1
|
Song J, Zhang S, Xing J, Zhang L, Wang J, Shan A. Optimizing therapeutic efficacy of antifungal peptides via strategic terminal amino acid modification. J Adv Res 2024:S2090-1232(24)00416-8. [PMID: 39322048 DOI: 10.1016/j.jare.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024] Open
Abstract
INTRODUCTION Antifungal peptides (AFPs) have the potential to treat antifungal-resistant infections; however, their structure-function relationship remains unknown, hindering their rapid development. Therefore, it is imperative to investigate and clarify the structure-function relationships of AFPs. OBJECTIVES This study aimed to investigate the impact of end-tagging single hydrophobic amino acids and capping the N-terminus with glycine (Gly) on the antifungal activity of peptide W4. METHODS The antifungal efficacy of the engineered peptides was initially assessed by determining the minimum inhibitory concentration (MIC) /minimal fungicidal concentration (MFC), killing kinetics, and drug resistance induction, in addition to evaluating the biocompatibility and stability. Subsequently, the antifungal mechanism was investigated using fluorescence labeling, electron microscopy, reactive oxygen species (ROS) detection, and measurement of mitochondrial membrane potential and apoptosis. The impact of the engineered peptides on Candida albicans (C. albicans) biofilm and their potential application in the scratch keratomycosis model were investigated. RESULTS The antifungal activity of W4 was significantly enhanced by capping Gly at the N-terminus, resulting in a decrease in average activity from 11.86 μM to 6.25 μM (GW4) and an increase in TI values by 1.9-fold (TIGW4 = 40.99). Mechanistically, GW4 exerted its antifungal effect by disrupting the cellular membrane structure in C. albicans, forming pores and subsequent leakage of intracellular contents. Concurrently, it facilitated intracellular ROS accumulation while decreasing the mitochondrial membrane potential. Additionally, GW4 demonstrated an excellent ability to inhibit and eliminate biofilms of C. albicans. Notably, GW4 demonstrated significant therapeutic potential in a C. albicans-associated keratitis model. CONCLUSION Capping Gly at the N-terminus increased residue length while significantly enhancing the helical propensity of W4, thereby augmenting its antifungal activity. Our exploratory study demonstrated the potential strategies and avenues for optimizing the structure-function relationships of AFPs and developing highly effective antifungal drugs.
Collapse
Affiliation(s)
- Jing Song
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China
| | - Shanshan Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China
| | - Junya Xing
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China
| | - Jiajun Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China.
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, PR China.
| |
Collapse
|
2
|
Kumar D, Kumar A. Molecular Determinants Involved in Candida albicans Biofilm Formation and Regulation. Mol Biotechnol 2024; 66:1640-1659. [PMID: 37410258 DOI: 10.1007/s12033-023-00796-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Candida albicans is known for its pathogenicity, although it lives within the human body as a commensal member. The commensal nature of C. albicans is well controlled and regulated by the host's immune system as they live in the harmonized microenvironment. However, the development of certain unusual microhabitat conditions (change in pH, co-inhabiting microorganisms' population ratio, debilitated host-immune system) pokes this commensal fungus to transform into a pathogen in such a way that it starts to propagate very rapidly and tries to breach the epithelial barrier to enter the host's systemic circulations. In addition, Candida is infamous as a major nosocomial (hospital-acquired infection) agent because it enters the human body through venous catheters or medical prostheses. The hysterical mode of C. albicans growth builds its microcolony or biofilm, which is pathogenic for the host. Biofilms propose additional resistance mechanisms from host immunity or extracellular chemicals to aid their survival. Differential gene expressions and regulations within the biofilms cause altered morphology and metabolism. The genes associated with adhesiveness, hyphal/pseudo-hyphal growth, persister cell transformation, and biofilm formation by C. albicans are controlled by myriads of cell-signaling regulators. These genes' transcription is controlled by different molecular determinants like transcription factors and regulators. Therefore, this review has focused discussion on host-immune-sensing molecular determinants of Candida during biofilm formation, regulatory descriptors (secondary messengers, regulatory RNAs, transcription factors) of Candida involved in biofilm formation that could enable small-molecule drug discovery against these molecular determinants, and lead to disrupt the well-structured Candida biofilms effectively.
Collapse
Affiliation(s)
- Dushyant Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India.
| |
Collapse
|
3
|
Lee J, Song H, Kim K. Inhibition of Candida albicans Biofilm Formation and Attenuation of Its Virulence by Liriope muscari. Antibiotics (Basel) 2024; 13:434. [PMID: 38786162 PMCID: PMC11117302 DOI: 10.3390/antibiotics13050434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
(1) Background: Although Candida albicans accounts for the majority of fungal infections, therapeutic options are limited and require alternative antifungal agents with new targets; (2) Methods: A biofilm formation assay with RPMI1640 medium was performed with Liriope muscari extract. A combination antifungal assay, dimorphic transition assay, and adhesion assay were performed under the biofilm formation condition to determine the anti-biofilm formation effect. qRT-PCR analysis was accomplished to confirm changes in gene expression; (3) Results: L. muscari extract significantly reduces biofilm formation by 51.65% at 1.56 μg/mL use and therefore increases susceptibility to miconazole. L. muscari extract also inhibited the dimorphic transition of Candida; nearly 50% of the transition was inhibited when 1.56 μg/mL of the extract was treated. The extract of L. muscari inhibited the expression of genes related to hyphal development and extracellular matrix of 34.4% and 36.0%, respectively, as well as genes within the Ras1-cAMP-PKA, Cph2-Tec1, and MAP kinase signaling pathways of 25.58%, 7.1% and 15.8%, respectively, at 1.56 μg/mL of L. muscari extract treatment; (4) Conclusions: L. muscari extract significantly reduced Candida biofilm formation, which lead to induced antifungal susceptibility to miconazole. It suggests that L. muscari extract is a promising anti-biofilm candidate of Candida albicans since the biofilm formation of Candida albicans is an excellent target for candidiasis regulation.
Collapse
Affiliation(s)
- Jeonghoon Lee
- Department of Medical Science of Meridian, College of Korean Medicine, Graduate School, Kyung Hee University, Kyungheedae-ro 6-gil, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Hyunchan Song
- Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea;
| | - Kiyoung Kim
- Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea;
| |
Collapse
|
4
|
Bravo-Chaucanés CP, Chitiva LC, Vargas-Casanova Y, Diaz-Santoyo V, Hernández AX, Costa GM, Parra-Giraldo CM. Exploring the Potential Mechanism of Action of Piperine against Candida albicans and Targeting Its Virulence Factors. Biomolecules 2023; 13:1729. [PMID: 38136600 PMCID: PMC10742119 DOI: 10.3390/biom13121729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 12/24/2023] Open
Abstract
Plant-derived compounds have proven to be a source of inspiration for new drugs. In this study, piperine isolated from the fruits of Piper nigrum showed anti-Candida activity. Furthermore, the mechanisms of action of piperine and its impact on virulence factors in Candida albicans, which have not been comprehensively understood, were also assessed. Initially, piperine suppressed the hyphal transition in both liquid and solid media, hindered biofilm formation, and resulted in observable cell distortions in scanning electron microscope (SEM) samples, for both fluconazole-sensitive and fluconazole-resistant C. albicans strains. Additionally, the morphogenetic switches triggered by piperine were found to rely on the activity of mutant C. albicans strains. Secondly, piperine treatment increased cell membrane permeability and disrupted mitochondrial membrane potential, as evidenced by propidium iodine and Rhodamine 123 staining, respectively. Moreover, it induced the accumulation of intracellular reactive oxygen species in C. albicans. Synergy was obtained between the piperine and the fluconazole against the fluconazole-sensitive strain. Interestingly, there were no hemolytic effects of piperine, and it resulted in reduced cytotoxicity on fibroblast cells at low concentrations. The results suggest that piperine could have a dual mode of action inhibiting virulence factors and modulating cellular processes, leading to cell death in C. albicans.
Collapse
Affiliation(s)
- Claudia Patricia Bravo-Chaucanés
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (C.P.B.-C.); (Y.V.-C.); (V.D.-S.)
| | - Luis Carlos Chitiva
- Grupo de Investigación Fitoquímica Universidad Javeriana (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (L.C.C.); (A.X.H.); (G.M.C.)
| | - Yerly Vargas-Casanova
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (C.P.B.-C.); (Y.V.-C.); (V.D.-S.)
| | - Valentina Diaz-Santoyo
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (C.P.B.-C.); (Y.V.-C.); (V.D.-S.)
| | - Andrea Ximena Hernández
- Grupo de Investigación Fitoquímica Universidad Javeriana (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (L.C.C.); (A.X.H.); (G.M.C.)
| | - Geison M. Costa
- Grupo de Investigación Fitoquímica Universidad Javeriana (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (L.C.C.); (A.X.H.); (G.M.C.)
| | - Claudia Marcela Parra-Giraldo
- Unidad de Proteómica y Micosis Humanas, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, DC, Colombia; (C.P.B.-C.); (Y.V.-C.); (V.D.-S.)
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
5
|
Schena NC, Baker KM, Stark AA, Thomas DP, Cleary IA. Constitutive ALS3 expression in Candida albicans enhances adhesion and biofilm formation of efg1, but not cph1 mutant strains. PLoS One 2023; 18:e0286547. [PMID: 37440498 PMCID: PMC10343153 DOI: 10.1371/journal.pone.0286547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/17/2023] [Indexed: 07/15/2023] Open
Abstract
Adhesion to living and non-living surfaces is an important virulence trait of the fungal pathogen Candida albicans. Biofilm formation in this organism depends on the expression of a number of cell surface proteins including the hypha-specific protein Als3p. Loss of ALS3 impairs biofilm formation and decreases cell-cell adhesion. We wanted to test whether constitutively expressing ALS3 could compensate for defects in adhesion and biofilm formation observed in mutant strains that lack key transcriptional regulators of biofilm formation Efg1p and Cph1p. We found that ALS3 improved adhesion and biofilm formation in the efg1Δ and efg1Δ cph1Δ mutant strains, but had less effect on the cph1Δ strain.
Collapse
Affiliation(s)
- Nicholas C. Schena
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Kassandra M. Baker
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Anna A. Stark
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Derek P. Thomas
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| | - Ian A. Cleary
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, United States of America
| |
Collapse
|
6
|
Kim C, Kim JG, Kim KY. Anti- Candida Potential of Sclareol in Inhibiting Growth, Biofilm Formation, and Yeast-Hyphal Transition. J Fungi (Basel) 2023; 9:jof9010098. [PMID: 36675919 PMCID: PMC9862543 DOI: 10.3390/jof9010098] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Even though Candida albicans commonly colonizes on most mucosal surfaces including the vaginal and gastrointestinal tract, it can cause candidiasis as an opportunistic infectious fungus. The emergence of resistant Candida strains and the toxicity of anti-fungal agents have encouraged the development of new classes of potential anti-fungal agents. Sclareol, a labdane-type diterpene, showed anti-Candida activity with a minimum inhibitory concentration of 50 μg/mL in 24 h based on a microdilution anti-fungal susceptibility test. Cell membrane permeability with propidium iodide staining and mitochondrial membrane potential with JC-1 staining were increased in C. albicans by treatment of sclareol. Sclareol also suppressed the hyphal formation of C. albicans in both liquid and solid media, and reduced biofilm formation. Taken together, sclareol induces an apoptosis-like cell death against Candida spp. and suppressed biofilm and hyphal formation in C. albicans. Sclareol is of high interest as a novel anti-fungal agent and anti-virulence factor.
Collapse
Affiliation(s)
- Chaerim Kim
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-do, Republic of Korea
| | - Jae-Goo Kim
- Graduate School of Biotechnology, Kyung Hee University, Yingin 17104, Gyeonggi-do, Republic of Korea
| | - Ki-Young Kim
- Graduate School of Biotechnology, Kyung Hee University, Yingin 17104, Gyeonggi-do, Republic of Korea
- College of Life Science, Kyung Hee University, Yongin 17104, Gyeonggi-do, Republic of Korea
- Correspondence: ; Tel.: +82-312012633
| |
Collapse
|
7
|
The Role of Glycoside Hydrolases in S. gordonii and C. albicans Interactions. Appl Environ Microbiol 2022; 88:e0011622. [PMID: 35506689 DOI: 10.1128/aem.00116-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Candida albicans can coaggregate with Streptococcus gordonii and cocolonize in the oral cavity. Saliva provides a vital microenvironment for close interactions of oral microorganisms. However, the level of fermentable carbohydrates in saliva is not sufficient to support the growth of multiple species. Glycoside hydrolases (GHs) that hydrolyze glycoproteins are critical for S. gordonii growth in low-fermentable-carbohydrate environments such as saliva. However, whether GHs are involved in the cross-kingdom interactions between C. albicans and S. gordonii under such conditions remains unknown. In this study, C. albicans and S. gordonii were cocultured in heart infusion broth with a low level of fermentable carbohydrate. Planktonic growth, biofilm formation, cell aggregation, and GH activities of monocultures and cocultures were examined. The results revealed that the planktonic growth of cocultured S. gordonii in a low-carbohydrate environment was elevated, while that of cocultured C. albicans was reduced. The biomass of S. gordonii in dual-species biofilms was higher than that of monocultures, while that of cocultured C. albicans was decreased. GH activity was observed in S. gordonii, and elevated activity of GHs was detected in S. gordonii-C. albicans cocultures, with elevated expression of GH-related genes of S. gordonii. By screening a mutant library of C. albicans, we identified a tec1Δ/Δ mutant strain that showed reduced ability to promote the growth and GH activities of S. gordonii compared with the wild-type strain. Altogether, the findings of this study demonstrate the involvement of GHs in the cross-kingdom metabolic interactions between C. albicans and S. gordonii in an environment with low level of fermentable carbohydrates. IMPORTANCE Cross-kingdom interactions between Candida albicans and oral streptococci such as Streptococcus gordonii have been reported. However, their interactions in a low-fermentable-carbohydrate environment like saliva is not clear. The current study revealed glycoside hydrolase-related cross-kingdom communications between S. gordonii and C. albicans under the low-fermentable-carbohydrate condition. We demonstrate that C. albicans can promote the growth and metabolic activities of S. gordonii by elevating the activities of cell-wall-anchored glycoside hydrolases of S. gordonii. C. albicans gene TEC1 is critical for this cross-kingdom metabolic communication.
Collapse
|
8
|
Adenophora triphylla var. japonica Inhibits Candida Biofilm Formation, Increases Susceptibility to Antifungal Agents and Reduces Infection. Int J Mol Sci 2021; 22:ijms222212523. [PMID: 34830415 PMCID: PMC8624521 DOI: 10.3390/ijms222212523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/31/2021] [Accepted: 11/18/2021] [Indexed: 12/04/2022] Open
Abstract
(1) Background: Candida is the most common cause of fungal infections worldwide, but due to the limited option of antifungal therapies, alternative strategies are required. (2) Methods: Adenophora triphylla var. japonica extract was used for the biofilm formation assay using RPMI1640. The combinatorial antifungal assay, the dimorphic transition assay, and the adherence assay were done to see the influence of inhibition of biofilm formation. qRT-PCR analysis were performed to check the gene expression. (3) Results: Adenophora triphylla var. japonica extract inhibited the Candida biofilm formation. Treatment of extract increased the antifungal susceptibility of miconazole from a 37% reduction in fungal growth to 99.05%, and also dose-dependently reduced the dimorphic transition of Candida and the attachment of Candida to HaCaT cells. The extract blocked the expression of hyphal-related genes, extracellular matrix genes, Ras1-cAMP-PKA pathway genes, Cph2-Tec1 pathway gene, and MAP kinase pathway gene. (4) Conclusions: In this study, the treatment of Adenophora triphylla var. japonica extract showed inhibition of fungal biofilm formation, activation of antifungal susceptibility, and reduction of infection. These results suggest that fungal biofilm formation is a good target for the development of antifungal adjuvants, and Adenophora triphylla var. japonica extract should be a good candidate for biofilm-associated fungal infections.
Collapse
|
9
|
Osés-Ruiz M, Cruz-Mireles N, Martin-Urdiroz M, Soanes DM, Eseola AB, Tang B, Derbyshire P, Nielsen M, Cheema J, Were V, Eisermann I, Kershaw MJ, Yan X, Valdovinos-Ponce G, Molinari C, Littlejohn GR, Valent B, Menke FLH, Talbot NJ. Appressorium-mediated plant infection by Magnaporthe oryzae is regulated by a Pmk1-dependent hierarchical transcriptional network. Nat Microbiol 2021; 6:1383-1397. [PMID: 34707224 DOI: 10.1038/s41564-021-00978-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 09/09/2021] [Indexed: 01/18/2023]
Abstract
Rice blast is a devastating disease caused by the fungal pathogen Magnaporthe oryzae that threatens rice production around the world. The fungus produces a specialized infection cell, called the appressorium, that enables penetration through the plant cell wall in response to surface signals from the rice leaf. The underlying biology of plant infection, including the regulation of appressorium formation, is not completely understood. Here we report the identification of a network of temporally coregulated transcription factors that act downstream of the Pmk1 mitogen-activated protein kinase pathway to regulate gene expression during appressorium-mediated plant infection. We show that this tiered regulatory mechanism involves Pmk1-dependent phosphorylation of the Hox7 homeobox transcription factor, which regulates genes associated with induction of major physiological changes required for appressorium development-including cell-cycle control, autophagic cell death, turgor generation and melanin biosynthesis-as well as controlling a additional set of virulence-associated transcription factor-encoding genes. Pmk1-dependent phosphorylation of Mst12 then regulates gene functions involved in septin-dependent cytoskeletal re-organization, polarized exocytosis and effector gene expression, which are necessary for plant tissue invasion. Identification of this regulatory cascade provides new potential targets for disease intervention.
Collapse
Affiliation(s)
- Míriam Osés-Ruiz
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK.
| | - Neftaly Cruz-Mireles
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | | | | | - Alice Bisola Eseola
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Bozeng Tang
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Paul Derbyshire
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | | | | | - Vincent Were
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Iris Eisermann
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | | | - Xia Yan
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Guadalupe Valdovinos-Ponce
- Department of Plant Pathology, Kansas State University, Manhattan, KS, USA.,Department of Plant Pathology, Colegio de Postgraduados, Montecillo, Texcoco, Mexico
| | - Camilla Molinari
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - George R Littlejohn
- School of Biosciences, University of Exeter, Exeter, UK.,Department of Biological and Marine Sciences, University of Plymouth, Drakes Circus, Plymouth, UK
| | - Barbara Valent
- Department of Plant Pathology, Kansas State University, Manhattan, KS, USA
| | - Frank L H Menke
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Nicholas J Talbot
- The Sainsbury Laboratory, Norwich Research Park, University of East Anglia, Norwich, UK.
| |
Collapse
|
10
|
Hedera rhombea inhibits the biofilm formation of Candida, thereby increases the susceptibility to antifungal agent, and reduces infection. PLoS One 2021; 16:e0258108. [PMID: 34614005 PMCID: PMC8494327 DOI: 10.1371/journal.pone.0258108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/17/2021] [Indexed: 12/27/2022] Open
Abstract
Candida is an opportunistic pathogen and a common cause of fungal infections worldwide. Anti-fungal use against Candida infections has resulted in the appearance of resistant strains. The limited choice of anti-fungal therapy means alternative strategies are needed to control fungal infectious diseases. The aim of this study was to evaluate the inhibition of Candida biofilm formation by Hedera rhombea (Korean name: songak) extract. Biofilm formation was assessed using the crystal violet assay which showed a dose dependent reduction in the presence of extract with the biofilm formation inhibitory concentration of C. albicans (IC50 = 12.5μg/ml), C. tropicalis var. tropicalis (IC50 = 25μg/ml), C. parapsilosis var. parapsilosis (IC50 = 6.25μg/ml), C. glabrata (IC50 = 6.25μg/ml), C. tropicalis (IC50 = 12.5μg/ml), and C. parapsilosis (IC50 = 12.5μg/ml) without directly reducing Candida growth. Treatment with 6.25μg/mL of extract increased the antifungal susceptibility to miconazole from 32% decreasing of fungal growth to 98.8% of that based on the fungal growth assay. Treatment of extract dose-dependently reduced the dimorphic transition of Candida based on the dimorphic transition assay and treatment of 3.125μg/mL of extract completely blocked the adherence of Candida to the HaCaT cells. To know the molecular mechanisms of biofilm formation inhibition by extract, qRT-PCR analysis was done, and the extract was found to dose dependently reduce the expression of hyphal-associated genes (ALS3, ECE1, HWP1, PGA50, and PBR1), extracellular matrix genes (GSC1, ZAP1, ADH5, and CSH1), Ras1-cAMP-PKA pathway genes (CYR1, EFG1, and RAS1), Cph2-Tec1 pathway gene (TEC1) and MAP kinases pathway gene (HST7). In this study, Hedera rhombea extract showed inhibition of fungal biofilm formation, activation of antifungal susceptibility, and reduction of infection. These results suggest that fungal biofilm formation is good screen for developing the antifungal adjuvant and Hedera rhombea extract should be a good candidate against biofilm-related fungal infection.
Collapse
|
11
|
Rai LS, Wijlick LV, Bougnoux ME, Bachellier-Bassi S, d'Enfert C. Regulators of commensal and pathogenic life-styles of an opportunistic fungus-Candida albicans. Yeast 2021; 38:243-250. [PMID: 33533498 DOI: 10.1002/yea.3550] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
The yeast Candida albicans is primarily a commensal of humans that colonizes the mucosal surfaces of the gastrointestinal and genital tracts. Yet, C. albicans can under certain circumstances undergo a shift from commensalism to pathogenicity. This transition is governed by fungal factors such as morphological transitions, environmental cues for instance relationships with gut microbiota and the host immune system. C. albicans utilizes distinct sets of regulatory programs to colonize or infect its host and to evade the host defense systems. Moreover, an orchestrated iron acquisition mechanism operates to adapt to specific niches with variable iron availability. Studies on regulatory networks and morphogenesis of these two distinct modes of C. albicans growth, suggest that both yeast and hyphal forms exist in both growth patterns and the regulatory circuits are inter-connected. Here, we summarize current knowledge about C. albicans commensal-to-pathogen shift, its regulatory elements and their contribution to human disease.
Collapse
Affiliation(s)
- Laxmi Shanker Rai
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, USC2019 INRA, Paris, France
| | - Lasse Van Wijlick
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, USC2019 INRA, Paris, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, USC2019 INRA, Paris, France.,Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Hôpital Necker-Enfants-Malades, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | | | - Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, USC2019 INRA, Paris, France
| |
Collapse
|
12
|
Qasim MN, Valle Arevalo A, Nobile CJ, Hernday AD. The Roles of Chromatin Accessibility in Regulating the Candida albicans White-Opaque Phenotypic Switch. J Fungi (Basel) 2021; 7:37. [PMID: 33435404 PMCID: PMC7826875 DOI: 10.3390/jof7010037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans, a diploid polymorphic fungus, has evolved a unique heritable epigenetic program that enables reversible phenotypic switching between two cell types, referred to as "white" and "opaque". These cell types are established and maintained by distinct transcriptional programs that lead to differences in metabolic preferences, mating competencies, cellular morphologies, responses to environmental signals, interactions with the host innate immune system, and expression of approximately 20% of genes in the genome. Transcription factors (defined as sequence specific DNA-binding proteins) that regulate the establishment and heritable maintenance of the white and opaque cell types have been a primary focus of investigation in the field; however, other factors that impact chromatin accessibility, such as histone modifying enzymes, chromatin remodelers, and histone chaperone complexes, also modulate the dynamics of the white-opaque switch and have been much less studied to date. Overall, the white-opaque switch represents an attractive and relatively "simple" model system for understanding the logic and regulatory mechanisms by which heritable cell fate decisions are determined in higher eukaryotes. Here we review recent discoveries on the roles of chromatin accessibility in regulating the C. albicans white-opaque phenotypic switch.
Collapse
Affiliation(s)
- Mohammad N. Qasim
- Department of Molecular and Cell Biology, University of California-Merced, Merced, CA 95343, USA; (M.N.Q.); (A.V.A.); (C.J.N.)
- Quantitative and Systems Biology Graduate Program, University of California-Merced, Merced, CA 95343, USA
| | - Ashley Valle Arevalo
- Department of Molecular and Cell Biology, University of California-Merced, Merced, CA 95343, USA; (M.N.Q.); (A.V.A.); (C.J.N.)
- Quantitative and Systems Biology Graduate Program, University of California-Merced, Merced, CA 95343, USA
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, University of California-Merced, Merced, CA 95343, USA; (M.N.Q.); (A.V.A.); (C.J.N.)
- Health Sciences Research Institute, University of California-Merced, Merced, CA 95343, USA
| | - Aaron D. Hernday
- Department of Molecular and Cell Biology, University of California-Merced, Merced, CA 95343, USA; (M.N.Q.); (A.V.A.); (C.J.N.)
- Health Sciences Research Institute, University of California-Merced, Merced, CA 95343, USA
| |
Collapse
|
13
|
Mba IE, Nweze EI. Mechanism of Candida pathogenesis: revisiting the vital drivers. Eur J Clin Microbiol Infect Dis 2020; 39:1797-1819. [PMID: 32372128 DOI: 10.1007/s10096-020-03912-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Candida is the most implicated fungal pathogen in the clinical setting. Several factors play important roles in the pathogenesis of Candida spp. Multiple transcriptional circuits, morphological and phenotypic switching, biofilm formation, tissue damaging extracellular hydrolytic enzymes, metabolic flexibility, genome plasticity, adaptation to environmental pH fluctuation, robust nutrient acquisition system, adherence and invasions (mediated by adhesins and invasins), heat shock proteins (HSPs), cytolytic proteins, escape from phagocytosis, evasion from host immune system, synergistic coaggregation with resident microbiota, resistance to antifungal agents, and the ability to efficiently respond to multiple stresses are some of the major pathogenic determinants of Candida species. The existence of multiple connections, in addition to the interactions and associations among all of these factors, are distinctive features that play important roles in the establishment of Candida infections. This review describes all the underlying factors and mechanisms involved in Candida pathogenesis by evaluating pathogenic determinants of Candida species. It reinforces the already available pool of data on the pathogenesis of Candida species by providing a clear and simplified understanding of the most important factors implicated in the pathogenesis of Candida species. The Candida pathogenesis network, an illustration linking all the major determinants of Candida pathogenesis, is also presented. Taken together, they will further improve our current understanding of how these factors modulate virulence and consequent infection(s). Development of new antifungal drugs and better therapeutic approaches to candidiasis can be achieved in the near future with continuing progress in the understanding of the mechanisms of Candida pathogenesis.
Collapse
|
14
|
Abstract
The ability for cells to maintain homeostasis in the presence of extracellular stress is essential for their survival. Stress adaptations are especially important for microbial pathogens to respond to rapidly changing conditions, such as those encountered during the transition from the environment to the infected host. Many fungal pathogens have acquired the ability to quickly adapt to changes in extracellular pH to promote their survival in the various microenvironments encountered during a host infection. For example, the fungus-specific Rim/Pal alkaline response pathway has been well characterized in many fungal pathogens, including Cryptococcus neoformans However, alternative mechanisms for sensing and responding to host pH have yet to be extensively studied. Recent observations from a genetic screen suggest that the C. neoformans sterol homeostasis pathway is required for growth at elevated pH. This work explores interactions among mechanisms of membrane homeostasis, alkaline pH tolerance, and Rim pathway activation. We find that the sterol homeostasis pathway is necessary for growth in an alkaline environment and that an elevated pH is sufficient to induce Sre1 activation. This pH-mediated activation of the Sre1 transcription factor is linked to the biosynthesis of ergosterol but is not dependent on Rim pathway signaling, suggesting that these two pathways are responding to alkaline pH independently. Furthermore, we discover that C. neoformans is more susceptible to membrane-targeting antifungals under alkaline conditions, highlighting the impact of microenvironmental pH on the treatment of invasive fungal infections. Together, these findings further connect membrane integrity and composition with the fungal pH response and pathogenesis.IMPORTANCE The work described here further elucidates how microorganisms sense and adapt to changes in their environment to establish infections in the human host. Specifically, we uncover a novel mechanism by which an opportunistic human fungal pathogen, Cryptococcus neoformans, responds to increases in extracellular pH in order to survive and thrive within the relatively alkaline environment of the human lung. This mechanism, which is intimately linked with fungal membrane sterol homeostasis, is independent of the previously well-studied alkaline response Rim pathway. Furthermore, this ergosterol-dependent alkaline pH response is present in Candida albicans, indicating that this mechanism spans diverse fungal species. These results are also relevant for novel antimicrobial drug development as we show that currently used ergosterol-targeting antifungals are more active in alkaline environments.
Collapse
|
15
|
Han G, Liu N, Li C, Tu J, Li Z, Sheng C. Discovery of Novel Fungal Lanosterol 14α-Demethylase (CYP51)/Histone Deacetylase Dual Inhibitors to Treat Azole-Resistant Candidiasis. J Med Chem 2020; 63:5341-5359. [PMID: 32347094 DOI: 10.1021/acs.jmedchem.0c00102] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Invasive fungal infections (particularly candidiasis) are emerging as severe infectious diseases worldwide. Because of serious antifungal drug resistance, therapeutic efficacy of the current treatment for candidiasis is limited and associated with high mortality. However, it is highly challenging to develop novel strategies and effective therapeutic agents to combat drug resistance. Herein, the first generation of lanosterol 14α-demethylase (CYP51)-histone deacetylase (HDAC) dual inhibitors was designed, which exhibited potent antifungal activity against azole-resistant clinical isolates. In particular, compounds 12h and 15j were highly active both in vitro and in vivo to treat azole-resistant candidiasis. Antifungal mechanism studies revealed that they acted by blocking ergosterol biosynthesis and HDAC catalytic activity in fungus, suppressing the function of efflux pump, yeast-to-hypha morphological transition, and biofilm formation. Therefore, CYP51-HDAC dual inhibitors represent a promising strategy to develop novel antifungal agents against azole-resistant candidiasis.
Collapse
Affiliation(s)
- Guiyan Han
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Na Liu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chenglan Li
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Jie Tu
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhuang Li
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
16
|
Polonio Á, Seoane P, Claros MG, Pérez-García A. The haustorial transcriptome of the cucurbit pathogen Podosphaera xanthii reveals new insights into the biotrophy and pathogenesis of powdery mildew fungi. BMC Genomics 2019; 20:543. [PMID: 31272366 PMCID: PMC6611051 DOI: 10.1186/s12864-019-5938-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022] Open
Abstract
Background Podosphaera xanthii is the main causal agent of powdery mildew disease in cucurbits and is responsible for important yield losses in these crops worldwide. Powdery mildew fungi are obligate biotrophs. In these parasites, biotrophy is determined by the presence of haustoria, which are specialized structures of parasitism developed by these fungi for the acquisition of nutrients and the delivery of effectors. Detailed molecular studies of powdery mildew haustoria are scarce due mainly to difficulties in their isolation. Therefore, their analysis is considered an important challenge for powdery mildew research. The aim of this work was to gain insights into powdery mildew biology by analysing the haustorial transcriptome of P. xanthii. Results Prior to RNA isolation and massive-scale mRNA sequencing, a flow cytometric approach was developed to isolate P. xanthii haustoria free of visible contaminants. Next, several commercial kits were used to isolate total RNA and to construct the cDNA and Illumina libraries that were finally sequenced by the Illumina NextSeq system. Using this approach, the maximum amount of information from low-quality RNA that could be obtained was used to accomplish the de novo assembly of the P. xanthii haustorial transcriptome. The subsequent analysis of this transcriptome and comparison with the epiphytic transcriptome allowed us to identify the importance of several biological processes for haustorial cells such as protection against reactive oxygen species, the acquisition of different nutrients and genetic regulation mediated by non-coding RNAs. In addition, we could also identify several secreted proteins expressed exclusively in haustoria such as cell adhesion proteins that have not been related to powdery mildew biology to date. Conclusions This work provides a novel approach to study the molecular aspects of powdery mildew haustoria. In addition, the results of this study have also allowed us to identify certain previously unknown processes and proteins involved in the biology of powdery mildews that could be essential for their biotrophy and pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12864-019-5938-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Álvaro Polonio
- Departamento de Microbiología, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur 31, 29071, Málaga, Spain.,Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga, Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Bulevar Louis Pasteur 31, 29071, Málaga, Spain
| | - Pedro Seoane
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur 31, 29071, Málaga, Spain
| | - M Gonzalo Claros
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur 31, 29071, Málaga, Spain
| | - Alejandro Pérez-García
- Departamento de Microbiología, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur 31, 29071, Málaga, Spain. .,Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga, Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Bulevar Louis Pasteur 31, 29071, Málaga, Spain.
| |
Collapse
|
17
|
Fonseca D, Leal-Pinto SM, Roa-Cordero MV, Vargas JD, Moreno-Moreno EM, Macías MA, Suescun L, Muñoz-Castro Á, Hurtado JJ. Inhibition of C. albicans Dimorphic Switch by Cobalt(II) Complexes with Ligands Derived from Pyrazoles and Dinitrobenzoate: Synthesis, Characterization and Biological Activity. Int J Mol Sci 2019; 20:E3237. [PMID: 31266213 PMCID: PMC6651002 DOI: 10.3390/ijms20133237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 01/27/2023] Open
Abstract
Seven cobalt(II) complexes of pyrazole derivatives and dinitrobenzoate ligands were synthesized and characterized. The single-crystal X-ray diffraction structure was determined for one of the ligands and one of the complexes. The analysis and spectral data showed that all the cobalt complexes had octahedral geometries, which was supported by DFT calculations. The complexes and their free ligands were evaluated against fungal strains of Candida albicans and emerging non-albicans species and epimastigotes of Trypanosoma cruzi. We obtained antifungal activity with a minimum inhibitory concentration (MIC) ranging from 31.3 to 250 µg mL-1. The complexes were more active against C. krusei, showing MIC values between 31.25 and 62.5 µg mL-1. In addition, some ligands (L1-L6) and complexes (5 and Co(OAc)2 · 4H2O) significantly reduced the yeast to hypha transition of C. albicans at 500 µg mL-1 (inhibition ranging from 30 to 54%). Finally, the complexes and ligands did not present trypanocidal activity and were not toxic to Vero cells. Our results suggest that complexes of cobalt(II) with ligands derived from pyrazoles and dinitrobenzoate may be an attractive alternative for the treatment of diseases caused by fungi, especially because they target one of the most important virulence factors of C. albicans.
Collapse
Affiliation(s)
- Daniela Fonseca
- Department of Chemistry, Universidad de los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia
| | - Sandra M Leal-Pinto
- Grupo de Investigación en Manejo Clínico-CLINIUDES, Facultad de Ciencias de la Salud, Universidad de Santander, 680002 Bucaramanga, Colombia
| | - Martha V Roa-Cordero
- Grupo de Investigación en Manejo Clínico-CLINIUDES, Facultad de Ciencias de la Salud, Universidad de Santander, 680002 Bucaramanga, Colombia
| | - José D Vargas
- Grupo de Investigación en Manejo Clínico-CLINIUDES, Facultad de Ciencias de la Salud, Universidad de Santander, 680002 Bucaramanga, Colombia
| | - Erika M Moreno-Moreno
- Grupo de Investigación en Biotecnología Agroambiente y Salud-MICROBIOTA, Facultad de Ciencias de la Salud, Universidad de Santander, 680002 Bucaramanga, Colombia
| | - Mario A Macías
- Department of Chemistry, Universidad de los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia
| | - Leopoldo Suescun
- Cryssmat-Lab, DETEMA, Facultad de Química, Universidad de la República, Av. 18 de Julio 1824-1850, 11200 Montevideo, Uruguay
| | - Álvaro Muñoz-Castro
- Grupo de Química Inorgánica y Materiales Moleculares, Universidad Autónoma de Chile, El Llano Subercaseaux, Santiago 2801, Chile
| | - John J Hurtado
- Department of Chemistry, Universidad de los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia.
| |
Collapse
|
18
|
Regulation of Candida albicans Hyphal Morphogenesis by Endogenous Signals. J Fungi (Basel) 2019; 5:jof5010021. [PMID: 30823468 PMCID: PMC6463138 DOI: 10.3390/jof5010021] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/15/2022] Open
Abstract
Candida albicans is a human commensal fungus that is able to assume several morphologies, including yeast, hyphal, and pseudohyphal. Under a range of conditions, C. albicans performs a regulated switch to the filamentous morphology, characterized by the emergence of a germ tube from the yeast cell, followed by a mold-like growth of branching hyphae. This transition from yeast to hyphal growth has attracted particular attention, as it has been linked to the virulence of C. albicans as an opportunistic human pathogen. Signal transduction pathways that mediate the induction of the hyphal transcription program upon the imposition of external stimuli have been extensively investigated. However, the hyphal morphogenesis transcription program can also be induced by internal cellular signals, such as inhibition of cell cycle progression, and conversely, the inhibition of hyphal extension can repress hyphal-specific gene expression, suggesting that endogenous cellular signals are able to modulate hyphal gene expression as well. Here we review recent developments in the regulation of the hyphal morphogenesis of C. albicans, with emphasis on endogenous morphogenetic signals.
Collapse
|
19
|
del Olmo Toledo V, Puccinelli R, Fordyce PM, Pérez JC. Diversification of DNA binding specificities enabled SREBP transcription regulators to expand the repertoire of cellular functions that they govern in fungi. PLoS Genet 2018; 14:e1007884. [PMID: 30596634 PMCID: PMC6329520 DOI: 10.1371/journal.pgen.1007884] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 01/11/2019] [Accepted: 12/08/2018] [Indexed: 01/08/2023] Open
Abstract
The Sterol Regulatory Element Binding Proteins (SREBPs) are basic-helix-loop-helix transcription regulators that control the expression of sterol biosynthesis genes in higher eukaryotes and some fungi. Surprisingly, SREBPs do not regulate sterol biosynthesis in the ascomycete yeasts (Saccharomycotina) as this role was handed off to an unrelated transcription regulator in this clade. The SREBPs, nonetheless, expanded in fungi such as the ascomycete yeasts Candida spp., raising questions about their role and evolution in these organisms. Here we report that the fungal SREBPs diversified their DNA binding preferences concomitantly with an expansion in function. We establish that several branches of fungal SREBPs preferentially bind non-palindromic DNA sequences, in contrast to the palindromic DNA motifs recognized by most basic-helix-loop-helix proteins (including SREBPs) in higher eukaryotes. Reconstruction and biochemical characterization of the likely ancestor protein suggest that an intrinsic DNA binding promiscuity in the family was resolved by alternative mechanisms in different branches of fungal SREBPs. Furthermore, we show that two SREBPs in the human commensal yeast Candida albicans drive a transcriptional cascade that inhibits a morphological switch under anaerobic conditions. Preventing this morphological transition enhances C. albicans colonization of the mammalian intestine, the fungus' natural niche. Thus, our results illustrate how diversification in DNA binding preferences enabled the functional expansion of a family of eukaryotic transcription regulators.
Collapse
Affiliation(s)
- Valentina del Olmo Toledo
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg, Würzburg, Germany
- Institute for Molecular Infection Biology, University Würzburg, Würzburg, Germany
| | - Robert Puccinelli
- Department of Genetics, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Polly M. Fordyce
- Department of Genetics, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Stanford CheM-H Institute, Stanford University, Stanford, California, United States of America
| | - J. Christian Pérez
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg, Würzburg, Germany
- Institute for Molecular Infection Biology, University Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
20
|
Ghorai P, Irfan M, Narula A, Datta A. A comprehensive analysis of Candida albicans phosphoproteome reveals dynamic changes in phosphoprotein abundance during hyphal morphogenesis. Appl Microbiol Biotechnol 2018; 102:9731-9743. [PMID: 30121747 DOI: 10.1007/s00253-018-9303-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/18/2022]
Abstract
The morphological plasticity of Candida albicans is a virulence determinant as the hyphal form has significant roles in the infection process. Recently, phosphoregulation of proteins through phosphorylation and dephosphorylation events has gained importance in studying the regulation of pathogenicity at the molecular level. To understand the importance of phosphorylation in hyphal morphogenesis, global analysis of the phosphoproteome was performed after hyphal induction with elevated temperature, serum, and N-acetyl-glucosamine (GlcNAc) treatments. The study identified 60, 20, and 53 phosphoproteins unique to elevated temperature-, serum-, and GlcNAc-treated conditions, respectively. Distribution of unique phosphorylation sites sorted by the modified amino acids revealed that predominant phosphorylation occurs in serine, followed by threonine and tyrosine residues in all the datasets. However, the frequency distribution of phosphorylation sites in the proteins varied with treatment conditions. Further, interaction network-based functional annotation of protein kinases of C. albicans as well as identified phosphoproteins was performed, which demonstrated the interaction of kinases with phosphoproteins during filamentous growth. Altogether, the present findings will serve as a base for further functional studies in the aspects of protein kinase-target protein interaction in effectuating phosphorylation of target proteins, and delineating the downstream signaling networks linked to virulence characteristics of C. albicans.
Collapse
Affiliation(s)
- Priyanka Ghorai
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India.,Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Mohammad Irfan
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Alka Narula
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Asis Datta
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
21
|
Chang P, Wang W, Igarashi Y, Luo F, Chen J. Efficient vector systems for economical and rapid epitope-tagging and overexpression in Candida albicans. J Microbiol Methods 2018; 149:14-19. [PMID: 29698691 DOI: 10.1016/j.mimet.2018.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 11/17/2022]
Abstract
Candida albicans is an opportunistic pathogenic fungus which causes superficial and systemic infections in immunocompromised patients. It is important to characterize the roles of genes involved in its pathogenesis, virulence, and drug resistance. Several genetic manipulation toolkits have been developed for gene function research in C. albicans. Here, we describe efficient vector systems that allow economical and rapid C-terminal and N-terminal epitope-tagging, inducible and constitutive promoter replacements, and ectopic gene overexpression in C. albicans. These systems use modularized genetic elements (conventional and non-conventional selection markers, epitope tags and promoters) and universal primers. These advantages should greatly reduce laboratory work and costs of strain construction for C. albicans.
Collapse
Affiliation(s)
- Peng Chang
- Research Center of Bioenergy & Bioremediation, College of Resources and Environment, Southwest University, Chongqing 400715, China.
| | - Wenjuan Wang
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yasuo Igarashi
- Research Center of Bioenergy & Bioremediation, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Feng Luo
- Research Center of Bioenergy & Bioremediation, College of Resources and Environment, Southwest University, Chongqing 400715, China
| | - Jiangye Chen
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
22
|
Cavalheiro M, Teixeira MC. Candida Biofilms: Threats, Challenges, and Promising Strategies. Front Med (Lausanne) 2018; 5:28. [PMID: 29487851 PMCID: PMC5816785 DOI: 10.3389/fmed.2018.00028] [Citation(s) in RCA: 418] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/26/2018] [Indexed: 12/19/2022] Open
Abstract
Candida species are fungal pathogens known for their ability to cause superficial and systemic infections in the human host. These pathogens are able to persist inside the host due to the development of pathogenicity and multidrug resistance traits, often leading to the failure of therapeutic strategies. One specific feature of Candida species pathogenicity is their ability to form biofilms, which protects them from external factors such as host immune system defenses and antifungal drugs. This review focuses on the current threats and challenges when dealing with biofilms formed by Candida albicans, Candida glabrata, Candida tropicalis, and Candida parapsilosis, highlighting the differences between the four species. Biofilm characteristics depend on the ability of each species to produce extracellular polymeric substances (EPS) and display dimorphic growth, but also on the biofilm substratum, carbon source availability and other factors. Additionally, the transcriptional control over processes like adhesion, biofilm formation, filamentation, and EPS production displays great complexity and diversity within pathogenic yeasts of the Candida genus. These differences not only have implications in the persistence of colonization and infections but also on antifungal resistance typically found in Candida biofilm cells, potentiated by EPS, that functions as a barrier to drug diffusion, and by the overexpression of drug resistance transporters. The ability to interact with different species in in vivo Candida biofilms is also a key factor to consider when dealing with this problem. Despite many challenges, the most promising strategies that are currently available or under development to limit biofilm formation or to eradicate mature biofilms are discussed.
Collapse
Affiliation(s)
- Mafalda Cavalheiro
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,iBB - Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Miguel Cacho Teixeira
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,iBB - Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
23
|
Chen M, Zhang J, Sampieri K, Clohessy JG, Mendez L, Gonzalez-Billalabeitia E, Liu XS, Lee YR, Fung J, Katon JM, Menon AV, Webster KA, Ng C, Palumbieri MD, Diolombi MS, Breitkopf SB, Teruya-Feldstein J, Signoretti S, Bronson RT, Asara JM, Castillo-Martin M, Cordon-Cardo C, Pandolfi PP. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat Genet 2018; 50:206-218. [PMID: 29335545 DOI: 10.1038/s41588-017-0027-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/01/2017] [Indexed: 12/15/2022]
Abstract
Lipids, either endogenously synthesized or exogenous, have been linked to human cancer. Here we found that PML is frequently co-deleted with PTEN in metastatic human prostate cancer (CaP). We demonstrated that conditional inactivation of Pml in the mouse prostate morphs indolent Pten-null tumors into lethal metastatic disease. We identified MAPK reactivation, subsequent hyperactivation of an aberrant SREBP prometastatic lipogenic program, and a distinctive lipidomic profile as key characteristic features of metastatic Pml and Pten double-null CaP. Furthermore, targeting SREBP in vivo by fatostatin blocked both tumor growth and distant metastasis. Importantly, a high-fat diet (HFD) induced lipid accumulation in prostate tumors and was sufficient to drive metastasis in a nonmetastatic Pten-null mouse model of CaP, and an SREBP signature was highly enriched in metastatic human CaP. Thus, our findings uncover a prometastatic lipogenic program and lend direct genetic and experimental support to the notion that a Western HFD can promote metastasis.
Collapse
Affiliation(s)
- Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jiangwen Zhang
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Katia Sampieri
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,GSK Vaccines, Antigen Identification and Molecular Biology, Siena, Italy
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lourdes Mendez
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Enrique Gonzalez-Billalabeitia
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xue-Song Liu
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yu-Ru Lee
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacqueline Fung
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jesse M Katon
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Archita Venugopal Menon
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kaitlyn A Webster
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christopher Ng
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria Dilia Palumbieri
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Moussa S Diolombi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Susanne B Breitkopf
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Julie Teruya-Feldstein
- Department of Pathology, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Roderick T Bronson
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mireia Castillo-Martin
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pathology, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Basso V, d'Enfert C, Znaidi S, Bachellier-Bassi S. From Genes to Networks: The Regulatory Circuitry Controlling Candida albicans Morphogenesis. Curr Top Microbiol Immunol 2018; 422:61-99. [PMID: 30368597 DOI: 10.1007/82_2018_144] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Candida albicans is a commensal yeast of most healthy individuals, but also one of the most prevalent human fungal pathogens. During adaptation to the mammalian host, C. albicans encounters different niches where it is exposed to several types of stress, including oxidative, nitrosative (e.g., immune system), osmotic (e.g., kidney and oral cavity) stresses and pH variation (e.g., gastrointestinal (GI) tract and vagina). C. albicans has developed the capacity to respond to the environmental changes by modifying its morphology, which comprises the yeast-to-hypha transition, white-opaque switching, and chlamydospore formation. The yeast-to-hypha transition has been very well characterized and was shown to be modulated by several external stimuli that mimic the host environment. For instance, temperature above 37 ℃, serum, alkaline pH, and CO2 concentration are all reported to enhance filamentation. The transition is characterized by the activation of an intricate regulatory network of signaling pathways, involving many transcription factors. The regulatory pathways that control either the stress response or morphogenesis are required for full virulence and promote survival of C. albicans in the host. Many of these transcriptional circuitries have been characterized, highlighting the complexity and the interconnections between the different pathways. Here, we present the major signaling pathways and the main transcription factors involved in the yeast-to-hypha transition. Furthermore, we describe the role of heat shock transcription factors in the morphogenetic transition, providing an edifying example of the complex cross talk between pathways involved in morphogenesis and stress response.
Collapse
Affiliation(s)
- Virginia Basso
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 25 Rue Du Docteur Roux, 75015, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, 25 Rue Du Docteur Roux, Paris, France.,Department of Pathology and Laboratory Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 25 Rue Du Docteur Roux, 75015, Paris, France
| | - Sadri Znaidi
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 25 Rue Du Docteur Roux, 75015, Paris, France. .,Institut Pasteur de Tunis, University of Tunis El Manar, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique, 13 Place Pasteur, 1002, Tunis-Belvédère, Tunisia.
| | - Sophie Bachellier-Bassi
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, INRA, 25 Rue Du Docteur Roux, 75015, Paris, France.
| |
Collapse
|
25
|
Burr R, Espenshade PJ. Oxygen-responsive transcriptional regulation of lipid homeostasis in fungi: Implications for anti-fungal drug development. Semin Cell Dev Biol 2017; 81:110-120. [PMID: 28851600 DOI: 10.1016/j.semcdb.2017.08.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/08/2017] [Accepted: 08/22/2017] [Indexed: 01/01/2023]
Abstract
Low oxygen adaptation is essential for aerobic fungi that must survive in varied oxygen environments. Pathogenic fungi in particular must adapt to the low oxygen host tissue environment in order to cause infection. Maintenance of lipid homeostasis is especially important for cell growth and proliferation, and is a highly oxygen-dependent process. In this review, we focus on recent advances in our understanding of the transcriptional regulation and coordination of the low oxygen response across fungal species, paying particular attention to pathogenic fungi. Comparison of lipid homeostasis pathways in these organisms suggests common mechanisms of transcriptional regulation and points toward untapped potential to target low oxygen adaptation in antifungal development.
Collapse
Affiliation(s)
- Risa Burr
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peter J Espenshade
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Basso V, Znaidi S, Lagage V, Cabral V, Schoenherr F, LeibundGut-Landmann S, d'Enfert C, Bachellier-Bassi S. The two-component response regulator Skn7 belongs to a network of transcription factors regulating morphogenesis in Candida albicans and independently limits morphogenesis-induced ROS accumulation. Mol Microbiol 2017; 106:157-182. [PMID: 28752552 DOI: 10.1111/mmi.13758] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2017] [Indexed: 01/01/2023]
Abstract
Skn7 is a conserved fungal heat shock factor-type transcriptional regulator. It participates in maintaining cell wall integrity and regulates the osmotic/oxidative stress response (OSR) in S. cerevisiae, where it is part of a two-component signal transduction system. Here, we comprehensively address the function of Skn7 in the human fungal pathogen Candida albicans. We provide evidence reinforcing functional divergence, with loss of the cell wall/osmotic stress-protective roles and acquisition of the ability to regulate morphogenesis on solid medium. Mapping of the Skn7 transcriptional circuitry, through combination of genome-wide expression and location technologies, pointed to a dual regulatory role encompassing OSR and filamentous growth. Genetic interaction analyses revealed close functional interactions between Skn7 and master regulators of morphogenesis, including Efg1, Cph1 and Ume6. Intracellular biochemical assays revealed that Skn7 is crucial for limiting the accumulation of reactive oxygen species (ROS) in filament-inducing conditions on solid medium. Interestingly, functional domain mapping using site-directed mutagenesis allowed decoupling of Skn7 function in morphogenesis from protection against intracellular ROS. Our work identifies Skn7 as an integral part of the transcriptional circuitry controlling C. albicans filamentous growth and illuminates how C. albicans relies on an evolutionarily-conserved regulator to protect itself from intracellular ROS during morphological development.
Collapse
Affiliation(s)
- Virginia Basso
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, rue du Dr. Roux, Paris, France
| | - Sadri Znaidi
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France.,Institut Pasteur de Tunis, Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique, 13 Place Pasteur, Tunis-Belvédère, B.P. 74, 1002, Tunisia.,University of Tunis El Manar, Tunis 1036, Tunisia
| | - Valentine Lagage
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France
| | - Vitor Cabral
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, rue du Dr. Roux, Paris, France
| | - Franziska Schoenherr
- Institute of Virology, Winterthurerstr. 266a, Zürich, Switzerland.,SUPSI, Laboratorio Microbiologia Applicata, via Mirasole 22a, Bellinzona, Switzerland
| | | | - Christophe d'Enfert
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France
| | - Sophie Bachellier-Bassi
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, 25 rue du Docteur Roux, Paris, France
| |
Collapse
|
27
|
Kurakado S, Takatori K, Sugita T. Minocycline Inhibits Candida albicans Budded-to-Hyphal-Form Transition and Biofilm Formation. Jpn J Infect Dis 2017; 70:490-494. [PMID: 28367877 DOI: 10.7883/yoken.jjid.2016.369] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Candida albicans frequently causes bloodstream infections; its budded-to-hyphalform transition (BHT) and biofilm formation are major contributors to virulence. During an analysis of antibacterial compounds that inhibit C. albicans BHT, we found that the tetracycline derivative minocycline inhibited BHT and subsequent biofilm formation. Minocycline decreased expression of hypha-specific genes HWP1 and ECE1, and adhesion factor gene ALS3 of C. albicans. In addition, minocycline decreased cell surface hydrophobicity and the extracellular β-glucan level in biofilms. Minocycline has been widely used for catheter antibiotic lock therapy to prevent bacterial infection; this compound may also be prophylactically effective against Candida infection.
Collapse
Affiliation(s)
- Sanae Kurakado
- Department of Microbiology, Meiji Pharmaceutical University
| | - Kazuhiko Takatori
- Department of Pharmaceutical Molecular Design, Meiji Pharmaceutical University
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University
| |
Collapse
|
28
|
Morici P, Fais R, Rizzato C, Tavanti A, Lupetti A. Inhibition of Candida albicans Biofilm Formation by the Synthetic Lactoferricin Derived Peptide hLF1-11. PLoS One 2016; 11:e0167470. [PMID: 27902776 PMCID: PMC5130267 DOI: 10.1371/journal.pone.0167470] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/15/2016] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to evaluate the in vitro activity of the synthetic peptide hLF1-11 against biofilm produced by clinical isolates of Candida albicans with different fluconazole susceptibility. The antibiofilm activity of the peptide hLF1-11 was assessed in terms of reduction of biofilm cellular density, metabolic activity and sessile cell viability. The extent of morphogenesis in hLF1-11 treated and untreated biofilms was also investigated microscopically. Transcription levels of genes related to cell adhesion, hyphal development and extracellular matrix production were analysed by qRT-PCR in hLF1-11 treated and untreated biofilms. Exogenous dibutyryl-cAMP (db-cAMP) was used to rescue morphogenesis in cells exposed to the peptide. The results revealed that hLF1-11 exhibited an inhibitory effect on biofilm formation by all C. albicans isolates tested in a dose-dependent manner, regardless of their fluconazole susceptibility. Visual inspection of treated or untreated biofilm cells with an inverted microscope revealed a significant reduction in hyphal formation by hLF1-11 treated cells, as early as 3 hours of incubation. Moreover, hLF1-11 showed a reduced activity on preadherent cells. hLF1-11 induced the down-regulation of biofilm and hyphal-associated genes, which were predominantly regulated via the Ras1-cAMP-Efg1 pathway. Indeed, exogenous db-cAMP restored morphogenesis in hLF1-11 treated cells. The hLF1-11 peptide significantly inhibited biofilm formation by C. albicans mainly at early stages, interfering with biofilm cellular density and metabolic activity, and affected morphogenesis through the Ras1-cAMP-Efg1 pathway. Our findings provide the first evidence that hLF1-11 could represent a potential candidate for the prevention of biofilm formation by C. albicans.
Collapse
Affiliation(s)
- Paola Morici
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberta Fais
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Cosmeri Rizzato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Antonella Lupetti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- * E-mail:
| |
Collapse
|
29
|
Noble SM, Gianetti BA, Witchley JN. Candida albicans cell-type switching and functional plasticity in the mammalian host. Nat Rev Microbiol 2016; 15:96-108. [PMID: 27867199 DOI: 10.1038/nrmicro.2016.157] [Citation(s) in RCA: 365] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Candida albicans is a ubiquitous commensal of the mammalian microbiome and the most prevalent fungal pathogen of humans. A cell-type transition between yeast and hyphal morphologies in C. albicans was thought to underlie much of the variation in virulence observed in different host tissues. However, novel yeast-like cell morphotypes, including opaque(a/α), grey and gastrointestinally induced transition (GUT) cell types, were recently reported that exhibit marked differences in vitro and in animal models of commensalism and disease. In this Review, we explore the characteristics of the classic cell types - yeast, hyphae, pseudohyphae and chlamydospores - as well as the newly identified yeast-like morphotypes. We highlight emerging knowledge about the associations of these different morphotypes with different host niches and virulence potential, as well as the environmental cues and signalling pathways that are involved in the morphological transitions.
Collapse
Affiliation(s)
- Suzanne M Noble
- Department of Microbiology and Immunology, University of California San Francisco (UCSF) School of Medicine.,Infectious Diseases Division, Department of Medicine, University of California San Francisco (UCSF) School of Medicine, San Francisco, California 94143, USA
| | - Brittany A Gianetti
- Department of Microbiology and Immunology, University of California San Francisco (UCSF) School of Medicine
| | - Jessica N Witchley
- Department of Microbiology and Immunology, University of California San Francisco (UCSF) School of Medicine
| |
Collapse
|
30
|
Pais P, Costa C, Cavalheiro M, Romão D, Teixeira MC. Transcriptional Control of Drug Resistance, Virulence and Immune System Evasion in Pathogenic Fungi: A Cross-Species Comparison. Front Cell Infect Microbiol 2016; 6:131. [PMID: 27812511 PMCID: PMC5072224 DOI: 10.3389/fcimb.2016.00131] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/29/2016] [Indexed: 12/26/2022] Open
Abstract
Transcription factors are key players in the control of the activation or repression of gene expression programs in response to environmental stimuli. The study of regulatory networks taking place in fungal pathogens is a promising research topic that can help in the fight against these pathogens by targeting specific fungal pathways as a whole, instead of targeting more specific effectors of virulence or drug resistance. This review is focused on the analysis of regulatory networks playing a central role in the referred mechanisms in the human fungal pathogens Aspergillus fumigatus, Cryptococcus neoformans, Candida albicans, Candida glabrata, Candida parapsilosis, and Candida tropicalis. Current knowledge on the activity of the transcription factors characterized in each of these pathogenic fungal species will be addressed. Particular focus is given to their mechanisms of activation, regulatory targets and phenotypic outcome. The review further provides an evaluation on the conservation of transcriptional circuits among different fungal pathogens, highlighting the pathways that translate common or divergent traits among these species in what concerns their drug resistance, virulence and host immune evasion features. It becomes evident that the regulation of transcriptional networks is complex and presents significant variations among different fungal pathogens. Only the oxidative stress regulators Yap1 and Skn7 are conserved among all studied species; while some transcription factors, involved in nutrient homeostasis, pH adaptation, drug resistance and morphological switching are present in several, though not all species. Interestingly, in some cases not very homologous transcription factors display orthologous functions, whereas some homologous proteins have diverged in terms of their function in different species. A few cases of species specific transcription factors are also observed.
Collapse
Affiliation(s)
- Pedro Pais
- Biological Sciences Research Group, Department of Bioengineering, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal; Biological Sciences Research Group, Institute for Bioengineering and Biosciences, Instituto Superior TécnicoLisboa, Portugal
| | - Catarina Costa
- Biological Sciences Research Group, Department of Bioengineering, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal; Biological Sciences Research Group, Institute for Bioengineering and Biosciences, Instituto Superior TécnicoLisboa, Portugal
| | - Mafalda Cavalheiro
- Biological Sciences Research Group, Department of Bioengineering, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal; Biological Sciences Research Group, Institute for Bioengineering and Biosciences, Instituto Superior TécnicoLisboa, Portugal
| | - Daniela Romão
- Biological Sciences Research Group, Department of Bioengineering, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal; Biological Sciences Research Group, Institute for Bioengineering and Biosciences, Instituto Superior TécnicoLisboa, Portugal
| | - Miguel C Teixeira
- Biological Sciences Research Group, Department of Bioengineering, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal; Biological Sciences Research Group, Institute for Bioengineering and Biosciences, Instituto Superior TécnicoLisboa, Portugal
| |
Collapse
|
31
|
Araújo D, Henriques M, Silva S. Portrait of Candida Species Biofilm Regulatory Network Genes. Trends Microbiol 2016; 25:62-75. [PMID: 27717660 DOI: 10.1016/j.tim.2016.09.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/01/2016] [Accepted: 09/15/2016] [Indexed: 11/15/2022]
Abstract
Most cases of candidiasis have been attributed to Candida albicans, but Candida glabrata, Candida parapsilosis and Candida tropicalis, designated as non-C. albicans Candida (NCAC), have been identified as frequent human pathogens. Moreover, Candida biofilms are an escalating clinical problem associated with significant rates of mortality. Biofilms have distinct developmental phases, including adhesion/colonisation, maturation and dispersal, controlled by complex regulatory networks. This review discusses recent advances regarding Candida species biofilm regulatory network genes, which are key components for candidiasis.
Collapse
Affiliation(s)
- Daniela Araújo
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Mariana Henriques
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Sónia Silva
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal.
| |
Collapse
|
32
|
Hwang J, Ribbens D, Raychaudhuri S, Cairns L, Gu H, Frost A, Urban S, Espenshade PJ. A Golgi rhomboid protease Rbd2 recruits Cdc48 to cleave yeast SREBP. EMBO J 2016; 35:2332-2349. [PMID: 27655872 DOI: 10.15252/embj.201693923] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 09/06/2016] [Indexed: 12/26/2022] Open
Abstract
Hypoxic growth of fungi requires sterol regulatory element-binding protein (SREBP) transcription factors, and human opportunistic fungal pathogens require SREBP activation for virulence. Proteolytic release of fission yeast SREBPs from the membrane in response to low oxygen requires the Golgi membrane-anchored Dsc E3 ligase complex. Using genetic interaction arrays, we identified Rbd2 as a rhomboid family protease required for SREBP proteolytic processing. Rbd2 is an active, Golgi-localized protease that cleaves the transmembrane segment of the TatA rhomboid model substrate. Epistasis analysis revealed that the Dsc E3 ligase acts on SREBP prior to cleavage by Rbd2. Using APEX2 proximity biotinylation, we demonstrated that Rbd2 binds the AAA-ATPase Cdc48 through a C-terminal SHP box. Interestingly, SREBP cleavage required Rbd2 binding of Cdc48, consistent with Cdc48 acting to recruit ubiquitinylated substrates. In support of this claim, overexpressing a Cdc48-binding mutant of Rbd2 bypassed the Cdc48 requirement for SREBP cleavage, demonstrating that Cdc48 likely plays a role in SREBP recognition. In the absence of functional Rbd2, SREBP precursor is degraded by the proteasome, indicating that Rbd2 activity controls the balance between SREBP activation and degradation.
Collapse
Affiliation(s)
- Jiwon Hwang
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diedre Ribbens
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leah Cairns
- Howard Hughes Medical Institute, Department of Molecular Biology & Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - He Gu
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Siniša Urban
- Howard Hughes Medical Institute, Department of Molecular Biology & Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter J Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Chen X, Zhang R, Takada A, Iwatani S, Oka C, Kitamoto T, Kajiwara S. The role of Bgl2p in the transition to filamentous cells during biofilm formation by Candida albicans. Mycoses 2016; 60:96-103. [PMID: 27597232 DOI: 10.1111/myc.12554] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Abstract
The fungal pathogen Candida albicans undergoes a transition from yeast cells to filamentous cells that is related to its pathogenicity. The complex multicellular processes involved in biofilm formation by this fungus also include this transition. In this work, we investigated the morphological role of the Bgl2 protein (Bgl2p) in the transition to filamentous cells during biofilm formation by C. albicans. Bgl2p has been identified as a β-1, 3-glucosyltransferase, and transcription of the CaBGL2 gene is upregulated during biofilm formation. We used scanning electron microscopy to observe the microstructure of a bgl2 null mutant during biofilm formation and found a delay in the transition to filamentous cells in the premature phase (24 hours) of biofilm formation. Deletion of the CaBGL2 gene led to a decrease in the expression of CPH2 and TEC1, which encode transcription factors required for the transition to the filamentous form. These findings indicate that Bgl2p plays a role in the transition to filamentous cells during biofilm formation by C. albicans.
Collapse
Affiliation(s)
- Xinyue Chen
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Ruoyu Zhang
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Ayako Takada
- Biomaterials Analysis Division, Technical Department, Tokyo Institute of Technology, Yokohama, Japan
| | - Shun Iwatani
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Chiemi Oka
- School of Materials and Chemical Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Toshitaka Kitamoto
- School of Materials and Chemical Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Susumu Kajiwara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
34
|
Moyes DL, Wilson D, Richardson JP, Mogavero S, Tang SX, Wernecke J, Höfs S, Gratacap RL, Robbins J, Runglall M, Murciano C, Blagojevic M, Thavaraj S, Förster TM, Hebecker B, Kasper L, Vizcay G, Iancu SI, Kichik N, Häder A, Kurzai O, Luo T, Krüger T, Kniemeyer O, Cota E, Bader O, Wheeler RT, Gutsmann T, Hube B, Naglik JR. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature 2016; 532:64-8. [PMID: 27027296 PMCID: PMC4851236 DOI: 10.1038/nature17625] [Citation(s) in RCA: 637] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/26/2016] [Indexed: 01/23/2023]
Abstract
Cytolytic proteins and peptide toxins are classical virulence factors of several bacterial pathogens which disrupt epithelial barrier function, damage cells and activate or modulate host immune responses. Such toxins have not been identified previously in human pathogenic fungi. Here we identify the first, to our knowledge, fungal cytolytic peptide toxin in the opportunistic pathogen Candida albicans. This secreted toxin directly damages epithelial membranes, triggers a danger response signalling pathway and activates epithelial immunity. Membrane permeabilization is enhanced by a positive charge at the carboxy terminus of the peptide, which triggers an inward current concomitant with calcium influx. C. albicans strains lacking this toxin do not activate or damage epithelial cells and are avirulent in animal models of mucosal infection. We propose the name 'Candidalysin' for this cytolytic peptide toxin; a newly identified, critical molecular determinant of epithelial damage and host recognition of the clinically important fungus, C. albicans.
Collapse
Affiliation(s)
- David L Moyes
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Duncan Wilson
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
| | - Jonathan P Richardson
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
| | - Shirley X Tang
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Julia Wernecke
- Research Center Borstel, Division of Biophysics, D-23845 Borstel, Germany
- Deutsches Elektronen-Synchrotron DESY, D-22607 Hamburg, Germany
| | - Sarah Höfs
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
| | - Remi L Gratacap
- Department of Molecular &Biomedical Sciences, University of Maine, Orono, Maine 04469, USA
| | - Jon Robbins
- Wolfson CARD, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Manohursingh Runglall
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Celia Murciano
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Mariana Blagojevic
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Selvam Thavaraj
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Toni M Förster
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
| | - Betty Hebecker
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
- Research Group Microbial Immunology, Hans Knöll Institute, D-07745 Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
| | - Gema Vizcay
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Simona I Iancu
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Nessim Kichik
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Antje Häder
- Septomics Research Center, Hans-Knöll Institute and Friedrich Schiller University, D-07745 Jena, Germany
| | - Oliver Kurzai
- Septomics Research Center, Hans-Knöll Institute and Friedrich Schiller University, D-07745 Jena, Germany
| | - Ting Luo
- Department of Molecular and Applied Microbiology, Hans Knöll Institute, D-07745 Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Hans Knöll Institute, D-07745 Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Hans Knöll Institute, D-07745 Jena, Germany
| | - Ernesto Cota
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Oliver Bader
- Institute for Medical Microbiology, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Robert T Wheeler
- Department of Molecular &Biomedical Sciences, University of Maine, Orono, Maine 04469, USA
| | - Thomas Gutsmann
- Research Center Borstel, Division of Biophysics, D-23845 Borstel, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knöll Institute, D-07745 Jena, Germany
- Friedrich Schiller University, D-07737 Jena, Germany
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, D-07747 Jena, Germany
| | - Julian R Naglik
- Mucosal &Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| |
Collapse
|
35
|
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
|
36
|
Chang P, Fan X, Chen J. Function and subcellular localization of Gcn5, a histone acetyltransferase in Candida albicans. Fungal Genet Biol 2015; 81:132-41. [DOI: 10.1016/j.fgb.2015.01.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/14/2015] [Accepted: 01/21/2015] [Indexed: 11/26/2022]
|
37
|
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
|
38
|
ChIP-seq and in vivo transcriptome analyses of the Aspergillus fumigatus SREBP SrbA reveals a new regulator of the fungal hypoxia response and virulence. PLoS Pathog 2014; 10:e1004487. [PMID: 25375670 PMCID: PMC4223079 DOI: 10.1371/journal.ppat.1004487] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/23/2014] [Indexed: 12/21/2022] Open
Abstract
The Aspergillus fumigatus sterol regulatory element binding protein (SREBP) SrbA belongs to the basic Helix-Loop-Helix (bHLH) family of transcription factors and is crucial for antifungal drug resistance and virulence. The latter phenotype is especially striking, as loss of SrbA results in complete loss of virulence in murine models of invasive pulmonary aspergillosis (IPA). How fungal SREBPs mediate fungal virulence is unknown, though it has been suggested that lack of growth in hypoxic conditions accounts for the attenuated virulence. To further understand the role of SrbA in fungal infection site pathobiology, chromatin immunoprecipitation followed by massively parallel DNA sequencing (ChIP-seq) was used to identify genes under direct SrbA transcriptional regulation in hypoxia. These results confirmed the direct regulation of ergosterol biosynthesis and iron uptake by SrbA in hypoxia and revealed new roles for SrbA in nitrate assimilation and heme biosynthesis. Moreover, functional characterization of an SrbA target gene with sequence similarity to SrbA identified a new transcriptional regulator of the fungal hypoxia response and virulence, SrbB. SrbB co-regulates genes involved in heme biosynthesis and demethylation of C4-sterols with SrbA in hypoxic conditions. However, SrbB also has regulatory functions independent of SrbA including regulation of carbohydrate metabolism. Loss of SrbB markedly attenuates A. fumigatus virulence, and loss of both SREBPs further reduces in vivo fungal growth. These data suggest that both A. fumigatus SREBPs are critical for hypoxia adaptation and virulence and reveal new insights into SREBPs' complex role in infection site adaptation and fungal virulence. Despite improvements in diagnostics and antifungal drug treatments, mortality rates from invasive mold infections remain high. Defining the fungal adaptation and growth mechanisms at the infection site microenvironment is one research focus that is expected to improve treatment of established invasive fungal infections. The Aspergillus fumigatus transcription factor SrbA is a major regulator of the fungal response to hypoxia found at sites of invasive fungal growth in vivo. In this study, new insights into how SrbA mediates hypoxia adaptation and virulence were revealed through identification of direct transcriptional targets of SrbA under hypoxic conditions. A major novel finding from these studies is the identification of a critical role in fungal hypoxia adaptation and virulence of an SrbA target gene, srbB, which is also in the SREBP family. SrbB plays a major role in regulation of heme biosynthesis and carbohydrate metabolism early in the response to hypoxia. The discovery of SrbA-dependent regulation of srbB gene expression, and the target genes they regulate opens new avenues to understand how SREBPs and their target genes mediate adaptation to the in vivo infection site microenvironment and responses to current antifungal therapies.
Collapse
|
39
|
Lu Y, Su C, Liu H. Candida albicans hyphal initiation and elongation. Trends Microbiol 2014; 22:707-14. [PMID: 25262420 DOI: 10.1016/j.tim.2014.09.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 01/01/2023]
Abstract
The fungus Candida albicans is a benign member of the mucosal microbiota, but can cause mucosal infections and life-threatening disseminated invasive infections in susceptible individuals. The ability to switch between yeast, pseudohyphal, and hyphal growth forms (polymorphism) is one of the most investigated virulence attributes of C. albicans. Recent studies suggest that hyphal development in C. albicans requires two temporally linked regulations for initiation and maintenance of the hyphal transcriptional program. Hyphal initiation requires a rapid but temporary disappearance of the Nrg1 transcriptional repressor of hyphal morphogenesis. Hyphal maintenance requires active sensing of the surrounding environment, leading to exclusion of Nrg1 binding to promoters of hypha-specific genes or reduced NRG1 expression. We discuss recent advances in understanding the complex transcriptional regulation of hyphal gene expression. These provide molecular mechanisms underpinning the phenotypic plasticity of C. albicans polymorphism.
Collapse
Affiliation(s)
- Yang Lu
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
| | - Chang Su
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
40
|
Holland LM, Schröder MS, Turner SA, Taff H, Andes D, Grózer Z, Gácser A, Ames L, Haynes K, Higgins DG, Butler G. Comparative phenotypic analysis of the major fungal pathogens Candida parapsilosis and Candida albicans. PLoS Pathog 2014; 10:e1004365. [PMID: 25233198 PMCID: PMC4169492 DOI: 10.1371/journal.ppat.1004365] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/28/2014] [Indexed: 01/15/2023] Open
Abstract
Candida parapsilosis and Candida albicans are human fungal pathogens that belong to the CTG clade in the Saccharomycotina. In contrast to C. albicans, relatively little is known about the virulence properties of C. parapsilosis, a pathogen particularly associated with infections of premature neonates. We describe here the construction of C. parapsilosis strains carrying double allele deletions of 100 transcription factors, protein kinases and species-specific genes. Two independent deletions were constructed for each target gene. Growth in >40 conditions was tested, including carbon source, temperature, and the presence of antifungal drugs. The phenotypes were compared to C. albicans strains with deletions of orthologous transcription factors. We found that many phenotypes are shared between the two species, such as the role of Upc2 as a regulator of azole resistance, and of CAP1 in the oxidative stress response. Others are unique to one species. For example, Cph2 plays a role in the hypoxic response in C. parapsilosis but not in C. albicans. We found extensive divergence between the biofilm regulators of the two species. We identified seven transcription factors and one protein kinase that are required for biofilm development in C. parapsilosis. Only three (Efg1, Bcr1 and Ace2) have similar effects on C. albicans biofilms, whereas Cph2, Czf1, Gzf3 and Ume6 have major roles in C. parapsilosis only. Two transcription factors (Brg1 and Tec1) with well-characterized roles in biofilm formation in C. albicans do not have the same function in C. parapsilosis. We also compared the transcription profile of C. parapsilosis and C. albicans biofilms. Our analysis suggests the processes shared between the two species are predominantly metabolic, and that Cph2 and Bcr1 are major biofilm regulators in C. parapsilosis. Candida species are among the most common causes of fungal infection worldwide. Infections can be both community-based and hospital-acquired, and are particularly associated with immunocompromised individuals. Candida albicans is the most commonly isolated species and is the best studied. However, other species are becoming of increasing concern. Candida parapsilosis causes outbreaks of infection in neonatal wards, and is one of the few Candida species that is transferred from the hands of healthcare workers. C. parapsilosis, like C. albicans, grows as biofilms (cell communities) on the surfaces of indwelling medical devices like feeding tubes. We describe here the construction of a set of tools that allow us to characterize the virulence properties of C. parapsilosis, and in particular its ability to grow as biofilms. We find that some of the regulatory mechanisms are shared with C. albicans, but others are unique to each species. Our tools, based on selectively deleting regulatory genes, will provide a major resource to the fungal research community.
Collapse
Affiliation(s)
- Linda M. Holland
- School of Biomedical and Biomolecular Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Markus S. Schröder
- School of Biomedical and Biomolecular Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Siobhán A. Turner
- School of Biomedical and Biomolecular Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Heather Taff
- Departments of Medicine and Microbiology and Immunology, University of Wisconsin, Madison, Madison, Wisconsin, United States of America
| | - David Andes
- Departments of Medicine and Microbiology and Immunology, University of Wisconsin, Madison, Madison, Wisconsin, United States of America
| | - Zsuzsanna Grózer
- Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Attila Gácser
- Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Lauren Ames
- School of Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Ken Haynes
- School of Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Desmond G. Higgins
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Geraldine Butler
- School of Biomedical and Biomolecular Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- * E-mail:
| |
Collapse
|
41
|
Plant-derived decapeptide OSIP108 interferes with Candida albicans biofilm formation without affecting cell viability. Antimicrob Agents Chemother 2014; 58:2647-56. [PMID: 24566179 DOI: 10.1128/aac.01274-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously identified a decapeptide from the model plant Arabidopsis thaliana, OSIP108, which is induced upon fungal pathogen infection. In this study, we demonstrated that OSIP108 interferes with biofilm formation of the fungal pathogen Candida albicans without affecting the viability or growth of C. albicans cells. OSIP108 displayed no cytotoxicity against various human cell lines. Furthermore, OSIP108 enhanced the activity of the antifungal agents amphotericin B and caspofungin in vitro and in vivo in a Caenorhabditis elegans-C. albicans biofilm infection model. These data point to the potential use of OSIP108 in combination therapy with conventional antifungal agents. In a first attempt to unravel its mode of action, we screened a library of 137 homozygous C. albicans mutants, affected in genes encoding cell wall proteins or transcription factors important for biofilm formation, for altered OSIP108 sensitivity. We identified 9 OSIP108-tolerant C. albicans mutants that were defective in either components important for cell wall integrity or the yeast-to-hypha transition. In line with these findings, we demonstrated that OSIP108 activates the C. albicans cell wall integrity pathway and that its antibiofilm activity can be blocked by compounds inhibiting the yeast-to-hypha transition. Furthermore, we found that OSIP108 is predominantly localized at the C. albicans cell surface. These data point to interference of OSIP108 with cell wall-related processes of C. albicans, resulting in impaired biofilm formation.
Collapse
|
42
|
O'Meara TR, Cowen LE. Hsp90-dependent regulatory circuitry controlling temperature-dependent fungal development and virulence. Cell Microbiol 2014; 16:473-81. [PMID: 24438186 DOI: 10.1111/cmi.12266] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/09/2014] [Accepted: 01/13/2014] [Indexed: 11/28/2022]
Abstract
The pathogenic fungi Candida albicans, Aspergillus fumigatus, and Cryptococcus neoformans are an increasing cause of human mortality, especially in immunocompromised populations. During colonization and adaptation to various host environments, these fungi undergo morphogenetic alterations that allow for survival within the host. One key environmental cue driving morphological changes is external temperature. The Hsp90 chaperone protein provides one mechanism to link temperature with the signalling cascades that regulate morphogenesis, fungal development and virulence. Candida albicans is a model system for understanding the connections between morphogenesis and Hsp90. Due to the high degree of conservation in Hsp90, many of the connections in C. albicans may be extrapolated to other fungal pathogens or parasites. Examining the role of Hsp90 during development and morphogenesis in these three major fungal pathogens may provide insight into key aspects of adaptation to the host, leading to additional avenues for therapy.
Collapse
Affiliation(s)
- Teresa R O'Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
43
|
Maguire SL, Wang C, Holland LM, Brunel F, Neuvéglise C, Nicaud JM, Zavrel M, White TC, Wolfe KH, Butler G. Zinc finger transcription factors displaced SREBP proteins as the major Sterol regulators during Saccharomycotina evolution. PLoS Genet 2014; 10:e1004076. [PMID: 24453983 PMCID: PMC3894159 DOI: 10.1371/journal.pgen.1004076] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 11/18/2013] [Indexed: 12/26/2022] Open
Abstract
In most eukaryotes, including the majority of fungi, expression of sterol biosynthesis genes is regulated by Sterol-Regulatory Element Binding Proteins (SREBPs), which are basic helix-loop-helix transcription activators. However, in yeasts such as Saccharomyces cerevisiae and Candida albicans sterol synthesis is instead regulated by Upc2, an unrelated transcription factor with a Gal4-type zinc finger. The SREBPs in S. cerevisiae (Hms1) and C. albicans (Cph2) have lost a domain, are not major regulators of sterol synthesis, and instead regulate filamentous growth. We report here that rewiring of the sterol regulon, with Upc2 taking over from SREBP, likely occurred in the common ancestor of all Saccharomycotina. Yarrowia lipolytica, a deep-branching species, is the only genome known to contain intact and full-length orthologs of both SREBP (Sre1) and Upc2. Deleting YlUPC2, but not YlSRE1, confers susceptibility to azole drugs. Sterol levels are significantly reduced in the YlUPC2 deletion. RNA-seq analysis shows that hypoxic regulation of sterol synthesis genes in Y. lipolytica is predominantly mediated by Upc2. However, YlSre1 still retains a role in hypoxic regulation; growth of Y. lipolytica in hypoxic conditions is reduced in a Ylupc2 deletion and is abolished in a Ylsre1/Ylupc2 double deletion, and YlSre1 regulates sterol gene expression during hypoxia adaptation. We show that YlSRE1, and to a lesser extent YlUPC2, are required for switching from yeast to filamentous growth in hypoxia. Sre1 appears to have an ancestral role in the regulation of filamentation, which became decoupled from its role in sterol gene regulation by the arrival of Upc2 in the Saccharomycotina.
Collapse
Affiliation(s)
- Sarah L. Maguire
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Can Wang
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Linda M. Holland
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - François Brunel
- INRA UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
- CNRS, Micalis, Jouy-en-Josas, France
| | - Cécile Neuvéglise
- INRA UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
- CNRS, Micalis, Jouy-en-Josas, France
| | - Jean-Marc Nicaud
- INRA UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
- CNRS, Micalis, Jouy-en-Josas, France
| | - Martin Zavrel
- University of Missouri-Kansas City, School of Biological Sciences, Cell Biology and Biophysics, Kansas City, Missouri, United States of America
| | - Theodore C. White
- University of Missouri-Kansas City, School of Biological Sciences, Cell Biology and Biophysics, Kansas City, Missouri, United States of America
| | - Kenneth H. Wolfe
- UCD School of Medicine and Medical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Geraldine Butler
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- * E-mail:
| |
Collapse
|
44
|
Zhao LX, Li DD, Hu DD, Hu GH, Yan L, Wang Y, Jiang YY. Effect of tetrandrine against Candida albicans biofilms. PLoS One 2013; 8:e79671. [PMID: 24260276 PMCID: PMC3832530 DOI: 10.1371/journal.pone.0079671] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023] Open
Abstract
Candida albicans is the most common human fungal pathogen and has a high propensity to develop biofilms that are resistant to traditional antifungal agents. In this study, we investigated the effect of tetrandrine (TET) on growth, biofilm formation and yeast-to-hypha transition of C. albicans. We characterized the inhibitory effect of TET on hyphal growth and addressed its possible mechanism of action. Treatment of TET at a low concentration without affecting fungal growth inhibited hyphal growth in both liquid and solid Spider media. Real-time RT-PCR revealed that TET down-regulated the expression of hypha-specific genes ECE1, ALS3 and HWP1, and abrogated the induction of EFG1 and RAS1, regulators of hyphal growth. Addition of cAMP restored the normal phenotype of the SC5314 strain. These results indicate that TET may inhibit hyphal growth through the Ras1p-cAMP-PKA pathway. In vivo, at a range of concentrations from 4 mg/L to 32 mg/L, TET prolonged the survival of C. albicans-infected Caenorhabditis elegans significantly. This study provides useful information for the development of new strategies to reduce the incidence of C. albicans biofilm-associated infections.
Collapse
Affiliation(s)
- Lan-Xue Zhao
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
- Department of Pharmacy, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - De-Dong Li
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Dan-Dan Hu
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Gan-Hai Hu
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lan Yan
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yan Wang
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
- * E-mail: (YW); (YYJ)
| | - Yuan-Ying Jiang
- New Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
- * E-mail: (YW); (YYJ)
| |
Collapse
|
45
|
Fan Y, He H, Dong Y, Pan H. Hyphae-specific genes HGC1, ALS3, HWP1, and ECE1 and relevant signaling pathways in Candida albicans. Mycopathologia 2013; 176:329-35. [PMID: 24002103 DOI: 10.1007/s11046-013-9684-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 07/22/2013] [Indexed: 10/26/2022]
Abstract
Fungal virulence mechanisms include adhesion to epithelia, morphogenesis, production of secretory hydrolytic enzymes, and phenotype switching, all of which contribute to the process of pathogenesis. A striking feature of the biology of Candida albicans is its ability to grow in yeast, pseudohyphal, and hyphal forms. The hyphal form plays an important role in causing disease, by invading epithelial cells and causing tissue damage. In this review, we illustrate some of the main hyphae-specific genes, namely HGC1, UME6, ALS3, HWP1, and ECE1, and their relevant and reversed signal transduction pathways in reactions stimulated by environmental factors, including pH, CO2, and serum.
Collapse
Affiliation(s)
- Yan Fan
- Affiliated Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | | | | | | |
Collapse
|
46
|
Soll DR. The evolution of alternative biofilms in an opportunistic fungal pathogen: an explanation for how new signal transduction pathways may evolve. INFECTION GENETICS AND EVOLUTION 2013; 22:235-43. [PMID: 23871837 DOI: 10.1016/j.meegid.2013.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 10/26/2022]
Abstract
The evolution of two types of biofilms, one pathogenic and one sexual, is unique for Candidaalbicans, the most pervasive fungal pathogen in humans. When in the predominant a/α configuration, cells can form a traditional biofilm made up of a basal layer of yeast cells and an extensive upper layer of hyphae and dense matrix. This a/α biofilm is impermeable, impenetrable and drug-resistant. When in the a/a or α/α configuration, white cells form a biofilm of similar architecture, but which is permeable, penetrable and drug-susceptible. The latter biofilm facilitates mating between minority opaque a/a and α/α cells. The two biofilms are regulated by different signal transduction pathways that provide clues for deducing not only how the sexual a/a or α/α biofilms evolved, but how the pathogenic a/α biofilm evolved as well. In the deduced evolutionary models, regulatory molecules, including components of the signal transduction pathways and transcription factors, are recruited from conserved pathways. The evolution of the alternative biofilms of C. albicans provides a rare glimpse into how new regulatory pathways may evolve in general.
Collapse
Affiliation(s)
- David R Soll
- The Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
47
|
Abstract
The response of eukaryotic microbes to low-oxygen (hypoxic) conditions is strongly regulated at the level of transcription. Comparative analysis shows that some of the transcriptional regulators (such as the sterol regulatory element-binding proteins, or SREBPs) are of ancient origin and probably regulate sterol synthesis in most eukaryotic microbes. However, in some fungi SREBPs have been replaced by a zinc-finger transcription factor (Upc2). Nuclear localization of fungal SREBPs is determined by regulated proteolysis, either by site-specific proteases or by an E3 ligase complex and the proteasome. The exact mechanisms of oxygen sensing are not fully characterized but involve responding to low levels of heme and/or sterols and possibly to levels of nitric oxide and reactive oxygen species. Changes in central carbon metabolism (glycolysis and respiration) are a core hypoxic response in some, but not all, fungal species. Adaptation to hypoxia is an important virulence characteristic of pathogenic fungi.
Collapse
Affiliation(s)
- Geraldine Butler
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland;
| |
Collapse
|
48
|
Yan L, Yang C, Tang J. Disruption of the intestinal mucosal barrier in Candida albicans infections. Microbiol Res 2013; 168:389-95. [PMID: 23545353 DOI: 10.1016/j.micres.2013.02.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/15/2013] [Accepted: 02/15/2013] [Indexed: 01/05/2023]
Abstract
Candida albicans is a common microorganism in the intestine. However, invasive C. albicans infection has emerged as a life-threatening disease in recent years. The mortality rate of invasive candidiasis is high in critically ill hosts. C. albicans can switch from the yeast to the hyphal morphology, and take advantage of the impaired intestinal mucosal barrier and insufficient immunity of the host to facilitate its colonization and penetration. Despite the availability of potent new antifungal drugs in recent years, the treatment of severe candidiasis, especially candidaemia, has not been substantially improved. In this review, the virulence factors of C. albicans, as well as the antagonistic role of the intestinal mucosal barrier will be discussed to illuminate the mechanisms of C. albicans enterogenic infections.
Collapse
Affiliation(s)
- Lei Yan
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | | | | |
Collapse
|
49
|
Pérez JC, Kumamoto CA, Johnson AD. Candida albicans commensalism and pathogenicity are intertwined traits directed by a tightly knit transcriptional regulatory circuit. PLoS Biol 2013; 11:e1001510. [PMID: 23526879 PMCID: PMC3601966 DOI: 10.1371/journal.pbio.1001510] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/06/2013] [Indexed: 12/21/2022] Open
Abstract
The identification of regulators, circuits, and target genes employed by the fungus Candida albicans to thrive in disparate niches in a mammalian host reveals interconnection between commensal and pathogenic lifestyles. Systemic, life-threatening infections in humans are often caused by bacterial or fungal species that normally inhabit a different locale in our body, particularly mucosal surfaces. A hallmark of these opportunistic pathogens, therefore, is their ability to thrive in disparate niches within the host. In this work, we investigate the transcriptional circuitry and gene repertoire that enable the human opportunistic fungal pathogen Candida albicans to proliferate in two different niches. By screening a library of transcription regulator deletion strains in mouse models of intestinal colonization and systemic infection, we identified eight transcription regulators that play roles in at least one of these models. Using genome-wide chromatin immunoprecipitation, we uncovered a network comprising ∼800 target genes and a tightly knit transcriptional regulatory circuit at its core. The network is enriched with genes upregulated in C. albicans cells growing in the host. Our findings indicate that many aspects of commensalism and pathogenicity are intertwined and that the ability of this microorganism to colonize multiple niches relies on a large, integrated circuit. Our skin and mouth, as well as our genital and gastrointestinal tracts, are laden with microorganisms belonging to all three domains of life (bacteria, archaea, and eukaryotes). Much of the time these commensal microorganisms are not only harmless but provide advantages to us. However, when the host's defenses are compromised, some members of the normal flora, such as the fungus C. albicans, can cross the host's protective barriers and colonize virtually every internal organ causing life-threatening conditions. The environment found in the bloodstream and internal organs is presumably distinct from the mucosal surfaces where our flora typically resides. Whether opportunistic pathogens such as C. albicans rely on common or separate gene repertoires to thrive in each of these locales is largely unknown. To address this question we carried out genetic screens in mouse models that recapitulate niches where C. albicans thrives and used genome-wide experimental approaches to uncover the genes required to proliferate in each environment. In fact, the ability of C. albicans to colonize disparate niches within a mammalian host relies on a large, integrated circuit. Our observations suggest that at least some key gene circuits are not dedicated to one niche or another. Rather, thriving in various locales of the host seems to involve the complex regulation of multiple processes, which may allow C. albicans to adjust to different environments.
Collapse
Affiliation(s)
- J Christian Pérez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America.
| | | | | |
Collapse
|
50
|
Normal adaptation of Candida albicans to the murine gastrointestinal tract requires Efg1p-dependent regulation of metabolic and host defense genes. EUKARYOTIC CELL 2012; 12:37-49. [PMID: 23125349 DOI: 10.1128/ec.00236-12] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although gastrointestinal colonization by the opportunistic fungal pathogen Candida albicans is generally benign, severe systemic infections are thought to arise due to escape of commensal C. albicans from the gastrointestinal (GI) tract. The C. albicans transcription factor Efg1p is a major regulator of GI colonization, hyphal morphogenesis, and virulence. The goals of this study were to identify the Efg1p regulon during GI tract colonization and to compare C. albicans gene expression during colonization of different organs of the GI tract. Our results identified significant differences in gene expression between cells colonizing the cecum and ileum. During colonization, efg1(-) null mutant cells expressed higher levels of genes involved in lipid catabolism, carnitine biosynthesis, and carnitine utilization than did colonizing wild-type (WT) cells. In addition, during laboratory growth, efg1(-) null mutant cells grew to a higher density than WT cells. The efg1(-) null mutant grew in depleted medium, while WT cells could grow only if the depleted medium was supplemented with carnitine, a compound that promotes the metabolism of fatty acids. Altered gene expression and altered growth capability support the ability of efg1(-) cells to hypercolonize naïve mice. Also, Efg1p was shown to be important for transcriptional responses to the stresses present in the cecum environment. For example, during colonization, SOD5, encoding a superoxide dismutase, was highly upregulated in an Efg1p-dependent manner. Ectopic expression of SOD5 in an efg1(-) null mutant increased the fitness of the efg1(-) null mutant cells during colonization. These data show that EFG1 is an important regulator of GI colonization.
Collapse
|