Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
ACS Catal ; 9(4): 2962-2968, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30984471

ABSTRACT

Biosynthesis of 6-deoxy sugars, including l-fucose, involves a mechanistically complex, enzymatic 4,6-dehydration of hexose nucleotide precursors as the first committed step. Here, we determined pre- and postcatalytic complex structures of the human GDP-mannose 4,6-dehydratase at atomic resolution. These structures together with results of molecular dynamics simulation and biochemical characterization of wildtype and mutant enzymes reveal elusive mechanistic details of water elimination from GDP-mannose C5″ and C6″, coupled to NADP-mediated hydride transfer from C4″ to C6″. We show that concerted acid-base catalysis from only two active-site groups, Tyr179 and Glu157, promotes a syn 1,4-elimination from an enol (not an enolate) intermediate. We also show that the overall multistep catalytic reaction involves the fewest position changes of enzyme and substrate groups and that it proceeds under conserved exploitation of the basic (minimal) catalytic machinery of short-chain dehydrogenase/reductases.

2.
Bone ; 81: 478-486, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318908

ABSTRACT

Farnesyl pyrophosphate synthase (FPPS) is the major molecular target of nitrogen-containing bisphosphonates (N-BPs), used clinically as bone resorption inhibitors. We investigated the role of threonine 201 (Thr201) and tyrosine 204 (Tyr204) residues in substrate binding, catalysis and inhibition by N-BPs, employing kinetic and crystallographic studies of mutated FPPS proteins. Mutants of Thr201 illustrated the importance of the methyl group in aiding the formation of the Isopentenyl pyrophosphate (IPP) binding site, while Tyr204 mutations revealed the unknown role of this residue in both catalysis and IPP binding. The interaction between Thr201 and the side chain nitrogen of N-BP was shown to be important for tight binding inhibition by zoledronate (ZOL) and risedronate (RIS), although RIS was also still capable of interacting with the main-chain carbonyl of Lys200. The interaction of RIS with the phenyl ring of Tyr204 proved essential for the maintenance of the isomerized enzyme-inhibitor complex. Studies with conformationally restricted analogues of RIS reaffirmed the importance of Thr201 in the formation of hydrogen bonds with N-BPs. In conclusion we have identified new features of FPPS inhibition by N-BPs and revealed unknown roles of the active site residues in catalysis and substrate binding.


Subject(s)
Diphosphonates/chemistry , Geranyltranstransferase/antagonists & inhibitors , Mutation , Nitrogen/chemistry , Bone Density Conservation Agents/therapeutic use , Catalysis , Catalytic Domain , Crystallization , Diphosphonates/therapeutic use , Drug Evaluation, Preclinical , Geranyltranstransferase/chemistry , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Imidazoles/therapeutic use , Inhibitory Concentration 50 , Molecular Conformation , Oligonucleotides/chemistry , Protein Binding , Recombinant Proteins/chemistry , Threonine/chemistry , Tyrosine/chemistry , Zoledronic Acid
3.
Chem Biol Interact ; 234: 114-25, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25526675

ABSTRACT

Short-chain dehydrogenases/reductases (SDRs) constitute a large, functionally diverse branch of enzymes within the class of NAD(P)(H) dependent oxidoreductases. In humans, over 80 genes have been identified with distinct metabolic roles in carbohydrate, amino acid, lipid, retinoid and steroid hormone metabolism, frequently associated with inherited genetic defects. Besides metabolic functions, a subset of atypical SDR proteins appears to play critical roles in adapting to redox status or RNA processing, and thereby controlling metabolic pathways. Here we present an update on the human SDR superfamily and a ligand identification strategy using differential scanning fluorimetry (DSF) with a focused library of oxidoreductase and metabolic ligands to identify substrate classes and inhibitor chemotypes. This method is applicable to investigate structure-activity relationships of oxidoreductases and ultimately to better understand their physiological roles.


Subject(s)
Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Humans , Ligands , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phylogeny , Structure-Activity Relationship , Substrate Specificity/genetics
4.
Nature ; 510(7505): 422-426, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24814345

ABSTRACT

2-Oxoglutarate (2OG)-dependent oxygenases have important roles in the regulation of gene expression via demethylation of N-methylated chromatin components and in the hydroxylation of transcription factors and splicing factor proteins. Recently, 2OG-dependent oxygenases that catalyse hydroxylation of transfer RNA and ribosomal proteins have been shown to be important in translation relating to cellular growth, TH17-cell differentiation and translational accuracy. The finding that ribosomal oxygenases (ROXs) occur in organisms ranging from prokaryotes to humans raises questions as to their structural and evolutionary relationships. In Escherichia coli, YcfD catalyses arginine hydroxylation in the ribosomal protein L16; in humans, MYC-induced nuclear antigen (MINA53; also known as MINA) and nucleolar protein 66 (NO66) catalyse histidine hydroxylation in the ribosomal proteins RPL27A and RPL8, respectively. The functional assignments of ROXs open therapeutic possibilities via either ROX inhibition or targeting of differentially modified ribosomes. Despite differences in the residue and protein selectivities of prokaryotic and eukaryotic ROXs, comparison of the crystal structures of E. coli YcfD and Rhodothermus marinus YcfD with those of human MINA53 and NO66 reveals highly conserved folds and novel dimerization modes defining a new structural subfamily of 2OG-dependent oxygenases. ROX structures with and without their substrates support their functional assignments as hydroxylases but not demethylases, and reveal how the subfamily has evolved to catalyse the hydroxylation of different residue side chains of ribosomal proteins. Comparison of ROX crystal structures with those of other JmjC-domain-containing hydroxylases, including the hypoxia-inducible factor asparaginyl hydroxylase FIH and histone N(ε)-methyl lysine demethylases, identifies branch points in 2OG-dependent oxygenase evolution and distinguishes between JmjC-containing hydroxylases and demethylases catalysing modifications of translational and transcriptional machinery. The structures reveal that new protein hydroxylation activities can evolve by changing the coordination position from which the iron-bound substrate-oxidizing species reacts. This coordination flexibility has probably contributed to the evolution of the wide range of reactions catalysed by oxygenases.


Subject(s)
Eukaryota/enzymology , Models, Molecular , Oxygenases/chemistry , Prokaryotic Cells/enzymology , Ribosomes/enzymology , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Eukaryota/classification , Humans , Oxygenases/metabolism , Phylogeny , Prokaryotic Cells/classification , Protein Folding , Protein Structure, Tertiary , Sequence Alignment
5.
J Biol Chem ; 287(37): 31349-58, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22810237

ABSTRACT

UDP-xylose synthase (UXS) catalyzes decarboxylation of UDP-D-glucuronic acid to UDP-xylose. In mammals, UDP-xylose serves to initiate glycosaminoglycan synthesis on the protein core of extracellular matrix proteoglycans. Lack of UXS activity leads to a defective extracellular matrix, resulting in strong interference with cell signaling pathways. We present comprehensive structural and mechanistic characterization of the human form of UXS. The 1.26-Å crystal structure of the enzyme bound with NAD(+) and UDP reveals a homodimeric short-chain dehydrogenase/reductase (SDR), belonging to the NDP-sugar epimerases/dehydratases subclass. We show that enzymatic reaction proceeds in three chemical steps via UDP-4-keto-D-glucuronic acid and UDP-4-keto-pentose intermediates. Molecular dynamics simulations reveal that the D-glucuronyl ring accommodated by UXS features a marked (4)C(1) chair to B(O,3) boat distortion that facilitates catalysis in two different ways. It promotes oxidation at C(4) (step 1) by aligning the enzymatic base Tyr(147) with the reactive substrate hydroxyl and it brings the carboxylate group at C(5) into an almost fully axial position, ideal for decarboxylation of UDP-4-keto-D-glucuronic acid in the second chemical step. The protonated side chain of Tyr(147) stabilizes the enolate of decarboxylated C(4) keto species ((2)H(1) half-chair) that is then protonated from the Si face at C(5), involving water coordinated by Glu(120). Arg(277), which is positioned by a salt-link interaction with Glu(120), closes up the catalytic site and prevents release of the UDP-4-keto-pentose and NADH intermediates. Hydrogenation of the C(4) keto group by NADH, assisted by Tyr(147) as catalytic proton donor, yields UDP-xylose adopting the relaxed (4)C(1) chair conformation (step 3).


Subject(s)
Carboxy-Lyases/chemistry , Uridine Diphosphate Glucuronic Acid/chemistry , Carboxy-Lyases/metabolism , Catalysis , Crystallography, X-Ray , Humans , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship
6.
BMC Struct Biol ; 12: 14, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22720794

ABSTRACT

BACKGROUND: Aspartyl aminopeptidase (DNPEP), with specificity towards an acidic amino acid at the N-terminus, is the only mammalian member among the poorly understood M18 peptidases. DNPEP has implicated roles in protein and peptide metabolism, as well as the renin-angiotensin system in blood pressure regulation. Despite previous enzyme and substrate characterization, structural details of DNPEP regarding ligand recognition and catalytic mechanism remain to be delineated. RESULTS: The crystal structure of human DNPEP complexed with zinc and a substrate analogue aspartate-ß-hydroxamate reveals a dodecameric machinery built by domain-swapped dimers, in agreement with electron microscopy data. A structural comparison with bacterial homologues identifies unifying catalytic features among the poorly understood M18 enzymes. The bound ligands in the active site also reveal the coordination mode of the binuclear zinc centre and a substrate specificity pocket for acidic amino acids. CONCLUSIONS: The DNPEP structure provides a molecular framework to understand its catalysis that is mediated by active site loop swapping, a mechanism likely adopted in other M18 and M42 metallopeptidases that form dodecameric complexes as a self-compartmentalization strategy. Small differences in the substrate binding pocket such as shape and positive charges, the latter conferred by a basic lysine residue, further provide the key to distinguishing substrate preference. Together, the structural knowledge will aid in the development of enzyme-/family-specific aminopeptidase inhibitors.


Subject(s)
Biocatalysis , Glutamyl Aminopeptidase/chemistry , Multigene Family , Amino Acid Sequence , Bacteria/enzymology , Catalytic Domain , Crystallography, X-Ray , Humans , Metals , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary , Static Electricity , Substrate Specificity
7.
J Biol Chem ; 287(3): 2119-29, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22123821

ABSTRACT

Biosynthesis of UDP-glucuronic acid by UDP-glucose 6-dehydrogenase (UGDH) occurs through the four-electron oxidation of the UDP-glucose C6 primary alcohol in two NAD(+)-dependent steps. The catalytic reaction of UGDH is thought to involve a Cys nucleophile that promotes formation of a thiohemiacetal enzyme intermediate in the course of the first oxidation step. The thiohemiacetal undergoes further oxidation into a thioester, and hydrolysis of the thioester completes the catalytic cycle. Herein we present crystallographic and kinetic evidence for the human form of UGDH that clarifies participation of covalent catalysis in the enzymatic mechanism. Substitution of the putative catalytic base for water attack on the thioester (Glu(161)) by an incompetent analog (Gln(161)) gave a UGDH variant (E161Q) in which the hydrolysis step had become completely rate-limiting so that a thioester enzyme intermediate accumulated at steady state. By crystallizing E161Q in the presence of 5 mm UDP-glucose and 2 mm NAD(+), we succeeded in trapping a thiohemiacetal enzyme intermediate and determined its structure at 2.3 Å resolution. Cys(276) was covalently modified in the structure, establishing its role as catalytic nucleophile of the reaction. The thiohemiacetal reactive C6 was in a position suitable to become further oxidized by hydride transfer to NAD(+). The proposed catalytic mechanism of human UGDH involves Lys(220) as general base for UDP-glucose alcohol oxidation and for oxyanion stabilization during formation and breakdown of the thiohemiacetal and thioester enzyme intermediates. Water coordinated to Asp(280) deprotonates Cys(276) to function as an aldehyde trap and also provides oxyanion stabilization. Glu(161) is the Brønsted base catalytically promoting the thioester hydrolysis.


Subject(s)
NAD/chemistry , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose/chemistry , Amino Acid Substitution , Catalysis , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Mutation, Missense , NAD/genetics , NAD/metabolism , Oxidation-Reduction , Structure-Activity Relationship , Uridine Diphosphate Glucose/genetics , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism
8.
Arch Biochem Biophys ; 513(1): 19-26, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21689631

ABSTRACT

Fungal methionine synthase, Met6p, transfers a methyl group from 5-methyl-tetrahydrofolate to homocysteine to generate methionine. The enzyme is essential to fungal growth and is a potential anti-fungal drug design target. We have characterized the enzyme from the pathogen Candida albicans but were unable to crystallize it in native form. We converted Lys103, Lys104, and Glu107 all to Tyr (Met6pY), Thr (Met6pT) and Ala (Met6pA). All variants showed wild-type kinetic activity and formed useful crystals, each with unique crystal packing. In each case the mutated residues participated in beneficial crystal contacts. We have solved the three structures at 2.0-2.8Å resolution and analyzed crystal packing, active-site residues, and similarity to other known methionine synthase structures. C. albicans Met6p has a two domain structure with each of the domains having a (ßα)(8)-barrel fold. The barrels are arranged face-to-face and the active site is located in a cleft between the two domains. Met6p utilizes a zinc ion for catalysis that is bound in the C-terminal domain and ligated by four conserved residues: His657, Cys659, Glu679 and Cys739.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , Candida albicans/enzymology , Fungal Proteins/chemistry , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Amino Acid Substitution , Candida albicans/genetics , Catalysis , Crystallography, X-Ray , Fungal Proteins/genetics , Mutation, Missense , Protein Structure, Tertiary , Structure-Activity Relationship
9.
Biochem Biophys Res Commun ; 408(4): 553-8, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21530488

ABSTRACT

Phytanoyl-CoA hydroxylase (PAHX) catalyzes an important step in the metabolism of the fatty acid side chain of chlorophyll. PHYHD1 exists in three isoforms and is the closest human homologue of PAHX. We show that like PAHX, the PHYHD1A but likely not the PHYHD1B/C isoforms, is a functional Fe(II) and 2-oxoglutarate (2OG) dependent oxygenase. Crystallographic and biochemical analyses reveal that PHYHD1A has the double-stranded ß-helix fold and Fe(II) and cosubstrate binding residues characteristic of the 2-oxoglutarate dependent oxygenases and catalyzes the conversion of 2-oxoglutarate to succinate and CO(2) in an iron-dependent manner. However, PHYHD1A did not couple 2OG turnover to the hydroxylation of acyl-coenzyme A derivatives that are substrates for PAHX, implying that it is not directly involved in phytanoyl coenzyme-A metabolism.


Subject(s)
Mixed Function Oxygenases/chemistry , Oxygenases/chemistry , Crystallography, X-Ray , Humans , Iron/chemistry , Mixed Function Oxygenases/genetics , Oxygenases/genetics , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Secondary
10.
J Biol Chem ; 286(27): 23877-87, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21502315

ABSTRACT

Elevated production of the matrix glycosaminoglycan hyaluronan is strongly implicated in epithelial tumor progression. Inhibition of synthesis of the hyaluronan precursor UDP-glucuronic acid (UDP-GlcUA) therefore presents an emerging target for cancer therapy. Human UDP-glucose 6-dehydrogenase (hUGDH) catalyzes, in two NAD(+)-dependent steps without release of intermediate aldehyde, the biosynthetic oxidation of UDP-glucose (UDP-Glc) to UDP-GlcUA. Here, we present a structural characterization of the hUGDH reaction coordinate using crystal structures of the apoenzyme and ternary complexes of the enzyme bound with UDP-Glc/NADH and UDP-GlcUA/NAD(+). The quaternary structure of hUGDH is a disc-shaped trimer of homodimers whose subunits consist of two discrete α/ß domains with the active site located in the interdomain cleft. Ternary complex formation is accompanied by rigid-body and restrained movement of the N-terminal NAD(+) binding domain, sequestering substrate and coenzyme in their reactive positions through interdomain closure. By alternating between conformations in and out of the active site during domain motion, Tyr(14), Glu(161), and Glu(165) participate in control of coenzyme binding and release during 2-fold oxidation. The proposed mechanism of hUGDH involves formation and breakdown of thiohemiacetal and thioester intermediates whereby Cys(276) functions as the catalytic nucleophile. Stopped-flow kinetic data capture the essential deprotonation of Cys(276) in the course of the first oxidation step, allowing the thiolate side chain to act as a trap of the incipient aldehyde. Because thiohemiacetal intermediate accumulates at steady state under physiological reaction conditions, hUGDH inhibition might best explore ligand binding to the NAD(+) binding domain.


Subject(s)
Coenzymes/chemistry , NAD/chemistry , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Catalysis , Coenzymes/metabolism , Humans , NAD/metabolism , Oxidation-Reduction , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship , Uridine Diphosphate Glucose Dehydrogenase/metabolism
11.
Proc Natl Acad Sci U S A ; 108(19): 7745-50, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21508329

ABSTRACT

Endoplasmatic reticulum aminopeptidase 1 (ERAP1) is a multifunctional enzyme involved in trimming of peptides to an optimal length for presentation by major histocompatibility complex (MHC) class I molecules. Polymorphisms in ERAP1 have been associated with chronic inflammatory diseases, including ankylosing spondylitis (AS) and psoriasis, and subsequent in vitro enzyme studies suggest distinct catalytic properties of ERAP1 variants. To understand structure-activity relationships of this enzyme we determined crystal structures in open and closed states of human ERAP1, which provide the first snapshots along a catalytic path. ERAP1 is a zinc-metallopeptidase with typical H-E-X-X-H-(X)(18)-E zinc binding and G-A-M-E-N motifs characteristic for members of the gluzincin protease family. The structures reveal extensive domain movements, including an active site closure as well as three different open conformations, thus providing insights into the catalytic cycle. A K(528)R mutant strongly associated with AS in GWAS studies shows significantly altered peptide processing characteristics, which are possibly related to impaired interdomain interactions.


Subject(s)
Aminopeptidases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Aminopeptidases/genetics , Aminopeptidases/metabolism , Antigen Presentation , Catalytic Domain/genetics , Crystallography, X-Ray , HLA-B27 Antigen/metabolism , Humans , Minor Histocompatibility Antigens , Models, Molecular , Mutagenesis, Site-Directed , Polymorphism, Single Nucleotide , Protein Conformation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spondylitis, Ankylosing/enzymology , Spondylitis, Ankylosing/genetics
12.
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
13.
Biochem J ; 433(2): 303-11, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21029046

ABSTRACT

Human GLRX5 (glutaredoxin 5) is an evolutionarily conserved thiol-disulfide oxidoreductase that has a direct role in the maintenance of normal cytosolic and mitochondrial iron homoeostasis, and its expression affects haem biosynthesis and erythropoiesis. We have crystallized the human GLRX5 bound to two [2Fe-2S] clusters and four GSH molecules. The crystal structure revealed a tetrameric organization with the [2Fe-2S] clusters buried in the interior and shielded from the solvent by the conserved ß1-α2 loop, Phe69 and the GSH molecules. Each [2Fe-2S] cluster is ligated by the N-terminal activesite cysteine (Cys67) thiols contributed by two protomers and two cysteine thiols from two GSH. The two subunits co-ordinating the cluster are in a more extended conformation compared with iron-sulfur-bound human GLRX2, and the intersubunit interactions are more extensive and involve conserved residues among monothiol GLRXs. Gel-filtration chromatography and analytical ultracentrifugation support a tetrameric organization of holo-GLRX5, whereas the apoprotein is monomeric. MS analyses revealed glutathionylation of the cysteine residues in the absence of the [2Fe-2S] cluster, which would protect them from further oxidation and possibly facilitate cluster transfer/acceptance. Apo-GLRX5 reduced glutathione mixed disulfides with a rate 100 times lower than did GLRX2 and was active as a glutathione-dependent electron donor for mammalian ribonucleotide reductase.


Subject(s)
Glutaredoxins/chemistry , Iron-Sulfur Proteins/chemistry , Protein Multimerization , Crystallography, X-Ray , Disulfides/chemistry , Disulfides/metabolism , Glutaredoxins/metabolism , Humans , Iron-Sulfur Proteins/metabolism , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Structure, Quaternary
14.
Biochem Soc Trans ; 38(5): 1378-85, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20863317

ABSTRACT

Biosynthesis of the glycosaminoglycan precursor UDP-α-D-glucuronic acid occurs through a 2-fold oxidation of UDP-α-D-glucose that is catalysed by UGDH (UDP-α-D-glucose 6-dehydrogenase). Structure-function relationships for UGDH and proposals for the enzymatic reaction mechanism are reviewed in the present paper, and structure-based sequence comparison is used for subclassification of UGDH family members. The eukaryotic group of enzymes (UGDH-II) utilize an extended C-terminal domain for the formation of complex homohexameric assemblies. The comparably simpler oligomerization behaviour of the prokaryotic group of enzymes (UGDH-I), in which dimeric forms prevail, is traced back to the lack of relevant intersubunit contacts and trimmings within the C-terminal region. The active site of UGDH contains a highly conserved cysteine residue, which plays a key role in covalent catalysis. Elevated glycosaminoglycan formation is implicated in a variety of human diseases, including the progression of tumours. The inhibition of synthesis of UDP-α-D-glucuronic acid using UGDH antagonists might therefore be a useful strategy for therapy.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Animals , Enzyme Inhibitors/therapeutic use , Humans , Hyaluronic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucuronic Acid/metabolism
15.
J Mol Biol ; 2010 May 31.
Article in English | MEDLINE | ID: mdl-20685276

ABSTRACT

Lysyl and prolyl hydroxylations are well-known post-translational modifications to animal and plant proteins with extracellular roles. More recent work has indicated that the hydroxylation of intracellular animal proteins may be common. JMJD6 catalyses the iron- and 2-oxoglutarate-dependent hydroxylation of lysyl residues in arginine-serine-rich domains of RNA-splicing-related proteins. We report crystallographic studies on the catalytic domain of JMJD6 in complex with Ni(II) substituting for Fe(II). Together with mutational studies, the structural data suggest how JMJD6 binds its lysyl residues such that it can catalyse C-5 hydroxylation rather than N(varepsilon)-demethylation, as for analogous enzymes.

16.
J Mol Biol ; 401(2): 211-22, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20684070

ABSTRACT

Lysyl and prolyl hydroxylations are well-known post-translational modifications to animal and plant proteins with extracellular roles. More recent work has indicated that the hydroxylation of intracellular animal proteins may be common. JMJD6 catalyses the iron- and 2-oxoglutarate-dependent hydroxylation of lysyl residues in arginine-serine-rich domains of RNA splicing-related proteins. We report crystallographic studies on the catalytic domain of JMJD6 in complex with Ni(II) substituting for Fe(II). Together with mutational studies, the structural data suggest how JMJD6 binds its lysyl residues such that it can catalyse C-5 hydroxylation rather than Nepsilon-demethylation, as for analogous enzymes.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/chemistry , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/chemistry , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Catalytic Domain , Crystallography, X-Ray , DNA Primers/genetics , Humans , In Vitro Techniques , Iron/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Ketoglutaric Acids/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nickel/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity
17.
J Biol Chem ; 285(24): 18452-63, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20207735

ABSTRACT

Mammalian ALDH7A1 is homologous to plant ALDH7B1, an enzyme that protects against various forms of stress, such as salinity, dehydration, and osmotic stress. It is known that mutations in the human ALDH7A1 gene cause pyridoxine-dependent and folic acid-responsive seizures. Herein, we show for the first time that human ALDH7A1 protects against hyperosmotic stress by generating osmolytes and metabolizing toxic aldehydes. Human ALDH7A1 expression in Chinese hamster ovary cells attenuated osmotic stress-induced apoptosis caused by increased extracellular concentrations of sucrose or sodium chloride. Purified recombinant ALDH7A1 efficiently metabolized a number of aldehyde substrates, including the osmolyte precursor, betaine aldehyde, lipid peroxidation-derived aldehydes, and the intermediate lysine degradation product, alpha-aminoadipic semialdehyde. The crystal structure for ALDH7A1 supports the enzyme's substrate specificities. Tissue distribution studies in mice showed the highest expression of ALDH7A1 protein in liver, kidney, and brain, followed by pancreas and testes. ALDH7A1 protein was found in the cytosol, nucleus, and mitochondria, making it unique among the aldehyde dehydrogenase enzymes. Analysis of human and mouse cDNA sequences revealed mitochondrial and cytosolic transcripts that are differentially expressed in a tissue-specific manner in mice. In conclusion, ALDH7A1 is a novel aldehyde dehydrogenase expressed in multiple subcellular compartments that protects against hyperosmotic stress by generating osmolytes and metabolizing toxic aldehydes.


Subject(s)
Aldehyde Dehydrogenase/physiology , Osmosis , Proteins/physiology , Aldehyde Dehydrogenase/metabolism , Aldehydes/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray/methods , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Mice , Proteins/metabolism , Tissue Distribution
18.
FEBS Lett ; 584(4): 825-30, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20067792

ABSTRACT

Crystallographic analysis of the catalytic domain of PHD finger protein 8 (PHF8), an N(epsilon)-methyl lysine histone demethylase associated with mental retardation and cleft lip/palate, reveals a double-stranded beta-helix fold with conserved Fe(II) and cosubstrate binding sites typical of the 2-oxoglutarate dependent oxygenases. The PHF8 active site is highly conserved with those of the FBXL10/11demethylases, which are also selective for the di-/mono-methylated lysine states, but differs from that of the JMJD2 demethylases which are selective for tri-/di-methylated states. The results rationalize the lack of activity for the clinically observed F279S PHF8 variant and they will help to identify inhibitors selective for specific N(epsilon)-methyl lysine demethylase subfamilies.


Subject(s)
Protein Structure, Tertiary , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites/genetics , Crystallography, X-Ray , Histone Demethylases , Humans , Iron/chemistry , Iron/metabolism , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Ketoglutaric Acids/chemistry , Ketoglutaric Acids/metabolism , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Transcription Factors/genetics
19.
EMBO Mol Med ; 2(2): 51-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20077426

ABSTRACT

Deficiency of the mitochondrial enzyme 2-methyl-3-hydroxybutyryl-CoA dehydrogenase involved in isoleucine metabolism causes an organic aciduria with atypical neurodegenerative course. The disease-causing gene is HSD17B10 and encodes 17beta-hydroxysteroid dehydrogenase type 10 (HSD10), a protein also implicated in the pathogenesis of Alzheimer's disease. Here we show that clinical symptoms in patients are not correlated with residual enzymatic activity of mutated HSD10. Loss-of-function and rescue experiments in Xenopus embryos and cells derived from conditional Hsd17b10(-/-) mice demonstrate that a property of HSD10 independent of its enzymatic activity is essential for structural and functional integrity of mitochondria. Impairment of this function in neural cells causes apoptotic cell death whilst the enzymatic activity of HSD10 is not required for cell survival. This finding indicates that the symptoms in patients with mutations in the HSD17B10 gene are unrelated to accumulation of toxic metabolites in the isoleucine pathway and, rather, related to defects in general mitochondrial function. Therefore alternative therapeutic approaches to an isoleucine-restricted diet are required.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/deficiency , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Hydroxysteroid Dehydrogenases/deficiency , Hydroxysteroid Dehydrogenases/metabolism , Mitochondria/physiology , Animals , Apoptosis , Cell Survival , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Deletion , Genetic Complementation Test , Humans , Infant , Mice , Mice, Knockout , Mitochondria/ultrastructure , Models, Molecular , Neurons/physiology , Protein Structure, Tertiary , Xenopus
20.
Chem Biol ; 16(6): 667-75, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19549604

ABSTRACT

Animals employ two systems for the de novo biosynthesis of fatty acids: a megasynthase complex in the cytosol (type I) that produces mainly palmitate, and an ensemble of freestanding enzymes in the mitochondria (type II) that produces mainly octanoyl moieties. The acyltransferases responsible for initiation of fatty acid biosynthesis in the two compartments are distinguished by their different substrate specificities: the type I enzyme transfers both the acetyl primer and the malonyl chain extender, whereas the type II enzyme is responsible for translocation of only the malonyl substrate. Crystal structures for the type I and II enzymes, supported by in silico substrate docking studies and mutagenesis experiments that alter their respective specificities, reveal that, although the two enzymes adopt a similar overall fold, subtle differences at their catalytic centers account for their different specificities.


Subject(s)
Acyltransferases/chemistry , Fatty Acid Synthase, Type II/chemistry , Fatty Acid Synthase, Type I/chemistry , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Sequence , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Cytosol/enzymology , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type II/metabolism , Fatty Acids/biosynthesis , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...