
| | - Masahiro Abe
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Nobuko Nakayama
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Minoru Nagi
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshitsugu Miyazaki
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
- Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
20
|
Vanzolini T, Magnani M. Old and new strategies in therapy and diagnosis against fungal infections. Appl Microbiol Biotechnol 2024; 108:147. [PMID: 38240822 PMCID: PMC10799149 DOI: 10.1007/s00253-023-12884-8] [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: 06/26/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024]
Abstract
Fungal infections represent a serious global health threat. The new emerging pathogens and the spread of different forms of resistance are now hardly challenging the tools available in therapy and diagnostics. With the commonly used diagnoses, fungal identification is often slow and inaccurate, and, on the other hand, some drugs currently used as treatments are significantly affected by the decrease in susceptibility. Herein, the antifungal arsenal is critically summarized. Besides describing the old approaches and their mechanisms, advantages, and limitations, the focus is dedicated to innovative strategies which are designed, identified, and developed to take advantage of the discrepancies between fungal and host cells. Relevant pathways and their role in survival and virulence are discussed as their suitability as sources of antifungal targets. In a similar way, molecules with antifungal activity are reported as potential agents/precursors of the next generation of antimycotics. Particular attention was devoted to biotechnological entities, to their novelty and reliability, to drug repurposing and restoration, and to combinatorial applications yielding significant improvements in efficacy. KEY POINTS: • New antifungal agents and targets are needed to limit fungal morbidity and mortality. • Therapeutics and diagnostics suffer of delays in innovation and lack of targets. • Biologics, drug repurposing and combinations are the future of antifungal treatments.
Collapse
Affiliation(s)
- Tania Vanzolini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy.
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| |
Collapse
|
21
|
Janeczko M, Kochanowicz E. Biochanin A Inhibits the Growth and Biofilm of Candida Species. Pharmaceuticals (Basel) 2024; 17:89. [PMID: 38256922 PMCID: PMC10818846 DOI: 10.3390/ph17010089] [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: 12/09/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The aim of this study was to investigate the antifungal activity of biochanin A (BCA) against planktonic growth and biofilms of six Candida species, including C. albicans, C. parapsilosis, C. glabrata, C. tropicalis, C. auris, and C. krusei. We applied various assays that determined (a) the antimicrobial effect on growth of Candida species, (b) the effect on formation of hyphae and biofilm, (c) the effect on the expression of genes related to hyphal growth and biofilm formation, (d) the influence on cell wall structure, and (e) the effect on cell membrane integrity and permeability. Moreover, disk diffusion tests were used to investigate the effect of a combination of BCA with fluconazole to assess their possible synergistic effect on drug-resistant C. albicans, C. glabrata, and C. auris. Our results showed that the BCA MIC50 values against Candida species ranged between 125 µg/mL and 500 µg/mL, and the MIC90 values were in a concentration range from 250 µg/mL to 1000 µg/mL. The treatment with BCA inhibited adhesion of cells, cell surface hydrophobicity (CSH), and biofilm formation and reduced hyphal growth in all the analyzed Candida species. Real-time qRT-PCR revealed that BCA down-regulated the expression of biofilm-specific genes in C. albicans. Furthermore, physical destruction of C. albicans cell membranes and cell walls as a result of the treatment with BCA was observed. The combination of BCA and fluconazole did not exert synergistic effects against fluconazole-resistant Candida.
Collapse
Affiliation(s)
- Monika Janeczko
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland;
| | | |
Collapse
|
22
|
Aderibigbe BA. Nanotherapeutics for the delivery of antifungal drugs. Ther Deliv 2024. [PMID: 38174574 DOI: 10.4155/tde-2023-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
The treatment of fungal infections is challenging with high death rates reported among immunocompromised patients. The currently available antifungals suffer from poor bioavailability and solubility, pharmacokinetics, and drug resistance, with limited cellular uptake. The clinical pipeline of new antifungals is dry. The incorporation of antifungal drugs into polymer-based nanocarriers to form nanotherapeutics is a promising approach to enhance the therapeutic outcomes of the available antifungal drugs. This review summarizes different polymer-based nanotherapeutics strategies that have been explored for the delivery of antifungals, resulting in enhanced therapeutic outcomes, such as improved pharmacokinetics, targeted/sustained delivery, prolonged drug circulation, retention of the drugs at the localized site of action, and overcoming drug resistance when compared with the free antifungal drugs.
Collapse
|
23
|
Ardakani R, Jia L, Matthews E, Thakur KT. Therapeutic advances in neuroinfectious diseases. Ther Adv Infect Dis 2024; 11:20499361241274246. [PMID: 39314743 PMCID: PMC11418331 DOI: 10.1177/20499361241274246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/05/2024] [Indexed: 09/25/2024] Open
Abstract
There have been several major advances in therapeutic options for the treatment of neurological infections over the past two decades. These advances encompass both the development of new antimicrobial therapies and the repurposing of existing agents for new indications. In addition, advances in our understanding of the host immune response have allowed for the development of new immunomodulatory strategies in the treatment of neurological infections. This review focuses on the key advances in the treatment of neurological infections, including viral, bacterial, fungal, and prion diseases, with a particular focus on immunomodulatory treatment options. This review also highlights the process by which clinicians can request access to therapeutic agents on a compassionate or emergency basis when they may not be commercially available. While many therapeutic advances have been achieved in the past several years, there remains a pressing need for the continued development of additional therapeutic agents in the treatment of neurological infections.
Collapse
Affiliation(s)
- Rumyar Ardakani
- Neuro-Infectious Diseases Group, Department of Neurology and Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lucy Jia
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Elizabeth Matthews
- Neuro-Infectious Diseases Group, Department of Neurology and Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kiran T. Thakur
- Department of Neurology, Columbia University Irving Medical Center, 177 Fort Washington Avenue, Milstein Hospital, 8GS-300, New York, NY 10032, USA
- Program in Neuroinfectious Diseases, Department of Neurology, Columbia University Irving Medical Center-New York Presbyterian Hospital
| |
Collapse
|
24
|
Kaur A, Sharma K, Sharma N, Aggarwal G. An Insight into the Repurposing of Phytoconstituents obtained from Delhi's Aravalli Biodiversity Park as Antifungal Agents. Infect Disord Drug Targets 2024; 24:e020224226666. [PMID: 38305295 DOI: 10.2174/0118715265282411240119061441] [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] [Received: 09/15/2023] [Revised: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
The global prevalence of fungal infections is alarming in both the pre- and post- COVID period. Due to a limited number of antifungal drugs, there are hurdles in treatment strategies for fungal infections due to toxic potential, drug interactions, and the development of fungal resistance. All the antifungal targets (existing and newer) and pipeline molecules showing promise against these targets are reviewed. The objective was to predict or repurpose phyto-based antifungal compounds based on a dual target inhibition approach (Sterol-14-α- demethylase and HSP-90) using a case study. In pursuit of repurposing the phytochemicals as antifungal agents, a team of researchers visited Aravalli Biodiversity Park (ABP), Delhi, India, to collect information on available medicinal plants. From 45 plants, a total of 1149 ligands were collected, and virtual screening was performed using Schrodinger Suite 2016 software to get 83 hits against both the target proteins: Sterol-14-α-demethylase and HSP-90. After analysis of docking results, ligands were selected based on their interaction against both the target proteins and comparison with respective standard ligands (fluconazole and ganetespib). We have selected Isocarthamidin, Quercetin and Boeravinone B based on their docking score and binding interaction against the HSP-90 (Docking Score -9.65, -9.22 and -9.21, respectively) and 14-α-demethylase (Docking Score -9.19, -10.76 and -9.74 respectively). The docking protocol was validated and MM/GBSA studies depicted better stability of selected three ligands (Isocarthamidin, Quercetin, Boeravinone B) complex as compared to standard complex. Further, MD simulation studies were performed using the Desmond (67) software package version 2018-4. All the findings are presented as a case study for the prediction of dual targets for the repurposing of certain phytochemicals as antifungal agents.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Delhi Pharmaceutical Sciences and Research University, M.B. Road, Sector III, Pushp Vihar, New Delhi, 110017, India
| | - Kalicharan Sharma
- Delhi Pharmaceutical Sciences and Research University, M.B. Road, Sector III, Pushp Vihar, New Delhi, 110017, India
| | - Neetika Sharma
- Delhi Pharmaceutical Sciences and Research University, M.B. Road, Sector III, Pushp Vihar, New Delhi, 110017, India
| | - Geeta Aggarwal
- Delhi Pharmaceutical Sciences and Research University, M.B. Road, Sector III, Pushp Vihar, New Delhi, 110017, India
| |
Collapse
|
25
|
Wu M, Xu X, Hu R, Chen Q, Chen L, Yuan Y, Li J, Zhou L, Feng S, Wang L, Chen S, Gu M. A Membrane-Targeted Photosensitizer Prevents Drug Resistance and Induces Immune Response in Treating Candidiasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207736. [PMID: 37875397 PMCID: PMC10724446 DOI: 10.1002/advs.202207736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Candida albicans (C. albicans), a ubiquitous polymorphic fungus in humans, causes different types of candidiasis, including oral candidiasis (OC) and vulvovaginal candidiasis (VVC), which are physically and mentally concerning and financially costly. Thus, developing alternative antifungals that prevent drug resistance and induce immunity to eliminate Candida biofilms is crucial. Herein, a novel membrane-targeted aggregation-induced emission (AIE) photosensitizer (PS), TBTCP-QY, is developed for highly efficient photodynamic therapy (PDT) of candidiasis. TBTCP-QY has a high molar absorption coefficient and an excellent ability to generate 1 O2 and •OH, entering the interior of biofilms due to its high permeability. Furthermore, TBTCP-QY can efficiently inhibit biofilm formation by suppressing the expression of genes related to the adhesion (ALS3, EAP1, and HWP1), invasion (SAP1 and SAP2), and drug resistance (MDR1) of C. albicans, which is also advantageous for eliminating potential fungal resistance to treat clinical infectious diseases. TBTCP-QY-mediated PDT efficiently targets OC and VVC in vivo in a mouse model, induces immune response, relieves inflammation, and accelerates the healing of mucosal defects to combat infections caused by clinically isolated fluconazole-resistant strains. Moreover, TBTCP-QY demonstrates excellent biocompatibility, suggesting its potential applications in the clinical treatment of OC and VVC.
Collapse
Affiliation(s)
- Ming‐Yu Wu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Xiaoyu Xu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Rui Hu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Qingrong Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Luojia Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Yuncong Yuan
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Jie Li
- Department of Medical Intensive Care UnitMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430070China
| | - Li Zhou
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Lianrong Wang
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Shi Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Meijia Gu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| |
Collapse
|
26
|
Li C, Meng Y, Li H, Du W, Gao X, Suo C, Gao Y, Ni Y, Sun T, Yang S, Lan T, Xin M, Ding C. Immunization with a heat-killed prm1 deletion strain protects the host from Cryptococcus neoformans infection. Emerg Microbes Infect 2023; 12:2244087. [PMID: 37526401 PMCID: PMC10431737 DOI: 10.1080/22221751.2023.2244087] [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] [Received: 01/17/2023] [Revised: 07/02/2023] [Accepted: 07/30/2023] [Indexed: 08/02/2023]
Abstract
Systemic infection with Cryptococcus neoformans, a dangerous and contagious pathogen found throughout the world, frequently results in lethal cryptococcal pneumonia and meningoencephalitis, and no effective treatments and vaccination of cryptococcosis are available. Here, we describe Prm1, a novel regulator of C. neoformans virulence. C. neoformans prm1Δ cells exhibit extreme sensitivity to various environmental stress conditions. Furthermore, prm1Δ cells show deficiencies in the biosynthesis of chitosan and mannoprotein, which in turn result in impairment of cell wall integrity. Treatment of mice with heat-killed prm1Δ cells was found to facilitate the host immunological defence against infection with wild-type C. neoformans. Further investigation demonstrated that prm1Δ cells strongly promote pulmonary production of interferon-γ, leading to activation of macrophage M1 differentiation and inhibition of M2 polarization. Therefore, our findings suggest that C. neoformans Prm1 may be a viable target for the development of anti-cryptococcosis medications and, cells lacking Prm1 represent a promising candidate for a vaccine.
Collapse
Affiliation(s)
- Chao Li
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Yang Meng
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Hailong Li
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Wei Du
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Xindi Gao
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Chenhao Suo
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Yiru Gao
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Yue Ni
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Tianshu Sun
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People’s Republic of China
- Department of Scientific Research, Central Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People’s Republic of China
| | - Sheng Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Tian Lan
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Meiling Xin
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, People’s Republic of China
| |
Collapse
|
27
|
Padaraju A, Dwivedi F, Kumar G. Microemulsions, nanoemulsions and emulgels as carriers for antifungal antibiotics. Ther Deliv 2023; 14:721-740. [PMID: 38014430 DOI: 10.4155/tde-2023-0076] [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: 11/29/2023] Open
Abstract
According to estimates, up to 25% of the world's population has fungal skin diseases, making them the most prevalent infectious disease. Several chemical classes of antifungal drugs are available to treat fungal infections. However, the major challenges of conventional formulations of antifungal drugs include poor pharmacokinetic profiles like solubility, low permeability, side effects and decreased efficacy. Novel drug delivery is a promising approach for overcoming pharmacokinetic limitations and increasing the effectiveness of antibiotics. In this review, we have shed light on microemulsions, nanoemulsions, and emulgels as novel drug delivery approaches for the topical delivery of antifungal antibiotics. We believe these formulations have potential translational value and could be developed for treating fungal infections in humans.
Collapse
Affiliation(s)
- Annapurna Padaraju
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education & Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Falguni Dwivedi
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education & Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education & Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| |
Collapse
|
28
|
David H, Solomon AP. Molecular association of Candida albicans and vulvovaginal candidiasis: focusing on a solution. Front Cell Infect Microbiol 2023; 13:1245808. [PMID: 37900321 PMCID: PMC10611527 DOI: 10.3389/fcimb.2023.1245808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/31/2023] Open
Abstract
Candida albicans-mediated vulvovaginal candidiasis (VVC) is a significant challenge in clinical settings, owing to the inefficacy of current antifungals in modulating virulence, development of resistance, and poor penetration into the biofilm matrix. Various predisposition factors are molecular drivers that lead to the dysbiosis of normal microflora of the vagina, upregulation of central metabolic pathways, morphogenesis, hyphal extension, adhesion, invasion, and biofilm formation leading to chronic infection and recurrence. Hence, it is crucial to understand the molecular mechanism behind the virulence pathways driven by those drivers to decode the drug targets. Finding innovative solutions targeting fungal virulence/biofilm may potentiate the antifungals at low concentrations without affecting the recurrence of resistance. With this background, the present review details the critical molecular drivers and associated network of virulence pathways, possible drug targets, target-specific inhibitors, and probable mode of drug delivery to cross the preclinical phase by appropriate in vivo models.
Collapse
Affiliation(s)
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
29
|
Sun B, Liu W, Wang Q, Liu Y, Yu S, Liu M, Han J. Design, Synthesis, and Activity Evaluation of Novel Dual-Target Inhibitors with Antifungal and Immunoregulatory Properties. J Med Chem 2023; 66:13007-13027. [PMID: 37705322 DOI: 10.1021/acs.jmedchem.3c00942] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Dual-target (CYP51/PD-L1) plays an important role in the process of fungal proliferation and immune suppression. A series of novel quinazoline compounds with dual-target inhibition function was constructed using the skeleton growth method, and their structures were synthesized, characterized, and evaluated. Among them, the perfected compounds (L11, L20, L21) were selected for further study, which exhibited remarkable biological activity against different fungal strains (MIC50, 0.25-2.0 μg/mL) in vitro. On the one hand, these compounds inhibited CYP51 activity, induced ROS aggregation, and mitochondrial damage; this ultimately caused fungal lysis and death. On the other hand, they also effectively activated the body's immune ability by blocking the interaction between PD-L1 and PD-1, slowed down the inflammatory reaction, and accelerated the recovery process of fungal infections.
Collapse
Affiliation(s)
- Bin Sun
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Wenxia Liu
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Qingpeng Wang
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Yating Liu
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Shuai Yu
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Min Liu
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| | - Jun Han
- Institute of BioPharmaceutical Research, Liaocheng University, 1 Hunan Road, Liaocheng 252000, PR China
| |
Collapse
|
30
|
Chen L, Zhang L, Xie Y, Wang Y, Tian X, Fang W, Xue X, Wang L. Confronting antifungal resistance, tolerance, and persistence: Advances in drug target discovery and delivery systems. Adv Drug Deliv Rev 2023; 200:115007. [PMID: 37437715 DOI: 10.1016/j.addr.2023.115007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
Human pathogenic fungi pose a serious threat to human health and safety. Unfortunately, the limited number of antifungal options is exacerbated by the continuous emergence of drug-resistant variants, leading to frequent drug treatment failures. Recent studies have also highlighted the clinical importance of other modes of fungal survival of antifungal treatment, including drug tolerance and persistence, pointing to the complexity of the fungal response to antifungal drugs. A lack of understanding of the fungal drug response has hampered the identification of new targets, the development of alternative antifungal strategies and the design of appropriate delivery systems. In this review we summarize recent advances in the study of antifungal resistance, tolerance and persistence, with an emphasis on promising drug targets and drug delivery systems that may yield important insights into the development of new or improved antifungal therapies against fungal infections.
Collapse
Affiliation(s)
- Lei Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lanyue Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuyan Xie
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yiting Wang
- College of Life Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Xiuyun Tian
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenxia Fang
- Institute of Biological Science and Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University; Peking University Ninth School of Clinical Medicine, Beijing 100038, China; Department of Respiratory and Critical Care, Weifang Medical College, 261053, Weifang, Shandong, China.
| | - Linqi Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Zhang Z, Le GNT, Ge Y, Tang X, Chen X, Ejim L, Bordeleau E, Wright GD, Burns DC, Tran S, Axerio-Cilies P, Wang YT, Dong M, Woolley GA. Isomerization of bioactive acylhydrazones triggered by light or thiols. Nat Chem 2023; 15:1285-1295. [PMID: 37308709 DOI: 10.1038/s41557-023-01239-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/12/2023] [Indexed: 06/14/2023]
Abstract
The acylhydrazone unit is well represented in screening databases used to find ligands for biological targets, and numerous bioactive acylhydrazones have been reported. However, potential E/Z isomerization of the C=N bond in these compounds is rarely examined when bioactivity is assayed. Here we analysed two ortho-hydroxylated acylhydrazones discovered in a virtual drug screen for modulators of N-methyl-D-aspartate receptors and other bioactive hydroxylated acylhydrazones with structurally defined targets reported in the Protein Data Bank. We found that ionized forms of these compounds, which are populated under laboratory conditions, photoisomerize readily and the isomeric forms have markedly different bioactivity. Furthermore, we show that glutathione, a tripeptide involved with cellular redox balance, catalyses dynamic E⇄Z isomerization of acylhydrazones. The ratio of E to Z isomers in cells is determined by the relative stabilities of the isomers regardless of which isomer was applied. We conclude that E/Z isomerization may be a common feature of the bioactivity observed with acylhydrazones and should be routinely analysed.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Giang N T Le
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yang Ge
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaowen Tang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China
| | - Linda Ejim
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Emily Bordeleau
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Darcy C Burns
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Susannah Tran
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Peter Axerio-Cilies
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Yu Tian Wang
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Mingxin Dong
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China.
| | - G Andrew Woolley
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Qin Y, Wang J, Lv Q, Han B. Recent Progress in Research on Mitochondrion-Targeted Antifungal Drugs: a Review. Antimicrob Agents Chemother 2023; 67:e0000323. [PMID: 37195189 PMCID: PMC10269089 DOI: 10.1128/aac.00003-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Fungal infections, which commonly occur in immunocompromised patients, can cause high morbidity and mortality. Antifungal agents act by disrupting the cell membrane, inhibiting nucleic acid synthesis and function, or inhibiting β-1,3-glucan synthase. Because the incidences of life-threatening fungal infections and antifungal drug resistance are continuously increasing, there is an urgent need for the development of new antifungal agents with novel mechanisms of action. Recent studies have focused on mitochondrial components as potential therapeutic drug targets, owing to their important roles in fungal viability and pathogenesis. In this review, we discuss novel antifungal drugs targeting mitochondrial components and highlight the unique fungal proteins involved in the electron transport chain, which is useful for investigating selective antifungal targets. Finally, we comprehensively summarize the efficacy and safety of lead compounds in clinical and preclinical development. Although fungus-specific proteins in the mitochondrion are involved in various processes, the majority of the antifungal agents target dysfunction of mitochondria, including mitochondrial respiration disturbance, increased intracellular ATP, reactive oxygen species generation, and others. Moreover, only a few drugs are under clinical trials, necessitating further exploration of possible targets and development of effective antifungal agents. The unique chemical structures and targets of these compounds will provide valuable hints for further exploiting new antifungals.
Collapse
Affiliation(s)
- Yulin Qin
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Jinxin Wang
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Quanzhen Lv
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Diniz-Lima I, da Fonseca LM, Dos Reis JS, Decote-Ricardo D, Morrot A, Previato JO, Previato LM, Freire-de-Lima CG, Freire-de-Lima L. Non-self glycan structures as possible modulators of cancer progression: would polysaccharides from Cryptococcus spp. impact this phenomenon? Braz J Microbiol 2023; 54:907-919. [PMID: 36840821 PMCID: PMC10235250 DOI: 10.1007/s42770-023-00936-0] [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] [Received: 11/26/2022] [Accepted: 02/15/2023] [Indexed: 02/26/2023] Open
Abstract
Invasive fungal infections (IFI) are responsible for a large number of annual deaths. Most cases are closely related to patients in a state of immunosuppression, as is the case of patients undergoing chemotherapy. Cancer patients are severely affected by the worrisome proportions that an IFI can take during cancer progression, especially in an already immunologically and metabolically impaired patient. There is scarce knowledge about strategies to mitigate cancer progression in these cases, beyond conventional treatment with antifungal drugs with a narrow therapeutic range. However, in recent years, ample evidence has surfaced describing the possible interferences that IFI may have both on the progression of pre-existing cancers and in the induction of newly transformed cells. The leading gambit for modulation of tumor progression comes from the ability of fungal virulence factors to modulate the host's immune system, since they are found in considerable concentrations in the tumor microenvironment during infection. In this context, cryptococcosis is of particular concern, since the main virulence factor of the pathogenic yeast is its polysaccharide capsule, which carries constituents with high immunomodulatory properties and cytotoxic potential. Therefore, we open a discussion on what has already been described regarding the progression of cryptococcosis in the context of cancer progression, and the possible implications that fungal glycan structures may take in both cancer development and progression.
Collapse
Affiliation(s)
- Israel Diniz-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Leonardo Marques da Fonseca
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Jhenifer Santos Dos Reis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Debora Decote-Ricardo
- Departamento de Microbiologia E Imunologia Veterinária, Instituto de Veterinária, Universidade Federal Rural Do Rio de Janeiro, Rio de Janeiro, 23890-000, Brazil
| | - Alexandre Morrot
- Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Jose Osvaldo Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Lucia Mendonça Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Celio Geraldo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
34
|
Du C, Yang X, Long Y, Lang X, Liu L, Xu Y, Wu H, Chu Y, Hu X, Deng J, Ji Q. Design, synthesis and biological evaluation of novel spiro-quinazolinone derivatives as chitin synthase inhibitors and antifungal agents. Eur J Med Chem 2023; 255:115388. [PMID: 37141707 DOI: 10.1016/j.ejmech.2023.115388] [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: 02/03/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 05/06/2023]
Abstract
A series of spiro-quinazolinone scaffolds were constructed based on the bioactivity of quinazolinone and the inherent feature of spirocycle to design novel chitin synthase inhibitors that possess mode of action different from that of the currently used antifungal agents. Among them, the spiro[thiophen-quinazolin]-one derivatives containing α, β-unsaturated carbonyl fragments had shown inhibitory activities against chitin synthase and antifungal activities. The enzymatic experiments showed that among the sixteen compounds, compounds 12d, 12g, 12j, 12l and 12m exhibited inhibitions against chitin synthase with IC50 values of 116.7 ± 19.6 μM, 106.7 ± 14.2 μM, 102.3 ± 9.6 μM, 122.7 ± 22.2 μM and 136.8 ± 12.4 μM, respectively, which were comparable to that of polyoxin B (IC50 = 93.5 ± 11.1 μM). The assays of enzymatic Kinetic parameters showed that compound 12g was a non-competitive inhibitor of chitin synthase. The antifungal assays showed that compounds 12d, 12g, 12j, 12l and 12m exhibited a broad-spectrum of antifungal activity against the four strains tested in vitro. In which, compounds 12g and 12j had stronger antifungal activity against four tested strains than that of polyoxin B and similar to that of fluconazole, while compounds 12d, 12l and 12m showed antifungal activity comparable to that of polyoxin B against four tested strains. Meanwhile, compounds 12d, 12g, 12j, 12l and 12m exhibited good antifungal activity against fluconazole-resistant and micafungin-resistant fungi variants with MIC values ranging from 4 to 32 μg/mL while the MIC values of reference drugs were above 256 μg/mL. Furthermore, the results of drug-combination experiments showed that compounds 12d, 12g, 12j, 12l and 12m had synergistic or additive effects with fluconazole or polyoxin B. The results of sorbitol protection experiment and the experiment of antifungal activity against micafungin-resistant fungi further demonstrated that these compounds target chitin synthase. The result of cytotoxicity assay showed that compound 12g had low toxicity toward human lung cancer A549 cells and the ADME analysis in silico displayed that compound 12g possessed promising pharmacokinetic properties. The molecular docking indicated that compound 12g formed multiple hydrogen bond interactions binding to chitin synthase, which might be conductive to increasing the binding affinity and inhibiting the activity of chitin synthase. The above results indicated that the designed compounds were chitin synthase inhibitors with selectivity and broad-spectrum antifungal activity and could be act as the lead compounds against drug-resistant fungi.
Collapse
Affiliation(s)
- Chuanbiao Du
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Xinlong Yang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yan Long
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Xueqing Lang
- Key Laboratory of Laboratory Medicine Diagnostics, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lige Liu
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yajie Xu
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Hu Wu
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, PR China
| | - Xiaolei Hu
- Key Laboratory of Laboratory Medicine Diagnostics, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Junfeng Deng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, PR China.
| | - Qinggang Ji
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
35
|
Lv Q, Yan L, Wang J, Feng J, Gao L, Qiu L, Chao W, Qin YL, Jiang Y. Combined Transcriptome and Metabolome Analysis Reveals That the Potent Antifungal Pyrylium Salt Inhibits Mitochondrial Complex I in Candida albicans. Microbiol Spectr 2023; 11:e0320922. [PMID: 36790175 PMCID: PMC10100848 DOI: 10.1128/spectrum.03209-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/06/2023] [Indexed: 02/16/2023] Open
Abstract
Based on the structural modification of SM21, xy12, a new pyrylium salt derivative with enhanced antifungal activities, was synthesized. The MICs (MIC90) of xy12 against Candida albicans ranged from 0.125 to 0.25 μg/mL, about 2-fold lower than those of SM21. In addition, xy12 inhibited hypha and biofilm formation in C. albicans in a dose-dependent manner. A total of 3,454 differentially expressed genes and 260 differential metabolites were identified in the xy12-treated C. albicans by RNA-seq and non-targeted metabolomics. By integrating KEGG pathway enrichment analysis, we found that inhibition of oxidative phosphorylation was the important antifungal mechanism of action of xy12. Electron transport through mitochondrial respiratory complexes I to IV is the common process of oxidative phosphorylation. Compared with the sensitivity of the wild-type SC5314 to xy12, decreased sensitivities in mitochondrial complex I (CI)-deficient mutants and increased sensitivities in mitochondrial complex III- and IV-deficient mutants suggested that the antifungal effects of xy12 were dependent on CI. Consistently, xy12 exhibited antagonism with rotenone, an inhibitor of CI, and significantly inhibited the expression and activity of CI. Meanwhile, the phenotypes in the xy12-treated C. albicans were similar to those in the CI-deficient mutants, such as decreased ATP production, reduced mitochondrial membrane potential, loss of mitochondrial DNA, inability to utilize nonfermentative carbon sources, and decreased cell wall N-linked mannoproteins. Collectively, our results revealed that the pyrylium salt xy12 could constrain oxidative phosphorylation by inhibiting mitochondrial complex I in C. albicans, providing a novel lead compound for the development of mitochondria-targeted antifungal drugs. IMPORTANCE The development of new antifungal drugs is critical for solving the problem of antifungal resistance and expanding the limited variety of clinical antifungal drugs. Based on the modification of the pyrylium salt SM21, a new lead compound, xy12, was synthesized which was effective against Candida species both in vitro and in vivo. In this study, conjoined analysis of the transcriptome and metabolome elucidated the antifungal mechanism of action of xy12, which inhibited the activity of mitochondrial complex I in C. albicans. Targeting fungi-specific mitochondrial complex proteins has been reported as a promising antifungal strategy. Our study provided a new lead compound for targeting C. albicans mitochondrial complex I, which could be beneficial for discovering novel antifungal drugs.
Collapse
Affiliation(s)
- Quanzhen Lv
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Lan Yan
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Jinxin Wang
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Jia Feng
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Lu Gao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Lijuan Qiu
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Wen Chao
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
| | - Yu-Lin Qin
- Fudan University Minhang Hospital, Shanghai, People’s Republic of China
| | - Yuanying Jiang
- School of Pharmacy, Naval Medical University, Shanghai, People’s Republic of China
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
36
|
Controlling antifungal activity with light: Optical regulation of fungal ergosterol biosynthetic pathway with photo-responsive CYP51 inhibitors. Acta Pharm Sin B 2023. [PMID: 37521860 PMCID: PMC10372832 DOI: 10.1016/j.apsb.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Invasive fungal infections (IFIs) have been associated with high mortality, highlighting the urgent need for developing novel antifungal strategies. Herein the first light-responsive antifungal agents were designed by optical control of fungal ergosterol biosynthesis pathway with photocaged triazole lanosterol 14α-demethylase (CYP51) inhibitors. The photocaged triazoles completely shielded the CYP51 inhibition. The content of ergosterol in fungi before photoactivation and after photoactivation was 4.4% and 83.7%, respectively. Importantly, the shielded antifungal activity (MIC80 ≥ 64 μg/mL) could be efficiently recovered (MIC80 = 0.5-8 μg/mL) by light irradiation. The new chemical tools enable optical control of fungal growth arrest, morphological conversion and biofilm formation. The ability for high-precision antifungal treatment was validated by in vivo models. The light-activated compound A1 was comparable to fluconazole in prolonging survival in Galleria mellonella larvae with a median survival of 14 days and reducing fungal burden in the mouse skin infection model. Overall, this study paves the way for precise regulation of antifungal therapy with improved efficacy and safety.
Collapse
|
37
|
Raza A, Parveen S, Majeed MI, Nawaz H, Javed MR, Iqbal MA, Rashid N, Haider MZ, Ali MZ, Sabir A, Mahmood Ul Hasan H, Majeed B. Surface-enhanced Raman spectral characterization of antifungal activity of selenium and zinc based organometallic compounds. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 285:121903. [PMID: 36209714 DOI: 10.1016/j.saa.2022.121903] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Surface-enhanced Raman spectroscopy (SERS) is used to identify the biochemical changes associated with the antifungal activities of selenium and zinc organometallic complexes against Aspergillus niger fungus. These biochemical changes identified in the form of SERS peaks can help to understand the mechanism of action of these antifungal agents which is important for development of new antifungal drugs. The SERS spectral changes indicate the denaturation and conformational changes of proteins and fungal cell wall decomposition in complex exposed fungal samples. The SERS spectra of these organometallic complexes exposed fungi are analyzed by using statistical tools like principal component analysis (PCA) and partial least square discriminant analysis (PLS-DA). PCA is employed to differentiate the SERS spectra of fungal samples exposed to ligands and complexes. The PLS-DA discriminated different groups of spectra with 99.8% sensitivity, 100% specificity, 98% accuracy and 86 % area under receiver operating characteristic (AUROC) curve.
Collapse
Affiliation(s)
- Ali Raza
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Soneya Parveen
- Medicine and Allied, Faisalabad Medical University, Faisalabad 38000, Pakistan
| | - Muhammad Irfan Majeed
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan.
| | - Haq Nawaz
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan.
| | - Muhammad Rizwan Javed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Muhammad Adnan Iqbal
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Nosheen Rashid
- Department of Chemistry, University of Education, Faisalabad Campus, Faisalabad 38000, Pakistan
| | | | - Muhammad Zeeshan Ali
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Amina Sabir
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Hafiz Mahmood Ul Hasan
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Beenish Majeed
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| |
Collapse
|
38
|
Yadav M, Kumar A, Lal K, Singh MB, Kumari K. Facile synthesis, antimicrobial screening and docking studies of pyrrole-triazole hybrids as potential antimicrobial agents. RESEARCH ON CHEMICAL INTERMEDIATES 2023. [DOI: 10.1007/s11164-022-04948-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
39
|
Mossion A, Ourliac-Garnier I, Wielgosz-Collin G. Fungal Sterol Analyses by Gas Chromatography-Mass Spectrometry Using Different Derivatives. Methods Mol Biol 2023; 2704:143-156. [PMID: 37642842 DOI: 10.1007/978-1-0716-3385-4_8] [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: 08/31/2023]
Abstract
Sterols are the main components of the fungal membrane. Their study can be used to describe the chemical biodiversity among the strains and species or to work on antifungal treatment. Those molecules can be analyzed by gas chromatography coupled with mass spectrometry (GC-MS) as free molecules or after derivation as acetate or trimethylsilyl ether (TMSi). Here we describe how to extract sterols from fungal biomass according to its physiological form and the culture conditions (liquid and solid media). The different methodologies that can be used to obtain free sterols, acetate, and/or TMSi derivatives are presented. Identification keys using the fragmentation at 70 eV are also described.
Collapse
Affiliation(s)
- Aurélie Mossion
- Nantes Université, Institut des Substances et Organismes de la Mer, ISOMer, Nantes, France.
| | - Isabelle Ourliac-Garnier
- Nantes Université, Cibles et Médicaments des Infections et de l'Immunité, IICiMed, Nantes, France
| | | |
Collapse
|
40
|
De Bels D, Maillart E, Van Bambeke F, Redant S, Honoré PM. Existing and emerging therapies for the treatment of invasive candidiasis and candidemia. Expert Opin Emerg Drugs 2022; 27:405-416. [PMID: 36317695 DOI: 10.1080/14728214.2022.2142207] [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: 11/15/2022]
Abstract
INTRODUCTION Invasive candidiasis or candidemia is a severe infection affecting more than 250,000 people worldwide every year. It is present in up to 16% of ICU patients. The prognosis of these infections is unfavorable, with global death estimated around 50,000 per year, which corresponds to up to 40% depending on patient severity and comorbidities. Therapeutic failure is not rare due to the emergence of multiresistant strains and of new species poorly responsive to current therapies like Candida auris. AREAS COVERED We first review the positioning of antifungal drugs used to treat candidiasis, namely polyenes, azoles, echinocandins and pyrimidine analogues. We then discuss the progresses brought by new formulations, new derivatives within these classes, compounds acting on new targets or repurposed drugs in terms of pharmacokinetic profile, spectrum of activity, potency, safety or risk of drug-drug interactions. EXPERT OPINION While new formulations (amphotericin B cochleate) improve oral bioavailability of the corresponding drugs, new azoles or echinocandins offer higher potency including against strains resistant to former generations of drugs. Repurposed drugs show synergism with current therapies in vitro. Results from ongoing and future clinical trials will be decisive to establish the interest for these drugs in our arsenal.
Collapse
Affiliation(s)
- David De Bels
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Evelyne Maillart
- Department of Infectious Disease, Brugmann University Hospital, Brussels, Belgium
| | - Françoise Van Bambeke
- Louvain Drug Research Institute, Department of Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Sebastien Redant
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Patrick M Honoré
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of ICU, CHU UCL Godinne-Namur, UCL Louvain Medical School
| |
Collapse
|
41
|
Pintea A, Vlad RA, Antonoaea P, Rédai EM, Todoran N, Barabás EC, Ciurba A. Structural Characterization and Optimization of a Miconazole Oral Gel. Polymers (Basel) 2022; 14:polym14225011. [PMID: 36433136 PMCID: PMC9692734 DOI: 10.3390/polym14225011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
The development of semisolid formulations, gels in particular, has raised the attention of scientists more and more over the last decades. Because of their biocompatibility, hydrophilic nature, and capacity of absorbing large quantities of water, hydrogels are still one of the most promising pharmaceutical formulations in the pharmaceutical industry. The purpose of this study is to develop an optimal formulation capable of incorporating a water-poorly soluble active ingredient such as miconazole used in the treatment of fungal infections with Candida albicans and Candida parapsilosis. A D-optimal design was applied to study the relationship between the formulation parameter and the gel characteristics. The independent parameters used in this study were the Carbopol 940 concentration (the polymer used to obtain the gel matrix), the sodium hydroxide amount, and the presence/absence of miconazole. Ten different dependent parameters (Y1-Y10) were evaluated (penetrometry, spreadability, viscosity, and tangential tension at 1 and 11 levels of speed whilst destructuring and during the reorganization of the gel matrix). The consistency of the gels ranged from 23.2 mm (GO2) to 29.6 mm (GM5). The least spreadable gel was GO7 (1384 mm2), whilst the gel that presented the best spreadability was GO1 (3525 mm2). The viscosity and the tangential stress at the selected levels (1 and 11) varied due to the different compositions of the proposed gels. The gels were also tested for drug content and antifungal activity. All determinations had satisfying results; the drug content was within limits accepted by Ph. Eur. 10 and all formulations containing miconazole exhibited antifungal activity. An optimal formulation with miconazole was attained, consisting of 0.84% Carbopol 940 and 0.32% sodium hydroxide.
Collapse
Affiliation(s)
- Andrada Pintea
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Robert-Alexandru Vlad
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Paula Antonoaea
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- Correspondence:
| | - Emöke Margit Rédai
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Nicoleta Todoran
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Enikő-Csilla Barabás
- Cellular Biology and Microbiology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- Department of Laboratory Medicine, Mures, County Hospital, 540136 Targu Mures, Romania
| | - Adriana Ciurba
- Pharmaceutical Technology and Cosmetology Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| |
Collapse
|
42
|
Fungal calcineurin complex as an antifungal target: From past to present to future. FUNGAL BIOL REV 2022. [DOI: 10.1016/j.fbr.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
43
|
Application of the Mutant Libraries for Candida albicans Functional Genomics. Int J Mol Sci 2022; 23:ijms232012307. [PMID: 36293157 PMCID: PMC9603287 DOI: 10.3390/ijms232012307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Candida albicans is a typical opportunistic pathogen in humans that causes serious health risks in clinical fungal infections. The construction of mutant libraries has made remarkable developments in the study of C. albicans molecular and cellular biology with the ongoing advancements of gene editing, which include the application of CRISPR-Cas9 and novel high-efficient transposon. Large-scale genetic screens and genome-wide functional analysis accelerated the investigation of new genetic regulatory mechanisms associated with the pathogenicity and resistance to environmental stress in C. albicans. More importantly, sensitivity screening based on C. albicans mutant libraries is critical for the target identification of novel antifungal compounds, which leads to the discovery of Sec7p, Tfp1p, Gwt1p, Gln4p, and Erg11p. This review summarizes the main types of C. albicans mutant libraries and interprets their applications in morphogenesis, biofilm formation, fungus-host interactions, antifungal drug resistance, and target identification.
Collapse
|
44
|
Fioriti S, Brescini L, Pallotta F, Canovari B, Morroni G, Barchiesi F. Antifungal Combinations against Candida Species: From Bench to Bedside. J Fungi (Basel) 2022; 8:jof8101077. [PMID: 36294642 PMCID: PMC9605143 DOI: 10.3390/jof8101077] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Candida spp. is the major causative agent of fungal infections in hospitalized patients and the fourth most common cause of nosocomial bloodstream infection (BSI). The availability of standardized methods for testing the in vitro activity of antifungals along with the expanding of antifungal armamentarium, the rising of drug-resistance and the persistence of a high mortality rate in systemic candidiasis have led to an increased interest in combination therapy. Therefore, we aimed to review the scientific literature concerning the antifungal combinations against Candida. A literature search performed in PubMed yielded 92 studies published from 2000 to 2021: 29 articles referring to in vitro studies, six articles referring to either in vitro and in vivo (i.e., animal models) studies and 57 clinical articles. Pre-clinical studies involved 735 isolates of Candida species and 12 unique types of antifungal combination approaches including azoles plus echinocandins (19%), polyenes plus echinocandins (16%), polyenes plus azoles (13%), polyenes plus 5-flucytosine ([5-FC], 13%), azoles plus 5-FC (11%) and other types of combinations (28%). Results varied greatly, often being species-, drug- and methodology-dependent. Some combinatorial regimens exerted a synergistic effect against difficult-to-treat Candida species (i.e., azoles plus echinocandins; polyenes plus 5-FC) or they were more effective than monotherapy in prevent or reducing biofilm formation and in speeding the clearance of infected tissues (i.e., polyenes plus echinocandins). In 283 patients with documented Candida infections (>90% systemic candidiasis/BSI), an antifungal combination approach could be evaluated. Combinations included: azoles plus echinocandins (36%), 5-FC-combination therapies (24%), polyenes plus azoles (18%), polyenes plus echinocandins (16%) and other types of combination therapy (6%). Case reports describing combination therapies yielded favorable response in most cases, including difficult-to-treat fungal infections (i.e., endocarditis, osteoarticular infections, CNS infections) or difficult-to-treat fungal pathogens. The only randomized trial comparing amphotericin-B deoxycholate (AMB) plus FLU vs. AMB alone for treatment of BSI in nonneutropenic patients showed that the combination trended toward improved success and more-rapid clearance from the bloodstream. In summary, antifungal combinations against Candida have produced great interest in the past two decades. To establish whether this approach can become a reliable treatment option, additional in vitro and clinical data are warranted.
Collapse
Affiliation(s)
- Simona Fioriti
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lucia Brescini
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
- Infectious Disease Clinic, Azienda Ospedaliero Universitaria “Ospedali Riuniti”, 60126 Ancona, Italy
| | - Francesco Pallotta
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
- Infectious Disease Clinic, Azienda Ospedaliero Universitaria “Ospedali Riuniti”, 60126 Ancona, Italy
| | - Benedetta Canovari
- Infectious Diseases Unit, Azienda Ospedaliera Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy
| | - Gianluca Morroni
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
- Correspondence: ; Tel.: +39-071-220-6298; Fax: +39-071-220-6297
| | - Francesco Barchiesi
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
- Infectious Diseases Unit, Azienda Ospedaliera Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy
| |
Collapse
|
45
|
da Silva Neto JX, Dias LP, Lopes de Souza LA, Silva da Costa HP, Vasconcelos IM, Pereira ML, de Oliveira JTA, Cardozo CJP, Gonçalves Moura LFW, de Sousa JS, Carneiro RF, Lopes TDP, Bezerra de Sousa DDO. Insights into the structure and mechanism of action of the anti-candidal lectin Mo-CBP2 and evaluation of its synergistic effect and antibiofilm activity. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Langfeldt A, Gold JAW, Chiller T. Emerging Fungal Infections: from the Fields to the Clinic, Resistant Aspergillus fumigatus and Dermatophyte Species: a One Health Perspective on an Urgent Public Health Problem. CURRENT CLINICAL MICROBIOLOGY REPORTS 2022; 9:46-51. [PMID: 36188157 PMCID: PMC9512973 DOI: 10.1007/s40588-022-00181-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/27/2022]
Abstract
Purpose of Review For this review, we use a One Health approach to examine two globally emerging public health threats related to antifungal drug resistance: triazole-resistant Aspergillus fumigatus infections, which can cause a life-threatening illness in immunocompromised hosts, and antifungal-resistant dermatophytosis, which is an aggressive skin infection caused by dermatophyte molds. We describe the state of current scientific knowledge and outline necessary public health actions to address each issue. Recent Findings Recent evidence has identified the agricultural use of triazole fungicides as an important driver of triazole-resistant A. fumigatus infections. Antifungal-resistant dermatophyte infections are likely driven by the inappropriate use of antifungal drugs and antibacterial and corticosteroid creams. Summary This review highlights the need for a One Health approach to address emerging antifungal resistant infections, emphasizing judicious antifungal use to preserve available treatments; strengthened laboratory capacity to identify antifungal resistance; and improved human, animal, and environmental surveillance to detect emerging resistance, monitor trends, and evaluate the effectiveness of efforts to decrease spread.
Collapse
Affiliation(s)
- Antonia Langfeldt
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Jeremy A. W. Gold
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Tom Chiller
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| |
Collapse
|
47
|
Basha NJ. Therapeutic Efficacy of Benzimidazole and Its Analogs: An Update. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2118334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- N. Jeelan Basha
- Department of Chemistry, Indian Academy Degree College-Autonomous Bengaluru, India
| |
Collapse
|
48
|
Lv QZ, Zhang XL, Gao L, Yan L, Jiang YY. iTRAQ-based proteomics revealed baicalein enhanced oxidative stress of Candida albicans by up-regulating CPD2 expression. Med Mycol 2022; 60:6687814. [PMID: 36055797 DOI: 10.1093/mmy/myac053] [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: 01/19/2022] [Revised: 05/05/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Baicalein could inhibit the growth and biofilm formation of Candida albicans, the most common clinical fungal pathogen. However, the antifungal mechanism of baicalein has not been elucidated. In this study, isobaric tags for relative and absolute quantification (iTRAQ) was used to verify the mechanism of antifungal fluconazole and baicalein. A total of 58 common proteins were detected in cells treated with fluconazole. These proteins encompassed fluconazole-targeted sterol synthesis pathway, including Erg11p, Erg6p, Erg3p, Erg25p, Erg5p, Erg10p and Ncp1p. Next, iTRAQ was applied to the comparison of baicalein-treated C. albicans proteins, which detected 16 common proteins. The putative NADH dehydrogenase Cpd2p and the ATP-binding cassette transporter Snq2p were the most up-regulated proteins with the treatment of baicalein. Our results showed that CPD2 disruption elevated C. albicans resistance to baicalein significantly both in vitro and in vivo. Further in-depth studies revealed that CPD2 disruption reduced the activation of C. albicans metacaspase and partially restored the mitochondrial membrane potential reduction caused by the treatment of baicalein, which indicated that CPD2 was involved in the apoptosis induced by baicalein. Consistently, under the treatment of baicalein, CPD2Δ/Δ mutant produced lower reactive oxygen species (ROS) that was critical in causing oxidative damage and apoptosis in C. albicans. These results indicated that baicalein could increase intracellular oxidative damage by up-regulating the expression of Cpd2p so as to inhibit the growth of C. albicans, which provides new insights for investigating the antifungal target of baicalein.
Collapse
Affiliation(s)
- Q-Z Lv
- School of Pharmacy, Naval Medical University, Shanghai, P.R.Chi
| | - X-L Zhang
- School of Pharmacy, Naval Medical University, Shanghai, P.R.Chi.,91034 PLA troops, Xuancheng, P.R.Chi
| | - L Gao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R.Chi
| | - L Yan
- School of Pharmacy, Naval Medical University, Shanghai, P.R.Chi
| | - Y-Y Jiang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R.Chi
| |
Collapse
|
49
|
Nicoletti G, White K. The Anti-Fungal Activity of Nitropropenyl Benzodioxole (NPBD), a Redox-Thiol Oxidant and Tyrosine Phosphatase Inhibitor. Antibiotics (Basel) 2022; 11:antibiotics11091188. [PMID: 36139967 PMCID: PMC9495065 DOI: 10.3390/antibiotics11091188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Phylogenetically diverse fungal species are an increasing cause of severe disease and mortality. Identification of new targets and development of new fungicidal drugs are required to augment the effectiveness of current chemotherapy and counter increasing resistance in pathogens. Nitroalkenyl benzene derivatives are thiol oxidants and inhibitors of cysteine-based molecules, which show broad biological activity against microorganisms. Nitropropenyl benzodioxole (NPBD), one of the most active antimicrobial derivatives, shows high activity in MIC assays for phylogenetically diverse saprophytic, commensal and parasitic fungi. NPBD was fungicidal to all species except the dermatophytic fungi, with an activity profile comparable to that of Amphotericin B and Miconazole. NPBD showed differing patterns of dynamic kill rates under different growth conditions for Candida albicans and Aspergillus fumigatus and was rapidly fungicidal for non-replicating vegetative forms and microconidia. It did not induce resistant or drug tolerant strains in major pathogens on long term exposure. A literature review highlights the complexity and interactivity of fungal tyrosine phosphate and redox signaling pathways, their differing metabolic effects in fungal species and identifies some targets for inhibition. A comparison of the metabolic activities of Amphotericin B, Miconazole and NPBD highlights the multiple cellular functions of these agents and the complementarity of many mechanisms. The activity profile of NPBD illustrates the functional diversity of fungal tyrosine phosphatases and thiol-based redox active molecules and contributes to the validation of tyrosine phosphatases and redox thiol molecules as related and complementary selective targets for antimicrobial drug development. NPBD is a selective antifungal agent with low oral toxicity which would be suitable for local treatment of skin and mucosal infections.
Collapse
|
50
|
Zupin L, dos Santos-Silva CA, Al Mughrbi ARH, Vilela LMB, Benko-Iseppon AM, Crovella S. Bioactive Antimicrobial Peptides: A New Weapon to Counteract Zoonosis. Microorganisms 2022; 10:1591. [PMID: 36014009 PMCID: PMC9414035 DOI: 10.3390/microorganisms10081591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Zoonoses have recently become the center of attention of the general population and scientific community. Notably, more than 30 new human pathogens have been identified in the last 30 years, 75% of which can be classified as zoonosis. The complete eradication of such types of infections is far out of reach, considering the limited understanding of animal determinants in zoonoses and their causes of emergence. Therefore, efforts must be doubled in examining the spread, persistence, and pathogenicity of zoonosis and studying possible clinical interventions and antimicrobial drug development. The search for antimicrobial bioactive compounds has assumed great emphasis, considering the emergence of multi-drug-resistant microorganisms. Among the biomolecules of emerging scientific interest are antimicrobial peptides (AMPs), potent biomolecules that can potentially act as important weapons against infectious diseases. Moreover, synthetic AMPs are easily tailored (bioinformatically) to target specific features of the pathogens to hijack, inducing no or very low resistance. Although very promising, previous studies on SAMPs' efficacy are still at their early stages. Indeed, further studies and better characterization on their mechanism of action with in vitro and in vivo assays are needed so as to proceed to their clinical application on human beings.
Collapse
Affiliation(s)
- Luisa Zupin
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | | | | | - Livia Maria Batista Vilela
- Centro de Biociências, Departamento de Genética, Universidade Federal de Pernambuco, Recife 50670-420, Brazil
| | - Ana Maria Benko-Iseppon
- Centro de Biociências, Departamento de Genética, Universidade Federal de Pernambuco, Recife 50670-420, Brazil
| | - Sergio Crovella
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| |
Collapse
|