Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Microbiol ; 116(5): 1315-1327, 2021 11.
Article in English | MEDLINE | ID: mdl-34597430

ABSTRACT

Biotin is an essential metabolic cofactor and de novo biotin biosynthetic pathways are widespread in microorganisms and plants. Biotin synthetic genes are generally found clustered into bio operons to facilitate tight regulation since biotin synthesis is a metabolically expensive process. Dethiobiotin synthetase (DTBS) catalyzes the penultimate step of biotin biosynthesis, the formation of 7,8-diaminononanoate (DAPA). In Escherichia coli, DTBS is encoded by the bio operon gene bioD. Several studies have reported transcriptional activation of ynfK a gene of unknown function, under anaerobic conditions. Alignments of YnfK with BioD have led to suggestions that YnfK has DTBS activity. We report that YnfK is a functional DTBS, although an enzyme of poor activity that is poorly expressed. Supplementation of growth medium with DAPA or substitution of BioD active site residues for the corresponding YnfK residues greatly improved the DTBS activity of YnfK. We confirmed that FNR activates transcriptional level of ynfK during anaerobic growth and identified the FNR binding site of ynfK. The ynfK gene is well conserved in γ-proteobacteria.


Subject(s)
Biotin/biosynthesis , Biotin/genetics , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Amino Acid Sequence , Amino Acids, Diamino/metabolism , Anaerobiosis , Binding Sites , Biosynthetic Pathways , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Iron-Sulfur Proteins/metabolism , Operon , Phylogeny
2.
J Biochem ; 170(4): 453-461, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-33982090

ABSTRACT

Proximity-dependent biotin identification (BioID) is a useful method to identify unknown protein-protein interactions. Few reports have described genetically engineered knock-in mouse models for in vivo BioID. Thus, little is known about the proper method for biotin administration and which tissues are applicable. Here, we established a BioID knock-in mouse model of Brain and Muscle ARNT-Like 1 (BMAL1) and the BirA biotin ligase with R118G mutation (BirA*). The BMAL1-BioID mouse model was used to investigate the effect of biotin diet feeding on protein biotinylation in several tissues. The BMAL1-BirA* fusion protein-retained proper intracellular localization of BMAL1 and binding to CLOCK protein in HEK293T cells. A biotin labelling assay in mouse embryonic fibroblasts revealed the protein biotinylation activity of BMAL1-BirA* expressed in knock-in mouse cells depending on biotin supplementation. Lastly, feeding a 0.5% biotin diet for 7 days induced protein biotinylation in the brain, heart, testis and liver of BMAL1-BioID mice without adverse effects on spermatogenesis. In the kidney, the biotin diet increased biotinylated protein levels in BMAL1-BioID and control mice, suggesting the existence of endogenous biotinylation activity. These results provide valuable information to optimize the in vivo BioID procedure.


Subject(s)
ARNTL Transcription Factors/metabolism , Biotin/pharmacology , Protein Interaction Mapping/methods , Animals , Biotin/administration & dosage , Biotinylation/methods , Brain/metabolism , CLOCK Proteins/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Diet/methods , Fibroblasts/metabolism , Genotype , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Muscles/metabolism , Staining and Labeling/methods
3.
Anal Chim Acta ; 1101: 120-128, 2020 Mar 08.
Article in English | MEDLINE | ID: mdl-32029102

ABSTRACT

Simple and easy to engineer metal-sensing molecules that are capable of differentiating metal ions and producing metal-specific signals are highly desirable. Metal ions affect the thermal stability of proteins by increasing or decreasing their resistance to unfolding. This work illustrates a new strategy for designing bivalent fluorescent fusion proteins capable of differentiating metal ions in solution through their distinct effects on a protein's thermal stability. A new dual purpose metal sensor was developed consisting of biotin protein ligase (BirA) from B. pseudomallei (Bp) fused to green fluorescent protein (GFP). When coupled with differential scanning fluorimetry of GFP-tagged proteins (DSF-GTP) for signal-transduction detection, Bp BirA-GFP yields distinct protein unfolding signatures with Zn(II) and Cu(II) ions in aqueous solutions. The limit of detection of the system is ∼1 µM for both metal species. The system can be used in a variety of high-throughput assay formats including for the screening of metal-binding proteins and chelators. Bp BirA-GFP has also the additional benefit of being useful in Cu(II) ion field-testing applications through simple visual observation of a temperature-dependent loss of fluorescence. Bp BirA-GFP is the first example of a 2protein-based dual purpose Cu(II) and Zn(II) ion sensor compatible with two different yet complementary signal-transduction detection systems.


Subject(s)
Carbon-Nitrogen Ligases/chemistry , Copper/analysis , Green Fluorescent Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Zinc/analysis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biosensing Techniques/methods , Biotin/metabolism , Burkholderia pseudomallei/enzymology , Carbon-Nitrogen Ligases/metabolism , Copper/metabolism , Fluorometry/methods , Green Fluorescent Proteins/metabolism , Limit of Detection , Proof of Concept Study , Protein Binding , Recombinant Fusion Proteins/metabolism , Zinc/metabolism
4.
Appl Microbiol Biotechnol ; 104(1): 119-130, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31776607

ABSTRACT

L-Theanine is a unique non-protein amino acid found in tea plants that has been shown to possess numerous functional properties relevant to food science and human nutrition. L-Theanine has been commercially developed as a valuable additive for use in food and beverages, and its market is expected to expand substantially if the production cost can be lowered. Although the enzymatic approach holds considerable potential for use in L-theanine production, demand exists for developing more tractable methods (than those currently available) that can be implemented under mild conditions and will reduce operational procedures and cost. Here, we sought to engineer fermentative production of L-theanine in Corynebacterium glutamicum, an industrially safe host. For L-theanine synthesis, we used γ-glutamylmethylamide synthetase (GMAS), which catalyzes the ATP-dependent ligation of L-glutamate and ethylamine. First, distinct GMASs were expressed in C. glutamicum wild-type ATCC 13032 strain and GDK-9, an L-glutamate overproducing strain, to produce L-theanine upon ethylamine addition to the hosts. Second, the L-glutamate exporter in host cells was disrupted, which markedly increased the L-theanine titer in GDK-9 cells and almost eliminated the accumulation of L-glutamate in the culture medium. Third, a chromosomally gmasMm-integrated L-alanine producer was constructed and used, attempting to synthesize ethylamine endogenously by expressing plant-derived L-serine/L-alanine decarboxylases; however, these enzymes showed no L-alanine decarboxylase activity under our experimental conditions. The optimal engineered strain that we ultimately created produced ~ 42 g/L L-theanine, with a yield of 19.6%, in a 5-L fermentor. This is the first report of fermentative production of L-theanine achieved using ethylamine supplementation.


Subject(s)
Corynebacterium glutamicum/metabolism , Fermentation , Glutamates/biosynthesis , Metabolic Engineering/methods , Adenosine Triphosphate/metabolism , Carbon-Nitrogen Ligases/metabolism , Ethylamines/metabolism , Glutamic Acid/metabolism , Industrial Microbiology
5.
ACS Chem Biol ; 14(8): 1760-1766, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31260252

ABSTRACT

The Gram-negative bacterium Francisella tularensis secretes the siderophore rhizoferrin to scavenge necessary iron from the environment. Rhizoferrin, also produced by a variety of fungi and bacteria, comprises two citrate molecules linked by amide bonds to a central putrescine (diaminobutane) moiety. Genetic analysis has determined that rhizoferrin production in F. tularensis requires two enzymes: FslA, a siderophore synthetase of the nonribosomal peptide synthetase-independent siderophore synthetase (NIS) family, and FslC, a pyridoxal-phosphate-dependent decarboxylase. To discern the steps in the biosynthetic pathway, we tested F. tularensis strain LVS and its ΔfslA and ΔfslC mutants for the ability to incorporate potential precursors into rhizoferrin. Unlike putrescine supplementation, supplementation with ornithine greatly enhanced siderophore production by LVS. Radioactivity from L-[U-14C] ornithine, but not from L-[1-14C] ornithine, was efficiently incorporated into rhizoferrin by LVS. Although neither the ΔfslA nor the ΔfslC mutant produced rhizoferrin, a putative siderophore intermediate labeled by both [U-14C] ornithine and [1-14C] ornithine was secreted by the ΔfslC mutant. Rhizoferrin was identified by liquid chromatography and mass spectrometry in LVS culture supernatants, while citryl-ornithine was detected as the siderophore intermediate in the culture supernatant of the ΔfslC mutant. Our findings support a three-step pathway for rhizoferrin production in Francisella; unlike the fungus Rhizopus delemar, where putrescine functions as a primary precursor for rhizoferrin, biosynthesis in Francisella preferentially starts with ornithine as the substrate for FslA-mediated condensation with citrate. Decarboxylation of this citryl ornithine intermediate by FslC is necessary for a second condensation reaction with citrate to produce rhizoferrin.


Subject(s)
Citrates/metabolism , Ferric Compounds/metabolism , Francisella tularensis/metabolism , Ornithine/analogs & derivatives , Ornithine/metabolism , Siderophores/biosynthesis , Bacterial Proteins/metabolism , Carbon Radioisotopes , Carbon-Nitrogen Ligases/metabolism , Carboxy-Lyases/metabolism , Francisella tularensis/enzymology
6.
J Cell Sci ; 132(11)2019 05 31.
Article in English | MEDLINE | ID: mdl-31064814

ABSTRACT

The use of proximity-dependent biotinylation assays coupled to mass spectrometry (PDB-MS) has changed the field of protein-protein interaction studies. However, despite the recurrent and successful use of BioID-based protein-protein interactions screening in mammalian cells, the implementation of PDB-MS in yeast has not been effective. Here, we report a simple and rapid approach in yeast to effectively screen for proximal and interacting proteins in their natural cellular environment by using TurboID, a recently described version of the BirA biotin ligase. Using the protein arginine methyltransferase Rmt3 and the RNA exosome subunits, Rrp6 and Dis3, the application of PDB-MS in yeast by using TurboID was able to recover protein-protein interactions previously identified using other biochemical approaches and provided new complementary information for a given protein bait. The development of a rapid and effective PDB assay that can systematically analyze protein-protein interactions in living yeast cells opens the way for large-scale proteomics studies in this powerful model organism.


Subject(s)
Biotinylation/methods , Protein Interaction Mapping/methods , Protein Interaction Maps/physiology , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Carbon-Nitrogen Ligases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Mass Spectrometry/methods , Protein Interaction Maps/genetics , Protein-Arginine N-Methyltransferases/metabolism , Proteomics/methods , Ribonucleases/metabolism , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism
7.
ACS Synth Biol ; 8(2): 251-256, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30702274

ABSTRACT

The antisense RNA (asRNA) strategy is commonly used to block protein expression and downregulate the contents of metabolites in several microorganisms. Here, we show that the asRNA strategy can also be used to block gfp expression in Bacillus subtilis TS1726, which could further be utilized in the identification of new genes and functions. Via application of this strategy, biotin carboxylase II encoded by yngH (GeneID 939474) was identified to play a more significant role in maintaining acetyl-CoA carboxylase (ACCase) activity and enhancing surfactin synthesis compared to those of other ACCase subunits. The yngH gene was then overexpressed in the engineered strain B. subtilis TS1726(yngH). The surfactin titer of TS1726(yngH) increased to 13.37 g/L in a flask culture, representing a 43% increase compared to that of parental strain TS1726. This strategy opens the door to achieving large-scale production and broad application of surfactin.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Bacillus subtilis/genetics , Carbon-Nitrogen Ligases/metabolism , RNA, Antisense/genetics , Acetyl-CoA Carboxylase/genetics , Biotin/genetics , Biotin/metabolism , Carbon-Nitrogen Ligases/genetics
8.
Sci Rep ; 8(1): 12538, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135446

ABSTRACT

Cell penetrating peptides (CPPs) offer great potential to deliver therapeutic molecules to previously inaccessible intracellular targets. However, many CPPs are inefficient and often leave their attached cargo stranded in the cell's endosome. We report a versatile platform for the isolation of peptides delivering a wide range of cargos into the cytoplasm of cells. We used this screening platform to identify multiple "Phylomer" CPPs, derived from bacterial and viral genomes. These peptides are amenable to conventional sequence optimization and engineering approaches for cell targeting and half-life extension. We demonstrate potent, functional delivery of protein, peptide, and nucleic acid analog cargos into cells using Phylomer CPPs. We validate in vivo activity in the cytoplasm, through successful transport of an oligonucleotide therapeutic fused to a Phylomer CPP in a disease model for Duchenne's muscular dystrophy. This report thus establishes a discovery platform for identifying novel, functional CPPs to expand the delivery landscape of druggable intracellular targets for biological therapeutics.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods , Animals , Bacteriophage T7 , Biotinylation , CHO Cells , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/toxicity , Circular Dichroism , Cricetulus , Disease Models, Animal , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscular Dystrophy, Duchenne/drug therapy , Peptide Library , Repressor Proteins/genetics , Repressor Proteins/metabolism
9.
Appl Microbiol Biotechnol ; 102(8): 3793-3803, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29508030

ABSTRACT

Biotin (Vitamin B7) is a critical enzyme co-factor in metabolic pathways important for bacterial survival. Biotin is obtained either from the environment or by de novo synthesis, with some bacteria capable of both. In certain species, the bifunctional protein BirA plays a key role in biotin homeostasis as it regulates expression of biotin biosynthetic enzymes in response to biotin demand and supply. Here, we compare the effect of biotin on the growth of two bacteria that possess a bifunctional BirA, namely Escherichia coli and Staphylococcus aureus. Unlike E. coli that could fulfill its biotin requirements through de novo synthesis, S. aureus showed improved growth rates in media supplemented with 10 nM biotin. S. aureus also accumulated more radiolabeled biotin from the media highlighting its ability to efficiently scavenge exogenous material. These data are consistent with S. aureus colonizing low biotin microhabitats. We also demonstrate that the S. aureus BirA protein is a transcriptional repressor of BioY, a subunit of the biotin transporter, and an operon containing yhfT and yhfS, the products of which have a putative role in fatty acid homeostasis. Increased expression of bioY is proposed to help cue S. aureus for efficient scavenging in low biotin environments.


Subject(s)
Biotin/pharmacology , Environmental Microbiology , Gene Expression Regulation, Bacterial/drug effects , Staphylococcus aureus/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biotin/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development
10.
Methods Mol Biol ; 1475: 161-9, 2016.
Article in English | MEDLINE | ID: mdl-27631805

ABSTRACT

Posttranslational regulation of proteins by conjugation of ubiquitin- and ubiquitin-like molecules is a common theme in almost every known biological pathway. SUMO (small ubiquitin-related modifier) is dynamically added and deleted from many cellular substrates to control activity, localization, and recruitment of other SUMO-recognizing protein complexes. The dynamic nature of this modification and its low abundance in resting cells make it challenging to study, with susceptibility to deSUMOylases further complicating its analysis. Here we describe bioSUMO, a general method to isolate and analyze SUMOylated proteins from cultured cells, using Drosophila as a highlighted example. The method also has been validated in transgenic flies, as well as human cells. SUMOylated substrates are labeled by in vivo biotinylation, which facilitates their subsequent purification using streptavidin-based affinity chromatography under stringent conditions and with very low background. The bioSUMO approach can be used to validate whether a specific protein is modified, or used to analyze an entire SUMO subproteome. If coupled to quantitative proteomics methods, it may reveal how the SUMO landscape changes with different stimuli, or in diverse cell or tissue types. This technique offers a complementary approach to study SUMO biology and we expect that the strategy can be extended to other ubiquitin-like proteins.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Escherichia coli Proteins/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Animals , Biotin/chemistry , Biotinylation , Carbon-Nitrogen Ligases/genetics , Cloning, Molecular , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Escherichia coli Proteins/genetics , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Larva/genetics , Larva/metabolism , Macrophages/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Transcription Factors/genetics
11.
Mol Biol Cell ; 27(8): 1188-96, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26912792

ABSTRACT

The BioID method uses a promiscuous biotin ligase to detect protein-protein associations as well as proximate proteins in living cells. Here we report improvements to the BioID method centered on BioID2, a substantially smaller promiscuous biotin ligase. BioID2 enables more-selective targeting of fusion proteins, requires less biotin supplementation, and exhibits enhanced labeling of proximate proteins. Thus BioID2 improves the efficiency of screening for protein-protein associations. We also demonstrate that the biotinylation range of BioID2 can be considerably modulated using flexible linkers, thus enabling application-specific adjustment of the biotin-labeling radius.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Molecular Biology/methods , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Animals , Biotin/metabolism , Biotinylation , Carbon-Nitrogen Ligases/genetics , Escherichia coli Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NIH 3T3 Cells , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Engineering/methods , Protein Interaction Mapping/methods , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics
12.
PLoS One ; 10(11): e0141796, 2015.
Article in English | MEDLINE | ID: mdl-26536126

ABSTRACT

Schizosaccharomyces pombe Δura4 cells lyse when grown on YPD medium. A S. pombe non-essential gene deletion library was screened to determine suppressors of the lysis phenotype. Deletion of the pub1 gene, which encoded E3 ubiquitin ligase, strongly suppressed cell lysis in Δura4 cells. The Δpub1 cells displayed high sensitivity to 5-fluorouracil, a toxic analog of uracil, and this sensitivity was suppressed by deletion of fur4, which encoded a uracil transporter. Fur4 localized primarily to the Golgi apparatus and vacuoles in wild-type cells, but localization was predominantly at the plasma membrane in Δpub1 cells. Fur4 was necessary for the utilization of extracellular uracil, cytosine, or UMP. Uracil uptake activity increased in the Δpub1 strain in a Fur4-dependent manner. In addition, uracil starvation was critical for induction of cell lysis of Δura4 strains and uracil supplementation suppressed lysis. In summary, the increased uracil uptake ability of Δpub1 cells, where Fur4 was predominantly localized to the plasma membrane, resulted in suppression of cell lysis in the Δura4 background.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Carbon-Nitrogen Ligases/genetics , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Down-Regulation , Fluorouracil/analysis , Fluorouracil/metabolism , Gene Deletion , Golgi Apparatus/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Mutation , Schizosaccharomyces pombe Proteins/genetics , Ubiquitination , Uracil/analysis , Vacuoles/metabolism
13.
Mol Genet Metab ; 116(3): 204-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26343941

ABSTRACT

Certain inborn errors of metabolism result from deficiencies in biotin containing enzymes. These disorders are mimicked by dietary absence or insufficiency of biotin, ATP deficit being a major effect,whose responsible mechanisms have not been thoroughly studied. Here we show that in rats and cultured cells it is the result of reduced TCA cycle flow, partly due to deficient anaplerotic biotin-dependent pyruvate carboxylase. This is accompanied by diminished flow through the electron transport chain, augmented by deficient cytochrome c oxidase (complex IV) activity with decreased cytochromes and reduced oxidative phosphorylation. There was also severe mitochondrial damage accompanied by decrease of mitochondria, associated with toxic levels of propionyl CoA as shown by carnitine supplementation studies, which explains the apparently paradoxical mitochondrial diminution in the face of the energy sensor AMPK activation, known to induce mitochondria biogenesis. This idea was supported by experiments on AMPK knockout mouse embryonic fibroblasts (MEFs). The multifactorial ATP deficit also provides a plausible basis for the cardiomyopathy in patients with propionic acidemia, and other diseases.Additionally, systemic inflammation concomitant to the toxic state might explain our findings of enhanced IL-6, STAT3 and HIF-1α, associated with an increase of mitophagic BNIP3 and PINK proteins, which may further increase mitophagy. Together our results imply core mechanisms of energy deficit in several inherited metabolic disorders.


Subject(s)
Biotin/deficiency , Biotin/metabolism , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Mitochondria/metabolism , Mitochondria/ultrastructure , Animals , Carbon-Nitrogen Ligases/metabolism , Carnitine/administration & dosage , Carnitine/metabolism , Cells, Cultured , Citric Acid Cycle , Electron Transport Complex IV/metabolism , Energy Metabolism , Interleukin-6/metabolism , Metabolism, Inborn Errors/genetics , Mice, Knockout , Mitophagy , Oxidative Phosphorylation , Pyruvate Carboxylase/metabolism , Rats
14.
Chem Biol ; 22(7): 917-27, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26097035

ABSTRACT

To combat the emergence of drug-resistant strains of Mycobacterium tuberculosis, new antitubercular agents and novel drug targets are needed. Phenotypic screening of a library of 594 hit compounds uncovered two leads that were active against M. tuberculosis in its replicating, non-replicating, and intracellular states: compounds 7947882 (5-methyl-N-(4-nitrophenyl)thiophene-2-carboxamide) and 7904688 (3-phenyl-N-[(4-piperidin-1-ylphenyl)carbamothioyl]propanamide). Mutants resistant to both compounds harbored mutations in ethA (rv3854c), the gene encoding the monooxygenase EthA, and/or in pyrG (rv1699) coding for the CTP synthetase, PyrG. Biochemical investigations demonstrated that EthA is responsible for the activation of the compounds, and by mass spectrometry we identified the active metabolite of 7947882, which directly inhibits PyrG activity. Metabolomic studies revealed that pharmacological inhibition of PyrG strongly perturbs DNA and RNA biosynthesis, and other metabolic processes requiring nucleotides. Finally, the crystal structure of PyrG was solved, paving the way for rational drug design with this newly validated drug target.


Subject(s)
Antitubercular Agents/pharmacology , Carbon-Nitrogen Ligases/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Oxidoreductases/metabolism , Thiophenes/pharmacology , Activation, Metabolic , Animals , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/metabolism , Drug Design , Drug Evaluation, Preclinical/methods , Hep G2 Cells , High-Throughput Screening Assays/methods , Humans , Mice , Microbial Sensitivity Tests , Models, Molecular , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Oxidoreductases/chemistry , Protein Conformation , Thiophenes/chemistry
15.
PLoS One ; 10(3): e0121224, 2015.
Article in English | MEDLINE | ID: mdl-25822522

ABSTRACT

The African lungfish, Protopterus annectens, can undergo aestivation during drought. Aestivation has three phases: induction, maintenance and arousal. The objective of this study was to examine the differential gene expression in the liver of P. annectens after 6 months (the maintenance phase) of aestivation as compared with the freshwater control, or after 1 day of arousal from 6 months aestivation as compared with 6 months of aestivation using suppression subtractive hybridization. During the maintenance phase of aestivation, the mRNA expression of argininosuccinate synthetase 1 and carbamoyl phosphate synthetase III were up-regulated, indicating an increase in the ornithine-urea cycle capacity to detoxify ammonia to urea. There was also an increase in the expression of betaine homocysteine-S-transferase 1 which could reduce and prevent the accumulation of hepatic homocysteine. On the other hand, the down-regulation of superoxide dismutase 1 expression could signify a decrease in ROS production during the maintenance phase of aestivation. In addition, the maintenance phase was marked by decreases in expressions of genes related to blood coagulation, complement fixation and iron and copper metabolism, which could be strategies used to prevent thrombosis and to conserve energy. Unlike the maintenance phase of aestivation, there were increases in expressions of genes related to nitrogen, carbohydrate and lipid metabolism and fatty acid transport after 1 day of arousal from 6 months aestivation. There were also up-regulation in expressions of genes that were involved in the electron transport system and ATP synthesis, indicating a greater demand for metabolic energy during arousal. Overall, our results signify the importance of sustaining a low rate of waste production and conservation of energy store during the maintenance phase, and the dependence on internal energy store for repair and structural modification during the arousal phase, of aestivation in the liver of P. annectens.


Subject(s)
Arousal/physiology , Energy Metabolism/physiology , Estivation/physiology , Fishes/genetics , Gene Expression Regulation, Enzymologic/physiology , Liver/metabolism , Metabolic Networks and Pathways/physiology , Africa , Animals , Argininosuccinate Synthase/metabolism , Base Sequence , Betaine-Homocysteine S-Methyltransferase/metabolism , Carbon-Nitrogen Ligases/metabolism , DNA, Complementary/genetics , Fishes/physiology , Gene Library , Molecular Sequence Data , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Superoxide Dismutase/metabolism
16.
J Proteomics ; 118: 81-94, 2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25281560

ABSTRACT

Mapping protein-protein interactions for chromatin-associated proteins remains challenging. Here we explore the use of BioID, a proximity biotinylation approach in which a mutated biotin ligase (BirA*) is fused to a bait of interest, allowing for the local activation of biotin and subsequent biotinylation of proteins in the bait vicinity. BioID allowed for successful interactome mapping of core histones and members of the mediator complex. We explored the background signal produced by the BioID approach and found that using distinct types of controls increased the stringency of our statistical analysis with SAINTexpress. A direct comparison of BioID with our AP-MS protocol optimized for chromatin-associated protein complexes revealed that the approaches identified few shared interaction partners and enriched for distinct biological processes; yet, both approaches permitted the recovery of biologically meaningful interactions. While no clear bias could be observed for either technique toward protein complexes of particular functions, BioID allowed for the purification of proteins of lower cellular abundance. Finally, we were able to identify a strong association of MED4 with the centrosome by BioID and validated this finding by immunofluorescence. In summary, BioID complements AP-MS for the study of chromatin-associated protein complexes. BIOLOGICAL SIGNIFICANCE: This manuscript describes the application of BioID, a proximity biotinylation approach, to chromatin-associated proteins, namely core histones and members of the mediator complex. We observed that BioID was successful at identifying known interaction partners for the baits tested, but also allowed novel putative interaction partners to be identified. By performing a detailed comparison of BioID versus a standard method for interactome mapping (affinity purification coupled to mass spectrometry, AP-MS), we show that the approaches were complementary, allowing for purification of different interaction partners. These interaction partners were different in the biological processes they are associated with, but also in their abundance. BioID represents a significant technical development in the field of chromatin research by expanding the search space for interactome mapping beyond what is possible with AP-MS. This article is part of a Special Issue entitled: Protein dynamics in health and disease. Guest Editors: Pierre Thibault and Anne-Claude Gingras.


Subject(s)
Carbon-Nitrogen Ligases , Chromatin , Escherichia coli Proteins , Escherichia coli , Mediator Complex , Repressor Proteins , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Chromatin/genetics , Chromatin/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
Cell Mol Life Sci ; 71(15): 2963-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24477477

ABSTRACT

Rods and rings (RR) are protein assemblies composed of cytidine triphosphate synthetase type 1 (CTPS1) and inosine monophosphate dehydrogenase type 2 (IMPDH2), key enzymes in CTP and GTP biosynthesis. Small-molecule inhibitors of CTPS1 or IMPDH2 induce RR assembly in various cancer cell lines within 15 min to hours. Since glutamine is an essential amide nitrogen donor in these nucleotide biosynthetic pathways, glutamine deprivation was examined to determine whether it leads to RR formation. HeLa cells cultured in normal conditions did not show RR, but after culturing in media lacking glutamine, short rods (<2 µm) assembled after 24 h, and longer rods (>5 µm) formed after 48 h. Upon supplementation with glutamine or guanosine, these RR underwent almost complete disassembly within 15 min. Inhibition of glutamine synthetase with methionine sulfoximine also increased RR assembly in cells deprived of glutamine. Taken together, our data support the hypothesis that CTP/GTP biosynthetic enzymes polymerize to form RR in response to a decreased intracellular level of glutamine. We speculate that rod and ring formation is an adaptive metabolic response linked to disruption of glutamine homeostasis.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Glutamine/metabolism , IMP Dehydrogenase/metabolism , Biosynthetic Pathways , Cytidine Triphosphate/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans
18.
Scand J Immunol ; 78(5): 419-25, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24007195

ABSTRACT

The protein biotin ligase, holocarboxylase synthetase (HLCS), is a chromatin protein that interacts physically with the DNA methyltransferase DNMT1, the methylated cytosine-binding protein MeCP2 and the histone H3 K9-methyltransferase EHMT1, all of which participate in folate-dependent gene repression. Here we tested the hypothesis that biotin and folate synergize in the repression of pro-inflammatory cytokines and long-terminal repeats (LTRs), mediated by interactions between HLCS and other chromatin proteins. Biotin and folate supplementation could compensate for each other's deficiency in the repression of LTRs in Jurkat and U937 cells. For example, when biotin-deficient Jurkat cells were supplemented with folate, the expression of LTRs decreased by >70%. Epigenetic synergies were more complex in the regulation of cytokines compared with LTRs. For example, the abundance of TNF-α was 100% greater in folate- and biotin-supplemented U937 cells compared with biotin-deficient and folate-supplemented cells. The NF-κB inhibitor curcumin abrogated the effects of folate and biotin in cytokine regulation, suggesting that transcription factor signalling adds an extra layer of complexity to the regulation of cytokine genes by epigenetic phenomena. We conclude that biotin and folate synergize in the repression of LTRs and that these interactions are probably mediated by HLCS-dependent epigenetic mechanisms. In contrast, synergies between biotin and folate in the regulation of cytokines need to be interpreted in the context of transcription factor signalling.


Subject(s)
Biotin/metabolism , Carbon-Nitrogen Ligases/metabolism , Cytokines/metabolism , Folic Acid/metabolism , Terminal Repeat Sequences/drug effects , Carbon-Nitrogen Ligases/genetics , Cell Line, Tumor , Curcumin/pharmacology , Cytokines/biosynthesis , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jurkat Cells , Methyl-CpG-Binding Protein 2/metabolism , NF-kappa B/antagonists & inhibitors , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , U937 Cells
19.
Br J Nutr ; 110(2): 321-9, 2013 Jul 28.
Article in English | MEDLINE | ID: mdl-23302490

ABSTRACT

Human biotin requirements are unknown and the identification of reliable markers of biotin status is necessary to fill this knowledge gap. Here, we used an outpatient feeding protocol to create states of biotin deficiency, sufficiency and supplementation in sixteen healthy men and women. A total of twenty possible markers of biotin status were assessed, including the abundance of biotinylated carboxylases in lymphocytes, the expression of genes from biotin metabolism and the urinary excretion of biotin and organic acids. Only the abundance of biotinylated 3-methylcrotonyl-CoA carboxylase (holo-MCC) and propionyl-CoA carboxylase (holo-PCC) allowed for distinguishing biotin-deficient and biotin-sufficient individuals. The urinary excretion of biotin reliably identified biotin-supplemented subjects, but did not distinguish between biotin-depleted and biotin-sufficient individuals. The urinary excretion of 3-hydroxyisovaleric acid detected some biotin-deficient subjects, but produced a meaningful number of false-negative results and did not distinguish between biotin-sufficient and biotin-supplemented individuals. None of the other organic acids that were tested were useful markers of biotin status. Likewise, the abundance of mRNA coding for biotin transporters, holocarboxylase synthetase and biotin-dependent carboxylases in lymphocytes were not different among the treatment groups. Generally, datasets were characterised by variations that exceeded those seen in studies in cell cultures. We conclude that holo-MCC and holo-PCC are the most reliable, single markers of biotin status tested in the present study.


Subject(s)
Biotin/metabolism , Carbon-Carbon Ligases/metabolism , Carbon-Nitrogen Ligases/metabolism , Lymphocytes/metabolism , Methylmalonyl-CoA Decarboxylase/metabolism , Nutritional Status , Vitamin B Deficiency/metabolism , Adult , Biomarkers/metabolism , Dietary Supplements , Female , Humans , Male , Middle Aged , Nutritional Requirements , RNA, Messenger/metabolism , Reference Values , Young Adult
20.
Protein Expr Purif ; 82(1): 162-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22227598

ABSTRACT

The extremely tight binding between biotin and avidin or streptavidin makes labeling proteins with biotin a useful tool for many applications. BirA is the Escherichia coli biotin ligase that site-specifically biotinylates a lysine side chain within a 15-amino acid acceptor peptide (also known as Avi-tag). As a complementary approach to in vivo biotinylation of Avi-tag-bearing proteins, we developed a protocol for producing recombinant BirA ligase for in vitro biotinylation. The target protein was expressed as both thioredoxin and MBP fusions, and was released from the corresponding fusion by TEV protease. The liberated ligase was separated from its carrier using HisTrap HP column. We obtained 24.7 and 27.6 mg BirA ligase per liter of culture from thioredoxin and MBP fusion constructs, respectively. The recombinant enzyme was shown to be highly active in catalyzing in vitro biotinylation. The described protocol provides an effective means for making BirA ligase that can be used for biotinylation of different Avi-tag-bearing substrates.


Subject(s)
Biotin/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Base Sequence , Biotinylation , Carbon-Nitrogen Ligases/isolation & purification , Endopeptidases/metabolism , Escherichia coli Proteins/isolation & purification , Genetic Vectors/genetics , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/isolation & purification , Maltose-Binding Proteins/metabolism , Molecular Sequence Data , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Repressor Proteins/isolation & purification , Thioredoxins/genetics , Thioredoxins/isolation & purification , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL