16 results on '"Caroline Mota Fernandes"'
Search Results
2. Targeting Sterylglucosidase A to Treat Aspergillus fumigatus Infections
- Author
-
Nivea Pereira de Sa, Kalani Jayanetti, Dominick Rendina, Timothy Clement, Veronica Soares Brauer, Caroline Mota Fernandes, Iwao Ojima, Michael V. Airola, and Maurizio Del Poeta
- Subjects
Virology ,Microbiology - Abstract
Invasive fungal infections are a leading cause of death in immunocompromised patients. Aspergillus fumigatus is a fungus ubiquitously found in the environment that, upon inhalation, causes both acute and chronic illnesses in at-risk individuals. A. fumigatus is recognized as one of the critical fungal pathogens for which a substantive treatment breakthrough is urgently needed. Here, we studied a fungus-specific enzyme, sterylglucosidase A (SglA), as a therapeutic target.
- Published
- 2023
- Full Text
- View/download PDF
3. Vaccination with Live or Heat-Killed Aspergillus fumigatus Δ sglA Conidia Fully Protects Immunocompromised Mice from Invasive Aspergillosis
- Author
-
Caroline Mota Fernandes, Tyler G. Normile, Joao Henrique Tadini Marilhano Fabri, Veronica Soares Brauer, Glauber R. de S. Araújo, Susana Frases, Leonardo Nimrichter, Iran Malavazi, and Maurizio Del Poeta
- Subjects
Virology ,Microbiology - Abstract
Infections by Aspergillus fumigatus occur by the inhalation of environmental fungal spores called conidia. We found that live mutant conidia accumulating glycolipids named sterylglucosides are not able to cause disease when injected into the lung.
- Published
- 2022
- Full Text
- View/download PDF
4. Vaccination with Live or Heat-Killed Aspergillus fumigatus Δ
- Author
-
Caroline Mota, Fernandes, Tyler G, Normile, Joao Henrique Tadini Marilhano, Fabri, Veronica Soares, Brauer, Glauber R, de S Araújo, Susana, Frases, Leonardo, Nimrichter, Iran, Malavazi, and Maurizio, Del Poeta
- Subjects
Mice ,Immunocompromised Host ,Hot Temperature ,Aspergillus fumigatus ,Vaccination ,Animals ,Aspergillosis ,Spores, Fungal ,Glycolipids ,Cyclophosphamide ,Invasive Fungal Infections - Abstract
Aspergillus fumigatus causes invasive aspergillosis (IA) in immunocompromised patients, resulting in high mortality rates. Currently, no vaccine formulations to promote immune protection in at-risk individuals have been developed. In this work, we deleted the sterylglucosidase-encoding gene
- Published
- 2022
5. Microdomain Protein Nce102 Is a Local Sensor of Plasma Membrane Sphingolipid Balance
- Author
-
Jakub Zahumenský, Caroline Mota Fernandes, Petra Veselá, Maurizio Del Poeta, James B. Konopka, and Jan Malínský
- Subjects
Microbiology (medical) ,Sphingolipids ,Saccharomyces cerevisiae Proteins ,General Immunology and Microbiology ,Ecology ,Physiology ,Cell Membrane ,Cell Biology ,Saccharomyces cerevisiae ,carbohydrates (lipids) ,Fungal Proteins ,Infectious Diseases ,Candida albicans ,Genetics ,lipids (amino acids, peptides, and proteins) - Abstract
Sphingolipids are essential building blocks of eukaryotic membranes and important signalling molecules, tightly regulated in response to environmental and physiological inputs. Mechanism of sphingolipid level perception at the plasma membrane remains unclear. In Saccharomyces cerevisiae, Nce102 protein has been proposed to function as sphingolipid sensor as it changes its plasma membrane distribution in response to sphingolipid biosynthesis inhibition. We show that Nce102 redistributes specifically in regions of increased sphingolipid demand, e.g., membranes of nascent buds. Furthermore, we report that production of Nce102 increases following sphingolipid biosynthesis inhibition and Nce102 is internalized when excess sphingolipid precursors are supplied. This suggests that the total amount of Nce102 in the plasma membrane is a measure of the current need for sphingolipids, whereas its local distribution marks sites of high sphingolipid demand. Physiological role of Nce102 in regulation of sphingolipid synthesis is demonstrated by mass spectrometry analysis showing reduced levels of complex sphingolipids and long-chain bases in nce102Δ deletion mutant. Nce102 behaves analogously in human fungal pathogen Candida albicans, suggesting a conserved principle of local sphingolipid control across species.
- Published
- 2022
6. Fungal sphingolipids: role in the regulation of virulence and potential as targets for future antifungal therapies
- Author
-
Maurizio Del Poeta and Caroline Mota Fernandes
- Subjects
0301 basic medicine ,Microbiology (medical) ,Antifungal ,Antifungal Agents ,medicine.drug_class ,030106 microbiology ,Virulence ,Biology ,Microbiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Drug Development ,Drug Resistance, Fungal ,Virology ,medicine ,Animals ,Humans ,030212 general & internal medicine ,Sphingolipids ,Fungi ,Sphingolipid ,Infectious Diseases ,Mycoses ,Echinocandins - Abstract
INTRODUCTION: The antifungal therapy currently available includes three major classes of drugs: polyenes, azoles and echinocandins. However, the clinical use of these compounds faces several challenges: while polyenes are toxic to the host, antifungal resistance to azoles and echinocandins has been reported. AREAS COVERED: Fungal sphingolipids (SL) play a pivotal role in growth, morphogenesis and virulence. In addition, fungi possess unique enzymes involved in SL synthesis, leading to the production of lipids which are absent or differ structurally from the mammalian counterparts. In this review, we address the enzymatic reactions involved in the SL synthesis and their relevance to the fungal pathogenesis, highlighting their potential as targets for novel drugs and the inhibitors described so far. EXPERT OPINION: The pharmacological inhibition of fungal serine palmitoyltransferase depends on the development of specific drugs, as myriocin also targets the mammalian enzyme. Inhibitors of ceramide synthase might constitute potent antifungals, by depleting the pool of complex SL and leading to the accumulation of the toxic intermediates. Acylhydrazones and aureobasidin A, which inhibit GlcCer and IPC synthesis, are not toxic to the host and effectively treat invasive mycoses, emerging as promising new classes of antifungal drugs.
- Published
- 2020
- Full Text
- View/download PDF
7. Sphingolipid depletion suppresses UPR activation and promotes galactose hypersensitivity in yeast models of classic galactosemia
- Author
-
Felipe S.A. Pimentel, Caio M. Machado, Evandro A. De-Souza, Caroline Mota Fernandes, Ana Luiza F.V. De-Queiroz, Guilherme F.S. Silva, Maurizio Del Poeta, Monica Montero-Lomeli, and Claudio A. Masuda
- Subjects
Galactosemias ,Sphingolipids ,Molecular Medicine ,Galactose ,Humans ,UTP-Hexose-1-Phosphate Uridylyltransferase ,Saccharomyces cerevisiae ,Molecular Biology ,Article - Abstract
Classic galactosemia is an inborn error of metabolism caused by deleterious mutations on the GALT gene, which encodes the Leloir pathway enzyme galactose-1-phosphate uridyltransferase. Previous studies have shown that the endoplasmic reticulum unfolded protein response (UPR) is relevant to galactosemia, but the molecular mechanism behind the endoplasmic reticulum stress that triggers this response remains elusive. In the present work, we show that the activation of the UPR in yeast models of galactosemia does not depend on the binding of unfolded proteins to the ER stress sensor protein Ire1p since the protein domain responsible for unfolded protein binding to Ire1p is not necessary for UPR activation. Interestingly, myriocin - an inhibitor of the de novo sphingolipid synthesis pathway - inhibits UPR activation and causes galactose hypersensitivity in these models, indicating that myriocin-mediated sphingolipid depletion impairs yeast adaptation to galactose toxicity. Supporting the interpretation that the effects observed after myriocin treatment were due to a reduction in sphingolipid levels, the addition of phytosphingosine to the culture medium reverses all myriocin effects tested. Surprisingly, constitutively active UPR signaling did not prevent myriocin-induced galactose hypersensitivity suggesting multiple roles for sphingolipids in the adaptation of yeast cells to galactose toxicity. Therefore, we conclude that sphingolipid homeostasis has an important role in UPR activation and cellular adaptation in yeast models of galactosemia, highlighting the possible role of lipid metabolism in the pathophysiology of this disease.
- Published
- 2022
8. Screening of Chemical Libraries for New Antifungal Drugs against Aspergillus fumigatus Reveals Sphingolipids Are Involved in the Mechanism of Action of Miltefosine
- Author
-
Rafael Wesley Bastos, Thaila Fernanda dos Reis, Ana Cristina Colabardini, Caroline Mota Fernandes, Cristina Silva Pereira, Marcio L. Rodrigues, Koon Ho Wong, Fang Wang, Maria Vitória de Lazari Fonseca, Maria Augusta Crivelente Horta, Lilian Pereira Silva, Maurizio Del Poeta, Celso Martins, and Gustavo H. Goldman
- Subjects
Drug ,Antifungal Agents ,Phosphorylcholine ,media_common.quotation_subject ,Mutant ,Microbial Sensitivity Tests ,Moths ,Aspergillosis ,Microbiology ,Aspergillus fumigatus ,Fungal Proteins ,Small Molecule Libraries ,Drug Resistance, Fungal ,Virology ,medicine ,Animals ,skin and connective tissue diseases ,transcription factor ,media_common ,Miltefosine ,sphingolipids ,Virulence ,drug repurposing ,biology ,biology.organism_classification ,medicine.disease ,Sphingolipid ,QR1-502 ,High-Throughput Screening Assays ,Drug repositioning ,Phenotype ,Mechanism of action ,Larva ,medicine.symptom ,miltefosine ,Research Article ,medicine.drug - Abstract
Aspergillus fumigatus is an important fungal pathogen and the main etiological agent of aspergillosis, a disease characterized by a noninvasive process that can evolve to a more severe clinical manifestation, called invasive pulmonary aspergillosis (IPA), in immunocompromised patients. The antifungal arsenal to threat aspergillosis is very restricted. Azoles are the main therapeutic approach to control IPA, but the emergence of azole-resistant A. fumigatus isolates has significantly increased over recent decades. Therefore, new strategies are necessary to combat aspergillosis, and drug repurposing has emerged as an efficient and alternative approach for identifying new antifungal drugs. Here, we used a screening approach to analyze A. fumigatus in vitro susceptibility to 1,127 compounds. A. fumigatus was susceptible to 10 compounds, including miltefosine, a drug that displayed fungicidal activity against A. fumigatus. By screening an A. fumigatus transcription factor null library, we identified a single mutant, which has the smiA (sensitive to miltefosine) gene deleted, conferring a phenotype of susceptibility to miltefosine. The transcriptional profiling (RNA-seq) of the wild-type and ΔsmiA strains and chromatin immunoprecipitation coupled to next-generation sequencing (ChIP-Seq) of an SmiA-tagged strain exposed to miltefosine revealed genes of the sphingolipid pathway that are directly or indirectly regulated by SmiA. Sphingolipid analysis demonstrated that the mutant has overall decreased levels of sphingolipids when growing in the presence of miltefosine. The identification of SmiA represents the first genetic element described and characterized that plays a direct role in miltefosine response in fungi. IMPORTANCE The filamentous fungus Aspergillus fumigatus causes a group of diseases named aspergillosis, and their development occurs after the inhalation of conidia dispersed in the environment. Very few classes of antifungal drugs are available for aspergillosis treatment, e.g., azoles, but the emergence of global resistance to azoles in A. fumigatus clinical isolates has increased over recent decades. Repositioning or repurposing drugs already available on the market is an interesting and faster opportunity for the identification of novel antifungal agents. By using a repurposing strategy, we identified 10 different compounds that impact A. fumigatus survival. One of these compounds, miltefosine, demonstrated fungicidal activity against A. fumigatus. The mechanism of action of miltefosine is unknown, and, aiming to get more insights about it, we identified a transcription factor, SmiA (sensitive to miltefosine), important for miltefosine resistance. Our results suggest that miltefosine displays antifungal activity against A. fumigatus, interfering in sphingolipid biosynthesis.
- Published
- 2021
- Full Text
- View/download PDF
9. Screening of chemical libraries for new antifungal drugs against Aspergillus fumigatus reveals the potential mechanism of action of miltefosine
- Author
-
Gustavo H. Goldman, Ana Cristina Colabardini, Lilian Pereira Silva, Caroline Mota Fernandes, Silva Pereira C, Fonseca Mvl, Koon Ho Wong, Rafael Wesley Bastos, Marcio L. Rodrigues, Del Poeta M, Celso Martins, Horta Mac, Fang Wang, and dos Reis Tf
- Subjects
Drug ,Miltefosine ,biology ,media_common.quotation_subject ,Mutant ,Aspergillosis ,medicine.disease ,biology.organism_classification ,Sphingolipid ,Microbiology ,Aspergillus fumigatus ,Drug repositioning ,Mechanism of action ,medicine ,medicine.symptom ,media_common ,medicine.drug - Abstract
Aspergillus fumigatus is an important fungal pathogen and the main etiological agent of aspergillosis, a disease characterized by a noninvasive process that can evolve to a more severe clinical manifestation called invasive pulmonary aspergillosis (IPA) in immunocompromised patients. The antifungal arsenal to threat aspergillosis is very restricted. Azoles are the main therapeutic approach to control IPA, but the emergence of azole-resistant A. fumigatus isolates has significantly increased over the last decades. Therefore, new strategies are necessary to combat aspergillosis and drug repurposing has emerged as an efficient and alternative approach for identifying new antifungal drugs. Here, we used a screening approach to analyze A. fumigatus in vitro susceptibility to 1,127 compounds. A. fumigatus was more susceptible to 10 compounds, including miltefosine, a drug that displayed fungicidal activity against A. fumigatus. By screening an A. fumigatus transcription factor null library, we identified a single mutant, which has the rmiA (resistant to miltefosine) gene deleted, conferring a phenotype of susceptibility to miltefosine. The transcriptional profiling (RNA-seq) of the wild-type and the ΔrmiA strains and the Chromatin Immunoprecipitation coupled to next generation sequencing (ChIP-Seq) of a RmiA-tagged strain exposed to miltefosine revealed genes of the sphingolipids pathway that are directly or indirectly regulated by RmiA. Sphingolipids analysis demonstrated that the mutant has overall decreased levels of sphingolipids when growing in the presence of miltefosine. The identification of RmiA represents the first genetic element described and characterized which plays a direct role in miltefosine response in fungi.Author summaryThe filamentous fungus Aspergillus fumigatus causes a group of diseases named aspergillosis and their development occurs after the inhalation of conidia dispersed in the environment. Very few classes of antifungal drugs are available for aspergillosis treatment, e.g., azoles, but the emergence of global resistance to azoles in A. fumigatus clinical isolates has increased over the last decades. Repositioning or repurposing drugs already available on the market is an interesting and faster opportunity for the identification of novel antifungals agents. By using a repurposing strategy, we identified 10 different compounds that impact A. fumigatus survival. One of these compounds, miltefosine, demonstrated fungicidal activity against A. fumigatus. The mechanism of action of miltefosine is unknown and aiming to get more insights about it, we identified a transcription factor RmiA (Resistant to miltefosine) important for miltefosine resistance. Our results suggest that miltefosine plays antifungal activity against A. fumigatus interfering in the sphingolipids biosynthesis.
- Published
- 2021
- Full Text
- View/download PDF
10. The Heat Shock Transcription Factor HsfA Is Essential for Thermotolerance and Regulates Cell Wall Integrity in Aspergillus fumigatus
- Author
-
João Henrique Tadini Marilhano Fabri, Marina Campos Rocha, Caroline Mota Fernandes, Gabriela Felix Persinoti, Laure Nicolas Annick Ries, Anderson Ferreira da Cunha, Gustavo Henrique Goldman, Maurizio Del Poeta, and Iran Malavazi
- Subjects
Microbiology (medical) ,biology ,Aspergillus fumigatus ,lcsh:QR1-502 ,heat shock (HS) ,biology.organism_classification ,Microbiology ,Hsp90 ,thermotolerance ,lcsh:Microbiology ,Cell biology ,Heat shock factor ,Chaperone (protein) ,Heat shock protein ,biology.protein ,cell wall integrity (CWI) ,HsfA ,Signal transduction ,Gene ,Transcription factor ,transcription factor - Abstract
The deleterious effects of human-induced climate change have long been predicted. However, the imminent emergence and spread of new diseases, including fungal infections through the rise of thermotolerant strains, is still neglected, despite being a potential consequence of global warming. Thermotolerance is a remarkable virulence attribute of the mold Aspergillus fumigatus. Under high-temperature stress, opportunistic fungal pathogens deploy an adaptive mechanism known as heat shock (HS) response controlled by heat shock transcription factors (HSFs). In eukaryotes, HSFs regulate the expression of several heat shock proteins (HSPs), such as the chaperone Hsp90, which is part of the cellular program for heat adaptation and a direct target of HSFs. We recently observed that the perturbation in cell wall integrity (CWI) causes concomitant susceptibility to elevated temperatures in A. fumigatus, although the mechanisms underpinning the HS response and CWI cross talking are not elucidated. Here, we aim at further deciphering the interplay between HS and CWI. Our results show that cell wall ultrastructure is severely modified when A. fumigatus is exposed to HS. We identify the transcription factor HsfA as essential for A. fumigatus viability, thermotolerance, and CWI. Indeed, HS and cell wall stress trigger the coordinated expression of both hsfA and hsp90. Furthermore, the CWI signaling pathway components PkcA and MpkA were shown to be important for HsfA and Hsp90 expression in the A. fumigatus biofilms. Lastly, RNA-sequencing confirmed that hsfA regulates the expression of genes related to the HS response, cell wall biosynthesis and remodeling, and lipid homeostasis. Our studies collectively demonstrate the connection between the HS and the CWI pathway, with HsfA playing a crucial role in this cross-pathway regulation, reinforcing the importance of the cell wall in A. fumigatus thermophily.
- Published
- 2021
- Full Text
- View/download PDF
11. The Heat Shock Transcription Factor HsfA Is Essential for Thermotolerance and Regulates Cell Wall Integrity in
- Author
-
João Henrique Tadini Marilhano, Fabri, Marina Campos, Rocha, Caroline Mota, Fernandes, Gabriela Felix, Persinoti, Laure Nicolas Annick, Ries, Anderson Ferreira, da Cunha, Gustavo Henrique, Goldman, Maurizio, Del Poeta, and Iran, Malavazi
- Subjects
Aspergillus fumigatus ,cell wall integrity (CWI) ,heat shock (HS) ,HsfA ,Microbiology ,thermotolerance ,transcription factor ,Original Research - Abstract
The deleterious effects of human-induced climate change have long been predicted. However, the imminent emergence and spread of new diseases, including fungal infections through the rise of thermotolerant strains, is still neglected, despite being a potential consequence of global warming. Thermotolerance is a remarkable virulence attribute of the mold Aspergillus fumigatus. Under high-temperature stress, opportunistic fungal pathogens deploy an adaptive mechanism known as heat shock (HS) response controlled by heat shock transcription factors (HSFs). In eukaryotes, HSFs regulate the expression of several heat shock proteins (HSPs), such as the chaperone Hsp90, which is part of the cellular program for heat adaptation and a direct target of HSFs. We recently observed that the perturbation in cell wall integrity (CWI) causes concomitant susceptibility to elevated temperatures in A. fumigatus, although the mechanisms underpinning the HS response and CWI cross talking are not elucidated. Here, we aim at further deciphering the interplay between HS and CWI. Our results show that cell wall ultrastructure is severely modified when A. fumigatus is exposed to HS. We identify the transcription factor HsfA as essential for A. fumigatus viability, thermotolerance, and CWI. Indeed, HS and cell wall stress trigger the coordinated expression of both hsfA and hsp90. Furthermore, the CWI signaling pathway components PkcA and MpkA were shown to be important for HsfA and Hsp90 expression in the A. fumigatus biofilms. Lastly, RNA-sequencing confirmed that hsfA regulates the expression of genes related to the HS response, cell wall biosynthesis and remodeling, and lipid homeostasis. Our studies collectively demonstrate the connection between the HS and the CWI pathway, with HsfA playing a crucial role in this cross-pathway regulation, reinforcing the importance of the cell wall in A. fumigatus thermophily.
- Published
- 2021
12. The Future of Antifungal Drug Therapy: Novel Compounds and Targets
- Author
-
Caroline Mota Fernandes, Iwao Ojima, Maurizio Del Poeta, J. Brian McCarthy, John P. Mallamo, Krupanandan Haranahalli, and Deveney Dasilva
- Subjects
Antifungal Agents ,Antifungal drug ,Drug resistance ,Bioinformatics ,03 medical and health sciences ,High morbidity ,chemistry.chemical_compound ,Pharmacotherapy ,Ergosterol ,Drug Discovery ,Medicine ,Humans ,Pharmacology (medical) ,030304 developmental biology ,Fungal pathogenesis ,Pharmacology ,0303 health sciences ,030306 microbiology ,business.industry ,Fungi ,Infectious Diseases ,Drug development ,chemistry ,Mycoses ,Molecular targets ,Minireview ,business - Abstract
Fungal infections are a universal problem and are routinely associated with high morbidity and mortality rates in immunocompromised patients. Existing therapies comprise five different classes of antifungal agents, four of which target the synthesis of ergosterol and cell wall glucans. However, the currently available antifungals have many limitations, including poor oral bioavailability, narrow therapeutic indices, and emerging drug resistance resulting from their use, thus making it essential to investigate the development of novel drugs which can overcome these limitations and add to the antifungal armamentarium. Advances have been made in antifungal drug discovery research and development over the past few years as evidenced by the presence of several new compounds currently in various stages of development. In the following minireview, we provide a comprehensive summary of compounds aimed at one or more novel molecular targets. We also briefly describe potential pathways relevant for fungal pathogenesis that can be considered for drug development in the near future.
- Published
- 2021
13. Internalization of the host alkaline pH signal in a fungal pathogen
- Author
-
Kaila M. Pianalto, K. D. Mueller, M Del Poeta, Hannah E. Brown, J. A. Alspaugh, and Caroline Mota Fernandes
- Subjects
Cryptococcus neoformans ,Cell signaling ,biology ,media_common.quotation_subject ,Extracellular ,Context (language use) ,Signal transduction ,Internalization ,biology.organism_classification ,Endocytosis ,Pathogen ,media_common ,Cell biology - Abstract
The ability for cells to internalize extracellular cues allows them to adapt to novel and stressful environments. This adaptability is especially important for microbial pathogens that must sense and respond to drastic changes when encountering the human host.Cryptococcus neoformansis an environmental fungus and opportunistic pathogen that naturally lives in slightly acidic reservoirs, but must adapt to the relative increase in alkalinity in the human host in order to effectively cause disease. The fungal-specific Rim alkaline response signaling pathway effectively converts this extracellular signal into an adaptive cellular response allowing the pathogen to survive in its new environment. The newly identified Rra1 protein, the most upstream component of theC. neoformansRim pathway, is an essential component of this alkaline response. Previous work connected Rra1-mediated signaling to the dynamics of the plasma membrane. Here we identify the specific mechanisms of Rim pathway signaling through detailed studies of the activation of the Rra1 protein. Specifically, we observe that the Rra1 protein is internalized and recycled in a pH-dependent manner, and that this dynamic pattern of localization further depends on specific residues in its C-terminal tail, clathrin-mediated endocytosis, and the integrity of the plasma membrane. The data presented here continue to unravel the complex and intricate processes of pH-sensing in a relevant human fungal pathogen. These studies will further elucidate general mechanisms by which cells respond to and internalize extracellular stress signals.Author SummaryThe work described here explores the genetics and mechanics of a cellular signaling pathway in a relevant human fungal pathogen,Cryptococcus neoformans. The findings presented in this manuscript untangle the complex interactions involved in the activation of a fungal-specific alkaline response pathway, the Rim pathway. Specifically, we find thatC. neoformansis able to sense an increase in pH within the human host, internalize a membrane-bound pH-sensor, and activate a downstream signaling pathway enabling this pathogen to adapt to a novel host environment and effectively cause disease. Revealing the mechanisms of Rim pathway activation within the larger context of the fungal cell allows us to understand how and when this microorganism interprets relevant host signals. Furthermore, understanding how this pathogenic organism converts extracellular stress signals into an adaptive cellular response will elucidate more general mechanisms of microbial environmental sensing and stress response.
- Published
- 2020
- Full Text
- View/download PDF
14. Identification of Antifungal Compounds against Multidrug-Resistant Candida auris Utilizing a High-Throughput Drug-Repurposing Screen
- Author
-
Patricia S. Tsang, Jose Santinni O Roma, Maurizio Del Poeta, Helena I. Boshoff, Peter R. Williamson, Caroline Mota Fernandes, Min Shen, Wei Zheng, He Eun Forbes, Yu-Shan Cheng, and Cristina Lazzarini
- Subjects
Drug ,Antifungal ,Antifungal Agents ,medicine.drug_class ,media_common.quotation_subject ,Drug resistance ,Microbial Sensitivity Tests ,Flucytosine ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Myriocin ,Medicine ,Pharmacology (medical) ,030304 developmental biology ,media_common ,Candida ,Pharmacology ,0303 health sciences ,030306 microbiology ,business.industry ,Drug Repositioning ,Multiple drug resistance ,Drug repositioning ,Infectious Diseases ,Candida auris ,chemistry ,Pharmaceutical Preparations ,Susceptibility ,business ,medicine.drug - Abstract
Candida auris is an emerging fatal fungal infection that has resulted in several outbreaks in hospitals and care facilities. Current treatment options are limited by the development of drug resistance. Identification of new pharmaceuticals to combat these drug-resistant infections will thus be required to overcome this unmet medical need. We have established a bioluminescent ATP-based assay to identify new compounds and potential drug combinations showing effective growth inhibition against multiple strains of multidrug-resistant Candida auris. The assay is robust and suitable for assessing large compound collections by high-throughput screening (HTS). Utilizing this assay, we conducted a screen of 4,314 approved drugs and pharmacologically active compounds that yielded 25 compounds, including 6 novel anti-Candida auris compounds and 13 sets of potential two-drug combinations. Among the drug combinations, the serine palmitoyltransferase inhibitor myriocin demonstrated a combinational effect with flucytosine against all tested isolates during screening. This combinational effect was confirmed in 13 clinical isolates of Candida auris.
- Published
- 2020
15. Regulation of sphingolipid synthesis by the G1/S transcription factor Swi4
- Author
-
Caroline Mota Fernandes, Deveney Dasilva, Maurizio Del Poeta, Claudio A. Masuda, Gabriel S. Matos, Mónica Montero-Lomelí, and Juliana B. Madeira
- Subjects
Ceramide ,Saccharomyces cerevisiae Proteins ,Saccharomyces cerevisiae ,Mitosis ,Article ,S Phase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Mitotic cell cycle ,Transcription (biology) ,Gene Expression Regulation, Fungal ,Myriocin ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Sphingolipids ,0303 health sciences ,biology ,Chemistry ,G1 Phase ,Cell Biology ,Cell cycle ,biology.organism_classification ,Sphingolipid ,Cell biology ,DNA-Binding Proteins ,030220 oncology & carcinogenesis ,Transcription Factors - Abstract
SBF (Swi4/Swi6 Binding Factor) complex is a crucial regulator of G1/S transition in Saccharomyces cerevisiae. Here, we show that SBF complex is required for myriocin resistance, an inhibitor of sphingolipid synthesis. This phenotype was not shared with MBF complex mutants nor with deletion of the Swi4p downstream targets, CLN1/CLN2. Based on data mining results, we selected putative Swi4p targets related to sphingolipid metabolism and studied their gene transcription as well as metabolite levels during progression of the cell cycle. Genes which encode key enzymes for the synthesis of long chain bases (LCBs) and ceramides were periodically transcribed during the mitotic cell cycle, having a peak at G1/S, and required SWI4 for full transcription at this stage. In addition, HPLC-MS/MS data indicated that swi4Δ cells have decreased levels of sphingolipids during progression of the cell cycle, particularly, dihydrosphingosine (DHS), C24-phytoceramides and C24-inositolphosphoryl ceramide (IPC) while it had increased levels of mannosylinositol phosphorylceramide (MIPC). Furthermore, we demonstrated that both inhibition of de novo sphingolipid synthesis by myriocin or SWI4 deletion caused partial arrest at the G2/M phase. Importantly, our lipidomic data demonstrated that the sphingolipid profile of WT cells treated with myriocin resembled that of swi4Δ cells, with lower levels of DHS, IPC and higher levels of MIPC. Taken together, these results show that SBF complex plays an essential role in the regulation of sphingolipid homeostasis, which reflects in the correct progression through the G2/M phase of the cell cycle.
- Published
- 2021
- Full Text
- View/download PDF
16. Metabolomics Analysis Identifies Sphingolipids as Key Signaling Moieties in Appressorium Morphogenesis and Function in Magnaporthe oryzae
- Author
-
Zheng Qingxia, Yun-Yun Wei, Caroline Mota Fernandes, Lin Li, Xiao-Hong Liu, Xue-Ming Zhu, Fu-Cheng Lin, Naweed I. Naqvi, Pingping Liu, Li-Juan Mao, Maurizio Del Poeta, Huina Zhou, Shuang Liang, and Yong Zhang
- Subjects
Molecular Biology and Physiology ,Antifungal Agents ,appressorium development ,chemistry.chemical_compound ,Cell Wall ,Gene Expression Regulation, Fungal ,Morphogenesis ,pathogenicity ,Protein Kinase C ,2. Zero hunger ,0303 health sciences ,Virulence ,early sphingolipid signaling ,food and beverages ,QR1-502 ,Cell biology ,Magnaporthe ,Phenotype ,Metabolome ,Functional genomics ,metabolomics analysis ,Signal Transduction ,Research Article ,Ceramide ,Genes, Fungal ,Mitosis ,Biology ,Glucosylceramides ,Poaceae ,Microbiology ,Fungal Proteins ,03 medical and health sciences ,Metabolomics ,Virology ,inducer ,Plant Diseases ,030304 developmental biology ,Sphingolipids ,Appressorium ,030306 microbiology ,Membrane Proteins ,Oryza ,Sphingolipid ,Biosynthetic Pathways ,Metabolic pathway ,chemistry ,rice blast fungus ,Edible Grain ,Transcriptome ,Gene Deletion ,Function (biology) - Abstract
Our untargeted analysis of metabolomics throughout the course of pathogenic development gave us an unprecedented high-resolution view of major shifts in metabolism that occur in the topmost fungal pathogen that infects rice, wheat, barley, and millet. Guided by these metabolic insights, we demonstrated their practical application by using two different small-molecule inhibitors of sphingolipid biosynthesis enzymes to successfully block the pathogenicity of M. oryzae. Our study thus defines the sphingolipid biosynthesis pathway as a key step and potential target that can be exploited for the development of antifungal agents. Furthermore, future investigations that exploit such important metabolic intermediates will further deepen our basic understanding of the molecular mechanisms underlying the establishment of fungal blast disease in important cereal crops., The blast fungus initiates infection using a heavily melanized, dome-shaped infection structure known as the appressorium, which forcibly ruptures the cuticle to enter the rice leaf tissue. How this process takes place remains not fully understood. Here, we used untargeted metabolomics analyses to profile the metabolome of developing appressoria and identified significant changes in six key metabolic pathways, including early sphingolipid biosynthesis. Analyses employing small molecule inhibitors, gene disruption, or genetic and chemical complementation demonstrated that ceramide compounds of the sphingolipid biosynthesis pathway are essential for normal appressorial development controlled by mitosis. In addition, ceramide was found to act upstream from the protein kinase C-mediated cell wall integrity pathway during appressorium repolarization and pathogenicity in rice blast. Further discovery of the sphingolipid biosynthesis pathway revealed that glucosylceramide (GlcCer) synthesized by ceramide is the key substance affecting the pathogenicity of Magnaporthe oryzae. Our results provide new insights into the chemical moieties involved in the infection-related signaling networks, thereby revealing a potential target for the development of novel control agents against the major disease of rice and other cereals.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.