Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 173
Filter
Add more filters

Publication year range
1.
J Mol Evol ; 92(3): 266-277, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38683367

ABSTRACT

Ascorbic acid functions as an antioxidant and facilitates other biochemical processes such as collagen triple helix formation, and iron uptake by cells. Animals which endogenously produce ascorbic acid have a functional gulonolactone oxidase gene (GULO); however, humans have a GULO pseudogene (GULOP) and depend on dietary ascorbic acid. In this study, the conservation of GULOP sequences in the primate haplorhini suborder were investigated and compared to the GULO sequences belonging to the primates strepsirrhini suborder. Phylogenetic analysis suggested that the conserved GULOP exons in the haplorhini primates experienced a high rate of mutations following the haplorhini/strepsirrhini divergence. This high mutation rate has decreased during the evolution of the haplorhini primates. Additionally, indels of the haplorhini GULOP sequences were conserved across the suborder. A separate analysis for GULO sequences and well-conserved GULOP sequences focusing on placental mammals identified an in-frame GULO sequence in the Brazilian guinea pig, and a potential GULOP sequence in the pika. Similar to haplorhini primates, the guinea pig and lagomorph species have experienced a high substitution rate when compared to the mammals used in this study. A shared synteny to examine the conservation of local genes near GULO/GULOP identified a conserved inversion around the GULO/GULOP locus between the haplorhini and strepsirrhini primates. Fischer's exact test did not support an association between GULOP and the chromosomal inversion. Mauve alignment showed that the inversion of the length of the syntenic block that the GULO/GULOP genes belonged to was variable. However, there were frequent rearrangements around ~ 2 million base pairs adjacent to GULOP involving the KIF13B and MSRA genes. These data may suggest that genes acquiring deleterious mutations in the coding sequence may respond to these deleterious mutations with rapid substitution rates.


Subject(s)
Chromosome Inversion , Evolution, Molecular , Exons , L-Gulonolactone Oxidase , Mutation , Phylogeny , Primates , Animals , Exons/genetics , Primates/genetics , Mutation/genetics , Humans , L-Gulonolactone Oxidase/genetics , Chromosome Inversion/genetics , Pseudogenes/genetics , Conserved Sequence/genetics
2.
Biol Res ; 57(1): 26, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735981

ABSTRACT

BACKGROUND: Vitamin C (ascorbate) is a water-soluble antioxidant and an important cofactor for various biosynthetic and regulatory enzymes. Mice can synthesize vitamin C thanks to the key enzyme gulonolactone oxidase (Gulo) unlike humans. In the current investigation, we used Gulo-/- mice, which cannot synthesize their own ascorbate to determine the impact of this vitamin on both the transcriptomics and proteomics profiles in the whole liver. The study included Gulo-/- mouse groups treated with either sub-optimal or optimal ascorbate concentrations in drinking water. Liver tissues of females and males were collected at the age of four months and divided for transcriptomics and proteomics analysis. Immunoblotting, quantitative RT-PCR, and polysome profiling experiments were also conducted to complement our combined omics studies. RESULTS: Principal component analyses revealed distinctive differences in the mRNA and protein profiles as a function of sex between all the mouse cohorts. Despite such sexual dimorphism, Spearman analyses of transcriptomics data from females and males revealed correlations of hepatic ascorbate levels with transcripts encoding a wide array of biological processes involved in glucose and lipid metabolisms as well as in the acute-phase immune response. Moreover, integration of the proteomics data showed that ascorbate modulates the abundance of various enzymes involved in lipid, xenobiotic, organic acid, acetyl-CoA, and steroid metabolism mainly at the transcriptional level, especially in females. However, several proteins of the mitochondrial complex III significantly correlated with ascorbate concentrations in both males and females unlike their corresponding transcripts. Finally, poly(ribo)some profiling did not reveal significant enrichment difference for these mitochondrial complex III mRNAs between Gulo-/- mice treated with sub-optimal and optimal ascorbate levels. CONCLUSIONS: Thus, the abundance of several subunits of the mitochondrial complex III are regulated by ascorbate at the post-transcriptional levels. Our extensive omics analyses provide a novel resource of altered gene expression patterns at the transcriptional and post-transcriptional levels under ascorbate deficiency.


Subject(s)
Ascorbic Acid , Liver , Proteomics , Animals , Ascorbic Acid/metabolism , Liver/metabolism , Liver/drug effects , Female , Male , Mice , L-Gulonolactone Oxidase/genetics , L-Gulonolactone Oxidase/metabolism , Gene Expression Profiling , Transcriptome , Principal Component Analysis , Antioxidants/metabolism
3.
Plant Mol Biol ; 110(3): 287-300, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35918559

ABSTRACT

KEY MESSAGE: Association genetic analysis empowered us to identify candidate genes underlying natural variation of morpho-physiological, antioxidants, and grain yield-related traits in barley. Novel intriguing genomic regions were identified and dissected. Salinity stress is one of the abiotic stresses that influence the morpho-physiological, antioxidants, and yield-related traits in crop plants. The plants of a core set of 138 diverse barley accessions were analyzed after exposure to salt stress under field conditions during the reproductive phase. A genome-wide association scan (GWAS) was then conducted using 19,276 single nucleotide polymorphisms (SNPs) to uncover the genetic basis of morpho-physiological and grain-related traits. A wide range of responses to salt stress by the accessions was explored in the current study. GWAS detected 263 significantly associated SNPs with the antioxidants, K+/Na+ content ratio, and agronomic traits. Five genomic regions harbored interesting putative candidate genes within LD ± 1.2 Mbp. Choromosome 2H harbored many candidate genes associated with the antioxidants ascorbic acid (AsA) and glutathione (GSH), such as superoxide dismutase (SOD), ascorbate peroxidase (APX), and glutathione reductase (GR), under salt stress. Markedly, an A:C SNP at 153,773,211 bp on chromosome 7H is located inside the gene HORVU.MOREX.r3.7HG0676830 (153,772,300-153,774,057 bp) that was annotated as L-gulonolactone oxidase, regulating the natural variation of SOD_S and APX_S. The allelic variation at this SNP reveals a negative selection of accessions carrying the C allele, predominantly found in six-rowed spring landraces originating from Far-, Near-East, and central Asia carrying photoperiod sensitive alleles having lower activity of enzymatic antioxidants. The SNP-trait associations detected in the current study constitute a benchmark for developing molecular selection tools for antioxidant compound selection in barley.


Subject(s)
Hordeum , Antioxidants , Ascorbate Peroxidases/genetics , Ascorbic Acid , Edible Grain/genetics , Genome-Wide Association Study , Glutathione , Glutathione Reductase/genetics , Hordeum/genetics , L-Gulonolactone Oxidase/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Salt Stress/genetics , Superoxide Dismutase/genetics
4.
Hum Mol Genet ; 29(9): 1476-1488, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32307537

ABSTRACT

Arterial tortuosity syndrome (ATS) is a recessively inherited connective tissue disorder, mainly characterized by tortuosity and aneurysm formation of the major arteries. ATS is caused by loss-of-function mutations in SLC2A10, encoding the facilitative glucose transporter GLUT10. Former studies implicated GLUT10 in the transport of dehydroascorbic acid, the oxidized form of ascorbic acid (AA). Mouse models carrying homozygous Slc2a10 missense mutations did not recapitulate the human phenotype. Since mice, in contrast to humans, are able to intracellularly synthesize AA, we generated a novel ATS mouse model, deficient for Slc2a10 as well as Gulo, which encodes for L-gulonolactone oxidase, an enzyme catalyzing the final step in AA biosynthesis in mouse. Gulo;Slc2a10 double knock-out mice showed mild phenotypic anomalies, which were absent in single knock-out controls. While Gulo;Slc2a10 double knock-out mice did not fully phenocopy human ATS, histological and immunocytochemical analysis revealed compromised extracellular matrix formation. Transforming growth factor beta signaling remained unaltered, while mitochondrial function was compromised in smooth muscle cells derived from Gulo;Slc2a10 double knock-out mice. Altogether, our data add evidence that ATS is an ascorbate compartmentalization disorder, but additional factors underlying the observed phenotype in humans remain to be determined.


Subject(s)
Arteries/abnormalities , Ascorbic Acid Deficiency/genetics , Glucose Transport Proteins, Facilitative/genetics , Joint Instability/genetics , L-Gulonolactone Oxidase/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , Animals , Arteries/metabolism , Arteries/pathology , Ascorbic Acid/biosynthesis , Ascorbic Acid/genetics , Ascorbic Acid Deficiency/metabolism , Ascorbic Acid Deficiency/pathology , Disease Models, Animal , Homozygote , Humans , Joint Instability/metabolism , Joint Instability/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Respiration/genetics , Signal Transduction/genetics , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology , Vascular Malformations/metabolism , Vascular Malformations/pathology
5.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36142515

ABSTRACT

Reactive oxygen species (ROS), which are exceptionally high in IBD lesions, are known to cause abnormal immune responses to inflammatory reactions in inflammatory bowel diseases (IBD) through damage to the intestinal mucosal linings. Moreover, they are theorized to be an agent of IBD development. Vitamin C is widely known to be an effective antioxidant for its ability to regulate inflammatory responses through its ROS scavenging effect. Therefore, we examined vitamin C's influence on the development and progression of IBD in Gulo(-/-) mice, which cannot synthesize vitamin C like humans due to a defect in the expression of L-gulono-γ-lactone oxidase, an essential enzyme for vitamin C production. First, we found extensive oxidative stress and an inflammation increase in the colon of vitamin C-insufficient Gulo(-/-) mice. We also found decreased IL-22 production and NKp46(+) cell recruitment and the impaired activation of the p38MAPK pathway. Additionally, comparing vitamin C-insufficient Gulo(-/-) mice to vitamin C-sufficient Gulo(-/-) mice and wild-type mice, the insufficient group faced a decrease in mucin-1 expression, accompanied by an increase in IL-6 production, followed by the activation of the STAT3 and Akt pathways. The results suggest that vitamin C insufficiency induces severe colitis, meaning vitamin C could also take on a preventative role by regulating the production of cytokines and the induction of inflammation.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Mustelidae , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Colitis/pathology , Cytokines , Dextran Sulfate/toxicity , Humans , Inflammation , Interleukin-6/adverse effects , Interleukins , L-Gulonolactone Oxidase , Mice , Mice, Inbred C57BL , Mucin-1 , Mustelidae/metabolism , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species/metabolism , Vitamins , Interleukin-22
6.
J Proteome Res ; 20(11): 5036-5053, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34643398

ABSTRACT

A suboptimal blood vitamin C (ascorbate) level increases the risk of several chronic diseases. However, the detection of hypovitaminosis C is not a simple task, as ascorbate is unstable in blood samples. In this study, we examined the serum proteome of mice lacking the gulonolactone oxidase (Gulo) required for the ascorbate biosynthesis. Gulo-/- mice were supplemented with different concentrations of ascorbate in drinking water, and serum was collected to identify proteins correlating with serum ascorbate levels using an unbiased label-free liquid chromatography-tandem mass spectrometry global quantitative proteomic approach. Parallel reaction monitoring was performed to validate the correlations. We uncovered that the serum proteome profiles differ significantly between male and female mice. Also, unlike Gulo-/- males, a four-week ascorbate treatment did not entirely re-establish the serum proteome profile of ascorbate-deficient Gulo-/- females to the optimal profile exhibited by Gulo-/- females that never experienced an ascorbate deficiency. Finally, the serum proteins involved in retinoid metabolism, cholesterol, and lipid transport were similarly affected by ascorbate levels in males and females. In contrast, the proteins regulating serum peptidases and the protein of the acute phase response were different between males and females. These proteins are potential biomarkers correlating with blood ascorbate levels and require further study in standard clinical settings. The complete proteomics data set generated in this study has been deposited to the public repository ProteomeXchange with the data set identifier: PXD027019.


Subject(s)
Ascorbic Acid , Proteome , Animals , Dietary Supplements , Female , L-Gulonolactone Oxidase/metabolism , Male , Mice , Proteome/genetics , Proteomics
7.
J Biol Chem ; 295(47): 15870-15882, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32913121

ABSTRACT

Ascorbic acid, a water-soluble antioxidant, regulates various biological processes and is thought to influence cholesterol. However, little is known about the mechanisms underpinning ascorbic acid-mediated cholesterol metabolism. Here, we determined if ascorbic acid can regulate expression of proprotein convertase subtilisin/kexin 9 (PCSK9), which binds low-density lipoprotein receptor (LDLR) leading to its intracellular degradation, to influence low-density lipoprotein (LDL) metabolism. At cellular levels, ascorbic acid inhibited PCSK9 expression in HepG2 and Huh7 cell lines. Consequently, LDLR expression and cellular LDL uptake were enhanced. Similar effects of ascorbic acid on PCSK9 and LDLR expression were observed in mouse primary hepatocytes. Mechanistically, ascorbic acid suppressed PCSK9 expression in a forkhead box O3-dependent manner. In addition, ascorbic acid increased LDLR transcription by regulating sterol regulatory element-binding protein 2. In vivo, administration of ascorbic acid reduced serum PCSK9 levels and enhanced liver LDLR expression in C57BL/6J mice. Reciprocally, lack of ascorbic acid supplementation in L-gulono-γ-lactone oxidase deficient (Gulo-/-) mice increased circulating PCSK9 and LDL levels, and decreased liver LDLR expression, whereas ascorbic acid supplementation decreased PCSK9 and increased LDLR expression, ameliorating LDL levels in Gulo-/- mice fed a high fat diet. Moreover, ascorbic acid levels were negatively correlated to PCSK9, total and LDL levels in human serum samples. Taken together, these findings suggest that ascorbic acid reduces PCSK9 expression, leading to increased LDLR expression and cellular LDL uptake. Thus, supplementation of ascorbic acid may ameliorate lipid profiles in ascorbic acid-deficient species.


Subject(s)
Ascorbic Acid/pharmacology , Gene Expression Regulation/drug effects , Proprotein Convertase 9/biosynthesis , Receptors, LDL/biosynthesis , Animals , Hep G2 Cells , Humans , L-Gulonolactone Oxidase/genetics , L-Gulonolactone Oxidase/metabolism , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout , Proprotein Convertase 9/genetics , Receptors, LDL/genetics
8.
BMC Evol Biol ; 19(1): 126, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31215418

ABSTRACT

BACKGROUND: L-ascorbate (Vitamin C) is an important antioxidant and co-factor in eukaryotic cells, and in mammals it is indispensable for brain development and cognitive function. Vertebrates usually become L-ascorbate auxothrophs when the last enzyme of the synthetic pathway, an L-gulonolactone oxidase (GULO), is lost. Since Protostomes were until recently thought not to have a GULO gene, they were considered to be auxothrophs for Vitamin C. RESULTS: By performing phylogenetic analyses with tens of non-Bilateria and Protostomian genomes, it is shown, that a GULO gene is present in the non-Bilateria Placozoa, Myxozoa (here reported for the first time) and Anthozoa groups, and in Protostomians, in the Araneae family, the Gastropoda class, the Acari subclass (here reported for the first time), and the Priapulida, Annelida (here reported for the first time) and Brachiopoda phyla lineages. GULO is an old gene that predates the separation of Animals and Fungi, although it could be much older. We also show that within Protostomes, GULO has been lost multiple times in large taxonomic groups, namely the Pancrustacea, Nematoda, Platyhelminthes and Bivalvia groups, a pattern similar to that reported for Vertebrate species. Nevertheless, we show that Drosophila melanogaster seems to be capable of synthesizing L-ascorbate, likely through an alternative pathway, as recently reported for Caenorhabditis elegans. CONCLUSIONS: Non-Bilaterian and Protostomians seem to be able to synthesize Vitamin C either through the conventional animal pathway or an alternative pathway, but in this animal group, not being able to synthesize L-ascorbate seems to be the exception rather than the rule.


Subject(s)
Ascorbic Acid/metabolism , Eukaryota/enzymology , Eukaryota/genetics , Evolution, Molecular , L-Gulonolactone Oxidase/genetics , Animals , Drosophila melanogaster/genetics , Eukaryota/classification , Eukaryota/metabolism , Genome , L-Gulonolactone Oxidase/chemistry , L-Gulonolactone Oxidase/metabolism , Models, Molecular , Phylogeny , Vertebrates/classification , Vertebrates/genetics
9.
Biochem Biophys Res Commun ; 504(4): 885-891, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30219227

ABSTRACT

During kidney development, the balance between self-renewal and differentiation of metanephric mesenchyme (MM) cells, mainly regulated by Sine oculis-related homeobox 2 (Six2), is critical for forming mature kidney. L-gulono-γ-lactone oxidase (Gulo), a crucial enzyme for vitamin C synthesis, reveals a different expression at various stages during kidney development, but its function in the early renal development remains unknown. In this work, we aim to study the role of Gulo in MM cells at two differentiation stages. We found that Gulo expression in undifferentiated MM (mK3) cells was lower than in differentiated MM (mK4) cells. Over-expression of Gulo can promote mesenchymal-to-epithelial transformation (MET) and apoptosis and inhibit the proliferation in mK3 cells. Knock-down of Gulo in mK4 cells made its epithelial character cells unstabilized, facilitated the proliferation and restrained the apoptosis. Furthermore, we found that Six2 was negatively regulated by Gulo, and over-expression or knock-down of Six2 was able to rescue partially the MET, proliferation and apoptosis of MM cells caused by Gulo. In conclusion, these findings reveal that Gulo promotes the MET and apoptosis, and inhibits proliferation in MM cells by down-regulating Six2.


Subject(s)
Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , L-Gulonolactone Oxidase/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Transcription Factors/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Kidney/cytology , Kidney/embryology , L-Gulonolactone Oxidase/genetics , Mice , Transcription Factors/genetics
10.
Mediators Inflamm ; 2017: 4024672, 2017.
Article in English | MEDLINE | ID: mdl-28210072

ABSTRACT

Cellular immunosuppression appears to be involved in sepsis and sepsis-induced multiple organ dysfunction syndrome (MODS). Recent evidence showed that parenteral vitamin C (Vit C) had the ability to attenuate sepsis and sepsis-induced MODS. Herein, we investigated the impact of parenteral Vit C on cellular immunosuppression and the therapeutic value in sepsis. Using cecal ligation and puncture (CLP), sepsis was induced in WT and Gulo-/- mice followed with 200 mg/Kg parenteral Vit C administration. The immunologic functions of CD4+CD25+ regulatory T cells (Tregs) and CD4+CD25- T cells, as well as the organ functions, were determined. Administration of parenteral Vit C per se markedly improved the outcome of sepsis and sepsis-induced MODS of WT and Gulo-/- mice. The negative immunoregulation of Tregs was inhibited, mainly including inhibiting the expression of forkhead helix transcription factor- (Foxp-) 3, cytotoxic T lymphocyte associated antigen- (CTLA-) 4, membrane associated transforming growth factor-ß (TGF-ßm+), and the secretion of inhibitory cytokines [including TGF-ß and interleukin- (IL-) 10], as well as CD4+ T cells-mediated cellular immunosuppression which was improved by parenteral Vit C in WT and Gulo-/- septic mice. These results suggested that parenteral Vit C has the ability to improve the outcome of sepsis and sepsis-induced MODS and is associated with improvement in cellular immunosuppression.


Subject(s)
Ascorbic Acid/therapeutic use , Multiple Organ Failure/drug therapy , Sepsis/drug therapy , Animals , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/metabolism , Forkhead Transcription Factors/metabolism , Immunosuppression Therapy , Interleukin-10/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , L-Gulonolactone Oxidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Sepsis/immunology , Sepsis/metabolism , Transforming Growth Factor beta/metabolism
11.
Kidney Blood Press Res ; 41(6): 794-801, 2016.
Article in English | MEDLINE | ID: mdl-27832650

ABSTRACT

BACKGROUNDS/AIMS: Vitamin C is an antioxidant and acts as a cofactor for several key enzymatic catalytic reactions in animals. Amphibians produce vitamin C in their kidneys, as opposed to mammals that produce vitamin C in their liver. Gulo serves as a crucial enzyme for vitamin C synthesis in mammals, but the characteristics and localization of its homologous genes during kidney development in Xenopus laevis, an amphibian, remains unknown. METHODS: We aligned amino acid sequences of Gulo across different species by using bioinformatics methods and detected patterns of expression for Gulo during kidney development by using RT-PCR and in situ hybridization. RESULTS: We identified a new site on the X. laevis genome, LOC495407. Sequence alignment analysis indicated this fragment is highly conserved and homologous to gulo genes in mammals. RT-PCR and in situ hybridization results reveal that X. laevis gulo is maternally expressed during the early stages of embryonic development, particularly, in the tubules of the pronephros from the middle tail-bud stage and onward in embryos. CONCLUSION: Gulo is a novel specific marker for pronephros tubules in X. laevis, and may be used as a potential marker for kidney development studies and disease diagnosis in mammals.


Subject(s)
Kidney Tubules/growth & development , L-Gulonolactone Oxidase/analysis , Pronephros/growth & development , Animals , Biomarkers/analysis , Female , Kidney Tubules/embryology , Kidney Tubules/enzymology , Mammals , Pronephros/embryology , Pronephros/enzymology , Sequence Alignment , Xenopus laevis
12.
IUBMB Life ; 67(2): 69-76, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25865952

ABSTRACT

Biosynthesis of ascorbate is known to occur in liver and/or kidney of some vertebrates; however, a recent study discovered the expression of l-gulono-γ-lactone oxidase, an enzyme essential for ascorbate synthesis, in the brain of the African lungfish, Protopterus annectens. This report provides an up-to-date review on ascorbate synthesis in fishes and the possible future directions of study in view of the discovery of the unusual site of ascorbate biosynthesis.


Subject(s)
Ascorbic Acid/biosynthesis , Fishes/metabolism , L-Gulonolactone Oxidase/metabolism , Animals , Ascorbic Acid/metabolism , Brain/metabolism , Kidney/metabolism , Liver/metabolism , Vertebrates/metabolism
13.
FASEB J ; 28(8): 3506-17, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24769670

ABSTRACT

This study aimed to test the hypothesis that the brain of Protopterus annectens expressed L-gulono-γ-lactone oxidase (gulo/Gulo), the enzyme catalyzing the last step of ascorbate biosynthesis, and could maintain high concentrations of ascorbate during estivation. We cloned and sequenced gulo from the kidney of P. annectens and performed quantitative PCR to determine its mRNA expression in kidney and brain. Gulo activity was assayed and its protein abundance was determined by Western blot using custom-made anti-Gulo antibody. Effects of estivation on concentrations of ascorbate and dehydroascorbate in the kidney and brain were also determined. Both brain and kidney, but not liver, of P. annectens expressed gulo/Gulo. Desiccation induced P. annectens to estivate, and 6 mo of estivation led to drastic decreases in gulo/Gulo expression and ascorbate concentration in the kidney. However, high concentrations of ascorbate and ascorbate + dehydroascorbate were maintained in the brain during estivation, probably resulting from in situ ascorbate synthesis. Control fish were placed in freshwater, where they were fully active in a favorable environment unlike estivation on land. The ability to synthesize ascorbate to ameliorate oxidative stress directly in the brain might contribute to the ability of P. annectens to undergo prolonged estivation on land.


Subject(s)
Ascorbic Acid/biosynthesis , Brain/enzymology , Estivation/physiology , Fishes/physiology , Kidney/enzymology , L-Gulonolactone Oxidase/biosynthesis , Amino Acid Sequence , Animals , Body Water , Conserved Sequence , L-Gulonolactone Oxidase/genetics , Molecular Sequence Data , Organ Specificity , Oxidative Stress , Phylogeny , RNA, Messenger/biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid
14.
Nucleic Acids Res ; 41(Web Server issue): W29-33, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23609542

ABSTRACT

The Basic Local Alignment Search Tool (BLAST) website at the National Center for Biotechnology (NCBI) is an important resource for searching and aligning sequences. A new BLAST report allows faster loading of alignments, adds navigation aids, allows easy downloading of subject sequences and reports and has improved usability. Here, we describe these improvements to the BLAST report, discuss design decisions, describe other improvements to the search page and database documentation and outline plans for future development. The NCBI BLAST URL is http://blast.ncbi.nlm.nih.gov.


Subject(s)
Sequence Alignment/methods , Software , Animals , Genomics , Internet , L-Gulonolactone Oxidase/genetics , Rats
15.
Article in English | MEDLINE | ID: mdl-25645296

ABSTRACT

Intense temperature change often leads to increased oxidative stress in many animals with a few exceptions, including the turtle. To date, little is known about the mechanism of protective antioxidative defenses in turtles during acute temperature change, specifically the role that the antioxidant ascorbic acid (AA) plays. In this study, Chinese soft-shelled turtles (Pelodiscus sinensis) were initially acclimated at 28°C (3 wks), exposed to acute cold condition (8°C, 8 h) and finally placed in recovery (28°C, 24 h). L-Gulonolactone oxidase (GLO) mRNA exhibited a stable transcription pattern during the intense thermal fluctuation. GLO activity also remained stable, which validated the mRNA expression pattern. The similar Q10 values for GLO activity in the different treatment groups at incubation temperatures of 28°C and 8°C indicated that the GLO activity response to thermal change exhibited a temperature-dependent enzymatic kinetic characteristic. The AA storage was tissue-specific as well as the AA re-supply in the recovery period, with brain as the priority. Despite the insufficient transport during cold exposure, the plasma AA reservoir greatly contributed to the redistribution of AA during recovery. Depending on the prominent GLO activity, the high level of tissue-specific AA storage and the extraordinary plasma AA transport potential, the Chinese soft-shelled turtle endured severe thermal fluctuations with no apparent oxidative stress. However, the significant decrease in AA concentration in the brain tissue during acute cold exposure suggested that such a strategy may not be sufficient for prolonged cold exposure.


Subject(s)
Ascorbic Acid/physiology , Cold Temperature , Stress, Physiological , Turtles/physiology , Animals , Gene Expression , L-Gulonolactone Oxidase/genetics , L-Gulonolactone Oxidase/metabolism , Oxidative Stress , RNA, Messenger/genetics
16.
Int J Biol Macromol ; 268(Pt 1): 131733, 2024 May.
Article in English | MEDLINE | ID: mdl-38649080

ABSTRACT

Up to now, it has been believed that invertebrates are unable to synthesize ascorbic acid (AA) in vivo. However, in the present study, the full-length CDs (Coding sequence) of L-gulonolactone oxidase (GLO) from Pacific abalone (Haliotis discus hannai Ino) were obtained through molecular cloning. The Pacific abalone GLO contained a FAD-binding domain in the N-termination, and ALO domain and conserved HWAK motif in the C-termination. The GLO gene possesses 12 exons and 11 introns. The Pacific abalone GLO was expressed in various tissues, including the kidney, digestive gland, gill, intestine, muscle and mantle. The GLO activity assay revealed that GLO activity was only detected in the kidney of Pacific abalone. After a 100-day feeding trial, dietary AA levels did not significantly affect the survival, weight gain, daily increment in shell length, and feed conversion ratio of Pacific abalone. The expression of GLO in the kidney was downregulated by dietary AA. These results implied that the ability to synthesize AA in abalone had not been lost. From the evolutionary perspective, the loss of GLO occurred independently as an independent event by matching with the genomes of various species. The positive selection analysis revealed that the GLO gene underwent purifying selective pressure during its evolution. In conclusion, the present study provided direct evidence to prove that the GLO activity and the ability to synthesize AA exist in abalone. The AA synthesis ability in vertebrates might have originated from invertebrates dating back 930.31 million years.


Subject(s)
Ascorbic Acid , Gastropoda , L-Gulonolactone Oxidase , Animals , Ascorbic Acid/biosynthesis , Ascorbic Acid/metabolism , Gastropoda/genetics , Gastropoda/enzymology , L-Gulonolactone Oxidase/genetics , L-Gulonolactone Oxidase/metabolism , Phylogeny , Amino Acid Sequence , Cloning, Molecular , Evolution, Molecular
17.
J Neurochem ; 124(3): 363-75, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23106783

ABSTRACT

Severe vitamin C deficiency (ascorbic acid; AA) was induced in gulo-/- mice incapable of synthesizing their own AA. A number of behavioral measures were studied before and during the deprivation period, including a scorbutic period, during which weight loss was observed in the mice. Mice were then resuscitated with AA supplements. During the scorbutic period, gulo-/- mice showed decreased voluntary locomotor activity, diminished physical strength, and increased preference for a highly palatable sucrose reward. These behaviors all returned to control levels following resuscitation. Altered trial times in subordinate mice in the tube test for social dominance in the AA-deprived mice persisted following resuscitation and may signify a depressive-like behavior in these mice. Biochemical analyses were undertaken following a second deprivation period. AA deficiency was accompanied by decreased blood glucose levels, oxidative damage to lipids and proteins in the cortex, and decreases in dopamine and serotonin metabolites in both the cortex and striatum. Given the reasonably high proportions of the population that do not consume sufficient AA in the diet, these data have important implications for physical and psychological function in the general population.


Subject(s)
Ascorbic Acid Deficiency/physiopathology , Biogenic Monoamines/metabolism , Severity of Illness Index , Animals , Ascorbic Acid/genetics , Ascorbic Acid/metabolism , Ascorbic Acid Deficiency/drug therapy , Ascorbic Acid Deficiency/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Female , L-Gulonolactone Oxidase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Blood ; 117(20): 5485-93, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21346252

ABSTRACT

Prolyl-4-hydroxylation is necessary for proper structural assembly of collagens and oxygen-dependent protein stability of hypoxia-inducible transcription factors (HIFs). In vitro function of HIF prolyl-4-hydroxylase domain (PHD) enzymes requires oxygen and 2-oxoglutarate as cosubstrates with iron(II) and vitamin C serving as cofactors. Although vitamin C deficiency is known to cause the collagen-disassembly disease scurvy, it is unclear whether cellular oxygen sensing is similarly affected. Here, we report that vitamin C-deprived Gulo(-/-) knockout mice show normal HIF-dependent gene expression. The systemic response of Gulo(-/-) animals to inspiratory hypoxia, as measured by plasma erythropoietin levels, was similar to that of animals supplemented with vitamin C. Hypoxic HIF induction was also essentially normal under serum- and vitamin C-free cell-culture conditions, suggesting that vitamin C is not required for oxygen sensing in vivo. Glutathione was found to fully substitute for vitamin C requirement of all 3 PHD isoforms in vitro. Consistently, glutathione also reduced HIF-1α protein levels, transactivation activity, and endogenous target gene expression in cells exposed to CoCl(2). A Cys201Ser mutation in PHD2 increased basal hydroxylation rates and conferred resistance to oxidative damage in vitro, suggesting that this surface-accessible PHD2 cysteine residue is a target of antioxidative protection by vitamin C and glutathione.


Subject(s)
Ascorbic Acid/metabolism , Oxygen/metabolism , Amino Acid Substitution , Animals , Ascorbic Acid Deficiency/metabolism , Cell Hypoxia , Cell Line , Cobalt/pharmacology , Glutathione/metabolism , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism
19.
Nutr Neurosci ; 16(4): 160-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23321552

ABSTRACT

OBJECTIVES: Key antioxidants, vitamins C and E, are necessary for normal brain development and neuronal function. In this study, we depleted both of these vitamins in two mouse models to determine if oxidative stress due to combined vitamin C and E dietary deficiency altered their neurological phenotype. The first model lacked both alleles for the Gulonolactone oxidase gene (Gulo(-/-)) and therefore was unable synthesize vitamin C. To obtain an additional cellular deficiency of vitamin C, the second model also lacked one allele for the cellular vitamin C transporter gene (Gulo(-/-)/SVCT2(+/-)). METHODS: The experimental treatment was 16 weeks of vitamin E deprivation followed by 3 weeks of vitamin C deprivation. Mice were assessed for motor coordination deficits, vitamin levels, and oxidative stress biomarkers. RESULTS: In the first model, defects in motor performance were more apparent in both vitamin C-deficient groups (VE+VC-, VE-VC-) compared to vitamin C-supplemented groups (VE+VC+, VE-VC+) regardless of vitamin E level. Analysis of brain cortex and liver confirmed decreases of at least 80% for each vitamin in mice on deficient diets. Vitamin E deficiency doubled oxidative stress biomarkers (F2-isoprostanes and malondialdehyde). In the second model, Gulo(-/-)/SVCT2(+/-) mice on the doubly deficient diets showed deficits in locomotor activity, Rota-rod performance, and other motor tasks, with no concomitant change in anxiety or spatial memory. DISCUSSION: Vitamin E deficiency alone caused a modest oxidative stress in brain that did not affect motor performance. Adding a cellular deficit in vitamin C to dietary deprivation of both vitamins significantly impaired motor performance.


Subject(s)
Ascorbic Acid/administration & dosage , Dietary Supplements , Psychomotor Performance/drug effects , Vitamin D Deficiency/pathology , Vitamin E Deficiency/pathology , Vitamin E/administration & dosage , Animals , Antioxidants/administration & dosage , Ascorbic Acid/blood , Biomarkers/blood , Brain/drug effects , Brain/metabolism , Disease Models, Animal , F2-Isoprostanes/blood , Female , L-Gulonolactone Oxidase/genetics , L-Gulonolactone Oxidase/metabolism , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/blood , Mice , Mice, Knockout , Oxidative Stress/drug effects , Vitamin D Deficiency/complications , Vitamin E/blood , Vitamin E Deficiency/complications
20.
Biol Pharm Bull ; 36(12): 2005-8, 2013.
Article in English | MEDLINE | ID: mdl-24292059

ABSTRACT

Senescence marker protein-30 (SMP30) was first described as a physiologic entity that decreases in the rat liver and kidney with aging. Previously, we established that SMP30 is the lactone-hydrolyzing enzyme gluconolactonase (GNL), which is involved in ascorbic acid (AA) biosynthesis. In the present study, we found SMP30/GNL mRNA expressed in the mouse ovary. To ascertain the reason for ovarian SMP30/GNL expression, we examined mice during gestation. SMP30/GNL mRNA expression was evident at the start of gestation, increased for the next eight days then decreased rapidly. Moreover, L-gulono-γ-lactone oxidase (Gulo) mRNA, which catalyzes the last step of AA, was found in the ovaries of these mice. The variations of these genes' expression showed an inverse pattern to that of Cyp19a1 (aromatase) mRNA expression. Therefore, the SMP30/GNL and Gulo mRNA expression might be regulated by estrogen levels in the ovary. Since the presence of both SMP30/GNL and Gulo mRNAs could indicate that AA synthesis occurs in the ovary, we quantified AA levels in mouse ovaries during gestation. However, no correlation was found between changes of AA content and SMP30/GNL or Gulo mRNAs expression at this site. Moreover, we compared the changes of AA content during gestation between wild-type and SMP30/GNL knockout mice, which cannot synthesize AA, and found no significant differences between them. These results indicated that, although AA synthesis might occur in the ovaries, the amount of AA which is synthesized in ovaries must be quite low and insufficient to influence the AA content in ovary.


Subject(s)
Ascorbic Acid/metabolism , Calcium-Binding Proteins/genetics , Carboxylic Ester Hydrolases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Ovary/metabolism , Animals , Aromatase/genetics , Brain/metabolism , Female , Kidney/metabolism , L-Gulonolactone Oxidase/genetics , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Pregnancy/metabolism , RNA, Messenger/metabolism , Testis/metabolism , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL