1
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.09.602714. [PMID: 40161774 PMCID: PMC11952329 DOI: 10.1101/2024.07.09.602714] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
2
|
Tosiano MA, Lanni F, Mitchell AP, McManus CJ. Roles of P-body factors in Candida albicans filamentation and stress response. PLoS Genet 2025; 21:e1011632. [PMID: 40096135 PMCID: PMC11975087 DOI: 10.1371/journal.pgen.1011632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 04/07/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
Hyphal growth is strongly associated with virulence in the human fungal pathogen Candida albicans. While hyphal transcriptional networks have been the subject of intense study, relatively little is known about post-transcriptional regulation. Previous work reported that P-Body (PB) factors Dhh1 and Edc3 were required for C. albicans virulence and filamentation, suggesting an essential role for post-transcriptional regulation of these processes. However, the molecular roles of these factors have not been determined. To further study the function of PB factors in filamentation, we generated homozygous deletions of DHH1 and EDC3 in diverse prototrophic clinical strains using transient CRISPR-Cas9. Homozygous DHH1 deletion strongly impaired growth, altered filamentation, and exhibited unusual colony morphology in response to heat stress in five strain backgrounds. Using RNA-seq, we found DHH1 deletion disrupts the regulation of thousands of genes under both yeast and hyphal growth conditions in SC5314 and P57055. This included upregulation of many stress response genes in the absence of external stress, similar to deletion of the S. cerevisiae DHH1 homolog. In contrast, we found EDC3 was not required for heat tolerance or filamentation in diverse strains. These results support a model in which DHH1, but not EDC3, represses hyphal stress response transcripts in yeast and remodels the transcriptome during filamentation. Our work supports distinct requirements for specific mRNA decay factors, bolstering evidence for post-transcriptional regulation of filamentation in C. albicans.
Collapse
Affiliation(s)
- Melissa A. Tosiano
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Frederick Lanni
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Aaron P. Mitchell
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
3
|
Nayak A, Khedri A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:23. [PMID: 39268078 PMCID: PMC11391927 DOI: 10.1038/s44259-024-00040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/19/2024] [Indexed: 09/15/2024]
Abstract
Candida albicans, an opportunistic fungal pathogen, causes life-threatening infections in immunocompromised patients. Current antifungals are limited by toxicity, drug-drug interactions, and emerging resistance, underscoring the importance of identifying novel treatment approaches. Here, we elucidate the impact of sinefungin, an analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 and clinical isolates. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal morphogenesis, biofilm formation, adhesion to epithelial cells, and virulence towards Galleria mellonella, highlighting sinefungin as an avenue for therapeutic intervention. We determine that sinefungin particularly disturbs N6-methyladenosine (m6A) formation. Transcriptome analysis of C. albicans hyphae upon sinefungin treatment reveals an increase in transcripts related to the yeast form and decrease in those associated with hyphae formation and virulence. Collectively, our data propose sinefungin as a potent molecule against C. albicans and emphasize further exploration of post-transcriptional control mechanisms of pathogenicity for antifungal design.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Azam Khedri
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Firdous Z, Kalra S, Chattopadhyay R, Bari VK. Current insight into the role of mRNA decay pathways in fungal pathogenesis. Microbiol Res 2024; 283:127671. [PMID: 38479232 DOI: 10.1016/j.micres.2024.127671] [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: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
Pathogenic fungal species can cause superficial and mucosal infections, to potentially fatal systemic or invasive infections in humans. These infections are more common in immunocompromised or critically ill patients and have a significant morbidity and fatality rate. Fungal pathogens utilize several strategies to adapt the host environment resulting in efficient and comprehensive alterations in their cellular metabolism. Fungal virulence is regulated by several factors and post-transcriptional regulation mechanisms involving mRNA molecules are one of them. Post-transcriptional controls have emerged as critical regulatory mechanisms involved in the pathogenesis of fungal species. The untranslated upstream and downstream regions of the mRNA, as well as RNA-binding proteins, regulate morphogenesis and virulence by controlling mRNA degradation and stability. The limited number of available therapeutic drugs, the emergence of multidrug resistance, and high death rates associated with systemic fungal illnesses pose a serious risk to human health. Therefore, new antifungal treatments that specifically target mRNA pathway components can decrease fungal pathogenicity and when combined increase the effectiveness of currently available antifungal drugs. This review summarizes the mRNA degradation pathways and their role in fungal pathogenesis.
Collapse
Affiliation(s)
- Zulikha Firdous
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda 151401, India
| | - Sapna Kalra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda 151401, India
| | - Rituja Chattopadhyay
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda 151401, India
| | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda 151401, India.
| |
Collapse
|
5
|
Woodruff AL, Berman J, Anderson M. Strain background of Candida albicans interacts with SIR2 to alter phenotypic switching. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001444. [PMID: 38446018 PMCID: PMC10999749 DOI: 10.1099/mic.0.001444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
The genetic background between strains of a single species and within a single strain lineage can significantly impact the expression of biological traits. This genetic variation may also reshape epigenetic mechanisms of cell identity and environmental responses that are controlled by interconnected transcriptional networks and chromatin-modifying enzymes. Histone deacetylases, including sirtuins, are critical regulators of chromatin state and have been directly implicated in governing the phenotypic transition between the 'sterile' white state and the mating-competent opaque state in Candida albicans, a common fungal commensal and pathogen of humans. Here, we found that a previously ambiguous role for the sirtuin SIR2 in C. albicans phenotypic switching is likely linked to the genetic background of mutant strains produced in the RM lineage of SC5314. SIR2 mutants in a specific lineage of BWP17 displayed increased frequencies of switching to the opaque state compared to the wild-type. Loss of SIR2 in other SC5314-derived backgrounds, including newly constructed BWP17 sir2Δ/Δ mutants, failed to recapitulate the increased white-opaque switching frequencies observed in the original BWP17 sir2Δ/Δ mutant background. Whole-genome sequencing revealed the presence of multiple imbalanced chromosomes and large loss of heterozygosity tracts that likely interact with SIR2 to increase phenotypic switching in this BWP17 sir2Δ/Δ mutant lineage. These genomic changes are not found in other SC5314-derived sir2Δ/Δ mutants that do not display increased opaque cell formation. Thus, complex karyotypes can emerge during strain construction that modify mutant phenotypes and highlight the importance of validating strain background when interpreting phenotypes.
Collapse
Affiliation(s)
- Andrew L. Woodruff
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, The George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Matthew Anderson
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
- Department of Medical Genetics, Laboratory of Genetics, University of Wisconsin – Madison, Madison, WI, 53706, USA
- Center for Genomic Science Innovation, University of Wisconsin – Madison, Madison, WI, 53706, USA
| |
Collapse
|
6
|
Nayak A, Chavarria A, Sanders KN, Ghalei H, Khoshnevis S. Sinefungin, a natural nucleoside analog of S-adenosyl methionine, impairs the pathogenicity of Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.562127. [PMID: 37873365 PMCID: PMC10592816 DOI: 10.1101/2023.10.12.562127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Candida albicans, an opportunistic fungal human pathogen, is a major threat to the healthcare system due to both infections in immunocompromised individuals and the emergence of antifungal resistance. Fungal infection caused by C. albicans, candidiasis, is a life-threatening condition in immunocompromised patients and the current treatments are mostly restricted to polyenes, azoles, and echinocandins. Use of these antifungals is limited by toxicity, drug-drug interactions, and the emergence of resistance, underscoring the importance of identifying novel therapeutic targets and the need for new treatment approaches. C. albicans can undergo a morphological transition from yeast to hyphae and this transition is central to C. albicans virulence. Here, we determine the impact of sinefungin, a natural nucleoside analog of S-adenosyl methionine, on the virulence of C. albicans strain SC5314 by evaluating treatment effects on the morphological transition, human epithelial cell adhesion, and biofilm formation. Our data indicate that sinefungin impairs pathogenic traits of C. albicans including hyphal lengthening, biofilm formation and the adhesion to the human epithelial cell lines, without adversely affecting human cells, therefore highlighting sinefungin as a potential avenue for therapeutic intervention. We determine that the formation of N6-methyladenosine (m6A) is particularly disturbed by sinefungin. More broadly, this study underscores the importance of considering the post-transcriptional control mechanisms of pathogenicity when designing therapeutic solutions to fungal infection.
Collapse
Affiliation(s)
- Anushka Nayak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Alejandro Chavarria
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Kyla N. Sanders
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Sohail Khoshnevis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
7
|
Oliveira M, Cunha E, Tavares L, Serrano I. P. aeruginosa interactions with other microbes in biofilms during co-infection. AIMS Microbiol 2023; 9:612-646. [PMID: 38173971 PMCID: PMC10758579 DOI: 10.3934/microbiol.2023032] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/10/2023] [Accepted: 07/26/2023] [Indexed: 01/05/2024] Open
Abstract
This review addresses the topic of biofilms, including their development and the interaction between different counterparts. There is evidence that various diseases, such as cystic fibrosis, otitis media, diabetic foot wound infections, and certain cancers, are promoted and aggravated by the presence of polymicrobial biofilms. Biofilms are composed by heterogeneous communities of microorganisms protected by a matrix of polysaccharides. The different types of interactions between microorganisms gives rise to an increased resistance to antimicrobials and to the host's defense mechanisms, with the consequent worsening of disease symptoms. Therefore, infections caused by polymicrobial biofilms affecting different human organs and systems will be discussed, as well as the role of the interactions between the gram-negative bacteria Pseudomonas aeruginosa, which is at the base of major polymicrobial infections, and other bacteria, fungi, and viruses in the establishment of human infections and diseases. Considering that polymicrobial biofilms are key to bacterial pathogenicity, it is fundamental to evaluate which microbes are involved in a certain disease to convey an appropriate and efficacious antimicrobial therapy.
Collapse
Affiliation(s)
- Manuela Oliveira
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Eva Cunha
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís Tavares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Isa Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| |
Collapse
|
8
|
Tarrés-Solé A, Battistini F, Gerhold JM, Piétrement O, Martínez-García B, Ruiz-López E, Lyonnais S, Bernadó P, Roca J, Orozco M, Le Cam E, Sedman J, Solà M. Structural analysis of the Candida albicans mitochondrial DNA maintenance factor Gcf1p reveals a dynamic DNA-bridging mechanism. Nucleic Acids Res 2023; 51:5864-5882. [PMID: 37207342 PMCID: PMC10287934 DOI: 10.1093/nar/gkad397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 04/01/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023] Open
Abstract
The compaction of mitochondrial DNA (mtDNA) is regulated by architectural HMG-box proteins whose limited cross-species similarity suggests diverse underlying mechanisms. Viability of Candida albicans, a human antibiotic-resistant mucosal pathogen, is compromised by altering mtDNA regulators. Among them, there is the mtDNA maintenance factor Gcf1p, which differs in sequence and structure from its human and Saccharomyces cerevisiae counterparts, TFAM and Abf2p. Our crystallographic, biophysical, biochemical and computational analysis showed that Gcf1p forms dynamic protein/DNA multimers by a combined action of an N-terminal unstructured tail and a long helix. Furthermore, an HMG-box domain canonically binds the minor groove and dramatically bends the DNA while, unprecedentedly, a second HMG-box binds the major groove without imposing distortions. This architectural protein thus uses its multiple domains to bridge co-aligned DNA segments without altering the DNA topology, revealing a new mechanism of mtDNA condensation.
Collapse
Affiliation(s)
- Aleix Tarrés-Solé
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Federica Battistini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Department of Biochemistry, University of Barcelona, Barcelona 08028, Spain
| | - Joachim M Gerhold
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Olivier Piétrement
- Genome Integrity and Cancer UMR 9019 CNRS, Université Paris Saclay, Gustave Roussy Campus, 114 rue Edouard Vaillant 94805VillejuifCedex, France
| | | | - Elena Ruiz-López
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Sébastien Lyonnais
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | - Pau Bernadó
- Centre de Biologie Structurale (CBS), Inserm, CNRS and Université de Montpellier, France, Sébastien Lyonnais, UAR 3725 CNRS, Université de Montpellier, 34000 Montpellier, France
| | - Joaquim Roca
- Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Department of Biochemistry, University of Barcelona, Barcelona 08028, Spain
| | - Eric Le Cam
- Genome Integrity and Cancer UMR 9019 CNRS, Université Paris Saclay, Gustave Roussy Campus, 114 rue Edouard Vaillant 94805VillejuifCedex, France
| | - Juhan Sedman
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Maria Solà
- Structural MitoLab, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona 08028, Spain
| |
Collapse
|
9
|
Biofilm Formation of Probiotic Saccharomyces cerevisiae var. boulardii on Glass Surface during Beer Bottle Ageing. BEVERAGES 2022. [DOI: 10.3390/beverages8040077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
While brewing probiotic beer using Saccharomyces cerevisiae var. boulardii, we noticed the yeast potentially makes biofilm in glass bottles as the bottles get hazy. In this study, S. cerevisiae var. boulardii CNCM I-745 was used as a starter culture to produce probiotic beer. We studied the biofilm parameters combined with FLO11 mRNA expression and used light and scanning electron microscopy to document biofilm formation and structure. Our results revealed that ageing the beer and maturing from a sugar-rich to a sugar-limited beer, along with the stress factors from the brewing process (pH reduction and produced metabolites), led to an increase in biofilm mass; however, the viable count remained relatively stable (approximately 7.1 log10 cells/mL). Biofilm S. boulardii cells showed significantly higher FLO11 mRNA expression in the exponential and stationary phase compared to the planktonic cells. This study, therefore, provides evidence that S. cerevisiae var. boulardii makes biofilm on glass surfaces during beer bottle ageing. The impact of complications caused by formed biofilms on returnable bottles emphasizes the significance of this study.
Collapse
|
10
|
Application of the Mutant Libraries for Candida albicans Functional Genomics. Int J Mol Sci 2022; 23:ijms232012307. [PMID: 36293157 PMCID: PMC9603287 DOI: 10.3390/ijms232012307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Candida albicans is a typical opportunistic pathogen in humans that causes serious health risks in clinical fungal infections. The construction of mutant libraries has made remarkable developments in the study of C. albicans molecular and cellular biology with the ongoing advancements of gene editing, which include the application of CRISPR-Cas9 and novel high-efficient transposon. Large-scale genetic screens and genome-wide functional analysis accelerated the investigation of new genetic regulatory mechanisms associated with the pathogenicity and resistance to environmental stress in C. albicans. More importantly, sensitivity screening based on C. albicans mutant libraries is critical for the target identification of novel antifungal compounds, which leads to the discovery of Sec7p, Tfp1p, Gwt1p, Gln4p, and Erg11p. This review summarizes the main types of C. albicans mutant libraries and interprets their applications in morphogenesis, biofilm formation, fungus-host interactions, antifungal drug resistance, and target identification.
Collapse
|
11
|
Abdulghani M, Iram R, Chidrawar P, Bhosle K, Kazi R, Patil R, Kharat K, Zore G. Proteomic profile of Candida albicans biofilm. J Proteomics 2022; 265:104661. [PMID: 35728770 DOI: 10.1016/j.jprot.2022.104661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/06/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
Candida albicans biofilms are characterized by structural and cellular heterogeneity that confers antifungal resistance and immune evasion. Despite this, biofilm formation remains poorly understood. In this study, we used proteomic analysis to understand biofilm formation in C. albicans related to morphophysiological and architectural features. LC-MS/MS analysis revealed that 64 proteins were significantly modulated, of which 31 were upregulated and 33 were downregulated. The results indicate that metabolism (25 proteins), gene expression (13 proteins), stress response (7 proteins), and cell wall (5 proteins) composition are modulated. The rate of oxidative phosphorylation (OxPhos) and biosynthesis of UDP-N-acetylglucosamine, vitamin B6, and thiamine increased, while the rate of methionine biosynthesis decreased. There was a significant modification of the cell wall architecture due to higher levels of Sun41, Pir1 and Csh1 and increased glycosylation of proteins. It was observed that C. albicans induces hyphal growth by upregulating the expression of genes involved in cAMP-PKA and MAPK pathways. This study is significant in that it suggests an increase in OxPhos and alteration of cell wall architecture that could be contributing to the recalcitrance of C. albicans cells growing in biofilms. Nevertheless, a deeper investigation is needed to explore it further. SIGNIFICANCE: Candida sps is included in the list of pathogens with potential drug resistance threat due to the increased frequency especially colonization of medical devices, and tissues among the patients, in recent years. Significance of our study is that we are reporting traits like modulation in cell wall composition, amino acid and vitamin biosynthesis and importantly energy generation (OxPhos) etc. These traits could be conferring antifungal resistance, host immune evasion etc. and thus survival, in addition to facilitating biofilm formation. These findings are expected to prime the further studies on devising potent strategy against biofilm growth among the patients.
Collapse
Affiliation(s)
- Mazen Abdulghani
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Rasiqua Iram
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Priti Chidrawar
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Kajal Bhosle
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India
| | - Rubina Kazi
- Division of Biochemical Sciences, CSIR-NCL, Pune 8, MS, India
| | - Rajendra Patil
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune 411007, MS, India
| | - Kiran Kharat
- Department of Biotechnology, Deogiri College, Aurangabad, MS, India
| | - Gajanan Zore
- School of Life Sciences, Swami Ramanand Teerth Marathwada University, Nanded 431606, MS, India.
| |
Collapse
|
12
|
D’Auria FD, Casciaro B, De Angelis M, Marcocci ME, Palamara AT, Nencioni L, Mangoni ML. Antifungal Activity of the Frog Skin Peptide Temporin G and Its Effect on Candida albicans Virulence Factors. Int J Mol Sci 2022; 23:ijms23116345. [PMID: 35683025 PMCID: PMC9181532 DOI: 10.3390/ijms23116345] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/26/2023] Open
Abstract
The increasing resistance to conventional antifungal drugs is a widespread concern, and a search for new compounds, active against different species of fungi, is demanded. Antimicrobial peptides (AMPs) hold promises in this context. Here we investigated the activity of the frog skin AMP Temporin G (TG) against a panel of fungal strains, by following the Clinical and Laboratory Standards Institute protocols. TG resulted to be active against (i) Candida species and Cryptococcus neoformans, with MIC50 between 4 µM and 64 µM after 24 h of incubation; (ii) dermatophytes with MIC80 ranging from 4 to 32 µM, and (iii) Aspergillus strains with MIC80 of 128 µM. In addition, our tests revealed that TG reduced the metabolic activity of Candida albicans cells, with moderate membrane perturbation, as proven by XTT and Sytox Green assays, respectively. Furthermore, TG was found to be effective against some C. albicans virulence factors; indeed, at 64 µM it was able to inhibit ~90% of yeast-mycelial switching, strongly prevented biofilm formation, and led to a 50% reduction of metabolic activity in mature biofilm cells, and ~30-35% eradication of mature biofilm biomass. Even though further studies are needed to deepen our knowledge of the mechanisms of TG antifungal activity, our results suggest this AMP as an attractive lead compound for treatment of fungal diseases.
Collapse
Affiliation(s)
- Felicia Diodata D’Auria
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Bruno Casciaro
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy;
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy; (F.D.D.); (M.D.A.); (M.E.M.); (A.T.P.)
- Correspondence: (L.N.); (M.L.M.); Tel.: +39-0649914608 (L.N.); +39-0649910838 (M.L.M.)
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (L.N.); (M.L.M.); Tel.: +39-0649914608 (L.N.); +39-0649910838 (M.L.M.)
| |
Collapse
|
13
|
Rana A, Gupta N, Thakur A. Post-transcriptional and translational control of the morphology and virulence in human fungal pathogens. Mol Aspects Med 2021; 81:101017. [PMID: 34497025 DOI: 10.1016/j.mam.2021.101017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022]
Abstract
Host-pathogen interactions at the molecular level are the key to fungal pathogenesis. Fungal pathogens utilize several mechanisms such as adhesion, invasion, phenotype switching and metabolic adaptations, to survive in the host environment and respond. Post-transcriptional and translational regulations have emerged as key regulatory mechanisms ensuring the virulence and survival of fungal pathogens. Through these regulations, fungal pathogens effectively alter their protein pool, respond to various stress, and undergo morphogenesis, leading to efficient and comprehensive changes in fungal physiology. The regulation of virulence through post-transcriptional and translational regulatory mechanisms is mediated through mRNA elements (cis factors) or effector molecules (trans factors). The untranslated regions upstream and downstream of the mRNA, as well as various RNA-binding proteins involved in translation initiation or circularization of the mRNA, play pivotal roles in the regulation of morphology and virulence by influencing protein synthesis, protein isoforms, and mRNA stability. Therefore, post-transcriptional and translational mechanisms regulating the morphology, virulence and drug-resistance processes in fungal pathogens can be the target for new therapeutics. With improved "omics" technologies, these regulatory mechanisms are increasingly coming to the forefront of basic biology and drug discovery. This review aims to discuss various modes of post-transcriptional and translation regulations, and how these mechanisms exert influence in the virulence and morphogenesis of fungal pathogens.
Collapse
Affiliation(s)
- Aishwarya Rana
- Regional Centre for Biotechnology, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad 121001, India
| | - Nidhi Gupta
- Regional Centre for Biotechnology, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad 121001, India
| | - Anil Thakur
- Regional Centre for Biotechnology, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad 121001, India.
| |
Collapse
|
14
|
Łabędzka-Dmoch K, Kolondra A, Karpińska MA, Dębek S, Grochowska J, Grochowski M, Piątkowski J, Hoang Diu Bui T, Golik P. Pervasive transcription of the mitochondrial genome in Candida albicans is revealed in mutants lacking the mtEXO RNase complex. RNA Biol 2021; 18:303-317. [PMID: 34229573 PMCID: PMC8677008 DOI: 10.1080/15476286.2021.1943929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The mitochondrial genome of the pathogenic yeast Candida albicans displays a typical organization of several (eight) primary transcription units separated by noncoding regions. Presence of genes encoding Complex I subunits and the stability of its mtDNA sequence make it an attractive model to study organellar genome expression using transcriptomic approaches. The main activity responsible for RNA degradation in mitochondria is a two-component complex (mtEXO) consisting of a 3ʹ-5ʹ exoribonuclease, in yeasts encoded by the DSS1 gene, and a conserved Suv3p helicase. In C. albicans, deletion of either DSS1 or SUV3 gene results in multiple defects in mitochondrial genome expression leading to the loss of respiratory competence. Transcriptomic analysis reveals pervasive transcription in mutants lacking the mtEXO activity, with evidence of the entire genome being transcribed, whereas in wild-type strains no RNAs corresponding to a significant fraction of the noncoding genome can be detected. Antisense (‘mirror’) transcripts, absent from normal mitochondria are also prominent in the mutants. The expression of multiple mature transcripts, particularly those translated from bicistronic mRNAs, as well as those that contain introns is affected in the mutants, resulting in a decreased level of proteins and reduced respiratory complex activity. The phenotype is most severe in the case of Complex IV, where a decrease of mature COX1 mRNA level to ~5% results in a complete loss of activity. These results show that RNA degradation by mtEXO is essential for shaping the mitochondrial transcriptome and is required to maintain the functional demarcation between transcription units and non-coding genome segments.
Collapse
Affiliation(s)
- Karolina Łabędzka-Dmoch
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Adam Kolondra
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Magdalena A Karpińska
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Sonia Dębek
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Joanna Grochowska
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Grochowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Jakub Piątkowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Thi Hoang Diu Bui
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Paweł Golik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
15
|
Computational Drug Repurposing Resources and Approaches for Discovering Novel Antifungal Drugs against Candida albicans N-Myristoyl Transferase. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is a yeast that is an opportunistic fungal pathogen and also identified as ubiquitous polymorphic species that is mainly linked with major fungal infections in humans, particularly in the immunocompromised patients including transplant recipients, chemotherapy patients, HIV-infected patients as well as in low-birth-weight infants. Systemic Candida infections have a high mortality rate of around 29 to 76%. For reducing its infection, limited drugs are existing such as caspofungin, fluconazole, terbinafine, and amphotericin B, etc. which contain unlikable side effects and also toxic. This review intends to utilize advanced bioinformatics technologies such as Molecular docking, Scaffold hopping, Virtual screening, Pharmacophore modeling, Molecular dynamics (MD) simulation for the development of potentially new drug candidates with a drug-repurpose approach against Candida albicans within a limited time frame and also cost reductive.
Collapse
|
16
|
van Gestel J, Wagner A. Cryptic surface-associated multicellularity emerges through cell adhesion and its regulation. PLoS Biol 2021; 19:e3001250. [PMID: 33983920 PMCID: PMC8148357 DOI: 10.1371/journal.pbio.3001250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/25/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
The repeated evolution of multicellularity led to a wide diversity of organisms, many of which are sessile, including land plants, many fungi, and colonial animals. Sessile organisms adhere to a surface for most of their lives, where they grow and compete for space. Despite the prevalence of surface-associated multicellularity, little is known about its evolutionary origin. Here, we introduce a novel theoretical approach, based on spatial lineage tracking of cells, to study this origin. We show that multicellularity can rapidly evolve from two widespread cellular properties: cell adhesion and the regulatory control of adhesion. By evolving adhesion, cells attach to a surface, where they spontaneously give rise to primitive cell collectives that differ in size, life span, and mode of propagation. Selection in favor of large collectives increases the fraction of adhesive cells until a surface becomes fully occupied. Through kin recognition, collectives then evolve a central-peripheral polarity in cell adhesion that supports a division of labor between cells and profoundly impacts growth. Despite this spatial organization, nascent collectives remain cryptic, lack well-defined boundaries, and would require experimental lineage tracking technologies for their identification. Our results suggest that cryptic multicellularity could readily evolve and originate well before multicellular individuals become morphologically evident.
Collapse
Affiliation(s)
- Jordi van Gestel
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Andreas Wagner
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- The Santa Fe Institute, Santa Fe, New Mexico, United States of America
| |
Collapse
|
17
|
Wang JM, Woodruff AL, Dunn MJ, Fillinger RJ, Bennett RJ, Anderson MZ. Intraspecies Transcriptional Profiling Reveals Key Regulators of Candida albicans Pathogenic Traits. mBio 2021; 12:e00586-21. [PMID: 33879584 PMCID: PMC8092256 DOI: 10.1128/mbio.00586-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/17/2021] [Indexed: 12/27/2022] Open
Abstract
The human commensal and opportunistic fungal pathogen Candida albicans displays extensive genetic and phenotypic variation across clinical isolates. Here, we performed RNA sequencing on 21 well-characterized isolates to examine how genetic variation contributes to gene expression differences and to link these differences to phenotypic traits. C. albicans adapts primarily through clonal evolution, and yet hierarchical clustering of gene expression profiles in this set of isolates did not reproduce their phylogenetic relationship. Strikingly, strain-specific gene expression was prevalent in some strain backgrounds. Association of gene expression with phenotypic data by differential analysis, linear correlation, and assembly of gene networks connected both previously characterized and novel genes with 23 C. albicans traits. Construction of de novo gene modules produced a gene atlas incorporating 67% of C. albicans genes and revealed correlations between expression modules and important phenotypes such as systemic virulence. Furthermore, targeted investigation of two modules that have novel roles in growth and filamentation supported our bioinformatic predictions. Together, these studies reveal widespread transcriptional variation across C. albicans isolates and identify genetic and epigenetic links to phenotypic variation based on coexpression network analysis.IMPORTANCE Infectious fungal species are often treated uniformly despite clear evidence of genotypic and phenotypic heterogeneity being widespread across strains. Identifying the genetic basis for this phenotypic diversity is extremely challenging because of the tens or hundreds of thousands of variants that may distinguish two strains. Here, we use transcriptional profiling to determine differences in gene expression that can be linked to phenotypic variation among a set of 21 Candida albicans isolates. Analysis of this transcriptional data set uncovered clear trends in gene expression characteristics for this species and new genes and pathways that were associated with variation in pathogenic processes. Direct investigation confirmed functional predictions for a number of new regulators associated with growth and filamentation, demonstrating the utility of these approaches in linking genes to important phenotypes.
Collapse
Affiliation(s)
- Joshua M Wang
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Andrew L Woodruff
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Matthew J Dunn
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Robert J Fillinger
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Richard J Bennett
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Matthew Z Anderson
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
18
|
Mancera E, Nocedal I, Hammel S, Gulati M, Mitchell KF, Andes DR, Nobile CJ, Butler G, Johnson AD. Evolution of the complex transcription network controlling biofilm formation in Candida species. eLife 2021; 10:e64682. [PMID: 33825680 PMCID: PMC8075579 DOI: 10.7554/elife.64682] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/06/2021] [Indexed: 12/30/2022] Open
Abstract
We examine how a complex transcription network composed of seven 'master' regulators and hundreds of target genes evolved over a span of approximately 70 million years. The network controls biofilm formation in several Candida species, a group of fungi that are present in humans both as constituents of the microbiota and as opportunistic pathogens. Using a variety of approaches, we observed two major types of changes that have occurred in the biofilm network since the four extant species we examined last shared a common ancestor. Master regulator 'substitutions' occurred over relatively long evolutionary times, resulting in different species having overlapping but different sets of master regulators of biofilm formation. Second, massive changes in the connections between the master regulators and their target genes occurred over much shorter timescales. We believe this analysis is the first detailed, empirical description of how a complex transcription network has evolved.
Collapse
Affiliation(s)
- Eugenio Mancera
- Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad IrapuatoIrapuatoMexico
| | - Isabel Nocedal
- Department of Microbiology and Immunology, University of California, San FranciscoSan FranciscoUnited States
| | - Stephen Hammel
- School of Biomolecular and Biomedical Science, Conway Institute, University College DublinDublinIreland
| | - Megha Gulati
- Department of Molecular and Cell Biology, University of California, MercedMercedUnited States
| | - Kaitlin F Mitchell
- Department of Medical Microbiology and Immunology, University of WisconsinMadisonUnited States
| | - David R Andes
- Department of Medical Microbiology and Immunology, University of WisconsinMadisonUnited States
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, University of California, MercedMercedUnited States
| | - Geraldine Butler
- School of Biomolecular and Biomedical Science, Conway Institute, University College DublinDublinIreland
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San FranciscoSan FranciscoUnited States
- Microbiome Initiative, Chan Zuckerberg BiohubSan FranciscoUnited States
| |
Collapse
|
19
|
Cell Surface Expression of Nrg1 Protein in Candida auris. J Fungi (Basel) 2021; 7:jof7040262. [PMID: 33807166 PMCID: PMC8066560 DOI: 10.3390/jof7040262] [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: 02/17/2021] [Revised: 03/21/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
Candida auris is an emerging antifungal resistant human fungal pathogen increasingly reported in healthcare facilities. It persists in hospital environments, and on skin surfaces, and can form biofilms readily. Here, we investigated the cell surface proteins from C. auris biofilms grown in a synthetic sweat medium mimicking human skin conditions. Cell surface proteins from both biofilm and planktonic control cells were extracted with a buffer containing β-mercaptoethanol and resolved by 2-D gel electrophoresis. Some of the differentially expressed proteins were excised and identified by mass spectrometry. C. albicans orthologs Spe3p, Tdh3p, Sod2p, Ywp1p, and Mdh1p were overexpressed in biofilm cells when compared to the planktonic cells of C. auris. Interestingly, several proteins with zinc ion binding activity were detected. Nrg1p is a zinc-binding transcription factor that negatively regulates hyphal growth in C. albicans. C. auris does not produce true hypha under standard in vitro growth conditions, and the role of Nrg1p in C. auris is currently unknown. Western blot analyses of cell surface and cytosolic proteins of C. auris against anti-CalNrg1 antibody revealed the Nrg1p in both locations. Cell surface localization of Nrg1p in C. auris, an unexpected finding, was further confirmed by immunofluorescence microscopy. Nrg1p expression is uniform across all four clades of C. auris and is dependent on growth conditions. Taken together, the data indicate that C. auris produces several unique proteins during its biofilm growth, which may assist in the skin-colonizing lifestyle of the fungus during its pathogenesis.
Collapse
|
20
|
Ponde NO, Lortal L, Ramage G, Naglik JR, Richardson JP. Candida albicans biofilms and polymicrobial interactions. Crit Rev Microbiol 2021; 47:91-111. [PMID: 33482069 PMCID: PMC7903066 DOI: 10.1080/1040841x.2020.1843400] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/05/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a common fungus of the human microbiota. While generally a harmless commensal in healthy individuals, several factors can lead to its overgrowth and cause a range of complications within the host, from localized superficial infections to systemic life-threatening disseminated candidiasis. A major virulence factor of C. albicans is its ability to form biofilms, a closely packed community of cells that can grow on both abiotic and biotic substrates, including implanted medical devices and mucosal surfaces. These biofilms are extremely hard to eradicate, are resistant to conventional antifungal treatment and are associated with high morbidity and mortality rates, making biofilm-associated infections a major clinical challenge. Here, we review the current knowledge of the processes involved in C. albicans biofilm formation and development, including the central processes of adhesion, extracellular matrix production and the transcriptional network that regulates biofilm development. We also consider the advantages of the biofilm lifestyle and explore polymicrobial interactions within multispecies biofilms that are formed by C. albicans and selected microbial species.
Collapse
Affiliation(s)
- Nicole O. Ponde
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Gordon Ramage
- School of Medicine, Dentistry & Nursing, Glasgow Dental School and Hospital, Faculty of Medicine, University of Glasgow, G2 3JZ, United Kingdom
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
21
|
Ke CL, Liao YT, Lin CH. MSS2 maintains mitochondrial function and is required for chitosan resistance, invasive growth, biofilm formation and virulence in Candida albicans. Virulence 2021; 12:281-297. [PMID: 33427576 PMCID: PMC7808435 DOI: 10.1080/21505594.2020.1870082] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is the most prevalent fungal pathogen in humans, particularly in immunocompromised patients. In this study, by screening a C. albicans mutant library, we first identified that the MSS2 gene, an ortholog of Saccharomyces cerevisiae MSS2 required for mitochondrial respiration, mediates chitosan resistance. Upon treatment with 0.2% chitosan, the growth of mss2Δ strains was strikingly impaired, and MSS2 expression was significantly repressed by chitosan. Furthermore, mss2Δ strains exhibited slow growth on medium supplemented with glycerol as the sole carbon source. Similar to the chitosan-treated wild-type strain, the mss2Δ strain exhibited a significantly impaired ATP production ability. These data suggest that an antifungal mechanism of chitosan against C. albicans acts by inhibiting MSS2 gene expression, leading to repression of mitochondrial function. Normal respiratory function is suggested to be required for fungal virulence. Interestingly, the mss2Δ mutant strains exhibited significantly impaired invasive ability in vitro and ex vivo but retained normal hyphal development ability in liquid medium. Furthermore, the MSS2 deletion strains could not form robust biofilms and exhibited significantly reduced virulence. Collectively, these results demonstrated that the antifungal effect of chitosan against C. albicans is mediated via inhibition of mitochondrial biogenesis. These data may provide another strategy for antifungal drug development via inhibition of fungal mitochondria.
Collapse
Affiliation(s)
- Cai-Ling Ke
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Taipei, Taiwan
| | - Yu-Ting Liao
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Taipei, Taiwan
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Taipei, Taiwan
| |
Collapse
|
22
|
Agarwalla SV, Ellepola K, Silikas N, Castro Neto AH, Seneviratne CJ, Rosa V. Persistent inhibition of Candida albicans biofilm and hyphae growth on titanium by graphene nanocoating. Dent Mater 2020; 37:370-377. [PMID: 33358443 DOI: 10.1016/j.dental.2020.11.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Candida albicanscolonizes biomaterial surfaces and are highly resistant to therapeutics. Graphene nanocoating on titanium compromises initial biofilm formation. However, its sustained antibiofilm potential is unknown. The objective of this study was to investigate the potential of graphene nanocoating to decrease long-term fungal biofilm development and hyphae growth on titanium. METHODS Graphene nanocoating was deposited twice (TiGD) or five times (TiGV) on grade 4 titanium with vacuum assisted technique and characterized with Raman spectroscopy and atomic force microscope. The biofilm formation and hyphae growth of C. albicans was monitored for seven days by CFU, XTT, confocal, mean cell density and scanning electronic microscopy (SEM). Uncoated titanium was the Control. All tests had three independent biological samples and were performed in independent triplicates. Data was analyzed with one- or two-way ANOVA and Tukey's HSD (α = 0.05). RESULTS Both TiGD and TiGV presented less biofilms at all times points compared with Control. The confocal and SEM images revealed few adhered cells on graphene coated samples, absence of hyphae and no features of a mature biofilm architecture. The increase in number of layers of graphene nanocoating did not improve its antibiofilm potential. SIGNIFICANCE The graphene nanocoating exerted a long-term persistent inhibitory effect on the biofilm formation on titanium. The fewer cells that were able to attach on graphene coated titanium were scattered and unable to form a mature biofilm with hyphae elements. The findings open opportunities to prevent microbial attachment and proliferation on implantable materials without the use of antibiotics.
Collapse
Affiliation(s)
| | - Kassapa Ellepola
- Louisiana State University Health Sciences Center, School of Dentistry, USA
| | - Nikolaos Silikas
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - A H Castro Neto
- Centre for Advanced 2D Materials and Graphene Research Centre, National University of Singapore, Singapore
| | - Chaminda Jayampath Seneviratne
- National Dental Centre Singapore, SingHealth, Duke NUS Medical School, 05, Hospital Avenue, National Dental Centre Singapor, Singapore.
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, 9 Lower Kent Ridge Road, Singapore; Centre for Advanced 2D Materials and Graphene Research Centre, National University of Singapore, Singapore; NUS Craniofacial Research and Innovation Center, National University of Singapore, Singapore.
| |
Collapse
|
23
|
Böttcher B, Hoffmann B, Garbe E, Weise T, Cseresnyés Z, Brandt P, Dietrich S, Driesch D, Figge MT, Vylkova S. The Transcription Factor Stp2 Is Important for Candida albicans Biofilm Establishment and Sustainability. Front Microbiol 2020; 11:794. [PMID: 32425915 PMCID: PMC7203782 DOI: 10.3389/fmicb.2020.00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/03/2020] [Indexed: 01/12/2023] Open
Abstract
The fungal pathogen Candida albicans forms polymorphic biofilms where hyphal morphogenesis and metabolic adaptation are tightly coordinated by a complex intertwined network of transcription factors. The sensing and metabolism of amino acids play important roles during various phases of biofilm development – from adhesion to maturation. Stp2 is a transcription factor that activates the expression of amino acid permease genes and is required for environmental alkalinization and hyphal growth in vitro and during macrophage phagocytosis. While it is well established that Stp2 is activated in response to external amino acids, its role in biofilm formation remains unknown. In addition to widely used techniques, we applied newly developed approaches for automated image analysis to quantify Stp2-regulated filamentation and biofilm growth. Our results show that in the stp2Δ deletion mutant adherence to abiotic surfaces and initial germ tube formation were strongly impaired, but formed mature biofilms with cell density and morphological structures comparable to the control strains. Stp2-dependent nutrient adaptation appeared to play an important role in biofilm development: stp2Δ biofilms formed under continuous nutrient flow displayed an overall reduction in biofilm formation, whereas under steady conditions the mutant strain formed biofilms with lower metabolic activity, resulting in increased cell survival and biofilm longevity. A deletion of STP2 led to increased rapamycin susceptibility and transcriptional activation of GCN4, the transcriptional regulator of the general amino acid control pathway, demonstrating a connection of Stp2 to other nutrient-responsive pathways. In summary, the transcription factor Stp2 is important for C. albicans biofilm formation, where it contributes to adherence and induction of morphogenesis, and mediates nutrient adaption and cell longevity in mature biofilms.
Collapse
Affiliation(s)
- Bettina Böttcher
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Bianca Hoffmann
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Enrico Garbe
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | | | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Philipp Brandt
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Stefanie Dietrich
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | | | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany.,Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Germany
| | - Slavena Vylkova
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| |
Collapse
|
24
|
Thomas G, Bain JM, Budge S, Brown AJP, Ames RM. Identifying Candida albicans Gene Networks Involved in Pathogenicity. Front Genet 2020; 11:375. [PMID: 32391057 PMCID: PMC7193023 DOI: 10.3389/fgene.2020.00375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/26/2020] [Indexed: 11/17/2022] Open
Abstract
Candida albicans is a normal member of the human microbiome. It is also an opportunistic pathogen, which can cause life-threatening systemic infections in severely immunocompromized individuals. Despite the availability of antifungal drugs, mortality rates of systemic infections are high and new drugs are needed to overcome therapeutic challenges including the emergence of drug resistance. Targeting known disease pathways has been suggested as a promising avenue for the development of new antifungals. However, <30% of C. albicans genes are verified with experimental evidence of a gene product, and the full complement of genes involved in important disease processes is currently unknown. Tools to predict the function of partially or uncharacterized genes and generate testable hypotheses will, therefore, help to identify potential targets for new antifungal development. Here, we employ a network-extracted ontology to leverage publicly available transcriptomics data and identify potential candidate genes involved in disease processes. A subset of these genes has been phenotypically screened using available deletion strains and we present preliminary data that one candidate, PEP8, is involved in hyphal development and immune evasion. This work demonstrates the utility of network-extracted ontologies in predicting gene function to generate testable hypotheses that can be applied to pathogenic systems. This could represent a novel first step to identifying targets for new antifungal therapies.
Collapse
Affiliation(s)
- Graham Thomas
- Biosciences, University of Exeter, Exeter, United Kingdom
| | - Judith M Bain
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Susan Budge
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Alistair J P Brown
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,MRC Centre for Medical Mycology at the University of Exeter, Biosciences, University of Exeter, Exeter, United Kingdom
| | - Ryan M Ames
- Biosciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
25
|
Zhou N, Jiang Y, Bergquist TR, Lee AJ, Kacsoh BZ, Crocker AW, Lewis KA, Georghiou G, Nguyen HN, Hamid MN, Davis L, Dogan T, Atalay V, Rifaioglu AS, Dalkıran A, Cetin Atalay R, Zhang C, Hurto RL, Freddolino PL, Zhang Y, Bhat P, Supek F, Fernández JM, Gemovic B, Perovic VR, Davidović RS, Sumonja N, Veljkovic N, Asgari E, Mofrad MRK, Profiti G, Savojardo C, Martelli PL, Casadio R, Boecker F, Schoof H, Kahanda I, Thurlby N, McHardy AC, Renaux A, Saidi R, Gough J, Freitas AA, Antczak M, Fabris F, Wass MN, Hou J, Cheng J, Wang Z, Romero AE, Paccanaro A, Yang H, Goldberg T, Zhao C, Holm L, Törönen P, Medlar AJ, Zosa E, Borukhov I, Novikov I, Wilkins A, Lichtarge O, Chi PH, Tseng WC, Linial M, Rose PW, Dessimoz C, Vidulin V, Dzeroski S, Sillitoe I, Das S, Lees JG, Jones DT, Wan C, Cozzetto D, Fa R, Torres M, Warwick Vesztrocy A, Rodriguez JM, Tress ML, Frasca M, Notaro M, Grossi G, Petrini A, Re M, Valentini G, Mesiti M, Roche DB, Reeb J, Ritchie DW, Aridhi S, Alborzi SZ, Devignes MD, Koo DCE, Bonneau R, Gligorijević V, Barot M, Fang H, Toppo S, Lavezzo E, et alZhou N, Jiang Y, Bergquist TR, Lee AJ, Kacsoh BZ, Crocker AW, Lewis KA, Georghiou G, Nguyen HN, Hamid MN, Davis L, Dogan T, Atalay V, Rifaioglu AS, Dalkıran A, Cetin Atalay R, Zhang C, Hurto RL, Freddolino PL, Zhang Y, Bhat P, Supek F, Fernández JM, Gemovic B, Perovic VR, Davidović RS, Sumonja N, Veljkovic N, Asgari E, Mofrad MRK, Profiti G, Savojardo C, Martelli PL, Casadio R, Boecker F, Schoof H, Kahanda I, Thurlby N, McHardy AC, Renaux A, Saidi R, Gough J, Freitas AA, Antczak M, Fabris F, Wass MN, Hou J, Cheng J, Wang Z, Romero AE, Paccanaro A, Yang H, Goldberg T, Zhao C, Holm L, Törönen P, Medlar AJ, Zosa E, Borukhov I, Novikov I, Wilkins A, Lichtarge O, Chi PH, Tseng WC, Linial M, Rose PW, Dessimoz C, Vidulin V, Dzeroski S, Sillitoe I, Das S, Lees JG, Jones DT, Wan C, Cozzetto D, Fa R, Torres M, Warwick Vesztrocy A, Rodriguez JM, Tress ML, Frasca M, Notaro M, Grossi G, Petrini A, Re M, Valentini G, Mesiti M, Roche DB, Reeb J, Ritchie DW, Aridhi S, Alborzi SZ, Devignes MD, Koo DCE, Bonneau R, Gligorijević V, Barot M, Fang H, Toppo S, Lavezzo E, Falda M, Berselli M, Tosatto SCE, Carraro M, Piovesan D, Ur Rehman H, Mao Q, Zhang S, Vucetic S, Black GS, Jo D, Suh E, Dayton JB, Larsen DJ, Omdahl AR, McGuffin LJ, Brackenridge DA, Babbitt PC, Yunes JM, Fontana P, Zhang F, Zhu S, You R, Zhang Z, Dai S, Yao S, Tian W, Cao R, Chandler C, Amezola M, Johnson D, Chang JM, Liao WH, Liu YW, Pascarelli S, Frank Y, Hoehndorf R, Kulmanov M, Boudellioua I, Politano G, Di Carlo S, Benso A, Hakala K, Ginter F, Mehryary F, Kaewphan S, Björne J, Moen H, Tolvanen MEE, Salakoski T, Kihara D, Jain A, Šmuc T, Altenhoff A, Ben-Hur A, Rost B, Brenner SE, Orengo CA, Jeffery CJ, Bosco G, Hogan DA, Martin MJ, O'Donovan C, Mooney SD, Greene CS, Radivojac P, Friedberg I. The CAFA challenge reports improved protein function prediction and new functional annotations for hundreds of genes through experimental screens. Genome Biol 2019; 20:244. [PMID: 31744546 PMCID: PMC6864930 DOI: 10.1186/s13059-019-1835-8] [Show More Authors] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The Critical Assessment of Functional Annotation (CAFA) is an ongoing, global, community-driven effort to evaluate and improve the computational annotation of protein function. RESULTS Here, we report on the results of the third CAFA challenge, CAFA3, that featured an expanded analysis over the previous CAFA rounds, both in terms of volume of data analyzed and the types of analysis performed. In a novel and major new development, computational predictions and assessment goals drove some of the experimental assays, resulting in new functional annotations for more than 1000 genes. Specifically, we performed experimental whole-genome mutation screening in Candida albicans and Pseudomonas aureginosa genomes, which provided us with genome-wide experimental data for genes associated with biofilm formation and motility. We further performed targeted assays on selected genes in Drosophila melanogaster, which we suspected of being involved in long-term memory. CONCLUSION We conclude that while predictions of the molecular function and biological process annotations have slightly improved over time, those of the cellular component have not. Term-centric prediction of experimental annotations remains equally challenging; although the performance of the top methods is significantly better than the expectations set by baseline methods in C. albicans and D. melanogaster, it leaves considerable room and need for improvement. Finally, we report that the CAFA community now involves a broad range of participants with expertise in bioinformatics, biological experimentation, biocuration, and bio-ontologies, working together to improve functional annotation, computational function prediction, and our ability to manage big data in the era of large experimental screens.
Collapse
Affiliation(s)
- Naihui Zhou
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.,Program in Bioinformatics and Computational Biology, Ames, IA, USA
| | - Yuxiang Jiang
- Indiana University Bloomington, Bloomington, Indiana, USA
| | - Timothy R Bergquist
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Alexandra J Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Balint Z Kacsoh
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Molecular and Systems Biology, Hanover, NH, USA
| | - Alex W Crocker
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Kimberley A Lewis
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - George Georghiou
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Huy N Nguyen
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.,Program in Computer Science, Ames, IA, USA
| | - Md Nafiz Hamid
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.,Program in Bioinformatics and Computational Biology, Ames, IA, USA
| | - Larry Davis
- Program in Bioinformatics and Computational Biology, Ames, IA, USA
| | - Tunca Dogan
- Department of Computer Engineering, Hacettepe University, Ankara, Turkey.,European Molecular Biolo gy Labora tory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Volkan Atalay
- Department of Computer Engineering, Middle East Technical University (METU), Ankara, Turkey
| | - Ahmet S Rifaioglu
- Department of Computer Engineering, Middle East Technical University (METU), Ankara, Turkey.,Department of Computer Engineering, Iskenderun Technical University, Hatay, Turkey
| | - Alperen Dalkıran
- Department of Computer Engineering, Middle East Technical University (METU), Ankara, Turkey
| | - Rengul Cetin Atalay
- CanSyL, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Chengxin Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Rebecca L Hurto
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Peter L Freddolino
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.,Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.,Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | | | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - José M Fernández
- INB Coordination Unit, Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, Spain.,(former) INB GN2, Structural and Computational Biology Programme, Spanish National Cancer Research Centre, Barcelona, Catalonia, Spain
| | - Branislava Gemovic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Vladimir R Perovic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Radoslav S Davidović
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Neven Sumonja
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Nevena Veljkovic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Ehsaneddin Asgari
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering, University of California Berkeley, Berkeley, CA, USA.,Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Berkeley, CA, USA
| | | | - Giuseppe Profiti
- Bologna Biocomputing Group, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,National Research Council, IBIOM, Bologna, Italy
| | - Castrense Savojardo
- Bologna Biocomputing Group, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Pier Luigi Martelli
- Bologna Biocomputing Group, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Rita Casadio
- Bologna Biocomputing Group, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Florian Boecker
- University of Bonn: INRES Crop Bioinformatics, Bonn, North Rhine-Westphalia, Germany
| | - Heiko Schoof
- INRES Crop Bioinformatics, University of Bonn, Bonn, Germany
| | - Indika Kahanda
- Gianforte School of Computing, Montana State University, Bozeman, Montana, USA
| | - Natalie Thurlby
- University of Bristol, Computer Science, Bristol, Bristol, United Kingdom
| | - Alice C McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Brunswick, Germany.,RESIST, DFG Cluster of Excellence 2155, Brunswick, Germany
| | - Alexandre Renaux
- Interuniversity Institute of Bioinformatics in Brussels, Université libre de Bruxelles - Vrije Universiteit Brussel, Brussels, Belgium.,Machine Learning Group, Université libre de Bruxelles, Brussels, Belgium.,Artificial Intelligence lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rabie Saidi
- European Molecular Biolo gy Labora tory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Julian Gough
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Alex A Freitas
- University of Kent, School of Computing, Canterbury, United Kingdom
| | - Magdalena Antczak
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Fabio Fabris
- University of Kent, School of Computing, Canterbury, United Kingdom
| | - Mark N Wass
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Jie Hou
- University of Missouri, Computer Science, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Zheng Wang
- University of Miami, Coral Gables, Florida, USA
| | - Alfonso E Romero
- Centre for Systems and Synthetic Biology, Department of Computer Science, Royal Holloway, University of London, Egham, Surrey, United Kingdom
| | - Alberto Paccanaro
- Centre for Systems and Synthetic Biology, Department of Computer Science, Royal Holloway, University of London, Egham, Surrey, United Kingdom
| | - Haixuan Yang
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Galway, Ireland.,Technical University of Munich, Garching, Germany
| | - Tatyana Goldberg
- Department of Informatics, Bioinformatics & Computational Biology-i12, Technische Universitat Munchen, Munich, Germany
| | - Chenguang Zhao
- Faculty for Informatics, Garching, Germany.,Department for Bioinformatics and Computational Biology, Garching, Germany.,School of Computing Sciences and Computer Engineering, Hattiesburg, Mississippi, USA
| | - Liisa Holm
- Institute of Biotechnology, Helsinki Institute of Life Sciences, University of Helsinki, Finland, Helsinki, Finland
| | - Petri Törönen
- Institute of Biotechnology, Helsinki Institute of Life Sciences, University of Helsinki, Finland, Helsinki, Finland
| | - Alan J Medlar
- Institute of Biotechnology, Helsinki Institute of Life Sciences, University of Helsinki, Finland, Helsinki, Finland
| | - Elaine Zosa
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Ilya Novikov
- Baylor College of Medicine, Department of Biochemistry and Molecular Biology, Houston, TX, USA
| | - Angela Wilkins
- Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, TX, USA
| | - Olivier Lichtarge
- Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, TX, USA
| | - Po-Han Chi
- National TsingHua University, Hsinchu, Taiwan
| | - Wei-Cheng Tseng
- Department of Electrical Engineering in National Tsing Hua University, Hsinchu City, Taiwan
| | - Michal Linial
- The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peter W Rose
- University of California San Diego, San Diego Supercomputer Center, La Jolla, California, USA
| | - Christophe Dessimoz
- Department of Computational Biology and Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Department of Genetics, Evolution & Environment, and Department of Computer Science, University College London, London, UK.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vedrana Vidulin
- Department of Knowledge Technologies, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Saso Dzeroski
- Jozef Stefan Institute, Ljubljana, Slovenia.,Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Ian Sillitoe
- Research Department of Structural and Molecular Biology, University College London, London, England
| | - Sayoni Das
- Research Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Jonathan Gill Lees
- Research Department of Structural and Molecular Biology, University College London, London, United Kingdom.,Department of Health and Life Sciences, Oxford Brookes University, London, UK
| | - David T Jones
- The Francis Crick Institute, Biomedical Data Science Laboratory, London, United Kingdom.,Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Cen Wan
- Department of Computer Science, University College London, London, United Kingdom.,The Francis Crick Institute, Biomedical Data Science Laboratory, London, United Kingdom
| | - Domenico Cozzetto
- Department of Computer Science, University College London, London, United Kingdom.,The Francis Crick Institute, Biomedical Data Science Laboratory, London, United Kingdom
| | - Rui Fa
- Department of Computer Science, University College London, London, United Kingdom.,The Francis Crick Institute, Biomedical Data Science Laboratory, London, United Kingdom
| | - Mateo Torres
- Centre for Systems and Synthetic Biology, Department of Computer Science, Royal Holloway, University of London, Egham, Surrey, United Kingdom
| | - Alex Warwick Vesztrocy
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, United Kingdom.,SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Jose Manuel Rodriguez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Michael L Tress
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marco Frasca
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Marco Notaro
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Giuliano Grossi
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Alessandro Petrini
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Matteo Re
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Giorgio Valentini
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy
| | - Marco Mesiti
- Università degli Studi di Milano - Computer Science Department - AnacletoLab, Milan, Milan, Italy.,Institut de Biologie Computationnelle, LIRMM, CNRS-UMR 5506, Universite de Montpellier, Montpellier, France
| | - Daniel B Roche
- Department of Informatics, Bioinformatics and Computational Biology-i12, Technische Universitat Munchen, Munich, Germany
| | - Jonas Reeb
- Department of Informatics, Bioinformatics and Computational Biology-i12, Technische Universitat Munchen, Munich, Germany
| | - David W Ritchie
- University of Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | - Sabeur Aridhi
- University of Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | | | - Marie-Dominique Devignes
- University of Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France.,University of Lorraine, Nancy, Lorraine, France.,Inria, Nancy, France
| | | | - Richard Bonneau
- NYU Center for Data Science, New York, 10010, NY, USA.,Flatiron Institute, CCB, New York, 10010, NY, USA
| | - Vladimir Gligorijević
- Center for Computational Biology (CCB), Flatiron Institute, Simons Foundation, New York, New York, USA
| | - Meet Barot
- Center for Data Science, New York University, New York, 10011, NY, USA
| | - Hai Fang
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Stefano Toppo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Enrico Lavezzo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marco Falda
- Department of Biology, University of Padova, Padova, Italy
| | - Michele Berselli
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Silvio C E Tosatto
- CNR Institute of Neuroscience, Padova, Italy.,Department of Biomedical Sciences, University of Padua, Padova, Italy
| | - Marco Carraro
- Department of Biomedical Sciences, University of Padua, Padova, Italy
| | - Damiano Piovesan
- Department of Biomedical Sciences, University of Padua, Padova, Italy
| | - Hafeez Ur Rehman
- Department of Computer Science, National University of Computer and Emerging Sciences, Peshawar, Khyber Pakhtoonkhwa, Pakistan
| | - Qizhong Mao
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA.,University of California, Riverside, Philadelphia, PA, USA
| | - Shanshan Zhang
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Slobodan Vucetic
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Gage S Black
- Department of Biology, Brigham Young University, Provo, UT, USA.,Bioinformatics Research Group, Provo, UT, USA
| | - Dane Jo
- Department of Biology, Brigham Young University, Provo, UT, USA.,Bioinformatics Research Group, Provo, UT, USA
| | - Erica Suh
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Jonathan B Dayton
- Department of Biology, Brigham Young University, Provo, UT, USA.,Bioinformatics Research Group, Provo, UT, USA
| | - Dallas J Larsen
- Department of Biology, Brigham Young University, Provo, UT, USA.,Bioinformatics Research Group, Provo, UT, USA
| | - Ashton R Omdahl
- Department of Biology, Brigham Young University, Provo, UT, USA.,Bioinformatics Research Group, Provo, UT, USA
| | - Liam J McGuffin
- School of Biological Sciences, University of Reading, Reading, England, United Kingdom
| | | | - Patricia C Babbitt
- Department of Pharmaceutical Chemistry, San Francisco, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 94158, CA, USA
| | - Jeffrey M Yunes
- UC Berkeley - UCSF Graduate Program in Bioengineering, University of California, San Francisco, 94158, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 94158, CA, USA
| | - Paolo Fontana
- Research and Innovation Center, Edmund Mach Foundation, San Michele all'Adige, Italy
| | - Feng Zhang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, Shanghai, China.,Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, Shanghai, China
| | - Shanfeng Zhu
- School of Computer Science and Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China.,Institute of Science and Technology for Brain-Inspired Intelligence and Shanghai Institute of Artificial Intelligence Algorithms, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Ronghui You
- School of Computer Science and Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China.,Institute of Science and Technology for Brain-Inspired Intelligence and Shanghai Institute of Artificial Intelligence Algorithms, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Zihan Zhang
- School of Computer Science and Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Suyang Dai
- School of Computer Science and Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Shuwei Yao
- School of Computer Science and Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China.,Institute of Science and Technology for Brain-Inspired Intelligence and Shanghai Institute of Artificial Intelligence Algorithms, Fudan University, Shanghai, China
| | - Weidong Tian
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, Shanghai, China.,Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Renzhi Cao
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, USA
| | - Caleb Chandler
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, USA
| | - Miguel Amezola
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, USA
| | - Devon Johnson
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, USA
| | - Jia-Ming Chang
- Department of Computer Science, National Chengchi University, Taipei, Taiwan
| | - Wen-Hung Liao
- Department of Computer Science, National Chengchi University, Taipei, Taiwan
| | - Yi-Wei Liu
- Department of Computer Science, National Chengchi University, Taipei, Taiwan
| | | | | | - Robert Hoehndorf
- Computer, Electrical and Mathematical Sciences & Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Jeddah, Saudi Arabia
| | - Maxat Kulmanov
- Computer, Electrical and Mathematical Sciences & Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Jeddah, Saudi Arabia
| | - Imane Boudellioua
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.,Computer, Electrical and Mathematical Sciences Engineering Division (CEMSE), King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Gianfranco Politano
- Control and Computer Engineering Department, Politecnico di Torino, Torino, TO, Italy
| | - Stefano Di Carlo
- Control and Computer Engineering Department, Politecnico di Torino, Torino, TO, Italy
| | - Alfredo Benso
- Control and Computer Engineering Department, Politecnico di Torino, Torino, TO, Italy
| | - Kai Hakala
- Department of Future Technologies, Turku NLP Group, University of Turku, Turku, Finland.,University of Turku Graduate School (UTUGS), Turku, Finland
| | - Filip Ginter
- Department of Future Technologies, Turku NLP Group, University of Turku, Turku, Finland.,University of Turku, Turku, Finland
| | - Farrokh Mehryary
- Department of Future Technologies, Turku NLP Group, University of Turku, Turku, Finland.,University of Turku Graduate School (UTUGS), Turku, Finland
| | - Suwisa Kaewphan
- Department of Future Technologies, Turku NLP Group, University of Turku, Turku, Finland.,University of Turku Graduate School (UTUGS), Turku, Finland.,Turku Centre for Computer Science (TUCS), Turku, Finland
| | - Jari Björne
- Department of Future Technologies, Faculty of Science and Engineering, University of Turku, Turku, FI-20014, Finland.,Turku Centre for Computer Science (TUCS), Agora, Vesilinnantie 3, Turku, FI-20500, Finland
| | | | | | - Tapio Salakoski
- Department of Future Technologies, Faculty of Science and Engineering, University of Turku, Turku, FI-20014, Finland.,Turku Centre for Computer Science (TUCS), Agora, Vesilinnantie 3, Turku, FI-20500, Finland
| | - Daisuke Kihara
- Department of Biological Sciences, Department of Computer Science, Purdue University, 47907, IN, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, 45229, OH, USA
| | - Aashish Jain
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Tomislav Šmuc
- Division of Electronics, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Adrian Altenhoff
- Department of Computer Science, ETH Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Asa Ben-Hur
- Department of Computer Science, Colorado State University, Fort Collins, CO, USA
| | - Burkhard Rost
- Department of Informatics, Bioinformatics & Computational Biology-i12, Technische Universitat Munchen, Munich, Germany.,Institute for Food and Plant Sciences WZW, Technische Universität München, Freising, Germany
| | | | - Christine A Orengo
- Research Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Constance J Jeffery
- Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Deborah A Hogan
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Maria J Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Claire O'Donovan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Sean D Mooney
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Childhood Cancer Data Lab, Alex's Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA.
| | - Iddo Friedberg
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
26
|
Inhibition of Dephosphorylation of Dolichyl Diphosphate Alters the Synthesis of Dolichol and Hinders Protein N-Glycosylation and Morphological Transitions in Candida albicans. Int J Mol Sci 2019; 20:ijms20205067. [PMID: 31614738 PMCID: PMC6829516 DOI: 10.3390/ijms20205067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 11/17/2022] Open
Abstract
The essential role of dolichyl phosphate (DolP) as a carbohydrate carrier during protein N-glycosylation is well established. The cellular pool of DolP is derived from de novo synthesis in the dolichol branch of the mevalonate pathway and from recycling of DolPP after each cycle of N-glycosylation, when the oligosaccharide is transferred from the lipid carrier to the protein and DolPP is released and then dephosphorylated. In Saccharomyces cerevisiae, the dephosphorylation of DolPP is known to be catalyzed by the Cwh8p protein. To establish the role of the Cwh8p orthologue in another distantly related yeast species, Candida albicans, we studied its mutant devoid of the CaCWH8 gene. A double Cacwh8∆/Cacwh8∆ strain was constructed by the URA-blaster method. As in S. cerevisiae, the mutant was impaired in DolPP recycling. This defect, however, was accompanied by an elevation of cis-prenyltransferase activity and higher de novo production of dolichols. Despite these compensatory changes, protein glycosylation, cell wall integrity, filamentous growth, and biofilm formation were impaired in the mutant. These results suggest that the defects are not due to the lack of DolP for the protein N-glycosylation but rather that the activity of oligosacharyltransferase could be inhibited by the excess DolPP accumulating in the mutant.
Collapse
|
27
|
Shih PY, Liao YT, Tseng YK, Deng FS, Lin CH. A Potential Antifungal Effect of Chitosan Against Candida albicans Is Mediated via the Inhibition of SAGA Complex Component Expression and the Subsequent Alteration of Cell Surface Integrity. Front Microbiol 2019; 10:602. [PMID: 30972050 PMCID: PMC6443709 DOI: 10.3389/fmicb.2019.00602] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/11/2019] [Indexed: 12/21/2022] Open
Abstract
Due to the high incidence of nosocomial Candida albicans infection, the first-line drugs for C. albicans infection have been heavily used, and the emergence of drug-resistant strains has gradually increased. Thus, a new antifungal drug or therapeutic method is needed. Chitosan, a product of chitin deacetylation, is considered to be potentially therapeutic for fungal infections because of its excellent biocompatibility, biodegradability and low toxicity. The biocidal action of chitosan against C. albicans shows great commercial potential, but the exact mechanisms underlying its antimicrobial activity are unclear. To reveal these mechanisms, mutant library screening was performed. ADA2 gene, which encodes a histone acetylation coactivator in the SAGA complex, was identified. Transmission electronic microscopy images showed that the surface of chitosan-treated ada2Δ cells was substantially disrupted and displayed an irregular morphology. Interestingly, the cell wall of ada2Δ cells was significantly thinner than that of wild-type cells, with a thickness similar to that seen in the chitosan-treated wild-type strain. Although ADA2 is required for chitosan tolerance, expression of ADA2 and several Ada2-mediated cell wall-related genes (ALS2, PGA45, and ACE2) and efflux transporter genes (MDR1 and CDR1) were significantly inhibited by chitosan. Furthermore, GCN5 encoding a SAGA complex catalytic subunit was inhibited by chitosan, and gcn5Δ cells exhibited phenotypes comparable to those of ada2Δ cells in response to chitosan and other cell surface-disrupting agents. This study demonstrated that a potential antifungal mechanism of chitosan against C. albicans operates by inhibiting SAGA complex gene expression, which decreases the protection of the cell surface against chitosan.
Collapse
Affiliation(s)
- Pei-Yu Shih
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Liao
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Kai Tseng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Fu-Sheng Deng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Phenotypic and genotypic evaluation of adherence and biofilm development in Candida albicans respiratory tract isolates from hospitalized patients. REV ROMANA MED LAB 2019. [DOI: 10.2478/rrlm-2019-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Abstract
In recent years, a significant number of epidemiological variations have been observed for fungal infections. In immunocompromised patients, Candida albicans is crucially involved in invasive infections, mostly originating in respiratory tract colonization. The global rise in candidiasis has led researchers to investigate possible correlations between fungal strains virulence profiles and their pathogenic potential, among the most investigated genes being those involved in adherence and biofilm development. In this study, we established the adherence gene profiles of C. albicans strains isolated from respiratory tract secretions in patients hospitalized for cardiovascular diseases and correlated them with the ability of the respective strains to colonize the epithelial cells and form biofilms on the inert substratum. The strains isolated from the lower respiratory tract exhibited the highest adherence capacity and were intensive biofilm producers. The SAP9, ALS3, ALS5, and ALS6 genes were the most frequently detected. There was a significant association between the presence of ALS 3 gene and the cellular substrate colonizing potential of the harboring strains. We also found that the strains expressing SAP9 were more virulent in the phenotypic assays. Detecting the presence of adherence genes from different clinical isolates is a cost-effective tool that would allow researchers to predict the virulence of a certain strain and estimate its potential to adhere to host cells and develop biofilms.
Collapse
|
29
|
Fedorova O, Jagdmann GE, Adams RL, Yuan L, Van Zandt MC, Pyle AM. Small molecules that target group II introns are potent antifungal agents. Nat Chem Biol 2018; 14:1073-1078. [PMID: 30323219 PMCID: PMC6239893 DOI: 10.1038/s41589-018-0142-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 08/29/2018] [Indexed: 11/09/2022]
Abstract
Specific RNA structures control numerous metabolic processes that impact human health, and yet efforts to target RNA structures de novo have been limited. In eukaryotes, the self-splicing group II intron is a mitochondrial RNA tertiary structure that is absent in vertebrates but essential for respiration in plants, fungi and yeast. Here we show that this RNA can be targeted through a process of high-throughput in vitro screening, SAR and lead optimization, resulting in high-affinity compounds that specifically inhibit group IIB intron splicing in vitro and in vivo and lack toxicity in human cells. The compounds are potent growth inhibitors of the pathogen Candida parapsilosis, displaying antifungal activity comparable to that of amphotericin B. These studies demonstrate that RNA tertiary structures can be successfully targeted de novo, resulting in pharmacologically valuable compounds.
Collapse
Affiliation(s)
- Olga Fedorova
- Howard Hughes Medical Institute, New Haven, CT, USA.,Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | | | - Rebecca L Adams
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Lin Yuan
- New England Discovery Partners, Branford, CT, USA
| | | | - Anna Marie Pyle
- Howard Hughes Medical Institute, New Haven, CT, USA. .,Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.
| |
Collapse
|
30
|
Candida albicans Sfl1/Sfl2 regulatory network drives the formation of pathogenic microcolonies. PLoS Pathog 2018; 14:e1007316. [PMID: 30252918 PMCID: PMC6173444 DOI: 10.1371/journal.ppat.1007316] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/05/2018] [Accepted: 09/04/2018] [Indexed: 11/19/2022] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can infect oral mucosal surfaces while being under continuous flow from saliva. Under specific conditions, C. albicans will form microcolonies that more closely resemble the biofilms formed in vivo than standard in vitro biofilm models. However, very little is known about these microcolonies, particularly genomic differences between these specialized biofilm structures and the traditional in vitro biofilms. In this study, we used a novel flow system, in which C. albicans spontaneously forms microcolonies, to further characterize the architecture of fungal microcolonies and their genomics compared to non-microcolony conditions. Fungal microcolonies arose from radially branching filamentous hyphae that increasingly intertwined with one another to form extremely dense biofilms, and closely resembled the architecture of in vivo oropharyngeal candidiasis. We identified 20 core microcolony genes that were differentially regulated in flow-induced microcolonies using RNA-seq. These genes included HWP1, ECE1, IHD1, PLB1, HYR1, PGA10, and SAP5. A predictive algorithm was utilized to identify ten transcriptional regulators potentially involved in microcolony formation. Of these transcription factors, we found that Rob1, Ndt80, Sfl1 and Sfl2, played a key role in microcolony formation under both flow and static conditions and to epithelial surfaces. Expression of core microcolony genes were highly up-regulated in Δsfl1 cells and down-regulated in both Δsfl2 and Δrob1 strains. Microcolonies formed on oral epithelium using C. albicans Δsfl1, Δsfl2 and Δrob1 deletion strains all had altered adhesion, invasion and cytotoxicity. Furthermore, epithelial cells infected with deletion mutants had reduced (SFL2, NDT80, and ROB1) or enhanced (SFL2) immune responses, evidenced by phosphorylation of MKP1 and c-Fos activation, key signal transducers in the hyphal invasion response. This profile of microcolony transcriptional regulators more closely reflects Sfl1 and Sfl2 hyphal regulatory networks than static biofilm regulatory networks, suggesting that microcolonies are a specialized pathogenic form of biofilm.
Collapse
|
31
|
Gulati M, Lohse MB, Ennis CL, Gonzalez RE, Perry AM, Bapat P, Arevalo AV, Rodriguez DL, Nobile CJ. In Vitro Culturing and Screening of Candida albicans Biofilms. ACTA ACUST UNITED AC 2018; 50:e60. [PMID: 29995344 DOI: 10.1002/cpmc.60] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Candida albicans is a normal member of the human microbiota that asymptomatically colonizes healthy individuals, however it is also an opportunistic pathogen that can cause severe infections, especially in immunocompromised individuals. The medical impact of C. albicans depends, in part, on its ability to form biofilms, communities of adhered cells encased in an extracellular matrix. Biofilms can form on both biotic and abiotic surfaces, such as tissues and implanted medical devices. Once formed, biofilms are highly resistant to antifungal agents and the host immune system, and can act as a protected reservoir to seed disseminated infections. Here, we present several in vitro biofilm protocols, including protocols that are optimized for high-throughput screening of mutant libraries and antifungal compounds. We also present protocols to examine specific stages of biofilm development and protocols to evaluate interspecies biofilms that C. albicans forms with interacting microbial partners. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Megha Gulati
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| | - Matthew B Lohse
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California.,Department of Biology, BioSynesis, Inc., San Francisco, California
| | - Craig L Ennis
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Ruth E Gonzalez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| | - Austin M Perry
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Priyanka Bapat
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Ashley Valle Arevalo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Diana L Rodriguez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, California
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, California
| |
Collapse
|
32
|
Jung SI, Rodriguez N, Irrizary J, Liboro K, Bogarin T, Macias M, Eivers E, Porter E, Filler SG, Park H. Yeast casein kinase 2 governs morphology, biofilm formation, cell wall integrity, and host cell damage of Candida albicans. PLoS One 2017; 12:e0187721. [PMID: 29107946 PMCID: PMC5673188 DOI: 10.1371/journal.pone.0187721] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/24/2017] [Indexed: 11/18/2022] Open
Abstract
The regulatory networks governing morphogenesis of a pleomorphic fungus, Candida albicans are extremely complex and remain to be completely elucidated. This study investigated the function of C. albicans yeast casein kinase 2 (CaYck2p). The yck2Δ/yck2Δ strain displayed constitutive pseudohyphae in both yeast and hyphal growth conditions, and formed enhanced biofilm under non-biofilm inducing condition. This finding was further supported by gene expression analysis of the yck2Δ/yck2Δ strain which showed significant upregulation of UME6, a key transcriptional regulator of hyphal transition and biofilm formation, and cell wall protein genes ALS3, HWP1, and SUN41, all of which are associated with morphogenesis and biofilm architecture. The yck2Δ/yck2Δ strain was hypersensitive to cell wall damaging agents and had increased compensatory chitin deposition in the cell wall accompanied by an upregulation of the expression of the chitin synthase genes, CHS2, CHS3, and CHS8. Absence of CaYck2p also affected fungal-host interaction; the yck2Δ/yck2Δ strain had significantly reduced ability to damage host cells. However, the yck2Δ/yck2Δ strain had wild-type susceptibility to cyclosporine and FK506, suggesting that CaYck2p functions independently from the Ca+/calcineurin pathway. Thus, in C. albicans, Yck2p is a multifunctional kinase that governs morphogenesis, biofilm formation, cell wall integrity, and host cell interactions.
Collapse
Affiliation(s)
- Sook-In Jung
- Division of Infectious Diseases, Chonnam National University Medical School, Gwangju, South Korea
| | - Natalie Rodriguez
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Jihyun Irrizary
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Karl Liboro
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Thania Bogarin
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Marlene Macias
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Edward Eivers
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Edith Porter
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
| | - Scott G. Filler
- Division of Infectious Diseases, Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Hyunsook Park
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Abstract
Candida albicans is among the most prevalent fungal species of the human microbiota and asymptomatically colonizes healthy individuals. However, it is also an opportunistic pathogen that can cause severe, and often fatal, bloodstream infections. The medical impact of C. albicans typically depends on its ability to form biofilms, which are closely packed communities of cells that attach to surfaces, such as tissues and implanted medical devices. In this Review, we provide an overview of the processes involved in the formation of C. albicans biofilms and discuss the core transcriptional network that regulates biofilm development. We also consider some of the advantages that biofilms provide to C. albicans in comparison with planktonic growth and explore polymicrobial biofilms that are formed by C. albicans and certain bacterial species.
Collapse
|
34
|
Abstract
We focus this article on turning a biofilm inside out. The "inside" of the biofilm comprises the individual biofilm-related phenotypes, their environmental drivers and genetic determinants, and the coordination of gene functions through transcriptional regulators. Investigators have viewed the inside of the biofilm through diverse approaches, and this article will attempt to capture the essence of many. The ultimate goal is to connect the inside to the "outside," which we view as biofilm structure, development, pharmacological attributes, and medical impact.
Collapse
|
35
|
Assessment and Optimizations of Candida albicans In Vitro Biofilm Assays. Antimicrob Agents Chemother 2017; 61:AAC.02749-16. [PMID: 28289028 DOI: 10.1128/aac.02749-16] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/03/2017] [Indexed: 01/07/2023] Open
Abstract
Candida albicans biofilms have a significant medical impact due to their rapid growth on implanted medical devices, their resistance to antifungal drugs, and their ability to seed disseminated infections. Biofilm assays performed in vitro allow for rapid, high-throughput screening of gene deletion libraries or antifungal compounds and typically serve as precursors to in vivo studies. Here, we compile and discuss the protocols for several recently published C. albicansin vitro biofilm assays. We also describe improved versions of these protocols as well as novel in vitro assays. Finally, we consider some of the advantages and disadvantages of these different types of assays.
Collapse
|
36
|
Niewiadomska M, Janik A, Perlińska-Lenart U, Piłsyk S, Palamarczyk G, Kruszewska JS. The role of Alg13 N-acetylglucosaminyl transferase in the expression of pathogenic features of Candida albicans. Biochim Biophys Acta Gen Subj 2017; 1861:789-801. [DOI: 10.1016/j.bbagen.2017.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 01/16/2017] [Accepted: 01/21/2017] [Indexed: 01/09/2023]
|
37
|
Abstract
We focus this article on turning a biofilm inside out. The "inside" of the biofilm comprises the individual biofilm-related phenotypes, their environmental drivers and genetic determinants, and the coordination of gene functions through transcriptional regulators. Investigators have viewed the inside of the biofilm through diverse approaches, and this article will attempt to capture the essence of many. The ultimate goal is to connect the inside to the "outside," which we view as biofilm structure, development, pharmacological attributes, and medical impact.
Collapse
Affiliation(s)
- Katherine Lagree
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Aaron P Mitchell
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
38
|
Abstract
Candida albicans, the most pervasive fungal pathogen that colonizes humans, forms biofilms that are architecturally complex. They consist of a basal yeast cell polylayer and an upper region of hyphae encapsulated in extracellular matrix. However, biofilms formed in vitro vary as a result of the different conditions employed in models, the methods used to assess biofilm formation, strain differences, and, in a most dramatic fashion, the configuration of the mating type locus (MTL). Therefore, integrating data from different studies can lead to problems of interpretation if such variability is not taken into account. Here we review the conditions and factors that cause biofilm variation, with the goal of engendering awareness that more attention must be paid to the strains employed, the methods used to assess biofilm development, every aspect of the model employed, and the configuration of the MTL locus. We end by posing a set of questions that may be asked in comparing the results of different studies and developing protocols for new ones. This review should engender the notion that not all biofilms are created equal.
Collapse
Affiliation(s)
- David R Soll
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| | - Karla J Daniels
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Abstract
In humans, microbial cells (including bacteria, archaea, and fungi) greatly outnumber host cells. Candida albicans is the most prevalent fungal species of the human microbiota; this species asymptomatically colonizes many areas of the body, particularly the gastrointestinal and genitourinary tracts of healthy individuals. Alterations in host immunity, stress, resident microbiota, and other factors can lead to C. albicans overgrowth, causing a wide range of infections, from superficial mucosal to hematogenously disseminated candidiasis. To date, most studies of C. albicans have been carried out in suspension cultures; however, the medical impact of C. albicans (like that of many other microorganisms) depends on its ability to thrive as a biofilm, a closely packed community of cells. Biofilms are notorious for forming on implanted medical devices, including catheters, pacemakers, dentures, and prosthetic joints, which provide a surface and sanctuary for biofilm growth. C. albicans biofilms are intrinsically resistant to conventional antifungal therapeutics, the host immune system, and other environmental perturbations, making biofilm-based infections a significant clinical challenge. Here, we review our current knowledge of biofilms formed by C. albicans and closely related fungal species.
Collapse
Affiliation(s)
- Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, California 95343;
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143;
| |
Collapse
|
40
|
Abstract
The fungus Candida albicans is a major source of device-associated infection because of its capacity for biofilm formation. It is part of the natural mucosal flora and thus has access to available niches that can lead to infection. In this chapter we discuss the major properties of C. albicans biofilms and the insight that has been gleaned from their genetic determinants. Our specific areas of focus include biofilm structure and development, cell morphology and biofilm formation, biofilm-associated gene expression, the cell surface and adherence, the extracellular matrix, biofilm metabolism, and biofilm drug resistance.
Collapse
|
41
|
Verma-Gaur J, Traven A. Post-transcriptional gene regulation in the biology and virulence of Candida albicans. Cell Microbiol 2016; 18:800-6. [PMID: 26999710 PMCID: PMC5074327 DOI: 10.1111/cmi.12593] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/28/2016] [Accepted: 03/16/2016] [Indexed: 11/27/2022]
Abstract
In the human fungal pathogen Candida albicans, remodelling of gene expression drives host adaptation and virulence. Recent studies revealed that in addition to transcription, post‐transcriptional mRNA control plays important roles in virulence‐related pathways. Hyphal morphogenesis, biofilm formation, stress responses, antifungal drug susceptibility and virulence in animal models require post‐transcriptional regulators. This includes RNA binding proteins that control mRNA localization, decay and translation, as well as the cytoplasmic mRNA decay pathway. Comprehensive understanding of how modulation of gene expression networks drives C. albicans virulence will necessitate integration of our knowledge on transcriptional and post‐transcriptional mRNA control.
Collapse
Affiliation(s)
- Jiyoti Verma-Gaur
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ana Traven
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
42
|
Tati S, Davidow P, McCall A, Hwang-Wong E, Rojas IG, Cormack B, Edgerton M. Candida glabrata Binding to Candida albicans Hyphae Enables Its Development in Oropharyngeal Candidiasis. PLoS Pathog 2016; 12:e1005522. [PMID: 27029023 PMCID: PMC4814137 DOI: 10.1371/journal.ppat.1005522] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 03/02/2016] [Indexed: 11/18/2022] Open
Abstract
Pathogenic mechanisms of Candida glabrata in oral candidiasis, especially because of its inability to form hyphae, are understudied. Since both Candida albicans and C. glabrata are frequently co-isolated in oropharyngeal candidiasis (OPC), we examined their co-adhesion in vitro and observed adhesion of C. glabrata only to C. albicans hyphae microscopically. Mice were infected sublingually with C. albicans or C. glabrata individually, or with both species concurrently, to study their ability to cause OPC. Infection with C. glabrata alone resulted in negligible infection of tongues; however, colonization by C. glabrata was increased by co-infection or a pre-established infection with C. albicans. Furthermore, C. glabrata required C. albicans for colonization of tongues, since decreasing C. albicans burden with fluconazole also reduced C. glabrata. C. albicans hyphal wall adhesins Als1 and Als3 were important for in vitro adhesion of C. glabrata and to establish OPC. C. glabrata cell wall protein coding genes EPA8, EPA19, AWP2, AWP7, and CAGL0F00181 were implicated in mediating adhesion to C. albicans hyphae and remarkably, their expression was induced by incubation with germinated C. albicans. Thus, we found a near essential requirement for the presence of C. albicans for both initial colonization and establishment of OPC infection by C. glabrata.
Collapse
Affiliation(s)
- Swetha Tati
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, United States of America
| | - Peter Davidow
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, United States of America
| | - Andrew McCall
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, United States of America
| | - Elizabeth Hwang-Wong
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Isolde G. Rojas
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, United States of America
| | - Brendan Cormack
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mira Edgerton
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
43
|
Li TX, Yang MH, Wang XB, Wang Y, Kong LY. Synergistic Antifungal Meroterpenes and Dioxolanone Derivatives from the Endophytic Fungus Guignardia sp. JOURNAL OF NATURAL PRODUCTS 2015; 78:2511-2520. [PMID: 26577190 DOI: 10.1021/acs.jnatprod.5b00008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Nine new meroterpenes (1-9) and one new dioxolanone derivative (10), along with seven known compounds (11-17), were isolated from solid cultures of the endophytic fungus Guignardia sp., obtained from Euphorbia sieboldiana. Their structures were elucidated by analysis of UV, IR, 1D and 2D NMR, and HRESIMS data, and their absolute configurations were determined by a combination of single-crystal X-ray studies, modified Mosher methods, and Rh2(OCOCF3)4- and Mo2(OCOCH3)4-induced electronic circular dichroism experiments. All compounds were evaluated for their inhibitory effects alone and with fluconazole on the growth and biofilms of Candida albicans. At 6.3 μg/mL combined with 0.031 μg/mL of fluconazole, compounds 8 and 16 were found to have prominent inhibition on the growth of C. albicans with fractional inhibitory concentration index values of 0.23 and 0.19, respectively. Combined with fluconazole, both of them (40 μg/mL for 8 and 20 μg/mL for 16) could also inhibit C. albicans biofilms and reverse the tolerance of C. albicans biofilms to fluconazole.
Collapse
Affiliation(s)
- Tian-Xiao Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Ming-Hua Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Xiao-Bing Wang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Ying Wang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University , 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| |
Collapse
|
44
|
Kolondra A, Labedzka-Dmoch K, Wenda JM, Drzewicka K, Golik P. The transcriptome of Candida albicans mitochondria and the evolution of organellar transcription units in yeasts. BMC Genomics 2015; 16:827. [PMID: 26487099 PMCID: PMC4618339 DOI: 10.1186/s12864-015-2078-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
Background Yeasts show remarkable variation in the organization of their mitochondrial genomes, yet there is little experimental data on organellar gene expression outside few model species. Candida albicans is interesting as a human pathogen, and as a representative of a clade that is distant from the model yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe. Unlike them, it encodes seven Complex I subunits in its mtDNA. No experimental data regarding organellar expression were available prior to this study. Methods We used high-throughput RNA sequencing and traditional RNA biology techniques to study the mitochondrial transcriptome of C. albicans strains BWP17 and SN148. Results The 14 protein-coding genes, two ribosomal RNA genes, and 24 tRNA genes are expressed as eight primary polycistronic transcription units. We also found transcriptional activity in the noncoding regions, and antisense transcripts that could be a part of a regulatory mechanism. The promoter sequence is a variant of the nonanucleotide identified in other yeast mtDNAs, but some of the active promoters show significant departures from the consensus. The primary transcripts are processed by a tRNA punctuation mechanism into the monocistronic and bicistronic mature RNAs. The steady state levels of various mature transcripts exhibit large differences that are a result of posttranscriptional regulation. Transcriptome analysis allowed to precisely annotate the positions of introns in the RNL (2), COB (2) and COX1 (4) genes, as well as to refine the annotation of tRNAs and rRNAs. Comparative study of the mitochondrial genome organization in various Candida species indicates that they undergo shuffling in blocks usually containing 2–3 genes, and that their arrangement in primary transcripts is not conserved. tRNA genes with their associated promoters, as well as GC-rich sequence elements play an important role in these evolutionary events. Conclusions The main evolutionary force shaping the mitochondrial genomes of yeasts is the frequent recombination, constantly breaking apart and joining genes into novel primary transcription units. The mitochondrial transcription units are constantly rearranged in evolution shaping the features of gene expression, such as the presence of secondary promoter sites that are inactive, or act as “booster” promoters, simplified transcriptional regulation and reliance on posttranscriptional mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2078-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adam Kolondra
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland.
| | - Karolina Labedzka-Dmoch
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland.
| | - Joanna M Wenda
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland.
| | - Katarzyna Drzewicka
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland.
| | - Pawel Golik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland. .,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106, Warsaw, Poland.
| |
Collapse
|
45
|
Potential Targets for Antifungal Drug Discovery Based on Growth and Virulence in Candida albicans. Antimicrob Agents Chemother 2015. [PMID: 26195510 DOI: 10.1128/aac.00726-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fungal infections, especially infections caused by Candida albicans, remain a challenging problem in clinical settings. Despite the development of more-effective antifungal drugs, their application is limited for various reasons. Thus, alternative treatments with drugs aimed at novel targets in C. albicans are needed. Knowledge of growth and virulence in fungal cells is essential not only to understand their pathogenic mechanisms but also to identify potential antifungal targets. This article reviews the current knowledge of the mechanisms of growth and virulence in C. albicans and examines potential targets for the development of new antifungal drugs.
Collapse
|
46
|
Cabral V, Znaidi S, Walker LA, Martin-Yken H, Dague E, Legrand M, Lee K, Chauvel M, Firon A, Rossignol T, Richard ML, Munro CA, Bachellier-Bassi S, d'Enfert C. Targeted changes of the cell wall proteome influence Candida albicans ability to form single- and multi-strain biofilms. PLoS Pathog 2014; 10:e1004542. [PMID: 25502890 PMCID: PMC4263760 DOI: 10.1371/journal.ppat.1004542] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 10/28/2014] [Indexed: 12/29/2022] Open
Abstract
Biofilm formation is an important virulence trait of the pathogenic yeast Candida albicans. We have combined gene overexpression, strain barcoding and microarray profiling to screen a library of 531 C. albicans conditional overexpression strains (∼10% of the genome) for genes affecting biofilm development in mixed-population experiments. The overexpression of 16 genes increased strain occupancy within a multi-strain biofilm, whereas overexpression of 4 genes decreased it. The set of 16 genes was significantly enriched for those encoding predicted glycosylphosphatidylinositol (GPI)-modified proteins, namely Ihd1/Pga36, Phr2, Pga15, Pga19, Pga22, Pga32, Pga37, Pga42 and Pga59; eight of which have been classified as pathogen-specific. Validation experiments using either individually- or competitively-grown overexpression strains revealed that the contribution of these genes to biofilm formation was variable and stage-specific. Deeper functional analysis of PGA59 and PGA22 at a single-cell resolution using atomic force microscopy showed that overexpression of either gene increased C. albicans ability to adhere to an abiotic substrate. However, unlike PGA59, PGA22 overexpression led to cell cluster formation that resulted in increased sensitivity to shear forces and decreased ability to form a single-strain biofilm. Within the multi-strain environment provided by the PGA22-non overexpressing cells, PGA22-overexpressing cells were protected from shear forces and fitter for biofilm development. Ultrastructural analysis, genome-wide transcript profiling and phenotypic analyses in a heterologous context suggested that PGA22 affects cell adherence through alteration of cell wall structure and/or function. Taken together, our findings reveal that several novel predicted GPI-modified proteins contribute to the cooperative behaviour between biofilm cells and are important participants during C. albicans biofilm formation. Moreover, they illustrate the power of using signature tagging in conjunction with gene overexpression for the identification of novel genes involved in processes pertaining to C. albicans virulence. Candida albicans is the most prevalent human fungal pathogen. Its ability to cause disease relies, in part, on the formation of biofilms, a protective structure of highly adherent cells tolerant to antifungal agents and the host immune response. The biofilm is considered as a persistent root of infection, disseminating infectious cells to other locations. In this study, we performed large-scale phenotypic analyses aimed at identifying genes whose overexpression affects biofilm development in C. albicans. Our screen relied on a collection of 531 C. albicans strains, each conditionally overexpressing one given gene and carrying one specific molecular tag allowing the quantification of strain abundance in mixed-population experiments. Our results strikingly revealed the enrichment of strains overproducing poorly-characterized surface proteins called Pgas (Putative GPI-Anchored proteins), within a 531-strain-containing biofilm model. We show that these PGA genes differentially contribute to single-strain and multi-strain biofilm formation and are involved in specific stages of the biofilm developmental process. Taken together, our results reveal the importance of C. albicans cell surface proteins during biofilm formation and reflect the powerful use of strain barcoding in combination with gene overexpression to identify genes and/or pathways involved in processes pertaining to virulence of pathogenic microbes.
Collapse
Affiliation(s)
- Vitor Cabral
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
- Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Sadri Znaidi
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Louise A. Walker
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Hélène Martin-Yken
- INSA, UPS, INP, ISAE, LAAS, Université de Toulouse, Toulouse, France
- UMR792 Ingénierie des Systèmes Biologiques et des Procédés, INRA, Toulouse, France
- UMR5504, CNRS, Toulouse, France
| | - Etienne Dague
- INSA, UPS, INP, ISAE, LAAS, Université de Toulouse, Toulouse, France
- LAAS, CNRS, Toulouse, France
| | - Mélanie Legrand
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Keunsook Lee
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Murielle Chauvel
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Arnaud Firon
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Tristan Rossignol
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Mathias L. Richard
- INRA, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Thiverval Grignon, France
| | - Carol A. Munro
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Sophie Bachellier-Bassi
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
| | - Christophe d'Enfert
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et Génétique, Paris, France
- INRA, USC2019, Paris, France
- * E-mail:
| |
Collapse
|
47
|
Rajendran R, Sherry L, Lappin DF, Nile CJ, Smith K, Williams C, Munro CA, Ramage G. Extracellular DNA release confers heterogeneity in Candida albicans biofilm formation. BMC Microbiol 2014; 14:303. [PMID: 25476750 PMCID: PMC4262977 DOI: 10.1186/s12866-014-0303-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/19/2014] [Indexed: 12/19/2022] Open
Abstract
Background Biofilm formation by Candida albicans has shown to be highly variable and is directly associated with pathogenicity and poor clinical outcomes in patients at risk. The aim of this study was to test the hypotheses that the extracellular DNA release by C. albicans is strain dependent and is associated with biofilm heterogeneity. Results Initially, biofilm formed by C. albicans high biofilm formers (HBF) or low biofilm formers (LBF) were treated with DNase to find whether eDNA play a role in their biofilm formation. Digestion of biofilm eDNA significantly reduced the HBF biofilm biomass by five fold compared to untreated controls. In addition, quantification of eDNA over the period of biofilm formation by SYBR green assay demonstrate a significantly higher level of 2 to 6 fold in HBF compared to LBF. Biochemical and transcriptional analyses showed that chitinase activity and mRNA levels of chitinase genes, a marker of autolysis, were upregulated in 24 h biofilm formation by HBF compared to LBF, indicating autolysis pathway possibly involved in causing variation. The biofilm biomass and eDNA release by single (∆cht2, ∆cht3) and double knockout (∆cht2/∆cht3) chitinase mutants were significantly less compared to their parental strain CA14, confirming the role of chitinases in eDNA release and biofilm formation. Correlation analysis found a positive correlation between chitinases and HWP1, suggesting eDNA may release during the hyphal growth. Finally, we showed a combinational treatment of biofilms with DNase or chitinase inhibitor (acetazolamide) plus amphotericin B significantly improved antifungal susceptibility by 2 to 8 fold. Conclusions Collectively, these data show that eDNA release by C. albicans clinical isolates is variable and is associated with differential biofilm formation. Digestion of biofilm eDNA by DNase may provide a novel therapeutic strategies to destabilise biofilm growth and improves antifungal sensitivity. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0303-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ranjith Rajendran
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| | - Leighann Sherry
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| | - David F Lappin
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| | - Chris J Nile
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| | - Karen Smith
- Institute of Healthcare Associated Infection, School of Health, Nursing and Midwifery, University of the West of Scotland, Paisley, UK.
| | - Craig Williams
- Institute of Healthcare Associated Infection, School of Health, Nursing and Midwifery, University of the West of Scotland, Paisley, UK.
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| | - Gordon Ramage
- Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| |
Collapse
|
48
|
Abstract
Morphogenesis in fungi is often induced by extracellular factors and executed by fungal genetic factors. Cell surface changes and alterations of the microenvironment often accompany morphogenetic changes in fungi. In this review, we will first discuss the general traits of yeast and hyphal morphotypes and how morphogenesis affects development and adaptation by fungi to their native niches, including host niches. Then we will focus on the molecular machinery responsible for the two most fundamental growth forms, yeast and hyphae. Last, we will describe how fungi incorporate exogenous environmental and host signals together with genetic factors to determine their morphotype and how morphogenesis, in turn, shapes the fungal microenvironment.
Collapse
Affiliation(s)
- Xiaorong Lin
- Department of Biology, Texas A&M University, College Station, Texas 77843-3258
| | - J Andrew Alspaugh
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina 27710
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, California 92697
| | - Steven Harris
- Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| |
Collapse
|
49
|
Lindsay AK, Morales DK, Liu Z, Grahl N, Zhang A, Willger SD, Myers LC, Hogan DA. Analysis of Candida albicans mutants defective in the Cdk8 module of mediator reveal links between metabolism and biofilm formation. PLoS Genet 2014; 10:e1004567. [PMID: 25275466 PMCID: PMC4183431 DOI: 10.1371/journal.pgen.1004567] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/30/2014] [Indexed: 12/29/2022] Open
Abstract
Candida albicans biofilm formation is a key virulence trait that involves hyphal growth and adhesin expression. Pyocyanin (PYO), a phenazine secreted by Pseudomonas aeruginosa, inhibits both C. albicans biofilm formation and development of wrinkled colonies. Using a genetic screen, we identified two mutants, ssn3Δ/Δ and ssn8Δ/Δ, which continued to wrinkle in the presence of PYO. Ssn8 is a cyclin-like protein and Ssn3 is similar to cyclin-dependent kinases; both proteins are part of the heterotetrameric Cdk8 module that forms a complex with the transcriptional co-regulator, Mediator. Ssn3 kinase activity was also required for PYO sensitivity as a kinase dead mutant maintained a wrinkled colony morphology in the presence of PYO. Furthermore, similar phenotypes were observed in mutants lacking the other two components of the Cdk8 module-Srb8 and Srb9. Through metabolomics analyses and biochemical assays, we showed that a compromised Cdk8 module led to increases in glucose consumption, glycolysis-related transcripts, oxidative metabolism and ATP levels even in the presence of PYO. In the mutant, inhibition of respiration to levels comparable to the PYO-treated wild type inhibited wrinkled colony development. Several lines of evidence suggest that PYO does not act through Cdk8. Lastly, the ssn3 mutant was a hyperbiofilm former, and maintained higher biofilm formation in the presence of PYO than the wild type. Together these data provide novel insights into the role of the Cdk8 module of Mediator in regulation of C. albicans physiology and the links between respiratory activity and both wrinkled colony and biofilm development.
Collapse
Affiliation(s)
- Allia K. Lindsay
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Diana K. Morales
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Zhongle Liu
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Nora Grahl
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Anda Zhang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Sven D. Willger
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Lawrence C. Myers
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Deborah A. Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
50
|
Desai JV, Mitchell AP, Andes DR. Fungal biofilms, drug resistance, and recurrent infection. Cold Spring Harb Perspect Med 2014; 4:4/10/a019729. [PMID: 25274758 DOI: 10.1101/cshperspect.a019729] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
A biofilm is a surface-associated microbial community. Diverse fungi are capable of biofilm growth. The significance of this growth form for infection biology is that biofilm formation on implanted devices is a major cause of recurrent infection. Biofilms also have limited drug susceptibility, making device-associated infection extremely difficult to treat. Biofilm-like growth can occur during many kinds of infection, even when an implanted device is not present. Here we summarize the current understanding of fungal biofilm formation, its genetic control, and the basis for biofilm drug resistance.
Collapse
Affiliation(s)
- Jigar V Desai
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Aaron P Mitchell
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - David R Andes
- Department of Medicine, University of Wisconsin, Madison, Wisconsin 53705
| |
Collapse
|