Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Plant Sci ; 347: 112184, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38996874

ABSTRACT

Nervonic acid (C24:1) is a very-long-chain fatty acid that plays an imperative role in human brain development and other health benefits. In plants, 3-ketoacyl-CoA synthase (KCS) is the key rate-limiting enzyme for C24:1 biosynthesis. Xanthoceras sorbifolium is a valuable oil-producing economic woody species with abundant C24:1 in seed oils, but the key KCS gene responsible for C24:1 accumulation remains unknown. In this work, a correlation analysis between the transcript profiles of KCS and dynamic change of C24:1 content in developing seeds of X. sorbifolium were conducted to screen out three members of KCS, namely XsKCS4, XsKCS7 and XsKCS8, potentially involved in C24:1 biosynthesis. Of which, the XsKCS7 was highly expressed in developing seeds, while XsKCS4 and XsKCS8 displayed the highest expression in fruits and flowers, respectively. Overexpression of XsKCS4, XsKCS7 and XsKCS8 in yeast Saccharomyces cerevisiae and plant Arabidopsis thaliana indicated that only XsKCS7 possessed the ability to facilitate the biosynthesis of C24:1. These findings collectively suggested that XsKCS7 played a crucial role in specific regulation of C24:1 biosynthesis in X. sorbifolium seeds.


Subject(s)
Fatty Acids, Monounsaturated , Plant Proteins , Sapindaceae , Seeds , Seeds/genetics , Seeds/metabolism , Seeds/growth & development , Plant Proteins/genetics , Plant Proteins/metabolism , Sapindaceae/genetics , Sapindaceae/metabolism , Sapindaceae/enzymology , Sapindaceae/growth & development , Fatty Acids, Monounsaturated/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/enzymology , Arabidopsis/metabolism , Plants, Genetically Modified/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
2.
Plant J ; 119(1): 490-507, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38666591

ABSTRACT

The cuticle, consisting of cuticular wax and cutin, is a lipid membrane that seals the plant surface against environmental stress. ß-Ketoacyl-CoA synthases (KCSs) are condensing enzymes catalyzing crucial reactions elongating hydrocarbon chains into precursors for various cuticular wax components. Although many KCS genes were well characterized in various species, the functions of the closely related Arabidopsis KCS3, KCS12, KCS19 enzymes remained unclear. Here, we found KCS3 preferentially expressed in growing organs, especially in guard cells. kcs3 mutants and kcs3kcs12 double mutants displayed sepal fusion phenotypes, suggesting defects in cuticle formation. The mutants had decreased amounts of wax components with relatively short hydrocarbon chains in the developing organs but increased levels of wax compounds in mature organs. In contrast, kcs19 mutants showed seed fusion phenotypes and altered chain length distributions in seed suberin. Taken together, our results show that KCS12 and KCS3 share redundant functions in flower development, while KCS19 is involved in seed coat formation. All three condensing enzymes are involved in the elongation of C>18 hydrocarbon chains in young, actively expanding tissues.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/genetics , Arabidopsis/enzymology , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant , Flowers/genetics , Flowers/enzymology , Flowers/growth & development , Flowers/metabolism , Seeds/genetics , Seeds/growth & development , Seeds/metabolism , Seeds/enzymology , Waxes/metabolism , Mutation , Phenotype , Lipids , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism
3.
Appl Environ Microbiol ; 90(3): e0225623, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38415624

ABSTRACT

The last step of the initiation phase of fatty acid biosynthesis in most bacteria is catalyzed by the 3-ketoacyl-acyl carrier protein (ACP) synthase III (FabH). Pseudomonas syringae pv. syringae strain B728a encodes two FabH homologs, Psyr_3467 and Psyr_3830, which we designated PssFabH1 and PssFabH2, respectively. Here, we explored the roles of these two 3-ketoacyl-ACP synthase (KAS) III proteins. We found that PssFabH1 is similar to the Escherichia coli FabH in using acetyl-acetyl-coenzyme A (CoA ) as a substrate in vitro, whereas PssFabH2 uses acyl-CoAs (C4-C10) or acyl-ACPs (C6-C10). Mutant analysis showed that neither KAS III protein is essential for the de novo fatty acid synthesis and cell growth. Loss of PssFabH1 reduced the production of an acyl homoserine lactone (AHL) quorum-sensing signal, and this production was partially restored by overexpressing FabH homologs from other bacteria. AHL production was also restored by inhibiting fatty acid elongation and providing exogenous butyric acid. Deletion of PssFabH1 supports the redirection of acyl-ACP toward biosurfactant synthesis, which in turn enhances swarming motility. Our study revealed that PssFabH1 is an atypical KAS III protein that represents a new KAS III clade that functions in providing a critical fatty acid precursor, butyryl-ACP, for AHL synthesis.IMPORTANCEAcyl homoserine lactones (AHLs) are important quorum-sensing compounds in Gram-negative bacteria. Although their formation requires acylated acyl carrier proteins (ACPs), how the acylated intermediate is shunted from cellular fatty acid synthesis to AHL synthesis is not known. Here, we provide in vivo evidence that Pseudomonas syringae strain B728a uses the enzyme PssFabH1 to provide the critical fatty acid precursor butyryl-ACP for AHL synthesis. Loss of PssFabH1 reduces the diversion of butyryl-ACP to AHL, enabling the accumulation of acyl-ACP for synthesis of biosurfactants that contribute to bacterial swarming motility. We report that PssFabH1 and PssFabH2 each encode a 3-ketoacyl-acyl carrier protein synthase (KAS) III in P. syringae B728a. Whereas PssFabH2 is able to function in redirecting intermediates from ß-oxidation to fatty acid synthesis, PssFabH1 is an atypical KAS III protein that represents a new KAS III clade based on its sequence, non-involvement in cell growth, and novel role in AHL synthesis.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Acyl-Butyrolactones , Pseudomonas syringae/genetics , Pseudomonas syringae/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Fatty Acids/metabolism , Bacteria/metabolism , Escherichia coli/metabolism , Acetyl Coenzyme A/metabolism
4.
J Biol Chem ; 300(2): 105600, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38335573

ABSTRACT

The condensation of acetyl-CoA with malonyl-acyl carrier protein (ACP) by ß-ketoacyl-ACP synthase III (KAS III, FabH) and decarboxylation of malonyl-ACP by malonyl-ACP decarboxylase are the two pathways that initiate bacterial fatty acid synthesis (FAS) in Escherichia coli. In addition to these two routes, we report that Pseudomonas putida F1 ß-ketoacyl-ACP synthase I (FabB), in addition to playing a key role in fatty acid elongation, also initiates FAS in vivo. We report that although two P. putida F1 fabH genes (PpfabH1 and PpfabH2) both encode functional KAS III enzymes, neither is essential for growth. PpFabH1 is a canonical KAS III similar to E. coli FabH whereas PpFabH2 catalyzes condensation of malonyl-ACP with short- and medium-chain length acyl-CoAs. Since these two KAS III enzymes are not essential for FAS in P. putida F1, we sought the P. putida initiation enzyme and unexpectedly found that it was FabB, the elongation enzyme of the oxygen-independent unsaturated fatty acid pathway. P. putida FabB decarboxylates malonyl-ACP and condenses the acetyl-ACP product with malonyl-ACP for initiation of FAS. These data show that P. putida FabB, unlike the paradigm E. coli FabB, can catalyze the initiation reaction in FAS.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Pseudomonas putida , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acyl Carrier Protein/metabolism , Escherichia coli/metabolism , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Fatty Acids , Glycogen Synthase , Pseudomonas putida/genetics , Pseudomonas putida/metabolism
5.
J Ovarian Res ; 16(1): 81, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-37087461

ABSTRACT

BACKGROUND: Heat shock protein 60 (HSP60) is essential for the folding and assembly of newly imported proteins to the mitochondria. HSP60 is overexpressed in most types of cancer, but its association with ovarian cancer is still in dispute. SKOV3 and OVCAR3 were used as experimental models after comparing the expression level of mitochondrial HSP60 in a normal human ovarian epithelial cell line and four ovarian cancer cell lines. RESULTS: Low HSPD1 (Heat Shock Protein Family D (HSP60) Member 1) expression was associated with unfavorable prognosis in ovarian cancer patients. Knockdown of HSPD1 significantly promoted the proliferation and migration of ovarian cancer cells. The differentially expressed proteins after HSPD1 knockdown were enriched in the lipoic acid (LA) biosynthesis and metabolism pathway, in which mitochondrial 3-oxoacyl-ACP synthase (OXSM) was the most downregulated protein and responsible for lipoic acid synthesis. HSP60 interacted with OXSM and overexpression of OXSM or LA treatment could reverse proliferation promotion mediated by HSPD1 knockdown. CONCLUSIONS: HSP60 interacted with OXSM and maintained its stability. Knockdown of HSPD1 could promote the proliferation and migration of SKOV3 and OVCAR3 via lowering the protein level of OXSM and LA synthesis.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Cell Proliferation , Chaperonin 60 , Ovarian Neoplasms , Thioctic Acid , Female , Humans , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , Chaperonin 60/genetics , Chaperonin 60/metabolism , Heat-Shock Proteins , Mitochondrial Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Thioctic Acid/pharmacology
6.
J Agric Food Chem ; 71(1): 276-287, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36588523

ABSTRACT

Exploring new herbicide targets based on natural product derivatives is an important research aspect for the generation of innovative pesticides. Ferulic acid ethyl ester (FAEE), a natural product derivative from ferulic acid, has significant herbicidal activity mainly by inhibiting the normal growth of weed seedling roots. However, the FAEE target protein underlying its herbicidal activity has not been identified. In this study, we synthesized an FAEE probe to locate its site of action. We discovered that FAEE entry point was via the root tips. Fourteen major binding proteins were identified using Drug affinity responsive target stability (DARTS) combined with LC-MS/MS, which included 3-ketoacyl-acyl carrier protein synthase I (KAS I) and phenylalanine ammonia-lyase I (PAL I). The KAS I and PAL I proteins/genes expression was changed significantly after exposure to FAEE, as evidenced by combined transcriptomic and proteomic analysis. A molecular docking assay indicated that KAS I and FAEE had a strong binding ability. Combined with previous studies on FAEE mechanism of action, and based on our results, we conclude that FAEE targeting KAS I lead to the blockage of the fatty acid synthesis pathway and result in plant death.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Herbicides , Plant Roots , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Chromatography, Liquid , Esters , Fatty Acids/metabolism , Herbicides/pharmacology , Molecular Docking Simulation , Proteomics , Tandem Mass Spectrometry , Plant Roots/drug effects
7.
Chem Biodivers ; 20(2): e202201060, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36579401

ABSTRACT

Fatty acid biosynthesis is essential for bacterial survival. Of these promising targets, ß-ketoacyl-acyl carrier protein (ACP) synthase III (FabH) is the most attractive target. A series of novel 1,3,4-oxadiazole-2(3H)-thione derivatives containing 1,4-benzodioxane skeleton targeting FabH were designed and synthesized. These compounds were determined by 1 H-NMR, 13 C-NMR, MS and further confirmed by crystallographic diffraction study for compound 7m and 7n. Most of the compounds exhibited good inhibitory activity against bacteria by computer-assisted screening, antibacterial activity test and E. coli FabH inhibitory activity test, wherein compounds 7e and 7q exhibited the most significant inhibitory activities. Besides, compound 7q showed the best E. coli FabH inhibitory activity (IC50 =2.45 µΜ). Computational docking studies also showed that compound 7q interacts with FabH critical residues in the active site.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Escherichia coli Proteins , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria , Enzyme Inhibitors/chemistry , Escherichia coli/metabolism , Molecular Docking Simulation , Skeleton/metabolism , Thiones
8.
Molecules ; 27(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36234941

ABSTRACT

Chlorothricin (CHL) belongs to a spirotetronate antibiotic family produced by Streptomyces antibioticus that inhibits pyruvate carboxylase and malate dehydrogenase. For the biosynthesis of CHL, ChlB3 plays a crucial role by introducing the 6-methylsalicylic acid (6MSA) moiety to ChlB2, an acyl carrier protein (ACP). However, the structural insight and catalytic mechanism of ChlB3 was unclear. In the current study, the crystal structure of ChlB3 was solved at 3.1 Å-resolution and a catalytic mechanism was proposed on the basis of conserved residues of structurally related enzymes. ChlB3 is a dimer having the same active sites as CerJ (a structural homologous enzyme) and uses a KSIII-like fold to work as an acyltransferase. The relaxed substrate specificity of ChlB3 was defined by its catalytic efficiencies (kcat/Km) for non-ACP tethered synthetic substrates such as 6MSA-SNAC, acetyl-SNAC, and cyclohexonyl-SNAC. ChlB3 successfully detached the 6MSA moiety from 6MSA-SNAC substrate and this hydrolytic activity demonstrated that ChlB3 has the potential to catalyze non-ACP tethered substrates. Structural comparison indicated that ChlB3 belongs to FabH family and showed 0.6-2.5 Å root mean square deviation (RMSD) with structural homologous enzymes. Molecular docking and dynamics simulations were implemented to understand substrate active site and structural behavior such as the open and closed conformation of the ChlB3 protein. The resultant catalytic and substrate recognition mechanism suggested that ChlB3 has the potential to use non-native substrates and minimize the labor of expressing ACP protein. This versatile acyltransferase activity may pave the way for manufacturing CHL variants and may help to hydrolyze several thioester-based compounds.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Acyl Carrier Protein , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Sequence , Aminoglycosides , Anti-Bacterial Agents , Malate Dehydrogenase/metabolism , Molecular Docking Simulation , Pyruvate Carboxylase/metabolism , Substrate Specificity
9.
Front Cell Infect Microbiol ; 12: 1008213, 2022.
Article in English | MEDLINE | ID: mdl-36189349

ABSTRACT

Recent studies have reported the ß-ketoacyl-acyl carrier protein KasA as a druggable target for Mycobacterium tuberculosis. This review summarizes the current status of major classes of KasA inhibitors with an emphasis on significant contributions from structure-based design methods leveraging X-ray crystal structures of KasA alone and in complex with inhibitors. The issues addressed within each inhibitor class are discussed while detailing the characterized interactions with KasA and structure-activity relationships. A critical analysis of these findings should lay the foundation for new KasA inhibitors to study the basic biology of M. tuberculosis and to form the basis of new antitubercular molecules of clinical significance with activity against drug-sensitive and drug-resistant infections.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Mycobacterium tuberculosis , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acyl Carrier Protein , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/metabolism
10.
J Biol Chem ; 298(10): 102496, 2022 10.
Article in English | MEDLINE | ID: mdl-36115459

ABSTRACT

Very long chain fatty acids (VLCFAs) are precursors to sphingolipids, glycerophospholipids, and plant cuticular waxes. In plants, members of a large 3-ketoacyl-CoA synthase (KCS) gene family catalyze the substrate-specific elongation of VLCFAs. Although it is well understood that KCSs have evolved to use diverse substrates, the underlying molecular determinants of their specificity are still unclear. In this study, we exploited the sequence similarity of a KCS gene cluster from Populus trichocarpa to examine the evolution and molecular determinants of KCS substrate specificity. Functional characterization of five members (PtKCS1, 2, 4, 8, 9) in yeast showed divergent product profiles based on VLCFA length, saturation, and position of the double bond. In addition, homology models, rationally designed chimeras, and site-directed mutants were used to identify two key regions (helix-4 and position 277) as being major determinants of substrate specificity. These results were corroborated with chimeras involving a more distantly related KCS, PtCER6 (the poplar ortholog of the Arabidopsis CER6), and used to show that helix-4 is necessary for the modulatory effect of PtCER2-like5 on KCS substrate specificity. The role of position 277 in limiting product length was further tested by substitution with smaller amino acids, which shifted specificity toward longer products. Finally, treatment with KCS inhibitors (K3 herbicides) showed varying inhibitor sensitivities between the duplicated paralogs despite their sequence similarity. Together, this work sheds light on the molecular mechanisms driving substrate diversification in the KCS family and lays the groundwork for tailoring the production of specific VLCFAs.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Arabidopsis , Populus , Substrate Specificity , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Populus/genetics , Populus/metabolism , Fatty Acids/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Multigene Family , Plants/metabolism , Coenzyme A/metabolism
11.
Acta Crystallogr D Struct Biol ; 78(Pt 9): 1171-1179, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36048156

ABSTRACT

Ketosynthases (KSs) catalyse essential carbon-carbon bond-forming reactions in fatty-acid biosynthesis using a two-step, ping-pong reaction mechanism. In Escherichia coli, there are two homodimeric elongating KSs, FabB and FabF, which possess overlapping substrate selectivity. However, FabB is essential for the biosynthesis of the unsaturated fatty acids (UFAs) required for cell survival in the absence of exogenous UFAs. Additionally, FabB has reduced activity towards substrates longer than 12 C atoms, whereas FabF efficiently catalyses the elongation of saturated C14 and unsaturated C16:1 acyl-acyl carrier protein (ACP) complexes. In this study, two cross-linked crystal structures of FabB in complex with ACPs functionalized with long-chain fatty-acid cross-linking probes that approximate catalytic steps were solved. Both homodimeric structures possess asymmetric substrate-binding pockets suggestive of cooperative relationships between the two FabB monomers when engaged with C14 and C16 acyl chains. In addition, these structures capture an unusual rotamer of the active-site gating residue, Phe392, which is potentially representative of the catalytic state prior to substrate release. These structures demonstrate the utility of mechanism-based cross-linking methods to capture and elucidate conformational transitions accompanying KS-mediated catalysis at near-atomic resolution.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , Escherichia coli Proteins , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Carbon/metabolism , Catalysis , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Fatty Acid Synthase, Type II , Fatty Acids, Unsaturated/metabolism
12.
Curr Drug Discov Technol ; 19(2): e110122200137, 2022.
Article in English | MEDLINE | ID: mdl-35021976

ABSTRACT

Since the early twentieth century, with the isolation of penicillin and streptomycin in the 1940s, the modern era of anti-infective drug development has gained momentum. Due to the enormous success of early drug discovery, many infectious diseases were successfully prevented and eradicated. However, this initial hope was wrongheaded, and pathogens evolved as a significant threat to human health. Drug resistance develops as a result of natural selection's relentless pressure, necessitating the identification of new drug targets and the creation of chemotherapeutics that bypass existing drug resistance mechanisms. Fatty acid biosynthesis (FAS) is a crucial metabolic mechanism for bacteria during their growth and development. Several crucial enzymes involved in this biosynthetic pathway have been identified as potential targets for new antibacterial agents. In Escherichia coli (E. coli), this pathway has been extensively investigated. The present review focuses on progress in the development of Kas A, Kas B, and Fab H inhibitors as mono-therapeutic antibiotics.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Escherichia coli , Fatty Acids/metabolism , Humans
13.
Plant Sci ; 310: 110972, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34315590

ABSTRACT

Cuticular wax covers the surface of fleshy fruit and plays a protective role in fruit development and postharvest storage, including reducing fruit water loss, resisting biotic and abiotic stress and affecting fruit glossiness. The ß-ketoacyl-CoA synthase (KCS) is the rate-limiting enzyme of very long chain fatty acids (VLCFAs) synthesis, which provides precursors for the synthesis of cuticular wax. In this study, a total of 96 KCS genes were identified in six Citrinae species, including 13, 16, 21, 14, 16 and 16 KCS genes in the primitive species (Atalantia buxifolia), the wild species (Citrus ichangensis), and four cultivated species (Citrus medica, Citrus grandis, Citrus sinensis and Citrus clementina), respectively. Compared with primitive species, wild and cultivated species showed expansion of KCS gene family. Evolutionary analysis of KCS gene family indicated that uneven gain and loss of genes resulted in variable numbers of KCS genes in Citrinae, and KCS genes have undergone purifying selection. Expression profiles in C. sinensis revealed that the KCS genes had diverse expression patterns among various tissues. Furthermore, CsKCS2 and CsKCS11 were predominantly expressed in the flavedo and their expression increased sharply with ripening. Subcellular localization analysis indicated that CsKCS2 and CsKCS11 were located in the endoplasmic reticulum. Further, heterologous expression of CsKCS2 and CsKCS11 in Arabidopsis significantly increased the content of cuticular wax in leaves. Thus, CsKCS2 and CsKCS11 are involved in the accumulation of fruit cuticular wax at ripening. This work will facilitate further functional verification and understanding of the evolution of KCS genes in Citrinae.


Subject(s)
Fruit/metabolism , Plant Leaves/metabolism , Plant Proteins/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Citrus/genetics , Citrus/metabolism , Fruit/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Plant Leaves/genetics , Plant Proteins/genetics , Waxes/metabolism
14.
ChemMedChem ; 16(17): 2715-2726, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34189850

ABSTRACT

FabF (3-oxoacyl-[acyl-carrier-protein] synthase 2), which catalyses the rate limiting condensation reaction in the fatty acid synthesis II pathway, is an attractive target for new antibiotics. Here, we focus on FabF from P. aeruginosa (PaFabF) as antibiotics against this pathogen are urgently needed. To facilitate exploration of this target we have set up an experimental toolbox consisting of binding assays using bio-layer interferometry (BLI) as well as saturation transfer difference (STD) and WaterLOGSY NMR in addition to robust conditions for structure determination. The suitability of the toolbox to support structure-based design of FabF inhibitors was demonstrated through the validation of hits obtained from virtual screening. Screening a library of almost 5 million compounds resulted in 6 compounds for which binding into the malonyl-binding site of FabF was shown. For one of the hits, the crystal structure in complex with PaFabF was determined. Based on the obtained binding mode, analogues were designed and synthesised, but affinity could not be improved. This work has laid the foundation for structure-based exploration of PaFabF.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Biological Products/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Pseudomonas aeruginosa/drug effects , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/isolation & purification , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Ligands , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pseudomonas aeruginosa/enzymology
15.
J Biol Chem ; 297(2): 100920, 2021 08.
Article in English | MEDLINE | ID: mdl-34181948

ABSTRACT

The Pseudomonas putida F1 genome contains five genes annotated as encoding 3-ketoacyl-acyl carrier protein (ACP) synthases. Four are annotated as encoding FabF (3-ketoacyl-ACP synthase II) proteins, and the fifth is annotated as encoding a FabB (3-ketoacyl-ACP synthase I) protein. Expression of one of the FabF proteins, FabF2, is cryptic in the native host and becomes physiologically important only when the repressor controlling fabF2 transcription is inactivated. When derepressed, FabF2 can functionally replace FabB, and when expressed from a foreign promoter, had weak FabF activity. Complementation of Escherichia coli fabB and fabF mutant strains with high expression showed that P. putida fabF1 restored E. coli fabF function, whereas fabB restored E. coli fabB function and fabF2 restored the functions of both E. coli fabF and fabB. The P. putida ΔfabF1 deletion strain was almost entirely defective in synthesis of cis-vaccenic acid, whereas the ΔfabB strain is an unsaturated fatty acid (UFA) auxotroph that accumulated high levels of spontaneous suppressors in the absence of UFA supplementation. This was due to increased expression of fabF2 that bypasses loss of fabB because of the inactivation of the regulator, Pput_2425, encoded in the same operon as fabF2. Spontaneous suppressor accumulation was decreased by high levels of UFA supplementation, whereas competition by the P. putida ß-oxidation pathway gave increased accumulation. The ΔfabB ΔfabF2 strain is a stable UFA auxotroph indicating that suppressor accumulation requires FabF2 function. However, at low concentrations of UFA supplementation, the ΔfabF2 ΔPput_2425 double-mutant strain still accumulated suppressors at low UFA concentrations.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Fatty Acids, Unsaturated/biosynthesis , Pseudomonas putida/metabolism , Genetic Complementation Test
16.
Acta Chim Slov ; 68(1): 88-101, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34057529

ABSTRACT

A detailed description of the two new pyridine ligands, (2E,3Z)-3-[2-(3-chloropyridin-2-yl)hydrazinylidene]-N-hydroxybutan-2-imine and 3-chloro-2-(2Z)-2-[1-(4 nitrophenyl)ethylidene]hydrazinyl, is reported. The synthesized compounds were characterized by spectroscopic studies, spectral features were performed by TD-DFT calculations. New-generation pyridine ligand of HL2 was also determinate by single-crystal X-ray diffraction and Hirshfeld surface analysis with two-dimensional fingerprint plots was used to analyze intermolecular interactions in crystals. Molecular-docking was performed to investigate the binding areas of chemical compounds, and the results showed the inhibitory activity of the studied HL1 and HL2 against E. coli. The results of the current study revealed the drug-likeness and bioactive properties of the ligands.


Subject(s)
Oximes/chemistry , Pyridines/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , DNA Gyrase/metabolism , Density Functional Theory , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Ligands , Models, Chemical , Molecular Docking Simulation , Molecular Structure , Oximes/chemical synthesis , Oximes/metabolism , Oximes/pharmacokinetics , Protein Binding , Pyridines/chemical synthesis , Pyridines/metabolism , Pyridines/pharmacokinetics , X-Ray Diffraction
17.
Plant Physiol ; 186(3): 1606-1615, 2021 07 06.
Article in English | MEDLINE | ID: mdl-33779764

ABSTRACT

Physical dormancy in seeds exists widely in seed plants and plays a vital role in maintaining natural seed banks. The outermost cuticle of the seed coat forms a water-impermeable layer, which is critical for establishing seed physical dormancy. We previously set up the legume plant Medicago truncatula as an excellent model for studying seed physical dormancy, and our studies revealed that a class II KNOTTED-like homeobox, KNOX4, is a transcription factor critical for controlling hardseededness. Here we report the function of a seed coat ß-ketoacyl-CoA synthase, KCS12. The expression level of KCS12 is significantly downregulated in the knox4 mutant. The KCS12 gene is predominantly expressed in the seed coat, and seed development in the M. truncatula kcs12 mutant is altered. Further investigation demonstrated that kcs12 mutant seeds lost physical dormancy and were able to absorb water without scarification treatment. Chemical analysis revealed that concentrations of C24:0 lipid polyester monomers are significantly decreased in mutant seeds, indicating that KCS12 is an enzyme that controls the production of very long chain lipid species in the seed coat. A chromatin immunoprecipitation assay demonstrated that the expression of KCS12 in the seed coat is directly regulated by the KNOX4 transcription factor. These findings define a molecular mechanism by which KNOX4 and KCS12 control formation of the seed coat and seed physical dormancy.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Germination/genetics , Medicago truncatula/growth & development , Medicago truncatula/genetics , Medicago truncatula/metabolism , Plant Dormancy/genetics , Seeds/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , Gene Expression Regulation, Plant , Genes, Homeobox , Genes, Plant , Genetic Variation , Genotype , Germination/physiology , Plant Dormancy/physiology , Seeds/growth & development , Seeds/metabolism
18.
Commun Biol ; 4(1): 340, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727677

ABSTRACT

Fatty acid biosynthesis (FAB) is an essential and highly conserved metabolic pathway. In bacteria, this process is mediated by an elaborate network of protein•protein interactions (PPIs) involving a small, dynamic acyl carrier protein that interacts with dozens of other partner proteins (PPs). These PPIs have remained poorly characterized due to their dynamic and transient nature. Using a combination of solution-phase NMR spectroscopy and protein-protein docking simulations, we report a comprehensive residue-by-residue comparison of the PPIs formed during FAB in Escherichia coli. This technique describes and compares the molecular basis of six discrete binding events responsible for E. coli FAB and offers insights into a method to characterize these events and those in related carrier protein-dependent pathways.


Subject(s)
Acyl Carrier Protein/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Fatty Acid Synthase, Type II/metabolism , Fatty Acids/biosynthesis , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acetyltransferases/metabolism , Alcohol Oxidoreductases/metabolism , Binding Sites , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Lysophospholipase/metabolism , Molecular Docking Simulation , Periplasmic Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proton Magnetic Resonance Spectroscopy
19.
F1000Res ; 102021.
Article in English | MEDLINE | ID: mdl-35136566

ABSTRACT

Background: FabB (3-oxoacyl-[acyl-carrier-protein] synthase 1) is part of the fatty acid synthesis II pathway found in bacteria and a potential target for antibiotics. The enzyme catalyses the Claisen condensation of malonyl-ACP (acyl carrier protein) with acyl-ACP via an acyl intermediate. Here, we report the crystal structure of the intermediate-mimicking Pseudomonas aeruginosa FabB ( PaFabB) C161A variant. Methods: His-tagged PaFabB C161A was expressed in E.coli Rosetta DE3 pLysS cells, cleaved by TEV protease and purified using affinity and size exclusion chromatography. Commercial screens were used to identify suitable crystallization conditions which were subsequently improved to obtain well diffracting crystals. Results: We developed a robust and efficient system for recombinant expression of PaFabB C161A. Conditions to obtain well diffracting crystals were established. The crystal structure of PaFabB C161A was solved by molecular replacement at 1.3 Å resolution. Conclusions: The PaFabB C161A crystal structure can be used as a template to facilitate the design of FabB inhibitors.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Anti-Bacterial Agents/pharmacology , Escherichia coli , Fatty Acids , Pseudomonas aeruginosa
20.
Med Chem ; 17(5): 474-484, 2021.
Article in English | MEDLINE | ID: mdl-31763975

ABSTRACT

BACKGROUND: Antimicrobial resistance is a persistent problem regarding infection treatment and calls for developing new antimicrobial agents. Inhibition of bacterial ß-ketoacyl acyl carrier protein synthase III (FabH), which catalyzes the condensation reaction between a CoAattached acetyl group and an ACP-attached malonyl group in bacteria is an interesting strategy to find new antibacterial agents. OBJECTIVE: The aim of this work was to design and synthesize arylsulfonylhydrazones potentially FabH inhibitors and evaluate their antimicrobial activity. METHODS: MIC50 values of sulfonylhydrazones against E. coli and S. aureus were determined. Antioxidant activity was evaluated by DPPH (1-1'-diphenyl-2-picrylhydrazyl) assay and cytotoxicity against LL24 lung fibroblast cells was verified by MTT method. Principal component analysis (PCA) was performed in order to suggest a structure-activity relationship. Molecular docking allowed to propose sulfonylhydrazones interactions with FabH. RESULTS: The most active compound showed activity against S. aureus and E. coli, with MIC50 = 0.21 and 0.44 µM, respectively. PCA studies correlated better activity to lipophilicity and molecular docking indicated that sulfonylhydrazone moiety is important to hydrogen-bond with FabH while methylcatechol ring performs π-π stacking interaction. The DPPH assay revealed that some sulfonylhydrazones derived from the methylcatechol series had antioxidant activity. None of the evaluated compounds was cytotoxic to human lung fibroblast cells, suggesting that the compounds might be considered safe at the tested concentration. CONCLUSION: Arylsufonylhydrazones is a promising scaffold to be explored for the design of new antimicrobial agents.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydrazones/pharmacology , Sulfonamides/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Catalytic Domain , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Fatty Acid Synthase, Type II/antagonists & inhibitors , Fatty Acid Synthase, Type II/chemistry , Fatty Acid Synthase, Type II/metabolism , Hydrazones/chemical synthesis , Hydrazones/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Principal Component Analysis , Protein Binding , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL