Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Oncotarget ; 14: 843-857, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37769033

ABSTRACT

UDP-glucose-6-dehydrogenase (UGDH) is a cytosolic, hexameric enzyme that converts UDP-glucose to UDP-glucuronic acid (UDP-GlcUA), a key reaction in hormone and xenobiotic metabolism and in the production of extracellular matrix precursors. In this review, we classify UGDH as a molecular indicator of tumor progression in multiple cancer types, describe its involvement in key canonical cancer signaling pathways, and identify methods to inhibit UGDH, its substrates, and its downstream products. As such, we position UGDH as an enzyme to be exploited as a potential prognostication marker in oncology and a therapeutic target in cancer biology.


Subject(s)
Neoplasms , Uridine Diphosphate Glucose Dehydrogenase , Humans , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose , Neoplasms/genetics , Medical Oncology , Glucose , Biology , Glucose Dehydrogenases
2.
PLoS One ; 17(9): e0274420, 2022.
Article in English | MEDLINE | ID: mdl-36107941

ABSTRACT

UDP-glucose dehydrogenase (UGDH) generates essential precursors of hyaluronic acid (HA) synthesis, however mechanisms regulating its activity are unclear. We used enzyme histostaining and quantitative image analysis to test whether cytokines that stimulate HA synthesis upregulate UGDH activity. Fibroblast-like synoviocytes (FLS, from N = 6 human donors with knee pain) were cultured, freeze-thawed, and incubated for 1 hour with UDP-glucose, NAD+ and nitroblue tetrazolium (NBT) which allows UGDH to generate NADH, and NADH to reduce NBT to a blue stain. Compared to serum-free medium, FLS treated with PDGF showed 3-fold higher UGDH activity and 6-fold higher HA release, but IL-1beta/TGF-beta1 induced 27-fold higher HA release without enhancing UGDH activity. In selected proliferating cells, UGDH activity was lost in the cytosol, but preserved in the nucleus. Cell-free assays led us to discover that diaphorase, a cytosolic enzyme, or glutathione reductase, a nuclear enzyme, was necessary and sufficient for NADH to reduce NBT to a blue formazan dye in a 1-hour timeframe. Primary synovial fibroblasts and transformed A549 fibroblasts showed constitutive diaphorase/GR staining activity that varied according to supplied NADH levels, with relatively stronger UGDH and diaphorase activity in A549 cells. Unilateral knee injury in New Zealand White rabbits (N = 3) stimulated a coordinated increase in synovial membrane UGDH and diaphorase activity, but higher synovial fluid HA in only 2 out of 3 injured joints. UGDH activity (but not diaphorase) was abolished by N-ethyl maleimide, and inhibited by peroxide or UDP-xylose. Our results do not support the hypothesis that UGDH is a rate-liming enzyme for HA synthesis under catabolic inflammatory conditions that can oxidize and inactivate the UGDH active site cysteine. Our novel data suggest a model where UGDH activity is controlled by a redox switch, where intracellular peroxide inactivates, and high glutathione and diaphorase promote UGDH activity by maintaining the active site cysteine in a reduced state, and by recycling NAD+ from NADH.


Subject(s)
Synoviocytes , Animals , Cysteine/metabolism , Fibroblasts/metabolism , Formazans , Glucose/pharmacology , Glucose Dehydrogenases/metabolism , Glutathione/metabolism , Glutathione Reductase/metabolism , Humans , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Maleimides , NAD/metabolism , Nitroblue Tetrazolium , Oxidation-Reduction , Peroxides , Rabbits , Synoviocytes/metabolism , Transforming Growth Factor beta1/metabolism , Uridine Diphosphate/metabolism , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Xylose
3.
Biochemistry ; 60(9): 725-734, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33621065

ABSTRACT

Campylobacter jejuni is a pathogenic organism that can cause campylobacteriosis in children and adults. Most commonly, campylobacter infection is brought on by consumption of raw or undercooked poultry, unsanitary drinking water, or pet feces. Surrounding the C. jejuni bacterium is a coat of sugar molecules known as the capsular polysaccharide (CPS). The capsular polysaccharide can be very diverse among the different strains of C. jejuni, and this diversity is considered important for evading the host immune system. Modifications to the CPS of C. jejuni NCTC 11168 include O-methylation, phosphoramidylation, and amidation of glucuronate with either serinol or ethanolamine. The enzymes responsible for amidation of glucuronate are currently unknown. In this study, Cj1441, an enzyme expressed from the CPS biosynthetic gene cluster in C. jejuni NCTC 11168, was shown to catalyze the oxidation of UDP-α-d-glucose into UDP-α-d-glucuronic acid with NAD+ as the cofactor. No amide products were found in an attempt to determine whether the putative thioester intermediate formed during the oxidation of UDP-glucose by Cj1441 could be captured in the presence of added amines. The three-dimensional crystal structure of Cj1441 was determined in the presence of NAD+ and UDP-glucose bound in the active site of the enzyme (Protein Data Bank entry 7KWS). A more thorough bioinformatic analysis of the CPS gene cluster suggests that the amidation activity is localized to the t-terminal half of Cj1438, a bifunctional enzyme that is currently annotated as a sugar transferase.


Subject(s)
Bacterial Capsules/metabolism , Campylobacter jejuni/enzymology , Polysaccharides/biosynthesis , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation
4.
J Histochem Cytochem ; 69(1): 13-23, 2021 01.
Article in English | MEDLINE | ID: mdl-32749901

ABSTRACT

Regulation of proteoglycan and glycosaminoglycan synthesis is critical throughout development, and to maintain normal adult functions in wound healing and the immune system, among others. It has become increasingly clear that these processes are also under tight metabolic control and that availability of carbohydrate and amino acid metabolite precursors has a role in the control of proteoglycan and glycosaminoglycan turnover. The enzyme uridine diphosphate (UDP)-glucose dehydrogenase (UGDH) produces UDP-glucuronate, an essential precursor for new glycosaminoglycan synthesis that is tightly controlled at multiple levels. Here, we review the cellular mechanisms that regulate UGDH expression, discuss the structural features of the enzyme, and use the structures to provide a context for recent studies that link post-translational modifications and allosteric modulators of UGDH to its function in downstream pathways.


Subject(s)
Proteoglycans/metabolism , Sugars/metabolism , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Allosteric Regulation , Animals , Biosynthetic Pathways , Humans , Models, Molecular , Neoplasms/metabolism , Protein Processing, Post-Translational , Uridine Diphosphate Glucose Dehydrogenase/chemistry
5.
Nature ; 571(7763): 127-131, 2019 07.
Article in English | MEDLINE | ID: mdl-31243371

ABSTRACT

Cancer metastasis is the primary cause of morbidity and mortality, and accounts for up to 95% of cancer-related deaths1. Cancer cells often reprogram their metabolism to efficiently support cell proliferation and survival2,3. However, whether and how those metabolic alterations contribute to the migration of tumour cells remain largely unknown. UDP-glucose 6-dehydrogenase (UGDH) is a key enzyme in the uronic acid pathway, and converts UDP-glucose to UDP-glucuronic acid4. Here we show that, after activation of EGFR, UGDH is phosphorylated at tyrosine 473 in human lung cancer cells. Phosphorylated UGDH interacts with Hu antigen R (HuR) and converts UDP-glucose to UDP-glucuronic acid, which attenuates the UDP-glucose-mediated inhibition of the association of HuR with SNAI1 mRNA and therefore enhances the stability of SNAI1 mRNA. Increased production of SNAIL initiates the epithelial-mesenchymal transition, thus promoting the migration of tumour cells and lung cancer metastasis. In addition, phosphorylation of UGDH at tyrosine 473 correlates with metastatic recurrence and poor prognosis of patients with lung cancer. Our findings reveal a tumour-suppressive role of UDP-glucose in lung cancer metastasis and uncover a mechanism by which UGDH promotes tumour metastasis by increasing the stability of SNAI1 mRNA.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , RNA Stability , Snail Family Transcription Factors/genetics , Uridine Diphosphate Glucose/metabolism , Animals , Cell Line, Tumor , Cell Movement , ELAV-Like Protein 1/deficiency , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phosphotyrosine/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Snail Family Transcription Factors/biosynthesis , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism
6.
Biochemistry ; 57(50): 6848-6859, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30457329

ABSTRACT

Human UDP-glucose dehydrogenase (hUGDH) oxidizes UDP-glucose to UDP-glucuronic acid, an essential substrate in the phase II metabolism of drugs. The activity of hUGDH is regulated by the conformation of a buried allosteric switch (T131 loop/α6 helix). Substrate binding induces the allosteric switch to slowly isomerize from an inactive E* conformation to the active E state, which can be observed as enzyme hysteresis. When the feedback inhibitor UDP-xylose binds, the allosteric switch and surrounding residues in the protein core repack, converting the hexamer into an inactive, horseshoe-shaped complex (EΩ). This allosteric transition is facilitated by large cavities and declivities in the protein core that provide the space required to accommodate the alternate packing arrangements. Here, we have used the A104L substitution to fill a cavity in the E state and sterically prevent repacking of the core into the EΩ state. Steady state analysis shows that hUGDHA104L binds UDP-xylose with lower affinity and that the inhibition is no longer cooperative. This means that the allosteric transition to the high-UDP-xylose affinity EΩ state is blocked by the substitution. The crystal structures of hUGDHA104L show that the allosteric switch still adopts the E and E* states, albeit with a more rigid protein core. However, the progress curves of hUGDHA104L do not show hysteresis, which suggests that the E* and E states are now in rapid equilibrium. Our data suggest that hysteresis in native hUGDH originates from the conformational entropy of the E* state protein core.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Substitution , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Substrate Specificity , Uridine Diphosphate Glucose Dehydrogenase/genetics
7.
Nature ; 563(7732): 584-588, 2018 11.
Article in English | MEDLINE | ID: mdl-30420606

ABSTRACT

Protein structures are dynamic and can explore a large conformational landscape1,2. Only some of these structural substates are important for protein function (such as ligand binding, catalysis and regulation)3-5. How evolution shapes the structural ensemble to optimize a specific function is poorly understood3,4. One of the constraints on the evolution of proteins is the stability of the folded 'native' state. Despite this, 44% of the human proteome contains intrinsically disordered peptide segments greater than 30 residues in length6, the majority of which have no known function7-9. Here we show that the entropic force produced by an intrinsically disordered carboxy terminus (ID-tail) shifts the conformational ensemble of human UDP-α-D-glucose-6-dehydrogenase (UGDH) towards a substate with a high affinity for an allosteric inhibitor. The function of the ID-tail does not depend on its sequence or chemical composition. Instead, the affinity enhancement can be accurately predicted based on the length of the intrinsically disordered segment, and is consistent with the entropic force generated by an unstructured peptide attached to the protein surface10-13. Our data show that the unfolded state of the ID-tail rectifies the dynamics and structure of UGDH to favour inhibitor binding. Because this entropic rectifier does not have any sequence or structural constraints, it is an easily acquired adaptation. This model implies that evolution selects for disordered segments to tune the energy landscape of proteins, which may explain the persistence of intrinsic disorder in the proteome.


Subject(s)
Entropy , Evolution, Molecular , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Allosteric Regulation/drug effects , Amino Acid Sequence , Humans , Intrinsically Disordered Proteins/antagonists & inhibitors , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Protein Folding , Protein Unfolding , Proteome/chemistry , Proteome/metabolism , Substrate Specificity , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors
8.
J Biol Chem ; 293(40): 15725-15732, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30089654

ABSTRACT

Folate derivatives are important cofactors for enzymes in several metabolic processes. Folate-related inhibition and resistance mechanisms in bacteria are potential targets for antimicrobial therapies and therefore a significant focus of current research. Here, we report that the activity of Escherichia coli poly-γ-glutamyl tetrahydrofolate/dihydrofolate synthase (FolC) is regulated by glutamate/glutamine-sensing uridylyltransferase (GlnD), THF-dependent tRNA modification enzyme (MnmE), and UDP-glucose dehydrogenase (Ugd) as shown by direct in vitro protein-protein interactions. Using kinetics analyses, we observed that GlnD, Ugd, and MnmE activate FolC many-fold by decreasing the Khalf of FolC for its substrate l-glutamate. Moreover, FolC inhibited the GTPase activity of MnmE at low GTP concentrations. The growth phenotypes associated with these proteins are discussed. These results, obtained using direct in vitro enzyme assays, reveal unanticipated networks of allosteric regulatory interactions in the folate pathway in E. coli and indicate regulation of polyglutamylated tetrahydrofolate biosynthesis by the availability of nitrogen sources, signaled by the glutamine-sensing GlnD protein.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/genetics , GTP Phosphohydrolases/chemistry , Gene Expression Regulation, Bacterial , Multienzyme Complexes/chemistry , Nucleotidyltransferases/chemistry , Peptide Synthases/chemistry , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Allosteric Regulation , Binding Sites , Enzyme Assays , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Folic Acid/biosynthesis , Folic Acid/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Kinetics , Molecular Docking Simulation , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pteroylpolyglutamic Acids/biosynthesis , Pteroylpolyglutamic Acids/chemistry , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Substrate Specificity , Thermodynamics , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism
9.
Biochemistry ; 56(1): 202-211, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27966912

ABSTRACT

Human UDP-glucose dehydrogenase (hUGDH) is regulated by an atypical allosteric mechanism in which the feedback inhibitor UDP-xylose (UDP-Xyl) competes with the substrate for the active site. Binding of UDP-Xyl triggers the T131-loop/α6 allosteric switch, which converts the hexameric structure of hUGDH into an inactive, horseshoe-shaped complex (EΩ). This allosteric transition buries residue A136 in the protein core to produce a subunit interface that favors the EΩ structure. Here we use a methionine substitution to prevent the burial of A136 and trap the T131-loop/α6 switch in the active conformation. We show that hUGDHA136M does not exhibit substrate cooperativity, which is strong evidence that the methionine substitution prevents the formation of the low-UDP-Glc-affinity EΩ state. In addition, the inhibitor affinity of hUGDHA136M is reduced 14-fold, which most likely represents the Ki for competitive inhibition in the absence of the allosteric transition to the higher-affinity EΩ state. hUGDH also displays a lag in progress curves, which is caused by a slow, substrate-induced isomerization that activates the enzyme. Stopped-flow analysis shows that hUGDHA136M does not exhibit hysteresis, which suggests that the T131-loop/α6 switch is the source of the slow isomerization. This interpretation is supported by the 2.05 Å resolution crystal structure of hUGDHA136M, which shows that the A136M substitution has stabilized the active conformation of the T131-loop/α6 allosteric switch. This work shows that the T131-loop/α6 allosteric switch couples allostery and hysteresis in hUGDH.


Subject(s)
Allosteric Regulation , Catalytic Domain , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate Xylose/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Binding, Competitive , Biocatalysis , Crystallization , Crystallography, X-Ray , Humans , Kinetics , Methionine/chemistry , Methionine/genetics , Methionine/metabolism , Models, Molecular , Mutation, Missense , Protein Conformation , Protein Multimerization , Substrate Specificity , Time Factors , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics
10.
Biochemistry ; 55(22): 3157-64, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27198584

ABSTRACT

The enzyme UDP-glucose dehydrogenase (UGDH) catalyzes the reaction of UDP-glucose to UDP-glucuronate through two successive NAD(+)-dependent oxidation steps. Human UGDH apoprotein is purified as a mixture of dimeric and hexameric species. Addition of substrate and cofactor stabilizes the oligomeric state to primarily the hexameric form. To determine if the dynamic conformations of hUGDH are required for catalytic activity, we used site-specific unnatural amino acid incorporation to facilitate cross-linking of monomeric subunits into predominantly obligate oligomeric species. Optimal cross-linking was achieved by encoding p-benzoyl-l-phenylalanine at position 458, normally a glutamine located within the dimer-dimer interface, and exposing the enzyme to long wavelength ultraviolet (UV) radiation in the presence of substrate and cofactor. Hexameric complexes were purified by gel filtration chromatography and found to contain significant fractions of dimer and trimer (approximately 50%) along with another 10% higher-molecular mass species. The activity of the cross-linked enzyme was reduced by almost 60% relative to that of the un-cross-linked UGDH mutant, and UV exposure had no effect on the activity of the wild-type enzyme. These results support a model for catalysis in which the ability to dissociate the dimer-dimer interface is as important for maximal enzyme function as has been previously shown for the formation of the hexamer.


Subject(s)
Amino Acids/chemistry , Cross-Linking Reagents , Light , Protein Multimerization/radiation effects , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Amino Acids/radiation effects , Catalysis , Humans , Kinetics , Models, Molecular , Oxidation-Reduction , Photochemical Processes , Protein Conformation , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose Dehydrogenase/metabolism
11.
Enzyme Microb Technol ; 85: 64-70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26920483

ABSTRACT

Hyaluronic acid (HA), a vital acid mucopolysaccharide, has immense applied value in foodstuffs, medicaments, and cosmetics among others. UDP-glucose dehydrogenase (UGDH, EC 1.1.1.22) is an essential enzyme for HA synthesis. In this study, a UGDH (PmuHasB, 45.9 kDa) from Pasteurella multocida CVCC 408 was expressed in Escherichia coli BL21 (DE3). It was purified by two chromatographic columns with a specific activity of 6.58 IU/mg. The optimum pH and temperature were determined to be 10.0 and 37°C, respectively. The activity was stable across the pH range 6-10, and had a half-life of about 3 h at 45°C. The estimated apparent Km values for UDP-glucose and NAD(+) were 0.11 and 0.069 mM, respectively. The results indicated that PmuHasB was an alkaline and mesophilic UGDH. PmuHasB and PmuHasA (HA synthase, HAS) were co-expressed in E. coli BW25113 to obtain a HA high-producing strain pBPAB/BW25113. It produced about 2.39 g/L HA in shake flask by using the method of whole-cell catalysis. Investigation of the different UGDHs on HA synthesis revealed that intracellular UGDH activity and HA total yield of pBPAB/BW25113 (0.15 IU/mg and 5.4 g/L) were higher than from pBPASB/BW25113 (0.013 IU/mg and 2.8 g/L) and pBPAEB/BW25113 (0.010 IU/mg and 2.22 g/L). These results indicated that the activity and stability of UGDH plays a significant role in HA production, and should prove useful for further genetic engineering research with a view to construct other glucuronic acid polysaccharide synthesis pathways.


Subject(s)
Bacterial Proteins/metabolism , Hyaluronic Acid/biosynthesis , Pasteurella multocida/enzymology , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Enzyme Stability , Escherichia coli/genetics , Genes, Bacterial , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics
12.
Protein Pept Lett ; 22(7): 628-34, 2015.
Article in English | MEDLINE | ID: mdl-26008638

ABSTRACT

UDP-glucose dehydrogenases (EC 1.1.1.22) are responsible for the conversion of UDP-glucose to UDP-glucuronic acid, a key precursor in the biosynthesis of glycoconjugates. Herein we report the discovery and characterization of a UDPglucose dehydrogenase (GbUGD) from Granulibacter bethesdensis, a bacterium originally isolated from the lymph nodes of patients with chronic granulomatous disease (CGD). The recombinant form of the protein was expressed in high yield and the purified enzyme showed highest activity at 37°C/pH 9.0 and was strongly inhibited by Zn(2+) ions, sodium dodecyl sulfate (SDS) and urea. UDP-xylose, an allosteric feedback inhibitor, reduced significantly the activity of the enzyme. High activities were observed using the co-substrates UDP-glucose and NAD+, whereas no activity could be detected using other nucleotide sugars or NADP(+) as potential alternative substrates. The high activity combined with the simple purification procedure used make GbUGD a valuable new alternative biocatalyst for the synthesis of UDP-glucuronic acid or the development of NAD+ regeneration systems.


Subject(s)
Acetobacteraceae/enzymology , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Acetobacteraceae/genetics , Cloning, Molecular , Detergents/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Metals/pharmacology , Protein Denaturation/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Temperature , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors , Uridine Diphosphate Glucose Dehydrogenase/genetics
13.
Prikl Biokhim Mikrobiol ; 51(1): 30-6, 2015.
Article in English | MEDLINE | ID: mdl-25842901

ABSTRACT

Biopolymer Ss of Sphingomonas sanxanigenens strain NX02 is an sphingan that can be extracted using a small quantity of acid, which is a low cost extraction process. A UDP-glucose dehydrogenase gene (ugdG), related to Ss biosynthesis, was cloned from S. sanxanigenens NX02 and expressed in Escherichia coli. It encoded a 454-residue protein of 48.2 kDa. The deduced amino acid sequence had 77% identity with UDP-glucose dehydrogenase (UgdG) from Sphingomonas sp. KC8, and 73% identity with UgdG from Sphingomonas elodea ATCC31461. Purified recombinant UgdG had maximum activity at 35°C and pH 8.0, with Km values of 0.47 and 0.38 mM for UDP-glucose and NAD+, respectively. Overexpression of the ugdG gene in S. sanxanigenens resulted in increased (14.9 ± 0.5)% Ss production and higher fermentation broth viscosity. Furthermore, the weight-average molecular weight of polymer Ss from the recombinant strain was (5.3 ± 0.16)% higher and the viscosity was (74 ± 0.15)% higher than those from the WT strain at a shear rate of 1 rev/min.


Subject(s)
Biopolymers/biosynthesis , Sphingomonas/enzymology , Uridine Diphosphate Glucose Dehydrogenase/genetics , Amino Acids/genetics , Cloning, Molecular , Gene Expression Regulation, Bacterial , Kinetics , Phylogeny , Sequence Alignment , Sequence Analysis, Protein , Uridine Diphosphate Glucose Dehydrogenase/biosynthesis , Uridine Diphosphate Glucose Dehydrogenase/chemistry
14.
Biochemistry ; 53(51): 8043-51, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25478983

ABSTRACT

Human UDP-α-d-glucose-6-dehydrogenase (hUGDH) displays hysteresis because of a slow isomerization from an inactive state (E*) to an active state (E). Here we show that the structure of E* constrains hUGDH in a conformation that favors feedback inhibition at physiological pH. The feedback inhibitor UDP-α-d-xylose (UDP-Xyl) competes with the substrate UDP-α-d-glucose for the active site. Upon binding, UDP-Xyl triggers an allosteric switch that changes the structure and affinity of the intersubunit interface to form a stable but inactive horseshoe-shaped hexamer. Using sedimentation velocity studies and a new crystal structure, we show that E* represents a stable conformational intermediate between the active and feedback-inhibited conformations. Because the allosteric switch occludes the cofactor and substrate binding sites in the inactive hexamer, the intermediate conformation observed in the crystal structure is consistent with the E* transient observed in relaxation studies. Steady-state analysis shows that the E* conformation enhances the affinity of hUGDH for the allosteric inhibitor UDP-Xyl by 8.6-fold (Ki = 810 nM). We present a model in which the constrained quaternary structure permits a small effector molecule to leverage a disproportionately large allosteric response.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/chemistry , Allosteric Regulation , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , Enzyme Stability , Feedback, Physiological , Humans , Hydrogen-Ion Concentration , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Ligands , Models, Molecular , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate Xylose/metabolism , Uridine Diphosphate Xylose/pharmacology
15.
J Biol Chem ; 288(49): 35049-57, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24145036

ABSTRACT

UDP-glucose dehydrogenase (UGDH) provides precursors for steroid elimination, hyaluronan production, and glycosaminoglycan synthesis. The wild-type UGDH enzyme purifies in a hexamer-dimer equilibrium and transiently undergoes dynamic motion that exposes the dimer-dimer interface during catalysis. In the current study we created and characterized point mutations that yielded exclusively dimeric species (obligate dimer, T325D), dimeric species that could be induced to form hexamers in the ternary complex with substrate and cofactor (T325A), and a previously described exclusively hexameric species (UGDHΔ132) to investigate the role of quaternary structure in regulation of the enzyme. Characterization of the purified enzymes revealed a significant decrease in the enzymatic activity of the obligate dimer and hexamer mutants. Kinetic analysis of wild-type UGDH and the inducible hexamer, T325A, showed that upon increasing enzyme concentration, which favors the hexameric species, activity was modestly decreased and exhibited cooperativity. In contrast, cooperative kinetic behavior was not observed in the obligate dimer, T325D. These observations suggest that the regulation of the quaternary assembly of the enzyme is essential for optimal activity and allosteric regulation. Comparison of kinetic and thermal stability parameters revealed structurally dependent properties consistent with a role for controlled assembly and disassembly of the hexamer in the regulation of UGDH. Finally, both T325A and T325D mutants were significantly less efficient in promoting downstream hyaluronan production by HEK293 cells. These data support a model that requires an operational dimer-hexamer equilibrium to function efficiently and preserve regulated activity in the cell.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Amino Acid Substitution , Enzyme Stability , HEK293 Cells , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits , Proteolysis , Thermodynamics , Uridine Diphosphate Glucose Dehydrogenase/genetics
16.
J Biol Chem ; 288(32): 23064-74, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23792965

ABSTRACT

UDP-glucose dehydrogenase (Ugd) generates UDP-glucuronic acid, an important precursor for the production of many hexuronic acid-containing bacterial surface glycostructures. In Escherichia coli K-12, Ugd is important for biosynthesis of the environmentally regulated exopolysaccharide known as colanic acid, whereas in other E. coli isolates, the same enzyme is required for production of the constitutive group 1 capsular polysaccharides, which act as virulence determinants. Recent studies have implicated tyrosine phosphorylation in the activation of Ugd from E. coli K-12, although it is not known if this is a feature shared by bacterial Ugd proteins. The activities of Ugd from E. coli K-12 and from the group 1 capsule prototype (serotype K30) were compared. Surprisingly, for both enzymes, site-directed Tyr → Phe mutants affecting the previously proposed phosphorylation site retained similar kinetic properties to the wild-type protein. Purified Ugd from E. coli K-12 had significant levels of NAD substrate inhibition, which could be alleviated by the addition of ATP and several other nucleotide triphosphates. Mutations in a previously identified UDP-glucuronic acid allosteric binding site decreased the binding affinity of the nucleotide triphosphate. Ugd from E. coli serotype K30 was not inhibited by NAD, but its activity still increased in the presence of ATP.


Subject(s)
Adenosine Triphosphate , Escherichia coli K12/enzymology , Escherichia coli Proteins , NAD , Uridine Diphosphate Glucose Dehydrogenase , Virulence Factors , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Kinetics , Mutagenesis, Site-Directed , Mutation, Missense , NAD/chemistry , NAD/genetics , NAD/metabolism , Polysaccharides/biosynthesis , Polysaccharides/chemistry , Polysaccharides/genetics , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
17.
Biochemistry ; 52(8): 1456-65, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23363239

ABSTRACT

Human UDP-α-d-glucose 6-dehydrogenase (hUGDH) forms a hexamer that catalyzes the NAD(+)-dependent oxidation of UDP-α-d-glucose (UDG) to produce UDP-α-d-glucuronic acid. Mammalian UGDH displays hysteresis (observed as a lag in progress curves), indicating that the enzyme undergoes a slow transition from an inactive to an active state. Here we show that hUGDH is sensitive to product inhibition during the lag. The inhibition results in a systematic decrease in steady-state velocity and makes the lag appear to have a second-order dependence on enzyme concentration. Using transient-state kinetics, we confirm that the lag is in fact due to a substrate and cofactor-induced isomerization of the enzyme. We also show that the cofactor binds to the hUGDH:UDG complex with negative cooperativity. This suggests that the isomerization may be related to the formation of an asymmetric enzyme complex. We propose that the hysteresis in hUGDH is the consequence of a functional adaptation; by slowing the response of hUGDH to sudden increases in the flux of UDG, the other biochemical pathways that use this important metabolite (i.e., glycolysis) will have a competitive edge.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/metabolism , Enzyme Activation , Humans , Isomerism , Kinetics , Models, Molecular , NAD/metabolism , Protein Binding , Protein Multimerization , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose Dehydrogenase/chemistry
18.
Appl Environ Microbiol ; 79(9): 2968-78, 2013 May.
Article in English | MEDLINE | ID: mdl-23435893

ABSTRACT

Pseudomonas aeruginosa produces three exopolysaccharides, Psl, Pel, and alginate, that play vital roles in biofilm formation. Pel is a glucose-rich, cellulose-like exopolysaccharide. The essential Pel biosynthesis proteins are encoded by seven genes, pelA to pelG. Bioinformatics analysis suggests that PelF is a cytosolic glycosyltransferase. Here, experimental evidence was provided to support this PelF function. A UDP-glucose dehydrogenase-based assay was developed to quantify UDP-glucose. UDP-glucose was proposed as the substrate for PelF. The isogenic pelF deletion mutant accumulated 1.8 times more UDP-glucose in its cytosol than the wild type. This suggested that PelF, which was found localized in the cystosol, uses UDP-glucose as substrate. Additionally, in vitro experiments confirmed that PelF uses UDP-glucose as substrate. To analyze the functional roles of conserved residues in PelF, site-directed mutagenesis was performed. The presence of the EX7E motif is characteristic for various glycosyltransferase families, and in PelF, E405/E413 are the conserved residues in this motif. Replacement of E405 with A resulted in a reduction of PelF activity to 30.35% ± 3.15% (mean ± standard deviation) of the wild-type level, whereas replacement of the second E, E413, with A did not produce a significant change in the activity of PelF. Moreover, replacement of both E residues did not result in a loss of PelF function, but replacement of the conserved R325 or K330 with A resulted in a complete loss of PelF activity. Overall, our data show that PelF is a soluble glycosyltransferase that uses UDP-glucose as the substrate for Pel synthesis and that conserved residues R325 and K330 are important for the activity of PelF.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Glycosyltransferases/metabolism , Polysaccharides/metabolism , Pseudomonas aeruginosa/enzymology , Alginates/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Glucuronic Acid/metabolism , Glycosyltransferases/chemistry , Glycosyltransferases/genetics , Glycosyltransferases/isolation & purification , Hexuronic Acids/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Recombinant Fusion Proteins , Sequence Deletion , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism
19.
Biochem Biophys Res Commun ; 430(1): 218-24, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23137539

ABSTRACT

Metabolic enzymes are usually characterized to have one specific function, and this is the case of UDP-glucose dehydrogenase that catalyzes the twofold NAD(+)-dependent oxidation of UDP-glucose into UDP-glucuronic acid. We have determined that this enzyme is also capable of participating in other cellular processes. Here, we report that the bacterial UDP-glucose dehydrogenase (UgdG) from Sphingomonas elodea ATCC 31461, which provides UDP-glucuronic acid for the synthesis of the exopolysaccharide gellan, is not only able to bind RNA but also acts as a ribonuclease. The ribonucleolytic activity occurs independently of the presence of NAD(+) and the RNA binding site does not coincide with the NAD(+) binding region. We have also performed the kinetics of interaction between UgdG and RNA. Moreover, computer analysis reveals that the N- and C-terminal domains of UgdG share structural features with ancient mitochondrial ribonucleases named MAR. MARs are present in lower eukaryotic microorganisms, have a Rossmannoid-fold and belong to the isochorismatase superfamily. This observation reinforces that the Rossmann structural motifs found in NAD(+)-dependent dehydrogenases can have a dual function working as a nucleotide cofactor binding domain and as a ribonuclease.


Subject(s)
Bacterial Proteins/metabolism , RNA/metabolism , Ribonucleases/metabolism , Sphingomonas/enzymology , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Kinetics , NAD/chemistry , NAD/metabolism , Protein Folding , RNA/chemistry , Ribonucleases/chemistry , Uridine Diphosphate Glucose Dehydrogenase/chemistry
20.
Biochemistry ; 51(46): 9364-74, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23106432

ABSTRACT

Human UDP-α-D-glucose dehydrogenase (hUGDH) catalyzes the NAD(+)-dependent oxidation of UDP-α-D-glucose (UDG) to produce UDP-α-D-glucuronic acid. The oligomeric structure of hUGDH is dynamic and can form two distinct hexameric complexes in solution. The active form of hUGDH consists of dimers that undergo a concentration-dependent association to form a hexamer with 32 symmetry. In the presence of the allosteric feedback inhibitor UDP-α-D-xylose (UDX), hUGDH changes shape to form an inactive, horseshoe-shaped complex. Previous studies have identified the UDX-induced allosteric mechanism that changes the hexameric structure to inhibit the enzyme. Here, we investigate the role of the 32 symmetry hexamer in the catalytic cycle. We engineered a stable hUGDH dimer by introducing a charge-switch substitution (K94E) in the hexamer-building interface (hUGDH(K94E)). The k(cat) of hUGDH(K94E) is ~160-fold lower than that of the wild-type enzyme, suggesting that the hexamer is the catalytically relevant state. We also show that cofactor binding triggers the formation of the 32 symmetry hexamer, but UDG is needed for the stability of the complex. The hUGDH(K94E) crystal structure at 2.08 Å resolution identifies loop(88-110) as the cofactor-responsive allosteric switch that drives hexamer formation; loop(88-110) directly links cofactor binding to the stability of the hexamer-building interface. In the interface, loop(88-110) packs against the Thr131-loop/α6 helix, the allosteric switch that responds to the feedback inhibitor UDX. We also identify a structural element (the S-loop) that explains the indirect stabilization of the hexamer by substrate and supports a sequential, ordered binding of the substrate and cofactor. These observations support a model in which (i) UDG binds to the dimer and stabilizes the S-loop to promote cofactor binding and (ii) cofactor binding orders loop(88-110) to induce formation of the catalytically active hexamer.


Subject(s)
Uridine Diphosphate Glucose Dehydrogenase/metabolism , Allosteric Regulation , Humans , Kinetics , Models, Molecular , Protein Binding , Uridine Diphosphate Glucose Dehydrogenase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...