Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 62(23): e202302490, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37014271

ABSTRACT

Lanthipeptides are ribosomally-synthesized natural products from bacteria featuring stable thioether-crosslinks and various bioactivities. Herein, we report on a new clade of tricyclic class-IV lanthipeptides with curvocidin from Thermomonospora curvata as its first representative. We obtained crystal structures of the corresponding lanthipeptide synthetase CuvL that showed a circular arrangement of its kinase, lyase and cyclase domains, forming a central reaction chamber for the iterative substrate processing involving nine catalytic steps. The combination of experimental data and artificial intelligence-based structural models identified the N-terminal subdomain of the kinase domain as the primary site of substrate recruitment. The ribosomal precursor peptide of curvocidin employs an amphipathic α-helix in its leader region as an anchor to CuvL, while its substrate core shuttles within the central reaction chamber. Our study thus reveals general principles of domain organization and substrate recruitment of class-IV and class-III lanthipeptide synthetases.


Subject(s)
Artificial Intelligence , Ligases , Ligases/chemistry , Peptides/chemistry
2.
Met Ions Life Sci ; 202020 Mar 23.
Article in English | MEDLINE | ID: mdl-32851832

ABSTRACT

Enzymes relying on the interplay of nickel, iron, and sulfur in their active sites are used by prokaryotes to catalyze reactions driving the global carbon and hydrogen cycles. The three enzymes, [NiFe] hydrogenases, Ni,Fe-containing carbon monoxide dehydrogenases and acetyl-CoA synthases share an ancient origin possibly derived from abiotic processes. Although their active sites have different compositions and assemble Ni, Fe, and S in different ways and for different purposes, they share a central role of Ni in substrate binding and activation, with sulfur linking the Ni ion to one or more Fe ions, which, although indispensable for function, supports the catalytic process in less understood ways. The review gives a short overview on the properties of the three individual enzymes highlighting their parallels and differences.


Subject(s)
Nickel/metabolism , Binding Sites , Catalytic Domain , Hydrogenase/metabolism , Iron/metabolism , Iron-Sulfur Proteins , Sulfur
3.
Chembiochem ; 21(12): 1710-1716, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32187824

ABSTRACT

Three different types of electron-transferring metallo-ATPases are able to couple ATP hydrolysis to the reduction of low-potential metal sites, thereby energizing an electron. Besides the Fe-protein known from nitrogenase and homologous enzymes, two other kinds of ATPase with different scaffolds and cofactors are used to achieve a unidirectional, energetic, uphill electron transfer to either reduce inactive Co-corrinoid-containing proteins (RACE-type activators) or a second iron-sulfur cluster-containing enzyme of a unique radical enzymes family (archerases). We have found a new cofactor in the latter enzyme family, that is, a double-cubane cluster with two [4Fe4S] subclusters bridged by a sulfido ligand. An enzyme containing this cofactor catalyzes the ATP-dependent reduction of small molecules, including acetylene. Thus, enzymes containing the double-cubane cofactor are analogous in function and share some structural features with nitrogenases.


Subject(s)
Iron-Sulfur Proteins/metabolism , Nitrogenase/chemistry , Acetylene/chemistry , Acetylene/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Biocatalysis , Iron-Sulfur Proteins/chemistry , Models, Molecular , Nitrogenase/metabolism , Oxidation-Reduction
4.
Methods Mol Biol ; 1876: 37-54, 2019.
Article in English | MEDLINE | ID: mdl-30317473

ABSTRACT

Carbon monoxide dehydrogenases (CODHs) catalyze the reversible oxidation of CO with water to CO2, two electrons, and two protons. Two classes of CODHs exist, having evolved from different scaffolds featuring active sites built from different transition metals. The basic properties of both classes are described in this overview chapter.


Subject(s)
Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , Carbon Monoxide/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Aldehyde Oxidoreductases/classification , Anaerobiosis , Archaea/enzymology , Bacteria, Anaerobic/enzymology , Catalytic Domain , Evolution, Molecular , Models, Molecular , Multienzyme Complexes/classification , Protein Conformation
5.
Methods Mol Biol ; 1876: 167-178, 2019.
Article in English | MEDLINE | ID: mdl-30317481

ABSTRACT

Carbon monoxide dehydrogenases (CODHs) are central players in the biogeochemical carbon monoxide (CO) cycle and have been extensively studied from the ecological level to the structural/molecular level. Of the two types of CODHs, the oxygen-tolerant CODHs use a bimetallic [CuSMo(=O)OH] center connected to the protein via a pyranopterin cofactor, whereas the oxygen-sensitive CODHs contain a [NiFe4S4-OHx]-cluster. Despite the fact that we have a basic understanding of how both types of CODHs use distinct active sites to catalyze the oxidation of CO with water to CO2, two protons, and two electrons (a reversible reaction in the cases of the oxygen-sensitive CODHs), many questions remain unanswered, especially concerning the electronic structures of the intermediate states. Since these states will likely be only revealed by the interplay of experimental and theoretical methods, there is a need to obtain accurate descriptions of the active site architectures in various states and, consequently, a need to generate crystals with good diffraction quality and collect data at element-specific wavelengths in order to determine the identity of elements in the case of mixed states. This chapter provides a description of the general working protocols for the crystallization and structural analysis of Cu,Mo-CODH and Ni,Fe-CODH that facilitates the mechanistic investigations of these important metalloenzymes.


Subject(s)
Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , Metalloproteins/chemistry , Metalloproteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Binding Sites , Carbon Monoxide/metabolism , Catalytic Domain , Crystallography, X-Ray , Iron/chemistry , Models, Molecular , Molybdenum/chemistry , Nickel/chemistry , Water/metabolism
6.
Angew Chem Int Ed Engl ; 58(9): 2899-2903, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30589180

ABSTRACT

Allosteric information transfer in proteins has been linked to distinct vibrational energy transfer (VET) pathways in a number of theoretical studies. Experimental evidence for such pathways, however, is sparse because site-selective injection of vibrational energy into a protein, that is, localized heating, is required for their investigation. Here, we solved this problem by the site-specific incorporation of the non-canonical amino acid ß-(1-azulenyl)-l-alanine (AzAla) through genetic code expansion. As an exception to Kasha's rule, AzAla undergoes ultrafast internal conversion and heating after S1 excitation while upon S2 excitation, it serves as a fluorescent label. We equipped PDZ3, a protein interaction domain of postsynaptic density protein 95, with this ultrafast heater at two distinct positions. We indeed observed VET from the incorporation sites in the protein to a bound peptide ligand on the picosecond timescale by ultrafast IR spectroscopy. This approach based on genetically encoded AzAla paves the way for detailed studies of VET and its role in a wide range of proteins.


Subject(s)
Alanine/chemistry , Energy Transfer , Alanine/genetics , Models, Molecular , Vibration
7.
Chembiochem ; 18(18): 1819-1823, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28650092

ABSTRACT

Marine mussels exhibit potent underwater adhesion abilities under hostile conditions by employing 3,4-dihydroxyphenylalanine (DOPA)-rich mussel adhesive proteins (MAPs). However, their recombinant production is a major biotechnological challenge. Herein, a novel strategy based on genetic code expansion has been developed by engineering efficient aminoacyl-transfer RNA synthetases (aaRSs) for the photocaged noncanonical amino acid ortho-nitrobenzyl DOPA (ONB-DOPA). The engineered ONB-DOPARS enables in vivo production of MAP type 5 site-specifically equipped with multiple instances of ONB-DOPA to yield photocaged, spatiotemporally controlled underwater adhesives. Upon exposure to UV light, these proteins feature elevated wet adhesion properties. This concept offers new perspectives for the production of recombinant bioadhesives.


Subject(s)
Bivalvia/metabolism , Genetic Code/genetics , Proteins/metabolism , Adhesives/radiation effects , Amino Acyl-tRNA Synthetases/metabolism , Animals , Biomimetic Materials/metabolism , Bivalvia/genetics , Dihydroxyphenylalanine/metabolism , Microscopy, Atomic Force , Microscopy, Scanning Probe , Mutagenesis, Site-Directed , Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Ultraviolet Rays
8.
J Mol Microbiol Biotechnol ; 26(1-3): 165-79, 2016.
Article in English | MEDLINE | ID: mdl-26959729

ABSTRACT

Short-chain fatty acids such as acetic, propionic, butyric or lactic acids are typical primary fermentation products in the anaerobic feeding chain. Fifteen years ago, a novel fermentation type was discovered in the obligately anaerobic Deltaproteobacterium Syntrophus aciditrophicus. During fermentative growth with crotonate and/or benzoate, acetate is formed in the oxidative branch and cyclohexane carboxylate in the reductive branch. In both cases cyclohexa-1,5-diene-1-carboxyl-CoA (Ch1,5CoA) is a central intermediate that is either formed by a class II benzoyl-CoA reductase (fermentation of benzoate) or by reverse reactions of the benzoyl-CoA degradation pathway (fermentation of crotonate). Here, we summarize the current knowledge of the enzymology involved in fermentations yielding cyclohexane carboxylate as an excreted product. The characteristic enzymes involved are two acyl-CoA dehydrogenases specifically acting on Ch1,5CoA and cyclohex-1-ene-1-carboxyl-CoA. Both enzymes are also employed during the syntrophic growth of S. aciditrophicus with cyclohexane carboxylate as the carbon source in coculture with a methanogen. An investigation of anabolic pathways in S. aciditrophicus revealed a rather unusual pathway for glutamate synthesis involving a Re-citrate synthase. Future work has to address the unresolved question concerning which components are involved in reoxidation of the NADH formed in the oxidative branch of the unique cyclohexane carboxylate fermentation pathway in S. aciditrophicus.


Subject(s)
Cyclohexanecarboxylic Acids/metabolism , Deltaproteobacteria/metabolism , Bacteria, Anaerobic/metabolism , Cyclohexanecarboxylic Acids/chemistry , Deltaproteobacteria/enzymology , Deltaproteobacteria/genetics , Enzyme Activation , Fermentation , Metabolic Networks and Pathways , Oxidation-Reduction
9.
J Mol Microbiol Biotechnol ; 26(1-3): 76-91, 2016.
Article in English | MEDLINE | ID: mdl-26959876

ABSTRACT

4-Hydroxyphenylacetate decarboxylase (4Hpad) is the prototype of a new class of Fe-S cluster-dependent glycyl radical enzymes (Fe-S GREs) acting on aromatic compounds. The two-enzyme component system comprises a decarboxylase responsible for substrate conversion and a dedicated activating enzyme (4Hpad-AE). The decarboxylase uses a glycyl/thiyl radical dyad to convert 4-hydroxyphenylacetate into p-cresol (4-methylphenol) by a biologically unprecedented Kolbe-type decarboxylation. In addition to the radical dyad prosthetic group, the decarboxylase unit contains two [4Fe-4S] clusters coordinated by an extra small subunit of unknown function. 4Hpad-AE reductively cleaves S-adenosylmethionine (SAM or AdoMet) at a site-differentiated [4Fe-4S]2+/+ cluster (RS cluster) generating a transient 5'-deoxyadenosyl radical that produces a stable glycyl radical in the decarboxylase by the abstraction of a hydrogen atom. 4Hpad-AE binds up to two auxiliary [4Fe-4S] clusters coordinated by a ferredoxin-like insert that is C-terminal to the RS cluster-binding motif. The ferredoxin-like domain with its two auxiliary clusters is not vital for SAM-dependent glycyl radical formation in the decarboxylase, but facilitates a longer lifetime for the radical. This review describes the 4Hpad and cognate AE families and focuses on the recent advances and open questions concerning the structure, function and mechanism of this novel Fe-S-dependent class of GREs.


Subject(s)
Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Enzyme Activation , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Hydrocarbons, Aromatic/metabolism
10.
J Mol Microbiol Biotechnol ; 26(1-3): 5-28, 2016.
Article in English | MEDLINE | ID: mdl-26960061

ABSTRACT

Hydrocarbons are abundant in anoxic environments and pose biochemical challenges to their anaerobic degradation by microorganisms. Within the framework of the Priority Program 1319, investigations funded by the Deutsche Forschungsgemeinschaft on the anaerobic microbial degradation of hydrocarbons ranged from isolation and enrichment of hitherto unknown hydrocarbon-degrading anaerobic microorganisms, discovery of novel reactions, detailed studies of enzyme mechanisms and structures to process-oriented in situ studies. Selected highlights from this program are collected in this synopsis, with more detailed information provided by theme-focused reviews of the special topic issue on 'Anaerobic biodegradation of hydrocarbons' [this issue, pp. 1-244]. The interdisciplinary character of the program, involving microbiologists, biochemists, organic chemists and environmental scientists, is best exemplified by the studies on alkyl-/arylalkylsuccinate synthases. Here, research topics ranged from in-depth mechanistic studies of archetypical toluene-activating benzylsuccinate synthase, substrate-specific phylogenetic clustering of alkyl-/arylalkylsuccinate synthases (toluene plus xylenes, p-cymene, p-cresol, 2-methylnaphthalene, n-alkanes), stereochemical and co-metabolic insights into n-alkane-activating (methylalkyl)succinate synthases to the discovery of bacterial groups previously unknown to possess alkyl-/arylalkylsuccinate synthases by means of functional gene markers and in situ field studies enabled by state-of-the-art stable isotope probing and fractionation approaches. Other topics are Mo-cofactor-dependent dehydrogenases performing O2-independent hydroxylation of hydrocarbons and alkyl side chains (ethylbenzene, p-cymene, cholesterol, n-hexadecane), degradation of p-alkylated benzoates and toluenes, glycyl radical-bearing 4-hydroxyphenylacetate decarboxylase, novel types of carboxylation reactions (for acetophenone, acetone, and potentially also benzene and naphthalene), W-cofactor-containing enzymes for reductive dearomatization of benzoyl-CoA (class II benzoyl-CoA reductase) in obligate anaerobes and addition of water to acetylene, fermentative formation of cyclohexanecarboxylate from benzoate, and methanogenic degradation of hydrocarbons.


Subject(s)
Bacteria, Anaerobic/metabolism , Biodegradation, Environmental , Hydrocarbons/metabolism , Anaerobiosis , Bacteria, Anaerobic/enzymology , Bacteria, Anaerobic/genetics , Biodiversity , Phylogeny
11.
J Mol Microbiol Biotechnol ; 26(1-3): 29-44, 2016.
Article in English | MEDLINE | ID: mdl-26959246

ABSTRACT

The pathway of anaerobic toluene degradation is initiated by a remarkable radical-type enantiospecific addition of the chemically inert methyl group to the double bond of a fumarate cosubstrate to yield (R)-benzylsuccinate as the first intermediate, as catalyzed by the glycyl radical enzyme benzylsuccinate synthase. In recent years, it has become clear that benzylsuccinate synthase is the prototype enzyme of a much larger family of fumarate-adding enzymes, which play important roles in the anaerobic metabolism of further aromatic and even aliphatic hydrocarbons. We present an overview on the biochemical properties of benzylsuccinate synthase, as well as its recently solved structure, and present the results of an initial structure-based modeling study on the reaction mechanism. Moreover, we compare the structure of benzylsuccinate synthase with those predicted for different clades of fumarate-adding enzymes, in particular the paralogous enzymes converting p-cresol, 2-methylnaphthalene or n-alkanes.


Subject(s)
Carbon-Carbon Lyases/chemistry , Carbon-Carbon Lyases/metabolism , Fumarates/metabolism , Anaerobiosis , Bacteria, Anaerobic/enzymology , Bacteria, Anaerobic/genetics , Bacteria, Anaerobic/metabolism , Bacterial Proteins/metabolism , Biodegradation, Environmental , Carbon-Carbon Lyases/genetics , Isoenzymes , Metabolic Networks and Pathways , Models, Molecular , Phylogeny , Structure-Activity Relationship , Toluene/metabolism
12.
Appl Environ Microbiol ; 81(3): 1071-84, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452282

ABSTRACT

4-Hydroxybutyryl-coenzyme A (CoA) dehydratase (4HBD) from Clostridium aminobutyricum catalyzes the reversible dehydration of 4-hydroxybutyryl-CoA to crotonyl-CoA and the irreversible isomerization of vinylacetyl-CoA to crotonyl-CoA. 4HBD is an oxygen-sensitive homotetrameric enzyme with one [4Fe-4S](2+) cluster and one flavin adenine dinucleotide (FAD) in each subunit. Upon the addition of crotonyl-CoA or the analogues butyryl-CoA, acetyl-CoA, and CoA, UV-visible light and electron paramagnetic resonance (EPR) spectroscopy revealed an internal one-electron transfer to FAD and the [4Fe-4S](2+) cluster prior to hydration. We describe an active recombinant 4HBD and variants produced in Escherichia coli. The variants of the cluster ligands (H292C [histidine at position 292 is replaced by cysteine], H292E, C99A, C103A, and C299A) had no measurable dehydratase activity and were composed of monomers, dimers, and tetramers. Variants of other potential catalytic residues were composed only of tetramers and exhibited either no measurable (E257Q, E455Q, and Y296W) hydratase activity or <1% (Y296F and T190V) dehydratase activity. The E455Q variant but not the Y296F or E257Q variant displayed the same spectral changes as the wild-type enzyme after the addition of crotonyl-CoA but at a much lower rate. The results suggest that upon the addition of a substrate, Y296 is deprotonated by E455 and reduces FAD to FADH·, aided by protonation from E257 via T190. In contrast to FADH·, the tyrosyl radical could not be detected by EPR spectroscopy. FADH· appears to initiate the radical dehydration via an allylic ketyl radical that was proposed 19 years ago. The mode of radical generation in 4HBD is without precedent in anaerobic radical chemistry. It differs largely from that in enzymes, which use coenzyme B12, S-adenosylmethionine, ATP-driven electron transfer, or flavin-based electron bifurcation for this purpose.


Subject(s)
Clostridium/enzymology , Free Radicals/metabolism , Hydro-Lyases/metabolism , Acetyl Coenzyme A/metabolism , Acyl Coenzyme A/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Flavin-Adenine Dinucleotide/metabolism , Iron-Sulfur Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrum Analysis
13.
J Biol Inorg Chem ; 19(8): 1317-26, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25156152

ABSTRACT

4-Hydroxyphenylacetate decarboxylase-activating enzyme (4Hpad-AE) uses S-adenosylmethionine (SAM or AdoMet) and a [4Fe-4S] ²âº/⁺cluster (RS cluster) to generate a stable glycyl radical on the decarboxylase. 4Hpad-AE might bind up to two auxiliary [4Fe-4S] clusters coordinated by a ferredoxin-like insert C-terminal to the RS cluster-binding motif. Except for the AEs of pyruvate formate-lyase and anaerobic ribonucleotide reductase, all glycyl radical-activating enzymes possess a similar ferredoxin-like domain, whose functional role is still poorly understood. To assess the role of the putative ferredoxin clusters from 4Hpad-AE, we combined biochemical and spectroscopic methods to characterize a truncated version of the protein (Δ66-AE) devoid of the ferredoxin-like domain. We found that Δ66-AE is stable, harbors a fully active RS cluster and can activate the decarboxylase. From the similar cleavage rates for S-adenosylmethionine of Δ66-AE and wild-type AE, we infer the reactivity of the RS cluster is unperturbed by the absence of the ferredoxin-like domain. Thus, the auxiliary clusters are not required as electron conduit to the RS cluster for effective reductive cleavage of SAM. The activation of the decarboxylase by Δ66-AE is almost as fast as with wild-type AE, but the generated glycyl radical is short living. We postulate that the ferredoxin-like domain is not required for SAM-dependent glycyl radical generation in the decarboxylase, but is necessary for producing a lasting glycyl radical.


Subject(s)
Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Ferredoxins/chemistry , Glycine/metabolism , Carboxy-Lyases/genetics , Cloning, Molecular , Enzyme Activation , Free Radicals/chemistry , Free Radicals/metabolism , Glycine/chemistry , Models, Molecular , Protein Structure, Tertiary
14.
J Am Chem Soc ; 135(39): 14574-85, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24028464

ABSTRACT

Using continuum electrostatics and QC/MM calculations, we investigate the catalytic cycle of the glycyl radical enzyme 4-hydroxyphenylacetate decarboxylase, an enzyme involved in the fermentative production of p-cresol from tyrosine in clostridia. On the basis of our calculations, we propose a five-step mechanism for the reaction. In the first step, the substrate 4-hydroxyphenylacetate is activated by an unusual concerted abstraction of an electron and a proton. Namely, Cys503 radical abstracts an electron from the substrate and Glu637 abstracts a proton. Thus in total, a hydrogen atom is abstracted from the substrate. In the second step, the carboxylic group readily splits off from the phenoxy-acetate radical anion to give carbon dioxide. This decarboxylation step is coupled to a proton transfer from Glu637 back to the phenolic hydroxyl group which leads to a p-hydroxybenzyl radical. The remaining steps of the reaction involve a rotation of the Cys503 side chain followed by a proton transfer from Glu505 to Cys503 and a hydrogen atom transfer from Cys503 to the p-hydroxybenzyl radical to form p-cresol. The calculated mechanism agrees with experimental data suggesting that both Cys503 and Glu637 are essential for the catalytic function of 4-hydroxyphenylacetate decarboxylase and that the substrate requires a hydroxyl group in para-position to the acetate moiety.


Subject(s)
Carboxy-Lyases/metabolism , Clostridium/enzymology , Carboxy-Lyases/chemistry , Catalytic Domain , Clostridium/chemistry , Decarboxylation , Models, Molecular , Quantum Theory , Static Electricity
15.
J Biol Inorg Chem ; 18(6): 633-43, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716017

ABSTRACT

4-Hydroxyphenylacetate decarboxylase (4Hpad) is an Fe/S cluster containing glycyl radical enzyme (GRE), which catalyses the last step of tyrosine fermentation in clostridia, generating the bacteriostatic p-cresol. The respective activating enzyme (4Hpad-AE) displays two cysteine-rich motifs in addition to the classical S-adenosylmethionine (SAM) binding cluster (RS cluster) motif. These additional motifs are also present in other glycyl radical activating enzymes (GR-AE) and it has been postulated that these orthologues may use an alternative SAM homolytic cleavage mechanism, generating a putative 3-amino-3-carboxypropyl radical and 5'-deoxy-5'-(methylthio)adenosine but not a 5'-deoxyadenosyl radical and methionine. 4Hpad-AE produced from a codon-optimized synthetic gene binds a maximum of two [4Fe-4S](2+/+) clusters as revealed by EPR and Mössbauer spectroscopy. The enzyme only catalyses the turnover of SAM under reducing conditions, and the reaction products were identified as 5'-deoxyadenosine (quenched form of 5'-deoxyadenosyl radical) and methionine. We demonstrate that the 5'-deoxyadenosyl radical is the activating agent for 4Hpad through p-cresol formation and correlation between the production of 5'-deoxyadenosine and the generation of glycyl radical in 4Hpad. Therefore, we conclude that 4Hpad-AE catalyses a classical SAM-dependent glycyl radical formation as reported for GR-AE without auxiliary clusters. Our observation casts doubt on the suggestion that GR-AE containing auxiliary clusters catalyse the alternative cleavage reaction detected for glycerol dehydratase activating enzyme.


Subject(s)
Biocatalysis , Carboxy-Lyases/metabolism , S-Adenosylmethionine/metabolism , Carboxy-Lyases/chemistry , Carboxy-Lyases/isolation & purification , Clostridium/enzymology , Electron Spin Resonance Spectroscopy , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , S-Adenosylmethionine/chemistry , Spectroscopy, Mossbauer
16.
Mol Microbiol ; 86(4): 908-20, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23013274

ABSTRACT

Under phosphate starvation conditions, Escherichia coli can utilize sn-glycerol-3-phosphate (G3P) and G3P diesters as phosphate source when transported by an ATP binding cassette importer composed of the periplasmic binding protein, UgpB, the transmembrane subunits, UgpA and UgpE, and a homodimer of the nucleotide binding subunit, UgpC. The current knowledge on the Ugp transporter is solely based on genetic evidence and transport assays using intact cells. Thus, we set out to characterize its properties at the level of purified protein components. UgpB was demonstrated to bind G3P and glycerophosphocholine with dissociation constants of 0.68 ± 0.02 µM and 5.1 ± 0.3 µM, respectively, while glycerol-2-phosphate (G2P) is not a substrate. The crystal structure of UgpB in complex with G3P was solved at 1.8 Å resolution and revealed the interaction with two tryptophan residues as key to the preferential binding of linear G3P in contrast to the branched G2P. Mutational analysis validated the crucial role of Trp-169 for G3P binding. The purified UgpAEC2 complex displayed UgpB/G3P-stimulated ATPase activity in proteoliposomes that was neither inhibited by phosphate nor by the signal transducing protein PhoU or the phosphodiesterase UgpQ. Furthermore, a hybrid transporter composed of MalFG-UgpC could be functionally reconstituted while a UgpAE-MalK complex was unstable.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Glycerophosphates/metabolism , Membrane Transport Proteins/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Crystallography, X-Ray , DNA Mutational Analysis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Esters/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/isolation & purification , Mutant Proteins/genetics , Mutant Proteins/isolation & purification , Mutant Proteins/metabolism , Protein Conformation , Substrate Specificity
17.
Biochim Biophys Acta ; 1824(11): 1278-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22178228

ABSTRACT

BACKGROUND: The steadily increasing field of radical biochemistry is dominated by the large family of S-adenosylmethionine dependent enzymes, the so-called radical SAM enzymes, of which several new members are discovered every year. Here we report on 2- and 4-hydroxyacyl-CoA dehydratases which apply a very different method of radical generation. In these enzymes ketyl radicals are formed by one-electron reduction or oxidation and are recycled after each turnover without further energy input. Earlier reviews on 2-hydroxyacyl-CoA dehydratases were published in 2004 [J. Kim, M. Hetzel, C.D. Boiangiu, W. Buckel, FEMS Microbiol. Rev. 28 (2004) 455-468. W. Buckel, M. Hetzel, J. Kim, Curr. Opin. Chem. Biol. 8 (2004) 462-467.] SCOPE OF REVIEW: The review focuses on four types of 2-hydroxyacyl-CoA dehydratases that are involved in the fermentation of amino acids by anaerobic bacteria, especially clostridia. These enzymes require activation by one-electron transfer from an iron-sulfur protein driven by hydrolysis of ATP. The review further describes the proposed mechanism that is highlighted by the identification of the allylic ketyl radical intermediate and the elucidation of the crystal structure of 2-hydroxyisocapryloyl-CoA dehydratase. With 4-hydroxybutyryl-CoA dehydratase the crystal structure, the complete stereochemistry and the function of several conserved residues around the active site could be identified. Finally potential biotechnological applications of the radical dehydratases are presented. GENERAL SIGNIFICANCE: The action of the activator as an 'Archerase' shooting electrons into difficultly reducible acceptors becomes an emerging principle in anaerobic metabolism. The dehydratases may provide useful tools in biotechnology. This article is part of a Special Issue entitled: Radical SAM enzymes and Radical Enzymology.


Subject(s)
Bacterial Proteins/metabolism , Hydro-Lyases/metabolism , Iron-Sulfur Proteins/metabolism , S-Adenosylmethionine/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Anaerobiosis , Bacteria, Anaerobic/metabolism , Bacterial Proteins/chemistry , Biocatalysis , Catalytic Domain , Free Radicals/chemistry , Free Radicals/metabolism , Hydro-Lyases/chemistry , Iron-Sulfur Proteins/chemistry , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Oxidation-Reduction , S-Adenosylmethionine/chemistry , Stereoisomerism
18.
J Am Chem Soc ; 133(37): 14666-74, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21823587

ABSTRACT

4-Hydroxyphenylacetate decarboxylase is a [4Fe-4S] cluster containing glycyl radical enzyme proposed to use a glycyl/thiyl radical dyad to catalyze the last step of tyrosine fermentation in clostridia. The decarboxylation product p-cresol (4-methylphenol) is a virulence factor of the human pathogen Clostridium difficile . Here we describe the crystal structures at 1.75 and 1.81 Å resolution of substrate-free and substrate-bound 4-hydroxyphenylacetate decarboxylase from the related Clostridium scatologenes . The structures show a (ßγ)(4) tetramer of heterodimers composed of a catalytic ß-subunit harboring the putative glycyl/thiyl dyad and a distinct small γ-subunit with two [4Fe-4S] clusters at 40 Å distance from the active site. The γ-subunit comprises two domains displaying pseudo-2-fold symmetry that are structurally related to the [4Fe-4S] cluster-binding scaffold of high-potential iron-sulfur proteins. The N-terminal domain coordinates one cluster with one histidine and three cysteines, and the C-terminal domain coordinates the second cluster with four cysteines. Whereas the C-terminal cluster is buried in the ßγ heterodimer interface, the N-terminal cluster is not part of the interface. The previously postulated decarboxylation mechanism required the substrate's hydroxyl group in the vicinity of the active cysteine residue. In contrast to expectation, the substrate-bound state shows a direct interaction between the substrate's carboxyl group and the active site Cys503, while His536 and Glu637 at the opposite side of the active site pocket anchor the hydroxyl group. This state captures a possible catalytically competent complex and suggests a Kolbe-type decarboxylation for p-cresol formation.


Subject(s)
Carboxy-Lyases/chemistry , Clostridium/enzymology , Iron-Sulfur Proteins/chemistry , Carboxy-Lyases/metabolism , Clostridium/chemistry , Cresols/metabolism , Crystallography, X-Ray , Decarboxylation , Iron-Sulfur Proteins/metabolism , Models, Molecular , Protein Binding , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Substrate Specificity
19.
Biochem J ; 425(2): 373-80, 2009 Dec 23.
Article in English | MEDLINE | ID: mdl-19860741

ABSTRACT

The eukaryotic transcription elongation factor DSIF [DRB (5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole) sensitivity-inducing factor] is composed of two subunits, hSpt4 and hSpt5, which are homologous to the yeast factors Spt4 and Spt5. DSIF is involved in regulating the processivity of RNA polymerase II and plays an essential role in transcriptional activation of eukaryotes. At several eukaryotic promoters, DSIF, together with NELF (negative elongation factor), leads to promoter-proximal pausing of RNA polymerase II. In the present paper we describe the crystal structure of hSpt4 in complex with the dimerization region of hSpt5 (amino acids 176-273) at a resolution of 1.55 A (1 A=0.1 nm). The heterodimer shows high structural similarity to its homologue from Saccharomyces cerevisiae. Furthermore, hSpt5-NGN is structurally similar to the NTD (N-terminal domain) of the bacterial transcription factor NusG. A homologue for hSpt4 has not yet been found in bacteria. However, the archaeal transcription factor RpoE" appears to be distantly related. Although a comparison of the NusG-NTD of Escherichia coli with hSpt5 revealed a similarity of the three-dimensional structures, interaction of E. coli NusG-NTD with hSpt4 could not be observed by NMR titration experiments. A conserved glutamate residue, which was shown to be crucial for dimerization in yeast, is also involved in the human heterodimer, but is substituted for a glutamine residue in Escherichia coli NusG. However, exchanging the glutamine for glutamate proved not to be sufficient to induce hSpt4 binding.


Subject(s)
Nuclear Proteins/chemistry , Repressor Proteins/chemistry , Transcription Factors/chemistry , Transcriptional Elongation Factors/chemistry , Crystallography, X-Ray , Glutamine , Humans , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Multimerization , Structural Homology, Protein
20.
J Mol Biol ; 368(4): 1122-31, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17395199

ABSTRACT

We have cloned, expressed, isolated and characterized a hexameric tyrosine-coordinated heme protein (HTHP) from the marine bacterium Silicibacter pomeroyi. HTHP shows peroxidase and catalase activity and has a high thermal stability. As-isolated HTHP has absorption maxima at 407, 495, 504, 532 and 622 nm wavelength. Upon reduction maxima at 430, 564 and 596 nm wavelength are discernible. The crystal structure of HTHP reveals a hexameric, ring-like arrangement of six monomers. Each monomer binds a solvent accessible heme group, which is stabilized by the interaction of three neighboring monomers. The pocket around the heme distal side is positively charged due to three conserved arginine residues in direct vicinity. The heme iron is penta-coordinated with a tyrosine residue as proximal ligand. The coordinating hydroxyl-group of the tyrosine ligand interacts with the guanidinium group of a nearby arginine residue, an arrangement closely resembling the catalytic dyad found in monofunctional heme-containing catalases and coral allene oxide synthases, which are b-type cytochromes with tyrosine coordination trans to an empty coordination site. Despite the similarity in heme coordination HTHP is functionally and structurally unrelated to catalases and other heme-containing proteins. Its hexameric arrangement, solvent accessible heme binding pocket and heme coordination by tyrosine render HTHP a unique protein with unusual properties. A database search against complete and incomplete genomes shows that the 76 amino acid residues sequence of HTHP is unrelated to characterized proteins, but is homologous to orfs found in a phylogenetically diverse set of bacteria with sequence identities of 30-76%. We therefore propose that HTHP is the prototype of a new class of heme proteins.


Subject(s)
Bacterial Proteins/chemistry , Hemeproteins/chemistry , Models, Molecular , Rhodobacteraceae/enzymology , Tyrosine/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Binding Sites , Catalase/chemistry , Enzyme Stability , Heme-Binding Proteins , Hemeproteins/genetics , Hemeproteins/isolation & purification , Molecular Sequence Data , Peroxidase/chemistry , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...