201
|
van Wijlick L, Swidergall M, Brandt P, Ernst JF. Candida albicansresponds to glycostructure damage by Ace2-mediated feedback regulation of Cek1 signaling. Mol Microbiol 2016; 102:827-849. [DOI: 10.1111/mmi.13494] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Lasse van Wijlick
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
- Manchot Graduate School Molecules of Infection, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Marc Swidergall
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Philipp Brandt
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Joachim F. Ernst
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
- Manchot Graduate School Molecules of Infection, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| |
Collapse
|
202
|
Polvi EJ, Averette AF, Lee SC, Kim T, Bahn YS, Veri AO, Robbins N, Heitman J, Cowen LE. Metal Chelation as a Powerful Strategy to Probe Cellular Circuitry Governing Fungal Drug Resistance and Morphogenesis. PLoS Genet 2016; 12:e1006350. [PMID: 27695031 PMCID: PMC5047589 DOI: 10.1371/journal.pgen.1006350] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 09/08/2016] [Indexed: 01/01/2023] Open
Abstract
Fungal pathogens have evolved diverse strategies to sense host-relevant cues and coordinate cellular responses, which enable virulence and drug resistance. Defining circuitry controlling these traits opens new opportunities for chemical diversity in therapeutics, as the cognate inhibitors are rarely explored by conventional screening approaches. This has great potential to address the pressing need for new therapeutic strategies for invasive fungal infections, which have a staggering impact on human health. To explore this approach, we focused on a leading human fungal pathogen, Candida albicans, and screened 1,280 pharmacologically active compounds to identify those that potentiate the activity of echinocandins, which are front-line therapeutics that target fungal cell wall synthesis. We identified 19 compounds that enhance activity of the echinocandin caspofungin against an echinocandin-resistant clinical isolate, with the broad-spectrum chelator DTPA demonstrating the greatest synergistic activity. We found that DTPA increases susceptibility to echinocandins via chelation of magnesium. Whole genome sequencing of mutants resistant to the combination of DTPA and caspofungin identified mutations in the histidine kinase gene NIK1 that confer resistance to the combination. Functional analyses demonstrated that DTPA activates the mitogen-activated protein kinase Hog1, and that NIK1 mutations block Hog1 activation in response to both caspofungin and DTPA. The combination has therapeutic relevance as DTPA enhanced the efficacy of caspofungin in a mouse model of echinocandin-resistant candidiasis. We found that DTPA not only reduces drug resistance but also modulates morphogenesis, a key virulence trait that is normally regulated by environmental cues. DTPA induced filamentation via depletion of zinc, in a manner that is contingent upon Ras1-PKA signaling, as well as the transcription factors Brg1 and Rob1. Thus, we establish a new mechanism by which metal chelation modulates morphogenetic circuitry and echinocandin resistance, and illuminate a novel facet to metal homeostasis at the host-pathogen interface, with broad therapeutic potential. Invasive fungal infections pose a serious threat to human health worldwide, with Candida albicans being a leading fungal pathogen. Mortality is in part due to the limited arsenal of effective antifungals, with drug resistance on the rise. The echinocandins, which target the fungal cell wall, are the newest class of antifungal, and echinocandin resistance has already emerged. Here, we screened a library of 1,280 pharmacologically active compounds to identify those that potentiate echinocandin activity against an echinocandin-resistant isolate. The lead compound was a chelator, DTPA, which affects resistance by depleting magnesium. Genome sequencing of mutants resistant to the combination of DTPA and echinocandin revealed mutations in the gene encoding Nik1, which signals upstream of the Hog1 stress response pathway. We established that DTPA acts through Nik1 to modulate Hog1 signaling and enhance echinocandin activity, and that this combination has therapeutic benefits in a murine model of candidiasis. We also discovered that DTPA modulates C. albicans morphogenesis, a key virulence trait. DTPA induced filamentation by chelating zinc, in a manner that is contingent upon core filamentation pathways and specialized circuitry. Thus, we establish novel roles for metal homeostasis in C. albicans pathogenesis, thereby illuminating new therapeutic strategies for life-threatening infectious disease.
Collapse
Affiliation(s)
- Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Anna F. Averette
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Soo Chan Lee
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Taeyup Kim
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Yong-Sun Bahn
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
203
|
Su C, Lu Y, Liu H. N-acetylglucosamine sensing by a GCN5-related N-acetyltransferase induces transcription via chromatin histone acetylation in fungi. Nat Commun 2016; 7:12916. [PMID: 27694804 PMCID: PMC5063960 DOI: 10.1038/ncomms12916] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 08/16/2016] [Indexed: 12/11/2022] Open
Abstract
N-acetylglucosamine (GlcNAc) exists ubiquitously as a component of the surface on a wide range of cells, from bacteria to humans. Many fungi are able to utilize environmental GlcNAc to support growth and induce cellular development, a property important for their survival in various host niches. However, how the GlcNAc signal is sensed and subsequently transduced is largely unknown. Here, we identify a gene that is essential for GlcNAc signalling (NGS1) in Candida albicans, a commensal and pathogenic yeast of humans. Ngs1 can bind GlcNAc through the N-terminal β-N-acetylglucosaminidase homology domain. This binding activates N-acetyltransferase activity in the C-terminal GCN5-related N-acetyltransferase domain, which is required for GlcNAc-induced promoter histone acetylation and transcription. Ngs1 is targeted to the promoters of GlcNAc-inducible genes constitutively by the transcription factor Rep1. Ngs1 is conserved in diverse fungi that have GlcNAc catabolic genes. Thus, fungi use Ngs1 as a GlcNAc-sensor and transducer for GlcNAc-induced transcription. Many fungi are able to metabolise environmental N-acetylglucosamine, however the mechanism by which this molecule is sensed is unclear. Su and Lu et al. show that Candida albicans NGS1 fulfils this function by mediating N-acetylglucosamine-dependent histone acetylation at target genes.
Collapse
Affiliation(s)
- Chang Su
- Department of Biological Chemistry, University of California, Irvine, California 92697, USA
| | - Yang Lu
- Department of Biological Chemistry, University of California, Irvine, California 92697, USA
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, California 92697, USA
| |
Collapse
|
204
|
Robledo-Márquez K, Gutiérrez-Escobedo G, Yáñez-Carrillo P, Vidal-Aguiar Y, Briones-Martín-del-Campo M, Orta-Zavalza E, De Las Peñas A, Castaño I. Candida glabrataencodes a longer variant of the mating type (MAT) alpha2 gene in the mating type-likeMTL3locus, which can form homodimers. FEMS Yeast Res 2016; 16:fow082. [DOI: 10.1093/femsyr/fow082] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2016] [Indexed: 01/22/2023] Open
|
205
|
Dalal CK, Zuleta IA, Mitchell KF, Andes DR, El-Samad H, Johnson AD. Transcriptional rewiring over evolutionary timescales changes quantitative and qualitative properties of gene expression. eLife 2016; 5. [PMID: 27614020 PMCID: PMC5067116 DOI: 10.7554/elife.18981] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/05/2016] [Indexed: 12/25/2022] Open
Abstract
Evolutionary changes in transcription networks are an important source of diversity across species, yet the quantitative consequences of network evolution have rarely been studied. Here we consider the transcriptional 'rewiring' of the three GAL genes that encode the enzymes needed for cells to convert galactose to glucose. In Saccharomyces cerevisiae, the transcriptional regulator Gal4 binds and activates these genes. In the human pathogen Candida albicans (which last shared a common ancestor with S. cerevisiae some 300 million years ago), we show that different regulators, Rtg1 and Rtg3, activate the three GAL genes. Using single-cell dynamics and RNA-sequencing, we demonstrate that although the overall logic of regulation is the same in both species-the GAL genes are induced by galactose-there are major differences in both the quantitative response of these genes to galactose and in the position of these genes in the overall transcription network structure of the two species.
Collapse
Affiliation(s)
- Chiraj K Dalal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Ignacio A Zuleta
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, United States
| | - Kaitlin F Mitchell
- Department of Medicine, University of Wisconsin, Madison, United States.,Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, United States
| | - David R Andes
- Department of Medicine, University of Wisconsin, Madison, United States.,Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, United States
| | - Hana El-Samad
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, United States
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
206
|
Urrialde V, Prieto D, Pla J, Alonso-Monge R. The Candida albicans Pho4 Transcription Factor Mediates Susceptibility to Stress and Influences Fitness in a Mouse Commensalism Model. Front Microbiol 2016; 7:1062. [PMID: 27458452 PMCID: PMC4935684 DOI: 10.3389/fmicb.2016.01062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/24/2016] [Indexed: 12/17/2022] Open
Abstract
The Pho4 transcription factor is required for growth under low environmental phosphate concentrations in Saccharomyces cerevisiae. A characterization of Candida albicans pho4 mutants revealed that these cells are more susceptible to both osmotic and oxidative stress and that this effect is diminished in the presence of 5% CO2 or anaerobiosis, reflecting the relevance of oxygen metabolism in the Pho4-mediated response. A pho4 mutant was as virulent as wild type strain when assayed in the Galleria mellonella infection model and was even more resistant to murine macrophages in ex vivo killing assays. The lack of Pho4 neither impairs the ability to colonize the murine gut nor alters the localization in the gastrointestinal tract. However, we found that Pho4 influenced the colonization of C. albicans in the mouse gut in competition assays; pho4 mutants were unable to attain high colonization levels when inoculated simultaneously with an isogenic wild type strain. Moreover, pho4 mutants displayed a reduced adherence to the intestinal mucosa in a competitive ex vivo assays with wild type cells. In vitro competitive assays also revealed defects in fitness for this mutant compared to the wild type strain. Thus, Pho4, a transcription factor involved in phosphate metabolism, is required for adaptation to stress and fitness in C. albicans.
Collapse
Affiliation(s)
- Verónica Urrialde
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Daniel Prieto
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Jesús Pla
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Rebeca Alonso-Monge
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| |
Collapse
|
207
|
Ikeh MAC, Kastora SL, Day AM, Herrero-de-Dios CM, Tarrant E, Waldron KJ, Banks AP, Bain JM, Lydall D, Veal EA, MacCallum DM, Erwig LP, Brown AJP, Quinn J. Pho4 mediates phosphate acquisition in Candida albicans and is vital for stress resistance and metal homeostasis. Mol Biol Cell 2016; 27:2784-801. [PMID: 27385340 PMCID: PMC5007097 DOI: 10.1091/mbc.e16-05-0266] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/23/2016] [Indexed: 12/02/2022] Open
Abstract
This study provides the first evidence that the phosphate-responsive transcription factor Pho4 is vital for survival of Candida albicans to diverse and physiologically relevant stresses. Pho4 is important for C. albicans pathogenesis, and thus these findings illustrate how metabolic adaptation promotes C. albicans survival in the host. During interactions with its mammalian host, the pathogenic yeast Candida albicans is exposed to a range of stresses such as superoxide radicals and cationic fluxes. Unexpectedly, a nonbiased screen of transcription factor deletion mutants revealed that the phosphate-responsive transcription factor Pho4 is vital for the resistance of C. albicans to these diverse stresses. RNA-Seq analysis indicated that Pho4 does not induce stress-protective genes directly. Instead, we show that loss of Pho4 affects metal cation toxicity, accumulation, and bioavailability. We demonstrate that pho4Δ cells are sensitive to metal and nonmetal cations and that Pho4-mediated polyphosphate synthesis mediates manganese resistance. Significantly, we show that Pho4 is important for mediating copper bioavailability to support the activity of the copper/zinc superoxide dismutase Sod1 and that loss of Sod1 activity contributes to the superoxide sensitivity of pho4Δ cells. Consistent with the key role of fungal stress responses in countering host phagocytic defenses, we also report that C. albicans pho4Δ cells are acutely sensitive to macrophage-mediated killing and display attenuated virulence in animal infection models. The novel connections between phosphate metabolism, metal homeostasis, and superoxide stress resistance presented in this study highlight the importance of metabolic adaptation in promoting C. albicans survival in the host.
Collapse
Affiliation(s)
- Mélanie A C Ikeh
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Stavroula L Kastora
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Alison M Day
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | - Emma Tarrant
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Kevin J Waldron
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - A Peter Banks
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Judith M Bain
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - David Lydall
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Elizabeth A Veal
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Donna M MacCallum
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Lars P Erwig
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Alistair J P Brown
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Janet Quinn
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| |
Collapse
|
208
|
Guo L, Zhao G, Xu J, Kistler HC, Gao L, Ma L. Compartmentalized gene regulatory network of the pathogenic fungus Fusarium graminearum. THE NEW PHYTOLOGIST 2016; 211:527-41. [PMID: 26990214 PMCID: PMC5069591 DOI: 10.1111/nph.13912] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/25/2016] [Indexed: 05/09/2023]
Abstract
Head blight caused by Fusarium graminearum threatens world-wide wheat production, resulting in both yield loss and mycotoxin contamination. We reconstructed the global F. graminearum gene regulatory network (GRN) from a large collection of transcriptomic data using Bayesian network inference, a machine-learning algorithm. This GRN reveals connectivity between key regulators and their target genes. Focusing on key regulators, this network contains eight distinct but interwoven modules. Enriched for unique functions, such as cell cycle, DNA replication, transcription, translation and stress responses, each module exhibits distinct expression profiles. Evolutionarily, the F. graminearum genome can be divided into core regions shared with closely related species and variable regions harboring genes that are unique to F. graminearum and perform species-specific functions. Interestingly, the inferred top regulators regulate genes that are significantly enriched from the same genomic regions (P < 0.05), revealing a compartmentalized network structure that may reflect network rewiring related to specific adaptation of this plant pathogen. This first-ever reconstructed filamentous fungal GRN primes our understanding of pathogenicity at the systems biology level and provides enticing prospects for novel disease control strategies involving the targeting of master regulators in pathogens. The program can be used to construct GRNs of other plant pathogens.
Collapse
Affiliation(s)
- Li Guo
- Department of Biochemistry and Molecular BiologyUniversity of Massachusetts AmherstAmherstMA01003USA
| | - Guoyi Zhao
- Department of Electrical & Computer EngineeringUniversity of Massachusetts AmherstAmherstMA01003USA
| | - Jin‐Rong Xu
- Department of Botany and Plant PathologyPurdue UniversityWest LafayetteIN47907USA
| | - H. Corby Kistler
- USDA‐ARSCereal Disease LaboratoryUniversity of MinnesotaSt PaulMN55108USA
| | - Lixin Gao
- Department of Electrical & Computer EngineeringUniversity of Massachusetts AmherstAmherstMA01003USA
| | - Li‐Jun Ma
- Department of Biochemistry and Molecular BiologyUniversity of Massachusetts AmherstAmherstMA01003USA
| |
Collapse
|
209
|
Saputo S, Norman KL, Murante T, Horton BN, Diaz JDLC, DiDone L, Colquhoun J, Schroeder JW, Simmons LA, Kumar A, Krysan DJ. Complex Haploinsufficiency-Based Genetic Analysis of the NDR/Lats Kinase Cbk1 Provides Insight into Its Multiple Functions in Candida albicans. Genetics 2016; 203:1217-33. [PMID: 27206715 PMCID: PMC4937472 DOI: 10.1534/genetics.116.188029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/11/2016] [Indexed: 01/11/2023] Open
Abstract
Although the analysis of genetic interactions and networks is a powerful approach to understanding biology, it has not been applied widely to the pathogenic yeast Candida albicans Here, we describe the use of both screening and directed genetic interaction studies based on complex haploinsufficiency to probe the function of the R: egulation of A: ce2 and M: orphogenesis (RAM) pathway in C. albicans A library of 5200 Tn7-mutagenized derivatives of a parental strain heterozygous at CBK1, the key kinase in the RAM pathway, was screened for alterations in serum-induced filamentation. Following confirmation of phenotypes and identification of insertion sites by sequencing, a set of 36 unique double heterozygous strains showing complex haploinsufficiency was obtained. In addition to a large set of genes regulated by the RAM transcription factor Ace2, genes related to cell wall biosynthesis, cell cycle, polarity, oxidative stress, and nitrogen utilization were identified. Follow-up analysis led to the first demonstration that the RAM pathway is required for oxidative stress tolerance in a manner related to the two-component-regulated kinase Chk1 and revealed a potential direct connection between the RAM pathway and the essential Mps1 spindle pole-related kinase. In addition, genetic interactions with CDC42-related genes MSB1, a putative scaffold protein, and RGD3, a putative Rho GTPase-activating protein (GAP) were identified. We also provide evidence that Rgd3 is a GAP for Cdc42 and show that its localization and phosphorylation are dependent on Cbk1.
Collapse
Affiliation(s)
- Sarah Saputo
- Department of Microbiology/Immunology University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Kaitlyn L Norman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas Murante
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Brooke N Horton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Jacinto De La Cruz Diaz
- Department of Microbiology/Immunology University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Louis DiDone
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jennifer Colquhoun
- Department of Microbiology/Immunology University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jeremy W Schroeder
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Lyle A Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Anuj Kumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Damian J Krysan
- Department of Microbiology/Immunology University of Rochester School of Medicine and Dentistry, Rochester, New York 14642 Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
210
|
The roles of zinc and copper sensing in fungal pathogenesis. Curr Opin Microbiol 2016; 32:128-134. [PMID: 27327380 PMCID: PMC4992176 DOI: 10.1016/j.mib.2016.05.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/13/2016] [Accepted: 05/19/2016] [Indexed: 12/17/2022]
Abstract
Zinc and copper are essential trace elements required for cell function. Nutrient Immunity restricts zinc and copper access and mediates toxicity. Divergent fungi integrate zinc and copper responsive regulons for pathogenesis.
All organisms must secure essential trace nutrients, including iron, zinc, manganese and copper for survival and proliferation. However, these very nutrients are also highly toxic if present at elevated levels. Mammalian immunity has harnessed both the essentiality and toxicity of micronutrients to defend against microbial invasion — processes known collectively as ‘nutritional immunity’. Therefore, pathogenic microbes must possess highly effective micronutrient assimilation and detoxification mechanisms to survive and proliferate within the infected host. In this review we compare and contrast the micronutrient homeostatic mechanisms of Cryptococcus and Candida — yeasts which, despite ancient evolutionary divergence, account for over a million life-threatening infections per year. We focus on two emerging arenas within the host–pathogen battle for essential trace metals: adaptive responses to zinc limitation and copper availability.
Collapse
|
211
|
Systematic Genetic Screen for Transcriptional Regulators of the Candida albicans White-Opaque Switch. Genetics 2016; 203:1679-92. [PMID: 27280690 DOI: 10.1534/genetics.116.190645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/23/2016] [Indexed: 02/04/2023] Open
Abstract
The human fungal pathogen Candida albicans can reversibly switch between two cell types named "white" and "opaque," each of which is stable through many cell divisions. These two cell types differ in their ability to mate, their metabolic preferences and their interactions with the mammalian innate immune system. A highly interconnected network of eight transcriptional regulators has been shown to control switching between these two cell types. To identify additional regulators of the switch, we systematically and quantitatively measured white-opaque switching rates of 196 strains, each deleted for a specific transcriptional regulator. We identified 19 new regulators with at least a 10-fold effect on switching rates and an additional 14 new regulators with more subtle effects. To investigate how these regulators affect switching rates, we examined several criteria, including the binding of the eight known regulators of switching to the control region of each new regulatory gene, differential expression of the newly found genes between cell types, and the growth rate of each mutant strain. This study highlights the complexity of the transcriptional network that regulates the white-opaque switch and the extent to which switching is linked to a variety of metabolic processes, including respiration and carbon utilization. In addition to revealing specific insights, the information reported here provides a foundation to understand the highly complex coupling of white-opaque switching to cellular physiology.
Collapse
|
212
|
O’Meara TR, Veri AO, Polvi EJ, Li X, Valaei SF, Diezmann S, Cowen LE. Mapping the Hsp90 Genetic Network Reveals Ergosterol Biosynthesis and Phosphatidylinositol-4-Kinase Signaling as Core Circuitry Governing Cellular Stress. PLoS Genet 2016; 12:e1006142. [PMID: 27341673 PMCID: PMC4920384 DOI: 10.1371/journal.pgen.1006142] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/27/2022] Open
Abstract
Candida albicans is a leading human fungal pathogen that causes life-threatening systemic infections. A key regulator of C. albicans stress response, drug resistance, morphogenesis, and virulence is the molecular chaperone Hsp90. Targeting Hsp90 provides a powerful strategy to treat fungal infections, however, the therapeutic utility of current inhibitors is compromised by toxicity due to inhibition of host Hsp90. To identify components of the Hsp90-dependent circuitry governing virulence and drug resistance that are sufficiently divergent for selective targeting in the pathogen, we pioneered chemical genomic profiling of the Hsp90 genetic network in C. albicans. Here, we screen mutant collections covering ~10% of the genome for hypersensitivity to Hsp90 inhibition in multiple environmental conditions. We identify 158 HSP90 chemical genetic interactors, most of which are important for growth only in specific environments. We discovered that the sterol C-22 desaturase gene ERG5 and the phosphatidylinositol-4-kinase (PI4K) gene STT4 are HSP90 genetic interactors under multiple conditions, suggesting a function upstream of Hsp90. By systematic analysis of the ergosterol biosynthetic cascade, we demonstrate that defects in ergosterol biosynthesis induce cellular stress that overwhelms Hsp90's functional capacity. By analysis of the phosphatidylinositol pathway, we demonstrate that there is a genetic interaction between the PI4K Stt4 and Hsp90. We also establish that Stt4 is required for normal actin polarization through regulation of Wal1, and suggest a model in which defects in actin remodeling induces stress that creates a cellular demand for Hsp90 that exceeds its functional capacity. Consistent with this model, actin inhibitors are synergistic with Hsp90 inhibitors. We highlight new connections between Hsp90 and virulence traits, demonstrating that Erg5 and Stt4 enable activation of macrophage pyroptosis. This work uncovers novel circuitry regulating Hsp90 functional capacity and new effectors governing drug resistance, morphogenesis and virulence, revealing new targets for antifungal drug development.
Collapse
Affiliation(s)
- Teresa R. O’Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O. Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J. Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Xinliu Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephanie Diezmann
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Claverton Down, Bath, United Kingdom
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
213
|
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: 15] [Impact Index Per Article: 1.7] [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
|
214
|
Koselny K, Green J, Favazzo L, Glazier V, DiDone L, Ransford S, Krysan DJ. Antitumor/Antifungal Celecoxib Derivative AR-12 is a Non-Nucleoside Inhibitor of the ANL-Family Adenylating Enzyme Acetyl CoA Synthetase. ACS Infect Dis 2016; 2:268-280. [PMID: 27088128 PMCID: PMC4828684 DOI: 10.1021/acsinfecdis.5b00134] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Indexed: 02/08/2023]
Abstract
AR-12/OSU-03012 is an antitumor celecoxib-derivative that has progressed to Phase I clinical trial as an anticancer agent and has activity against a number of infectious agents including fungi, bacteria and viruses. However, the mechanism of these activities has remained unclear. Based on a chemical-genetic profiling approach in yeast, we have found that AR-12 is an ATP-competitive, time-dependent inhibitor of yeast acetyl coenzyme A synthetase. AR-12-treated fungal cells show phenotypes consistent with the genetic reduction of acetyl CoA synthetase activity, including induction of autophagy, decreased histone acetylation, and loss of cellular integrity. In addition, AR-12 is a weak inhibitor of human acetyl CoA synthetase ACCS2. Acetyl CoA synthetase activity is essential in many fungi and parasites. In contrast, acetyl CoA is primarily synthesized by an alternate enzyme, ATP-citrate lyase, in mammalian cells. Taken together, our results indicate that AR-12 is a non-nucleoside acetyl CoA synthetase inhibitor and that acetyl CoA synthetase may be a feasible antifungal drug target.
Collapse
Affiliation(s)
- Kristy Koselny
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Julianne Green
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Lacey Favazzo
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Virginia
E. Glazier
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Louis DiDone
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Shea Ransford
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Damian J. Krysan
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| |
Collapse
|
215
|
Simões J, Bezerra AR, Moura GR, Araújo H, Gut I, Bayes M, Santos MAS. The Fungus Candida albicans Tolerates Ambiguity at Multiple Codons. Front Microbiol 2016; 7:401. [PMID: 27065968 PMCID: PMC4814463 DOI: 10.3389/fmicb.2016.00401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 12/31/2022] Open
Abstract
The ascomycete Candida albicans is a normal resident of the gastrointestinal tract of humans and other warm-blooded animals. It occurs in a broad range of body sites and has high capacity to survive and proliferate in adverse environments with drastic changes in oxygen, carbon dioxide, pH, osmolarity, nutrients, and temperature. Its biology is unique due to flexible reassignment of the leucine CUG codon to serine and synthesis of statistical proteins. Under standard growth conditions, CUG sites incorporate leucine (3% of the times) and serine (97% of the times) on a proteome wide scale, but leucine incorporation fluctuates in response to environmental stressors and can be artificially increased up to 98%. In order to determine whether such flexibility also exists at other codons, we have constructed several serine tRNAs that decode various non-cognate codons. Expression of these tRNAs had minor effects on fitness, but growth of the mistranslating strains at different temperatures, in medium with different pH and nutrients composition was often enhanced relatively to the wild type (WT) strain, supporting our previous data on adaptive roles of CUG ambiguity in variable growth conditions. Parallel evolution of the recombinant strains (100 generations) followed by full genome resequencing identified various strain specific single nucleotide polymorphisms (SNP) and one SNP in the deneddylase (JAB1) gene in all strains. Since JAB1 is a subunit of the COP9 signalosome complex, which interacts with cullin (Cdc53p) to mediate degradation of a variety of cellular proteins, our data suggest that neddylation plays a key role in tolerance and adaptation to codon ambiguity in C. albicans.
Collapse
Affiliation(s)
- João Simões
- Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro Aveiro, Portugal
| | - Ana R Bezerra
- Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro Aveiro, Portugal
| | - Gabriela R Moura
- Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro Aveiro, Portugal
| | - Hugo Araújo
- Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro Aveiro, Portugal
| | - Ivo Gut
- Centro Nacional de Análises Genómico, Parc Científic Barcelona, Spain
| | - Mónica Bayes
- Centro Nacional de Análises Genómico, Parc Científic Barcelona, Spain
| | - Manuel A S Santos
- Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro Aveiro, Portugal
| |
Collapse
|
216
|
Mechanisms Underlying the Delayed Activation of the Cap1 Transcription Factor in Candida albicans following Combinatorial Oxidative and Cationic Stress Important for Phagocytic Potency. mBio 2016; 7:e00331. [PMID: 27025253 PMCID: PMC4817257 DOI: 10.1128/mbio.00331-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Following phagocytosis, microbes are exposed to an array of antimicrobial weapons that include reactive oxygen species (ROS) and cationic fluxes. This is significant as combinations of oxidative and cationic stresses are much more potent than the corresponding single stresses, triggering the synergistic killing of the fungal pathogen Candida albicans by “stress pathway interference.” Previously we demonstrated that combinatorial oxidative plus cationic stress triggers a dramatic increase in intracellular ROS levels compared to oxidative stress alone. Here we show that activation of Cap1, the major regulator of antioxidant gene expression in C. albicans, is significantly delayed in response to combinatorial stress treatments and to high levels of H2O2. Cap1 is normally oxidized in response to H2O2; this masks the nuclear export sequence, resulting in the rapid nuclear accumulation of Cap1 and the induction of Cap1-dependent genes. Here we demonstrate that following exposure of cells to combinatorial stress or to high levels of H2O2, Cap1 becomes trapped in a partially oxidized form, Cap1OX-1. Notably, Cap1-dependent gene expression is not induced when Cap1 is in this partially oxidized form. However, while Cap1OX-1 readily accumulates in the nucleus and binds to target genes following high-H2O2 stress, the nuclear accumulation of Cap1OX-1 following combinatorial H2O2 and NaCl stress is delayed due to a cationic stress-enhanced interaction with the Crm1 nuclear export factor. These findings define novel mechanisms that delay activation of the Cap1 transcription factor, thus preventing the rapid activation of the stress responses vital for the survival of C. albicans within the host. Combinatorial stress-mediated synergistic killing represents a new unchartered area in the field of stress signaling. This phenomenon contrasts starkly with “stress cross-protection,” where exposure to one stress protects against subsequent exposure to a different stress. Previously we demonstrated that the pathogen Candida albicans is acutely sensitive to combinations of cationic and oxidative stresses, because the induction of H2O2-responsive genes is blocked in the presence of cationic stress. We reveal that this is due to novel mechanisms that delay activation of the Cap1 AP-1-like transcription factor, the major regulator of the H2O2-induced regulon. Cap1 becomes trapped in a partially oxidized form following simultaneous exposure to oxidative and cationic stresses. In addition, cationic stress promotes the interaction of Cap1 with the Crm1 nuclear export factor, thus inhibiting its nuclear accumulation. These mechanisms probably explain the potency of neutrophils, which employ multiple stresses to kill fungal pathogens.
Collapse
|
217
|
Böhm L, Muralidhara P, Pérez JC. ACandida albicansregulator of disseminated infection operates primarily as a repressor and governs cell surface remodeling. Mol Microbiol 2016; 100:328-44. [DOI: 10.1111/mmi.13320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Lena Böhm
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg; Würzburg Germany
- Institute for Molecular Infection Biology, University of Würzburg; Würzburg Germany
| | | | - J. Christian Pérez
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg; Würzburg Germany
- Institute for Molecular Infection Biology, University of Würzburg; Würzburg Germany
| |
Collapse
|
218
|
Ssn6 Defines a New Level of Regulation of White-Opaque Switching in Candida albicans and Is Required For the Stochasticity of the Switch. mBio 2016; 7:e01565-15. [PMID: 26814177 PMCID: PMC4742700 DOI: 10.1128/mbio.01565-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The human commensal and opportunistic pathogen Candida albicans can switch between two distinct, heritable cell types, named “white” and “opaque,” which differ in morphology, mating abilities, and metabolic preferences and in their interactions with the host immune system. Previous studies revealed a highly interconnected group of transcriptional regulators that control switching between the two cell types. Here, we identify Ssn6, the C. albicans functional homolog of the Saccharomyces cerevisiae transcriptional corepressor Cyc8, as a new regulator of white-opaque switching. In a or α mating type strains, deletion of SSN6 results in mass switching from the white to the opaque cell type. Transcriptional profiling of ssn6 deletion mutant strains reveals that Ssn6 represses part of the opaque cell transcriptional program in white cells and the majority of the white cell transcriptional program in opaque cells. Genome-wide chromatin immunoprecipitation experiments demonstrate that Ssn6 is tightly integrated into the opaque cell regulatory circuit and that the positions to which it is bound across the genome strongly overlap those bound by Wor1 and Wor2, previously identified regulators of white-opaque switching. This work reveals the next layer in the white-opaque transcriptional circuitry by integrating a transcriptional regulator that does not bind DNA directly but instead associates with specific combinations of DNA-bound transcriptional regulators. The most common fungal pathogen of humans, C. albicans, undergoes several distinct morphological transitions during interactions with its host. One such transition, between cell types named “white” and “opaque,” is regulated in an epigenetic manner, in the sense that changes in gene expression are heritably maintained without any modification of the primary genomic DNA sequence. Prior studies revealed a highly interconnected network of sequence-specific DNA-binding proteins that control this switch. We report the identification of Ssn6, which defines an additional layer of transcriptional regulation that is critical for this heritable switch. Ssn6 is necessary to maintain the white cell type and to properly express the opaque cell transcriptional program. Ssn6 does not bind DNA directly but rather associates with specific combinations of DNA-bound transcriptional regulators to control the switch. This work is significant because it reveals a new level of regulation of an important epigenetic switch in the predominant fungal pathogen of humans.
Collapse
|
219
|
Felice MR, Gulati M, Giuffrè L, Giosa D, Di Bella LM, Criseo G, Nobile CJ, Romeo O, Scordino F. Molecular Characterization of the N-Acetylglucosamine Catabolic Genes in Candida africana, a Natural N-Acetylglucosamine Kinase (HXK1) Mutant. PLoS One 2016; 11:e0147902. [PMID: 26808192 PMCID: PMC4726466 DOI: 10.1371/journal.pone.0147902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/11/2016] [Indexed: 01/09/2023] Open
Abstract
Background In this study we report the genetic characterization, including expression analysis, of the genes involved in the uptake (NGT1) and catabolism (HXK1/NAG5, DAC1/NAG2, NAG1) of the aminosugar N-acetylglucosamine (GlcNAc) in Candida africana, a pathogenic biovariant of Candida albicans that is naturally unable to assimilate the GlcNAc. Results DNA sequence analysis of these genes revealed a number of characteristic nucleotide substitutions including a unique and distinctive guanine insertion that shifts the reading frame and generates a premature stop codon (TGA) 154 bp downstream of the ATG start codon of the HXK1 gene encoding the GlcNAc-kinase, a key enzyme of the GlcNAc catabolic pathway. However, all examined genes produced transcripts even though different levels of expression were observed among the Candida isolates examined. In particular, we found an HXK1-idependent relationship of the NGT1 gene and a considerable influence of the GlcNAc-kinase functionality on the transcription of the DAC1 and NAG1 genes. Additional phenotypic analysis revealed that C. africana isolates are hyperfilamentous in the first 24-48h of growth on filament-inducing media and revert to the yeast morphological form after 72h of incubation on these media. Conclusions Our results show that C. africana is a natural HXK1 mutant, displaying a number of phenotypic characteristics distinct from typical C. albicans isolates.
Collapse
Affiliation(s)
- Maria Rosa Felice
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Megha Gulati
- Department of Molecular and Cell Biology, University of California Merced, Merced, California, United States of America
| | - Letterio Giuffrè
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Domenico Giosa
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Luca Marco Di Bella
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Giuseppe Criseo
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, University of California Merced, Merced, California, United States of America
| | - Orazio Romeo
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
- Scientific Institute for Research, Hospitalization and Health Care (IRCCS)—Centro Neurolesi "Bonino-Pulejo", Messina, Italy
- * E-mail:
| | - Fabio Scordino
- Scientific Institute for Research, Hospitalization and Health Care (IRCCS)—Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| |
Collapse
|
220
|
Nocedal I, Johnson AD. How Transcription Networks Evolve and Produce Biological Novelty. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2015; 80:265-74. [PMID: 26657905 DOI: 10.1101/sqb.2015.80.027557] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The rewiring of gene regulatory networks over evolutionary timescales produces changes in the patterns of gene expression and is a major source of diversity among species. Yet the molecular mechanisms underlying evolutionary rewiring are only beginning to be understood. Here, we discuss recent analyses in ascomycete yeasts that have revealed several general principles of network rewiring. Specifically, we discuss how transcription networks can maintain a functional output despite changes in mechanism, how specific types of constraints alter available evolutionary trajectories, and how regulatory rewiring can ultimately lead to phenotypic novelty. We also argue that the structure and "logic" of extant gene regulatory networks can largely be accounted for by constraints that shape their evolutionary trajectories.
Collapse
Affiliation(s)
- Isabel Nocedal
- Departments of Microbiology and Immunology and of Biochemistry and Biophysics, University of California, San Francisco, California 94158
| | - Alexander D Johnson
- Departments of Microbiology and Immunology and of Biochemistry and Biophysics, University of California, San Francisco, California 94158
| |
Collapse
|
221
|
Ghosh AK, Wangsanut T, Fonzi WA, Rolfes RJ. The GRF10 homeobox gene regulates filamentous growth in the human fungal pathogen Candida albicans. FEMS Yeast Res 2015; 15:fov093. [PMID: 26472755 PMCID: PMC4705307 DOI: 10.1093/femsyr/fov093] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/14/2015] [Accepted: 10/07/2015] [Indexed: 12/14/2022] Open
Abstract
Candida albicans is the most common human fungal pathogen and can cause life-threatening infections. Filamentous growth is critical in the pathogenicity of C. albicans, as the transition from yeast to hyphal forms is linked to virulence and is also a pivotal process in fungal biofilm development. Homeodomain-containing transcription factors have been linked to developmental processes in fungi and other eukaryotes. We report here on GRF10, a homeobox transcription factor-encoding gene that plays a role in C. albicans filamentation. Deletion of the GRF10 gene, in both C. albicans SN152 and BWP17 strain backgrounds, results in mutants with strongly decreased hyphal growth. The mutants are defective in chlamydospore and biofilm formation, as well as showing dramatically attenuated virulence in a mouse infection model. Expression of the GRF10 gene is highly induced during stationary phase and filamentation. In summary, our study emphasizes a new role for the homeodomain-containing transcription factor in morphogenesis and pathogenicity of C. albicans.
Collapse
Affiliation(s)
- Anup K Ghosh
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | | | - William A Fonzi
- Department of Microbiology and Immunology, Georgetown University, Washington, DC 20057, USA
| | - Ronda J Rolfes
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
222
|
Kim SH, Clark ST, Surendra A, Copeland JK, Wang PW, Ammar R, Collins C, Tullis DE, Nislow C, Hwang DM, Guttman DS, Cowen LE. Global Analysis of the Fungal Microbiome in Cystic Fibrosis Patients Reveals Loss of Function of the Transcriptional Repressor Nrg1 as a Mechanism of Pathogen Adaptation. PLoS Pathog 2015; 11:e1005308. [PMID: 26588216 PMCID: PMC4654494 DOI: 10.1371/journal.ppat.1005308] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/03/2015] [Indexed: 01/10/2023] Open
Abstract
The microbiome shapes diverse facets of human biology and disease, with the importance of fungi only beginning to be appreciated. Microbial communities infiltrate diverse anatomical sites as with the respiratory tract of healthy humans and those with diseases such as cystic fibrosis, where chronic colonization and infection lead to clinical decline. Although fungi are frequently recovered from cystic fibrosis patient sputum samples and have been associated with deterioration of lung function, understanding of species and population dynamics remains in its infancy. Here, we coupled high-throughput sequencing of the ribosomal RNA internal transcribed spacer 1 (ITS1) with phenotypic and genotypic analyses of fungi from 89 sputum samples from 28 cystic fibrosis patients. Fungal communities defined by sequencing were concordant with those defined by culture-based analyses of 1,603 isolates from the same samples. Different patients harbored distinct fungal communities. There were detectable trends, however, including colonization with Candida and Aspergillus species, which was not perturbed by clinical exacerbation or treatment. We identified considerable inter- and intra-species phenotypic variation in traits important for host adaptation, including antifungal drug resistance and morphogenesis. While variation in drug resistance was largely between species, striking variation in morphogenesis emerged within Candida species. Filamentation was uncoupled from inducing cues in 28 Candida isolates recovered from six patients. The filamentous isolates were resistant to the filamentation-repressive effects of Pseudomonas aeruginosa, implicating inter-kingdom interactions as the selective force. Genome sequencing revealed that all but one of the filamentous isolates harbored mutations in the transcriptional repressor NRG1; such mutations were necessary and sufficient for the filamentous phenotype. Six independent nrg1 mutations arose in Candida isolates from different patients, providing a poignant example of parallel evolution. Together, this combined clinical-genomic approach provides a high-resolution portrait of the fungal microbiome of cystic fibrosis patient lungs and identifies a genetic basis of pathogen adaptation. Microbial cells vastly outnumber human cells in our bodies, yet we are only beginning to understand how these microbes influence human health and disease. One disease for which microbial communities are especially important is cystic fibrosis, where persistent lung infections can be lethal. Fungi are associated with poor respiratory function, but how fungal communities change with disease progression or treatment remains enigmatic. Here, we assess the dynamics of fungal communities by combining high-throughput sequencing of sputum samples from 28 patients with detailed analysis of phenotypes and genotypes of 1,603 fungal isolates. We found stable communities dominated by Candida and Aspergillus, and diversity in traits important for host adaptation. Antifungal drug resistance varied largely between species, while morphogenesis varied within species. For Candida species, the capacity to transition between yeast and filaments is a key virulence trait that is normally regulated by inducing cues, however, 28 isolates grew as filaments without such cues. Filamentation was due to loss-of-function mutations in the transcriptional regulator NRG1 in most isolates, which conferred resistance to the filament-repressive effects of a common bacterial pathogen. This work provides a portrait of the fungal microbiome associated with a lethal disease, and illuminates a genetic basis of pathogen adaptation.
Collapse
Affiliation(s)
- Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Shawn T. Clark
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Anuradha Surendra
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Julia K. Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Pauline W. Wang
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ron Ammar
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Cathy Collins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Corey Nislow
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - David M. Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David S. Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
223
|
Zhang Q, Tao L, Guan G, Yue H, Liang W, Cao C, Dai Y, Huang G. Regulation of filamentation in the human fungal pathogenCandida tropicalis. Mol Microbiol 2015; 99:528-45. [DOI: 10.1111/mmi.13247] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Qiuyu Zhang
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
- University of Chinese Academy of Sciences; Beijing China
| | - Li Tao
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
| | - Guobo Guan
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
| | - Huizhen Yue
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
- University of Chinese Academy of Sciences; Beijing China
| | - Weihong Liang
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
- University of Chinese Academy of Sciences; Beijing China
| | - Chengjun Cao
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
- University of Chinese Academy of Sciences; Beijing China
| | - Yu Dai
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
| | - Guanghua Huang
- State Key Laboratory of Mycology; Institute of Microbiology; Chinese Academy of Sciences; Beijing China
| |
Collapse
|
224
|
Fedorovych D, Boretsky Y, Bobak Y, Prokopiv T, Sybirny A. Putative ferroxidases in the Flavinogenic yeast Pichia guilliermondii are regulated by iron acquisition. CYTOL GENET+ 2015. [DOI: 10.3103/s0095452715050035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
225
|
Abstract
Candida species are the most prevalent human fungal pathogens, with Candida albicans being the most clinically relevant species. Candida albicans resides as a commensal of the human gastrointestinal tract but is a frequent cause of opportunistic mucosal and systemic infections. Investigation of C. albicans virulence has traditionally relied on candidate gene approaches, but recent advances in functional genomics have now facilitated global, unbiased studies of gene function. Such studies include comparative genomics (both between and within Candida species), analysis of total RNA expression, and regulation and delineation of protein-DNA interactions. Additionally, large collections of mutant strains have begun to aid systematic screening of clinically relevant phenotypes. Here, we will highlight the development of functional genomics in C. albicans and discuss the use of these approaches to addressing both commensalism and pathogenesis in this species.
Collapse
|
226
|
In Vitro Activity of Miltefosine against Candida albicans under Planktonic and Biofilm Growth Conditions and In Vivo Efficacy in a Murine Model of Oral Candidiasis. Antimicrob Agents Chemother 2015; 59:7611-20. [PMID: 26416861 DOI: 10.1128/aac.01890-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
The generation of a new antifungal against Candida albicans biofilms has become a major priority, since biofilm formation by this opportunistic pathogenic fungus is usually associated with an increased resistance to azole antifungal drugs and treatment failures. Miltefosine is an alkyl phospholipid with promising antifungal activity. Here, we report that, when tested under planktonic conditions, miltefosine displays potent in vitro activity against multiple fluconazole-susceptible and -resistant C. albicans clinical isolates, including isolates overexpressing efflux pumps and/or with well-characterized Erg11 mutations. Moreover, miltefosine inhibits C. albicans biofilm formation and displays activity against preformed biofilms. Serial passage experiments confirmed that miltefosine has a reduced potential to elicit resistance, and screening of a library of C. albicans transcription factor mutants provided additional insight into the activity of miltefosine against C. albicans growing under planktonic and biofilm conditions. Finally, we demonstrate the in vivo efficacy of topical treatment with miltefosine in the murine model of oropharyngeal candidiasis. Overall, our results confirm the potential of miltefosine as a promising antifungal drug candidate, in particular for the treatment of azole-resistant and biofilm-associated superficial candidiasis.
Collapse
|
227
|
Sarda S, Hannenhalli S. High-Throughput Identification of Cis-Regulatory Rewiring Events in Yeast. Mol Biol Evol 2015; 32:3047-63. [PMID: 26399482 DOI: 10.1093/molbev/msv203] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A coregulated module of genes ("regulon") can have evolutionarily conserved expression patterns and yet have diverged upstream regulators across species. For instance, the ribosomal genes regulon is regulated by the transcription factor (TF) TBF1 in Candida albicans, while in Saccharomyces cerevisiae it is regulated by RAP1. Only a handful of such rewiring events have been established, and the prevalence or conditions conducive to such events are not well known. Here, we develop a novel probabilistic scoring method to comprehensively screen for regulatory rewiring within regulons across 23 yeast species. Investigation of 1,713 regulons and 176 TFs yielded 5,353 significant rewiring events at 5% false discovery rate (FDR). Besides successfully recapitulating known rewiring events, our analyses also suggest TF candidates for certain processes reported to be under distinct regulatory controls in S. cerevisiae and C. albicans, for which the implied regulators are not known: 1) Oxidative stress response (Sc-MSN2 to Ca-FKH2) and 2) nutrient modulation (Sc-RTG1 to Ca-GCN4/Ca-UME6). Furthermore, a stringent screen to detect TF rewiring at individual genes identified 1,446 events at 10% FDR. Overall, these events are supported by strong coexpression between the predicted regulator and its target gene(s) in a species-specific fashion (>50-fold). Independent functional analyses of rewiring TF pairs revealed greater functional interactions and shared biological processes between them (P = 1 × 10(-3)).Our study represents the first comprehensive assessment of regulatory rewiring; with a novel approach that has generated a unique high-confidence resource of several specific events, suggesting that evolutionary rewiring is relatively frequent and may be a significant mechanism of regulatory innovation.
Collapse
Affiliation(s)
- Shrutii Sarda
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park
| |
Collapse
|
228
|
N-Acetylglucosamine-Induced Cell Death in Candida albicans and Its Implications for Adaptive Mechanisms of Nutrient Sensing in Yeasts. mBio 2015; 6:e01376-15. [PMID: 26350972 PMCID: PMC4600118 DOI: 10.1128/mbio.01376-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Single-celled organisms have different strategies to sense and utilize nutrients in their ever-changing environments. The opportunistic fungal pathogen Candida albicans is a common member of the human microbiota, especially that of the gastrointestinal (GI) tract. An important question concerns how C. albicans gained a competitive advantage over other microbes to become a successful commensal and opportunistic pathogen. Here, we report that C. albicans uses N-acetylglucosamine (GlcNAc), an abundant carbon source present in the GI tract, as a signal for nutrient availability. When placed in water, C. albicans cells normally enter the G0 phase and remain viable for weeks. However, they quickly lose viability when cultured in water containing only GlcNAc. We term this phenomenon GlcNAc-induced cell death (GICD). GlcNAc triggers the upregulation of ribosomal biogenesis genes, alterations of mitochondrial metabolism, and the accumulation of reactive oxygen species (ROS), followed by rapid cell death via both apoptotic and necrotic mechanisms. Multiple pathways, including the conserved cyclic AMP (cAMP) signaling and GlcNAc catabolic pathways, are involved in GICD. GlcNAc acts as a signaling molecule to regulate multiple cellular programs in a coordinated manner and therefore maximizes the efficiency of nutrient use. This adaptive behavior allows C. albicans’ more efficient colonization of the gut. The ability to rapidly and appropriately respond to nutrients in the environment is crucial to free-living microorganisms. To maximize the use of available nutrients, microorganisms often use a limiting nutritional component as a signal to coordinate multiple biological processes. The human fungal pathogen Candida albicans uses N-acetylglucosamine (GlcNAc) as a signal for the availability of external nutrient resources. GlcNAc induces rapid cell death in C. albicans due to the constitutive activation of oxidative metabolism and accumulation of reactive oxygen species (ROS), and multiple pathways are involved in its regulation. This study sheds light on the mechanisms of niche specialization of pathogenic fungi and raises the possibility that this cell death pathway could be an unexplored therapeutic target.
Collapse
|
229
|
Function and Regulation of Cph2 in Candida albicans. EUKARYOTIC CELL 2015; 14:1114-26. [PMID: 26342020 DOI: 10.1128/ec.00102-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/31/2015] [Indexed: 01/03/2023]
Abstract
Candida albicans is associated with humans as both a harmless commensal organism and a pathogen. Cph2 is a transcription factor whose DNA binding domain is similar to that of mammalian sterol response element binding proteins (SREBPs). SREBPs are master regulators of cellular cholesterol levels and are highly conserved from fungi to mammals. However, ergosterol biosynthesis is regulated by the zinc finger transcription factor Upc2 in C. albicans and several other yeasts. Cph2 is not necessary for ergosterol biosynthesis but is important for colonization in the murine gastrointestinal (GI) tract. Here we demonstrate that Cph2 is a membrane-associated transcription factor that is processed to release the N-terminal DNA binding domain like SREBPs, but its cleavage is not regulated by cellular levels of ergosterol or oxygen. Chromatin immunoprecipitation sequencing (ChIP-seq) shows that Cph2 binds to the promoters of HMS1 and other components of the regulatory circuit for GI tract colonization. In addition, 50% of Cph2 targets are also bound by Hms1 and other factors of the regulatory circuit. Several common targets function at the head of the glycolysis pathway. Thus, Cph2 is an integral part of the regulatory circuit for GI colonization that regulates glycolytic flux. Transcriptome sequencing (RNA-seq) shows a significant overlap in genes differentially regulated by Cph2 and hypoxia, and Cph2 is important for optimal expression of some hypoxia-responsive genes in glycolysis and the citric acid cycle. We suggest that Cph2 and Upc2 regulate hypoxia-responsive expression in different pathways, consistent with a synthetic lethal defect of the cph2 upc2 double mutant in hypoxia.
Collapse
|
230
|
MIG1 Regulates Resistance of Candida albicans against the Fungistatic Effect of Weak Organic Acids. EUKARYOTIC CELL 2015; 14:1054-61. [PMID: 26297702 DOI: 10.1128/ec.00129-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 08/13/2015] [Indexed: 12/15/2022]
Abstract
Candida albicans is the leading cause of fungal infections; but it is also a member of the human microbiome, an ecosystem of thousands of microbial species potentially influencing the outcome of host-fungal interactions. Accordingly, antibacterial therapy raises the risk of candidiasis, yet the underlying mechanism is currently not fully understood. We hypothesize the existence of bacterial metabolites that normally control C. albicans growth and of fungal resistance mechanisms against these metabolites. Among the most abundant microbiota-derived metabolites found on human mucosal surfaces are weak organic acids (WOAs), such as acetic, propionic, butyric, and lactic acid. Here, we used quantitative growth assays to investigate the dose-dependent fungistatic properties of WOAs on C. albicans growth and found inhibition of growth to occur at physiologically relevant concentrations and pH values. This effect was conserved across distantly related fungal species both inside and outside the CTG clade. We next screened a library of transcription factor mutants and identified several genes required for the resistance of C. albicans to one or more WOAs. A single gene, MIG1, previously known for its role in glucose repression, conferred resistance against all four acids tested. Consistent with glucose being an upstream activator of Mig1p, the presence of this carbon source was required for WOA resistance in wild-type C. albicans. Conversely, a MIG1-complemented strain completely restored the glucose-dependent resistance against WOAs. We conclude that Mig1p plays a central role in orchestrating a transcriptional program to fight against the fungistatic effect of this class of highly abundant metabolites produced by the gastrointestinal tract microbiota.
Collapse
|
231
|
Candida albicans mutant construction and characterization of selected virulence determinants. J Microbiol Methods 2015; 115:153-65. [DOI: 10.1016/j.mimet.2015.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/22/2022]
|
232
|
Abstract
The fungal cell wall confers cell morphology and protection against environmental insults. For fungal pathogens, the cell wall is a key immunological modulator and an ideal therapeutic target. Yeast cell walls possess an inner matrix of interlinked β-glucan and chitin that is thought to provide tensile strength and rigidity. Yeast cells remodel their walls over time in response to environmental change, a process controlled by evolutionarily conserved stress (Hog1) and cell integrity (Mkc1, Cek1) signaling pathways. These mitogen-activated protein kinase (MAPK) pathways modulate cell wall gene expression, leading to the construction of a new, modified cell wall. We show that the cell wall is not rigid but elastic, displaying rapid structural realignments that impact survival following osmotic shock. Lactate-grown Candida albicans cells are more resistant to hyperosmotic shock than glucose-grown cells. We show that this elevated resistance is not dependent on Hog1 or Mkc1 signaling and that most cell death occurs within 10 min of osmotic shock. Sudden decreases in cell volume drive rapid increases in cell wall thickness. The elevated stress resistance of lactate-grown cells correlates with reduced cell wall elasticity, reflected in slower changes in cell volume following hyperosmotic shock. The cell wall elasticity of lactate-grown cells is increased by a triple mutation that inactivates the Crh family of cell wall cross-linking enzymes, leading to increased sensitivity to hyperosmotic shock. Overexpressing Crh family members in glucose-grown cells reduces cell wall elasticity, providing partial protection against hyperosmotic shock. These changes correlate with structural realignment of the cell wall and with the ability of cells to withstand osmotic shock. The C. albicans cell wall is the first line of defense against external insults, the site of immune recognition by the host, and an attractive target for antifungal therapy. Its tensile strength is conferred by a network of cell wall polysaccharides, which are remodeled in response to growth conditions and environmental stress. However, little is known about how cell wall elasticity is regulated and how it affects adaptation to stresses such as sudden changes in osmolarity. We show that elasticity is critical for survival under conditions of osmotic shock, before stress signaling pathways have time to induce gene expression and drive glycerol accumulation. Critical cell wall remodeling enzymes control cell wall flexibility, and its regulation is strongly dependent on host nutritional inputs. We also demonstrate an entirely new level of cell wall dynamism, where significant architectural changes and structural realignment occur within seconds of an osmotic shock.
Collapse
|
233
|
Abstract
The Candida albicans RHR2 gene, which specifies a glycerol biosynthetic enzyme, is required for biofilm formation in vitro and in vivo. Prior studies indicate that RHR2 is ultimately required for expression of adhesin genes, such as ALS1. In fact, RHR2 is unnecessary for biofilm formation when ALS1 is overexpressed from an RHR2-independent promoter. Here, we describe two additional biological processes that depend upon RHR2: invasion into an abiotic substrate and pathogenicity in an abdominal infection model. We report here that abiotic substrate invasion occurs concomitantly with biofilm formation, and a screen of transcription factor mutants indicates that biofilm and hyphal formation ability correlates with invasion ability. However, analysis presented here of the rhr2Δ/Δ mutant separates biofilm formation and invasion. We found that an rhr2Δ/Δ mutant forms a biofilm upon overexpression of the adhesin gene ALS1 or the transcription factor genes BRG1 or UME6. However, the biofilm-forming strains do not invade the substrate. These results indicate that RHR2 has an adhesin-independent role in substrate invasion, and mathematical modeling argues that RHR2 is required to generate turgor. Previous studies have shown that abdominal infection by C. albicans has two aspects: infection of abdominal organs and persistence in abscesses. We report here that an rhr2Δ/Δ mutant is defective in both of these infection phenotypes. We find here that overexpression of ALS1 in the mutant restores infection of organs, but does not improve persistence in abscesses. Therefore, RHR2 has an adhesin-independent role in abdominal infection, just as it does in substrate invasion. This report suggests that RHR2, through glycerol synthesis, coordinates adherence with host- or substrate-interaction activities that enable proliferation of the C. albicans population.
Collapse
|
234
|
Affiliation(s)
- Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
235
|
Gil-Bona A, Parra-Giraldo CM, Hernáez ML, Reales-Calderon JA, Solis NV, Filler SG, Monteoliva L, Gil C. Candida albicans cell shaving uncovers new proteins involved in cell wall integrity, yeast to hypha transition, stress response and host-pathogen interaction. J Proteomics 2015; 127:340-351. [PMID: 26087349 DOI: 10.1016/j.jprot.2015.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/25/2015] [Accepted: 06/09/2015] [Indexed: 01/09/2023]
Abstract
The ability to switch from yeast to hyphal growth is essential for virulence in Candida albicans. The cell surface is the initial point of contact between the fungus and the host. In this work, a free-gel proteomic strategy based on tryptic digestion of live yeast and hyphae cells and protein identification using LC-MS/MS methodology was used to identify cell surface proteins. Using this strategy, a total of 943 proteins were identified, of which 438 were in yeast and 928 were in hyphae. Of these proteins, 79 were closely related to the organization and biogenesis of the cell wall, including 28 GPI-anchored proteins, such as Hyr1 and Sod5 which were detected exclusively in hyphae, and Als2 and Sap10which were detected only in yeast. A group of 17 proteins of unknown function were subsequently studied by analysis of the corresponding deletion mutants. We found that four new proteins, Pst3, Tos1, Orf19.3060 and Orf19.5352 are involved in cell wall integrity and in C. albicans' engulfment by macrophages. Moreover, the putative NADH-ubiquinone-related proteins, Ali1, Mci4, Orf19.287 and Orf19.7590, are also involved in osmotic and oxidative resistance, yeast to hypha transition and the ability to damage and invade oral epithelial cells. This article is part of a Special Issue entitled: HUPO 2014.
Collapse
Affiliation(s)
- Ana Gil-Bona
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Claudia Marcela Parra-Giraldo
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - María Luisa Hernáez
- Unidad de Proteómica, Universidad Complutense de Madrid-Parque Científico de Madrid (UCM-PCM), Spain
| | - Jose Antonio Reales-Calderon
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Norma V Solis
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Scott G Filler
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lucia Monteoliva
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Concha Gil
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| |
Collapse
|
236
|
Kim K, Zilbermintz L, Martchenko M. Repurposing FDA approved drugs against the human fungal pathogen, Candida albicans. Ann Clin Microbiol Antimicrob 2015; 14:32. [PMID: 26054754 PMCID: PMC4462072 DOI: 10.1186/s12941-015-0090-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/27/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The high cost and prolonged timeline of new drug discovery and development are major roadblocks to creating therapies for infectious diseases. Candida albicans is an opportunistic fungal pathogen that is the most common cause of fatal fungal infections in humans and costs $2-4 billion dollars to treat in the US alone. METHODS To accelerate drug discovery, we screened a library of 1581 existing FDA approved drugs, as well as drugs approved abroad, for inhibitors of C. albicans. The screen was done on YPD yeast growth media as well as on the serum plate assay developed in this study. RESULTS We discovered that fifteen drugs, all which were originally approved for treating various infectious and non-infectious diseases, were able to kill Candida albicans. Additionally, one of those drugs, Octodrine, displays wide-spectrum anti-microbial activity. Compared to other selected anti-Candida drugs, Octodrine was shown to be one of the most effective drugs in killing serum-grown Candida albicans without significantly affecting the survival of host macrophages and skin cells. CONCLUSIONS This approach is useful for the discovery of economically viable new therapies against infectious diseases.
Collapse
Affiliation(s)
- Kevin Kim
- Keck Graduate Institute, Claremont, CA, 91711, USA.
| | | | | |
Collapse
|
237
|
Abstract
Fungal infections are responsible for millions of human deaths annually. Copper, an essential but toxic trace element, plays an important role at the host-pathogen axis during infection. In this review, we describe how the host uses either Cu compartmentalization within innate immune cells or Cu sequestration in other infected host niches such as in the brain to combat fungal infections. We explore Cu toxicity mechanisms and the Cu homeostasis machinery that fungal pathogens bring into play to succeed in establishing an infection. Finally, we address recent approaches that manipulate Cu-dependent processes at the host-pathogen axis for antifungal drug development.
Collapse
Affiliation(s)
| | - Dennis J Thiele
- From the Departments of Pharmacology & Cancer Biology and Biochemistry, Duke University, School of Medicine, Durham, North Carolina 27710
| |
Collapse
|
238
|
Calderone R, Li D, Traven A. System-level impact of mitochondria on fungal virulence: to metabolism and beyond. FEMS Yeast Res 2015; 15:fov027. [PMID: 26002841 PMCID: PMC4542695 DOI: 10.1093/femsyr/fov027] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/26/2015] [Accepted: 05/14/2015] [Indexed: 12/23/2022] Open
Abstract
The mitochondrion plays wide-ranging roles in eukaryotic cell physiology. In pathogenic fungi, this central metabolic organelle mediates a range of functions related to disease, from fitness of the pathogen to developmental and morphogenetic transitions to antifungal drug susceptibility. In this review, we present the latest findings in this area. We focus on likely mechanisms of mitochondrial impact on fungal virulence pathways through metabolism and stress responses, but also potentially via control over signaling pathways. We highlight fungal mitochondrial proteins that lack human homologs, and which could be inhibited as a novel approach to antifungal drug strategy.
Collapse
Affiliation(s)
- Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Monash University Clayton, 3800 VIC, Australia
| |
Collapse
|
239
|
Fox EP, Bui CK, Nett JE, Hartooni N, Mui MC, Andes DR, Nobile CJ, Johnson AD. An expanded regulatory network temporally controls Candida albicans biofilm formation. Mol Microbiol 2015; 96:1226-39. [PMID: 25784162 PMCID: PMC4464956 DOI: 10.1111/mmi.13002] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2015] [Indexed: 01/10/2023]
Abstract
Candida albicans biofilms are composed of highly adherent and densely arranged cells with properties distinct from those of free‐floating (planktonic) cells. These biofilms are a significant medical problem because they commonly form on implanted medical devices, are drug resistant and are difficult to remove. C. albicans biofilms are not static structures; rather they are dynamic and develop over time. Here we characterize gene expression in biofilms during their development, and by comparing them to multiple planktonic reference states, we identify patterns of gene expression relevant to biofilm formation. In particular, we document time‐dependent changes in genes involved in adhesion and metabolism, both of which are at the core of biofilm development. Additionally, we identify three new regulators of biofilm formation, Flo8, Gal4, and Rfx2, which play distinct roles during biofilm development over time. Flo8 is required for biofilm formation at all time points, and Gal4 and Rfx2 are needed for proper biofilm formation at intermediate time points.
Collapse
Affiliation(s)
- Emily P Fox
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA.,Tetrad Program, Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Catherine K Bui
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Jeniel E Nett
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA.,Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Nairi Hartooni
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Michael C Mui
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - David R Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA.,Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Clarissa J Nobile
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| |
Collapse
|
240
|
Jung KW, Yang DH, Maeng S, Lee KT, So YS, Hong J, Choi J, Byun HJ, Kim H, Bang S, Song MH, Lee JW, Kim MS, Kim SY, Ji JH, Park G, Kwon H, Cha S, Meyers GL, Wang LL, Jang J, Janbon G, Adedoyin G, Kim T, Averette AK, Heitman J, Cheong E, Lee YH, Lee YW, Bahn YS. Systematic functional profiling of transcription factor networks in Cryptococcus neoformans. Nat Commun 2015; 6:6757. [PMID: 25849373 PMCID: PMC4391232 DOI: 10.1038/ncomms7757] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/24/2015] [Indexed: 02/06/2023] Open
Abstract
Cryptococcus neoformans causes life-threatening meningoencephalitis in humans, but its overall biological and pathogenic regulatory circuits remain elusive, particularly due to the presence of an evolutionarily divergent set of transcription factors (TFs). Here, we report the construction of a high-quality library of 322 signature-tagged gene-deletion strains for 155 putative TF genes previously predicted using the DNA-binding domain TF database, and examine their in vitro and in vivo phenotypic traits under 32 distinct growth conditions. At least one phenotypic trait is exhibited by 145 out of 155 TF mutants (93%) and ∼85% of them (132/155) are functionally characterized for the first time in this study. The genotypic and phenotypic data for each TF are available in the C. neoformans TF phenome database (http://tf.cryptococcus.org). In conclusion, our phenome-based functional analysis of the C. neoformans TF mutant library provides key insights into transcriptional networks of basidiomycetous fungi and human fungal pathogens.
Collapse
Affiliation(s)
- Kwang-Woo Jung
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Dong-Hoon Yang
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Shinae Maeng
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Kyung-Tae Lee
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Yee-Seul So
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Joohyeon Hong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Jaeyoung Choi
- Department of Agricultural Biotechnology, Center for Fungal Pathogenesis, Seoul National University, Seoul 151-921, Korea
| | - Hyo-Jeong Byun
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Hyelim Kim
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Soohyun Bang
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Min-Hee Song
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Jang-Won Lee
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Min Su Kim
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Seo-Young Kim
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Je-Hyun Ji
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Goun Park
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Hyojeong Kwon
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Suyeon Cha
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Gena Lee Meyers
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Li Li Wang
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Jooyoung Jang
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Guilhem Janbon
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, Paris F-75015, France
| | - Gloria Adedoyin
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Taeyup Kim
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Anna K. Averette
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Yong-Hwan Lee
- Department of Agricultural Biotechnology, Center for Fungal Pathogenesis, Seoul National University, Seoul 151-921, Korea
| | - Yin-Won Lee
- Department of Agricultural Biotechnology, Center for Fungal Pathogenesis, Seoul National University, Seoul 151-921, Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
241
|
Merhej J, Delaveau T, Guitard J, Palancade B, Hennequin C, Garcia M, Lelandais G, Devaux F. Yap7 is a transcriptional repressor of nitric oxide oxidase in yeasts, which arose from neofunctionalization after whole genome duplication. Mol Microbiol 2015; 96:951-72. [DOI: 10.1111/mmi.12983] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2015] [Indexed: 11/27/2022]
Affiliation(s)
- Jawad Merhej
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
- CNRS, UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
| | - Thierry Delaveau
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
- CNRS, UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
| | - Juliette Guitard
- Sorbonne Universités, UPMC Univ Paris 06, CR7; Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris); 91 Bd de l'hôpital F-75013 Paris France
- Inserm; U1135; CIMI-Paris; 91 Bd de l'hôpital F-75013 Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital St Antoine; Service de Parasitologie-Mycologie; F-75012 Paris France
- CNRS; ERL 8255; CIMI-Paris; 91 Bd de l'hôpital F-75013 Paris France
| | - Benoit Palancade
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot; Sorbonne Paris Cité; F-75205 Paris France
| | - Christophe Hennequin
- Sorbonne Universités, UPMC Univ Paris 06, CR7; Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris); 91 Bd de l'hôpital F-75013 Paris France
- Inserm; U1135; CIMI-Paris; 91 Bd de l'hôpital F-75013 Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital St Antoine; Service de Parasitologie-Mycologie; F-75012 Paris France
- CNRS; ERL 8255; CIMI-Paris; 91 Bd de l'hôpital F-75013 Paris France
| | - Mathilde Garcia
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
- CNRS, UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
| | - Gaëlle Lelandais
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot; Sorbonne Paris Cité; F-75205 Paris France
| | - Frédéric Devaux
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
- CNRS, UMR 7238; Laboratoire de biologie computationnelle et quantitative; F-75006 Paris France
| |
Collapse
|
242
|
Urrialde V, Prieto D, Pla J, Alonso-Monge R. The Pho4 transcription factor mediates the response to arsenate and arsenite in Candida albicans. Front Microbiol 2015; 6:118. [PMID: 25717325 PMCID: PMC4324303 DOI: 10.3389/fmicb.2015.00118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/29/2015] [Indexed: 11/21/2022] Open
Abstract
Arsenate (As (V)) is the dominant form of the toxic metalloid arsenic (As). Microorganisms have consequently developed mechanisms to detoxify and tolerate this kind of compounds. In the present work, we have explored the arsenate sensing and signaling mechanisms in the pathogenic fungus Candida albicans. Although mutants impaired in the Hog1 or Mkc1-mediated pathways did not show significant sensitivity to this compound, both Hog1 and Mkc1 became phosphorylated upon addition of sodium arsenate to growing cells. Hog1 phosphorylation upon arsenate challenge was shown to be Ssk1-dependent. A screening designed for the identification of transcription factors involved in the arsenate response identified Pho4, a transcription factor of the myc-family, as pho4 mutants were susceptible to As (V). The expression of PHO4 was shortly induced in the presence of sodium arsenate in a Hog1-independent manner. Pho4 level affects Hog1 phosphorylation upon As (V) challenge, suggesting an indirect relationship between Pho4 activity and signaling in C. albicans. Pho4 also mediates the response to arsenite as revealed by the fact that pho4 defective mutants are sensitive to arsenite and Pho4 becomes phosphorylated upon sodium arsenite addition. Arsenite also triggers Hog1 phosphorylation by a process that is, in this case, independent of the Ssk1 kinase. These results indicate that the HOG pathway mediates the response to arsenate and arsenite in C. albicans and that the Pho4 transcription factor can differentiate among As (III), As (V) and Pi, triggering presumably specific responses.
Collapse
Affiliation(s)
- Verónica Urrialde
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Daniel Prieto
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Jesús Pla
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| | - Rebeca Alonso-Monge
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Madrid, Spain
| |
Collapse
|
243
|
Pointer BR, Boyer MP, Schmidt M. Boric acid destabilizes the hyphal cytoskeleton and inhibits invasive growth ofCandida albicans. Yeast 2015; 32:389-98. [DOI: 10.1002/yea.3066] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/12/2014] [Accepted: 01/14/2015] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - Michael P. Boyer
- Department of Biochemistry and Nutrition; Des Moines University; IA USA
| | - Martin Schmidt
- Department of Biochemistry and Nutrition; Des Moines University; IA USA
| |
Collapse
|
244
|
Wells ML, Washington OL, Hicks SN, Nobile CJ, Hartooni N, Wilson GM, Zucconi BE, Huang W, Li L, Fargo DC, Blackshear PJ. Post-transcriptional regulation of transcript abundance by a conserved member of the tristetraprolin family in Candida albicans. Mol Microbiol 2015; 95:1036-53. [PMID: 25524641 DOI: 10.1111/mmi.12913] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2014] [Indexed: 11/29/2022]
Abstract
Members of the tristetraprolin (TTP) family of CCCH tandem zinc finger proteins bind to AU-rich regions in target mRNAs, leading to their deadenylation and decay. Family members in Saccharomyces cerevisiae influence iron metabolism, whereas the single protein expressed in Schizosaccharomyces pombe, Zfs1, regulates cell-cell interactions. In the human pathogen Candida albicans, deep sequencing of mutants lacking the orthologous protein, Zfs1, revealed significant increases (> 1.5-fold) in 156 transcripts. Of these, 113 (72%) contained at least one predicted TTP family member binding site in their 3'UTR, compared with only 3 of 56 (5%) down-regulated transcripts. The zfs1Δ/Δ mutant was resistant to 3-amino-1,2,4-triazole, perhaps because of increased expression of the potential target transcript encoded by HIS3. Sequences of the proteins encoded by the putative Zfs1 targets were highly conserved among other species within the fungal CTG clade, while the predicted Zfs1 binding sites in these mRNAs often 'disappeared' with increasing evolutionary distance from the parental species. C. albicans Zfs1 bound to the ideal mammalian TTP binding site with high affinity, and Zfs1 was associated with target transcripts after co-immunoprecipitation. Thus, the biochemical activities of these proteins in fungi are highly conserved, but Zfs1-like proteins may target different transcripts in each species.
Collapse
Affiliation(s)
- Melissa L Wells
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Abstract
In budding yeast, Saccharomyces cerevisiae, the phosphate signalling and response pathway, known as PHO pathway, monitors phosphate cytoplasmic levels by controlling genes involved in scavenging, uptake and utilization of phosphate. Recent attempts to understand the phosphate starvation response in other ascomycetes have suggested the existence of both common and novel components of the budding yeast PHO pathway in these ascomycetes. In this review, we discuss the components of PHO pathway, their roles in maintaining phosphate homeostasis in yeast and their conservation across ascomycetes. The role of high-affinity transporter, Pho84, in sensing and signalling of phosphate has also been discussed.
Collapse
Affiliation(s)
- Parul Tomar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400 005, India
| | | |
Collapse
|
246
|
Horton BN, Kumar A. Genome-wide synthetic genetic screening by transposon mutagenesis in Candida albicans. Methods Mol Biol 2015; 1279:125-35. [PMID: 25636616 DOI: 10.1007/978-1-4939-2398-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transposon-based mutagenesis is an effective method for genetic screening on a genome-wide scale, with particular applicability in organisms possessing compact genomes where transforming DNA tends to integrate by homologous recombination. Methods for transposon mutagenesis have been applied with great success in the budding yeast Saccharomyces cerevisiae and in the related pathogenic yeast Candida albicans. In C. albicans, we have implemented transposon mutagenesis to generate heterozygous mutations for the analysis of complex haploinsufficiency, a type of synthetic genetic interaction wherein a pair of non-complementing heterozygous mutations results in a stronger phenotype then either individual mutation in isolation. Genes exhibiting complex haploinsufficiency typically function within a regulatory pathway, in parallel pathways, or in parallel branches within a single pathway. Here, we present protocols to implement transposon mutagenesis for complex haploinsufficiency screening in C. albicans, indicating methods for transposon construction, mutagenesis, phenotypic screening, and identification of insertion sites in strains of interest. In total, the approach is a useful means to implement large-scale synthetic genetic screening in the diploid C. albicans.
Collapse
Affiliation(s)
- Brooke N Horton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1048, USA
| | | |
Collapse
|
247
|
De S, Pérez JC. Reshuffling transcriptional circuits: how microorganisms adapt to colonize the human body. Transcription 2014; 5:e976095. [PMID: 25483603 PMCID: PMC4581354 DOI: 10.4161/21541264.2014.976095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 11/19/2022] Open
Abstract
Several hundred taxa of microorganisms-including bacteria, archaea and eukaryotes-inhabit the human body. What did it take for these species to become stable residents of humans? Recent reports illustrate how evolutionary changes in transcriptional circuits played a pivotal role in the adaptation of single-celled eukaryotes to colonize mammals.
Collapse
Affiliation(s)
- Sonakshi De
- Institut für Molekulare Infektionsbiologie; Universität Würzburg; Würzburg, Germany
| | - J Christian Pérez
- Institut für Molekulare Infektionsbiologie; Universität Würzburg; Würzburg, Germany
| |
Collapse
|
248
|
Lin C, Lin CN, Wang YC, Liu FY, Chien YW, Chuang YJ, Lan CY, Hsieh WP, Chen BS. Robustness analysis on interspecies interaction network for iron and glucose competition between Candida albicans and zebrafish during infection. BMC SYSTEMS BIOLOGY 2014; 8 Suppl 5:S6. [PMID: 25603810 PMCID: PMC4305985 DOI: 10.1186/1752-0509-8-s5-s6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Candida albicans has emerged as an important model organism for the study of infectious disease. Using high-throughput simultaneously quantified time-course transcriptomics, this study constructed host-pathogen interspecies interaction networks between C. albicans and zebrafish during the adhesion, invasion, and damage stages. Given that iron and glucose have been identified as crucial resources required during the infection process between C. albicans and zebrafish, we focused on the construction of the interspecies networks associated with them. Furthermore, a randomization technique was proposed to identify differentially regulated proteins that are statistically eminent for the three infection stages. The behaviors of the highly connected or differentially regulated proteins identified from the resulting networks were further investigated. "Robustness" is an important system property that measures the ability of the system tolerating the intrinsic perturbations in a dynamic network. This characteristic provides a systematic and quantitative view to elucidate the dynamics of iron and glucose competition in terms of the interspecies interaction networks. Here, we further estimated the robustness of our constructed interspecies interaction networks for the three infection stages. The constructed networks and robustness analysis provided significant insight into dynamic interactions related to iron and glucose competition during infection and enabled us to quantify the system's intrinsic perturbation tolerance ability during iron and glucose competition throughout the three infection stages. Moreover, the networks also assist in elucidating the offensive and defensive mechanisms of C. albicans and zebrafish during their competition for iron and glucose. Our proposed method can be easily extended to identify other such networks involved in the competition for essential resources during infection.
Collapse
|
249
|
Goranov AI, Madhani HD. Functional profiling of human fungal pathogen genomes. Cold Spring Harb Perspect Med 2014; 5:a019596. [PMID: 25377143 DOI: 10.1101/cshperspect.a019596] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Fungal infections are challenging to diagnose and often difficult to treat, with only a handful of drug classes existing. Understanding the molecular mechanisms by which pathogenic fungi cause human disease is imperative. Here, we discuss how the development and use of genome-scale genetic resources, such as whole-genome knockout collections, can address this unmet need. Using work in Saccharomcyes cerevisiae as a guide, studies of Cryptococcus neoformans and Candida albicans have shown how the challenges of large-scale gene deletion can be overcome, and how such collections can be effectively used to obtain insights into mechanisms of pathogenesis. We conclude that, with concerted efforts, full genome-wide functional analysis of human fungal pathogen genomes is within reach.
Collapse
Affiliation(s)
- Alexi I Goranov
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94158
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
250
|
Fox EP, Cowley ES, Nobile CJ, Hartooni N, Newman DK, Johnson AD. Anaerobic bacteria grow within Candida albicans biofilms and induce biofilm formation in suspension cultures. Curr Biol 2014; 24:2411-6. [PMID: 25308076 PMCID: PMC4252622 DOI: 10.1016/j.cub.2014.08.057] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 07/31/2014] [Accepted: 08/22/2014] [Indexed: 12/28/2022]
Abstract
The human microbiome contains diverse microorganisms, which share and compete for the same environmental niches. A major microbial growth form in the human body is the biofilm state, where tightly packed bacterial, archaeal, and fungal cells must cooperate and/or compete for resources in order to survive. We examined mixed biofilms composed of the major fungal species of the gut microbiome, Candida albicans, and each of five prevalent bacterial gastrointestinal inhabitants: Bacteroides fragilis, Clostridium perfringens, Escherichia coli, Klebsiella pneumoniae, and Enterococcus faecalis. We observed that biofilms formed by C. albicans provide a hypoxic microenvironment that supports the growth of two anaerobic bacteria, even when cultured in ambient oxic conditions that are normally toxic to the bacteria. We also found that coculture with bacteria in biofilms induces massive gene expression changes in C. albicans, including upregulation of WOR1, which encodes a transcription regulator that controls a phenotypic switch in C. albicans, from the "white" cell type to the "opaque" cell type. Finally, we observed that in suspension cultures, C. perfringens induces aggregation of C. albicans into "mini-biofilms," which allow C. perfringens cells to survive in a normally toxic environment. This work indicates that bacteria and C. albicans interactions modulate the local chemistry of their environment in multiple ways to create niches favorable to their growth and survival.
Collapse
Affiliation(s)
- Emily P Fox
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16(th) Street, San Francisco, CA 94158, USA; Tetrad Program, Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16(th) Street, San Francisco, CA 94158, USA
| | - Elise S Cowley
- Division of Biology and Biological Engineering, California Institute of Technology, 147-75, 1200 East California Boulevard, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, 147-75, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Clarissa J Nobile
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16(th) Street, San Francisco, CA 94158, USA; School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, CA 95343, USA
| | - Nairi Hartooni
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16(th) Street, San Francisco, CA 94158, USA
| | - Dianne K Newman
- Division of Biology and Biological Engineering, California Institute of Technology, 147-75, 1200 East California Boulevard, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, 147-75, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16(th) Street, San Francisco, CA 94158, USA.
| |
Collapse
|