Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Brain Behav Immun ; 120: 557-570, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972487

ABSTRACT

Neuroinflammation is a major characteristic of pathology in several neurodegenerative diseases. Microglia, the brain's resident myeloid cells, shift between activation states under neuroinflammatory conditions, both responding to, but also driving damage in the brain. Vitamin C (ascorbate) is an essential antioxidant for central nervous system function that may have a specific role in the neuroinflammatory response. Uptake of ascorbate throughout the central nervous system is facilitated by the sodium-dependent vitamin C transporter 2 (SVCT2). SVCT2 transports the reduced form of ascorbate into neurons and microglia, however the contribution of altered SVCT2 expression to the neuroinflammatory response in microglia is not well understood. In this study we demonstrate that SVCT2 expression modifies microglial response, as shown through changes in cell morphology and mRNA expression, following a mild traumatic brain injury (mTBI) in mice with decreased or increased expression of SVCT2. Results were supported by in vitro studies in an immortalized microglial cell line and in primary microglial cultures derived from SVCT2-heterozygous and transgenic animals. Overall, this work demonstrates the importance of SVCT2 and ascorbate in modulating the microglial response to mTBI and suggests a potential role for both in response to neuroinflammatory challenges.


Subject(s)
Ascorbic Acid , Microglia , Sodium-Coupled Vitamin C Transporters , Animals , Sodium-Coupled Vitamin C Transporters/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Microglia/metabolism , Mice , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Mice, Transgenic , Mice, Inbred C57BL , Neuroinflammatory Diseases/metabolism , Male , Brain/metabolism , Neurons/metabolism , Brain Concussion/metabolism , Cell Line
2.
Pflugers Arch ; 475(4): 489-504, 2023 04.
Article in English | MEDLINE | ID: mdl-36749388

ABSTRACT

Uric acid, the end product of purine metabolism in humans, is crucial because of its anti-oxidant activity and a causal relationship with hyperuricemia and gout. Several physiologically important urate transporters regulate this water-soluble metabolite in the human body; however, the existence of latent transporters has been suggested in the literature. We focused on the Escherichia coli urate transporter YgfU, a nucleobase-ascorbate transporter (NAT) family member, to address this issue. Only SLC23A proteins are members of the NAT family in humans. Based on the amino acid sequence similarity to YgfU, we hypothesized that SLC23A1, also known as sodium-dependent vitamin C transporter 1 (SVCT1), might be a urate transporter. First, we identified human SVCT1 and mouse Svct1 as sodium-dependent low-affinity/high-capacity urate transporters using mammalian cell-based transport assays. Next, using the CRISPR-Cas9 system followed by the crossing of mice, we generated Svct1 knockout mice lacking both urate transporter 1 and uricase. In the hyperuricemic mice model, serum urate levels were lower than controls, suggesting that Svct1 disruption could reduce serum urate. Given that Svct1 physiologically functions as a renal vitamin C re-absorber, it could also be involved in urate re-uptake from urine, though additional studies are required to obtain deeper insights into the underlying mechanisms. Our findings regarding the dual-substrate specificity of SVCT1 expand the understanding of urate handling systems and functional evolutionary changes in NAT family proteins.


Subject(s)
Organic Anion Transporters , Uric Acid , Animals , Humans , Mice , Amino Acid Sequence , Ascorbic Acid/metabolism , Biological Transport , Mammals/metabolism , Organic Anion Transporters/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Sodium-Coupled Vitamin C Transporters/metabolism , Uric Acid/metabolism
3.
FASEB J ; 35(2): e21219, 2021 02.
Article in English | MEDLINE | ID: mdl-33236467

ABSTRACT

Liver fibrosis is characterized by the accumulation of extracellular matrix proteins, mainly composed of collagen. Hepatic stellate cells (HSCs) mediate liver fibrosis by secreting collagen. Vitamin C (ascorbic acid) is a cofactor of prolyl-hydroxylases that modify newly synthesized collagen on the route for secretion. Unlike most animals, humans cannot synthesize ascorbic acid and its role in liver fibrosis remains unclear. Here, we determined the effect of ascorbic acid and prolyl-hydroxylase inhibition on collagen production and secretion by human HSCs. Primary human HSCs (p-hHSCs) and the human HSCscell line LX-2 were treated with ascorbic acid, transforming growth factor-beta (TGFß) and/or the pan-hydroxylase inhibitor dimethyloxalylglycine (DMOG). Expression of collagen-I was analyzed by RT-qPCR (COL1A1), Western blotting, and immunofluorescence microscopy. Collagen secretion was determined in the medium by Western blotting for collagen-I and by HPLC for hydroxyproline concentrations. Expression of solute carrier family 23 members 1 and 2 (SLC23A1/SLC23A2), encoding sodium-dependent vitamin C transporters 1 and 2 (SVCT1/SVCT2) was quantified in healthy and cirrhotic human tissue. In the absence of ascorbic acid, collagen-I accumulated intracellularly in p-hHSCs and LX-2 cells, which was potentiated by TGFß. Ascorbic acid co-treatment strongly promoted collagen-I excretion and enhanced extracellular hydroxyproline concentrations, without affecting collagen-I (COL1A1) mRNA levels. DMOG inhibited collagen-I release even in the presence of ascorbic acid and suppressed COL1A1 and alpha-smooth muscle actin (αSMA/ACTA2) mRNA levels, also under hypoxic conditions. Hepatocytes express both ascorbic acid transporters, while p-hHSCs and LX-2 express the only SVCT2, which is selectively enhanced in cirrhotic livers. Human HSCs rely on ascorbic acid for the efficient secretion of collagen-I, which can be effectively blocked by hydroxylase antagonists, revealing new therapeutic targets to treat liver fibrosis.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Ascorbic Acid/metabolism , Collagen Type I/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Actins/genetics , Actins/metabolism , Animals , Ascorbic Acid/pharmacology , Cell Line , Cells, Cultured , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Hepatic Stellate Cells/drug effects , Humans , Rats , Sodium-Coupled Vitamin C Transporters/metabolism , Transforming Growth Factor beta/pharmacology
4.
Cell Physiol Biochem ; 55(5): 553-568, 2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34599650

ABSTRACT

BACKGROUND/AIMS: Maintenance of whole-body ascorbate levels and distribution is mediated via sodium-dependent vitamin C transporters (SVCTs). The kidney is one of a few organs that express both SVCT1 and SVCT2. Recent evidence suggests that accumulation of ascorbate may be different in tumour compared to normal tissue, but data on SVCT levels in tumours is sparse. METHODS: The role of the two SVCT isoforms in ascorbate uptake in renal cell carcinoma (RCC) was investigated in vitro and in clinical samples. In three human RCC cell lines, we investigated SVCT protein levels and cellular location in response to ascorbate supplementation and withdrawal. In clinical RCC samples (n=114), SVCT patterns of staining and protein levels were analysed and compared to ascorbate levels. RESULTS: In cell culture, transporter levels and cellular location were not modified by ascorbate availability at any time up to 8h, although basal SVCT2 levels governed maximal ascorbate accumulation. In clinical samples, SVCT1 protein levels in papillary RCC (pRCC) were similar to matched normal renal cortex, but were increased in clear-cell RCC (ccRCC). Native SVCT2 (72 kDa) was significantly decreased in both pRCC and ccRCC tissues compared to cortex (p<0.01), whereas a modified form of SVCT2 (100 kDa) was significantly increased (p<0.001). There was no association between the transporters (SVCT1, native or modified SVCT2) and ascorbate concentrations in either normal or tumour tissues. SVCT1 and SVCT2 displayed diffuse cytoplasmic staining in both pRCC and ccRCC tumour cells, with cortex showing distinct membrane staining for SVCT1. CONCLUSION: We observed a re-distribution of ascorbate transporters in tumour tissue compared to normal cortex and a shift from native to modified SVCT2 in cell culture and clinical samples. Data presented here show that SVCT protein levels do not appear to predict intracellular ascorbate accumulation in RCC.


Subject(s)
Ascorbic Acid/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , Kidney Neoplasms/pathology , Sodium-Coupled Vitamin C Transporters/analysis
5.
J Neurosci Res ; 99(1): 223-235, 2021 01.
Article in English | MEDLINE | ID: mdl-32754987

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a glutamine expansion at the first exon of the huntingtin gene. Huntingtin protein (Htt) is ubiquitously expressed and it is localized in several organelles, including endosomes. HD is associated with a failure in energy metabolism and oxidative damage. Ascorbic acid is a powerful antioxidant highly concentrated in the brain where it acts as a messenger, modulating neuronal metabolism. It is transported into neurons via the sodium-dependent vitamin C transporter 2 (SVCT2). During synaptic activity, ascorbic acid is released from glial reservoirs to the extracellular space, inducing an increase in SVCT2 localization at the plasma membrane. Here, we studied SVCT2 trafficking and localization in HD. SVCT2 is decreased at synaptic terminals in YAC128 male mice. Using cellular models for HD (STHdhQ7 and STHdhQ111 cells), we determined that SVCT2 trafficking through secretory and endosomal pathways is altered in resting conditions. We observed Golgi fragmentation and SVCT2/Htt-associated protein-1 mis-colocalization. Additionally, we observed altered ascorbic acid-induced calcium signaling that explains the reduced SVCT2 translocation to the plasma membrane in the presence of extracellular ascorbic acid (active conditions) described in our previous results. Therefore, SVCT2 trafficking to the plasma membrane is altered in resting and active conditions in HD, explaining the redox imbalance observed during early stages of the disease.


Subject(s)
Huntington Disease/metabolism , Protein Transport/physiology , Sodium-Coupled Vitamin C Transporters/metabolism , Synaptosomes/metabolism , Animals , Male , Mice , Mice, Transgenic , Neurons/metabolism , Oxidation-Reduction
6.
Dig Dis Sci ; 66(7): 2250-2260, 2021 07.
Article in English | MEDLINE | ID: mdl-32556816

ABSTRACT

BACKGROUND: Enteropathogenic Escherichia coli (EPEC) infection causes prolonged, watery diarrhea leading to morbidity and mortality. Although EPEC infection impacts nutrient transporter function and expression in intestinal epithelial cells, the effects of EPEC infection on intestinal absorption of ascorbic acid (AA) have not yet been investigated. AIMS: To investigate the effect of EPEC infection on intestinal AA uptake process and expression of both AA transporters. METHODS: We used two experimental models: human-derived intestinal epithelial Caco-2 cells and mice. 14C-AA uptake assay, Western blot, RT-qPCR, and promoter assay were performed. RESULTS: EPEC (WT) as well as ΔespF and ΔespG/G2 mutant-infected Caco-2 cells showed markedly inhibited AA uptake, while other mutants (ΔescN, ΔespA, ΔespB, and ΔespD) did not affect AA uptake. Infection also reduced protein and mRNA expression levels for both hSVCT1 and hSVCT2. EPEC-infected mice showed marked inhibitory effect on AA uptake and decreased protein and mRNA expression levels for both mSVCT1 and mSVCT2 in jejunum and colon. MicroRNA regulators of SVCT1 and SVCT2 (miR103a, miR141, and miR200a) were upregulated significantly upon EPEC infection in both Caco-2 and mouse jejunum and colon. In addition, expression of the accessory protein glyoxalate reductase/hydroxypyruvate reductase (GRHPR), which regulates SVCT1 function, was markedly decreased by EPEC infection in both models. CONCLUSIONS: These findings suggest that EPEC infection causes inhibition in AA uptake through a multifactorial dysregulation of SVCT1 and SVCT2 expression in intestinal epithelial cells.


Subject(s)
Ascorbic Acid/metabolism , Enteropathogenic Escherichia coli , Escherichia coli Infections/pathology , Intestinal Mucosa/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Animals , Biological Transport , Caco-2 Cells , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation , Humans , Mice , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Coupled Vitamin C Transporters/genetics
7.
Mediators Inflamm ; 2021: 4157132, 2021.
Article in English | MEDLINE | ID: mdl-34285658

ABSTRACT

Vitamin C (ascorbic acid: AA) uptake in neurons occurs via the sodium-dependent vitamin C transporter-2 (SVCT2), which is highly expressed in the central nervous system (CNS). During chronic neuroinflammation or infection, CNS levels of lipopolysaccharide (LPS) and LPS-induced tumor necrosis factor-α (TNFα) are increased. Elevated levels of LPS and TNFα have been associated with neurodegenerative diseases together with reduced levels of AA. However, little is known about the impacts of LPS and TNFα on neuronal AA uptake. The objective of this study was to examine the effect of LPS and TNFα on SVCT2 expression and function using in vitro and in vivo approaches. Treatment of SH-SY5Y cells with either LPS or TNFα inhibited AA uptake. This reduced uptake was associated with a significant decrease in SVCT2 protein and mRNA levels. In vivo exposure to LPS or TNFα also decreased SVCT2 protein and mRNA levels in mouse brains. Both LPS and TNFα decreased SLC23A2 promoter activity. Further, the inhibitory effect of LPS on a minimal SLC23A2 promoter was attenuated when either the binding site for the transcription factor Sp1 was mutated or cells were treated with the NF-κB inhibitor, celastrol. We conclude that inflammatory signals suppress AA uptake by impairing SLC23A2 transcription through opposing regulation of Sp1 and NF-κB factors.


Subject(s)
Ascorbic Acid , Lipopolysaccharides , Animals , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Mice , Neurons/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Sodium-Coupled Vitamin C Transporters/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Molecules ; 26(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809092

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disorder. The number of patients with AD is projected to reach 152 million by 2050. Donepezil, rivastigmine, galantamine, and memantine are the only four drugs currently approved by the United States Food and Drug Administration for AD treatment. However, these drugs can only alleviate AD symptoms. Thus, this research focuses on the discovery of novel lead compounds that possess multitarget regulation of AD etiopathology relating to amyloid cascade. The ascorbic acid structure has been designated as a core functional domain due to several characteristics, including antioxidant activities, amyloid aggregation inhibition, and the ability to be transported to the brain and neurons. Multifunctional ascorbic derivatives were synthesized by copper (I)-catalyzed azide-alkyne cycloaddition reaction (click chemistry). The in vitro and cell-based assays showed that compounds 2c and 5c exhibited prominent multifunctional activities as beta-secretase 1 inhibitors, amyloid aggregation inhibitors, and antioxidant, neuroprotectant, and anti-inflammatory agents. Significant changes in activities promoting neuroprotection and anti-inflammation were observed at a considerably low concentration at a nanomolar level. Moreover, an in silico study showed that compounds 2c and 5c were capable of being permeated across the blood-brain barrier by sodium-dependent vitamin C transporter-2.


Subject(s)
Amyloidogenic Proteins/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Ascorbic Acid/analogs & derivatives , Neuroprotective Agents/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Ascorbic Acid/chemistry , Ascorbic Acid/pharmacology , Binding Sites , Blood-Brain Barrier , Cells, Cultured , Computer Simulation , Cyclooxygenase 2/genetics , Gene Expression/drug effects , Humans , Mice , Molecular Docking Simulation , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Nitric Oxide Synthase Type II/genetics , RAW 264.7 Cells , Sodium-Coupled Vitamin C Transporters/chemistry , Sodium-Coupled Vitamin C Transporters/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology
9.
Pharmacol Res ; 159: 105042, 2020 09.
Article in English | MEDLINE | ID: mdl-32580031

ABSTRACT

The Na+-dependent Vitamin C transporter 2 (SVCT2) is expressed in the plasma and mitochondrial membranes of various cell types. This notion was also established in proliferating C2C12 myoblasts (Mb), in which the transporter was characterised by a high and low affinity in the plasma and mitochondrial membranes, respectively. In addition, the mitochondrial expression of SVCT2 appeared particularly elevated and, consistently, a brief pre-exposure to low concentrations of Ascorbic Acid (AA) abolished mitochondrial superoxide formation selectively induced by the cocktail arsenite/ATP. Early myotubes (Mt) derived from these cells after 4 days of differentiation presented evidence of slightly increased SVCT2 expression, and were characterised by kinetic parameters for plasma membrane transport of AA in line with those detected in Mb. Confocal microscopy studies indicated that the mitochondrial expression of SVCT2 is well preserved in Mt with one or two nuclei, but progressively reduced in Mt with three or more nuclei. Cellular and mitochondrial expression of SVCT2 was found reduced in day 7 Mt. While the uptake studies were compromised by the poor purity of the mitochondrial preparations obtained from day 4 Mt, we nevertheless obtained evidence of poor transport of the vitamin using the same functional studies successfully employed with Mb. Indeed, even greater concentrations of/longer pre-exposure to AA failed to induce scavenging of mitochondrial superoxide in Mt. These results are therefore indicative of a severely reduced mitochondrial uptake of the vitamin in early Mt, attributable to decreased expression as well as impaired activity of mitochondrial SVCT2.


Subject(s)
Ascorbic Acid/metabolism , Cell Differentiation , Cell Membrane/metabolism , Mitochondrial Membranes/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Adenosine Triphosphate/pharmacology , Animals , Arsenites/pharmacology , Ascorbic Acid/pharmacology , Biological Transport , Cell Differentiation/drug effects , Cell Line , Cell Membrane/drug effects , Kinetics , Mice , Mitochondrial Membranes/drug effects , Muscle Fibers, Skeletal/drug effects , Myoblasts, Skeletal/drug effects , Sodium Compounds/pharmacology , Sodium-Coupled Vitamin C Transporters/genetics
10.
Cell Mol Biol Lett ; 25: 32, 2020.
Article in English | MEDLINE | ID: mdl-32514268

ABSTRACT

Recent interest in the role of ascorbate in crucial metabolic processes is driven by the growing number of medical reports that show beneficial effects of ascorbate supplementation for maintaining general well-being and recovery from a variety of medical conditions. The effect of ascorbate on the local body environment highly depends on its local concentration; at low concentrations it can cause the reduction of reactive oxygen and facilitate activities of enzymes, while at high concentrations it generates free radicals by reducing ferric ions. Ascorbate serving as an electron donor assists the iron-containing proteins and the iron transfer between various aqueous compartments. These functions require effective and adjustable mechanisms responsible for ascorbate biodistribution. In the paper we propose a new biophysical model of ascorbate redistribution between various aqueous body compartments. It combines recent experimental evidence regarding the ability of ascorbate to cross the lipid bilayer by unassisted diffusion, with active transport by well-characterized sodium vitamin C transporter (SVCT) membrane proteins. In the model, the intracellular concentration of ascorbate is maintained by the balance of two opposing fluxes: fast active and slow passive transport. The model provides a mechanistic understanding of ascorbate flux across the epidermal barrier in the gut as well as the role of astrocytes in ascorbate recycling in the brain. In addition, ascorbate passive diffusion across biological membranes, which depends on membrane electric potentials and pH gradients, provides the rationale for the correlation between ascorbate distribution and the transfer of iron ions inside a cell. The proposed approach provides, for the first time, a mechanistic account of processes leading to ascorbate physiological and cellular distribution, which helps to explain numerous experimental and clinical observations.


Subject(s)
Ascorbic Acid/metabolism , Ascorbic Acid/pharmacokinetics , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Homeostasis/drug effects , Humans , Hydrogen-Ion Concentration , Lipid Bilayers , Models, Biological , Sodium-Coupled Vitamin C Transporters/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G55-G63, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30285481

ABSTRACT

Vitamin C is an antioxidant and acts as a cofactor for many enzymatic reactions. Humans obtain vitamin C from dietary sources via intestinal absorption, a process that involves the sodium-dependent vitamin C transporters-1 and -2 (SVCT1 and SVCT2). Enterotoxigenic Escherichia coli (ETEC) infection impacts intestinal absorption/secretory functions, but nothing is known about its effect on ascorbic acid (AA) uptake. Here we demonstrate that infection of Caco-2 cells with ETEC led to a significant inhibition in intestinal AA uptake. This inhibition was associated with a marked reduction in hSVCT1 and hSVCT2 protein, mRNA, and heterogeneous nuclear RNA (hnRNA) expression levels as well as significant inhibition in the activity of both the SLC23A1 and SLC23A2 promoters. Similarly, exposure of mice to ETEC led to a significant inhibition in intestinal AA uptake and reduction in mSVCT1 and mSVCT2 protein, mRNA, and hnRNA expression levels. Inhibition was caused by the action of heat labile enterotoxin (LT), since infecting Caco-2 cells with LT-deficient ETEC (ΔLT) failed to impact AA uptake. Because LT activates adenylate cyclase, we also examined the effect of dibutyryl-cAMP in AA uptake by Caco-2 cells and observed a significant inhibition. Furthermore, treating the cells with celastrol, a specific NF-κB inhibitor, significantly blocked the inhibition of AA uptake caused by ETEC infection. Together, these data demonstrate that ETEC infection impairs intestinal AA uptake through a cAMP-dependent NF-κB-mediated pathway that regulates both SLC23A1 and SLC23A2 transcription. NEW & NOTEWORTHY Our findings demonstrate that heat-labile enterotoxin produced by enterotoxigenic Escherichia coli inhibits AA uptake in intestinal epithelial cells and mouse intestine. This effect is mediated through transcriptional repression of SLC23A1 (SVCT1) and SLC23A2 (SVCT2) via a cAMP-dependent NF-κB signaling pathway.


Subject(s)
Ascorbic Acid/pharmacology , Enterotoxigenic Escherichia coli/chemistry , Animals , Biological Transport/drug effects , Caco-2 Cells , Enterotoxins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Escherichia coli Infections/metabolism , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , NF-kappa B/metabolism , Sodium-Coupled Vitamin C Transporters/drug effects , Sodium-Coupled Vitamin C Transporters/metabolism , Vitamins/metabolism
12.
Biochim Biophys Acta Biomembr ; 1860(2): 556-565, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29030247

ABSTRACT

Ascorbic acid (AA) accumulation in intestinal epithelial cells is an active transport process mainly mediated by two sodium-dependent vitamin C transporters (SVCT-1 and SVCT-2). To date, little is known about the effect of gut microbiota generated lipopolysaccharide (LPS) on intestinal absorption of water-soluble vitamins. Therefore, the objective of this study was to investigate the effects of bacterially-derived LPS on AA homeostasis in enterocytes using Caco-2 cells, mouse intestine and intestinal enteroids models. Pre-treating Caco-2 cells and mice with LPS led to a significant decrease in carrier-mediated AA uptake. This inhibition was associated with a significant reduction in SVCT-1 and SVCT-2 protein, mRNA, and hnRNA expression. Furthermore, pre-treating enteroids with LPS also led to a marked decrease in SVCT-1 and SVCT-2 protein and mRNA expression. Inhibition of SVCT-1 and SVCT-2 occurred at least in part at the transcriptional level as promoter activity of SLC23A1 and SLC23A2 was attenuated following LPS treatment. Subsequently, we examined the protein and mRNA expression levels of HNF1α and Sp1 transcription factors, which are needed for basal SLC23A1 and SLC23A2 promoter activity, and found that they were significantly decreased in the LPS treated Caco-2 cells and mouse jejunum; this was reflected on level of the observed reduction in the interaction of these transcription factors with their respective promoters in Caco-2 cells treated with LPS. Our findings indicate that LPS inhibits intestinal carrier- mediated AA uptake by down regulating the expression of both vitamin C transporters and transcriptional regulation of SLC23A1 and SLC23A2 genes.


Subject(s)
Ascorbic Acid/metabolism , Gene Expression Regulation/drug effects , Intestinal Absorption/drug effects , Intestines/drug effects , Lipopolysaccharides/pharmacology , Animals , Ascorbic Acid/pharmacokinetics , Biological Transport/drug effects , Caco-2 Cells , Cells, Cultured , Enterocytes/cytology , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Male , Mice, Inbred C57BL , Sodium-Coupled Vitamin C Transporters/genetics , Sodium-Coupled Vitamin C Transporters/metabolism , Vitamins/metabolism , Vitamins/pharmacokinetics
13.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G241-G248, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29631379

ABSTRACT

Sodium-dependent vitamin C transporter-1 (SVCT-1) is the major transporter mediating intestinal vitamin C uptake. Intestinal inflammation and prolonged infection are associated with increased serum and intestinal mucosa levels of tumor necrosis factor-α (TNF-α), which also exerts profound effects on the intestinal absorption process. Elevated levels of TNF-α have been linked to the pathogenesis of inflammatory bowel disease (IBD) and malabsorption of nutrients, and patients with this condition have low levels of vitamin C. To date, little is known about the effect of TNF-α on intestinal absorption of vitamin C. We studied the impact of TNF-α on ascorbic acid (AA) transport using a variety of intestinal preparations. The expression level of human SVCT-1 mRNA is significantly lower in patients with IBD. TNF-α treated Caco-2 cells and mice showed a significant inhibition of intestinal 14C-AA uptake. This inhibition was associated with significant decreases in SVCT-1 protein, mRNA, and heterogeneous nuclear RNA levels in TNF-α treated Caco-2 cells, mouse jejunum, and enteroids. Also, TNF-α caused a significant inhibition in the SLC23A1 promoter activity. Furthermore, treatment of Caco-2 cells with celastrol (NF-κB inhibitor) blocked the inhibitory effect caused by TNF-α on AA uptake, SVCT-1 protein, and mRNA expression, as well as the activity of SLC23A1 promoter. Treatment of TNF-α also led to a significant decrease in the expression of hepatocyte nuclear factor-1-α, which drives the basal activity of SLC23A1 promoter, and this effect was reversed by celastrol. Together, these findings show that TNF-α inhibits intestinal AA uptake, and this effect is mediated, at least in part, at the level of transcription of the SLC23A1 gene via the NF-κB pathway. NEW & NOTEWORTHY Our findings show that tumor necrosis factor-α inhibits intestinal ascorbic acid uptake in both in vitro and in vivo systems, and this inhibitory effect is mediated, at least in part, at the level of transcription of the SLC23A1 (sodium-dependent vitamin C transporter-1) gene via the NF-κB pathway.


Subject(s)
Ascorbic Acid , Intestinal Absorption , Animals , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Biological Transport/physiology , Caco-2 Cells/physiology , Humans , Inflammatory Bowel Diseases/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , NF-kappa B/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vitamins/metabolism , Vitamins/pharmacology
14.
J Anat ; 232(4): 699-715, 2018 04.
Article in English | MEDLINE | ID: mdl-29315537

ABSTRACT

Its high metabolic rate and high polyunsaturated fatty acid content make the brain very sensitive to oxidative damage. In the brain, neuronal metabolism occurs at a very high rate and generates considerable amounts of reactive oxygen species and free radicals, which accumulate inside neurons, leading to altered cellular homeostasis and integrity and eventually irreversible damage and cell death. A misbalance in redox metabolism and the subsequent neurodegeneration increase throughout the course of normal aging, leading to several age-related changes in learning and memory as well as motor functions. The neuroprotective function of antioxidants is crucial to maintain good brain homeostasis and adequate neuronal functions. Vitamins E and C are two important antioxidants that are taken up by brain cells via the specific carriers αTTP and SVCT2, respectively. The aim of this study was to use immunohistochemistry to determine the distribution pattern of these vitamin transporters in the brain in a mouse model that shows fewer signs of brain aging and a higher resistance to oxidative damage. Both carriers were distributed widely throughout the entire brain in a pattern that remained similar in 4-, 12-, 18- and 24-month-old mice. In general, αTTP and SVCT2 were located in the same regions, but they seemed to have complementary distribution patterns. Double-labeled cell bodies were detected only in the inferior colliculus, entorhinal cortex, dorsal subiculum, and several cortical areas. In addition, the presence of αTTP and SVCT2 in neurons was analyzed using double immunohistochemistry for NeuN and the results showed that αTTP but not SVCT2 was present in Bergmann's glia. The presence of these transporters in brain regions implicated in learning, memory and motor control provides an anatomical basis that may explain the higher resistance of this animal model to brain oxidative stress, which is associated with better motor performance and learning abilities in old age.


Subject(s)
Aging/metabolism , Brain/metabolism , Carrier Proteins/metabolism , Oxidative Stress , Sodium-Coupled Vitamin C Transporters/metabolism , Animals , Antigens, Nuclear/metabolism , Antioxidants/metabolism , Ascorbic Acid/metabolism , Biomarkers/metabolism , Brain/diagnostic imaging , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Learning , Male , Memory , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Vitamin E/metabolism
15.
Mol Cell Biochem ; 446(1-2): 209-219, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29356987

ABSTRACT

In humans, vitamin C (VC) accumulates at higher concentrations in cells than in plasma, and this intracellular accumulation appears critical to several important physiological functions. However, although VC accumulation decreases in the elderly, the influence of cellular senescence on the transport, accumulation, and function of VC is poorly understood. In this study, we investigated the effects of supplementation with both ascorbic acid (AsA) and dehydroascorbic acid (DehAsA) on the uptake and accumulation of VC, AsA, and DehAsA into cells and the effect of AsA on the levels of intracellular reactive oxygen species (ROS) in human fibroblast TIG-1 cells. We also assessed how that supplementation affected senescence-associated changes in intracellular VC transport and accumulation. AsA supplementation significantly increased intracellular levels of AsA, DehAsA, and total VC (i.e., reduced AsA plus oxidized DehAsA) in senescent cells compared with young cells. DehAsA supplementation also significantly increased intracellular AsA and total VC levels in senescent cells, but not DehAsA, and the increases were less than after adding AsA. Among the molecules related to VC accumulation, the mRNA and protein expressions of sodium-dependent VC transporter 2 (SLC23A2) were increased in senescent cells. Furthermore, intracellular peroxide and superoxide anion levels were higher in senescent cells, with AsA supplementation markedly attenuating spontaneous intracellular peroxide accumulation. Overall, our results therefore suggest that VC transport and accumulation improved in senescent human fibroblast TIG-1 cells due to the adaptive upregulation of sodium-dependent VC transporter 2 in response to increased ROS levels. We conclude that adequate supplementation with AsA can effectively mitigate senescence-associated intracellular ROS.


Subject(s)
Ascorbic Acid , Cellular Senescence/drug effects , Fibroblasts/metabolism , Reactive Oxygen Species/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Ascorbic Acid/pharmacokinetics , Ascorbic Acid/pharmacology , Biological Transport, Active/drug effects , Cell Line , Fibroblasts/cytology , Humans , Sodium-Coupled Vitamin C Transporters/genetics
16.
J Cell Physiol ; 232(9): 2418-2426, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27463513

ABSTRACT

In the kidney, vitamin C is reabsorbed from the glomerular ultrafiltrate by sodium-vitamin C cotransporter isoform 1 (SVCT1) located in the brush border membrane of the proximal tubules. Although we know that vitamin C levels decrease with age, the adaptive physiological mechanisms used by the kidney for vitamin C reabsorption during aging remain unknown. In this study, we used an animal model of accelerated senescence (SAMP8 mice) to define the morphological alterations and aging-induced changes in the expression of vitamin C transporters in renal tissue. Aging induced significant morphological changes, such as periglomerular lymphocytic infiltrate and glomerular congestion, in the kidneys of SAMP8 mice, although no increase in collagen deposits was observed using 2-photon microscopy analysis and second harmonic generation. The most characteristic histological alteration was the dilation of intracellular spaces in the basolateral region of proximal tubule epithelial cells. Furthermore, a combination of laser microdissection, qRT-PCR, and immunohistochemical analyses allowed us to determine that SVCT1 expression specifically increased in the proximal tubules from the outer strip of the outer medulla (segment S3) and cortex (segment S2) during aging and that these tubules also express GLUT1. We conclude that aging modulates vitamin C transporter expression and that renal over-expression of SVCT1 enhances vitamin C reabsorption in aged animals that may synthesize less vitamin C. J. Cell. Physiol. 232: 2418-2426, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Aging/metabolism , Ascorbic Acid/metabolism , Kidney/metabolism , Renal Reabsorption , Sodium-Coupled Vitamin C Transporters/metabolism , Adaptation, Physiological , Age Factors , Aging/genetics , Aging/pathology , Animals , Cellular Senescence , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Kidney/ultrastructure , Male , Mice, Inbred BALB C , Models, Animal , Sodium-Coupled Vitamin C Transporters/genetics , Up-Regulation
17.
Am J Physiol Gastrointest Liver Physiol ; 312(4): G340-G347, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27932501

ABSTRACT

Mammalian cells utilize two transporters for the uptake of ascorbic acid (AA), Na+-dependent vitamin C transporter SVCT-1 and SVCT-2. In the intestine, these transporters are involved in AA absorption and are expressed at the apical and basolateral membrane domains of the polarized epithelia, respectively. Little is known about the differential expression of these two transporters along the anterior-posterior axis of the intestinal tract and the molecular mechanism(s) that dictate this pattern of expression. We used mouse and human intestinal cDNAs to address these issues. The results showed a significantly lower rate of carrier-mediated AA uptake by mouse colon than jejunum. This was associated with a significantly lower level of expression of SVCT-1 and SVCT-2 at the protein, mRNA, and heterogeneous nuclear RNA (hnRNA) levels in the colon than the jejunum, implying the involvement of transcriptional mechanism(s). Similarly, expression levels of SVCT-1 and SVCT-2 mRNA and hnRNA were significantly lower in human colon. We also examined the levels of expression of hepatocyte nuclear factor 1α and specificity protein 1, which drive transcription of the Slc23a1 and Slc23a2 promoters, respectively, and found them to be markedly lower in the colon. Furthermore, significantly lower levels of the activating markers for histone (H3) modifications [H3 trimethylation of lysine 4 (H3K4me3) and H3 triacetylation of lysine 9 (H3K9ac)] were observed in the Slc23a1 and Slc23a2 promoters in the colon. These findings show, for the first time, that SVCT-1 and SVCT-2 are differentially expressed along the intestinal tract and that this pattern of expression is, at least in part, mediated via transcriptional/epigenetic mechanisms.NEW & NOTEWORTHY Our findings show, for the first time, that transporters of the water-soluble vitamin ascorbic acid (i.e., the vitamin C transporters SVCT-1 and SVCT-2) are differentially expressed along the length of the intestinal tract and that the pattern of expression is mediated, at least in part, by transcriptional and epigenetic mechanism(s) affecting both Slc23a1 and Slc23a2 genes.


Subject(s)
Colon/metabolism , Gene Expression Regulation , Jejunum/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Adolescent , Adult , Animals , DNA Methylation , Female , Humans , Male , Mice , Middle Aged , Organ Specificity , Promoter Regions, Genetic , Young Adult
18.
Cell Physiol Biochem ; 44(1): 377-387, 2017.
Article in English | MEDLINE | ID: mdl-29132138

ABSTRACT

BACKGROUND/AIM: Colorectal cancer is still considered a leading cause of death in the United States and worldwide. One potential way to improve survival besides detection is to look to new therapeutic agents that can be taken prophylactically to reduce the risk of tumor formation. For cancer cells to grow and invade, a higher (more alkaline) intracellular pH must occur. We chose to examine a specific nutraceutical agent, which is Vitamin C. The acute effect of Vitamin C exposure on normal colonic crypts has been studied, providing some insight into how Vitamin C achieve its effect. METHODS: Distal colon was excised from rats. Following enzymatic digestion single colonic crypts were isolated. Colonic crypts were loaded with pH sensitive dye to measure the intracellular pH changes. Crypts were exposed to solutions +/- Vitamin C. RESULTS: 10 mM Vitamin C decreased Na+-dependent intracellular pH recovery. Vitamin C modulates SVCT leading to changes in proton extrusion. Vitamin C entry occurs via either SVCT2 on the basolateral membrane or by transcellular passive diffusion through tight junctions to the apical membrane and then active transport via SVCT1. CONCLUSION: Acute addition of Vitamin C to the basolateral membrane maintains low intracellular pH for a longer period which could halt and/or prevent tumor formation.


Subject(s)
Ascorbic Acid/pharmacology , Intestinal Mucosa/drug effects , Animals , Cell Membrane/metabolism , Colon/cytology , Hydrogen-Ion Concentration/drug effects , In Vitro Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism
19.
Nutr Cancer ; 69(4): 521-533, 2017.
Article in English | MEDLINE | ID: mdl-28362118

ABSTRACT

There are several extrinsic and intrinsic factors involving reactive oxygen species that play critical roles in tumor development and progression by inducing DNA mutations, genomic instability, and aberrant pro-tumorigenic signaling. There are various essential micronutrients including minerals and vitamins in the diet, which play pivotal roles in maintaining and reinforcing antioxidant performance, affecting the complex network of genes (nutrigenomic approach) and encoding proteins for carcinogenesis. A lot of these antioxidant agents are available as dietary supplements and are predominant worldwide. However, the best antioxidant micronutrient (or a combination of micronutrients) for reducing cancer risks is unknown. The purpose of this review is to survey the literature on modern biological theories of cancer and the roles of dietary antioxidants in cancer. The roles and functions of antioxidant micronutrients, such as vitamin C (ascorbate), vitamin E (alpha-tocopherol), selenium, and vitamin A, provided through diet for the prevention of cancer are discussed in the present work.


Subject(s)
Antioxidants/pharmacology , Dietary Supplements , Neoplasms/prevention & control , ATP Binding Cassette Transporter 1/metabolism , Angiogenesis Inhibitors/pharmacology , Anticarcinogenic Agents/pharmacology , Ascorbic Acid/pharmacokinetics , Ascorbic Acid/pharmacology , Diet , Humans , Neoplasms/diet therapy , Neoplasms/metabolism , Recommended Dietary Allowances , Selenium/pharmacology , Sodium-Coupled Vitamin C Transporters/metabolism , Vitamin A/pharmacology , Vitamin E/pharmacokinetics , Vitamin E/pharmacology , alpha-Tocopherol/pharmacology
20.
Am J Physiol Cell Physiol ; 311(1): C129-35, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27122159

ABSTRACT

Vitamin C (ascorbic acid, AA) is indispensable for normal metabolism of all mammalian cells including pancreatic acinar cells (PACs). PACs obtain AA from their surroundings via transport across the cell membrane. Chronic alcohol exposure negatively affects body AA homeostasis; it also inhibits uptake of other micronutrients into PACs, but its effect on AA uptake is not clear. We examined this issue using both in vitro (266-6 cells) and in vivo (mice) models of chronic alcohol exposure. First, we determined the relative expression of the AA transporters 1 and 2 [i.e., sodium-dependent vitamin C transporter-1 (SVCT-1) and SVCT-2] in mouse and human PACs and found SVCT-2 to be the predominant transporter. Chronic exposure of 266-6 cells to alcohol significantly inhibited AA uptake and caused a marked reduction in SVCT-2 expression at the protein, mRNA, and heterogeneous nuclear RNA (hnRNA) levels. Similarly, chronic alcohol feeding of mice significantly inhibited AA uptake and caused a marked reduction in level of expression of the SVCT-2 protein, mRNA, and hnRNA. These findings suggest possible involvement of transcriptional mechanism(s) in mediating chronic alcohol effect on AA uptake by PACs. We also observed significant epigenetic changes (histone modifications) in the Slc23a2 gene (reduction in H3K4me3 level and an increase in H3K27me3 level) in the alcohol-exposed 266-6 cells. These findings show that chronic alcohol exposure inhibits PAC AA uptake and that the effect is mediated, in part, at the level of transcription of the Slc23a2 gene and may involve epigenetic mechanism(s).


Subject(s)
Alcohol Drinking/adverse effects , Ascorbic Acid/metabolism , Ethanol/toxicity , Pancreas, Exocrine/drug effects , Sodium-Coupled Vitamin C Transporters/metabolism , Alcohol Drinking/metabolism , Animals , Biological Transport , Cell Line, Tumor , Down-Regulation , Epigenesis, Genetic , Humans , Mice , Models, Animal , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , RNA, Heterogeneous Nuclear/genetics , RNA, Heterogeneous Nuclear/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL