Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
Add more filters

Publication year range
1.
Arch Microbiol ; 206(3): 126, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38411730

ABSTRACT

Glyoxylate shunt is an important pathway for microorganisms to survive under multiple stresses. One of its enzymes, malate synthase (encoded by aceB gene), has been widely speculated for its contribution to both the pathogenesis and virulence of various microorganisms. We have previously demonstrated that malate synthase (MS) is required for the growth of Salmonella Typhimurium (S. Typhimurium) under carbon starvation and survival under oxidative stress conditions. The aceB gene is encoded by the acetate operon in S. Typhimurium. We attempted to study the activity of acetate promoter under both the starvation and oxidative stress conditions in a heterologous system. The lac promoter of the pUC19 plasmid was substituted with the putative promoter sequence of the acetate operon of S. Typhimurium upstream to the lacZ gene and transformed the vector construct into E. coli NEBα cells. The transformed cells were subjected to the stress conditions mentioned above. We observed a fourfold increase in the ß-galactosidase activity in these cells resulting from the upregulation of the lacZ gene in the stationary phase of cell growth (nutrient deprived) as compared to the mid-log phase. Following exposure of stationary phase cells to hypochlorite-induced oxidative stress, we further observed a 1.6-fold increase in ß galactosidase activity. These data suggest the induction of promoter activity of the acetate operon under carbon starvation and oxidative stress conditions. Thus, these observations corroborate our previous findings regarding the upregulation of aceB expression under stressful environments.


Subject(s)
Escherichia coli , Salmonella typhimurium , Salmonella typhimurium/genetics , Malate Synthase , Operon , Oxidative Stress/genetics , Acetates , Carbon , Nutrients
2.
J Struct Biol ; 215(2): 107958, 2023 06.
Article in English | MEDLINE | ID: mdl-36997036

ABSTRACT

Determination of sub-100 kDa (kDa) structures by cryo-electron microscopy (EM) is a longstanding but not straightforward goal. Here, we present a 2.9-Å cryo-EM structure of a 723-amino acid apo-form malate synthase G (MSG) from Escherichia coli. The cryo-EM structure of the 82-kDa MSG exhibits the same global folding as structures resolved by crystallography and nuclear magnetic resonance (NMR) spectroscopy, and the crystal and cryo-EM structures are indistinguishable. Analyses of MSG dynamics reveal consistent conformational flexibilities among the three experimental approaches, most notably that the α/ß domain exhibits structural heterogeneity. We observed that sidechains of F453, L454, M629, and E630 residues involved in hosting the cofactor acetyl-CoA and substrate rotate differently between the cryo-EM apo-form and complex crystal structures. Our work demonstrates that the cryo-EM technique can be used to determine structures and conformational heterogeneity of sub-100 kDa biomolecules to a quality as high as that obtained from X-ray crystallography and NMR spectroscopy.


Subject(s)
Escherichia coli , Malate Synthase , Cryoelectron Microscopy/methods , Molecular Conformation , Crystallography, X-Ray
3.
Yeast ; 40(7): 265-275, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37170862

ABSTRACT

Debaryomyces hansenii is a halotolerant/halophilic yeast usually found in salty environments. The yeast accumulated sodium at high concentrations, which improved growth in salty media. In contrast, lithium was toxic even at low concentrations and its presence prevented cell proliferation. To analyse the responses to both cations, metabolite levels, enzymatic activities and gene expression were determined, showing that NaCl and LiCl trigger different cellular responses. At high concentrations of NaCl (0.5 or 1.5 M) cells accumulated higher amounts of the intermediate metabolites glyoxylate and malate and, at the same time, the levels of intracellular oxoglutarate decreased. Additionally, 0.5 M NaCl increased the activity of the enzymes isocitrate lyase and malate synthase involved in the synthesis of glyoxylate and malate respectively and decreased the activity of isocitrate dehydrogenase. Moreover, transcription of the genes coding for isocitrate lyase and malate synthase was activated by NaCl. Also, cells accumulated phosphate upon NaCl exposure. None of these effects was provoked when LiCl (0.1 or 0.3 M) was used instead of NaCl. Lithium induced accumulation of higher amounts of oxoglutarate and decreased the concentrations of glyoxylate and malate to non-detectable levels. Cells incubated with lithium also showed higher activity of the isocitrate dehydrogenase and neither increased isocitrate lyase and malate synthase activities nor the transcription of the corresponding genes. In summary, we show that sodium, but not lithium, up regulates the shunt of the glyoxylic acid in D. hansenii and we propose that this is an important metabolic adaptation to thrive in salty environments.


Subject(s)
Debaryomyces , Sodium , Sodium Chloride/pharmacology , Malate Synthase/genetics , Malate Synthase/metabolism , Isocitrate Lyase/genetics , Isocitrate Lyase/metabolism , Malates , Debaryomyces/metabolism , Saccharomyces cerevisiae/metabolism , Isocitrate Dehydrogenase/genetics , Carbon , Ketoglutaric Acids , Glyoxylates/metabolism
4.
Proc Natl Acad Sci U S A ; 117(36): 22452-22461, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32820073

ABSTRACT

Carbon fixation via the Calvin cycle is constrained by the side activity of Rubisco with dioxygen, generating 2-phosphoglycolate. The metabolic recycling of phosphoglycolate was extensively studied in photoautotrophic organisms, including plants, algae, and cyanobacteria, where it is referred to as photorespiration. While receiving little attention so far, aerobic chemolithoautotrophic bacteria that operate the Calvin cycle independent of light must also recycle phosphoglycolate. As the term photorespiration is inappropriate for describing phosphoglycolate recycling in these nonphotosynthetic autotrophs, we suggest the more general term "phosphoglycolate salvage." Here, we study phosphoglycolate salvage in the model chemolithoautotroph Cupriavidus necator H16 (Ralstonia eutropha H16) by characterizing the proxy process of glycolate metabolism, performing comparative transcriptomics of autotrophic growth under low and high CO2 concentrations, and testing autotrophic growth phenotypes of gene deletion strains at ambient CO2 We find that the canonical plant-like C2 cycle does not operate in this bacterium, and instead, the bacterial-like glycerate pathway is the main route for phosphoglycolate salvage. Upon disruption of the glycerate pathway, we find that an oxidative pathway, which we term the malate cycle, supports phosphoglycolate salvage. In this cycle, glyoxylate is condensed with acetyl coenzyme A (acetyl-CoA) to give malate, which undergoes two oxidative decarboxylation steps to regenerate acetyl-CoA. When both pathways are disrupted, autotrophic growth is abolished at ambient CO2 We present bioinformatic data suggesting that the malate cycle may support phosphoglycolate salvage in diverse chemolithoautotrophic bacteria. This study thus demonstrates a so far unknown phosphoglycolate salvage pathway, highlighting important diversity in microbial carbon fixation metabolism.


Subject(s)
Chemoautotrophic Growth/physiology , Glycolates/metabolism , Photosynthesis/physiology , Acetyl Coenzyme A/metabolism , Bacterial Proteins/metabolism , Carbon Cycle/physiology , Cupriavidus necator/genetics , Cupriavidus necator/metabolism , Malate Synthase/metabolism , Malates/metabolism , Oxidation-Reduction
5.
Int J Mol Sci ; 25(1)2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38203573

ABSTRACT

Trichophyton rubrum is the primary causative agent of dermatophytosis worldwide. This fungus colonizes keratinized tissues and uses keratin as a nutritional source during infection. In T. rubrum-host interactions, sensing a hostile environment triggers the adaptation of its metabolic machinery to ensure its survival. The glyoxylate cycle has emerged as an alternative metabolic pathway when glucose availability is limited; this enables the conversion of simple carbon compounds into glucose via gluconeogenesis. In this study, we investigated the impact of stuA deletion on the response of glyoxylate cycle enzymes during fungal growth under varying culture conditions in conjunction with post-transcriptional regulation through alternative splicing of the genes encoding these enzymes. We revealed that the ΔstuA mutant downregulated the malate synthase and isocitrate lyase genes in a keratin-containing medium or when co-cultured with human keratinocytes. Alternative splicing of an isocitrate lyase gene yielded a new isoform. Enzymatic activity assays showed specific instances where isocitrate lyase and malate synthase activities were affected in the mutant strain compared to the wild type strain. Taken together, our results indicate a relevant balance in transcriptional regulation that has distinct effects on the enzymatic activities of malate synthase and isocitrate lyase.


Subject(s)
Arthrodermataceae , Transcription Factors , Humans , Isocitrate Lyase/genetics , Malate Synthase/genetics , Gluconeogenesis/genetics , Alternative Splicing , Carbon , Glucose , Keratins , Glyoxylates
6.
J Appl Microbiol ; 133(5): 2760-2778, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35665578

ABSTRACT

AIMS: Iron (Fe) deficiency in soil is a continuing problem for soybean (Glycine max L.) production, partly as a result of continuing climate change. This study elucidates how Trichoderma harzianum strain T22 (TH) mitigates growth retardation associated with Fe-deficiency in a highly sensitive soybean cultivar. METHODS AND RESULTS: Soil TH supplementation led to mycelial colonization and the presence of UAOX1 gene in roots that caused substantial improvement in chlorophyll score, photosynthetic efficiency and morphological parameters, indicating a positive influence on soybean health. Although rhizosphere acidification was found to be a common feature of Fe-deficient soybean, the upregulation of Fe-reductase activity (GmFRO2) and total phenol secretion were two of the mechanisms that substantially increased the Fe availability by TH. Heat-killed TH applied to soil caused no improvement in photosynthetic attributes and Fe-reductase activity, confirming the active role of TH in mitigating Fe-deficiency. Consistent increases in tissue Fe content and increased Fe-transporter (GmIRT1, GmNRAMP2a, GmNRAMP2b and GmNRAMP7) mRNA levels in roots following TH supplementation were observed only under Fe-deprivation. Root cell death, electrolyte leakage, superoxide (O2 •- ) and hydrogen peroxide (H2 O2 ) substantially declined due to TH in Fe-deprived plants. Further, the elevation of citrate and malate concentration along with the expression of citrate synthase (GmCs) and malate synthase (GmMs) caused by TH suggest improved chelation of Fe in Fe-deficient plants. Results also suggest that TH has a role in triggering antioxidant defence by increasing the activity of glutathione reductase (GR) along with elevated S-metabolites (glutathione and methionine) to stabilize redox status under Fe-deficiency. CONCLUSIONS: TH increases the availability and mobilization of Fe by inducing Fe-uptake pathways, which appears to help provide resistance to oxidative stress associated with Fe-shortage in soybean. SIGNIFICANCE AND IMPACT OF THE STUDY: These findings indicate that while Fe deficiency does not affect the rate or degree of TH hyphal association in soybean roots, the beneficial effects of TH alone may be Fe deficiency-dependent.


Subject(s)
Glycine max , Iron Deficiencies , Glycine max/metabolism , Malates/metabolism , Antioxidants/metabolism , Hydrogen Peroxide/metabolism , Glutathione Reductase/metabolism , Plant Roots/metabolism , Superoxides/metabolism , Citrate (si)-Synthase/metabolism , Malate Synthase/metabolism , Chlorophyll/metabolism , Iron/metabolism , Glutathione/metabolism , Phenols/metabolism , Soil , Citrates , Methionine/metabolism , RNA, Messenger/metabolism
7.
Chemphyschem ; 21(1): 13-19, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31703148

ABSTRACT

Optimized NMR experiments are developed for isolating magnetization belonging to the I=1/2 manifolds of 13 CH3 methyl groups in proteins, enabling the manipulation of the magnetization of a 13 CH3 moiety as if it were an AX (1 H-13 C) spin-system. These experiments result in the same 'simplification' of a 13 CH3 spin-system that would be obtained from the production of {13 CHD2 }-methyl-labeled protein samples. The sensitivity of I=1/2 manifold-selection experiments is a factor of approximately 2 less than that of the corresponding experiments acquired on {13 CHD2 }-labeled methyl groups. The methodology described here is primarily intended for small-to-medium sized proteins, where the losses in sensitivity associated with the isolation of I=1/2 manifold transitions can be tolerated. Several NMR applications that benefit from simplification of the 13 CH3 (AX3 ) spin-systems are described, with an emphasis on the measurements of methyl 1 H-13 C residual dipolar couplings in a {13 CH3 }-methyl-labeled deletion mutant of the human chaperone DNAJB6b, where modulation of NMR signal intensities due to evolution of methyl 1 H-13 C scalar and dipolar couplings follows a simple cosine function characteristic of an AX (1 H-13 C) spin-system, significantly simplifying data analysis.


Subject(s)
Malate Synthase/chemistry , Nuclear Magnetic Resonance, Biomolecular , Ubiquitin/chemistry , Humans , Malate Synthase/metabolism
8.
Microb Cell Fact ; 19(1): 144, 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32677952

ABSTRACT

BACKGROUND: Methylocella silvestris is a facultative aerobic methanotrophic bacterium which uses not only methane, but also other alkanes such as ethane and propane, as carbon and energy sources. Its high metabolic versatility, together with the availability of tools for its genetic engineering, make it a very promising platform for metabolic engineering and industrial biotechnology using natural gas as substrate. RESULTS: The first Genome Scale Metabolic Model for M. silvestris is presented. The model has been used to predict the ability of M. silvestris to grow on 12 different substrates, the growth phenotype of two deletion mutants (ΔICL and ΔMS), and biomass yield on methane and ethanol. The model, together with phenotypic characterization of the deletion mutants, revealed that M. silvestris uses the glyoxylate shuttle for the assimilation of C1 and C2 substrates, which is unique in contrast to published reports of other methanotrophs. Two alternative pathways for propane metabolism have been identified and validated experimentally using enzyme activity tests and constructing a deletion mutant (Δ1641), which enabled the identification of acetol as one of the intermediates of propane assimilation via 2-propanol. The model was also used to integrate proteomic data and to identify key enzymes responsible for the adaptation of M. silvestris to different substrates. CONCLUSIONS: The model has been used to elucidate key metabolic features of M. silvestris, such as its use of the glyoxylate shuttle for the assimilation of one and two carbon compounds and the existence of two parallel metabolic pathways for propane assimilation. This model, together with the fact that tools for its genetic engineering already exist, paves the way for the use of M. silvestris as a platform for metabolic engineering and industrial exploitation of methanotrophs.


Subject(s)
Beijerinckiaceae/growth & development , Beijerinckiaceae/genetics , Isocitrate Lyase/genetics , Malate Synthase/genetics , Models, Biological , Propane/metabolism , Carbon/metabolism , Ethanol/metabolism , Genes, Bacterial , Genetic Engineering , Glyoxylates/metabolism , Industrial Microbiology , Metabolic Networks and Pathways/genetics , Methane/metabolism , Mutation , Proteomics
9.
Proc Natl Acad Sci U S A ; 114(11): E2225-E2232, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28265055

ABSTRACT

The glyoxylate shunt is a metabolic pathway of bacteria, fungi, and plants used to assimilate even-chain fatty acids (FAs) and has been implicated in persistence of Mycobacterium tuberculosis (Mtb). Recent work, however, showed that the first enzyme of the glyoxylate shunt, isocitrate lyase (ICL), may mediate survival of Mtb during the acute and chronic phases of infection in mice through physiologic functions apart from fatty acid metabolism. Here, we report that malate synthase (MS), the second enzyme of the glyoxylate shunt, is essential for in vitro growth and survival of Mtb on even-chain fatty acids, in part, for a previously unrecognized activity: mitigating the toxicity of glyoxylate excess arising from metabolism of even-chain fatty acids. Metabolomic profiling revealed that MS-deficient Mtb cultured on fatty acids accumulated high levels of the ICL aldehyde endproduct, glyoxylate, and increased levels of acetyl phosphate, acetoacetyl coenzyme A (acetoacetyl-CoA), butyryl CoA, acetoacetate, and ß-hydroxybutyrate. These changes were indicative of a glyoxylate-induced state of oxaloacetate deficiency, acetate overload, and ketoacidosis. Reduction of intrabacterial glyoxylate levels using a chemical inhibitor of ICL restored growth of MS-deficient Mtb, despite inhibiting entry of carbon into the glyoxylate shunt. In vivo depletion of MS resulted in sterilization of Mtb in both the acute and chronic phases of mouse infection. This work thus identifies glyoxylate detoxification as an essential physiologic function of Mtb malate synthase and advances its validation as a target for drug development.


Subject(s)
Carbon/metabolism , Glyoxylates/metabolism , Inactivation, Metabolic , Malate Synthase/metabolism , Mycobacterium tuberculosis/metabolism , Animals , Disease Models, Animal , Fatty Acids/metabolism , Female , Gene Knockout Techniques , Macrophages/immunology , Macrophages/metabolism , Malate Synthase/genetics , Metabolic Networks and Pathways , Mice , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/drug therapy , Tuberculosis/microbiology , Tuberculosis/pathology , Virulence/genetics
10.
Int J Mol Sci ; 21(7)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260167

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen responsible for many hospital-acquired infections. P. aeruginosa can thrive in diverse infection scenarios by rewiring its central metabolism. An example of this is the production of biomass from C2 nutrient sources such as acetate via the glyoxylate shunt when glucose is not available. The glyoxylate shunt is comprised of two enzymes, isocitrate lyase (ICL) and malate synthase G (MS), and flux through the shunt is essential for the survival of the organism in mammalian systems. In this study, we characterized the mode of action and cytotoxicity of structural analogs of 2-aminopyridines, which have been identified by earlier work as being inhibitory to both shunt enzymes. Two of these analogs were able to inhibit ICL and MS in vitro and prevented growth of P. aeruginosa on acetate (indicating cell permeability). Moreover, the compounds exerted negligible cytotoxicity against three human cell lines and showed promising in vitro drug metabolism and safety profiles. Isothermal titration calorimetry was used to confirm binding of one of the analogs to ICL and MS, and the mode of enzyme inhibition was determined. Our data suggest that these 2-aminopyridine analogs have potential as anti-pseudomonal agents.


Subject(s)
Aminopyridines/pharmacology , Anti-Bacterial Agents/pharmacology , Isocitrate Lyase/antagonists & inhibitors , Malate Synthase/antagonists & inhibitors , Pseudomonas aeruginosa/growth & development , Aminopyridines/chemistry , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Calorimetry , Cell Line , Gene Expression Regulation, Bacterial/drug effects , Glyoxylates/metabolism , Humans , Isocitrate Lyase/chemistry , Malate Synthase/chemistry , Molecular Structure , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology
11.
J Biol Chem ; 293(34): 13270-13283, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29959230

ABSTRACT

Most protein folding studies until now focus on single domain or truncated proteins. Although great insights in the folding of such systems has been accumulated, very little is known regarding the proteins containing multiple domains. It has been shown that the high stability of domains, in conjunction with inter-domain interactions, manifests as a frustrated energy landscape, causing complexity in the global folding pathway. However, multidomain proteins despite containing independently foldable, loosely cooperative sections can fold into native states with amazing speed and accuracy. To understand the complexity in mechanism, studies were conducted previously on the multidomain protein malate synthase G (MSG), an enzyme of the glyoxylate pathway with four distinct and adjacent domains. It was shown that the protein refolds to a functionally active intermediate state at a fast rate, which slowly produces the native state. Although experiments decoded the nature of the intermediate, a full description of the folding pathway was not elucidated. In this study, we use a battery of biophysical techniques to examine the protein's folding pathway. By using multiprobe kinetics studies and comparison with the equilibrium behavior of protein against urea, we demonstrate that the unfolded polypeptide undergoes conformational compaction to a misfolded intermediate within milliseconds of refolding. The misfolded product appears to be stabilized under moderate denaturant concentrations. Further folding of the protein produces a stable intermediate, which undergoes partial unfolding-assisted large segmental rearrangements to achieve the native state. This study reveals an evolved folding pathway of the multidomain protein MSG, which involves surpassing the multiple misfolding traps during refolding.


Subject(s)
Escherichia coli/enzymology , Malate Synthase/chemistry , Protein Conformation , Protein Folding , Protein Refolding , Crystallography, X-Ray , Kinetics , Malate Synthase/metabolism , Models, Molecular , Protein Denaturation , Thermodynamics
12.
Plasmid ; 101: 20-27, 2019 01.
Article in English | MEDLINE | ID: mdl-30594649

ABSTRACT

The Gram-positive bacterium Corynebacterium glutamicum represents a promising platform for the production of amino acids, organic acids, and other bio-products. However, the availability of only few expression vectors limits its use for production purposes, using metabolic engineering approaches when co-expression of several target genes is desired. To widen the scope for co-expression, the pCG1/p15A and pBL1/colE1 replicons were employed to construct the two differentially-inducible and compatible expression vectors pRG_Duet1 and pRG_Duet2. To functionally validate these newly constructed expression vectors, target genes for easily measurable enzymes were cloned and over-expression of these genes was investigated using respective enzyme assays. Furthermore, functionality and co-existence of the pCG1-based C. glutamicum - E. coli shuttle vector pRG_Duet1 were confirmed with pBL1-based expression vectors pRG_Duet2 and pEKEx2, using co-transformation and enzyme assays. The novel shuttle expression vectors pRG_Duet1 and pRG_Duet2 are attractive additions to the existing set of vectors for co-expression studies and metabolic engineering of C. glutamicum.


Subject(s)
Corynebacterium glutamicum/genetics , Escherichia coli/genetics , Genetic Vectors/chemistry , Metabolic Engineering/methods , Plasmids/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cloning, Molecular , Corynebacterium glutamicum/metabolism , Enzyme Assays , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Malate Synthase/genetics , Malate Synthase/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Plasmids/metabolism , Promoter Regions, Genetic , Replicon , Transformation, Bacterial
13.
Curr Microbiol ; 76(11): 1298-1305, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31428805

ABSTRACT

Geobacillus thermoglucosidasius NY05 catalyzes calcite single crystal formation at 60 °C by using acetate and calcium. Endospores are embedded at the central part of the calcite single crystal and carbon atoms in the calcite lattice are derived from acetate carbon. Here, we synthesized 21-mer antisense DNA oligonucleotides targeting sporulation transcription factor, acetate-CoA ligase, isocitrate lyase, and malate synthase G mRNAs and evaluated the effect of these oligonucleotides on calcite formation in G. thermoglucosidasius NY05. G. thermoglucosidasius NY05 cells containing antisense DNA oligonucleotides targeting sporulation transcription factor, acetate-CoA ligase, isocitrate lyase, and malate synthase G mRNAs had reduced calcite single crystal formation by 18.7, 50.6, 55.7, and 82.3%, respectively, compared with cells without antisense DNA oligonucleotides. These results support that calcite formation needs endospores as the nucleus to grow, and carbon dioxide generated from acetate, which is metabolized via the glyoxylate pathway and glucogenesis, is supplied to the crystal lattice.


Subject(s)
Bacterial Proteins/genetics , Calcium Carbonate/metabolism , Gene Silencing , Geobacillus/genetics , Acetates/metabolism , Bacterial Proteins/metabolism , Calcium/metabolism , Calcium Carbonate/chemistry , Geobacillus/chemistry , Geobacillus/metabolism , Glyoxylates/metabolism , Isocitrate Lyase/genetics , Isocitrate Lyase/metabolism , Malate Synthase/genetics , Malate Synthase/metabolism
14.
J Cell Sci ; 129(21): 4057-4066, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27678487

ABSTRACT

Peroxisomal proteins carrying a type 1 peroxisomal targeting signal (PTS1) are recognized by the well-conserved cycling import receptor Pex5p. The yeast YMR018W gene encodes a Pex5p paralog and newly identified peroxin that is involved in peroxisomal import of a subset of matrix proteins. The new peroxin was designated Pex9p, and it interacts with the docking protein Pex14p and a subclass of PTS1-containing peroxisomal matrix enzymes. Unlike Pex5p, Pex9p is not expressed in glucose- or ethanol-grown cells, but it is strongly induced by oleate. Under these conditions, Pex9p acts as a cytosolic and membrane-bound peroxisome import receptor for both malate synthase isoenzymes, Mls1p and Mls2p. The inducible Pex9p-dependent import pathway provides a mechanism for the oleate-inducible peroxisomal targeting of malate synthases. The existence of two distinct PTS1 receptors, in addition to two PTS2-dependent import routes, contributes to the adaptive metabolic capacity of peroxisomes in response to environmental changes and underlines the role of peroxisomes as multi-purpose organelles. The identification of different import routes into peroxisomes contributes to the molecular understanding of how regulated protein targeting can alter the function of organelles according to cellular needs.


Subject(s)
Peroxisomes/metabolism , Protein Sorting Signals , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Malate Synthase/metabolism , Models, Biological , Oleic Acid/pharmacology , Peroxisomes/drug effects , Protein Binding/drug effects , Protein Sorting Signals/drug effects , Protein Transport/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Structural Homology, Protein , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
15.
Microbiology (Reading) ; 164(4): 587-599, 2018 04.
Article in English | MEDLINE | ID: mdl-29465342

ABSTRACT

The aceA and glcB genes, encoding isocitrate lyase (ICL) and malate synthase, respectively, are not in an operon in many bacteria, including Pseudomonas aeruginosa, unlike in Escherichia coli. Here, we show that expression of aceA in P. aeruginosa is specifically upregulated under H2O2-induced oxidative stress and under iron-limiting conditions. In contrast, the addition of exogenous redox active compounds or antibiotics increases the expression of glcB. The transcriptional start sites of aceA under iron-limiting conditions and in the presence of iron were found to be identical by 5' RACE. Interestingly, the enzymatic activities of ICL and isocitrate dehydrogenase had opposite responses under different iron conditions, suggesting that the glyoxylate shunt (GS) might be important under iron-limiting conditions. Remarkably, the intracellular iron concentration was lower while the iron demand was higher in the GS-activated cells growing on acetate compared to cells growing on glucose. Absence of GS dysregulated iron homeostasis led to changes in the cellular iron pool, with higher intracellular chelatable iron levels. In addition, GS mutants were found to have higher cytochrome c oxidase activity on iron-supplemented agar plates of minimal media, which promoted the growth of the GS mutants. However, deletion of the GS genes resulted in higher sensitivity to a high concentration of H2O2, presumably due to iron-mediated killing. In conclusion, the GS system appears to be tightly linked to iron homeostasis in the promotion of P. aeruginosa survival under oxidative stress.


Subject(s)
Glyoxylates/metabolism , Homeostasis , Iron/metabolism , Isocitrate Lyase/metabolism , Malate Synthase/metabolism , Oxidative Stress , Pseudomonas aeruginosa/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Citric Acid Cycle , Cytoplasm/chemistry , Electron Transport , Gene Expression Regulation, Bacterial/drug effects , Homeostasis/drug effects , Hydrogen Peroxide/pharmacology , Iron/chemistry , Isocitrate Dehydrogenase/metabolism , Isocitrate Lyase/genetics , Malate Synthase/genetics , Mutation , Oxidative Stress/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism
16.
Biochem Biophys Res Commun ; 495(1): 560-566, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29129690

ABSTRACT

The metabolic pathway such as glyoxylate cycle (GC) enables Candida albicans, to survive under glucose deficient conditions prevalent in the hostile niche. Thus its key enzymes (Isocitrate lyase; ICL and malate synthase; MLS) represent attractive targets against C. albicans. We have previously reported the antifungal potential of a natural monoterpenoid perillyl alcohol (PA). The present study uncovers additional role of PA as a potent GC inhibitor. We explored that PA phenocopied ICL1 deletion mutant and were hypersensitive under low carbon utilizing conditions. The effect of PA on GC was substantiated by molecular docking analyses, which reveals the in-silico binding affinity of PA with ICL and MLS and explored that PA binds to the active sites of both proteins with better binding energy in comparison to their known inhibitors 3-nitropropionate and bromopyruvate respectively. Enzyme kinetics by Lineweaver-Burk plot unravels that PA inhibits ICL and MLS enzymes in competitive and non-competitive manner respectively. Moreover, semi-quantitative RT-PCR indicated that PA inhibits ICL1 and MLS1 mRNA expressions. Lastly, we demonstrated the antifungal efficacy of PA by enhanced survival of Caenorhabditis elegans model and less hemolytic activity (10.6%) on human blood cells. Further studies are warranted for PA to be considered as viable drug candidate.


Subject(s)
Candida albicans/drug effects , Candida albicans/metabolism , Glyoxylates/metabolism , Isocitrate Lyase/metabolism , Malate Synthase/metabolism , Metabolic Networks and Pathways/physiology , Monoterpenes/administration & dosage , Anti-Bacterial Agents/administration & dosage , Bacterial Proteins/metabolism , Candida albicans/cytology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/physiology , Metabolic Flux Analysis , Metabolic Networks and Pathways/drug effects
17.
J Chem Inf Model ; 58(10): 2085-2091, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30137983

ABSTRACT

Human infection by Mycobacterium tuberculosis (Mtb) continues to be a global epidemic. Computer-aided drug design (CADD) methods are used to accelerate traditional drug discovery efforts. One noncovalent interaction that is being increasingly identified in biological systems but is neglected in CADD is the anion-π interaction. The study reported herein supports the conclusion that anion-π interactions play a central role in directing the binding of phenyl-diketo acid (PDKA) inhibitors to malate synthase (GlcB), an enzyme required for Mycobacterium tuberculosis virulence. Using density functional theory methods (M06-2X/6-31+G(d)), a GlcB active site template was developed for a predictive model through a comparative analysis of PDKA-bound GlcB crystal structures. The active site model includes the PDKA molecule and the protein determinants of the electrostatic, hydrogen-bonding, and anion-π interactions involved in binding. The predictive model accurately determines the Asp 633-PDKA structural position upon binding and precisely predicts the relative binding enthalpies of a series of 2-ortho halide-PDKAs to GlcB. A screening model was also developed to efficiently assess the propensity of each PDKA analog to participate in an anion-π interaction; this method is in good agreement with both the predictive model and the experimental binding enthalpies for the 2-ortho halide-PDKAs. With the screening and predictive models in hand, we have developed an efficient method for computationally screening and evaluating the binding enthalpy of variously substituted PDKA molecules. This study serves to illustrate the contribution of this overlooked interaction to binding affinity and demonstrates the importance of integrating anion-π interactions into structure-based CADD.


Subject(s)
Antitubercular Agents/pharmacology , Malate Synthase/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Antitubercular Agents/chemistry , Binding Sites , Computer Simulation , Malate Synthase/metabolism , Models, Molecular , Protein Binding , Protein Conformation
18.
J Bacteriol ; 199(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27920298

ABSTRACT

Haloarchaea are extremely halophilic heterotrophic microorganisms belonging to the class Halobacteria (Euryarchaeota). Almost half of the haloarchaea possesses the genes coding for enzymes of the methylaspartate cycle, a recently discovered anaplerotic acetate assimilation pathway. In this cycle, the enzymes of the tricarboxylic acid cycle together with the dedicated enzymes of the methylaspartate cycle convert two acetyl coenzyme A (acetyl-CoA) molecules to malate. The methylaspartate cycle involves two reactions catalyzed by homologous enzymes belonging to the CitE-like enzyme superfamily, malyl-CoA lyase/thioesterase (haloarchaeal malate synthase [hMS]; Hah_2476 in Haloarcula hispanica) and ß-methylmalyl-CoA lyase (haloarchaeal ß-methylmalyl-CoA lyase [hMCL]; Hah_1341). Although both enzymes catalyze the same reactions, hMS was previously proposed to preferentially catalyze the formation of malate from acetyl-CoA and glyoxylate (malate synthase activity) and hMCL was proposed to primarily cleave ß-methylmalyl-CoA to propionyl-CoA and glyoxylate. Here we studied the physiological functions of these enzymes during acetate assimilation in H. hispanica by using biochemical assays of the wild type and deletion mutants. Our results reveal that the main physiological function of hMS is malyl-CoA (not malate) formation and that hMCL catalyzes a ß-methylmalyl-CoA lyase reaction in vivo The malyl-CoA thioesterase activities of both enzymes appear to be not essential for growth on acetate. Interestingly, despite the different physiological functions of hMS and hMCL, structural comparisons predict that these two proteins have virtually identical active sites, thus highlighting the need for experimental validation of their catalytic functions. Our results provide further proof of the operation of the methylaspartate cycle and indicate the existence of a distinct, yet-to-be-discovered malyl-CoA thioesterase in haloarchaea. IMPORTANCE: Acetate is one of the most important substances in natural environments. The activated form of acetate, acetyl coenzyme A (acetyl-CoA), is the high-energy intermediate at the crossroads of central metabolism: its oxidation generates energy for the cell, and about a third of all biosynthetic fluxes start directly from acetyl-CoA. Many organic compounds enter the central carbon metabolism via this key molecule. To sustain growth on acetyl-CoA-generating compounds, a dedicated assimilation (anaplerotic) pathway is required. The presence of an anaplerotic pathway is a prerequisite for growth in many environments, being important for environmentally, industrially, and clinically important microorganisms. Here we studied specific reactions of a recently discovered acetate assimilation pathway, the methylaspartate cycle, functioning in extremely halophilic archaea.


Subject(s)
Aspartic Acid/analogs & derivatives , Gene Expression Regulation, Archaeal/physiology , Gene Expression Regulation, Enzymologic/physiology , Haloarcula/enzymology , Malate Synthase/metabolism , Oxo-Acid-Lyases/metabolism , Aspartic Acid/metabolism , Cell Extracts , Haloarcula/genetics , Haloarcula/metabolism , Malate Synthase/genetics , Mutation , Oxo-Acid-Lyases/genetics , Phylogeny
19.
Biochemistry ; 56(41): 5539-5549, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28985053

ABSTRACT

Pseudomonas aeruginosa is an opportunistic human pathogen recognized as a critical threat by the World Health Organization because of the dwindling number of effective therapies available to treat infections. Over the past decade, it has become apparent that the glyoxylate shunt plays a vital role in sustaining P. aeruginosa during infection scenarios. The glyoxylate shunt comprises two enzymes: isocitrate lyase and malate synthase isoform G. Inactivation of these enzymes has been reported to abolish the ability of P. aeruginosa to establish infection in a mammalian model system, yet we still lack the structural information to support drug design efforts. In this work, we describe the first X-ray crystal structure of P. aeruginosa malate synthase G in the apo form at 1.62 Å resolution. The enzyme is a monomer composed of four domains and is highly conserved with homologues found in other clinically relevant microorganisms. It is also dependent on Mg2+ for catalysis. Metal ion binding led to a change in the intrinsic fluorescence of the protein, allowing us to quantitate its affinity for Mg2+. We also identified putative drug binding sites in malate synthase G using computational analysis and, because of the high resolution of the experimental data, were further able to characterize its hydration properties. Our data reveal two promising binding pockets in malate synthase G that may be exploited for drug design.


Subject(s)
Bacterial Proteins/metabolism , Malate Synthase/metabolism , Models, Molecular , Pseudomonas aeruginosa/enzymology , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Amino Acid Sequence , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalytic Domain , Computational Biology , Conserved Sequence , Crystallography, X-Ray , Expert Systems , Glyoxylates/chemistry , Glyoxylates/metabolism , Indoles/chemistry , Indoles/metabolism , Ligands , Magnesium/chemistry , Magnesium/metabolism , Malate Synthase/chemistry , Malate Synthase/genetics , Molecular Docking Simulation , Molecular Structure , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Structural Homology, Protein
20.
J Biol Chem ; 291(53): 27421-27432, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27738104

ABSTRACT

Fragment screening and high throughput screening are complementary approaches that combine with structural biology to explore the binding capabilities of an active site. We have used a fragment-based approach on malate synthase (GlcB) from Mycobacterium tuberculosis and discovered several novel binding chemotypes. In addition, the crystal structures of GlcB in complex with these fragments indicated conformational changes in the active site that represent the enzyme conformations during catalysis. Additional structures of the complex with malate and of the apo form of GlcB supported that hypothesis. Comparative analysis of GlcB structures in complex with 18 fragments allowed us to characterize the preferred chemotypes and their binding modes. The fragment structures showed a hydrogen bond to the backbone carbonyl of Met-631. We successfully incorporated an indole group from a fragment into an existing phenyl-diketo acid series. The resulting indole-containing inhibitor was 100-fold more potent than the parent phenyl-diketo acid with an IC50 value of 20 nm.


Subject(s)
Malate Synthase/chemistry , Malate Synthase/metabolism , Malates/metabolism , Mycobacterium tuberculosis/enzymology , Binding Sites , Catalysis , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL