Academic literature on the topic 'GLYOXYLATE AMINOTRANSFERASE'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'GLYOXYLATE AMINOTRANSFERASE.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "GLYOXYLATE AMINOTRANSFERASE"

1

Han, Qian, Cihan Yang, Jun Lu, Yinai Zhang, and Jianyong Li. "Metabolism of Oxalate in Humans: A Potential Role Kynurenine Aminotransferase/Glutamine Transaminase/Cysteine Conjugate Betalyase Plays in Hyperoxaluria." Current Medicinal Chemistry 26, no. 26 (October 22, 2019): 4944–63. http://dx.doi.org/10.2174/0929867326666190325095223.

Full text
Abstract:
Hyperoxaluria, excessive urinary oxalate excretion, is a significant health problem worldwide. Disrupted oxalate metabolism has been implicated in hyperoxaluria and accordingly, an enzymatic disturbance in oxalate biosynthesis can result in the primary hyperoxaluria. Alanine-glyoxylate aminotransferase-1 and glyoxylate reductase, the enzymes involving glyoxylate (precursor for oxalate) metabolism, have been related to primary hyperoxalurias. Some studies suggest that other enzymes such as glycolate oxidase and alanine-glyoxylate aminotransferase-2 might be associated with primary hyperoxaluria as well, but evidence of a definitive link is not strong between the clinical cases and gene mutations. There are still some idiopathic hyperoxalurias, which require a further study for the etiologies. Some aminotransferases, particularly kynurenine aminotransferases, can convert glyoxylate to glycine. Based on biochemical and structural characteristics, expression level, and subcellular localization of some aminotransferases, a number of them appear able to catalyze the transamination of glyoxylate to glycine more efficiently than alanine glyoxylate aminotransferase-1. The aim of this minireview is to explore other undermining causes of primary hyperoxaluria and stimulate research toward achieving a comprehensive understanding of underlying mechanisms leading to the disease. Herein, we reviewed all aminotransferases in the liver for their functions in glyoxylate metabolism. Particularly, kynurenine aminotransferase-I and III were carefully discussed regarding their biochemical and structural characteristics, cellular localization, and enzyme inhibition. Kynurenine aminotransferase-III is, so far, the most efficient putative mitochondrial enzyme to transaminate glyoxylate to glycine in mammalian livers, which might be an interesting enzyme to look for in hyperoxaluria etiology of primary hyperoxaluria and should be carefully investigated for its involvement in oxalate metabolism.
APA, Harvard, Vancouver, ISO, and other styles
2

Orzechowski, S., J. Socha-Hanc, and A. Paszkowski. "Alanine aminotransferase and glycine aminotransferase from maize (Zea mays L.) leaves." Acta Biochimica Polonica 46, no. 2 (June 30, 1999): 447–57. http://dx.doi.org/10.18388/abp.1999_4176.

Full text
Abstract:
Alanine aminotransferase (AlaAT, EC 2.6.1.2) and glycine aminotransferase (GlyAT, EC 2.6.1.4), two different enzymes catalyzing transamination reactions with L-alanine as the amino-acid substrate, were examined in maize in which alanine participates substantially in nitrogen transport. Preparative PAGE of a partially purified preparation of aminotransferases from maize leaves gave 6 fractions differing in electrophoretic mobility. The fastest migrating fraction I represents AlaAT specific for L-alanine as amino donor and 2-oxoglutarate as amino acceptor. The remaining fractions showed three aminotransferase activities: L-alanine-2-oxoglutarate, L-alanine-glyoxylate and L-glutamate-glyoxylate. By means of molecular sieving on Zorbax SE-250 two groups of enzymes were distinguished in the PAGE fractions: of about 100 kDa and 50 kDa. Molecular mass of 104 kDa was ascribed to AlaAT in fraction I, while the molecular mass of the three enzymatic activities in 3 fractions of the low electrophoretic mobility was about 50 kDa. The response of these fractions to: aminooxyacetate, 3-chloro-L-alanine and competing amino acids prompted us to suggest that five out of the six preparative PAGE fractions represented GlyAT isoforms, differing from each other by the L-glutamate-glyoxylate:L-alanine-glyoxylate:L-alanine-2-oxoglutarate activity ratio.
APA, Harvard, Vancouver, ISO, and other styles
3

Donini, Stefano, Manuela Ferrari, Chiara Fedeli, Marco Faini, Ilaria Lamberto, Ada Serena Marletta, Lara Mellini, et al. "Recombinant production of eight human cytosolic aminotransferases and assessment of their potential involvement in glyoxylate metabolism." Biochemical Journal 422, no. 2 (August 13, 2009): 265–72. http://dx.doi.org/10.1042/bj20090748.

Full text
Abstract:
PH1 (primary hyperoxaluria type 1) is a severe inborn disorder of glyoxylate metabolism caused by a functional deficiency of the peroxisomal enzyme AGXT (alanine-glyoxylate aminotransferase), which converts glyoxylate into glycine using L-alanine as the amino-group donor. Even though pre-genomic studies indicate that other human transaminases can convert glyoxylate into glycine, in PH1 patients these enzymes are apparently unable to compensate for the lack of AGXT, perhaps due to their limited levels of expression, their localization in an inappropriate cell compartment or the scarcity of the required amino-group donor. In the present paper, we describe the cloning of eight human cytosolic aminotransferases, their recombinant expression as His6-tagged proteins and a comparative study on their ability to transaminate glyoxylate, using any standard amino acid as an amino-group donor. To selectively quantify the glycine formed, we have developed and validated an assay based on bacterial GO (glycine oxidase); this assay allows the detection of enzymes that produce glycine by transamination in the presence of mixtures of potential amino-group donors and without separation of the product from the substrates. We show that among the eight enzymes tested, only GPT (alanine transaminase) and PSAT1 (phosphoserine aminotransferase 1) can transaminate glyoxylate with good efficiency, using L-glutamate (and, for GPT, also L-alanine) as the best amino-group donor. These findings confirm that glyoxylate transamination can occur in the cytosol, in direct competition with the conversion of glyoxylate into oxalate. The potential implications for the treatment of primary hyperoxaluria are discussed.
APA, Harvard, Vancouver, ISO, and other styles
4

Pey, Angel L., Armando Albert, and Eduardo Salido. "Protein Homeostasis Defects of Alanine-Glyoxylate Aminotransferase: New Therapeutic Strategies in Primary Hyperoxaluria Type I." BioMed Research International 2013 (2013): 1–15. http://dx.doi.org/10.1155/2013/687658.

Full text
Abstract:
Alanine-glyoxylate aminotransferase catalyzes the transamination between L-alanine and glyoxylate to produce pyruvate and glycine using pyridoxal 5′-phosphate (PLP) as cofactor. Human alanine-glyoxylate aminotransferase is a peroxisomal enzyme expressed in the hepatocytes, the main site of glyoxylate detoxification. Its deficit causes primary hyperoxaluria type I, a rare but severe inborn error of metabolism. Single amino acid changes are the main type of mutation causing this disease, and considerable effort has been dedicated to the understanding of the molecular consequences of such missense mutations. In this review, we summarize the role of protein homeostasis in the basic mechanisms of primary hyperoxaluria. Intrinsic physicochemical properties of polypeptide chains such as thermodynamic stability, folding, unfolding, and misfolding rates as well as the interaction of different folding states with protein homeostasis networks are essential to understand this disease. The view presented has important implications for the development of new therapeutic strategies based on targeting specific elements of alanine-glyoxylate aminotransferase homeostasis.
APA, Harvard, Vancouver, ISO, and other styles
5

IZUMI, Yoshikazu, Toyokazu YOSNIDA, and Hideaki YAMADA. "An assay for serine-glyoxylate aminotransferase." Agricultural and Biological Chemistry 54, no. 6 (1990): 1573–74. http://dx.doi.org/10.1271/bbb1961.54.1573.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Izumi, Yoshikazu, Toyokazu Yoshida, and Hideaki Yamada. "An Assay for Serine-glyoxylate Aminotransferase." Agricultural and Biological Chemistry 54, no. 6 (June 1990): 1573–74. http://dx.doi.org/10.1080/00021369.1990.10870180.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Han, Qian, Howard Robinson, Yi Gui Gao, Nancy Vogelaar, Scott R. Wilson, Menico Rizzi, and Jianyong Li. "Crystal Structures ofAedes aegyptiAlanine Glyoxylate Aminotransferase." Journal of Biological Chemistry 281, no. 48 (September 21, 2006): 37175–82. http://dx.doi.org/10.1074/jbc.m607032200.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Rumsby, G., T. Weir, and C. T. Samuell. "A Semiautomated Alanine: Glyoxylate Aminotransferase Assay for the Tissue Diagnosis of Primary Hyperoxaluria Type 1." Annals of Clinical Biochemistry: International Journal of Laboratory Medicine 34, no. 4 (July 1997): 400–404. http://dx.doi.org/10.1177/000456329703400411.

Full text
Abstract:
We have developed a sensitive assay for the measurement of alanine:glyoxylate aminotransferase (EC 2.6.1.44) activity in human liver. The assay is partly automated, and takes into consideration the sensitivity of the reaction to pH and to glyoxylate concentration. It is less subject to interference from other enzymes utilizing glyoxylate and to chemical interference from glyoxylate itself and can therefore be used without correction for cross-over by glutamate:glyoxylate aminotransferase (EC 2.6.1.4). The assay allows clear discrimination between normal and affected livers and is sufficiently sensitive to measure enzyme activity in fetal liver samples. Enzyme activity ranged from 17·9 to 38·5 μmol/h/mg protein in control livers ( n = 9) and 0·8 to 9·5 μmol/h/mg protein in 30 of 39 hyperoxaluric patients studied. Normal alanine: glyoxylate aminotransferase activity (from 22·8 to 45·5 μmol/h/mg protein) allowed exclusion of primary hyperoxaluria type 1 in the other nine hyperoxaluric patients.
APA, Harvard, Vancouver, ISO, and other styles
9

Takada, Y., and T. Noguchi. "Characteristics of alanine: glyoxylate aminotransferase from Saccharomyces cerevisiae, a regulatory enzyme in the glyoxylate pathway of glycine and serine biosynthesis from tricarboxylic acid-cycle intermediates." Biochemical Journal 231, no. 1 (October 1, 1985): 157–63. http://dx.doi.org/10.1042/bj2310157.

Full text
Abstract:
Alanine: glyoxylate aminotransferase (EC 2.6.1.44), which is involved in the glyoxylate pathway of glycine and serine biosynthesis from tricarboxylic acid-cycle intermediates in Saccharomyces cerevisiae, was highly purified and characterized. The enzyme had Mr about 80 000, with two identical subunits. It was highly specific for L-alanine and glyoxylate and contained pyridoxal 5′-phosphate as cofactor. The apparent Km values were 2.1 mM and 0.7 mM for L-alanine and glyoxylate respectively. The activity was low (10 nmol/min per mg of protein) with glucose as sole carbon source, but was remarkably high with ethanol or acetate as carbon source (930 and 430 nmol/min per mg respectively). The transamination of glyoxylate is mainly catalysed by this enzyme in ethanol-grown cells. When glucose-grown cells were incubated in medium containing ethanol as sole carbon source, the activity markedly increased, and the increase was completely blocked by cycloheximide, suggesting that the enzyme is synthesized de novo during the incubation period. Similarity in the amino acid composition was observed, but immunological cross-reactivity was not observed among alanine: glyoxylate aminotransferases from yeast and vertebrate liver.
APA, Harvard, Vancouver, ISO, and other styles
10

Sakuraba, Haruhiko. "Studies on Avian Peroxisomal Alanine : Glyoxylate Aminotransferase." Journal of the Kyushu Dental Society 45, no. 3 (1991): 390–408. http://dx.doi.org/10.2504/kds.45.390.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "GLYOXYLATE AMINOTRANSFERASE"

1

Holbrook, Joanna Dawn. "Molecular evolution of the intracellular targeting of alanine glyoxylate aminotransferase." Thesis, University College London (University of London), 2001. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.272486.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Birdsey, Graeme Miles. "Molecular analysis of the peroxisomal targeting of guinea-pig alanine : glyoxylate aminotransferase." Thesis, University College London (University of London), 1998. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.300508.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Burdin, Dmitry V., Alexey A. Kolobov, Chad Brocker, Alexey A. Soshnev, Nikolay Samusik, Anton v. Demyanov, Silke Brilloff, et al. "Diabetes-linked transcription factor HNF4α regulates metabolism of endogenous methylarginines and β-aminoisobutyric acid by controlling expression of alanine-glyoxylate aminotransferase 2." Nature Publishing Group, 2016. https://tud.qucosa.de/id/qucosa%3A30404.

Full text
Abstract:
Elevated levels of circulating asymmetric and symmetric dimethylarginines (ADMA and SDMA) predict and potentially contribute to end organ damage in cardiovascular diseases. Alanine-glyoxylate aminotransferase 2 (AGXT2) regulates systemic levels of ADMA and SDMA, and also of beta-aminoisobutyric acid (BAIB)-a modulator of lipid metabolism. We identified a putative binding site for hepatic nuclear factor 4 α (HNF4α) in AGXT2 promoter sequence. In a luciferase reporter assay we found a 75% decrease in activity of Agxt2 core promoter after disruption of the HNF4α binding site. Direct binding of HNF4α to Agxt2 promoter was confirmed by chromatin immunoprecipitation assay. siRNA-mediated knockdown of Hnf4a led to an almost 50% reduction in Agxt2 mRNA levels in Hepa 1–6 cells. Liver-specific Hnf4a knockout mice exhibited a 90% decrease in liver Agxt2 expression and activity, and elevated plasma levels of ADMA, SDMA and BAIB, compared to wild-type littermates. Thus we identified HNF4α as a major regulator of Agxt2 expression. Considering a strong association between human HNF4A polymorphisms and increased risk of type 2 diabetes our current findings suggest that downregulation of AGXT2 and subsequent impairment in metabolism of dimethylarginines and BAIB caused by HNF4α deficiency might contribute to development of cardiovascular complications in diabetic patients.
APA, Harvard, Vancouver, ISO, and other styles
4

Burdin, Dmitry V., Alexey A. Kolobov, Chad Brocker, Alexey A. Soshnev, Nikolay Samusik, Anton v. Demyanov, Silke Brilloff, et al. "Diabetes-linked transcription factor HNF4α regulates metabolism of endogenous methylarginines and β-aminoisobutyric acid by controlling expression of alanine-glyoxylate aminotransferase 2." Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2017. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-226882.

Full text
Abstract:
Elevated levels of circulating asymmetric and symmetric dimethylarginines (ADMA and SDMA) predict and potentially contribute to end organ damage in cardiovascular diseases. Alanine-glyoxylate aminotransferase 2 (AGXT2) regulates systemic levels of ADMA and SDMA, and also of beta-aminoisobutyric acid (BAIB)-a modulator of lipid metabolism. We identified a putative binding site for hepatic nuclear factor 4 α (HNF4α) in AGXT2 promoter sequence. In a luciferase reporter assay we found a 75% decrease in activity of Agxt2 core promoter after disruption of the HNF4α binding site. Direct binding of HNF4α to Agxt2 promoter was confirmed by chromatin immunoprecipitation assay. siRNA-mediated knockdown of Hnf4a led to an almost 50% reduction in Agxt2 mRNA levels in Hepa 1–6 cells. Liver-specific Hnf4a knockout mice exhibited a 90% decrease in liver Agxt2 expression and activity, and elevated plasma levels of ADMA, SDMA and BAIB, compared to wild-type littermates. Thus we identified HNF4α as a major regulator of Agxt2 expression. Considering a strong association between human HNF4A polymorphisms and increased risk of type 2 diabetes our current findings suggest that downregulation of AGXT2 and subsequent impairment in metabolism of dimethylarginines and BAIB caused by HNF4α deficiency might contribute to development of cardiovascular complications in diabetic patients.
APA, Harvard, Vancouver, ISO, and other styles
5

Roberts, Thomas Hugh. "Glyoxylate aminotransferases and ureide catabolism in the developing fruits of legumes." Thesis, The University of Sydney, 1994. http://hdl.handle.net/2123/9998.

Full text
Abstract:
The metabolism of glyoxylate released as a result of ureide catabolism in the developing fruits of legumes was examined through an investigation of alanine-, glutamate- and serine-glyoxylate aminotransferase activity in these tissues. A study of the distribution of these activities among the tissues of the developing fruits of two ureide-transporting legumes, French bean (Phaseolus vulgaris) and cowpea (Vigna unguiculata) and an amide-transporter, snow pea (Pisum sativum), was conducted. Serine-glyoxylate aminotransferase activity in each species was very low relative to the alanine- and glutamate-glyoxylate aminotransferase activity. In all three species most of the aminotransferase activity was in the pods. In cowpea, alanine- and glutamate-glyoxylate aminotransferase activity was high in the embryos relative to the seed coats, whereas in French bean and snow pea the seed coats had higher activity than the embryos. Three glyoxylate aminotransferases were isolated and partially purified from extracts of French bean and snow pea developing fruits. These were a serine-glyoxylate aminotransferase (SGAT), which primarily catalyzes the serine glyoxylate aminotransferase reaction; a glutamate-glyoxylate aminotransferase (GGAT), which catalyzes both the glutamate- and alanine-glyoxylate aminotransferase reactions; and an alanine-glyoxylate aminotransferase (AGAT), which catalyzes the alanine-glyoxylate aminotransferase reaction. Sucrose-density gradient ultracentrifugation indicated that the SGAT and GGAT from French bean fruits are located in the peroxisomes, and that the AGAT is located outside the peroxisomes, probably in the mitochondria or chloroplasts. These results confirm the existence of an alanine-glyoxylate aminotransferase (AGAT) as an enzyme distinct from the GGAT. The results of initial velocity kinetic studies of the enzymes isolated from French bean and snow pea, conducted at pH 7.1, were consistent with a Ping Pong reaction mechanism involving pyridoxal phosphate as a co-enzyme. The Km values obtained for the substrates of the enzymes were in all cases higher for the amino acid than for glyoxylate. For the French bean enzymes the Km for the amino acids was about ten-fold higher than that for glyxoylate. The affinity of the snow pea enzymes for glyoxylate was less than that of the French bean enzymes. Alanine--ketoglutarate aminotransferase activity was associated with the GGAT and this activity was reversible, in contrast to the alanine- and glutamate-glyoxylate aminotransferase activity of this enzyme. The reversible aspartate--ketoglutarate aminotransferase activity of the French bean developing fruits was not catalyzed by the glyoxylate aminotransferases. The pH optimum of the GGAT and the SGAT from French bean was approximately 8.3. The molecular weight of the GGAT and AGAT from French bean estimated by gel filtration was 104 kDa. The results are discussed in the context of the metabolic pathway of ureide catabolism, and its location within the fruits of different ureide transporting legumes.
APA, Harvard, Vancouver, ISO, and other styles
6

DINDO, MIRCO. "Molecular analysis of the dimerization and aggregation processes of human alanine:glyoxylate aminotransferase and effect of mutations leading to Primary Hyperoxaluria Type I." Doctoral thesis, 2017. http://hdl.handle.net/11562/960999.

Full text
Abstract:
Primary Hyperoxaluria Type 1 (PH1) is a rare autosomal recessive disorder characterized by the deposition of insoluble calcium oxalate crystals at first in the kidneys and urinary tract and then in the whole body. PH1 is caused by the deficiency of human liver peroxisomal alanine:glyoxylate aminotransferase (AGT). AGT is a pyridoxal 5'-phosphate (PLP)-dependent enzyme, which converts glyoxylate to glycine, thus preventing glyoxylate oxidation to oxalate and calcium oxalate formation. Only two curative therapeutic approaches are currently available for PH1: the administration of pyridoxine (PN), a precursor of PLP, which is only effective in a minority of patients (25- 35%), and liver transplantation, a very invasive procedure. AGT is encoded by the AGXT gene, which is present in humans as two polymorphic forms: the major allele (encoding AGT-Ma) and the minor allele (encoding AGT-Mi). PH1 is a very heterogeneous disease with respect to the clinical manifestations, the response to treatment and the pathogenic mechanisms. In fact, more than 200 pathogenic mutations have been identified so far and the molecular mechanisms by which missense mutations cause AGT deficiency span from functional, to structural and to subcellular localization defects or to a combination of them. Several lines of evidence at both molecular and cellular level, indicate that many disease-causing missense mutations interfere with AGT dimer stability and/or aggregation propensity. However, neither the dimerization nor the aggregation process of AGT have been analyzed in detail. Therefore, we engineered a mutant form of AGT stable in solution in the monomeric form and studied its biochemical properties and dimerization kinetics. We found that monomeric AGT is able to bind PLP and that the coenzyme stabilizes the dimeric structure. Moreover, the identification of key dimerization hot-spots at the monomer-monomer interface allowed us to unravel the mechanisms at the basis of the aberrant mitochondrial mistargeting of two of the most common PH1-causing variants. We also elucidated the molecular and cellular consequences of the pathogenic mutations R36H, G42E, I56N, G63R and G216R, involving residues located at the dimer interface, and tested their in-vitro responsiveness to the treatment with PN. The latter results allowed us to suggest a possible correlation between the structural defect of a variant and its degree of responsiveness to PN. Finally, by combining bioinformatic and biochemical approaches, we analyzed in detail the tendency of AGT to undergo an electrostatically-driven aggregation. We found that the polymorphic changes typical of the minor allele have opposite effect on the aggregation propensity of the protein, and we predicted the possible effect/s of pathogenic mutations of residues located on the AGT surface. Overall, the results obtained allow not only to better understand PH1 pathogenesis, but also to predict the response of the patients to the available therapies as well as to pave the way for the development of new therapeutic strategies.
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "GLYOXYLATE AMINOTRANSFERASE"

1

Danpure, C. J., and P. R. Jennings. "Deficiency of Peroxisomal Alanine: Glyoxylate Aminotransferase in Primary Hyperoxaluria Type 1." In Proceedings in Life Sciences, 374–78. Berlin, Heidelberg: Springer Berlin Heidelberg, 1987. http://dx.doi.org/10.1007/978-3-642-71325-5_40.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Danpure, C. J., and P. R. Jennings. "Enzymatic Heterogeneity in Primary Hyperoxaluria Type 1 (Hepatic Peroxisomal Alanine: Glyoxylate Aminotransferase Deficiency)." In Studies in Inherited Metabolic Disease, 205–7. Dordrecht: Springer Netherlands, 1988. http://dx.doi.org/10.1007/978-94-009-1259-5_32.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Petrarulo, M., S. Pellegrino, M. Marangella, D. Cosseddu, and F. Linari. "Sensitive High-Performance Liquid Chromatographic Microassay for Human Liver L-Glutamate: Glyoxylate Aminotransferase Activity." In Urolithiasis 2, 87. Boston, MA: Springer US, 1994. http://dx.doi.org/10.1007/978-1-4615-2556-1_24.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Wanders, R. J. A., C. W. T. van Roermund, S. Jurriaans, R. B. H. Schutgens, J. M. Tager, H. van den Bosch, E. D. Wolff, et al. "Diversity in Residual Alanine Glyoxylate Aminotransferase Activity in Hyperoxaluria Type I: Correlation with Pyridoxine Responsiveness." In Studies in Inherited Metabolic Disease, 208–11. Dordrecht: Springer Netherlands, 1988. http://dx.doi.org/10.1007/978-94-009-1259-5_33.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Suzuki, Toshiaki, Kozo Nishiyama, Tsuneyoshi Funai, Keiji Tanaka, Akira Ichihara, and Arata Ichiyama. "Energy-Dependent Degration of a Mutant Serine:Pyruvate/Alanin: Glyoxylate Aminotransferase in a Primary Hyperoxaluria Type 1 C." In Intracellular Protein Catabolism, 137–40. Boston, MA: Springer US, 1996. http://dx.doi.org/10.1007/978-1-4613-0335-0_16.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Takada, Yoshikazu, and Tomoo Noguchi. "[35] Aromatic-amino acid-glyoxylate aminotransferase from rat liver." In Methods in Enzymology, 273–79. Elsevier, 1987. http://dx.doi.org/10.1016/s0076-6879(87)42037-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography