Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1822(10): 1544-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22771891

ABSTRACT

Mutations in the gene encoding for 4-hydroxy-2-oxoglutarate aldolase (HOGA) are associated with an excessive production of oxalate in Primary Hyperoxaluria type 3 (PH3). This enzyme is the final step of the hydroxyproline degradation pathway within the mitochondria and catalyzes the cleavage of 4-hydroxy-2-oxoglutarate (HOG) to pyruvate and glyoxylate. No analyses have been performed to assess the consequences of the mutations identified, particularly for those variants that produce either full-length or nearly full-length proteins. In this study, the expression, stability, and activity of nine PH3 human HOGA variants were examined. Using recombinant protein produced in Escherichia coli as well as transfected Chinese hamster ovary (CHO) cells, it was found that all nine PH3 variants are quite unstable, have a tendency to aggregate, and retain no measurable activity. A buildup of HOG was confirmed in the urine, sera and liver samples from PH3 patients. To determine how HOG is cleaved in the absence of HOGA activity, the ability of N-acetylneuraminate aldolase (NAL) to cleave HOG was evaluated. NAL showed minimal activity towards HOG. Whether the expected buildup of HOG in mitochondria could inhibit glyoxylate reductase (GR), the enzyme mutated in PH2, was also evaluated. GR was inhibited by HOG but not by 2-hydroxyglutarate or 2-oxoglutarate. Thus, one hypothetical component of the molecular basis for the excessive oxalate production in PH3 appears to be the inhibition of GR by HOG, resulting in a phenotype similar to PH2.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Hyperoxaluria, Primary/enzymology , Oxo-Acid-Lyases/antagonists & inhibitors , Oxo-Acid-Lyases/metabolism , Alcohol Oxidoreductases/genetics , Animals , CHO Cells , Cells, Cultured , Cricetinae , Glutarates/metabolism , Humans , Hydroxyproline/metabolism , Hyperoxaluria, Primary/genetics , Ketoglutaric Acids/metabolism , Liver/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Oxalates/metabolism , Oxo-Acid-Lyases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection/methods
2.
Am J Physiol Gastrointest Liver Physiol ; 302(6): G637-43, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22207577

ABSTRACT

Hydroxyproline (Hyp) metabolism is a key source of glyoxylate production in the body and may be a major contributor to excessive oxalate production in the primary hyperoxalurias where glyoxylate metabolism is impaired. Important gaps in our knowledge include identification of the tissues with the capacity to degrade Hyp and the development of model systems to study this metabolism and how to suppress it. The expression of mRNA for enzymes in the pathway was examined in 15 different human tissues. Expression of the complete pathway was identified in liver, kidney, pancreas, and small intestine. HepG2 cells also expressed these mRNAs and enzymes and were shown to metabolize Hyp in the culture medium to glycolate, glycine, and oxalate. [(18)O]- and [(13)C(5)]Hyp were synthesized and evaluated for their use with in vitro and in vivo models. [(18)O]Hyp was not suitable because of an apparent tautomerism of [(18)O]glyoxylate between enol and hydrated forms, which resulted in a loss of isotope. [(13)C(5)]Hyp, however, was metabolized to [(13)C(2)]glycolate, [(13)C(2)]glycine, and [(13)C(2)]oxalate in vitro in HepG2 cells and in vivo in mice infused with [(13)C(5)]Hyp. These model systems should be valuable tools for exploring various aspects of Hyp metabolism and will be useful in determining whether blocking Hyp catabolism is an effective therapy in the treatment of primary hyperoxaluria.


Subject(s)
Hydroxyproline/metabolism , Hyperoxaluria/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Carbon Isotopes , Gene Expression Regulation/physiology , Hep G2 Cells , Humans , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
PLoS One ; 6(10): e26021, 2011.
Article in English | MEDLINE | ID: mdl-21998747

ABSTRACT

BACKGROUND: 4-hydroxy-2-oxoglutarate (HOG) aldolase is a unique enzyme in the hydroxyproline degradation pathway catalyzing the cleavage of HOG to pyruvate and glyoxylate. Mutations in this enzyme are believed to be associated with the excessive production of oxalate in primary hyperoxaluria type 3 (PH3), although no experimental data is available to support this hypothesis. Moreover, the identity, oligomeric state, enzymatic activity, and crystal structure of human HOGA have not been experimentally determined. METHODOLOGY/PRINCIPAL FINDINGS: In this study human HOGA (hHOGA) was identified by mass spectrometry of the mitochondrial enzyme purified from bovine kidney. hHOGA performs a retro-aldol cleavage reaction reminiscent of the trimeric 2-keto-3-deoxy-6-phosphogluconate aldolases. Sequence comparisons, however, show that HOGA is related to the tetrameric, bacterial dihydrodipicolinate synthases, but the reaction direction is reversed. The 1.97 Å resolution crystal structure of hHOGA bound to pyruvate was determined and enabled the modeling of the HOG-Schiff base intermediate and the identification of active site residues. Kinetic analyses of site-directed mutants support the importance of Lys196 as the nucleophile, Tyr168 and Ser77 as components of a proton relay, and Asn78 and Ser198 as unique residues that facilitate substrate binding. CONCLUSIONS/SIGNIFICANCE: The biochemical and structural data presented support that hHOGA utilizes a type I aldolase reaction mechanism, but employs novel residue interactions for substrate binding. A mapping of the PH3 mutations identifies potential rearrangements in either the active site or the tetrameric assembly that would likely cause a loss in activity. Altogether, these data establish a foundation to assess mutant forms of hHOGA and how their activity could be pharmacologically restored.


Subject(s)
Hydroxyproline/metabolism , Hyperoxaluria, Primary/metabolism , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Amino Acid Sequence , Animals , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/isolation & purification , Apoenzymes/metabolism , Bacteria/enzymology , Catalytic Domain , Cattle , Crystallography, X-Ray , Humans , Hyperoxaluria, Primary/genetics , Hyperoxaluria, Primary/pathology , Kidney/pathology , Mass Spectrometry , Mitochondria/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/isolation & purification , Protein Multimerization , Protein Structure, Quaternary , Pyruvic Acid/metabolism , Schiff Bases/metabolism , Solutions , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL