Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Poult Sci ; 97(7): 2249-2257, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29672771

ABSTRACT

Footpad dermatitis is a condition that causes lesions on the plantar surface of the footpads in growing turkeys. Potential inflammatory processes and pain associated with increasing severity of footpad dermatitis raise animal welfare concerns. This study investigated whether the temperature of the plantar surface of the foot (the footpads and the entire plantar foot including interdigital membranes) assessed with infrared thermography reflects severity of mild footpad dermatitis as assessed with a Visual Analogue Scale in 80 turkey toms at 10 weeks of age. In order to study effects of a potential emotional arousal due to the testing procedures, effects of sequential testing order and duration of handling of the turkeys was included in the model. Footpad temperatures were significantly lower than foot temperatures (P < 0.001, R2 = 0.57, -3.36°C ± 0.28°C), and higher visual analogue scale scores were anti-correlated with footpad (-0.06°C ± 0.037°C) and foot temperatures (-0.07°C ± 0.066°C). Furthermore, a negative association between footpad temperature and handling time (-0.02 ± 0.0227, P = 0.048), and a non-linear association between foot and footpad temperatures and sequential testing order, were found (P<0.001). The results indicate that severity of mild footpad dermatitis as scored visually was associated with the temperatures of the plantar surface of the foot and footpads, and that thermal imaging therefore represents a novel tool for the reliable and non-invasive early detection of subclinical foot pathologies in turkeys. The association was negative, and the findings therefore indicate that potential inflammatory processes in the epidermis at this early stage of footpad dermatitis are negligible, and/or that the hyperkeratosis of the surface keratin shielded heat emission from the footpads. The associations between surface temperatures, handling time, and sequential testing order suggest an emotional arousal in response to the experimental procedures, and these factors need to be considered when applying infrared thermography in future studies of leg health in turkeys.


Subject(s)
Arousal/physiology , Dermatitis/veterinary , Emotions/physiology , Poultry Diseases/physiopathology , Temperature , Turkeys , Animals , Asymptomatic Infections , Dermatitis/physiopathology , Foot/physiopathology , Foot Diseases/physiopathology , Foot Diseases/veterinary , Male , Thermography/veterinary
2.
J Inherit Metab Dis ; 24(2): 213-30, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11405341

ABSTRACT

Deficiencies in the human enzyme phenylalanine hydroxylase (PAH) due to mutations in the PAH gene (PAH) result in the inborn error of metabolism phenylketonuria (PKU). The clinical symptom of this disease is an elevated concentration of L-phenylalanine (L-Phe) in blood serum. To prevent mental retardation due to the buildup of neurotoxic metabolites of L-Phe, patients with severe PKU must be treated with a low-L-Phe diet starting early in their life. Owing to extensive newborn screening programmes and genotyping efforts, more than 400 different mutations have been identified in the PAH gene. Recently, there have been several reports of PKU patients showing a normalization of their L-Phe concentrations upon oral administration of the natural cofactor to PAH, (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin (BH4). In an attempt to correlate the clinical responsiveness to BH4 administration with PKU genotype, we propose specific structural consequences for this subset of PAH mutations. Based on the location and proximity of this subset of mutations to the cofactor-binding site in the three-dimensional structure of PAH, a hypothesis for BH4 responsiveness in PKU patients is presented. It is believed that some of these mutations result in expressed mutant enzymes that are Km variants (with a lower binding affinity for BH4) of the standard PAH enzyme phenotype. Oral administration of excess BH4 thus makes it possible for these mutant enzymes to suppress their low binding affinity for BH4, enabling this subset of PAH mutations to perform the L-Phe hydroxylation reaction. Most of the BH4-responsive PAH mutations map to the catalytic domain of PAH in either of two categories. Residues are located in cofactor-binding regions or in regions that interact with the secondary structural elements involved in cofactor binding. Based on the series of known mutations that have been found to be responsive to BH4, we propose that other subsets of PAH mutations will have a high likelihood of being responsive to oral BH4 administration.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Biopterins/analogs & derivatives , Biopterins/therapeutic use , Phenylalanine/metabolism , Phenylketonurias/drug therapy , Phenylketonurias/metabolism , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Sequence , Animals , Biopterins/chemistry , Humans , Molecular Sequence Data , Phenylalanine Hydroxylase/chemistry , Phenylketonurias/genetics , Protein Conformation
3.
Curr Opin Struct Biol ; 10(6): 719-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11114510

ABSTRACT

An important goal of structural genomics is to complete the structural analysis of all the enzymes in metabolic pathways and to understand the structural similarities and differences. A preliminary glimpse of this type of analysis was achieved before structural genomics efforts with the glycolytic pathway and efforts are underway for many other pathways, including that of catecholamine metabolism. Structural enzymology necessitates a complete structural characterization, even for highly homologous proteins (greater than 80% sequence homology), as every active site has distinct structural features and it is these active site differences that distinguish one enzyme from another. Short cuts with homology modeling cannot be taken with our current knowledge base. Each enzyme structure in a pathway needs to be determined, including structures containing bound substrates, cofactors, products and transition state analogs, in order to obtain a complete structural and functional understanding of pathway-related enzymes.


Subject(s)
Enzymes/metabolism , Genome , Binding Sites , Catecholamines/metabolism , Enzymes/chemistry , Glycolysis , Protein Conformation
4.
Biochemistry ; 39(9): 2208-17, 2000 Mar 07.
Article in English | MEDLINE | ID: mdl-10694386

ABSTRACT

The crystal structure of the dimeric catalytic domain (residues 118-424) of human PheOH (hPheOH), cocrystallized with the oxidized form of the cofactor (7,8-dihydro-L-biopterin, BH(2)), has been determined at 2.0 A resolution. The pterin binds in the second coordination sphere of the catalytic iron (the C4a atom is 6.1 A away), and interacts through several hydrogen bonds to two water molecules coordinated to the iron, as well as to the main chain carbonyl oxygens of Ala322, Gly247, and Leu249 and the main chain amide of Leu249. Some important conformational changes are seen in the active site upon pterin binding. The loop between residues 245 and 250 moves in the direction of the iron, and thus allows for several important hydrogen bonds to the pterin ring to be formed. The pterin cofactor is in an ideal orientation for dioxygen to bind in a bridging position between the iron and the pterin. The pterin ring forms an aromatic pi-stacking interaction with Phe254, and Tyr325 contributes to the positioning of the pterin ring and its dihydroxypropyl side chain by hydrophobic interactions. Of particular interest in the hPheOH x BH(2) binary complex structure is the finding that Glu286 hydrogen bonds to one of the water molecules coordinated to the iron as well as to a water molecule which hydrogen bonds to N3 of the pterin ring. Site-specific mutations of Glu286 (E286A and E286Q), Phe254 (F254A and F254L), and Tyr325 (Y325F) have confirmed the important contribution of Glu286 and Phe254 to the normal positioning of the pterin cofactor and catalytic activity of hPheOH. Tyr325 also contributes to the correct positioning of the pterin, but has no direct function in the catalytic reaction, in agreement with the results obtained with rat TyrOH [Daubner, S. C., and Fitzpatrick, P. F. (1998) Biochemistry 37, 16440-16444]. Superposition of the binary hPheOH.BH(2) complex onto the crystal structure of the ligand-free rat PheOH (which contains the regulatory and catalytic domains) [Kobe, B., Jennings, I. G., House, C. M., Michell, B. J., Goodwill, K. E., Santarsiero, B. D., Stevens, R. C., Cotton, R. G. H., and Kemp, B. E. (1999) Nat. Struct. Biol. 6, 442-448] reveals that the C2'-hydroxyl group of BH(2) is sufficiently close to form hydrogen bonds to Ser23 in the regulatory domain. Similar interactions are seen with the hPheOH.adrenaline complex and Ser23. These interactions suggest a structural explanation for the specific regulatory properties of the dihydroxypropyl side chain of BH(4) (negative effector) in the full-length enzyme in terms of phosphorylation of Ser16 and activation by L-Phe.


Subject(s)
Mutagenesis, Site-Directed , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/genetics , Pterins/chemistry , Binding Sites/genetics , Biopterins/analogs & derivatives , Biopterins/chemistry , Biopterins/metabolism , Catalysis , Catecholamines/antagonists & inhibitors , Crystallography, X-Ray , Dimerization , Humans , Phenylalanine/metabolism , Phenylalanine Hydroxylase/metabolism , Pterins/metabolism
5.
Mol Genet Metab ; 68(2): 103-25, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10527663

ABSTRACT

The human phenylalanine hydroxylase gene (PAH) (locus on human chromosome 12q24.1) contains the expressed nucleotide sequence which encodes the hepatic enzyme phenylalanine hydroxylase (PheOH). The PheOH enzyme hydroxylates the essential amino acid l-phenylalanine resulting in another amino acid, tyrosine. This is the major pathway for catabolizing dietary l-phenylalanine and accounts for approximately 75% of the disposal of this amino acid. The autosomal recessive disease phenylketonuria (PKU) is the result of a deficiency of PheOH enzymatic activity due to mutations in the PAH gene. Of the mutant alleles that cause hyperphenylalaninemia or PKU 99% map to the PAH gene. The remaining 1% maps to several genes that encode enzymes involved in the biosynthesis or regeneration of the cofactor ((6R)-l-erythro-5,6,7,8-tetrahydrobiopterin) regenerating the cofactor (tetrahydrobiopterin) necessary for the hydroxylation reaction. The recently solved crystal structures of human phenylalanine hydroxylase provide a structural scaffold for explaining the effects of some of the mutations in the PAH gene and suggest future biochemical studies that may increase our understanding of the PKU mutations.


Subject(s)
Phenylalanine Hydroxylase/genetics , Phenylketonurias/enzymology , Amino Acid Sequence , Humans , Molecular Sequence Data , Mutation , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/deficiency , Phenylketonurias/genetics , Protein Conformation
6.
Biochemistry ; 37(45): 15638-46, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9843368

ABSTRACT

The aromatic amino acid hydroxylases represent a superfamily of structurally and functionally closely related enzymes, one of those functions being reversible inhibition by catechol derivatives. Here we present the crystal structure of the dimeric catalytic domain (residues 117-424) of human phenylalanine hydroxylase (hPheOH), cocrystallized with various potent and well-known catechol inhibitors and refined at a resolution of 2.0 A. The catechols bind by bidentate coordination to each iron in both subunits of the dimer through the catechol hydroxyl groups, forming a blue-green colored ligand-to-metal charge-transfer complex. In addition, Glu330 and Tyr325 are identified as determinant residues in the recognition of the inhibitors. In particular, the interaction with Glu330 conforms to the structural explanation for the pH dependence of catecholamine binding to PheOH, with a pKa value of 5.1 (20 degreesC). The overall structure of the catechol-bound enzyme is very similar to that of the uncomplexed enzyme (rms difference of 0.2 A for the Calpha atoms). Most striking is the replacement of two iron-bound water molecules with catechol hydroxyl groups. This change is consistent with a change in the ligand field symmetry of the high-spin (S = 5/2) Fe(III) from a rhombic to a nearly axial ligand field symmetry as seen upon noradrenaline binding using EPR spectroscopy [Martinez, A., Andersson, K. K., Haavik, J., and Flatmark, T. (1991) Eur. J. Biochem. 198, 675-682]. Crystallographic comparison with the structurally related rat tyrosine hydroxylase binary complex with the oxidized cofactor 7,8-dihydrobiopterin revealed overlapping binding sites for the catechols and the cofactor, compatible with a competitive type of inhibition of the catechols versus BH4. The comparison demonstrates some structural differences at the active site as the potential basis for the different substrate specificity of the two enzymes.


Subject(s)
Catecholamines/chemistry , Phenylalanine Hydroxylase/antagonists & inhibitors , Phenylalanine Hydroxylase/chemistry , Binding, Competitive , Catalysis , Catecholamines/metabolism , Catecholamines/pharmacology , Catechols/chemistry , Catechols/metabolism , Computer Simulation , Crystallization , Crystallography, X-Ray , Humans , Macromolecular Substances , Models, Molecular , Phenylalanine Hydroxylase/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary
7.
J Biol Chem ; 273(27): 16962-7, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9642259

ABSTRACT

Phenylalanine hydroxylase (PheOH) catalyzes the conversion of L-phenylalanine to L-tyrosine, the rate-limiting step in the oxidative degradation of phenylalanine. Mutations in the human PheOH gene cause phenylketonuria, a common autosomal recessive metabolic disorder that in untreated patients often results in varying degrees of mental retardation. We have determined the crystal structure of human PheOH (residues 118-452). The enzyme crystallizes as a tetramer with each monomer consisting of a catalytic and a tetramerization domain. The tetramerization domain is characterized by the presence of a domain swapping arm that interacts with the other monomers forming an antiparallel coiled-coil. The structure is the first report of a tetrameric PheOH and displays an overall architecture similar to that of the functionally related tyrosine hydroxylase. In contrast to the tyrosine hydroxylase tetramer structure, a very pronounced asymmetry is observed in the phenylalanine hydroxylase, caused by the occurrence of two alternate conformations in the hinge region that leads to the coiled-coil helix. Examination of the mutations causing PKU shows that some of the most frequent mutations are located at the interface of the catalytic and tetramerization domains. Their effects on the structural and cellular stability of the enzyme are discussed.


Subject(s)
Phenylalanine Hydroxylase/chemistry , Phenylketonurias/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Enzyme Stability , Humans , Molecular Sequence Data , Phenylalanine Hydroxylase/deficiency , Phenylalanine Hydroxylase/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
8.
Nat Struct Biol ; 4(12): 995-1000, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9406548

ABSTRACT

The 2.0 A crystal structure of the catalytic domain of human phenylalanine hydroxylase reveals a fold similar to that of tyrosine hydroxylase. It provides the first structural view of where mutations occur and a rationale to explain molecular mechanisms of the enzymatic phenotypes in the autosomal recessive disorder phenylketoneuria.


Subject(s)
Phenylalanine Hydroxylase/chemistry , Phenylketonurias/enzymology , Amino Acid Sequence , Animals , Binding Sites/genetics , Crystallization , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics , Protein Folding , Tyrosine 3-Monooxygenase/chemistry , Tyrosine 3-Monooxygenase/genetics
9.
FEBS Lett ; 406(1-2): 171-4, 1997 Apr 07.
Article in English | MEDLINE | ID: mdl-9109411

ABSTRACT

A recombinant truncated form (delta1-102/delta428-452) of the non-heme iron-dependent metalloenzyme human phenylalanine hydroxylase (hPAH, phenylalanine 4-monooxygenase; EC 1.14.16.1) was expressed in E. coli, purified to homogeneity as a homodimer (70 kDa) and crystallized using the hanging drop vapour diffusion method. The crystals are orthorhombic, space group C222 with cell dimensions of a = 66.6 A, b = 108.4 A, c = 125.7 A. The calculated packing parameter (Vm) is 3.24 A3/Da with four 2-fold symmetric dimers (or eight momomers) in the unit cell. Data have been collected to 2.0 A resolution.


Subject(s)
Phenylalanine Hydroxylase/chemistry , Crystallization , Humans , Recombinant Proteins/chemistry , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...