Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34083440

ABSTRACT

Class-II fumarases (fumarate hydratase, FH) are dual-targeted enzymes occurring in the mitochondria and cytosol of all eukaryotes. They are essential components in the DNA damage response (DDR) and, more specifically, protect cells from DNA double-strand breaks. Similarly, the gram-positive bacterium Bacillus subtilis class-II fumarase, in addition to its role in the tricarboxylic acid cycle, participates in the DDR. Escherichia coli harbors three fumarase genes: class-I fumA and fumB and class-II fumC Notably, class-I fumarases show no sequence similarity to class-II fumarases and are of different evolutionary origin. Strikingly, here we show that E. coli fumarase functions are distributed between class-I fumarases, which participate in the DDR, and the class-II fumarase, which participates in respiration. In E. coli, we discover that the signaling molecule, alpha-ketoglutarate (α-KG), has a function, complementing DNA damage sensitivity of fum-null mutants. Excitingly, we identify the E. coli α-KG-dependent DNA repair enzyme AlkB as the target of this interplay of metabolite signaling. In addition to α-KG, fumarate (fumaric acid) is shown to affect DNA damage repair on two different levels, first by directly inhibiting the DNA damage repair enzyme AlkB demethylase activity, both in vitro and in vivo (countering α-KG). The second is a more global effect on transcription, because fum-null mutants exhibit a decrease in transcription of key DNA damage repair genes. Together, these results show evolutionary adaptable metabolic signaling of the DDR, in which fumarases and different metabolites are recruited regardless of the evolutionary enzyme class performing the function.


Subject(s)
DNA Damage , Escherichia coli/genetics , Fumarate Hydratase/metabolism , Fumarates/metabolism , Ketoglutaric Acids/metabolism , AlkB Enzymes , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Citric Acid Cycle , DNA Breaks, Double-Stranded , DNA, Bacterial/genetics , Fumarate Hydratase/chemistry , Genes, Bacterial
2.
Biochemistry ; 62(2): 476-493, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36595439

ABSTRACT

Fumarate hydratase (FH) is a remarkable catalyst that decreases the free energy of the catalyzed reaction by 30 kcal mol-1, much larger than most exceptional enzymes with extraordinary catalytic rates. Two classes of FH are observed in nature: class-I and class-II, which have different folds, yet catalyze the same reversible hydration/dehydration reaction of the dicarboxylic acids fumarate/malate, with equal efficiencies. Using class-I FH from the hyperthermophilic archaeon Methanocaldococcus jannaschii (Mj) as a model along with comparative analysis with the only other available class-I FH structure from Leishmania major (Lm), we provide insights into the molecular mechanism of catalysis in this class of enzymes. The structure of MjFH apo-protein has been determined, revealing that large intersubunit rearrangements occur across apo- and holo-protein forms, with a largely preorganized active site for substrate binding. Site-directed mutagenesis of active site residues, kinetic analysis, and computational studies, including density functional theory (DFT) and natural population analysis, together show that residues interacting with the carboxylate group of the substrate play a pivotal role in catalysis. Our study establishes that an electrostatic network at the active site of class-I FH polarizes the substrate fumarate through interactions with its carboxylate groups, thereby permitting an easier addition of a water molecule across the olefinic bond. We propose a mechanism of catalysis in FH that occurs through transition-state stabilization involving the distortion of the electronic structure of the substrate olefinic bond mediated by the charge polarization of the bound substrate at the enzyme active site.


Subject(s)
Fumarate Hydratase , Fumarates , Fumarate Hydratase/chemistry , Kinetics , Catalytic Domain , Catalysis
3.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163394

ABSTRACT

The FH gene encodes the fumarate hydratase of the Krebs cycle and functions as a homotetramer to catalyze the hydration of fumarate to malate. Mutations in FH result in uterine leiomyomas, a rare autosomal dominant inherited metabolic disease. However, how FH mutations result in this disease is poorly understood. Here, the FH mutation c.557G>A (p.S186N) was identified in a family with uterine leiomyomas phenotype. A series of studies were performed to confirm the pathogenicity of this mutation. Results showed that the FH mutant exhibited significantly lower fumarase enzyme activity and increased the fumarates level compared with the wildtype, which might be due to the impaired homotetramer formation in the native gel electrophoresis. Interestingly, the immunofluorescence study revealed that the overexpressed FH mutant exhibited puncta structures compared with the evenly expressed FH wildtype in cytoplasm suggesting that the altered amino acid might result in dysfunctional proteins which were accumulated to reduce its cytotoxicity. Importantly, the cells overexpressing the FH mutant exhibited higher proliferation and extracellular acidification rate value (ECAR) which might be caused by the upregulated HIF-1α indicating the tumor phenotype. Notably, phospho-mTOR was significantly increased and autophagy was inhibited in the FH mutant overexpression cells compared with the wildtype. Our work provides new insight into the FH mutation c.557G>A (p.S186N) underlies uterine leiomyomas and important information for accurate genetic counseling and clinical diagnosis of the disease.


Subject(s)
Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation/genetics , Uterine Neoplasms/genetics , Adult , Autophagy , Base Sequence , Female , Fumarate Hydratase/chemistry , Fumarates/metabolism , HEK293 Cells , Humans , Male , Pedigree , Protein Multimerization , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
4.
Biochem Biophys Res Commun ; 557: 329-333, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33895474

ABSTRACT

Glutathione (GSH) is the most abundant non-protein thiol and its cellular concentration has been reported as 17 mM in Escherichia coli. This study introduces a label-free method to determine the binding affinity of GSH to proteins, utilizing the intrinsic fluorescence of proteins; the dissociation constants of GSH for d-arabinose 5-phosphate isomerase KdsD, fumarase C, malate dehydrogenase, and RNA polymerase subunit α have been determined as 96 ± 8, 246 ± 42, 292 ± 78, and 296 ± 97 µM, respectively. The dissociation constants, less than 2% of the cellular concentration of GSH, suggests that protein-GSH interactions are strong enough to make all of the GSH-binding sites occupied fully. The method described here may be applicable to other proteins.


Subject(s)
Aldose-Ketose Isomerases/chemistry , DNA-Directed RNA Polymerases/chemistry , Fumarate Hydratase/chemistry , Glutathione/chemistry , Malate Dehydrogenase/chemistry , Spectrometry, Fluorescence/methods , Escherichia coli/metabolism , Fluorescence , Gene Expression , Glutathione/metabolism , Kinetics , Ligands , Oxidative Stress , Recombinant Proteins
5.
Biochemistry ; 58(49): 5011-5021, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31743022

ABSTRACT

Class I fumarate hydratases (FHs) are central metabolic enzymes that use a [4Fe-4S] cluster to catalyze the reversible conversion of fumarate to S-malate. The parasite Leishmania major, which is responsible for leishmaniasis, expresses two class I FH isoforms: mitochondrial LmFH-1 and cytosolic LmFH-2. In this study, we present kinetic characterizations of both LmFH isoforms, present 13 crystal structures of LmFH-2 variants, and employ site-directed mutagenesis to investigate the enzyme's mechanism. Our kinetic data confirm that both LmFH-1 and LmFH-2 are susceptible to oxygen-dependent inhibition, with data from crystallography and electron paramagnetic resonance spectroscopy showing that oxygen exposure converts an active [4Fe-4S] cluster to an inactive [3Fe-4S] cluster. Our anaerobically conducted kinetic studies reveal a preference for fumarate over S-malate. Our data further reveal that single alanine substitutions of T467, R421, R471, D135, and H334 decrease kcat values 9-16000-fold without substantially affecting Km values, suggesting that these residues function in catalytic roles. Crystal structures of LmFH-2 variants are consistent with this idea, showing similar bidentate binding to the unique iron of the [4Fe-4S] cluster for substrate S-malate as observed in wild type FH. We further present LmFH-2 structures with substrate fumarate and weak inhibitors succinate and malonate bound in the active site and the first structure of an LmFH that is substrate-free and inhibitor-free, the latter showing increased mobility in the C-terminal domain. Collectively, these data provide insight into the molecular basis for the reaction catalyzed by LmFHs, enzymes that are potential drug targets against leishmaniasis.


Subject(s)
Fumarate Hydratase/chemistry , Fumarate Hydratase/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Leishmania major/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Amino Acid Substitution , Catalytic Domain , Fumarate Hydratase/genetics , Iron-Sulfur Proteins/genetics , Kinetics , Leishmania major/chemistry , Leishmania major/genetics , Multigene Family , Oxygen/chemistry , Oxygen/metabolism , Protozoan Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 113(35): 9804-9, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27528683

ABSTRACT

Fumarate hydratases (FHs) are essential metabolic enzymes grouped into two classes. Here, we present the crystal structure of a class I FH, the cytosolic FH from Leishmania major, which reveals a previously undiscovered protein fold that coordinates a catalytically essential [4Fe-4S] cluster. Our 2.05 Å resolution data further reveal a dimeric architecture for this FH that resembles a heart, with each lobe comprised of two domains that are arranged around the active site. Besides the active site, where the substrate S-malate is bound bidentate to the unique iron of the [4Fe-4S] cluster, other binding pockets are found near the dimeric enzyme interface, some of which are occupied by malonate, shown here to be a weak inhibitor of this enzyme. Taken together, these data provide a framework both for investigations of the class I FH catalytic mechanism and for drug design aimed at fighting neglected tropical diseases.


Subject(s)
Fumarate Hydratase/chemistry , Iron-Sulfur Proteins/chemistry , Leishmania major/chemistry , Malates/chemistry , Protozoan Proteins/chemistry , Amino Acid Motifs , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Leishmania major/enzymology , Malates/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
7.
Crit Rev Biotechnol ; 36(1): 99-107, 2016.
Article in English | MEDLINE | ID: mdl-25025277

ABSTRACT

l-Malic acid has many uses in food, beverage, pharmaceutical, chemical and medical industries. It can be produced by one-step fermentation, enzymatic transformation of fumaric acid to l-malate and acid hydrolysis of polymalic acid. However, the process for one-step fermentation is preferred as it has many advantages over any other process. The pathways of l-malic acid biosynthesis in microorganisms are partially clear and three metabolic pathways including non-oxidative pathway, oxidative pathway and glyoxylate cycle for the production of l-malic acid from glucose have been identified. Usually, high levels of l-malate are produced under the nitrogen starvation conditions, l-malate, as a calcium salt, is secreted from microbial cells and CaCO3 can play an important role in calcium malate biosynthesis and regulation. However, it is still unclear how it is secreted into the medium. To enhance l-malate biosynthesis and secretion by microbial cells, it is very important to study the mechanisms of l-malic acid biosynthesis and secretion at enzymatic and molecular levels.


Subject(s)
Bacteria/metabolism , Fermentation , Fumarate Hydratase/metabolism , Malates/metabolism , Bacteria/cytology , Food Industry , Fumarate Hydratase/chemistry , Glucose/metabolism , Glyoxylates/metabolism , Hydrolysis , Malates/chemistry
8.
Appl Environ Microbiol ; 81(16): 5632-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26070669

ABSTRACT

Pseudomonas aeruginosa, Yersinia pestis, and many other bacteria are able to utilize the C5-dicarboxylic acid itaconate (methylenesuccinate). Itaconate degradation starts with its activation to itaconyl coenzyme A (itaconyl-CoA), which is further hydrated to (S)-citramalyl-CoA, and citramalyl-CoA is finally cleaved into acetyl-CoA and pyruvate. The xenobiotic-degrading betaproteobacterium Burkholderia xenovorans possesses a P. aeruginosa-like itaconate degradation gene cluster and is able to grow on itaconate and its isomer mesaconate (methylfumarate). Although itaconate degradation proceeds in B. xenovorans in the same way as in P. aeruginosa, the pathway of mesaconate utilization is not known. Here, we show that mesaconate is metabolized through its hydration to (S)-citramalate. The latter compound is then metabolized to acetyl-CoA and pyruvate with the participation of two enzymes of the itaconate degradation pathway, a promiscuous itaconate-CoA transferase able to activate (S)-citramalate in addition to itaconate and (S)-citramalyl-CoA lyase. The first reaction of the pathway, the mesaconate hydratase (mesaconase) reaction, is catalyzed by a class I fumarase. As this enzyme (Bxe_A3136) has similar efficiencies (kcat/Km) for both fumarate and mesaconate hydration, we conclude that B. xenovorans class I fumarase is in fact a promiscuous fumarase/mesaconase. This promiscuity is physiologically relevant, as it allows the growth of this bacterium on mesaconate as a sole carbon and energy source.


Subject(s)
Burkholderia/enzymology , Burkholderia/metabolism , Fumarate Hydratase/metabolism , Fumarates/metabolism , Hydro-Lyases/metabolism , Maleates/metabolism , Acetyl Coenzyme A/metabolism , Burkholderia/genetics , Fumarate Hydratase/chemistry , Hydro-Lyases/chemistry , Kinetics , Malates/metabolism , Metabolic Networks and Pathways/genetics , Pyruvic Acid/metabolism , Substrate Specificity , Succinates/metabolism
9.
Mol Biol Rep ; 41(1): 497-504, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24307253

ABSTRACT

Fumarase is a key enzyme that catalyzes the reversible hydration of fumarate to L-malate in the tricarboxylic acid cycle. This reaction has been extensively utilized for industrial applications in producing L-malate. In this study, a fumarase C gene from Streptomyces lividans TK54 (slFumC) was cloned and expressed as a fused protein (SlFumC) in Escherichia coli. The molecular mass of SlFumC was about 49 kDa determined by SDS-PAGE. Kinetic studies showed that the K m value of SlFumC for L-malate increased by approximately 8.5-fold at pH 6.5 (6.7 ± 0.81 mM) to 8.0 (57.0 ± 1.12 mM), which was higher than some known fumarases. The catalytic efficiency (k cat) and the specific activity increased by about 9.5-fold at pH 6.5 (65 s(-1)) to 8.0 (620 s(-1)) and from 79 U/mg at pH 6.5 to 752 U/mg at pH 8.0, respectively. Therefore, SlFumC may acquire strong catalytic ability by increasing pH to partially compensate for the loss of substrate affinity. The enzyme also showed substrate inhibition phenomenon, which is pH-dependent. Specific activity of SlFumC was gradually enhanced with increasing phosphate concentrations. However, no inhibition was observed at high concentration of phosphate ion, which was distinctly different in case of other Class II fumarases. In industrial process, the reaction temperatures for L-malate production are usually set between 40 and 60 °C. The recombinant SlFumC displayed maximal activity at 45 °C and remained over 85 % of original activity after 48 h incubation at 40 °C, which was more thermostable than other fumarases from Streptomyces and make it an efficient enzyme for use in the industrial production of L-malate.


Subject(s)
Malates/chemical synthesis , Streptomyces lividans/enzymology , Amino Acid Sequence , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Enzyme Stability , Escherichia coli , Fumarate Hydratase/biosynthesis , Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Homology, Amino Acid
10.
Protein Sci ; 32(10): e4779, 2023 10.
Article in English | MEDLINE | ID: mdl-37695939

ABSTRACT

Malate (2-hydroxysuccinic acid) and tartrate (2,3-dihydroxysuccinic acid) are chiral substrates; the former existing in two enantiomeric forms (R and S) while the latter exists as three stereoisomers (R,R; S,S; and R,S). Dehydration by stereospecific hydrogen abstraction and antielimination of the hydroxyl group yield the achiral products fumarate and oxaloacetate, respectively. Class-I fumarate hydratase (FH) and L-tartrate dehydratase (L-TTD) are two highly conserved enzymes belonging to the iron-sulfur cluster hydrolyase family of enzymes that catalyze reactions on specific stereoisomers of malate and tartrate. FH from Methanocaldococcus jannaschii accepts only (S)-malate and (S,S)-tartrate as substrates while the structurally similar L-TTD from Escherichia coli accepts only (R)-malate and (R,R)-tartrate as substrates. Phylogenetic analysis reveals a common evolutionary origin of L-TTDs and two-subunit archaeal FHs suggesting a divergence during evolution that may have led to the switch in substrate stereospecificity preference. Due to the high conservation of their sequences, a molecular basis for switch in stereospecificity is not evident from analysis of crystal structures of FH and predicted structure of L-TTD. The switch in enantiomer preference may be rationalized by invoking conformational plasticity of the amino acids interacting with the substrate, together with substrate reorientation and conformer selection about the C2C3 bond of the dicarboxylic acid substrates. Although classical models of enzyme-substrate binding are insufficient to explain such a phenomenon, the enantiomer superposition model suggests that a minor reorientation in the active site residues could lead to the switch in substrate stereospecificity.


Subject(s)
Malates , Tartrates , Humans , Tartrates/metabolism , Malates/metabolism , Phylogeny , Dehydration , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Escherichia coli/metabolism , Catalytic Domain , Substrate Specificity , Kinetics
11.
Biochemistry ; 51(21): 4237-43, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22551392

ABSTRACT

Members of the aspartase/fumarase superfamily share a common tertiary and quaternary fold, as well as a similar active site architecture; the superfamily includes aspartase, fumarase, argininosuccinate lyase, adenylosuccinate lyase, δ-crystallin, and 3-carboxy-cis,cis-muconate lactonizing enzyme (CMLE). These enzymes all process succinyl-containing substrates, leading to the formation of fumarate as the common product (except for the CMLE-catalyzed reaction, which results in the formation of a lactone). In the past few years, X-ray crystallographic analysis of several superfamily members in complex with substrate, product, or substrate analogues has provided detailed insights into their substrate binding modes and catalytic mechanisms. This structural work, combined with earlier mechanistic studies, revealed that members of the aspartase/fumarase superfamily use a common catalytic strategy, which involves general base-catalyzed formation of a stabilized aci-carboxylate (or enediolate) intermediate and the participation of a highly flexible loop, containing the signature sequence GSSxxPxKxN (named the SS loop), in substrate binding and catalysis.


Subject(s)
Aspartate Ammonia-Lyase/chemistry , Aspartate Ammonia-Lyase/metabolism , Fumarate Hydratase/chemistry , Fumarate Hydratase/metabolism , Adenylosuccinate Lyase/chemistry , Adenylosuccinate Lyase/genetics , Adenylosuccinate Lyase/metabolism , Amino Acid Sequence , Argininosuccinate Lyase/chemistry , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Aspartate Ammonia-Lyase/genetics , Catalysis , Catalytic Domain , Conserved Sequence , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fumarate Hydratase/genetics , Humans , Intramolecular Lyases/chemistry , Intramolecular Lyases/genetics , Intramolecular Lyases/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , delta-Crystallins/chemistry , delta-Crystallins/genetics , delta-Crystallins/metabolism
12.
J Biol Chem ; 286(24): 21100-9, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21498518

ABSTRACT

The enzyme fumarase catalyzes the reversible hydration of fumarate to malate. The reaction catalyzed by fumarase is critical for cellular energetics as a part of the tricarboxylic acid cycle, which produces reducing equivalents to drive oxidative ATP synthesis. A catalytic mechanism for the fumarase reaction that can account for the kinetic behavior of the enzyme observed in both isotope exchange studies and initial velocity studies has not yet been identified. In the present study, we develop an 11-state kinetic model of the enzyme based on the current consensus on its catalytic mechanism and design a series of experiments to estimate the model parameters and identify the major flux routes through the mechanism. The 11-state mechanism accounts for competitive binding of inhibitors and activation by different anions, including phosphate and fumarate. The model is identified from experimental time courses of the hydration of fumarate to malate obtained over a wide range of buffer and substrate concentrations. Further, the 11-state model is found to effectively reduce to a five-state model by lumping certain successive steps together to yield a mathematically less complex representation that is able to match the data. Analysis suggests the primary reaction route of the catalytic mechanism, with fumarate binding to the free unprotonated enzyme and a proton addition prior to malate release in the fumarate hydration reaction. In the reverse direction (malate dehydration), malate binds the protonated form of the enzyme, and a proton is generated before fumarate is released from the active site.


Subject(s)
Fumarate Hydratase/chemistry , Gene Expression Regulation, Enzymologic , Animals , Biochemistry/methods , Catalysis , Catalytic Domain , Citric Acid Cycle , Heart/physiology , Kinetics , Malates/chemistry , Models, Chemical , Myocardium/metabolism , Swine , Thermodynamics , Time Factors
13.
Biometals ; 25(1): 95-102, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21818585

ABSTRACT

Helicobacter pylori causes various gastric diseases, such as gastritis, peptic ulcerations and gastric cancer. Triple therapy combining bismuth compounds with two antibiotics is the cornerstone of the treatment of H. pylori infections. Up to now, the molecular mechanisms by which bismuth inhibits the growth of H. pylori are far from clear. In the bacterial tricarboxylic acid (TCA) cycle, fumarase catalyses the reversible hydration of fumarate to malic acid. Our previous proteomic work indicated that fumarase was capable of bismuth-binding. The interactions as well as the inhibitory effects of bismuth to fumarase have been characterized in this study. The titration of bismuth showed that each fumarase monomer binds one mol equiv of Bi(3+), with negligible secondary structural change. Bismuth-binding results in a near stoichiometric inactivation of the enzyme, leading to an apparent non-competitive mechanism as reflected by the Lineweaver-Burk plots. Our collective data indicate that the TCA cycle is a potential molecular target of bismuth drugs in H. pylori.


Subject(s)
Bacterial Proteins/metabolism , Bismuth/pharmacology , Citric Acid Cycle/drug effects , Fumarate Hydratase/antagonists & inhibitors , Helicobacter pylori/drug effects , Helicobacter pylori/enzymology , Bacterial Proteins/genetics , Bismuth/therapeutic use , Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Helicobacter Infections/drug therapy , Humans , Models, Molecular , Protein Structure, Tertiary
14.
Curr Microbiol ; 64(5): 412-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22302452

ABSTRACT

Little is known about the association among the transcription, post-transcription, and protein production of the fumA gene. This study demonstrates that increasing growth rate (k) from 0.24/h to 0.96/h causes a marked eightfold reduction in fumA transcription as assessed using the ß-galactosidase activity from fumA promoter fused with a lacZ reporter. It was further confirmed using Northern blot analysis. Most interestingly, the FumA protein levels remained unchanged over the growth rate, as indicated by Western blot analysis. Therefore, whether the reduced fumA mRNA expression under the high growth rate can be overcome by increasing the stability of the fumA mRNA was tested. The half-life of fumA mRNA was established to significantly increase by fivefold when the growth rate was increased to 0.96/h. This finding suggests that the cells could turn down the expression of fumA mRNA because of increased stability of its mRNA under the high growth rate. This notion indicates that mRNA stability plays an essential role in maintaining a critical cellular level of a given protein when the mRNA transcript is downregulated by a metabolic event.


Subject(s)
Down-Regulation , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Escherichia coli/growth & development , Fumarate Hydratase/genetics , Gene Expression Regulation, Bacterial , RNA, Messenger/chemistry , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Fumarate Hydratase/chemistry , Fumarate Hydratase/metabolism , Kinetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
15.
J Biomol Struct Dyn ; 40(3): 1260-1272, 2022 02.
Article in English | MEDLINE | ID: mdl-32969324

ABSTRACT

Fumarate hydratase (FH), one of the members of TCA cycle, acts as a catalyte for the synthesis of malate from fumarate. FH has been proposed to play as a tumour suppressor leading to the pathogenicity of leiomyomas, renal cell carcinoma and paraganglioma. Mutations in the active site of FH lead to alteration in the protein structure. Similarly, binding of several chemical inhibitors to the active site also leads to the disruption of protein structural integrity thereby leading to protein dysfunction. Therefore, in order to address this mechanism leading to cancer, the binding efficiency of potential human FH inhibitor citrate to zebrafish fh has been extensively analysed in this study by molecular docking and simulation experiments followed by quantification of fumarate hydratase enzyme activity to validate and confirm the findings. Molecular docking revealed stronger interaction of zebrafish fh protein with inhibitor citrate when compared to natural substrate fumarate. Study on the dynamics of docked structures further confirmed that citrate was found to possess more binding affinity than fumarate. In vitro biochemical analysis also revealed concentration dependent potential inhibitory effect of citrate on zebrafish fh, thus confirming the findings of the in-silico experiments.Communicated by Ramaswamy H. Sarma.


Subject(s)
Fumarate Hydratase , Zebrafish Proteins/chemistry , Animals , Catalytic Domain , Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Molecular Docking Simulation , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
16.
Hum Mutat ; 32(9): 1046-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21560188

ABSTRACT

Fumarase deficiency (FD), caused by biallelic alteration of the Fumarase Hydratase gene (FH), and a rare metabolic disorder that affects the Krebs cycle, causes severe neurological impairment and fumaric aciduria. Less than 30 unrelated cases are known to date. In addition, heterozygous mutations of the FH gene are responsible for hereditary leiomyomatosis and renal cell cancer (HLRCC). We report three additional patients with dramatically different clinical presentations of FD and novel missense mutations in the FH gene. One patient had severe neonatal encephalopathy, polymicrogyria, <1% enzyme activity, and mildly increased levels of urinary fumarate. The second patient had microcephaly, mental retardation, 20% of fumarase activity, and intermediate levels of urinary fumarate. The third patient had mild mental retardation, polymicrogyria, 42-61% enzyme activity in different cell types and massive amounts of urinary fumarate. In silico analysis predicted minor yet significant structural changes in the encoded proteins. The nuclear translocation of hypoxia-inducible factor (HIF)-1alpha (HIF1A) in cultured fibroblasts was similar to controls. These results extend the range of clinical and biochemical variation associated with FD, supporting the notion that patients with moderate increases in fumarate excretion should be investigated for this disease. The tumoral risk in the patients and their relatives requires adequate screening protocols.


Subject(s)
Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Malformations of Cortical Development/enzymology , Malformations of Cortical Development/pathology , Cell Hypoxia , Child , Child, Preschool , Computer Simulation , Female , Fumarate Hydratase/chemistry , Fumarates/urine , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mutation/genetics , Signal Transduction
17.
J Inherit Metab Dis ; 34(3): 671-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21445611

ABSTRACT

Fumarate hydratase catalyzes the stereospecific hydration across the olefinic double bond in fumarate leading to L-malate. The enzyme is expressed in mitochondrial and cytosolic compartments, and participates in the Krebs cycle in mitochondria, as well as in regulation of cytosolic fumarate levels. Fumarate hydratase deficiency is an autosomal recessive trait presenting as metabolic disorder with severe encephalopathy, seizures and poor neurological outcome. Heterozygous mutations are associated with a predisposition to cutaneous and uterine leiomyomas and to renal cancer. The crystal structure of human fumarate hydratase shows that mutations can be grouped into two distinct classes either affecting structural integrity of the core enzyme architecture, or are localized around the enzyme active site. An interactive version of this manuscript (which may contain additional mutations appended after acceptance of this manuscript) may be found on the SSIEM website at: http://www.ssiem.org/resources/structures/FH .


Subject(s)
Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Metabolism, Inborn Errors/etiology , Catalytic Domain/genetics , Crystallography, X-Ray , Fumarate Hydratase/deficiency , Humans , Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Models, Molecular , Mutant Proteins/chemistry , Mutation/physiology , Protein Conformation , Protein Folding , Structure-Activity Relationship
18.
Article in English | MEDLINE | ID: mdl-21904061

ABSTRACT

Rickettsiae are obligate intracellular parasites of eukaryotic cells that are the causative agents responsible for spotted fever and typhus. Their small genome (about 800 protein-coding genes) is highly conserved across species and has been postulated as the ancestor of the mitochondria. No genes that are required for glycolysis are found in the Rickettsia prowazekii or mitochondrial genomes, but a complete set of genes encoding components of the tricarboxylic acid cycle and the respiratory-chain complex is found in both. A 2.4 Å resolution crystal structure of R. prowazekii fumarate hydratase, an enzyme catalyzing the third step of the tricarboxylic acid cycle pathway that ultimately converts phosphoenolpyruvate into succinyl-CoA, has been solved. A structure alignment with human mitochondrial fumarate hydratase highlights the close similarity between R. prowazekii and mitochondrial enzymes.


Subject(s)
Fumarate Hydratase/chemistry , Mitochondria/chemistry , Rickettsia prowazekii/enzymology , Crystallography, X-Ray , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary
19.
Microb Cell Fact ; 9: 91, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092234

ABSTRACT

BACKGROUND: Fumarase catalyzes the reversible hydration of fumarate to L-malate and is a key enzyme in the tricarboxylic acid (TCA) cycle and in amino acid metabolism. Fumarase is also used for the industrial production of L-malate from the substrate fumarate. Thermostable and high-activity fumarases from organisms that inhabit extreme environments may have great potential in industry, biotechnology, and basic research. The marine environment is highly complex and considered one of the main reservoirs of microbial diversity on the planet. However, most of the microorganisms are inaccessible in nature and are not easily cultivated in the laboratory. Metagenomic approaches provide a powerful tool to isolate and identify enzymes with novel biocatalytic activities for various biotechnological applications. RESULTS: A plasmid metagenomic library was constructed from uncultivated marine microorganisms within marine water samples. Through sequence-based screening of the DNA library, a gene encoding a novel fumarase (named FumF) was isolated. Amino acid sequence analysis revealed that the FumF protein shared the greatest homology with Class II fumarate hydratases from Bacteroides sp. 2_1_33B and Parabacteroides distasonis ATCC 8503 (26% identical and 43% similar). The putative fumarase gene was subcloned into pETBlue-2 vector and expressed in E. coli BL21(DE3)pLysS. The recombinant protein was purified to homogeneity. Functional characterization by high performance liquid chromatography confirmed that the recombinant FumF protein catalyzed the hydration of fumarate to form L-malate. The maximum activity for FumF protein occurred at pH 8.5 and 55°C in 5 mM Mg(2+). The enzyme showed higher affinity and catalytic efficiency under optimal reaction conditions: K(m) = 0.48 mM, V(max) = 827 µM/min/mg, and k(cat)/K(m) = 1900 mM/s. CONCLUSIONS: We isolated a novel fumarase gene, fumF, from a sequence-based screen of a plasmid metagenomic library from uncultivated marine microorganisms. The properties of FumF protein may be ideal for the industrial production of L-malate under higher temperature conditions. The identification of FumF underscores the potential of marine metagenome screening for novel biomolecules.


Subject(s)
Fumarate Hydratase/genetics , Amino Acid Sequence , Aquatic Organisms/enzymology , Aquatic Organisms/genetics , Cloning, Molecular , Computational Biology , Fumarate Hydratase/chemistry , Fumarate Hydratase/classification , Fumarates/metabolism , Kinetics , Malates/metabolism , Metagenome , Molecular Sequence Data , Phylogeny , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Temperature
20.
FEBS Lett ; 594(2): 337-357, 2020 01.
Article in English | MEDLINE | ID: mdl-31514245

ABSTRACT

Fumarase C (FumC) catalyzes the reversible conversion of fumarate to S-malate. Previous structural investigations within the superfamily have reported a dynamic structural segment, termed the SS Loop. To date, active-site asymmetry has raised the question of how SS Loop placement affects participation of key residues during the reaction. Herein, we report structural and kinetic analyses from Escherichia coli FumC variants to understand the contribution of SS Loop residues S318, K324, and N326. High-resolution X-ray crystallographic results reveal three distinct FumC active-site conformations; disordered-open, ordered-open, and the newly discovered ordered-closed. Surprisingly, each SS Loop variant has unaffected Michaelis constants coupled to reductions in turnover number. Based upon our structural and functional analyses, we propose structural and catalytic roles for each of the aforementioned residues.


Subject(s)
Catalysis , Fumarate Hydratase/ultrastructure , Protein Conformation , Amino Acid Sequence/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , Escherichia coli/enzymology , Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Kinetics , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL