Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 542
Filter
1.
Structure ; 32(9): 1296-1298, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241761

ABSTRACT

Fumonisin B1 (FB1) targets sphingolipid biosynthesis, inhibiting ceramide synthases. In this issue of Structure, Zhang et al.1 determined the cryoelectron microscopic structures of yeast ceramide synthase in complex with FB1 and its acylated derivative, acyl-FB1, revealing a two-step "ping-pong" mechanism for the N-acylation of FB1 and how it inhibits ceramide synthase.


Subject(s)
Cryoelectron Microscopy , Fumonisins , Oxidoreductases , Fumonisins/chemistry , Fumonisins/metabolism , Oxidoreductases/metabolism , Oxidoreductases/chemistry , Oxidoreductases/antagonists & inhibitors , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Acylation , Models, Molecular , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Sphingolipids/metabolism , Sphingolipids/chemistry
2.
Structure ; 32(9): 1419-1428.e4, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38964337

ABSTRACT

Ceramide synthases (CerSs) play crucial roles in sphingolipid metabolism and have emerged as promising drug targets for metabolic diseases, cancers, and antifungal therapy. However, the therapeutic targeting of CerSs has been hindered by a limited understanding of their inhibition mechanisms by small molecules. Fumonisin B1 (FB1) has been extensively studied as a potent inhibitor of eukaryotic CerSs. In this study, we characterize the inhibition mechanism of FB1 on yeast CerS (yCerS) and determine the structures of both FB1-bound and N-acyl-FB1-bound yCerS. Through our structural analysis and the observation of N-acylation of FB1 by yCerS, we propose a potential ping-pong catalytic mechanism for FB1 N-acylation by yCerS. Lastly, we demonstrate that FB1 exhibits lower binding affinity for yCerS compared to the C26- coenzyme A (CoA) substrate, suggesting that the potent inhibitory effect of FB1 on yCerS may primarily result from the N-acyl-FB1 catalyzed by yCerS, rather than through direct binding of FB1.


Subject(s)
Fumonisins , Oxidoreductases , Protein Binding , Saccharomyces cerevisiae , Fumonisins/metabolism , Fumonisins/chemistry , Oxidoreductases/metabolism , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/enzymology , Acylation , Models, Molecular , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Crystallography, X-Ray , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Catalytic Domain , Binding Sites , Coenzyme A/metabolism , Coenzyme A/chemistry , Substrate Specificity
3.
FEBS Lett ; 598(13): 1655-1666, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750637

ABSTRACT

Cymoxanil (CYM) is a widely used synthetic acetamide fungicide, but its biochemical mode of action remains elusive. Since CYM inhibits cell growth, biomass production, and respiration in Saccharomyces cerevisiae, we used this model to characterize the effect of CYM on mitochondria. We found it inhibits oxygen consumption in both whole cells and isolated mitochondria, specifically inhibiting cytochrome c oxidase (CcO) activity during oxidative phosphorylation. Based on molecular docking, we propose that CYM blocks the interaction of cytochrome c with CcO, hampering electron transfer and inhibiting CcO catalytic activity. Although other targets cannot be excluded, our data offer valuable insights into the mode of action of CYM that will be instrumental in driving informed management of the use of this fungicide.


Subject(s)
Electron Transport Complex IV , Fungicides, Industrial , Mitochondria , Molecular Docking Simulation , Saccharomyces cerevisiae , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/enzymology , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/antagonists & inhibitors , Fungicides, Industrial/pharmacology , Fungicides, Industrial/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects , Oxidative Phosphorylation/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors
4.
J Microbiol Biotechnol ; 34(6): 1348-1355, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38755008

ABSTRACT

The eukaryotic translation initiation factor eIF5B is a bacterial IF2 ortholog that plays an important role in ribosome joining and stabilization of the initiator tRNA on the AUG start codon during the initiation of translation. We identified the fluorophenyl oxazole derivative 2,2-dibromo-1-(2-(4-fluorophenyl)benzo[d]oxazol-5-yl)ethanone quinolinol as an inhibitor of fungal protein synthesis using an in vitro translation assay in a fungal system. Mutants resistant to this compound were isolated in Saccharomyces cerevisiae and were demonstrated to contain amino acid substitutions in eIF5B that conferred the resistance. These results suggest that eIF5B is a target of potential antifungal compound and that mutation of eIF5B can confer resistance. Subsequent identification of 16 other mutants revealed that primary mutations clustered mainly on domain 2 of eIF5B and secondarily mainly on domain 4. Domain 2 has been implicated in the interaction with the small ribosomal subunit during initiation of translation. The tested translation inhibitor could act by weakening the functional contact between eIF5B and the ribosome complex. This data provides the basis for the development of a new family of antifungals.


Subject(s)
Antifungal Agents , Eukaryotic Initiation Factors , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/drug effects , Eukaryotic Initiation Factors/metabolism , Eukaryotic Initiation Factors/genetics , Antifungal Agents/pharmacology , Mutation , Protein Biosynthesis/drug effects , Drug Resistance, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Amino Acid Substitution
5.
Nature ; 616(7955): 190-198, 2023 04.
Article in English | MEDLINE | ID: mdl-36949198

ABSTRACT

The membrane-integrated synthase FKS is involved in the biosynthesis of ß-1,3-glucan, the core component of the fungal cell wall1,2. FKS is the target of widely prescribed antifungal drugs, including echinocandin and ibrexafungerp3,4. Unfortunately, the mechanism of action of FKS remains enigmatic and this has hampered development of more effective medicines targeting the enzyme. Here we present the cryo-electron microscopy structures of Saccharomyces cerevisiae FKS1 and the echinocandin-resistant mutant FKS1(S643P). These structures reveal the active site of the enzyme at the membrane-cytoplasm interface and a glucan translocation path spanning the membrane bilayer. Multiple bound lipids and notable membrane distortions are observed in the FKS1 structures, suggesting active FKS1-membrane interactions. Echinocandin-resistant mutations are clustered at a region near TM5-6 and TM8 of FKS1. The structure of FKS1(S643P) reveals altered lipid arrangements in this region, suggesting a drug-resistant mechanism of the mutant enzyme. The structures, the catalytic mechanism and the molecular insights into drug-resistant mutations of FKS1 revealed in this study advance the mechanistic understanding of fungal ß-1,3-glucan biosynthesis and establish a foundation for developing new antifungal drugs by targeting FKS.


Subject(s)
Cryoelectron Microscopy , Glucosyltransferases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Antifungal Agents/pharmacology , beta-Glucans/metabolism , Catalytic Domain , Cell Membrane/chemistry , Cell Membrane/metabolism , Drug Resistance, Fungal/drug effects , Drug Resistance, Fungal/genetics , Echinocandins/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glucosyltransferases/ultrastructure , Microbial Sensitivity Tests , Mutation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure
6.
J Biol Chem ; 299(2): 102861, 2023 02.
Article in English | MEDLINE | ID: mdl-36603766

ABSTRACT

Phosphatidylinositol (PtdIns) transfer proteins (PITPs) enhance the activities of PtdIns 4-OH kinases that generate signaling pools of PtdIns-4-phosphate. In that capacity, PITPs serve as key regulators of lipid signaling in eukaryotic cells. Although the PITP phospholipid exchange cycle is the engine that stimulates PtdIns 4-OH kinase activities, the underlying mechanism is not understood. Herein, we apply an integrative structural biology approach to investigate interactions of the yeast PITP Sec14 with small-molecule inhibitors (SMIs) of its phospholipid exchange cycle. Using a combination of X-ray crystallography, solution NMR spectroscopy, and atomistic MD simulations, we dissect how SMIs compete with native Sec14 phospholipid ligands and arrest phospholipid exchange. Moreover, as Sec14 PITPs represent new targets for the development of next-generation antifungal drugs, the structures of Sec14 bound to SMIs of diverse chemotypes reported in this study will provide critical information required for future structure-based design of next-generation lead compounds directed against Sec14 PITPs of virulent fungi.


Subject(s)
Antifungal Agents , Drug Design , Phospholipid Transfer Proteins , Saccharomyces cerevisiae Proteins , Biological Transport/drug effects , Phosphatidylinositols/metabolism , Phospholipid Transfer Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Signal Transduction , Antifungal Agents/chemistry , Antifungal Agents/pharmacology
7.
Sci Rep ; 12(1): 1429, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082341

ABSTRACT

The passive transport of glucose and related hexoses in human cells is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT3 is a high-affinity glucose transporter primarily responsible for glucose entry in neurons. Changes in its expression have been implicated in neurodegenerative diseases and cancer. GLUT3 inhibitors can provide new ways to probe the pathophysiological role of GLUT3 and tackle GLUT3-dependent cancers. Through in silico screening of an ~ 8 million compounds library against the inward- and outward-facing models of GLUT3, we selected ~ 200 ligand candidates. These were tested for in vivo inhibition of GLUT3 expressed in hexose transporter-deficient yeast cells, resulting in six new GLUT3 inhibitors. Examining their specificity for GLUT1-5 revealed that the most potent GLUT3 inhibitor (G3iA, IC50 ~ 7 µM) was most selective for GLUT3, inhibiting less strongly only GLUT2 (IC50 ~ 29 µM). None of the GLUT3 inhibitors affected GLUT5, three inhibited GLUT1 with equal or twofold lower potency, and four showed comparable or two- to fivefold better inhibition of GLUT4. G3iD was a pan-Class 1 GLUT inhibitor with the highest preference for GLUT4 (IC50 ~ 3.9 µM). Given the prevalence of GLUT1 and GLUT3 overexpression in many cancers and multiple myeloma's reliance on GLUT4, these GLUT3 inhibitors may discriminately hinder glucose entry into various cancer cells, promising novel therapeutic avenues in oncology.


Subject(s)
Drug Discovery , Glucose Transporter Type 3/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Binding Sites , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/antagonists & inhibitors , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 5/antagonists & inhibitors , Glucose Transporter Type 5/chemistry , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry
8.
Molecules ; 26(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34641343

ABSTRACT

(-)-Epigallocatechin gallate (EGCG), the chief dietary constituent in green tea (Camellia sinensis), is relatively unstable under oxidative conditions. This study evaluated the use of non-thermal dielectric barrier discharge (DBD) plasma to improve the anti-digestive enzyme capacities of EGCG oxidation products. Pure EGCG was dissolved in an aqueous solution and irradiated with DBD plasma for 20, 40, and 60 min. The reactant, irradiated for 60 min, exhibited improved inhibitory properties against α-glucosidase and α-amylase compared with the parent EGCG. The chemical structures of these oxidation products 1-3 from the EGCG, irradiated with the plasma for 60 min, were characterized using spectroscopic methods. Among the oxidation products, EGCG quinone dimer A (1) showed the most potent inhibitory effects toward α-glucosidase and α-amylase with IC50 values of 15.9 ± 0.3 and 18.7 ± 0.3 µM, respectively. These values were significantly higher than that of the positive control, acarbose. Compound 1, which was the most active, was the most abundant in the plasma-irradiated reactant for 60 min according to quantitative high-performance liquid chromatography analysis. These results suggest that the increased biological capacity of EGCG can be attributed to the structural changes to EGCG in H2O, induced by cold plasma irradiation.


Subject(s)
Camellia sinensis/chemistry , Catechin/analogs & derivatives , Glycoside Hydrolase Inhibitors/chemistry , Plasma Gases/adverse effects , alpha-Amylases/antagonists & inhibitors , Animals , Catechin/chemistry , Catechin/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Molecular Structure , Oxidation-Reduction , Pancreas/enzymology , Plant Leaves/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Swine , Water/chemistry , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
9.
Open Biol ; 11(8): 200415, 2021 08.
Article in English | MEDLINE | ID: mdl-34343464

ABSTRACT

Protein S-acylation or palmitoylation is a widespread post-translational modification that consists of the addition of a lipid molecule to cysteine residues of proteins through a thioester bond. Palmitoylation and palmitoyltransferases (PATs) have been linked to several types of cancers, diseases of the central nervous system and many infectious diseases where pathogens use the host cell machinery to palmitoylate their effectors. Despite the central importance of palmitoylation in cell physiology and disease, progress in the field has been hampered by the lack of potent-specific inhibitors of palmitoylation in general, and of individual PATs in particular. Herein, we present a yeast-based method for the high-throughput identification of small molecules that inhibit protein palmitoylation. The system is based on a reporter gene that responds to the acylation status of a palmitoylation substrate fused to a transcription factor. The method can be applied to heterologous PATs such as human DHHC20, mouse DHHC21 and also a PAT from the parasite Giardia lamblia. As a proof-of-principle, we screened for molecules that inhibit the palmitoylation of Yck2, a substrate of the yeast PAT Akr1. We tested 3200 compounds and were able to identify a candidate molecule, supporting the validity of our method.


Subject(s)
Acyltransferases/antagonists & inhibitors , Lipoylation , Protozoan Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/metabolism , Small Molecule Libraries/pharmacology , Animals , Giardia lamblia/drug effects , Giardia lamblia/growth & development , Giardia lamblia/metabolism , High-Throughput Screening Assays , Humans , Mice , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Substrate Specificity
10.
Molecules ; 26(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299598

ABSTRACT

In this work we introduce a novel filtering and molecular modeling pipeline based on a fingerprint and descriptor similarity procedure, coupled with molecular docking and molecular dynamics (MD), to select potential novel quoinone outside inhibitors (QoI) of cytochrome bc1 with the aim of determining the same or different chromophores to usual. The study was carried out using the yeast cytochrome bc1 complex with its docked ligand (stigmatellin), using all the fungicides from FRAC code C3 mode of action, 8617 Drugbank compounds and 401,624 COCONUT compounds. The introduced drug repurposing pipeline consists of compound similarity with C3 fungicides and molecular docking (MD) simulations with final QM/MM binding energy determination, while aiming for potential novel chromophores and perserving at least an amide (R1HN(C=O)R2) or ester functional group of almost all up to date C3 fungicides. 3D descriptors used for a similarity test were based on the 280 most stable Padel descriptors. Hit compounds that passed fingerprint and 3D descriptor similarity condition and had either an amide or an ester group were submitted to docking where they further had to satisfy both Chemscore fitness and specific conformation constraints. This rigorous selection resulted in a very limited number of candidates that were forwarded to MD simulations and QM/MM binding affinity estimations by the ORCA DFT program. In this final step, stringent criteria based on (a) sufficiently high frequency of H-bonds; (b) high interaction energy between protein and ligand through the whole MD trajectory; and (c) high enough QM/MM binding energy scores were applied to further filter candidate inhibitors. This elaborate search pipeline led finaly to four Drugbank synthetic lead compounds (DrugBank) and seven natural (COCONUT database) lead compounds-tentative new inhibitors of cytochrome bc1. These eleven lead compounds were additionally validated through a comparison of MM/PBSA free binding energy for new leads against those obtatined for 19 QoIs.


Subject(s)
Electron Transport Complex III/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/enzymology , Drug Evaluation, Preclinical , Electron Transport Complex III/chemistry , Saccharomyces cerevisiae Proteins/chemistry
11.
Nat Commun ; 12(1): 3483, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108481

ABSTRACT

The hexameric AAA-ATPase Drg1 is a key factor in eukaryotic ribosome biogenesis and initiates cytoplasmic maturation of the large ribosomal subunit by releasing the shuttling maturation factor Rlp24. Drg1 monomers contain two AAA-domains (D1 and D2) that act in a concerted manner. Rlp24 release is inhibited by the drug diazaborine which blocks ATP hydrolysis in D2. The mode of inhibition was unknown. Here we show the first cryo-EM structure of Drg1 revealing the inhibitory mechanism. Diazaborine forms a covalent bond to the 2'-OH of the nucleotide in D2, explaining its specificity for this site. As a consequence, the D2 domain is locked in a rigid, inactive state, stalling the whole Drg1 hexamer. Resistance mechanisms identified include abolished drug binding and altered positioning of the nucleotide. Our results suggest nucleotide-modifying compounds as potential novel inhibitors for AAA-ATPases.


Subject(s)
ATPases Associated with Diverse Cellular Activities/chemistry , Adenosine Triphosphatases/chemistry , Boron Compounds/chemistry , Saccharomyces cerevisiae Proteins/chemistry , AAA Domain , ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Binding Sites , Boron Compounds/pharmacology , Drug Resistance/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Mutation , Nucleotides/chemistry , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
Iran Biomed J ; 25(4): 255-64, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33992037

ABSTRACT

Background: The methylotrophic yeast Pichia pastoris is an appealing production host for a variety of recombinant proteins, including biologics. In this sense, various genetic- and non-genetic-based techniques have been implemented to improve the production efficiency of this expression platform. Loss of supression (Los1) encodes a non-essential nuclear tRNA exporter in Saccharomyces cerevisiae, which its deletion extends replicative lifespan. Herein, a los1-deficient strain of P. pastoris was generated and characterized. Methods: A gene disruption cassette was prepared and transformed into an anti-CD22-expressing strain of P. pastoris. A δ los1 mutant was isolated and confirmed. The drug sensitivity of the mutant was also assessed. The growth pattern and the level of anti-CD22 single-chain variable fragment (scFv) expression were compared between the parent and mutant strains. Resuults: The los1 homologue was found to be a non-essential gene in P. pastoris. Furthermore, the susceptibility of los1 deletion strain to protein synthesis inhibitors was altered. This strain showed an approximately 1.85-fold increase in the extracellular level of anti-CD22 scFv (p < 0.05). The maximum concentrations of total proteins secreted by δ los1 and parent strains were 125 mg/L and 68 mg/L, respectively. Conclusion: The presented data suggest that the targeted disruption of los1 homologue in P. pastoris can result in a higher expression level of our target protein. Findings of this study may improve the current strategies used in optimizing the productivity of recombinant P. pastoris strains.


Subject(s)
Gene Deletion , Gene Targeting/methods , Nuclear Pore Complex Proteins/genetics , Recombinant Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales/genetics , Cell Survival/physiology , Nuclear Pore Complex Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors
13.
Cells ; 10(4)2021 04 13.
Article in English | MEDLINE | ID: mdl-33924665

ABSTRACT

Lithium hexafluorophosphate (LiPF6) is one of the leading electrolytes in lithium-ion batteries, and its usage has increased tremendously in the past few years. Little is known, however, about its potential environmental and biological impacts. In order to improve our understanding of the cytotoxicity of LiPF6 and the specific cellular response mechanisms to it, we performed a genome-wide screen using a yeast (Saccharomyces cerevisiae) deletion mutant collection and identified 75 gene deletion mutants that showed LiPF6 sensitivity. Among these, genes associated with mitochondria showed the most enrichment. We also found that LiPF6 is more toxic to yeast than lithium chloride (LiCl) or sodium hexafluorophosphate (NaPF6). Physiological analysis showed that a high concentration of LiPF6 caused mitochondrial damage, reactive oxygen species (ROS) accumulation, and ATP content changes. Compared with the results of previous genome-wide screening for LiCl-sensitive mutants, we found that oxidative phosphorylation-related mutants were specifically hypersensitive to LiPF6. In these deletion mutants, LiPF6 treatment resulted in higher ROS production and reduced ATP levels, suggesting that oxidative phosphorylation-related genes were important for counteracting LiPF6-induced toxicity. Taken together, our results identified genes specifically involved in LiPF6-modulated toxicity, and demonstrated that oxidative stress and ATP imbalance maybe the driving factors in governing LiPF6-induced toxicity.


Subject(s)
Fluorides/toxicity , Lithium/toxicity , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Phosphates/toxicity , Saccharomyces cerevisiae/drug effects , Adaptation, Physiological/drug effects , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Gene Expression Regulation, Fungal/drug effects , Gene Ontology , Genome-Wide Association Study , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Sequence Annotation , Oxidative Stress , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
14.
Int J Biol Macromol ; 166: 259-267, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33115652

ABSTRACT

Inhibition of α-glucosidase and non-enzymatic glycation is regarded as an effective method to prevent and treat type 2 diabetes and its complications. In this study, the inhibition of sinensetin on α-glucosidase and non-enzymatic glycation was studied with multi-spectroscopic techniques and molecular docking analysis. The results of fluorescence spectroscopy analysis indicated that sinensetin quenched the endogenous fluorescence of α-glucosidase in static manner. The binding of sinensetin with α-glucosidase was a spontaneous process primarily driven by hydrophobic interaction. At 298 K, the binding constant was (5.70 ± 0.12) × 104 L·mol-1 and the binding site number was 1. The conformation of α-glucosidase was altered by sinensetin, which was revealed by circular dichroism (CD), FTIR spectra, synchronous fluorescence and three-dimensional (3D) fluorescence spectroscopy methods. Molecular docking analysis demonstrated that sinensetin interacted with the amino acid residues of α-glucosidase, which might prevent the entrance of substrate, leading to the decrease of catalytic efficiency of α-glucosidase. Furthermore, glycation assays showed that sinensetin stabilized the structure of bovine serum albumins (BSA), interacted with BSA, strongly inhibited the formation of dityrosine, N'-formylkynurenine and advanced glycation end products (AGEs). This study provided useful information concerning sinensetin preventing and treating type 2 diabetes and its related complications.


Subject(s)
Flavonoids/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Molecular Docking Simulation , Saccharomyces cerevisiae Proteins/chemistry , alpha-Glucosidases/chemistry , Binding Sites , Flavonoids/pharmacology , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Kinetics , Kynurenine/analogs & derivatives , Kynurenine/chemistry , Kynurenine/metabolism , Protein Binding , Protein Stability , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Tyrosine/metabolism , alpha-Glucosidases/metabolism
15.
J Biochem Mol Toxicol ; 35(4): e22688, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33368871

ABSTRACT

A series of new benzofuran-1,3,4-oxadiazole containing 1,2,3-triazole-acetamides 12a-n as potential anti-α-glucosidase agents were designed and synthesized. α-Glucosidase inhibition assay demonstrated that all the synthesized compounds 12a-n (half-maximal inhibitory concentration [IC50 ] values in the range of 40.7 ± 0.3-173.6 ± 1.9 µM) were more potent than standard inhibitor acarbose (IC50 = 750.0 ± 12.5 µM). Among them, the most potent compound was compound 12c, with inhibitory activity around 19-fold higher than acarbose. Since the most potent compound inhibited α-glucosidase in a competitive mode, a docking study of this compound was also performed into the active site of α-glucosidase. In vitro and in silico toxicity assays of the title compounds were also performed.


Subject(s)
Acetamides , Glycoside Hydrolase Inhibitors , Oxadiazoles , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , alpha-Glucosidases/chemistry , Acetamides/chemical synthesis , Acetamides/chemistry , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/chemistry , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry
16.
Int J Mol Sci ; 21(21)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33143019

ABSTRACT

Environmental and intracellular factors often damage DNA, but multiple DNA repair pathways maintain genome integrity. In yeast, the 26S proteasome and its transcriptional regulator and substrate Rpn4 are involved in DNA damage resistance. Paradoxically, while proteasome dysfunction may induce hyper-resistance to DNA-damaging agents, Rpn4 malfunction sensitizes yeasts to these agents. Previously, we proposed that proteasome inhibition causes Rpn4 stabilization followed by the upregulation of Rpn4-dependent DNA repair genes and pathways. Here, we aimed to elucidate the key Rpn4 targets responsible for DNA damage hyper-resistance in proteasome mutants. We impaired the Rpn4-mediated regulation of candidate genes using the CRISPR/Cas9 system and tested the sensitivity of mutant strains to 4-NQO, MMS and zeocin. We found that the separate or simultaneous deregulation of 19S or 20S proteasome subcomplexes induced MAG1, DDI1, RAD23 and RAD52 in an Rpn4-dependent manner. Deregulation of RAD23, DDI1 and RAD52 sensitized yeast to DNA damage. Genetic, epigenetic or dihydrocoumarin-mediated RAD52 repression restored the sensitivity of the proteasome mutants to DNA damage. Our results suggest that the Rpn4-mediated overexpression of DNA repair genes, especially RAD52, defines the DNA damage hyper-resistant phenotype of proteasome mutants. The developed yeast model is useful for characterizing drugs that reverse the DNA damage hyper-resistance phenotypes of cancers.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal , Proteasome Endopeptidase Complex/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , CRISPR-Cas Systems , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Mutation , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
17.
Mech Ageing Dev ; 192: 111381, 2020 12.
Article in English | MEDLINE | ID: mdl-33045248

ABSTRACT

Saccharomyces cerevisiae MED2/YDL005C is a subunit of the mediator complex (Mediator), which is responsible for tightly controlling the transcription of protein-coding genes by mediating the interaction of RNA polymerase II with gene-specific transcription factors. Although a high-throughput analysis in yeast showed that the MED2 protein exhibits altered cellular localization under hypoxic stress, no specific function of MED2 has been described to date. In this study, we first provided evidence that MED2 is involved in the endoplasmic reticulum (ER) stress response and modulation of the replicative life span. We showed that deletion of MED2 leads to sensitivity to the ER stress inducer tunicamycin (TM) as well as a shortened replicative lifespan (RLS), accompanied by increased intracellular ROS levels and hyperpolarization of mitochondria. On the other hand, overexpression of MED2 in wild-type (WT) yeast enhanced TM resistance and extended the RLS. In addition, the IRE1-HAC1 pathway was essential for the TM resistance of MED2-overexpressing cells. Moreover, we showed that MED2 deficiency enhances ER unfolded protein response (UPR) activity compared to that in WT cells. Collectively, these results suggest the novel role of MED2 as a regulator in maintaining ER homeostasis and longevity.


Subject(s)
Cellular Senescence/genetics , Endoplasmic Reticulum Stress , Mediator Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tunicamycin/pharmacology , Antiviral Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Gene Expression Regulation , Mediator Complex/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Polymerase II , Repressor Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Stress, Physiological/physiology , Unfolded Protein Response/physiology
18.
Sci Rep ; 10(1): 14605, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32884066

ABSTRACT

Recent advances in CRISPR/Cas9 based genome editing have considerably advanced genetic engineering of industrial yeast strains. In this study, we report the construction and characterization of a toolkit for CRISPR activation and interference (CRISPRa/i) for a polyploid industrial yeast strain. In the CRISPRa/i plasmids that are available in high and low copy variants, dCas9 is expressed alone, or as a fusion with an activation or repression domain; VP64, VPR or Mxi1. The sgRNA is introduced to the CRISPRa/i plasmids from a double stranded oligonucleotide by in vivo homology-directed repair, allowing rapid transcriptional modulation of new target genes without cloning. The CRISPRa/i toolkit was characterized by alteration of expression of fluorescent protein-encoding genes under two different promoters allowing expression alterations up to ~ 2.5-fold. Furthermore, we demonstrated the usability of the CRISPRa/i toolkit by improving the tolerance towards wheat straw hydrolysate of our industrial production strain. We anticipate that our CRISPRa/i toolkit can be widely used to assess novel targets for strain improvement and thus accelerate the design-build-test cycle for developing various industrial production strains.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Genetic Engineering/methods , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Genome, Fungal , Plasmids , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics
19.
Nat Commun ; 11(1): 3387, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636417

ABSTRACT

Biosynthesis of glycosylphosphatidylinositol (GPI) is required for anchoring proteins to the plasma membrane, and is essential for the integrity of the fungal cell wall. Here, we use a reporter gene-based screen in Saccharomyces cerevisiae for the discovery of antifungal inhibitors of GPI-anchoring of proteins, and identify the oligocyclopropyl-containing natural product jawsamycin (FR-900848) as a potent hit. The compound targets the catalytic subunit Spt14 (also referred to as Gpi3) of the fungal UDP-glycosyltransferase, the first step in GPI biosynthesis, with good selectivity over the human functional homolog PIG-A. Jawsamycin displays antifungal activity in vitro against several pathogenic fungi including Mucorales, and in vivo in a mouse model of invasive pulmonary mucormycosis due to Rhyzopus delemar infection. Our results provide a starting point for the development of Spt14 inhibitors for treatment of invasive fungal infections.


Subject(s)
Antifungal Agents/pharmacology , Glycosyltransferases/antagonists & inhibitors , Polyketides/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Animals , Cell Proliferation , Disease Models, Animal , Fermentation , Genes, Reporter , Glycosylphosphatidylinositols/biosynthesis , HCT116 Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , K562 Cells , Lung/microbiology , Male , Mice , Mice, Inbred ICR , Mucorales , Multigene Family , Rhizopus , Saccharomyces cerevisiae
20.
PLoS Genet ; 16(6): e1008865, 2020 06.
Article in English | MEDLINE | ID: mdl-32603360

ABSTRACT

Fpr1 (FK506-sensitive proline rotamase 1), a protein of the FKBP12 (FK506-binding protein 12 kDa) family in Saccharomyces cerevisiae, is a primary target for the immunosuppressive agents FK506 and rapamycin. Fpr1 inhibits calcineurin and TORC1 (target of rapamycin complex 1) when bound to FK506 and rapamycin, respectively. Although Fpr1 is recognised to play a crucial role in the efficacy of these drugs, its physiological functions remain unclear. In a hmo1Δ (high mobility group family 1-deleted) yeast strain, deletion of FPR1 induced severe growth defects, which could be alleviated by increasing the copy number of RPL25 (ribosome protein of the large subunit 25), suggesting that RPL25 expression was affected in hmo1Δfpr1Δ cells. In the current study, extensive chromatin immunoprecipitation (ChIP) and ChIP-sequencing analyses revealed that Fpr1 associates specifically with the upstream activating sequences of nearly all RPG (ribosomal protein gene) promoters, presumably in a manner dependent on Rap1 (repressor/activator site binding protein 1). Intriguingly, Fpr1 promotes the binding of Fhl1/Ifh1 (forkhead-like 1/interacts with forkhead 1), two key regulators of RPG transcription, to certain RPG promoters independently of and/or cooperatively with Hmo1. Furthermore, mutation analyses of Fpr1 indicated that for transcriptional function on RPG promoters, Fpr1 requires its N-terminal domain and the binding surface for rapamycin, but not peptidyl-prolyl isomerase activity. Notably, Fpr1 orthologues from other species also inhibit TORC1 when bound to rapamycin, but do not regulate transcription in yeast, which suggests that these two functions of Fpr1 are independent of each other.


Subject(s)
High Mobility Group Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Calcineurin/metabolism , Chromatin Immunoprecipitation Sequencing , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Fungal , Genes, Fungal , High Mobility Group Proteins/genetics , Peptidylprolyl Isomerase/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Sirolimus/pharmacology , Tacrolimus/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL