Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Biomol Struct Dyn ; 41(24): 15124-15136, 2023.
Article in English | MEDLINE | ID: mdl-36883880

ABSTRACT

Diabetes, the cause of colossal economic and disease burden, is a key area of research in drug discovery programs. Elevated blood glucose levels in diabetes lead to several adverse consequences due to the formation of advanced glycation end products and free radicals. Vitamin C, a potent antioxidant, protects the body's cells and tissues from oxidative damage and dysfunctions. Glucose is the precursor of Vitamin C synthesis in plants and some mammals. L-gulono lactone oxidase (GULO) is the rate-limiting enzyme in producing Vitamin C. However, it is not synthesized in bats, primates, humans, and guinea pigs because of the pseudogene. Several phytomolecules having antioxidant properties are hypothesized to be promising and selective activators of GULO. Therefore, the present study focused on screening agonists of GULO from phytomolecules as an effective augmentor for Vitamin C synthesis, thereby suppressing the sequela of diabetic events. The 3D structure of GULO was generated by the ab-initio method. Subsequently, molecular docking explored the possible binding patterns of GULO protein with different plant phenolic compounds, followed by supplementation of the potent phytomolecules to diabetic guinea pigs. It is noteworthy that Resveratrol and Hydroxytyrosol showed better binding affinity. The molecular simulation also confirmed that Resveratrol is an activator of the GULO enzyme. Interestingly, it was also established that Vitamin C levels were improved in diabetic guinea pigs supplemented with the phytomolecules and comparatively Resveratrol modulates the concentration of glucose and Vitamin C levels substantially, thereby alleviating hyperglycemia. However, further studies are warranted to study the mechanisms.Communicated by Ramaswamy H. Sarma.


Subject(s)
Diabetes Mellitus , Mustelidae , Humans , Animals , Guinea Pigs , Antioxidants/pharmacology , Resveratrol , Molecular Docking Simulation , L-Gulonolactone Oxidase , Ascorbic Acid , Glucose
2.
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
3.
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
4.
Aging (Albany NY) ; 8(3): 458-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26922388

ABSTRACT

Suboptimal intake of dietary vitamin C (ascorbate) increases the risk of several chronic diseases but the exact metabolic pathways affected are still unknown. In this study, we examined the metabolic profile of mice lacking the enzyme gulonolactone oxidase (Gulo) required for the biosynthesis of ascorbate. Gulo-/- mice were supplemented with 0%, 0.01%, and 0.4% ascorbate (w/v) in drinking water and serum was collected for metabolite measurements by targeted mass spectrometry. We also quantified 42 serum cytokines and examined the levels of different stress markers in liver. The metabolic profiles of Gulo-/- mice treated with ascorbate were different from untreated Gulo-/- and normal wild type mice. The cytokine profiles of Gulo-/-mice, in return, overlapped the profile of wild type animals upon 0.01% or 0.4% vitamin C supplementation. The life span of Gulo-/- mice increased with the amount of ascorbate in drinking water. It also correlated significantly with the ratios of serum arginine/lysine, tyrosine/phenylalanine, and the ratio of specific species of saturated/unsaturated phosphatidylcholines. Finally, levels of hepatic phosphorylated endoplasmic reticulum associated stress markers IRE1α and eIF2α correlated inversely with serum ascorbate and life span suggesting that vitamin C modulates endoplasmic reticulum stress response and longevity in Gulo-/- mice.


Subject(s)
Antioxidants/administration & dosage , Ascorbic Acid Deficiency/blood , Ascorbic Acid/administration & dosage , Longevity/drug effects , Metabolome , Amino Acids/blood , Animals , Ascorbic Acid Deficiency/drug therapy , Body Weight/drug effects , Cytokines/blood , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Hormones/blood , L-Gulonolactone Oxidase/genetics , Male , Membrane Lipids/blood , Mice , Mice, Knockout , Mitochondria, Liver/drug effects , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
5.
Antioxid Redox Signal ; 23(16): 1270-83, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-25977985

ABSTRACT

AIMS: The developing brain of a neonate is particularly susceptible to damage by vitamin C deficiency because of its rapid growth and immature antioxidant system. Cognitive impairment and sensory motor deficits are found in the adult brain upon vitamin C deficiency. Therefore, the aim of this study was to clarify the role of vitamin C in its own right and its related mechanisms in Gulo(-/-) mice incapable of synthesizing vitamin C. RESULTS: When vitamin C supplementation was ceased for 2 weeks until delivery, stillbirths and a significant reduction in neonatal mice were observed and the growth of neonates was remarkably decreased. In addition, intraparenchymal hemorrhages were found in most of the brains, especially in the stillborn neonates. In addition, the levels of malondialdehyde (MDA) and 8-isoprostanes were increased and structural abnormalities were found in the cortex, hippocampus, and cerebellum. Especially, vitamin C deficiency caused the failure of or a delay in the formation of cerebellar fissures accompanied by abnormal foliation and altered Purkinje cell alignment. In the developed adult brains from vitamin C-deficient Gulo(-/-) mice, the levels of glutathione, MDA, nitrate, IL-6, TNF-α, and Bax were increased and the expression of the GABRA6 and calbindin-28k was decreased. Due to atrophy of the granule and Purkinje cells, the motor behavior of vitamin C-deficient Gulo(-/-) mice declined. INNOVATION AND CONCLUSION: Vitamin C deficiency during gestation induces intraparenchymal hemorrhages and severe defects in the development of the cerebellum. In fully developed brains, it induces the functional impairment by altering the cellular composition in the cerebellum.


Subject(s)
Ascorbic Acid Deficiency/complications , Cerebellum/metabolism , Cerebellum/physiopathology , L-Gulonolactone Oxidase/deficiency , Motor Activity/genetics , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/physiopathology , Animals , Animals, Newborn , Ascorbic Acid/metabolism , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Interleukin-6/metabolism , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/pathology , Mice , Mice, Knockout , Neurodevelopmental Disorders/pathology , Oxidative Stress , Stillbirth , Tumor Necrosis Factor-alpha/metabolism
6.
Cell Death Dis ; 5: e1083, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24577081

ABSTRACT

Blood-brain barrier (BBB) breakdown and mitochondrial dysfunction have been implicated in the pathogenesis of Alzheimer's disease (AD), a neurodegenerative disease characterized by cognitive deficits and neuronal loss. Besides vitamin C being as one of the important antioxidants, recently, it has also been reported as a modulator of BBB integrity and mitochondria morphology. Plasma levels of vitamin C are decreased in AD patients, which can affect disease progression. However, investigation using animal models on the role of vitamin C in the AD pathogenesis has been hampered because rodents produce with no dependence on external supply. Therefore, to identify the pathogenic importance of vitamin C in an AD mouse model, we cross-bred 5 familial Alzheimer's disease mutation (5XFAD) mice (AD mouse model) with ι-gulono-γ-lactone oxidase (Gulo) knockout (KO) mice, which are unable to synthesize their own vitamin C, and produced Gulo KO mice with 5XFAD mice background (KO-Tg). These mice were maintained on either low (0.66 g/l) or high (3.3 g/l) supplementation of vitamin C. We found that the higher supplementation of vitamin C had reduced amyloid plaque burden in the cortex and hippocampus in KO-Tg mice, resulting in amelioration of BBB disruption and mitochondrial alteration. These results suggest that intake of a larger amount of vitamin C could be protective against AD-like pathologies.


Subject(s)
Alzheimer Disease/prevention & control , Ascorbic Acid/administration & dosage , Cerebral Cortex/drug effects , Dietary Supplements , Hippocampus/drug effects , Plaque, Amyloid , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Ascorbic Acid/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Capillaries/drug effects , Capillaries/metabolism , Capillaries/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Female , Gliosis , Hippocampus/enzymology , Hippocampus/pathology , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology
7.
Free Radic Biol Med ; 67: 81-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24184603

ABSTRACT

Given the involvement of oxidative stress in liver-disease- or hepato-toxicant-induced hepatic damage and fibrosis, antioxidants are an effective preventive and therapeutic tool. The beneficial results of vitamin C, one of the physiological antioxidants, have been observed both in experimental animals and in humans. However, most of these studies have been concerned with supplementary vitamin C; the effects of under vitamin C insufficiency, which humans sometimes confront, have not been substantially investigated. In the present study, we established a vitamin C-insufficient animal model (half-to-normal serum vitamin C concentration) with gulo(-/-) mice that cannot synthesize vitamin C, and induced hepatotoxicity by means of thioacetamide (TAA) injections twice a week for 18 weeks. Additionally, we explored the direct effects of vitamin C both on immortalized human hepatic stellate LX-2 cells and on rat primary hepatic stellate cells. Vitamin C insufficiency resulted in a decreased survival rate and increased serum markers for hepatocyte damage, such as alanine aminotransferase and aspartate aminotransferase. Concomitantly, the levels of reactive oxygen species (ROS) and lipid peroxides in the liver were increased. Histological examinations of the vitamin C-insufficient liver revealed increases in collagen fiber deposition and activated-hepatic-stellate-cell number. Vitamin C, when directly applied to the LX-2 cells as well as the rat primary hepatic stellate cells, suppressed not only proliferation but hydrogen peroxide-induced collagen expression as well. In conclusion, vitamin C insufficiency exacerbated TAA-induced hepatotoxicity. These effects seem to be mainly from insufficient scavenging of ROS in the liver, and possibly in part, by directly affecting hepatic stellate cells.


Subject(s)
Ascorbic Acid Deficiency/metabolism , Ascorbic Acid/administration & dosage , L-Gulonolactone Oxidase/genetics , Liver Cirrhosis/metabolism , Alanine Transaminase/blood , Animals , Ascorbic Acid Deficiency/complications , Ascorbic Acid Deficiency/genetics , Ascorbic Acid Deficiency/pathology , Aspartate Aminotransferases/blood , Collagen/biosynthesis , Collagen/genetics , Gene Expression , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , L-Gulonolactone Oxidase/deficiency , Lipid Peroxidation/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/complications , Liver Cirrhosis/genetics , Male , Mice , Mice, Knockout , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Thioacetamide
8.
Antioxid Redox Signal ; 19(17): 2040-53, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23472641

ABSTRACT

AIM: l-ascorbic acid (vitamin C) insufficiency is considered one of the major risk factors for the development of liver disease. However, its specific effects and related mechanisms in vivo are largely unknown. The objective of this study was to investigate the in vivo protective role of vitamin C and its related mechanisms in liver injury with Gulo(-/-) mice that cannot synthesize vitamin C like humans due to the lack of l-gulonolactone-γ-oxidase (Gulo), an essential enzyme for vitamin C synthesis. RESULTS: When liver injury was induced in Gulo(-/-) mice by injection of concanavalin A (Con A), there was greater extensive liver damage accompanied by an increased number of apoptotic hepatocytes in vitamin C-insufficient Gulo(-/-) mice. Additionally, the plasma and hepatic levels of the proinflammatory cytokines, such as TNF-α and IFN-γ, were much higher in the vitamin C-insufficient Gulo(-/-) mice than in the control mice. Moreover, increased numbers of liver-infiltrating T-cells in the vitamin C-insufficient Gulo(-/-) mice were related to the increased hepatic levels of IFN-inducible factor (IP-10). Although the vitamin C-insufficient Gulo(-/-) mice had higher amounts of interleukin-22 (IL-22), a hepatoprotective cytokine, a defect in IL-22Rα expression and its downstream STAT3 activation in hepatocytes were found. INNOVATION: We first demonstrate the novel in vivo action mechanisms of vitamin C on the prevention of disease development in the liver, through the regulation of excessive immune activation and maintenance of the IL-22Rα signaling pathways. CONCLUSION: These results suggest that severe liver damage induced by inflammation could be prevented by sufficient supplementation with vitamin C.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid Deficiency/pathology , Ascorbic Acid/therapeutic use , Chemical and Drug Induced Liver Injury/metabolism , Hepatitis/metabolism , Animals , Ascorbic Acid Deficiency/enzymology , Ascorbic Acid Deficiency/immunology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/immunology , Cytokines/metabolism , Enzyme Activation , Hepatitis/immunology , Inflammation Mediators/metabolism , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , p38 Mitogen-Activated Protein Kinases
9.
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
10.
Int J Oncol ; 42(1): 55-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23175106

ABSTRACT

Degradation of the extracellular matrix (ECM) plays a critical role in the formation of tumors and metastasis and has been found to correlate with the aggressiveness of tumor growth and invasiveness of cancer. Ascorbic acid, which is known to be essential for the structural integrity of the intercellular matrix, is not produced by humans and must be obtained from the diet. Cancer patients have been shown to have very low reserves of ascorbic acid. Our main objective was to determine the effect of ascorbate supplementation on metastasis, tumor growth and tumor immunohistochemistry in mice unable to synthesize ascorbic acid [gulonolactone oxidase (gulo) knockout (KO)] when challenged with B16FO melanoma or 4T1 breast cancer cells. Gulo KO female mice 36-38 weeks of age were deprived of or maintained on ascorbate in food and water for 4 weeks prior to and 2 weeks post intraperitoneal (IP) injection of 5x105 B16FO murine melanoma cells or to injection of 5x105 4T1 breast cancer cells into the mammary pad of mice. Ascorbate-supplemented gulo KO mice injected with B16FO melanoma cells demonstrated significant reduction (by 71%, p=0.005) in tumor metastasis compared to gulo KO mice on the control diet. The mean tumor weight in ascorbate supplemented mice injected with 4T1 cells was reduced by 28% compared to tumor weight in scorbutic mice. Scorbutic tumors demonstrated large dark cores, associated with increased necrotic areas and breaches to the tumor surface, apoptosis and matrix metalloproteinase-9 (MMP-9), and weak, disorganized or missing collagen I tumor capsule. In contrast, the ascorbate-supplemented group tumors had smaller fainter colored cores and confined areas of necrosis/apoptosis with no breaches from the core to the outside of the tumor and a robust collagen I tumor capsule. In both studies, ascorbate supplementation of gulo KO mice resulted in profoundly decreased serum inflammatory cytokine interleukin (IL)-6 (99% decrease, p=0.01 in the B16F0 study and 85% decrease, p=0.08 in the 4T1 study) compared to the levels in gulo KO mice deprived of ascorbate. In the B16FO study, ascorbate supplementation of gulo KO mice resulted in profoundly decreased serum VEGF (98% decrease, p=0.019 than in the scorbutic gulo KO mice). As expected, mean serum ascorbate level in ascorbate-restricted mice was 2% (p<0.001) of the mean ascorbate levels in supplemented mice. In conclusion, ascorbate supplementation hinders metastasis, tumor growth and inflammatory cytokine secretion as well as enhanced encapsulation of tumors elicited by melanoma and breast cancer cell challenge in gulo KO mice.


Subject(s)
Antioxidants/administration & dosage , Ascorbic Acid Deficiency/prevention & control , Ascorbic Acid/administration & dosage , Breast Neoplasms/prevention & control , Dietary Supplements , L-Gulonolactone Oxidase/physiology , Melanoma, Experimental/prevention & control , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Ascorbic Acid/metabolism , Ascorbic Acid Deficiency/metabolism , Ascorbic Acid Deficiency/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Immunoenzyme Techniques , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Metastasis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
11.
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
12.
Genes Brain Behav ; 11(3): 269-77, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22296218

ABSTRACT

Organisms using oxygen for aerobic respiration require antioxidants to balance the production of reactive oxygen species during metabolic processes. Various species--including humans and other primates--suffer mutations in the GULO gene encoding L-gulono-γ-lactone oxidase; GULO is the rate-limiting enzyme in the biosynthesis of ascorbate, an important cellular antioxidant. Animals lacking the ability to synthesize vitamin C develop scurvy without dietary supplementation. The Gulo-/- knockout (KO) mouse requires oral supplemental vitamin C; without this supplementation the animal dies with a scorbutic condition within several weeks. Vitamin C is known to be most abundant in the brain, where it is believed to play important roles in neuroprotection, neurotransmission and neuromodulation. We therefore hypothesized that ascorbate deficiency in Gulo-/- KO mice might lead to an abnormal behavioral phenotype. We established the amount of ascorbate in the drinking water (220 ppm) necessary for generating a chronic low-ascorbate status in the brain, yet clinically the mice appeared healthy throughout 100 days postpartum at which time all behavioral-phenotyping tests were completed. Compared with Gulo+/+ wild-type littermates, ascorbate-deficient Gulo-/- mice were found to be less active in moving in their environment; when in water, these mice swam more slowly in some tests, consistent with a mild motor deficit. We found no evidence of cognitive, anxiety or sensorimotor-gating problems. Despite being less active, Gulo-/- mice exhibited exaggerated hyperactivity to the dopaminergic agonist methamphetamine. The subnormal movement, combined with hypersensitivity to a dopamine agonist, point to developmental ascorbate deficiency causing long-term striatal dysfunction.


Subject(s)
Ascorbic Acid Deficiency/enzymology , Ascorbic Acid Deficiency/genetics , Behavior, Animal/physiology , L-Gulonolactone Oxidase/deficiency , Animals , Animals, Newborn , Ascorbic Acid/genetics , Ascorbic Acid Deficiency/physiopathology , Disease Models, Animal , Female , L-Gulonolactone Oxidase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy
13.
Toxicol Appl Pharmacol ; 257(1): 32-7, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21878346

ABSTRACT

The aim of this study was to test a hypothesis that ascorbate depletion could enhance carcinogenicity and acute toxicity of nickel. Homozygous L-gulono--lactone oxidase gene knock-out mice (Gulo-/- mice) unable to produce ascorbate and wild-type C57BL mice (WT mice) were injected intramuscularly with carcinogenic nickel subsulfide (Ni3S2), and observed for the development of injection site tumors for 57 weeks. Small pieces of one of the induced tumors were transplanted subcutaneously into separate groups of Gulo-/- and WT mice and the growth of these tumors was measured for up to 3 months. The two strains of mice differed significantly with regard to (1) Ni3S2 carcinogenesis: Gulo-/- mice were 40% more susceptible than WT mice; and (2) transplanted tumors development: Gulo-/- mice were more receptive to tumor growth than WT mice, but only in terms of a much shorter tumor latency; later in the exponential phase of growth, the growth rates were the same. And, with adequate ascorbate supplementation, the two strains were equally susceptible to acute toxicity of Ni3S2. Statistically significant effects of dietary ascorbate dosing levels were the following: (1) reduction in ascorbate supplementation increased acute toxicity of Ni3S2 in Gulo-/- mice; (2) ascorbate supplementation extended the latency of transplanted tumors in WT mice. In conclusion, the lack of endogenous ascorbate synthesis makes Gulo-/- mice more susceptible to Ni3S2 carcinogenesis. Dietary ascorbate tends to attenuate acute toxicity of Ni3S2 and to extend the latency of transplanted tumors. The latter effects may be of practical importance to humans and thus deserve further studies.


Subject(s)
Ascorbic Acid/pharmacology , Carcinogens/toxicity , L-Gulonolactone Oxidase/metabolism , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Nickel/toxicity , Animals , Ascorbic Acid/metabolism , Carcinogens/administration & dosage , Drug Interactions , Injections, Intramuscular , L-Gulonolactone Oxidase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/pathology , Nickel/administration & dosage
14.
J Biomol NMR ; 49(3-4): 165-73, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21360155

ABSTRACT

Nutrient deficiencies are an ongoing problem in many populations and ascorbic acid is a key vitamin whose mild or acute absence leads to a number of conditions including the famously debilitating scurvy. As such, the biochemical effects of ascorbate deficiency merit ongoing scrutiny, and the Gulo knockout mouse provides a useful model for the metabolomic examination of vitamin C deficiency. Like humans, these animals are incapable of synthesizing ascorbic acid but with dietary supplements are otherwise healthy and grow normally. In this study, all vitamin C sources were removed after weaning from the diet of Gulo-/- mice (n = 7) and wild type controls (n = 7) for 12 weeks before collection of serum. A replicate study was performed with similar parameters but animals were harvested pre-symptomatically after 2-3 weeks. The serum concentration of 50 metabolites was determined by quantitative profiling of 1D proton NMR spectra. Multivariate statistical models were used to describe metabolic changes as compared to control animals; replicate study animals were used for external validation of the resulting models. The results of the study highlight the metabolites and pathways known to require ascorbate for proper flux.


Subject(s)
Ascorbic Acid Deficiency/metabolism , Magnetic Resonance Spectroscopy , Metabolome , Animals , Ascorbic Acid/metabolism , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/metabolism , Metabolic Networks and Pathways , Mice , Mice, Knockout
15.
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
16.
Genet. mol. biol ; Genet. mol. biol;34(3): 386-395, 2011. ilus, tab
Article in English | LILACS | ID: lil-595988

ABSTRACT

The aim of this study was to test the hypothesis that hepatic vitamin C (VC) levels in VC deficient mice rescued with high doses of VC supplements still do not reach the optimal levels present in wild-type mice. For this, we used a mouse scurvy model (sfx) in which the L-gulonolactone oxidase gene (Gulo) is deleted. Six age- (6 weeks old) and gender- (female) matched wild-type (WT) and sfx mice (rescued by administering 500 mg of VC/L) were used as the control (WT) and treatment (MT) groups (n = 3 for each group), respectively. Total hepatic RNA was used in triplicate microarray assays for each group. EDGE software was used to identify differentially expressed genes and transcriptomic analysis was used to assess the potential genetic regulation of Gulo gene expression. Hepatic VC concentrations in MT mice were significantly lower than in WT mice, even though there were no morphological differences between the two groups. In MT mice, 269 differentially expressed transcripts were detected (> twice the difference between MT and WT mice), including 107 up-regulated and 162 down-regulated genes. These differentially expressed genes included stress-related and exclusively/predominantly hepatocyte genes. Transcriptomic analysis identified a major locus on chromosome 18 that regulates Gulo expression. Since three relevant oxidative genes are located within the critical region of this locus we suspect that they are involved in the down-regulation of oxidative activity in sfx mice.


Subject(s)
Animals , Mice , Ascorbic Acid , Gene Expression , L-Gulonolactone Oxidase , Liver , Oxidative Stress
17.
Reprod Sci ; 17(12): 1081-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861395

ABSTRACT

MicroRNAs (miRs) are known to repress target genes at posttranscriptional level and play important roles in the maturation of cells. However, the expression profiles of miRs during follicular maturation have not been fully elucidated. This study was designed to investigate the expression profiles of miRs in murine follicles according to human chorionic gonadotropin (hCG) treatment and vitamin C status during in vitro culture. Ovaries were removed from the 12-day-old wild-type and vitamin C-deficient (L-gulonogammalactone oxidase knockout, Gulo-/-) C57BL6 mice. Preantral follicles were isolated and cultured in 20 µL droplets of culture media supplemented with follicle-stimulating hormone and luteinizing hormone (FSH + LH). After their full maturation, follicles were divided into 2 groups: with and without hCG treatment. Real-time polymerase chain reaction (PCR) was performed using oocytes and granulosa cells (G-cells) to evaluate the miRs known to be expressed mainly in the mouse ovary. After the addition of hCG, miR profiles showed divergent changes between oocytes and G-cells. These profiles significantly differed from those of hCG(-) group. Compared to wild type, Gulo-/- mice showed altered miR profiles in matured oocytes and G-cells. Conclusively, hCG supplementation and vitamin C status alter the miR expression profiles in oocytes and G-cells during in vitro growth of murine follicles.


Subject(s)
Ascorbic Acid Deficiency/metabolism , Gene Expression/drug effects , MicroRNAs/genetics , Ovarian Follicle/growth & development , Animals , Ascorbic Acid Deficiency/genetics , Chorionic Gonadotropin/pharmacology , Culture Media , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/chemistry , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Luteinizing Hormone/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/analysis , Oocytes/chemistry , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Polymerase Chain Reaction , Tissue Culture Techniques
18.
Arterioscler Thromb Vasc Biol ; 30(9): 1751-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20558818

ABSTRACT

OBJECTIVE: To assess the role of combined deficiencies of vitamins C and E on the earliest stages of atherosclerosis (an inflammatory condition associated with oxidative stress), 4 combinations of vitamin supplementation (low C/low E, low C/high E, high C/low E, and high C/high E) were studied in atherosclerosis-prone apolipoprotein E-deficient mice also unable to synthesize their own vitamin C (gulonolactone oxidase(-/-)); and to evaluate the effect of a more severe depletion of vitamin C alone in a second experiment using gulonolactone oxidase(-/-) mice carrying the hemizygous deletion of SVCT2 (the vitamin C transporter). METHODS AND RESULTS: After 8 weeks of a high-fat diet (16% lard and 0.2% cholesterol), atherosclerosis developed in the aortic sinus areas of mice in all diet groups. Each vitamin-deficient diet significantly decreased liver and brain contents of the corresponding vitamin. Combined deficiency of both vitamins increased lipid peroxidation, doubled plaque size, and increased plaque macrophage content by 2- to 3-fold in male mice, although only plaque macrophage content was increased in female mice. A more severe deficiency of vitamin C in gulonolactone oxidase(-/-) mice with defective cellular uptake of vitamin C increased both oxidative stress and atherosclerosis in apolipoprotein E(-/-) mice compared with littermates receiving a diet replete in vitamin C, again most clearly in males. CONCLUSIONS: Combined deficiencies of vitamins E and C are required to worsen early atherosclerosis in an apolipoprotein E-deficient mouse model. However, a more severe cellular deficiency of vitamin C alone promotes atherosclerosis when vitamin E is replete.


Subject(s)
Aortic Diseases/etiology , Apolipoproteins E/deficiency , Ascorbic Acid Deficiency/complications , Atherosclerosis/etiology , Vitamin E Deficiency/complications , Animals , Aortic Diseases/drug therapy , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Ascorbic Acid Deficiency/drug therapy , Ascorbic Acid Deficiency/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brain/metabolism , Dietary Supplements , Disease Models, Animal , Disease Progression , Female , L-Gulonolactone Oxidase/deficiency , L-Gulonolactone Oxidase/genetics , Lipid Peroxidation , Liver/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Organic Anion Transporters, Sodium-Dependent/deficiency , Organic Anion Transporters, Sodium-Dependent/genetics , Oxidative Stress , Severity of Illness Index , Sex Factors , Sodium-Coupled Vitamin C Transporters , Symporters/deficiency , Symporters/genetics , Time Factors , Vitamin E/metabolism , Vitamin E/pharmacology , Vitamin E Deficiency/drug therapy , Vitamin E Deficiency/metabolism , Vitamins/metabolism , Vitamins/pharmacology
19.
Brain Res ; 1348: 181-6, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20570663

ABSTRACT

Vitamin C (VC) is a crucial antioxidant in the brain. To assess whether different brain regions vary in their sensitivity to oxidative stress induced by VC depletion, we used the gulonolactone oxidase (gulo) knockout mouse. This mouse, like humans, cannot synthesize VC and thus its tissue VC levels can be varied by dietary VC intake. Gulo knockout mice were fed drinking water containing standard (0.33g/L), low (0.033g/L) or zero (0g/L) VC supplementation levels. After 4weeks, mice were sacrificed and different brain regions removed for assay of VC and malondialdehyde, a marker of lipid peroxidation. Compared to age-matched wild-type controls, the cerebellum, olfactory bulbs and frontal cortex had the highest VC content, whereas the pons and spinal chord had the lowest. However, in mice that did not receive VC, area differences were no longer significant as all values trended towards zero. Malondialdehyde increased in the cortex as VC supplementation was decreased. The same changes were not observed in the cerebellum or pons, suggesting that cortex is more susceptible to oxidative damage from low VC. These results suggest enhanced susceptibility of the cortex to oxidative stress induced by low VC compared to other brain regions.


Subject(s)
Ascorbic Acid Deficiency/pathology , Ascorbic Acid/metabolism , Brain/metabolism , Oxidative Stress/physiology , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid Deficiency/etiology , Disease Models, Animal , L-Gulonolactone Oxidase/deficiency , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Statistics, Nonparametric , Tissue Distribution
20.
Biotechnol Lett ; 32(2): 321-30, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19821071

ABSTRACT

L-ascorbic acid (Vitamin C, AsA) is an important component of human nutrition. Plants and several animals can synthesize their own ascorbic acid, whereas humans lack the gene essential for ascorbic acid biosynthesis and must acquire from their diet. In the present study, we developed transgenic potato (Solanum tuberosum L. cv. Taedong Valley) over-expressing L-gulono-gamma-lactone oxidase (GLOase gene; NCBI Acc. No. NM022220), isolated from rat cells driven by CaMV35S constitutive promoter that showed enhanced AsA accumulation. Molecular analyses of four independent transgenic lines performed by PCR, Southern and RT-PCR revealed the stable integration of the transgene in the progeny. The transformation frequency was ca. 7.5% and the time required for the generation of transgenic plants was 6-7 weeks. Transgenic tubers showed significantly enhanced AsA content (141%) and GLOase activity as compared to untransformed tubers. These transgenics were also found to withstand various abiotic stresses caused by Methyl Viologen (MV), NaCl or mannitol, respectively. The T(1) transgenic plants exposed to salt stress (100 mM NaCl) survived better with increased shoot and root length when compared to untransformed plants. The elevated level of AsA accumulation in transgenics was directly correlated with their ability to withstand abiotic stresses. These results further demonstrated that the overexpression of GLOase gene enhanced basal levels of AsA in potato tubers and also the transgenics showed better survival under various abiotic stresses.


Subject(s)
Adaptation, Physiological/physiology , Ascorbic Acid/metabolism , Genetic Enhancement/methods , Heat-Shock Response/physiology , L-Gulonolactone Oxidase/metabolism , Plants, Genetically Modified/physiology , Solanum tuberosum/physiology , Animals , Base Sequence , L-Gulonolactone Oxidase/genetics , Molecular Sequence Data , Rats
SELECTION OF CITATIONS
SEARCH DETAIL