1
|
Boucher MJ, Banerjee S, Joshi MB, Wei AL, Nalley MJ, Huang MY, Lei S, Ciranni M, Condon A, Langen A, Goddard TD, Caradonna I, Goranov AI, Homer CM, Mortensen Y, Petnic S, Reilly MC, Xiong Y, Susa KJ, Pastore VP, Zaro BW, Madhani HD. Phenotypic landscape of an invasive fungal pathogen reveals its unique biology. Cell 2025:S0092-8674(25)00567-7. [PMID: 40505656 DOI: 10.1016/j.cell.2025.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/27/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025]
Abstract
Cryptococcus neoformans is the most common cause of fungal meningitis and the top-ranking WHO fungal priority pathogen. Only distantly related to model fungi, C. neoformans is also a powerful experimental system for exploring conserved eukaryotic mechanisms lost from specialist model yeast lineages. To decipher its biology globally, we constructed 4,328 gene deletions and measured-with exceptional precision-the fitness of each mutant under 141 diverse growth-limiting in vitro conditions and during murine infection. We defined functional modules by clustering genes based on their phenotypic signatures. In-depth studies leveraged these data in two ways. First, we defined and investigated new components of key signaling pathways, which revealed metazoan-like cellular machinery not present in model yeasts. Second, we identified environmental adaptation mechanisms repurposed to promote mammalian virulence by C. neoformans, which lacks a known animal reservoir. Our work provides an unprecedented resource for deciphering a deadly human pathogen.
Collapse
Affiliation(s)
- Michael J Boucher
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sanjita Banerjee
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Meenakshi B Joshi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Angela L Wei
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Matthew J Nalley
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Manning Y Huang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Susan Lei
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Massimiliano Ciranni
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16145 Genoa, Italy
| | - Andrew Condon
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andreas Langen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Thomas D Goddard
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ippolito Caradonna
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alexi I Goranov
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christina M Homer
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yasaman Mortensen
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sarah Petnic
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Morgann C Reilly
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ying Xiong
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Katherine J Susa
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Vito Paolo Pastore
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16145 Genoa, Italy
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
2
|
Billmyre RB, Craig CJ, Lyon JW, Reichardt C, Kuhn AM, Eickbush MT, Zanders SE. Landscape of essential growth and fluconazole-resistance genes in the human fungal pathogen Cryptococcus neoformans. PLoS Biol 2025; 23:e3003184. [PMID: 40402997 DOI: 10.1371/journal.pbio.3003184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Fungi can cause devastating invasive infections, typically in immunocompromised patients. Treatment is complicated both by the evolutionary similarity between humans and fungi and by the frequent emergence of drug resistance. Studies in fungal pathogens have long been slowed by a lack of high-throughput tools and community resources that are common in model organisms. Here we demonstrate a high-throughput transposon mutagenesis and sequencing (TN-seq) system in Cryptococcus neoformans that enables genome-wide determination of gene essentiality. We employed a random forest machine learning approach to classify the C. neoformans genome as essential or nonessential, predicting 1,465 essential genes, including 302 that lack human orthologs. These genes are ideal targets for new antifungal drug development. TN-seq also enables genome-wide measurement of the fitness contribution of genes to phenotypes of interest. As proof of principle, we demonstrate the genome-wide contribution of genes to growth in fluconazole, a clinically used antifungal. We show a novel role for the well-studied RIM101 pathway in fluconazole susceptibility. We also show that insertions of transposons into the 5' upstream region can drive sensitization of essential genes, enabling screenlike assays of both essential and nonessential components of the genome. Using this approach, we demonstrate a role for mitochondrial function in fluconazole sensitivity, such that tuning down many essential mitochondrial genes via 5' insertions can drive resistance to fluconazole. Our assay system will be valuable in future studies of C. neoformans, particularly in examining the consequences of genotypic diversity.
Collapse
Affiliation(s)
- R Blake Billmyre
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, GeorgiaUnited States of America
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GeorgiaUnited States of America
- Department of Microbiology, Franklin College of Arts and Sciences, University of Georgia, Athens, GeorgiaUnited States of America
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Caroline J Craig
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Joshua W Lyon
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GeorgiaUnited States of America
- Department of Pharmaceutical and Biological Sciences, College of Pharmacy, University of Georgia, Athens, GeorgiaUnited States of America
| | - Claire Reichardt
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, GeorgiaUnited States of America
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GeorgiaUnited States of America
- Department of Microbiology, Franklin College of Arts and Sciences, University of Georgia, Athens, GeorgiaUnited States of America
| | - Amy M Kuhn
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, GeorgiaUnited States of America
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GeorgiaUnited States of America
| | - Michael T Eickbush
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Sarah E Zanders
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
3
|
Wang X, Li X, Li D, Zhang Y, Bai B, Chai B, Wen Z. A novel lncRNA YIL163C enhances genomic stability and antifungal resistance via the DNA damage response in Saccharomyces cerevisiae. Front Microbiol 2025; 16:1571797. [PMID: 40376465 PMCID: PMC12078222 DOI: 10.3389/fmicb.2025.1571797] [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: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators in cellular processes, including the DNA damage response (DDR). In Saccharomyces cerevisiae, DDR is critical for maintaining genomic integrity under stress, mediated by proteins like Mec1 and Rad53. However, the involvement of lncRNAs in DDR pathways, remains largely unexplored. This study investigates the function of a novel lncRNA, YIL163C, in promoting cell survival and genomic stability under DNA damage conditions. Methods Genetic suppressor screening was employed to assess the role of YIL163C in rescuing lethality in mec1Δ sml1Δ and rad53Δ sml1Δ exposed to DNA damage. Proteomic and phosphoproteomic analyses were conducted to evaluate changes in protein abundance and phosphorylation states. The impact of YIL163C on DDR and antifungal drug tolerance, specifically to 5-fluorocytosine, was also examined. Results Overexpression of YIL163C was found to rescue lethality in mec1Δ sml1Δ and rad53Δ sml1Δ under DNA damage conditions. Proteomic analyses revealed that YIL163C modulates pathways related to DNA replication, ER stress response, and ribosome biogenesis, enhancing cellular resilience to HU-induced stress. Additionally, YIL163C reduced sensitivity to 5-fluorocytosine, indicating a role in antifungal drug tolerance. Phosphoproteomic data suggested YIL163C influences phosphorylation states, potentially acting downstream of the Mec1-Rad53 signaling pathway. Conclusion This study provides new insights into the regulatory mechanisms of lncRNAs in DDR, with broader implications for antifungal therapy and genomic stability research, emphasizing the role of lncRNAs in stress responses beyond traditional protein-centric mechanisms.
Collapse
Affiliation(s)
- Xueting Wang
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for New Technology Research of Vegetables, Vegetable Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xuemei Li
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Duoyun Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Yiying Zhang
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Bing Bai
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Bao Chai
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Zewen Wen
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Pharkjaksu S, Cai H, Walter PJ, Chang YC, Kwon-Chung KJ. Elevated UDP-glucuronic acid levels mend drug resistance and stress responses via a protease and a transporter in Cryptococcus gattii. Proc Natl Acad Sci U S A 2025; 122:e2503960122. [PMID: 40267138 PMCID: PMC12054807 DOI: 10.1073/pnas.2503960122] [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: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
UDP-glucuronic acid (UDP-GlcUA) is a nucleotide sugar essential for various biological processes in many organisms, and its excess within the cell can disrupt cellular functions. In Cryptococcus, mutations in the UXS1 gene which encodes an enzyme responsible for converting UDP-GlcUA into UDP-xylose, result in excessive accumulation of UDP-GlcUA and confer resistance to the antifungal drug 5-fluorocytosine. Here, we demonstrate that elevation of UDP-GlcUA affects several cellular processes in Cryptococcus gattii, including growth rate, ability to grow under various stress conditions and resistance to fluorinated pyrimidine analogs. RNA-seq analyses of the uxs1Δ mutant identify three acid protease genes, notably PEP401, that are differentially expressed. The absence of PEP401 in the uxs1Δ background significantly reduces UDP-GlcUA levels and reverts all the phenotypes of the uxs1Δ mutant to the wild-type characteristics. High levels of UDP-GlcUA not only regulate expression of PEP401 at RNA and protein levels but also enhance the proteolytic activity of total protein extracts in a PEP401-dependent manner, establishing a functional link between nucleotide sugar metabolism and proteolytic regulation. Moreover, the UDP-GlcUA transporter gene, UUT1, can further modulate the levels of UDP-GlcUA in the uxs1Δ pep401Δ double mutant and manifests drug resistance phenotypes observed in the uxs1Δ mutant. Collectively, these findings reveal a previously unrecognized regulatory network that links UDP-GlcUA metabolism to protease-mediated cellular processes and the transport of UDP-GlcUA. This interaction provides a foundation for targeting nucleotide sugar metabolism and protease regulation in the development of enhanced therapeutic strategies against cryptococcosis.
Collapse
Affiliation(s)
- Sujiraphong Pharkjaksu
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Hongyi Cai
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD20892
| | - Peter J. Walter
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD20892
| | - Yun C. Chang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Kyung J. Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
5
|
Phan-Canh T, Nguyen-Le DM, Luu PL, Khunweeraphong N, Kuchler K. Rapid in vitro evolution of flucytosine resistance in Candida auris. mSphere 2025; 10:e0097724. [PMID: 40099908 PMCID: PMC12039228 DOI: 10.1128/msphere.00977-24] [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: 12/27/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
The pan-antifungal-resistant pathogen Candida auris has been causing high-mortality infection outbreaks in hospitals and healthcare settings. The prodrug 5-fluorocytosine (5FC) is one of four chemical entities, but its clinical use as an antifungal drug has been limited owing to pronounced resistance. However, antifungal combination therapy with 5FC appears as a promising strategy for treating C. auris infections. Here, we show that a C. auris clinical isolate can rapidly acquire genetic mutations to mount 5FC resistance after only one to two passages under drug selection. We exploit a new bioinformatics workflow to identify genetic polymorphisms from RNA-seq data. Strikingly, we identify several mutations in the FUR1 gene encoding the 5-fluorouracil convertase that normally generates the active drug. A single nonsense mutation truncates the enzyme at residue Q30*, leading to 5FC resistance due to inactive Fur1. Whole-genome sequencing analysis revealed that an indel mutation in FCY2 also contributes to 5FC resistance. Furthermore, at least one out of seven adapted strains acquired enhanced 5FC tolerance without mutations in the 5FC conversion pathway. Thus, we demonstrate that FUR1 mutations are critical drivers of 5FC resistance in C. auris.IMPORTANCECandida auris is a high-priority human fungal pathogen, causing infection outbreaks of high mortality in healthcare settings. Antifungal combination therapy with 5-fluorocytosine (5FC) is one of the emerging approaches in treatment. However, acquired 5FC resistance traits have been a matter of concern. 5FC is taken up by fungal cells via a cytosine permease and further metabolized by a cytosine deaminase to 5-fluorouracil (5FU). 5FU is then converted by the Fur1 uracil phosphoribosyltransferase into a toxic antimetabolite that disrupts fungal RNA and DNA syntheses. Mutations in these proteins are commonly associated with 5FC resistance in fungal species. Here, we show that C. auris can rapidly develop resistance under 5FC selective stress owing to mutational inactivation of Fur1 function. Moreover, other mechanisms that bypass mutations in the 5FC conversion pathway may also contribute to 5FC resistance traits. Finally, we have developed a tailored bioinformatics workflow that facilitates the identification of polymorphisms associated with 5FC resistance in clinical isolates.
Collapse
Affiliation(s)
- Trinh Phan-Canh
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| | - Duc-Minh Nguyen-Le
- Institute for Applied Research in Health Sciences and Aging (ARiHA)-Thong Nhat Hospital, Ho Chi Minh City, Vietnam
| | - Phuc-Loi Luu
- Institute for Applied Research in Health Sciences and Aging (ARiHA)-Thong Nhat Hospital, Ho Chi Minh City, Vietnam
- Mathematics Department, Faculty of Fundamental Sciences, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Narakorn Khunweeraphong
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| |
Collapse
|
6
|
Huang X, Dong Q, Zhou Q, Fang S, Xu Y, Long H, Chen J, Li X, Qin H, Mu D, Cai X. Genomics insights of candidiasis: mechanisms of pathogenicity and drug resistance. Front Microbiol 2025; 16:1531543. [PMID: 40083780 PMCID: PMC11903725 DOI: 10.3389/fmicb.2025.1531543] [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: 11/20/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Candidiasis, a prevalent class of human infections caused by fungi belonging to the Candida genus, is garnering increasing attention due to its pathogenicity and the emergence of drug resistance. The advancement of genomics technologies has offered powerful tools for investigating the pathogenic mechanisms and drug resistance characteristics of Candida. This comprehensive review provides an overview of the applications of genomics in candidiasis research, encompassing genome sequencing, comparative genomics, and functional genomics, along with the pathogenic features and core virulence factors of Candida. Moreover, this review highlights the role of genomic variations in the emergence of drug resistance, further elucidating the evolutionary and adaptive mechanisms of Candida. In conclusion, the review underscores the current state of research and prospective avenues for exploration of candidiasis, providing a theoretical basis for clinical treatments and public health strategies.
Collapse
Affiliation(s)
- Xin Huang
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qin Dong
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Qi Zhou
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Shitao Fang
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Yiheng Xu
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Hongjie Long
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Jingyi Chen
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Xiao Li
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Huaguang Qin
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Dan Mu
- Key Laboratory of Biodiversity Conservation and Characteristic Resource Utilization in Southwest Anhui, Anqing Forestry Technology Innovation Research Institute, School of Life Sciences, Anqing Normal University, Anqing, China
| | - Xunchao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
Su Y, Li Y, Yi Q, Xu Y, Sun T, Li Y. Insight into the Mechanisms and Clinical Relevance of Antifungal Heteroresistance. J Fungi (Basel) 2025; 11:143. [PMID: 39997437 PMCID: PMC11856953 DOI: 10.3390/jof11020143] [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: 01/04/2025] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Antifungal resistance poses a critical global health threat, particularly in immuno-compromised patients. Beyond the traditional resistance mechanisms rooted in heritable and stable mutations, a distinct phenomenon known as heteroresistance has been identified, wherein a minority of resistant fungal cells coexist within a predominantly susceptible population. Heteroresistance may be induced by pharmacological factors or non-pharmacological agents. The reversible nature of it presents significant clinical challenges, as it can lead to undetected resistance during standard susceptibility testing. As heteroresistance allows fungal pathogens to survive antifungal treatment, this adaptive strategy often leads to treatment failure and recurring infection. Though extensively studied in bacteria, limited research has explored its occurrence in fungi. This review summarizes the current findings on antifungal heteroresistance mechanisms, highlighting the clinical implications of fungal heteroresistance and the pressing need for deeper mechanism insights. We aim to bring together the latest research advances in the field of antifungal heteroresistance, summarizing in detail its known characteristics, inducing factors, molecular mechanisms, and clinical significance, and describing the similarities and differences between heteroresistance, tolerance and persistence. Further research is needed to understand this phenomenon and develop more effective antifungal therapies to combat fungal infections.
Collapse
Affiliation(s)
- Yanyu Su
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Graduate School, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yi Li
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Qiaolian Yi
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China; (Y.S.); (Y.L.); (Q.Y.); (Y.X.)
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
| | - Tianshu Sun
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- Clinical Biobank, Center for Biomedical Technology, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yingxing Li
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing 100730, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
8
|
Razumovskaya A, Silkina M, Poloznikov A, Kulagin T, Raigorodskaya M, Gorban N, Kudryavtseva A, Fedorova M, Alekseev B, Tonevitsky A, Nikulin S. Predicting patient outcomes with gene-expression biomarkers from colorectal cancer organoids and cell lines. Front Mol Biosci 2025; 12:1531175. [PMID: 39886381 PMCID: PMC11774744 DOI: 10.3389/fmolb.2025.1531175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is characterized by an extremely high mortality rate, mainly caused by the high metastatic potential of this type of cancer. To date, chemotherapy remains the backbone of the treatment of metastatic colorectal cancer. Three main chemotherapeutic drugs used for the treatment of metastatic colorectal cancer are 5-fluorouracil, oxaliplatin and irinotecan which is metabolized to an active compound SN-38. The main goal of this study was to find the genes connected to the resistance to the aforementioned drugs and to construct a predictive gene expression-based classifier to separate responders and non-responders. Methods In this study, we analyzed gene expression profiles of seven patient-derived CRC organoids and performed correlation analyses between gene expression and IC50 values for the three standard-of-care chemotherapeutic drugs. We also included in the study publicly available datasets of colorectal cancer cell lines, thus combining two different in vitro models relevant to cancer research. Logistic regression was used to build gene expression-based classifiers for metastatic Stage IV and non-metastatic Stage II/III CRC patients. Prognostic performance was evaluated through Kaplan-Meier survival analysis and log-rank tests, while independent prognostic significance was assessed using multivariate Cox proportional hazards modeling. Results A small set of genes showed consistent correlation with resistance to chemotherapy across different datasets. While some genes were previously implicated in cancer prognosis and drug response, several were linked to drug resistance for the first time. The resulting gene expression signatures successfully stratified Stage II/III and Stage IV CRC patients, with potential clinical utility for improving treatment outcomes after further validation. Discussion This study highlights the advantages of integrating diverse experimental models, such as organoids and cell lines, to identify novel prognostic biomarkers and enhance the understanding of chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Alexandra Razumovskaya
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Mariia Silkina
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Poloznikov
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Timur Kulagin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Maria Raigorodskaya
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nina Gorban
- Central Clinical Hospital with Polyclinic, Administration of the President of the Russian Federation, Moscow, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Alekseev
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Art Photonics GmbH, Berlin, Germany
| | - Sergey Nikulin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
9
|
Chen X, Duan HD, Hoy MJ, Koteva K, Spitzer M, Guitor AK, Puumala E, Hu G, Yiu B, Chou S, Bian Z, Guo ABY, Sun S, Robbins N, Cook MA, Truant R, MacNeil LT, Brown ED, Kronstad JW, Cowen LE, Heitman J, Li H, Wright GD. Butyrolactol A is a phospholipid flippase inhibitor that potentiates the bioactivity of caspofungin against resistant fungi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.630955. [PMID: 39829750 PMCID: PMC11741340 DOI: 10.1101/2025.01.06.630955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Fungal infections cause millions of deaths annually and are challenging to treat due to limited antifungal options and increasing drug resistance. Cryptococci are intrinsically resistant to the latest generation of antifungals, echinocandins, while Candida auris , a notorious global threat, is also increasingly resistant. We performed a natural product extract screen for rescue of the activity of the echinocandin caspofungin against Cryptococcus neoformans H99, identifying butyrolactol A, which restores echinocandin efficacy against resistant fungal pathogens, including C. auris . Mode of action studies revealed that butyrolactol A inhibits the phospholipid flippase Apt1-Cdc50, blocking phospholipid transport. Cryoelectron-microscopy analysis of the Apt1●butyrolactol A complex revealed that the flippase is locked in a dead-end state. Apt1 inhibition disrupts membrane asymmetry, vesicular trafficking, and cytoskeletal organization, thereby enhancing echinocandin uptake and potency. This study identifies flippases as promising antifungal targets and demonstrates the potential of revisiting natural products to expand the antifungal arsenal and combat resistance.
Collapse
|
10
|
Iracane E, Buscaino A. Transposon load and RNAi loss synergize to drive intraspecies diversity in Cryptococcus. Proc Natl Acad Sci U S A 2024; 121:e2422896121. [PMID: 39680781 PMCID: PMC11670079 DOI: 10.1073/pnas.2422896121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Affiliation(s)
- Elise Iracane
- Kent Fungal Group, School of Biosciences, Division of Natural Sciences, University of Kent Canterbury, KentCT2 7NZ, United Kingdom
| | - Alessia Buscaino
- Kent Fungal Group, School of Biosciences, Division of Natural Sciences, University of Kent Canterbury, KentCT2 7NZ, United Kingdom
| |
Collapse
|
11
|
Bottery MJ, van Rhijn N, Chown H, Rhodes JL, Celia-Sanchez BN, Brewer MT, Momany M, Fisher MC, Knight CG, Bromley MJ. Elevated mutation rates in multi-azole resistant Aspergillus fumigatus drive rapid evolution of antifungal resistance. Nat Commun 2024; 15:10654. [PMID: 39681549 PMCID: PMC11649685 DOI: 10.1038/s41467-024-54568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
The environmental use of azole fungicides has led to selective sweeps across multiple loci in the Aspergillus fumigatus genome causing the rapid global expansion of a genetically distinct cluster of resistant genotypes. Isolates within this cluster are also more likely to be resistant to agricultural antifungals with unrelated modes of action. Here we show that this cluster is not only multi-azole resistant but has increased propensity to develop resistance to next generation antifungals because of variants in the DNA mismatch repair system. A variant in msh6-G233A is found almost exclusively within azole resistant isolates harbouring the canonical cyp51A azole resistance allelic variant TR34/L98H. Naturally occurring isolates with this msh6 variant display up to 5-times higher rate of mutation, leading to an increased likelihood of evolving resistance to other antifungals. Furthermore, unlike hypermutator strains, the G233A variant conveys no measurable fitness cost and has become globally distributed. Our findings further suggest that resistance to next-generation antifungals is more likely to emerge within organisms that are already multi-azole resistant due to close linkage between TR34/L98H and msh6-G233A, posing a major problem due to the prospect of dual use of novel antifungals in clinical and agricultural settings.
Collapse
Affiliation(s)
- Michael J Bottery
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Norman van Rhijn
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Harry Chown
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Research Council Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
| | - Johanna L Rhodes
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Brandi N Celia-Sanchez
- Fungal Biology Group and Department of Plant Biology, University of Georgia, Athens, GA, 30602, USA
| | - Marin T Brewer
- Fungal Biology Group and Department of Plant Pathology, University of Georgia, Athens, GA, 30602, USA
| | - Michelle Momany
- Fungal Biology Group and Department of Plant Biology, University of Georgia, Athens, GA, 30602, USA
| | - Matthew C Fisher
- Medical Research Council Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
| | - Christopher G Knight
- Department of Earth and Environmental Sciences, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Michael J Bromley
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
12
|
Lathakumari RH, Vajravelu LK, Satheesan A, Thulukanam J. Advancing cryptococcal treatment: The role of nanoparticles in mitigating antifungal resistance. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100323. [PMID: 39678065 PMCID: PMC11638651 DOI: 10.1016/j.crmicr.2024.100323] [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] [Indexed: 12/17/2024] Open
Abstract
Cryptococcus, a ubiquitous and formidable fungal pathogen, contributes to a substantial global disease burden, with nearly 250,000 cases and 181,000 fatalities attributed to cryptococcal meningitis annually worldwide. The invasive nature of Cryptococcus presents significant challenges in treatment and management, as it mostly affects vulnerable populations, including HIV patients, organ transplant recipients, pregnant women, and elderly individuals. Moreover, these difficulties are exacerbated by the development of antifungal resistance, which emphasizes the need for efficient control measures. In this context, research efforts focusing on infection control and novel therapeutic strategies become paramount. Nanoparticle-based therapies emerge as a solution, offering advanced antifungal properties and improved efficacy. Developing effective treatment options requires understanding the complex landscape of cryptococcal infections and the innovative potential of nanoparticle-based therapies. This review highlights the urgent need for novel strategies to combat the growing threat posed by antifungal resistance while offering insights into the intricate realm of cryptococcal infections, particularly focusing on the promising role of nanoparticle-based therapies.
Collapse
Affiliation(s)
- Rahul Harikumar Lathakumari
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Leela Kakithakara Vajravelu
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| | - Jayaprakash Thulukanam
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, Chennai, Tamil Nadu, India
| |
Collapse
|
13
|
Huang J, Larmore CJ, Priest SJ, Xu Z, Dietrich FS, Yadav V, Magwene PM, Sun S, Heitman J. Distinct evolutionary trajectories following loss of RNA interference in Cryptococcus neoformans. Proc Natl Acad Sci U S A 2024; 121:e2416656121. [PMID: 39536081 PMCID: PMC11588098 DOI: 10.1073/pnas.2416656121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
While increased mutation rates typically have negative consequences in multicellular organisms, hypermutation can be advantageous for microbes adapting to the environment. Previously, we identified two hypermutator Cryptococcus neoformans clinical isolates that rapidly develop drug resistance due to transposition of a retrotransposon, Cnl1. Cnl1-mediated hypermutation is caused by a nonsense mutation in a gene encoding an RNA interference (RNAi) component, ZNF3, combined with a tremendous transposon burden. To elucidate adaptive mechanisms following RNAi loss, two bioinformatic pipelines were developed to identify RNAi loss-of-function (LOF) mutations in a collection of 387 sequenced C. neoformans isolates. Remarkably, several RNAi-loss isolates were identified that are not hypermutators and have not accumulated transposons. To test whether these RNAi LOF mutations can cause hypermutation, the mutations were introduced into a nonhypermutator strain with a high transposon burden, which resulted in a hypermutator phenotype. To further investigate whether RNAi-loss isolates can become hypermutators, in vitro passaging was performed. Although no hypermutators were found in two C. neoformans RNAi-loss strains after short-term passage, hypermutation was observed in a passaged Cryptococcus deneoformans strain with an increased transposon burden. Consistent with a two-step evolution, when an RNAi-loss isolate was crossed with an isolate containing a high Cnl1 burden, F1 hypermutator progeny inheriting a high transposon burden were identified. In addition to Cnl1 transpositions, insertions of a gigantic DNA transposon KDZ1 (~11 kb) contributed to hypermutation in the progeny. Our results suggest that RNAi loss is relatively common (7/387, ~1.8%) and enables distinct evolutionary trajectories: hypermutation following transposon accumulation or survival without hypermutation.
Collapse
Affiliation(s)
- Jun Huang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Connor J. Larmore
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Shelby J. Priest
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Ziyan Xu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Fred S. Dietrich
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Vikas Yadav
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | | | - Sheng Sun
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
14
|
Boucher MJ, Banerjee S, Joshi MB, Wei AL, Huang MY, Lei S, Ciranni M, Condon A, Langen A, Goddard TD, Caradonna I, Goranov AI, Homer CM, Mortensen Y, Petnic S, Reilly MC, Xiong Y, Susa KJ, Pastore VP, Zaro BW, Madhani HD. Phenotypic landscape of a fungal meningitis pathogen reveals its unique biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619677. [PMID: 39484549 PMCID: PMC11526942 DOI: 10.1101/2024.10.22.619677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Cryptococcus neoformans is the most common cause of fungal meningitis and the top-ranked W.H.O. priority fungal pathogen. Only distantly related to model fungi, C. neoformans is also a powerful experimental system for exploring conserved eukaryotic mechanisms lost from specialist model yeast lineages. To decipher its biology globally, we constructed 4328 gene deletions and measured-with exceptional precision--the fitness of each mutant under 141 diverse growth-limiting in vitro conditions and during murine infection. We defined functional modules by clustering genes based on their phenotypic signatures. In-depth studies leveraged these data in two ways. First, we defined and investigated new components of key signaling pathways, which revealed animal-like pathways/components not predicted from studies of model yeasts. Second, we identified environmental adaptation mechanisms repurposed to promote mammalian virulence by C. neoformans, which lacks a known animal reservoir. Our work provides an unprecedented resource for deciphering a deadly human pathogen.
Collapse
Affiliation(s)
- Michael J Boucher
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Sanjita Banerjee
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Meenakshi B Joshi
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Angela L Wei
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Manning Y Huang
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Susan Lei
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Massimiliano Ciranni
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, via alla Opera Pia 13, 16145 Genoa, Italy
| | - Andrew Condon
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Andreas Langen
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Thomas D Goddard
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Ippolito Caradonna
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Alexi I Goranov
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Christina M Homer
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Yassaman Mortensen
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Sarah Petnic
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Morgann C Reilly
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Ying Xiong
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Katherine J Susa
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Vito Paolo Pastore
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, via alla Opera Pia 13, 16145 Genoa, Italy
| | - Balyn W Zaro
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Hiten D Madhani
- Dept. of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
15
|
Stott KE, Mohabir JT, Bowers K, Tenor JL, Toffaletti DL, Unsworth J, Jimenez-Valverde A, Ahmadu A, Moyo M, Gondwe E, Chimang’anga W, Chasweka M, Lawrence DS, Jarvis JN, Harrison T, Hope W, Lalloo DG, Mwandumba HC, Perfect JR, Cuomo CA, The AMBITION Study Group. Integration of genomic and pharmacokinetic data to predict clinical outcomes in HIV-associated cryptococcal meningitis. mBio 2024; 15:e0159224. [PMID: 39189739 PMCID: PMC11481554 DOI: 10.1128/mbio.01592-24] [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: 05/30/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
Cryptococcal meningitis causes an estimated 112,000 global deaths per annum. Genomic and phenotypic features of the infecting strain of Cryptococcus spp. have been associated with outcomes from cryptococcal meningitis. Additionally, population-level pharmacokinetic variability is well documented in these patient cohorts. The relative contribution of these factors to clinical outcomes is unknown. Based in Malawi, we conducted a sub-study of the phase 3 Ambition-CM trial (ISRCTN72509687), collecting plasma and cerebrospinal fluid at serial time points during the first 14 days of antifungal therapy. We explored the relative contribution of pathogen genotype, drug resistance phenotype, and pharmacokinetics on clinical outcomes including lumbar opening pressure, pharmacodynamic effect, and mortality. We report remarkable genomic homogeneity among infecting strains of Cryptococcus spp., within and between patients. There was no evidence of acquisition of antifungal resistance in our isolates. Genotypic features of the infecting strain were not consistently associated with adverse or favorable clinical outcomes. However, baseline fungal burden and early fungicidal activity (EFA) were associated with mortality. The strongest predictor of EFA was the level of exposure to amphotericin B. Our analysis suggests the most effective means of improving clinical outcomes from HIV-associated cryptococcal meningitis is to optimize exposure to potent antifungal therapy. IMPORTANCE HIV-associated cryptococcal meningitis is associated with a high burden of mortality. Research into the different strain types causing this disease has yielded inconsistent findings in terms of which strains are associated with worse clinical outcomes. Our study suggests that the exposure of patients to potent anti-cryptococcal drugs has a more significant impact on clinical outcomes than the strain type of the infecting organism. Future research should focus on optimizing drug exposure, particularly in the context of novel anticryptococcal drugs coming into clinical use.
Collapse
Affiliation(s)
- Katharine E. Stott
- Antimicrobial Pharmacodynamics and Therapeutics Group, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
| | - Jason T. Mohabir
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Katharine Bowers
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jennifer L. Tenor
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dena L. Toffaletti
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jennifer Unsworth
- Antimicrobial Pharmacodynamics and Therapeutics Group, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Ana Jimenez-Valverde
- Antimicrobial Pharmacodynamics and Therapeutics Group, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Ajisa Ahmadu
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
| | - Melanie Moyo
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
- Department of Medicine, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Ebbie Gondwe
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
| | - Wezi Chimang’anga
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
| | | | - David S. Lawrence
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Tropical Medicine, London, United Kingdom
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Joseph N. Jarvis
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Tropical Medicine, London, United Kingdom
- Botswana Harvard Health Partnership, Gaborone, Botswana
| | - Tom Harrison
- Institute of Infection and Immunity, St George’s University London, London, United Kingdom
| | - William Hope
- Antimicrobial Pharmacodynamics and Therapeutics Group, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - David G. Lalloo
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - John R. Perfect
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - The AMBITION Study Group
- Antimicrobial Pharmacodynamics and Therapeutics Group, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Clinical Research Programme, Blantyre, Malawi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Tropical Medicine, London, United Kingdom
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Institute of Infection and Immunity, St George’s University London, London, United Kingdom
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
16
|
Rhodes J, Jacobs J, Dennis EK, Manjari SR, Banavali NK, Marlow R, Rokebul MA, Chaturvedi S, Chaturvedi V. What makes Candida auris pan-drug resistant? Integrative insights from genomic, transcriptomic, and phenomic analysis of clinical strains resistant to all four major classes of antifungal drugs. Antimicrob Agents Chemother 2024; 68:e0091124. [PMID: 39297640 PMCID: PMC11459930 DOI: 10.1128/aac.00911-24] [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: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024] Open
Abstract
The global epidemic of drug-resistant Candida auris continues unabated. The initial report on pan-drug resistant (PDR) C. auris strains in a hospitalized patient in New York was unprecedented. PDR C. auris showed both known and unique mutations in the prominent gene targets of azoles, amphotericin B, echinocandins, and flucytosine. However, the factors that allow C. auris to acquire pan-drug resistance are not known. Therefore, we conducted a genomic, transcriptomic, and phenomic analysis to better understand PDR C. auris. Among 1,570 genetic variants in drug-resistant C. auris, 299 were unique to PDR strains. The whole-genome sequencing results suggested perturbations in genes associated with nucleotide biosynthesis, mRNA processing, and nuclear export of mRNA. Whole transcriptome sequencing of PDR C. auris revealed two genes to be significantly differentially expressed-a DNA repair protein and DNA replication-dependent chromatin assembly factor 1. Of 59 novel transcripts, 12 transcripts had no known homology. We observed no fitness defects among multi-drug resistant (MDR) and PDR C. auris strains grown in nutrient-deficient or -enriched media at different temperatures. Phenotypic profiling revealed wider adaptability to nitrogenous nutrients and increased utilization of substrates critical in upper glycolysis and tricarboxylic acid cycle. Structural modeling of a 33-amino acid deletion in the gene for uracil phosphoribosyl transferase suggested an alternate route in C. auris to generate uracil monophosphate that does not accommodate 5-fluorouracil as a substrate. Overall, we find evidence of metabolic adaptations in MDR and PDR C. auris in response to antifungal drug lethality without deleterious fitness costs.
Collapse
Affiliation(s)
- Johanna Rhodes
- Department of Medical Microbiology, Radboudumc, Nijmegen, the Netherlands
- MRC GIDA, Imperial College London, London, United Kingdom
| | - Jonathan Jacobs
- American Type Culture Collection, University Blvd, Manassas, Virginia, USA
| | - Emily K. Dennis
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Swati R. Manjari
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Nilesh K. Banavali
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- School of Public Health, University of Albany, Albany, New York, USA
| | - Robert Marlow
- American Type Culture Collection, University Blvd, Manassas, Virginia, USA
| | | | - Sudha Chaturvedi
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- School of Public Health, University of Albany, Albany, New York, USA
| | - Vishnu Chaturvedi
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Westchester Medical Center/New York Medical College, Valhalla, New York, USA
| |
Collapse
|
17
|
Floyd HEE, Kavanagh AM, Lowe GJ, Amado M, Fraser JA, Blaskovich MAT, Elliott AG, Zuegg J. Standardisation of high throughput microdilution antifungal susceptibility testing for Candida albicans and Cryptococcus neoformans. Sci Rep 2024; 14:23407. [PMID: 39379501 PMCID: PMC11461513 DOI: 10.1038/s41598-024-74068-2] [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: 06/03/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
The Clinical and Laboratory Standards Institute (CLSI) M27 guidelines are the recommended and most commonly used protocols for broth microdilution antifungal susceptibility testing of yeasts. However, these guidelines are limited to the use of 96-well assay plates, limiting assay capacity. With the increased risk of fungal resistance emerging in the community, it is important to have alternative protocols available, that offer higher throughput and can screen more than eight to ten potential antifungal compounds per plate. This study presents an optimised broth microdilution minimum inhibitory concentration (MIC) method for testing the susceptibility of yeasts in an efficient high throughput screening setup, with minimal growth variability and maximum reproducibility. We extend the M27 guidelines and optimise the conditions for 384-well plates. Validation of the assay was performed with ten clinically used antifungals (fluconazole, amphotericin B, 5-fluorocytosine, posaconazole, voriconazole, ketoconazole, itraconazole, caspofungin diacetate, anidulafungin and micafungin) against Candida albicans and Cryptococcus neoformans.
Collapse
Affiliation(s)
- Holly E E Floyd
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Angela M Kavanagh
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Gabrielle J Lowe
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Maite Amado
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - James A Fraser
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Mark A T Blaskovich
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Alysha G Elliott
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Johannes Zuegg
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
18
|
Huang J, Larmore CJ, Priest SJ, Xu Z, Dietrich FS, Yadav V, Magwene PM, Sun S, Heitman J. Distinct evolutionary trajectories following loss of RNA interference in Cryptococcus neoformans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608186. [PMID: 39185155 PMCID: PMC11343200 DOI: 10.1101/2024.08.15.608186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
While increased mutation rates typically have negative consequences in multicellular organisms, hypermutation can be advantageous for microbes adapting to the environment. Previously, we identified two hypermutator Cryptococcus neoformans clinical isolates that rapidly develop drug resistance due to transposition of a retrotransposon, Cnl1. Cnl1-mediated hypermutation is caused by a nonsense mutation in the gene encoding a novel RNAi component, Znf3, combined with a tremendous transposon burden. To elucidate adaptative mechanisms following RNAi loss, two bioinformatic pipelines were developed to identify RNAi loss-of-function mutations in a collection of 387 sequenced C. neoformans isolates. Remarkably, several RNAi-loss isolates were identified that are not hypermutators and have not accumulated transposons. To test if these RNAi loss-of-function mutations can cause hypermutation, the mutations were introduced into a non-hypermutator strain with a high transposon burden, which resulted in a hypermutator phenotype. To further investigate if RNAi-loss isolates can become hypermutators, in vitro passaging was performed. Although no hypermutators were found in two C. neoformans RNAi-loss strains after short-term passage, hypermutation was observed in a passaged C. deneoformans strain with increased transposon burden. Consistent with a two-step evolution, when an RNAi-loss isolate was crossed with an isolate containing a high Cnl1 burden, F1 hypermutator progeny inheriting a high transposon burden were identified. In addition to Cnl1 transpositions, insertions of a novel gigantic DNA transposon KDZ1 (~11 kb), contributed to hypermutation in the progeny. Our results suggest that RNAi loss is relatively common (7/387, ~1.8%) and enables distinct evolutionary trajectories: hypermutation following transposon accumulation or survival without hypermutation.
Collapse
Affiliation(s)
- Jun Huang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Connor J. Larmore
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Shelby J. Priest
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ziyan Xu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Fred S. Dietrich
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Vikas Yadav
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Paul M. Magwene
- Department of Biology, Duke University, Durham, NC 27710, USA
| | - Sheng Sun
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
19
|
Delma FZ, Yang DH, Cabrera-Orefice A, Coolen J, Al-Hatmi AMS, Ahmed SA, Melchers WJG, Chang YC, Kwon-Chung KJ, de Hoog S, Verweij PE, Buil JB. Genetic mutations in Cryptococcus neoformans pyrimidine salvage pathway enzymes contribute to reduced susceptibility against 5-fluorocytosine. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:24. [PMID: 39843955 PMCID: PMC11721657 DOI: 10.1038/s44259-024-00041-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/19/2024] [Indexed: 01/24/2025]
Abstract
Cryptococcal meningitis is a high-mortality infection. Adding 5-fluorocytosine (5-FC) to its treatment improves outcomes, but resistance to 5-FC presents a significant challenge. We conducted whole-genome sequencing on seven C. neoformans isolates with varying 5-FC susceptibility, along with proteomic and in silico analyses. Our findings indicate that mutations in genes of the pyrimidine salvage pathway are responsible for 5-FC resistance. Specifically, we identified an E64G missense mutation in the FUR1 gene, a large deletion in the FCY1 gene, and a point mutation in FCY1 leading to a truncated protein. The proteomic data indicated that these mutations resulted in the absence or reduction of crucial enzymes in resistant isolates. Genetic transformations confirmed the association between these mutations and 5-FC resistance. Resistance to 5-FC can develop during treatment and is closely tied to mutations in key metabolic enzymes. Understanding in vivo resistance development is crucial for combating resistance and enhancing patient outcomes.
Collapse
Affiliation(s)
- Fatima Zohra Delma
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Dong-Hoon Yang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Alfredo Cabrera-Orefice
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jordy Coolen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Abdullah M S Al-Hatmi
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Sarah A Ahmed
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Willem J G Melchers
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Yun C Chang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Kyung J Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Sybren de Hoog
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Paul E Verweij
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Jochem B Buil
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.
- Radboudumc-CWZ Center of Expertise for Mycology, Radboudumc Community for Infectious Diseases (RCI), Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Billmyre RB, Craig CJ, Lyon J, Reichardt C, Eickbush MT, Zanders SE. Saturation transposon mutagenesis enables genome-wide identification of genes required for growth and fluconazole resistance in the human fungal pathogen Cryptococcus neoformans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605507. [PMID: 39131341 PMCID: PMC11312461 DOI: 10.1101/2024.07.28.605507] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Fungi can cause devastating invasive infections, typically in immunocompromised patients. Treatment is complicated both by the evolutionary similarity between humans and fungi and by the frequent emergence of drug resistance. Studies in fungal pathogens have long been slowed by a lack of high-throughput tools and community resources that are common in model organisms. Here we demonstrate a high-throughput transposon mutagenesis and sequencing (TN-seq) system in Cryptococcus neoformans that enables genome-wide determination of gene essentiality. We employed a random forest machine learning approach to classify the Cryptococcus neoformans genome as essential or nonessential, predicting 1,465 essential genes, including 302 that lack human orthologs. These genes are ideal targets for new antifungal drug development. TN-seq also enables genome-wide measurement of the fitness contribution of genes to phenotypes of interest. As proof of principle, we demonstrate the genome-wide contribution of genes to growth in fluconazole, a clinically used antifungal. We show a novel role for the well-studied RIM101 pathway in fluconazole susceptibility. We also show that 5' insertions of transposons can drive sensitization of essential genes, enabling screenlike assays of both essential and nonessential components of the genome. Using this approach, we demonstrate a role for mitochondrial function in fluconazole sensitivity, such that tuning down many essential mitochondrial genes via 5' insertions can drive resistance to fluconazole. Our assay system will be valuable in future studies of C. neoformans, particularly in examining the consequences of genotypic diversity.
Collapse
Affiliation(s)
- R. Blake Billmyre
- Department of Pharmaceutical and Biological Sciences, College of Pharmacy, University of Georgia, GA, United States
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, GA, United States
- Department of Microbiology, Franklin College of Arts and Sciences, University of Georgia, GA, United States
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, GA, United States
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Joshua Lyon
- Department of Pharmaceutical and Biological Sciences, College of Pharmacy, University of Georgia, GA, United States
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, GA, United States
| | - Claire Reichardt
- Department of Pharmaceutical and Biological Sciences, College of Pharmacy, University of Georgia, GA, United States
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, GA, United States
- Department of Microbiology, Franklin College of Arts and Sciences, University of Georgia, GA, United States
| | | | - Sarah E. Zanders
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, KS, United States
| |
Collapse
|
21
|
Wang X, Zhou WT, Dong HH, Li CY, Jiang YY, Xie P, Xu ZY, Xie SH, Yang SX, Huang L, Chen H, Wang LY, Wei X, Huang YQ. Isobavachalcone: A redox antifungal agent impairs the mitochondria protein of Cryptococcus neoformans. Int J Antimicrob Agents 2024; 64:107253. [PMID: 38925229 DOI: 10.1016/j.ijantimicag.2024.107253] [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: 07/27/2023] [Revised: 04/30/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Isobavachalcone (IBC) is a natural small molecule with various biological activities; however, its inhibitory effects on Cryptococcus neoformans remain unclear. In our study, IBC showed a good antifungal effect. Through in vitro experiments, its minimum inhibitory concentration was 0.5-1 µg/mL. It exhibited the same antifungal effect as Amphotericin B in brain and lung infections in in vivo experiments. IBC also showed a synergistic antifungal effect with emodin with lower toxicity, and C. neoformans did not develop drug resistance to IBC. In the mechanistic study, significantly damaged mitochondria of C. neoformans, a significant reduction in mitochondrial membrane potential and adenosine triphosphate production, and an increase in hydrogen peroxide (H2O2) caused by IBC were observed using transmission electron microscopy. Through drug affinity-responsive target stability combined with phenotype detection, riboflavin synthases of aconitase and succinate dehydrogenase were screened. Molecular docking, quantitative polymerase chain reaction experiments, target inhibitor and agonist intervention, molecular interaction measurements, and minimum inhibitory concentration detection of the constructed expression strains revealed that IBC targeted the activity of these two enzymes, interfered by the tricarboxylic acid cycle, inhibited the production of adenosine triphosphate, blocked electron transport, reduced mitochondrial membrane potential, and induced antioxidation imbalance and reactive oxygen species accumulation, thus producing an antifungal effect. Therefore, IBC is a promising lead drug and redox antifungal agent for C. neoformans.
Collapse
Affiliation(s)
- Xue Wang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Wen-Ting Zhou
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Hui-Hua Dong
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Chen-Yan Li
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Yu-Ying Jiang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Ping Xie
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Zhen-Yi Xu
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Shuo-Hua Xie
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China; Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Shi-Xian Yang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China
| | - Liang Huang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China
| | - Hao Chen
- Department of Pathology, Wannan Medical College, Wuhu, China
| | - Lu-Yao Wang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China.
| | - Xian Wei
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China.
| | - Yan-Qiang Huang
- Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise, China; Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
22
|
Sarpong AK, Odoi H, Boakye YD, Boamah VE, Agyare C. Resistant C. albicans implicated in recurrent vulvovaginal candidiasis (RVVC) among women in a tertiary healthcare facility in Kumasi, Ghana. BMC Womens Health 2024; 24:412. [PMID: 39030542 PMCID: PMC11264716 DOI: 10.1186/s12905-024-03217-6] [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: 06/13/2023] [Accepted: 06/19/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Vulvovaginal candidiasis is a common fungal infection that affects the female lower genital tract. This study determined the major risk factors associated with vulvovaginal infection (VVI) in the Ashanti region of Ghana and also determined the antifungal resistance patterns of Candida albicans isolates to some antifungals. METHODS Three hundred and fifty (350) high vaginal swab (HVS) samples were collected from women who presented with signs and symptoms of VVI. A structured questionnaire was administered to one hundred and seventy-two (172) of the women. HVS samples were cultured on Sabouraud dextrose agar with 2% chloramphenicol. The polymerase chain reaction was employed to confirm C. albicans. Antifungal susceptibility testing was performed and the susceptibility of C. albicans isolates to fluconazole, clotrimazole, amphotericin B, nystatin, miconazole and 5-flurocytosine were assessed. RESULTS Vaginal infection was most prevalent amongst females in their reproductive age (21 to 30 years; 63.0%). The study found a significant association between vaginal infections and some risk factors such as sexual practices (p < 0.001), antibiotic misuse (p < 0.05), poor personal hygiene (p < 0.005) and birth control methods (p < 0.049). Out of the 350 HVS samples collected, 112 yielded yeast cells with 65 (58%) identified as C. albicans. The C. albicans isolates were resistant to 5' flucytosine (100%), fluconazole (70%), voriconazole (69.2%), miconazole (58.5%) and nystatin (49.2%). C. albicans isolates were more susceptible to amphotericin B (53.8%) and clotrimazole (45.1%), although an appreciable number of isolates showed resistance (46.1% and 52.3%, respectively). CONCLUSION There should be nationwide education on all associated risk factors of VVI. Also, use of the various antifungal agents in vaginal candidiasis should proceed after antifungal susceptibility testing to ensure efficacious use of these agents.
Collapse
Affiliation(s)
- Abena Kyeraa Sarpong
- Pharmaceutical Microbiology Section, Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Laboratory Technology Department , Kumasi Technical University, Kumasi, Ghana
| | - Hayford Odoi
- Department of Pharmaceutical Microbiology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Yaw Duah Boakye
- Pharmaceutical Microbiology Section, Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Vivian Etsiapa Boamah
- Pharmaceutical Microbiology Section, Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
- Pharmaceutical Microbiology Section, Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | - Christian Agyare
- Pharmaceutical Microbiology Section, Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
23
|
Santana de Carvalho D, Bastos RW, Rossato L, Teixeira de Aguiar Peres N, Assis Santos D. ResFungi: A Novel Protein Database of Antifungal Drug Resistance Genes Using a Hidden Markov Model Profile. ACS OMEGA 2024; 9:30559-30570. [PMID: 39035910 PMCID: PMC11256324 DOI: 10.1021/acsomega.4c02198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Fungal infections vary from superficial to invasive and can be life-threatening in immunocompromised and healthy individuals. Antifungal resistance is one of the main reasons for an increasing concern about fungal infections as they become more complex and harder to treat. The fungal "omics" databases help us find drug resistance genes, which is of great importance and extremely necessary. With that in mind, we built a new platform for drug resistance genes. We added seven drug classes of resistance genes to our database: azoles (without specifying which drug), fluconazole, voriconazole, itraconazole, flucytosine, micafungin, and caspofungin. Species with known resistance genes were used to validate the results from our database. This study describes a list of 261 candidate genes related to antifungal resistance, with several genes displaying transport functions involved in azole resistance. Over 65% of the candidate genes found were related to at least one type of azole. Overall, the candidate genes found have functional annotations consistent with genes or enzymes that have been linked to antifungal resistance in previous studies. Also, candidate antifungal resistance genes found exhibit functional annotations consistent with previously described resistance mechanisms. The existence of an HMM profile focusing on antifungal resistance genes allows in silico searches for candidate genes, helping future wet lab experiments, and hence, reducing costs when studying candidate antifungal genes without prior knowledge of the species or genes. Finally, ResFungi has proven to be a powerful tool to narrow down candidate antifungal-related genes and unravel mechanisms related to resistance to help in the design of experiments focusing on the genetic basis of antifungal resistance.
Collapse
Affiliation(s)
- Daniel Santana de Carvalho
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rafael Wesley Bastos
- Bioscience Center, Federal University of Rio Grande do Norte, 59064-741 Natal, Brazil
| | - Luana Rossato
- Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, 79825-070 Dourados, Brazil
| | - Nalu Teixeira de Aguiar Peres
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Assis Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
24
|
Zhang ZH, Sun LL, Fu BQ, Deng J, Jia CL, Miao MX, Yang F, Cao YB, Yan TH. Aneuploidy underlies brefeldin A-induced antifungal drug resistance in Cryptococcus neoformans. Front Cell Infect Microbiol 2024; 14:1397724. [PMID: 38966251 PMCID: PMC11222406 DOI: 10.3389/fcimb.2024.1397724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024] Open
Abstract
Cryptococcus neoformans is at the top of the list of "most wanted" human pathogens. Only three classes of antifungal drugs are available for the treatment of cryptococcosis. Studies on antifungal resistance mechanisms are limited to the investigation of how a particular antifungal drug induces resistance to a particular drug, and the impact of stresses other than antifungals on the development of antifungal resistance and even cross-resistance is largely unexplored. The endoplasmic reticulum (ER) is a ubiquitous subcellular organelle of eukaryotic cells. Brefeldin A (BFA) is a widely used chemical inducer of ER stress. Here, we found that both weak and strong selection by BFA caused aneuploidy formation in C. neoformans, mainly disomy of chromosome 1, chromosome 3, and chromosome 7. Disomy of chromosome 1 conferred cross-resistance to two classes of antifungal drugs: fluconazole and 5-flucytosine, as well as hypersensitivity to amphotericin B. However, drug resistance was unstable, due to the intrinsic instability of aneuploidy. We found overexpression of AFR1 on Chr1 and GEA2 on Chr3 phenocopied BFA resistance conferred by chromosome disomy. Overexpression of AFR1 also caused resistance to fluconazole and hypersensitivity to amphotericin B. Furthermore, a strain with a deletion of AFR1 failed to form chromosome 1 disomy upon BFA treatment. Transcriptome analysis indicated that chromosome 1 disomy simultaneously upregulated AFR1, ERG11, and other efflux and ERG genes. Thus, we posit that BFA has the potential to drive the rapid development of drug resistance and even cross-resistance in C. neoformans, with genome plasticity as the accomplice.
Collapse
Affiliation(s)
- Zhi-hui Zhang
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu-liu Sun
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bu-qing Fu
- Laboratory Department, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jie Deng
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng-lin Jia
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming-xing Miao
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng Yang
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yong-bing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
25
|
Meng Y, Ni Y, Li Z, Jiang T, Sun T, Li Y, Gao X, Li H, Suo C, Li C, Yang S, Lan T, Liao G, Liu T, Wang P, Ding C. Interplay between acetylation and ubiquitination of imitation switch chromatin remodeler Isw1 confers multidrug resistance in Cryptococcus neoformans. eLife 2024; 13:e85728. [PMID: 38251723 PMCID: PMC10834027 DOI: 10.7554/elife.85728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/21/2024] [Indexed: 01/23/2024] Open
Abstract
Cryptococcus neoformans poses a threat to human health, but anticryptococcal therapy is hampered by the emergence of drug resistance, whose underlying mechanisms remain poorly understood. Herein, we discovered that Isw1, an imitation switch chromatin remodeling ATPase, functions as a master modulator of genes responsible for in vivo and in vitro multidrug resistance in C. neoformans. Cells with the disrupted ISW1 gene exhibited profound resistance to multiple antifungal drugs. Mass spectrometry analysis revealed that Isw1 is both acetylated and ubiquitinated, suggesting that an interplay between these two modification events exists to govern Isw1 function. Mutagenesis studies of acetylation and ubiquitination sites revealed that the acetylation status of Isw1K97 coordinates with its ubiquitination processes at Isw1K113 and Isw1K441 through modulating the interaction between Isw1 and Cdc4, an E3 ligase. Additionally, clinical isolates of C. neoformans overexpressing the degradation-resistant ISW1K97Q allele showed impaired drug-resistant phenotypes. Collectively, our studies revealed a sophisticated acetylation-Isw1-ubiquitination regulation axis that controls multidrug resistance in C. neoformans.
Collapse
Affiliation(s)
- Yang Meng
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Yue Ni
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Zhuoran Li
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Tianhang Jiang
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Tianshu Sun
- Department of Scientific Research, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yanjian Li
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Xindi Gao
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Hailong Li
- NHC Key Laboratory of AIDS Immunology, The First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Chenhao Suo
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Chao Li
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Sheng Yang
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Tian Lan
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| | - Guojian Liao
- College of Pharmaceutical Sciences, Southwest UniversityChongqingChina
| | - Tongbao Liu
- Medical Research Institute, Southwest UniversityChongqingChina
| | - Ping Wang
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New OrleansNew OrleansUnited States
| | - Chen Ding
- College of Life and Health Sciences, Northeastern UniversityShenyangChina
| |
Collapse
|
26
|
Yu J, Liu X, Guo D, Yang W, Chen X, Zou G, Wang T, Pang S, Zhang G, Dong J, Xu Y, Zhao Y, on behalf of the National China Hospital Invasive Fungal Surveillance Network (CHIF-NET). Antifungal susceptibility profile and local epidemiological cut-off values of Yarrowia ( Candida) lipolytica: an emergent and rare opportunistic yeast. Microbiol Spectr 2024; 12:e0320323. [PMID: 38084981 PMCID: PMC10783140 DOI: 10.1128/spectrum.03203-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/15/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE Yarrowia lipolytica, also known as Candida lipolytica, is an emerging opportunistic "rare pathogenic yeast". Due to the limited data on its antifungal susceptibility, the clinical treatments become challenging. Based on the China Hospital Invasive Fungal Surveillance Network (2009-2022), we conducted a comprehensive multi-method study on clinical isolates from various central hospitals. This study is currently the largest study carried out to assess the antifungal susceptibility of Y. lipolytica. It is also the first to establish local epidemiological cut-off values (L-ECOFFs), identify its ERG11 mutations, and assess the consistency between the three prevalent commercial antifungal susceptibility testing methods and the broth microdilution method. We recommend the Sensititre YeastOne as the best option for antifungal susceptibility testing for Y. lipolytica, followed by the ATB FUNGUS 3. Nevertheless, practitioners should use the MIC test strip with discretion.
Collapse
Affiliation(s)
- Jinhan Yu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xueqing Liu
- Department of Clinical Laboratory, Yongzhou Central Hospital, Yongzhou, China
| | - Dawen Guo
- Department of Microbiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wenhang Yang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Xinfei Chen
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Guiling Zou
- Department of Microbiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tong Wang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Shichao Pang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Ge Zhang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Jingjing Dong
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Ying Zhao
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - on behalf of the National China Hospital Invasive Fungal Surveillance Network (CHIF-NET)
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Clinical Laboratory, Yongzhou Central Hospital, Yongzhou, China
- Department of Microbiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Microbiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| |
Collapse
|
27
|
Boakye-Yiadom E, Odoom A, Osman AH, Ntim OK, Kotey FCN, Ocansey BK, Donkor ES. Fungal Infections, Treatment and Antifungal Resistance: The Sub-Saharan African Context. Ther Adv Infect Dis 2024; 11:20499361241297525. [PMID: 39544852 PMCID: PMC11562003 DOI: 10.1177/20499361241297525] [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: 01/17/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
Fungal pathogens cause a wide range of infections in humans, from superficial to disfiguring, allergic syndromes, and life-threatening invasive infections, affecting over a billion individuals globally. With an estimated 1.5 million deaths annually attributable to them, fungal pathogens are a major cause of mortality in humans, especially people with underlying immunosuppression. The continuous increase in the population of individuals at risk of fungal infections in sub-Saharan Africa, such as HIV patients, tuberculosis patients, intensive care patients, patients with haematological malignancies, transplant (haematopoietic stem cell and organ) recipients and the growing global threat of multidrug-resistant fungal strains, raise the need for an appreciation of the region's perspective on antifungal usage and resistance. In addition, the unavailability of recently introduced novel antifungal drugs in sub-Saharan Africa further calls for regular evaluation of resistance to antifungal agents in these settings. This is critical for ensuring appropriate and optimal use of the limited available arsenal to minimise antifungal resistance. This review, therefore, elaborates on the multifaceted nature of fungal resistance to the available antifungal drugs on the market and further provides insights into the prevalence of fungal infections and the use of antifungal agents in sub-Saharan Africa.
Collapse
Affiliation(s)
- Emily Boakye-Yiadom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
- Department of Microbiology and Immunology, University of Health and Allied Sciences, Ho, Ghana
| | - Alex Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Onyansaniba K. Ntim
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Fleischer C. N. Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Bright K. Ocansey
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, P.O. Box KB 4236, Ghana
| |
Collapse
|
28
|
Zhu X, Chen Y, Yu D, Fang W, Liao W, Pan W. Progress in the application of nanoparticles for the treatment of fungal infections: A review. Mycology 2023; 15:1-16. [PMID: 38558835 PMCID: PMC10977003 DOI: 10.1080/21501203.2023.2285764] [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: 05/21/2023] [Accepted: 10/28/2023] [Indexed: 04/04/2024] Open
Abstract
The burden of fungal infections on human health is increasing worldwide. Aspergillus, Candida, and Cryptococcus are the top three human pathogenic fungi that are responsible for over 90% of infection-related deaths. Moreover, effective antifungal therapeutics are lacking, primarily due to host toxicity, pathogen resistance, and immunodeficiency. In recent years, nanomaterials have proved not only to be more efficient antifungal therapeutic agents but also to overcome resistance against fungal medication. This review will examine the limitations of standard antifungal therapy as well as focus on the development of nanomaterials.
Collapse
Affiliation(s)
- Xinlin Zhu
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Youming Chen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Dan Yu
- Department of General Practice, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wenjie Fang
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wanqing Liao
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weihua Pan
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
29
|
Boyce KJ. The Microevolution of Antifungal Drug Resistance in Pathogenic Fungi. Microorganisms 2023; 11:2757. [PMID: 38004768 PMCID: PMC10673521 DOI: 10.3390/microorganisms11112757] [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: 10/11/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
The mortality rates of invasive fungal infections remain high because of the limited number of antifungal drugs available and antifungal drug resistance, which can rapidly evolve during treatment. Mutations in key resistance genes such as ERG11 were postulated to be the predominant cause of antifungal drug resistance in the clinic. However, recent advances in whole genome sequencing have revealed that there are multiple mechanisms leading to the microevolution of resistance. In many fungal species, resistance can emerge through ERG11-independent mechanisms and through the accumulation of mutations in many genes to generate a polygenic resistance phenotype. In addition, genome sequencing has revealed that full or partial aneuploidy commonly occurs in clinical or microevolved in vitro isolates to confer antifungal resistance. This review will provide an overview of the mutations known to be selected during the adaptive microevolution of antifungal drug resistance and focus on how recent advances in genome sequencing technology have enhanced our understanding of this process.
Collapse
Affiliation(s)
- Kylie J Boyce
- School of Science, RMIT University, Melbourne, VIC 3085, Australia
| |
Collapse
|
30
|
Wang Y, Guo X, Zhang X, Chen P, Wang W, Hu S, Ma T, Zhou X, Li D, Yang Y. In Vivo Microevolutionary Analysis of a Fatal Case of Rhinofacial and Disseminated Mycosis Due to Azole-Drug-Resistant Candida Species. J Fungi (Basel) 2023; 9:815. [PMID: 37623586 PMCID: PMC10455694 DOI: 10.3390/jof9080815] [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: 06/27/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Ten Candida species strains were isolated from the first known fatal case of rhinofacial and rhino-orbital-cerebral candidiasis. Among them, five strains of Candida parapsilosis complex were isolated during the early stage of hospitalization, while five strains of Candida tropicalis were isolated in the later stages of the disease. Using whole-genome sequencing, we distinguished the five strains of C. parapsilosis complex as four Candida metapsilosis strains and one Candida parapsilosis strain. Antifungal susceptibility testing showed that the five strains of C. parapsilosis complex were susceptible to all antifungal drugs, while five C. tropicalis strains had high minimum inhibitory concentrations to azoles, whereas antifungal-drug resistance gene analysis revealed the causes of azole resistance in such strains. For the first time, we analyzed the microevolutionary characteristics of pathogenic fungi in human hosts and inferred the infection time and parallel evolution of C. tropicalis strains. Molecular clock analysis revealed that azole-resistant C. tropicalis infection occurred during the first round of therapy, followed by divergence via parallel evolution in vivo. The presence/absence variations indicated a potential decrease in the virulence of genomes in strains isolated following antifungal drug treatment, despite the absence of observed clinical improvement in the conditions of the patient. These results suggest that genomic analysis could serve as an auxiliary tool in guiding clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Yuchen Wang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
| | - Xi Guo
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjing 300457, China;
| | - Xinran Zhang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
| | - Ping Chen
- Division of Dermatology and Mycological Lab, Peking University Third Hospital, Beijing 100191, China
| | - Wenhui Wang
- Division of Dermatology and Mycological Lab, Peking University Third Hospital, Beijing 100191, China
| | - Shan Hu
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
| | - Teng Ma
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
| | - Xingchen Zhou
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
- School of Life Science & Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Dongming Li
- Division of Dermatology and Mycological Lab, Peking University Third Hospital, Beijing 100191, China
| | - Ying Yang
- Bioinformatics Center of AMMS, Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Microbiology and Epidemiology, Beijing 100850, China; (Y.W.); (X.Z.)
| |
Collapse
|
31
|
Sousa NSOD, Almeida JDRD, Frickmann H, Lacerda MVG, Souza JVBD. Searching for new antifungals for the treatment of cryptococcosis. Rev Soc Bras Med Trop 2023; 56:e01212023. [PMID: 37493736 PMCID: PMC10367226 DOI: 10.1590/0037-8682-0121-2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/19/2023] [Indexed: 07/27/2023] Open
Abstract
There is a consensus that the antifungal repertoire for the treatment of cryptococcal infections is limited. Standard treatment involves the administration of an antifungal drug derived from natural sources (i.e., amphotericin B) and two other drugs developed synthetically (i.e., flucytosine and fluconazole). Despite treatment, the mortality rates associated with fungal cryptococcosis are high. Amphotericin B and flucytosine are toxic, require intravenous administration, and are usually unavailable in low-income countries because of their high cost. However, fluconazole is cost-effective, widely available, and harmless with regard to its side effects. However, fluconazole is a fungistatic agent that has contributed considerably to the increase in fungal resistance and frequent relapses in patients with cryptococcal meningitis. Therefore, there is an unquestionable need to identify new alternatives or adjuvants to conventional drugs for the treatment of cryptococcosis. A potential antifungal agent should be able to kill cryptococci and "bypass" the virulence mechanism of the yeast. Furthermore, it should have fungicidal action, low toxicity, high selectivity, easily penetrate the central nervous system, and widely available. In this review, we describe cryptococcosis, its conventional therapy, and failures arising from the use of drugs traditionally considered to be the reference standard. Additionally, we present the approaches used for the discovery of new drugs to counteract cryptococcosis, ranging from the conventional screening of natural products to the inclusion of structural modifications to optimize anticryptococcal activity, as well as drug repositioning and combined therapies.
Collapse
Affiliation(s)
| | | | - Hagen Frickmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Germany
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Germany
| | - Marcus Vinícius Guimarães Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, AM, Brasil
- Instituto de Pesquisas Leônidas & Maria Deane, Fiocruz, Manaus, AM, Brasil
- University of Texas Medical Branch, Galveston, USA
| | - João Vicente Braga de Souza
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Rede BIONORTE, Manaus, AM, Brasil
- Instituto Nacional de Pesquisas da Amazônia, Manaus, AM, Brasil
| |
Collapse
|
32
|
Zhu P, Li Y, Guo T, Liu S, Tancer RJ, Hu C, Zhao C, Xue C, Liao G. New antifungal strategies: drug combination and co-delivery. Adv Drug Deliv Rev 2023; 198:114874. [PMID: 37211279 DOI: 10.1016/j.addr.2023.114874] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
The growing occurrence of invasive fungal infections and the mounting rates of drug resistance constitute a significant menace to human health. Antifungal drug combinations have garnered substantial interest for their potential to improve therapeutic efficacy, reduce drug doses, reverse, or ameliorate drug resistance. A thorough understanding of the molecular mechanisms underlying antifungal drug resistance and drug combination is key to developing new drug combinations. Here we discuss the mechanisms of antifungal drug resistance and elucidate how to discover potent drug combinations to surmount resistance. We also examine the challenges encountered in developing such combinations and discuss prospects, including advanced drug delivery strategies.
Collapse
Affiliation(s)
- Ping Zhu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Yan Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Ting Guo
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Simei Liu
- Department of Traditional Chinese Medicine, Chongqing College of Traditional Chinese Medicine, Chongqing 402760, China; Institute of Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Robert J Tancer
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Changhua Hu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Chengzhi Zhao
- Chongqing Health Center for Women and Children, Chongqing, 400700, PR China.
| | - Chaoyang Xue
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Guojian Liao
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China.
| |
Collapse
|
33
|
Carmo A, Rocha M, Pereirinha P, Tomé R, Costa E. Antifungals: From Pharmacokinetics to Clinical Practice. Antibiotics (Basel) 2023; 12:884. [PMID: 37237787 PMCID: PMC10215229 DOI: 10.3390/antibiotics12050884] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The use of antifungal drugs started in the 1950s with polyenes nystatin, natamycin and amphotericin B-deoxycholate (AmB). Until the present day, AmB has been considered to be a hallmark in the treatment of invasive systemic fungal infections. Nevertheless, the success and the use of AmB were associated with severe adverse effects which stimulated the development of new antifungal drugs such as azoles, pyrimidine antimetabolite, mitotic inhibitors, allylamines and echinochandins. However, all of these drugs presented one or more limitations associated with adverse reactions, administration route and more recently the development of resistance. To worsen this scenario, there has been an increase in fungal infections, especially in invasive systemic fungal infections that are particularly difficult to diagnose and treat. In 2022, the World Health Organization (WHO) published the first fungal priority pathogens list, alerting people to the increased incidence of invasive systemic fungal infections and to the associated risk of mortality/morbidity. The report also emphasized the need to rationally use existing drugs and develop new drugs. In this review, we performed an overview of the history of antifungals and their classification, mechanism of action, pharmacokinetic/pharmacodynamic (PK/PD) characteristics and clinical applications. In parallel, we also addressed the contribution of fungi biology and genetics to the development of resistance to antifungal drugs. Considering that drug effectiveness also depends on the mammalian host, we provide an overview on the roles of therapeutic drug monitoring and pharmacogenomics as means to improve the outcome, prevent/reduce antifungal toxicity and prevent the emergence of antifungal resistance. Finally, we present the new antifungals and their main characteristics.
Collapse
Affiliation(s)
- Anália Carmo
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| | - Marilia Rocha
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Pharmacy Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal (P.P.)
| | - Patricia Pereirinha
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Pharmacy Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal (P.P.)
| | - Rui Tomé
- Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal;
| | - Eulália Costa
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| |
Collapse
|
34
|
Li-Zhulanov NS, Zaikova NP, Sari S, Gülmez D, Sabuncuoğlu S, Ozadali-Sari K, Arikan-Akdagli S, Nefedov AA, Rybalova TV, Volcho KP, Salakhutdinov NF. Rational Design of New Monoterpene-Containing Azoles and Their Antifungal Activity. Antibiotics (Basel) 2023; 12:antibiotics12050818. [PMID: 37237723 DOI: 10.3390/antibiotics12050818] [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: 04/10/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Azole antifungals, including fluconazole, have long been the first-line antifungal agents in the fight against fungal infections. The emergence of drug-resistant strains and the associated increase in mortality from systemic mycoses has prompted the development of new agents based on azoles. We reported a synthesis of novel monoterpene-containing azoles with high antifungal activity and low cytotoxicity. These hybrids demonstrated broad-spectrum activity against all tested fungal strains, with excellent minimum inhibitory concentration (MIC) values against both fluconazole-susceptible and fluconazole-resistant strains of Candida spp. Compounds 10a and 10c with cuminyl and pinenyl fragments demonstrated up to 100 times lower MICs than fluconazole against clinical isolates. The results indicated that the monoterpene-containing azoles had much lower MICs against fluconazole-resistant clinical isolates of Candida parapsilosis than their phenyl-containing counterpart. In addition, the compounds did not exhibit cytotoxicity at active concentrations in the MTT assay, indicating potential for further development as antifungal agents.
Collapse
Affiliation(s)
- Nikolai S Li-Zhulanov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Nadezhda P Zaikova
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Suat Sari
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Sihhiye, Ankara 06100, Turkey
| | - Dolunay Gülmez
- Department of Medical Microbiology, Hacettepe University Faculty of Medicine, Sihhiye, Ankara 06100, Turkey
| | - Suna Sabuncuoğlu
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Sihhiye, Ankara 06100, Turkey
| | - Keriman Ozadali-Sari
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Sihhiye, Ankara 06100, Turkey
| | - Sevtap Arikan-Akdagli
- Department of Medical Microbiology, Hacettepe University Faculty of Medicine, Sihhiye, Ankara 06100, Turkey
| | - Andrey A Nefedov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Tatyana V Rybalova
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Konstantin P Volcho
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Nariman F Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| |
Collapse
|
35
|
de Oliveira H, Bezerra BT, Rodrigues ML. Antifungal Development and the Urgency of Minimizing the Impact of Fungal Diseases on Public Health. ACS BIO & MED CHEM AU 2023; 3:137-146. [PMID: 37101810 PMCID: PMC10125384 DOI: 10.1021/acsbiomedchemau.2c00055] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 04/28/2023]
Abstract
Fungal infections are a major public health problem resulting from the lack of public policies addressing these diseases, toxic and/or expensive therapeutic tools, scarce diagnostic tests, and unavailable vaccines. In this Perspective, we discuss the need for novel antifungal alternatives, highlighting new initiatives based on drug repurposing and the development of novel antifungals.
Collapse
Affiliation(s)
| | - Bárbara T. Bezerra
- Instituto
Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba81310-020, Brazil
| | - Marcio L. Rodrigues
- Instituto
Carlos Chagas, Fundação Oswaldo Cruz (Fiocruz), Curitiba81310-020, Brazil
- Instituto
de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, Rio de Janeiro21941-902, Brazil
| |
Collapse
|
36
|
Zhao Y, Ye L, Zhao F, Zhang L, Lu Z, Chu T, Wang S, Liu Z, Sun Y, Chen M, Liao G, Ding C, Xu Y, Liao W, Wang L. Cryptococcus neoformans, a global threat to human health. Infect Dis Poverty 2023; 12:20. [PMID: 36932414 PMCID: PMC10020775 DOI: 10.1186/s40249-023-01073-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Emerging fungal pathogens pose important threats to global public health. The World Health Organization has responded to the rising threat of traditionally neglected fungal infections by developing a Fungal Priority Pathogens List (FPPL). Taking the highest-ranked fungal pathogen in the FPPL, Cryptococcus neoformans, as a paradigm, we review progress made over the past two decades on its global burden, its clinical manifestation and management of cryptococcal infection, and its antifungal resistance. The purpose of this review is to drive research efforts to improve future diagnoses, therapies, and interventions associated with fungal infections. METHODS We first reviewed trends in the global burden of HIV-associated cryptococcal infection, mainly based on a series of systematic studies. We next conducted scoping reviews in accordance with the guidelines described in the Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for Scoping Reviews using PubMed and ScienceDirect with the keyword Cryptococcus neoformans to identify case reports of cryptococcal infections published since 2000. We then reviewed recent updates on the diagnosis and antifungal treatment of cryptococcal infections. Finally, we summarized knowledge regarding the resistance and tolerance of C. neoformans to approved antifungal drugs. RESULTS There has been a general reduction in the estimated global burden of HIV-associated cryptococcal meningitis since 2009, probably due to improvements in highly active antiretroviral therapies. However, cryptococcal meningitis still accounts for 19% of AIDS-related deaths annually. The incidences of CM in Europe and North America and the Latin America region have increased by approximately two-fold since 2009, while other regions showed either reduced or stable numbers of cases. Unfortunately, diagnostic and treatment options for cryptococcal infections are limited, and emerging antifungal resistance exacerbates the public health burden. CONCLUSION The rising threat of C. neoformans is compounded by accumulating evidence for its ability to infect immunocompetent individuals and the emergence of antifungal-resistant variants. Emphasis should be placed on further understanding the mechanisms of pathogenicity and of antifungal resistance and tolerance. The development of novel management strategies through the identification of new drug targets and the discovery and optimization of new and existing diagnostics and therapeutics are key to reducing the health burden.
Collapse
Affiliation(s)
- Youbao Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China.
| | - Leixin Ye
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Fujie Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Lanyue Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Zhenguo Lu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Tianxin Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Siyu Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Zhanxiang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Yukai Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Min Chen
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Changzheng Hospital, Shanghai, 200003, China
| | - Guojian Liao
- The Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Yingchun Xu
- Department of Laboratory Medicine, and Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Wanqing Liao
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Changzheng Hospital, Shanghai, 200003, China
| | - Linqi Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100039, China.
| |
Collapse
|
37
|
Zheng L, Xu Y, Dong Y, Ma X, Wang C, Yang F, Guo L. Chromosome 1 trisomy confers resistance to aureobasidin A in Candida albicans. Front Microbiol 2023; 14:1128160. [PMID: 37007527 PMCID: PMC10063858 DOI: 10.3389/fmicb.2023.1128160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
IntroductionCandida albicans is a prevalent opportunistic human fungal pathogen. However, there are currently very few antifungal treatments available. Inositol phosphoryl ceramide synthase is an essential and fungal-specific protein that also provides a novel and promising antifungal target. Aureobasidin A is a widely used inhibitor of inositol phosphoryl ceramide synthase, however the mechanism of resistance to aureobasidin A is largely unknown in pathogenic fungi.MethodsHere we investigated how C. albicans adapted to low and high concentrations of aureobasidin A.Results and discussionsWe identified trisomy of chromosome 1 as the predominant mechanism of rapid adaptation. Resistance to aureobasidin A was unstable because of the inherent instability of aneuploids. Importantly, chromosome 1 trisomy simultaneously regulated genes which were associated with aureobasidin A resistance that are on this aneuploid chromosome as well as on other chromosomes. Furthermore, the pleiotropic effect of aneuploidy caused altered resistance not only to aureobasidin A but also to other antifungal drugs including caspofungin and 5-flucytosine. We posit aneuploidy provides a rapid and reversible mechanism of development of drug resistance and cross resistance in C. albicans.
Collapse
Affiliation(s)
- Lijun Zheng
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Xu
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Yubo Dong
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Xiaowen Ma
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Chen Wang
- Department of Pharmacy, The 960 Hospital of PLA, Jinan, China
| | - Feng Yang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liangsheng Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Liangsheng Guo,
| |
Collapse
|
38
|
Factors Influencing the Nitrogen-Source Dependent Flucytosine Resistance in Cryptococcus Species. mBio 2023; 14:e0345122. [PMID: 36656038 PMCID: PMC9973006 DOI: 10.1128/mbio.03451-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Flucytosine (5-FC) is an antifungal agent commonly used for treatment of cryptococcosis and several other systemic mycoses. In fungi, cytosine permease and cytosine deaminase are known major players in flucytosine resistance by regulating uptake and deamination of 5-FC, respectively. Cryptococcus species have three paralogs each of cytosine permease (FCY2, FCY3, and FCY4) and cytosine deaminase (FCY1, FCY5 and FCY6). As in other fungi, we found FCY1 and FCY2 to be the primary cytosine deaminase and permease gene, respectively, in C. neoformans H99 (VNI), C. gattii R265 (VGIIa) and WM276 (VGI). However, when various amino acids were used as the sole nitrogen source, C. neoformans and C. gattii diverged in the function of FCY3 and FCY6. Though there was some lineage-dependent variability, the two genes functioned as the secondary permease and deaminase, respectively, only in C. gattii when the nitrogen source was arginine, asparagine, or proline. Additionally, the expression of FCY genes, excluding FCY1, was under nitrogen catabolic repression in the presence of NH4. Functional analysis of GAT1 and CIR1 gene deletion constructs demonstrated that these two genes regulate the expression of each permease and deaminase genes individually. Furthermore, the expression levels of FCY3 and FCY6 under different amino acids corroborated the 5-FC susceptibility in fcy2Δ or fcy1Δ background. Thus, the mechanism of 5-FC resistance in C. gattii under diverse nitrogen conditions is orchestrated by two transcription factors of GATA family, cytosine permease and deaminase genes. IMPORTANCE 5-FC is a commonly used antifungal drug for treatment of cryptococcosis caused by Cryptococcus neoformans and C. gattii species complexes. When various amino acids were used as the sole nitrogen source for growth, we found lineage dependent differences in 5-FC susceptibility. Deletion of the classical cytosine permease (FCY2) and deaminase (FCY1) genes caused increased 5-FC resistance in all tested nitrogen sources in C. neoformans but not in C. gattii. Furthermore, we demonstrate that the two GATA family transcription factor genes GAT1 and CIR1 are involved in the nitrogen-source dependent 5-FC resistance by regulating the expression of the paralogs of cytosine permease and deaminase genes. Our study not only identifies the new function of paralogs of the cytosine permease and deaminase and the role of their regulatory transcription factors but also denotes the differences in the mechanism of 5-FC resistance among the two etiologic agents of cryptococcosis under different nitrogen sources.
Collapse
|
39
|
Deng H, Song J, Huang Y, Yang C, Zang X, Zhou Y, Li H, Dai B, Xue X. Combating increased antifungal drug resistance in Cryptococcus, what should we do in the future? Acta Biochim Biophys Sin (Shanghai) 2023; 55:540-547. [PMID: 36815374 PMCID: PMC10195138 DOI: 10.3724/abbs.2023011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/08/2022] [Indexed: 02/05/2023] Open
Abstract
Few therapeutic drugs and increased drug resistance have aggravated the current treatment difficulties of Cryptococcus in recent years. To better understand the antifungal drug resistance mechanism and treatment strategy of cryptococcosis. In this review, by combining the fundamental features of Cryptococcus reproduction leading to changes in its genome, we review recent research into the mechanism of four current anti-cryptococcal agents, coupled with new therapeutic strategies and the application of advanced technologies WGS and CRISPR-Cas9 in this field, hoping to provide a broad idea for the future clinical therapy of cryptococcosis.
Collapse
Affiliation(s)
- Hengyu Deng
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Jialin Song
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Yemei Huang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Chen Yang
- Department of Laboratory Medicinethe First Medical CentreChinese PLA General HospitalBeijing100853China
| | - Xuelei Zang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Yangyu Zhou
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Hongli Li
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Bin Dai
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Xinying Xue
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| |
Collapse
|
40
|
Suo C, Gao Y, Ding C, Sun T. The function and regulation of heat shock transcription factor in Cryptococcus. Front Cell Infect Microbiol 2023; 13:1195968. [PMID: 37168390 PMCID: PMC10165103 DOI: 10.3389/fcimb.2023.1195968] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Cryptococcus species are opportunistic human fungal pathogens. Survival in a hostile environment, such as the elevated body temperatures of transmitting animals and humans, is crucial for Cryptococcus infection. Numerous intriguing investigations have shown that the Hsf family of thermotolerance transcription regulators plays a crucial role in the pathogen-host axis of Cryptococcus. Although Hsf1 is known to be a master regulator of the heat shock response through the activation of gene expression of heat shock proteins (Hsps). Hsf1 and other Hsfs are multifaceted transcription regulators that regulate the expression of genes involved in protein chaperones, metabolism, cell signal transduction, and the electron transfer chain. In Saccharomyces cerevisiae, a model organism, Hsf1's working mechanism has been intensively examined. Nonetheless, the link between Hsfs and Cryptococcus pathogenicity remains poorly understood. This review will focus on the transcriptional regulation of Hsf function in Cryptococcus, as well as potential antifungal treatments targeting Hsf proteins.
Collapse
Affiliation(s)
- Chenhao Suo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yiru Gao
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, China
- *Correspondence: Tianshu Sun, ; Chen Ding,
| | - Tianshu Sun
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- *Correspondence: Tianshu Sun, ; Chen Ding,
| |
Collapse
|
41
|
Sun LL, Li H, Yan TH, Cao YB, Jiang YY, Yang F. Aneuploidy enables cross-tolerance to unrelated antifungal drugs in Candida parapsilosis. Front Microbiol 2023; 14:1137083. [PMID: 37113223 PMCID: PMC10126355 DOI: 10.3389/fmicb.2023.1137083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Candida parapsilosis is an emerging major human fungal pathogen. Echinocandins are first-line antifungal drugs for the treatment of invasive Candida infections. In clinical isolates, tolerance to echinocandins in Candida species is mostly due to point mutations of FKS genes, which encode the target protein of echinocandins. However, here, we found chromosome 5 trisomy was the major mechanism of adaptation to the echinocandin drug caspofungin, and FKS mutations were rare events. Chromosome 5 trisomy conferred tolerance to echinocandin drugs caspofungin and micafungin and cross-tolerance to 5-flucytosine, another class of antifungal drugs. The inherent instability of aneuploidy caused unstable drug tolerance. Tolerance to echinocandins might be due to increased copy number and expression of CHS7, which encodes chitin synthase. Although copy number of chitinase genes CHT3 and CHT4 was also increased to the trisomic level, the expression was buffered to the disomic level. Tolerance to 5-flucytosine might be due to the decreased expression of FUR1. Therefore, the pleiotropic effect of aneuploidy on antifungal tolerance was due to the simultaneous regulation of genes on the aneuploid chromosome and genes on euploid chromosomes. In summary, aneuploidy provides a rapid and reversible mechanism of drug tolerance and cross-tolerance in C. parapsilosis.
Collapse
Affiliation(s)
- Liu-liu Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong-bing Cao
- Department of Vascular Diseases, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-ying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Yuan-ying Jiang
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Feng Yang
| |
Collapse
|
42
|
Garvey M, Meade E, Rowan NJ. Effectiveness of front line and emerging fungal disease prevention and control interventions and opportunities to address appropriate eco-sustainable solutions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:158284. [PMID: 36029815 DOI: 10.1016/j.scitotenv.2022.158284] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/21/2022] [Accepted: 08/21/2022] [Indexed: 06/15/2023]
Abstract
Fungal pathogens contribute to significant disease burden globally; however, the fact that fungi are eukaryotes has greatly complicated their role in fungal-mediated infections and alleviation. Antifungal drugs are often toxic to host cells and there is increasing evidence of adaptive resistance in animals and humans. Existing fungal diagnostic and treatment regimens have limitations that has contributed to the alarming high mortality rates and prolonged morbidity seen in immunocompromised cohorts caused by opportunistic invasive infections as evidenced during HIV and COVID-19 pandemics. There is a need to develop real-time monitoring and diagnostic methods for fungal pathogens and to create a greater awareness as to the contribution of fungal pathogens in disease causation. Greater information is required on the appropriate selection and dose of antifungal drugs including factors governing resistance where there is commensurate need to discover more appropriate and effective solutions. Popular azole fungal drugs are widely detected in surface water and sediment due to incomplete removal in wastewater treatment plants where they are resistant to microbial degradation and may cause toxic effects on aquatic organisms such as algae and fish. UV has limited effectiveness in destruction of anti-fungal drugs where there is increased interest in the combination approaches such as novel use of pulsed-plasma gas-discharge technologies for environmental waste management. There is growing interest in developing alternative and complementary green eco-biocides and disinfection innovation. Fungi present challenges for cleaning, disinfection and sterilization of reusable medical devices such as endoscopes where they (example, Aspergillus and Candida species) can be protected when harboured in build-up biofilm from lethal processing. Information on the efficacy of established disinfection and sterilization technologies to address fungal pathogens including bottleneck areas that present high risk to patients is lacking. There is a need to address risk mitigation and modelling to inform efficacy of appropriate intervention technologies that must consider all contributing factors where there is potential to adopt digital technologies to enable real-time analysis of big data, such as use of artificial intelligence and machine learning. International consensus on standardised protocols for developing and reporting on appropriate alternative eco-solutions must be reached, particularly in order to address fungi with increasing drug resistance where research and innovation can be enabled using a One Health approach.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, Sligo, Ireland; Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, Sligo, Ireland
| | - Elaine Meade
- Department of Life Science, Atlantic Technological University, Sligo, Ireland; Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, Sligo, Ireland
| | - Neil J Rowan
- Bioscience Research Institute, Technological University of the Shannon Midlands Midwest, Athlone, Ireland; Centre for Decontamination, Sterilization and Biosecurity, Technological University of the Shannon Midlands Midwest, Athlone, Ireland; Empower Eco Sustainability Hub, Technological University of the Shannon Midlands Midwest, Athlone, Ireland.
| |
Collapse
|
43
|
Huang JJ, Chen XF, Tsui CKM, Pang CJ, Hu ZD, Shi Y, Wang WP, Cui LY, Xiao YL, Gong J, Fan X, Li YX, Zhang G, Xiao M, Xu YC. Persistence of an epidemic cluster of Rhodotorula mucilaginosa in multiple geographic regions in China and the emergence of a 5-flucytosine resistant clone. Emerg Microbes Infect 2022; 11:1079-1089. [PMID: 35343400 PMCID: PMC9009924 DOI: 10.1080/22221751.2022.2059402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rhodotorula mucilaginosa, an environmental yeast widely used in industry and agriculture, is also an opportunistic pathogen resistant to multi-antifungals. During the national surveillance in China, R. mucilaginosa has been documented from various hospitals and regions. At present, the molecular epidemiology of invasive infections caused by R. mucilaginosa and their resistance profiles to antifungals were unknown. Here we collected 49 strains from four hospitals located in different geographic regions from 2009 to 2019 in China, determined their genotypes using different molecular markers and quantified susceptibilities to various antifungals. Sequencing of ITS and D1/D2 regions in rDNA indicated that 73.5% (36/49) of clinical strains belong to same sequence type (rDNA type 2). Microsatellite (MT) genotyping with 15 (recently developed) tandem repeat loci identified 5 epidemic MT types, which accounted for 44.9% (22/49) of clinical strains, as well as 27 sporadic MT types. Microsatellite data indicated that the presence of an epidemic cluster including 35 strains (71.4%) repeatedly isolated in four hospitals for eight years. Single nucleotide variants (SNVs) from the whole genome sequence data also supported the clustering of these epidemic strains due to low pairwise distance. In addition, phylogenetic analysis of SNVs from these clinical strains, together with environmental and animal strains showed that the closely related epidemic cluster strains may be opportunistic, zoonotic pathogens. Also, molecular data indicated a possible clonal transmission of pan echinocandins-azoles-5-flucytosine resistant R. mucilaginosa strains in hospital H01. Our study demonstrated that R. mucilaginosa is a multi-drug resistant pathogen with the ability to cause nosocomial infection.
Collapse
Affiliation(s)
- Jing-Jing Huang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| | - Xin-Fei Chen
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| | - Clement K M Tsui
- Department of Pathology, Sidra Medicine, Education City, Al Rayyan Municipality, Qatar.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar.,Division of Infectious Diseases, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Chong-Jie Pang
- Department of Infection Diseases, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Zhi-Dong Hu
- Department of Clinical Laboratories, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yi Shi
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Wei-Ping Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Lan-Ying Cui
- Department of Laboratory Diagnosis, the first Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yu-Ling Xiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jie Gong
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xin Fan
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ying-Xing Li
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China.,Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, People's Republic of China
| | - Ge Zhang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| | - Meng Xiao
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| | - Ying-Chun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| |
Collapse
|
44
|
Sastré-Velásquez LE, Dallemulle A, Kühbacher A, Baldin C, Alcazar-Fuoli L, Niedrig A, Müller C, Gsaller F. The fungal expel of 5-fluorocytosine derived fluoropyrimidines mitigates its antifungal activity and generates a cytotoxic environment. PLoS Pathog 2022; 18:e1011066. [PMID: 36574449 PMCID: PMC9829169 DOI: 10.1371/journal.ppat.1011066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/09/2023] [Accepted: 12/14/2022] [Indexed: 12/29/2022] Open
Abstract
Invasive aspergillosis remains one of the most devastating fungal diseases and is predominantly linked to infections caused by the opportunistic human mold pathogen Aspergillus fumigatus. Major treatment regimens for the disease comprise the administration of antifungals belonging to the azole, polyene and echinocandin drug class. The prodrug 5-fluorocytosine (5FC), which is the only representative of a fourth class, the nucleobase analogs, shows unsatisfactory in vitro activities and is barely used for the treatment of aspergillosis. The main route of 5FC activation in A. fumigatus comprises its deamination into 5-fluorouracil (5FU) by FcyA, which is followed by Uprt-mediated 5FU phosphoribosylation into 5-fluorouridine monophosphate (5FUMP). In this study, we characterized and examined the role of a metabolic bypass that generates this nucleotide via 5-fluorouridine (5FUR) through uridine phosphorylase and uridine kinase activities. Resistance profiling of mutants lacking distinct pyrimidine salvage activities suggested a minor contribution of the alternative route in 5FUMP formation. We further analyzed the contribution of drug efflux in 5FC tolerance and found that A. fumigatus cells exposed to 5FC reduce intracellular fluoropyrimidine levels through their export into the environment. This release, which was particularly high in mutants lacking Uprt, generates a toxic environment for cytosine deaminase lacking mutants as well as mammalian cells. Employing the broad-spectrum fungal efflux pump inhibitor clorgyline, we demonstrate synergistic properties of this compound in combination with 5FC, 5FU as well as 5FUR.
Collapse
Affiliation(s)
| | - Alex Dallemulle
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Kühbacher
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Clara Baldin
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura Alcazar-Fuoli
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC-CB21/13/00105), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Niedrig
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Christoph Müller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Fabio Gsaller
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
Teixeira MM, Almeida-Paes R, Bernardes-Engemann AR, Nicola AM, de Macedo PM, Valle ACF, Gutierrez-Galhardo MC, Freitas DFS, Barker BM, Matute DR, Stajich JE, Zancopé-Oliveira RM. Single nucleotide polymorphisms and chromosomal copy number variation may impact the Sporothrix brasiliensis antifungal susceptibility and sporotrichosis clinical outcomes. Fungal Genet Biol 2022; 163:103743. [PMID: 36152775 DOI: 10.1016/j.fgb.2022.103743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 02/05/2023]
Abstract
Feline-transmitted sporotrichosis has garnered attention due to the recent high incidence and the lack of efficient control in the epicenter of the epidemic, Rio de Janeiro, Brazil. Sporothrix brasiliensis is the major pathogen involved in feline-to-human sporotrichosis in Brazil and displays more virulent genotypes than the closely related species S. schenckii. Over the last two decades, several reports of antifungal-resistant strains have emerged. Sequencing and comparison analysis of the outbreak strains allowed us to observe that the azole non-wild-type S. brasiliensis strain CFP 1054 had significant chromosomal variations compared to wild-type strains. One of these variants includes a region of 231 Kb containing 75 duplicated genes, which were overrepresented for lipid and isoprenoid metabolism. We also identified an additional strain (CFP 1055) that was resistant to itraconazole and amphotericin B, which had a single nucleotide polymorphism in the tac1 gene. The patients infected with these two strains showed protracted clinical course and sequelae. Even though our sample size is modest, these results suggest the possibility of identifying specific point mutations and large chromosomal duplications potentially associated with antifungal resistance and clinical outcomes of sporotrichosis.
Collapse
Affiliation(s)
- Marcus M Teixeira
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA; Faculty of Medicine, University of Brasília-DF, Brazil
| | - Rodrigo Almeida-Paes
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Andréa R Bernardes-Engemann
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | | | - Priscila M de Macedo
- Laboratório de Pesquisa Clínica em Dermatologia Infecciosa, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Antonio Carlos F Valle
- Laboratório de Pesquisa Clínica em Dermatologia Infecciosa, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Maria Clara Gutierrez-Galhardo
- Laboratório de Pesquisa Clínica em Dermatologia Infecciosa, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Dayvison F S Freitas
- Laboratório de Pesquisa Clínica em Dermatologia Infecciosa, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Bridget M Barker
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Daniel R Matute
- Biology Department, University of North Carolina, Chapel Hill, NC, USA
| | - Jason E Stajich
- Department of Microbiology & Plant Pathology and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
| | - Rosely M Zancopé-Oliveira
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
46
|
Després PC, Cisneros AF, Alexander EMM, Sonigara R, Gagné-Thivierge C, Dubé AK, Landry CR. Asymmetrical dose responses shape the evolutionary trade-off between antifungal resistance and nutrient use. Nat Ecol Evol 2022; 6:1501-1515. [PMID: 36050399 DOI: 10.1038/s41559-022-01846-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/07/2022] [Indexed: 12/22/2022]
Abstract
Antimicrobial resistance is an emerging threat for public health. The success of resistance mutations depends on the trade-off between the benefits and costs they incur. This trade-off is largely unknown and uncharacterized for antifungals. Here, we systematically measure the effect of all amino acid substitutions in the yeast cytosine deaminase Fcy1, the target of the antifungal 5-fluorocytosine (5-FC, flucytosine). We identify over 900 missense mutations granting resistance to 5-FC, a large fraction of which appear to act through destabilization of the protein. The relationship between 5-FC resistance and growth sustained by cytosine deamination is characterized by a sharp trade-off, such that small gains in resistance universally lead to large losses in canonical enzyme function. We show that this steep relationship can be explained by differences in the dose-response functions of 5-FC and cytosine. Finally, we observe the same trade-off shape for the orthologue of FCY1 in Cryptoccocus neoformans, a human pathogen. Our results provide a powerful resource and platform for interpreting drug target variants in fungal pathogens as well as unprecedented insights into resistance-function trade-offs.
Collapse
Affiliation(s)
- Philippe C Després
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada.
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada.
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada.
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada.
| | - Angel F Cisneros
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada
| | - Emilie M M Alexander
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada
| | - Ria Sonigara
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
| | - Cynthia Gagné-Thivierge
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
| | - Alexandre K Dubé
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Canada
| | - Christian R Landry
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Canada.
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada.
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Canada.
- Centre de Recherche sur les Données Massives, Université Laval, Québec, Canada.
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Canada.
| |
Collapse
|
47
|
Tharappel AM, Li Z, Zhu YC, Wu X, Chaturvedi S, Zhang QY, Li H. Calcimycin Inhibits Cryptococcus neoformans In Vitro and In Vivo by Targeting the Prp8 Intein Splicing. ACS Infect Dis 2022; 8:1851-1868. [PMID: 35948057 PMCID: PMC9464717 DOI: 10.1021/acsinfecdis.2c00137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Drug resistance is a significant concern in the treatment of diseases, including cryptococcosis caused by Cryptococcus neoformans (Cne) and Cryptococcus gattii (Cga). Alternative drug targets are necessary to overcome drug resistance before it attains a critical stage. Splicing of inteins from pro-protein precursors is crucial for activities of essential proteins hosting intein elements in many organisms, including human pathogens such as Cne and Cga. Through a high-throughput screening, we identified calcimycin (CMN) as a potent Prp8 intein splicing inhibitor with a minimum inhibitory concentration (MIC) of 1.5 μg/mL against the wild-type Cne-H99 (Cne-WT or Cne). In contrast, CMN inhibited the intein-less mutant strain (Cne-Mut) with a 16-fold higher MIC. Interestingly, Aspergillus fumigatus and a few Candida species were resistant to CMN. Further studies indicated that CMN reduced virulence factors such as urease activity, melanin production, and biofilm formation in Cne. CMN also inhibited Cne intracellular infection in macrophages. In a target-specific split nanoluciferase assay, the IC50 of CMN was 4.6 μg/mL. Binding of CMN to recombinant Prp8 intein was demonstrated by thermal shift assay and microscale thermophoresis. Treating Cne cells with CMN reduced intein splicing. CMN was fungistatic and showed a synergistic effect with the known antifungal drug amphotericin B. Finally, CMN treatment at 20 mg/kg body weight led to 60% reduction in lung fungal load in a cryptococcal pulmonary infection mouse model. Overall, CMN represents a potent antifungal with a novel mechanism of action to treat Cne and possibly Cga infections.
Collapse
Affiliation(s)
- Anil Mathew Tharappel
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Zhong Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Yan Chun Zhu
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Xiangmeng Wu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
| | - Sudha Chaturvedi
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson Arizona 85721-0207, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
- The BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
48
|
Epigenetic Regulation of Antifungal Drug Resistance. J Fungi (Basel) 2022; 8:jof8080875. [PMID: 36012862 PMCID: PMC9409733 DOI: 10.3390/jof8080875] [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: 07/07/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022] Open
Abstract
In medical mycology, epigenetic mechanisms are emerging as key regulators of multiple aspects of fungal biology ranging from development, phenotypic and morphological plasticity to antifungal drug resistance. Emerging resistance to the limited therapeutic options for the treatment of invasive fungal infections is a growing concern. Human fungal pathogens develop drug resistance via multiple mechanisms, with recent studies highlighting the role of epigenetic changes involving the acetylation and methylation of histones, remodeling of chromatin and heterochromatin-based gene silencing, in the acquisition of antifungal resistance. A comprehensive understanding of how pathogens acquire drug resistance will aid the development of new antifungal therapies as well as increase the efficacy of current antifungals by blocking common drug-resistance mechanisms. In this article, we describe the epigenetic mechanisms that affect resistance towards widely used systemic antifungal drugs: azoles, echinocandins and polyenes. Additionally, we review the literature on the possible links between DNA mismatch repair, gene silencing and drug-resistance mechanisms.
Collapse
|
49
|
Priest SJ, Yadav V, Roth C, Dahlmann TA, Kück U, Magwene PM, Heitman J. Uncontrolled transposition following RNAi loss causes hypermutation and antifungal drug resistance in clinical isolates of Cryptococcus neoformans. Nat Microbiol 2022; 7:1239-1251. [PMID: 35918426 PMCID: PMC10840647 DOI: 10.1038/s41564-022-01183-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/23/2022] [Indexed: 02/07/2023]
Abstract
Cryptococcus neoformans infections cause approximately 15% of AIDS-related deaths owing to a combination of limited antifungal therapies and drug resistance. A collection of clinical and environmental C. neoformans isolates were assayed for increased mutation rates via fluctuation analysis, and we identified two hypermutator C. neoformans clinical isolates with increased mutation rates when exposed to the combination of rapamycin and FK506. Sequencing of drug target genes found that Cnl1 transposon insertions conferred the majority of resistance to rapamycin and FK506 and could also independently cause resistance to 5-fluoroorotic acid and the clinically relevant antifungal 5-flucytosine. Whole-genome sequencing revealed both hypermutator genomes harbour a nonsense mutation in the RNA-interference component ZNF3 and hundreds of Cnl1 elements organized into massive subtelomeric arrays on each of the fourteen chromosomes. Quantitative trait locus mapping in 28 progeny derived from a cross between a hypermutator and wild-type identified a locus associated with hypermutation that included znf3. CRISPR editing of the znf3 nonsense mutation abolished hypermutation and restored small-interfering-RNA production. We conclude that hypermutation and drug resistance in these clinical isolates result from RNA-interference loss and accumulation of Cnl1 elements.
Collapse
Affiliation(s)
- Shelby J Priest
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Vikas Yadav
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Cullen Roth
- Department of Biology, Duke University, Durham, NC, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Tim A Dahlmann
- Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Bochum, Germany
| | - Ulrich Kück
- Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Bochum, Germany
| | | | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
50
|
Zhou Z, Zhu C, Ip M, Liu M, Zhu Z, Liu R, Li X, Zeng L, Wu W. Molecular Epidemiology and Antifungal Resistance of Cryptococcus neoformans From Human Immunodeficiency Virus-Negative and Human Immunodeficiency Virus-Positive Patients in Eastern China. Front Microbiol 2022; 13:942940. [PMID: 35865921 PMCID: PMC9294546 DOI: 10.3389/fmicb.2022.942940] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Cryptococcosis is an opportunistic and potentially lethal infection caused by Cryptococcus neoformans and Cryptococcus gattii complex, which affects both immunocompromised and immunocompetent people, and it has become a major public health concern worldwide. In this study, we characterized the molecular epidemiology and antifungal susceptibility of 133 C. neoformans isolates from East China Invasive Fungal Infection Group (ECIFIG), 2017–2020. Isolates were identified to species level by matrix-assisted laser desorption ionization-time of flight mass spectrometry and confirmed by IGS1 sequencing. Whole-genome sequencing (WGS) was conducted on three multidrug-resistant isolates. Among the 133 strains, 61 (45.86%) were isolated from HIV-positive patients and 72 (54.16%) were isolated from HIV-negative patients. In total, C. neoformans var. grubii accounted for 97.74% (130/133), while C. neoformans var. neoformans was rare (2.06%, 3/133). The strains were further classified into nine sequence types (STs) dominated by ST5 (90.23%, 120/133) with low genetic diversity. No association was observed between STs and HIV status. All strains were wild type to voriconazole, while high antifungal minimal inhibitory concentrations (MICs) above the epidemiological cutoff values (ECVs) were observed in C. neoformans strains, and more than half of isolates were non-wild-type to amphotericin B (89.15%, 109/133). Eight isolates were resistant to fluconazole, and eight isolates were non-wild type to 5-fluorocytosine. Furthermore, WGS has verified the novel mutations of FUR1 in 5-fluorocytosine-resistant strains. In one isolate, aneuploidy of chromosome 1 with G484S mutation of ERG11 was observed, inducing high-level resistance (MIC: 32 μg/ml) to fluconazole. In general, our data showed that there was no significant difference between HIV-positive and HIV-negative patients on STs, and we elucidate the resistant mechanisms of C. neoformans from different perspectives. It is important for clinical therapy and drug usage in the future.
Collapse
Affiliation(s)
- Ziyi Zhou
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chendi Zhu
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Margaret Ip
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Manjiao Liu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Zhaoqin Zhu
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Ryon Liu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Xiaomin Li
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Lingbing Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Lingbing Zeng,
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Wenjuan Wu,
| |
Collapse
|