1
|
Akiyama M, Nonomura H, Kamil SH, Ignotz RA. Periosteal Cell Pellet Culture System: A New Technique for Bone Engineering. Cell Transplant 2017; 15:521-32. [PMID: 17121163 DOI: 10.3727/000000006783981765] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To treat bone loss that is induced by disease or wounds, bone grafts are commonly used. In dentistry, guided tissue regeneration is effective in the treatment of periodontal diseases. However, bone resorption after implantation is a major problem with the bone graft and guided tissue regeneration technique. This study examines a cell pellet culture system without exogenous scaffolds for bone regeneration. First, we examined the effect of ascorbic acid on cells. Transmission electron microscopic observation revealed that cells formed a three-dimensional structure of multiple cell layers after 5 weeks of culturing in medium containing 50 μg/ml ascorbic acid with the medium changed every 7 days. A single cell pellet was produced by centrifuging cells that were gathered from 10 tissue culture dishes. Van Gieson staining and collagen type I immunostaining showed that the pellet contained collagen fibers and cells that adhered to the collagen fibers. Several of these cell pellets were implanted subcutaneously on the backs of nude mice for 6 weeks. Histology and immunohistochemistry results indicated new bone formation, vascular invasion, and insular areas of calcification. Bone tissue was surrounded by osteoblasts. The appearance of new bone formation is similar to that seen in intramembranous ossification. The present pellet system is reliable and might solve problems of bone resorption after implantation.
Collapse
Affiliation(s)
- Mari Akiyama
- Center for Tissue Engineering, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | |
Collapse
|
2
|
Sun YQ, Liu J, Zhang H, Huo Y, Lv X, Shi Y, Guo W. A mitochondria-targetable fluorescent probe for dual-channel NO imaging assisted by intracellular cysteine and glutathione. J Am Chem Soc 2014; 136:12520-3. [PMID: 25122520 DOI: 10.1021/ja504156a] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A mitochondria-specific fluorescent probe for NO (1) was synthesized by the direct conjugation of a pyronin dye with one of the amino groups of o-phenylenediamino (OPD). The probe could selectively detect NO over dehydroascorbic acid (DHA), ascorbic acid (AA), and methylglyoxal (MGO) as well as the reactive oxygen/nitrogen species (ROS/RNS) with the significant off-on response due to the production of a red-emission triazole 2. In the presence of cysteine/glutathione (Cys/GSH), 2 could be further transformed into a green-emission aminopyronin 4 and a red-emission thiopyronin 5, respectively. Assisted by intracellular Cys and GSH, the probe demonstrated its potential to monitor mitochondrial NO in a dual-channel mode.
Collapse
Affiliation(s)
- Yuan-Qiang Sun
- School of Chemistry and Chemical Engineering and ‡Institute of Biotechnology, Shanxi University , Taiyuan 030006, China
| | | | | | | | | | | | | |
Collapse
|
3
|
Mortensen A, Lykkesfeldt J. Does vitamin C enhance nitric oxide bioavailability in a tetrahydrobiopterin-dependent manner? In vitro, in vivo and clinical studies. Nitric Oxide 2014; 36:51-7. [PMID: 24333161 DOI: 10.1016/j.niox.2013.12.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/09/2013] [Accepted: 12/03/2013] [Indexed: 12/31/2022]
Abstract
Ascorbate (Asc) has been shown to increase nitric oxide (NO) bioavailability and thereby improve endothelial function in patients showing signs of endothelial dysfunction. Tetrahydrobiopterin (BH₄) is a co-factor of endothelial nitric oxide synthase (eNOS) which may easily become oxidized to the inactive form dihydrobiopterin (BH₂). Asc may increase NO bioavailability by a number of mechanisms involving BH₄ and eNOS. Asc increases BH₄ bioavailability by either reducing oxidized BH₄ or preventing BH₄ from becoming oxidized in the first place. Asc could also increase NO bioavailability in a BH₄-independent manner by increasing eNOS activity by changing its phosphorylation and S-nitrosylation status or by upregulating eNOS expression. In this review, we discuss the putative mechanisms by which Asc may increase NO bioavailability through its interactions with BH₄ and eNOS.
Collapse
Affiliation(s)
- Alan Mortensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens Lykkesfeldt
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
4
|
Meredith ME, Qu ZC, May JM. Ascorbate reverses high glucose- and RAGE-induced leak of the endothelial permeability barrier. Biochem Biophys Res Commun 2014; 445:30-5. [PMID: 24472555 DOI: 10.1016/j.bbrc.2014.01.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/20/2014] [Indexed: 02/07/2023]
Abstract
High glucose concentrations due to diabetes increase leakage of plasma constituents across the endothelial permeability barrier. We sought to determine whether vitamin C, or ascorbic acid (ascorbate), could reverse such high glucose-induced increases in endothelial barrier permeability. Human umbilical vein endothelial cells and two brain endothelial cell lines cultured at 25 mM glucose showed increases in endothelial barrier permeability to radiolabeled inulin compared to cells cultured at 5mM glucose. Acute loading of the cells for 30-60 min with ascorbate before the permeability assay prevented the high glucose-induced increase in permeability and decreased basal permeability at 5mM glucose. High glucose-induced barrier leakage was mediated largely by activation of the receptor for advanced glycation end products (RAGE), since it was prevented by RAGE blockade and mimicked by RAGE ligands. Intracellular ascorbate completely prevented RAGE ligand-induced increases in barrier permeability. The high glucose-induced increase in endothelial barrier permeability was also acutely decreased by several cell-penetrant antioxidants, suggesting that at least part of the ascorbate effect could be due to its ability to act as an antioxidant.
Collapse
Affiliation(s)
- M Elizabeth Meredith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States
| | - Zhi-Chao Qu
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States
| | - James M May
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States.
| |
Collapse
|
5
|
Lv X, Wang Y, Zhang S, Liu Y, Zhang J, Guo W. A specific fluorescent probe for NO based on a new NO-binding group. Chem Commun (Camb) 2014; 50:7499-502. [DOI: 10.1039/c4cc03540b] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A highly selective fluorescent probe for NO was exploited based on a specific NO-binding group, 2-amino-3′-dimethylaminobiphenyl.
Collapse
Affiliation(s)
- Xin Lv
- School of Chemistry and Chemical Engineering
- Shanxi University
- Taiyuan 030006, China
| | - Yue Wang
- School of Chemistry and Chemical Engineering
- Shanxi University
- Taiyuan 030006, China
| | - Song Zhang
- Shanxi Academy of Analytical Sciences
- , China
| | - Yawei Liu
- School of Chemistry and Chemical Engineering
- Shanxi University
- Taiyuan 030006, China
| | - Jian Zhang
- School of Chemistry and Chemical Engineering
- Shanxi University
- Taiyuan 030006, China
| | - Wei Guo
- School of Chemistry and Chemical Engineering
- Shanxi University
- Taiyuan 030006, China
| |
Collapse
|
6
|
Pedersen TO, Blois AL, Xue Y, Xing Z, Cottler-Fox M, Fristad I, Leknes KN, Lorens JB, Mustafa K. Osteogenic stimulatory conditions enhance growth and maturation of endothelial cell microvascular networks in culture with mesenchymal stem cells. J Tissue Eng 2012; 3:2041731412443236. [PMID: 22511994 PMCID: PMC3324846 DOI: 10.1177/2041731412443236] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
To optimize culture conditions for in vitro prevascularization of tissue-engineered bone constructs, the development of organotypic blood vessels under osteogenic stimulatory conditions (OM) was investigated. Coculture of endothelial cells and mesenchymal stem cells was used to assess proangiogenic effects of mesenchymal stem cells on endothelial cells. Four different culture conditions were evaluated for their effect on development of microvascular endothelial cell networks. Mineralization, deposition of extracellular matrix, and perivascular gene expression were studied in OM. After 3 days, endothelial cells established elongated capillary-like networks, and upregulated expression of vascular markers was seen. After 15 days, all parameters evaluated were significantly increased for cultures in OM. Mature networks developed in OM presented lumens enveloped by basement membrane-like collagen IV, with obvious mineralization and upregulated perivascular gene expression from mesenchymal stem cells. Our results suggest osteogenic stimulatory conditions to be appropriate for in vitro development of vascularized bone implants for tissue engineering.
Collapse
Affiliation(s)
- Torbjorn O Pedersen
- Department of Clinical Dentistry-Center for Clinical Dental Research, University of Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Frikke-Schmidt H, Roursgaard M, Lykkesfeldt J, Loft S, Nøjgaard JK, Møller P. Effect of vitamin C and iron chelation on diesel exhaust particle and carbon black induced oxidative damage and cell adhesion molecule expression in human endothelial cells. Toxicol Lett 2011; 203:181-9. [PMID: 21421028 DOI: 10.1016/j.toxlet.2011.03.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 03/04/2011] [Accepted: 03/07/2011] [Indexed: 01/07/2023]
Abstract
Exposure to particulate matter is associated with oxidative stress and risk of cardiovascular diseases. We investigated if vitamin C and desferrioxamine (iron chelator) altered the levels of oxidative stress and expression of cell adhesion molecules upon exposure to diesel exhaust particles (DEP) and carbon black in cultured human umbilical vein endothelial cells (HUVECs). We found that the particles were only slightly cytotoxic in the high concentration ranges. Particle-induced intracellular reactive oxygen species (ROS) production was attenuated by vitamin C administration or iron chelation and particularly when combined (p<0.001). Only desferrioxamine protected the DNA from oxidative damage in terms of strand breaks and formamidopyrimidine DNA glycosylase sensitive sites induced by carbon black (p<0.01). Carbon black and small sized DEP generated from an Euro4 engine increased the surface expression of VCAM-1 and ICAM-1, whereas DEP from an engine representing an old combustion type engine (SRM2975) with larger particles did not affect the expression of cell adhesion molecules. These effects were also attenuated by desferrioxamine but not vitamin C. The study shows that exposure to carbon black and DEP in HUVECs can generate both oxidative stress and expression of cell surface adhesion molecules and that these effects can in part be attenuated by vitamin C and desferrioxamine.
Collapse
Affiliation(s)
- Henriette Frikke-Schmidt
- Section of Biomedicine, Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
8
|
May JM, Qu ZC. Nitric oxide mediates tightening of the endothelial barrier by ascorbic acid. Biochem Biophys Res Commun 2010; 404:701-5. [PMID: 21156160 DOI: 10.1016/j.bbrc.2010.12.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 12/08/2010] [Indexed: 11/29/2022]
Abstract
Vitamin C, or ascorbic acid, decreases paracellular endothelial permeability in a process that requires rearrangement of the actin cytoskeleton. To define the proximal mechanism of this effect, we tested whether it might involve enhanced generation and/or sparing of nitric oxide (NO) by the vitamin. EA.hy926 endothelial cells cultured on semi-porous filter supports showed decreased endothelial barrier permeability to radiolabeled inulin in response to exogenous NO provided by the NO donor spermine NONOATE, as well as to activation of the downstream NO pathway by 8-bromo-cyclic GMP, a cell-penetrant cyclic GMP analog. Inhibition of endothelial nitric oxide synthase (eNOS) with N(ω)-nitro-l-arginine methyl ester increased endothelial permeability, indicating a role constitutive NO generation by eNOS in maintaining the permeability barrier. Inhibition of guanylate cyclase by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one also increased endothelial permeability and blocked barrier tightening by spermine NONOATE. Loading cells with what are likely physiologic concentrations of ascorbate decreased endothelial permeability. This effect was blocked by inhibition of either eNOS or guanylate cyclase, suggesting that it involved generation of NO by eNOS and subsequent NO-dependent activation of guanylate cyclase. These results show that endothelial permeability barrier function depends on constitutive generation of NO and that ascorbate-dependent tightening of this barrier involves maintaining NO through the eNOS/guanylate cyclase pathway.
Collapse
Affiliation(s)
- James M May
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, USA.
| | | |
Collapse
|
9
|
Muellner MK, Schreier SM, Schmidbauer B, Moser M, Quehenberger P, Kapiotis S, Goldenberg H, Laggner H. Vitamin C inhibits NO-induced stabilization of HIF-1alpha in HUVECs. Free Radic Res 2010; 44:783-91. [PMID: 20380593 DOI: 10.3109/10715761003786172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
HIF-1alpha represents the oxygen-regulated sub-unit of the transcription factor HIF-1, which regulates the transcription of numerous genes involved in cellular response to hypoxia and oxidative stress. It is shown here that nitric oxide (NO) induces HIF-1alpha stabilization in human endothelial cells from umbilical cords (HUVECs) under normoxic conditions. HIF-1alpha protein was increased approximately 36-fold after incubation with 500 microM DETA-NO, which releases a steady state NO concentration of roughly one thousandth of the initial concentration of the donor. Loading of the cells with vitamin C counteracted NO-induced HIF-1alpha accumulation. Based on the observations that oxidative and nitrosative stress can influence the activity of the proteasomal system, which is responsible for the non-lysosomal degradation of proteins, among them HIF-1alpha, it was investigated whether NO-induced stabilization of HIF-1alpha might be due to reduced 20S proteasomal activity. This hypothesis could not be proved, because NO concentrations to inhibit 20S proteasomal activity were about one order of magnitude higher than that to inhibit HIF-1alpha degradation.
Collapse
Affiliation(s)
- Markus K Muellner
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Corti A, Casini AF, Pompella A. Cellular pathways for transport and efflux of ascorbate and dehydroascorbate. Arch Biochem Biophys 2010; 500:107-15. [PMID: 20494648 DOI: 10.1016/j.abb.2010.05.014] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 11/17/2022]
Abstract
The mechanisms allowing the cellular transport of ascorbic acid represent a primary aspect for the understanding of the roles played by this vitamin in pathophysiology. Considerable research effort has been spent in the field, on several animal models and different cell types. Several mechanisms have been described to date, mediating the movements of different redox forms of ascorbic acid across cell membranes. Vitamin C can enter cells both in its reduced and oxidized form, ascorbic acid (AA) and dehydroascorbate (DHA), utilizing respectively sodium-dependent transporters (SVCT) or glucose transporters (GLUT). Modulation of SVCT expression and function has been described by cytokines, steroids and post-translational protein modification. Cellular uptake of DHA is followed by its intracellular reduction to AA by several enzymatic and non-enzymatic systems. Efflux of vitamin C has been also described in a number of cell types and different pathophysiological functions were proposed for this phenomenon, in dependence of the cell model studied. Cellular efflux of AA is mediated through volume-sensitive (VSOAC) and Ca(2+)-dependent anion channels, gap-junction hemichannels, exocytosis of secretory vesicles and possibly through homo- and hetero-exchange systems at the plasma membrane level. Altogether, available data suggest that cellular efflux of ascorbic acid - besides its uptake - should be taken into account when evaluating the cellular homeostasis and functions of this important vitamin.
Collapse
Affiliation(s)
- Alessandro Corti
- Dipartimento di Patologia Sperimentale, Università di Pisa, Italy.
| | | | | |
Collapse
|
11
|
May JM, Qu ZC, Qiao H. Transfer of ascorbic acid across the vascular endothelium: mechanism and self-regulation. Am J Physiol Cell Physiol 2009; 297:C169-78. [PMID: 19419995 DOI: 10.1152/ajpcell.00674.2008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine how ascorbic acid moves from the bloodstream into tissues, we assessed transfer of the vitamin across the barrier generated by EA.hy926 endothelial cells when these were cultured on semipermeable filter supports. Ascorbate transfer from the luminal to the abluminal compartment was time dependent, inhibited by anion channel blockers and by activation of protein kinase A, but was increased by thrombin. Ascorbate transfer occurred by a paracellular route, since it did not correlate with intracellular ascorbate contents and was not rectified or saturable. Nonetheless, intracellular ascorbate inhibited the transfer of both ascorbate and radiolabeled inulin across the endothelial barrier. The increase in barrier function due to ascorbate was dependent on its intracellular concentration, significant by 15 min of incubation, prevented by the cytoskeletal inhibitor colchicine, associated with F-actin stress fiber formation, and not due to collagen deposition. These results show that ascorbate traverses the endothelial barrier by a paracellular route that is regulated by cell metabolism, ion channels, and ascorbate itself. Since the latter effect occurred over the physiological range of ascorbate plasma concentrations, it could reflect a role for the vitamin in control of endothelial barrier function in vivo.
Collapse
Affiliation(s)
- James M May
- Department of Medicine, Vanderbilt Univ. School of Medicine, Nashville, TN 37232-0475, USA.
| | | | | |
Collapse
|
12
|
Aguirre R, May JM. Inflammation in the vascular bed: importance of vitamin C. Pharmacol Ther 2008; 119:96-103. [PMID: 18582947 DOI: 10.1016/j.pharmthera.2008.05.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 05/09/2008] [Indexed: 02/07/2023]
Abstract
Despite decreases in atherosclerotic coronary vascular disease over the last several decades, atherosclerosis remains a major cause of mortality in developed nations. One possible contributor to this residual risk is oxidant stress, which is generated by the inflammatory response of atherosclerosis. Although there is a wealth of in vitro, cellular, and animal data supporting a protective role for antioxidant vitamins and nutrients in the atherosclerotic process, the best clinical trials have been negative. This may be due to the fact that antioxidant therapies are applied "too little and too late." This review considers the role of vitamin C, or ascorbic acid in preventing the earliest inflammatory changes in atherosclerosis. It focuses on the three major vascular cell types involved in atherosclerosis: endothelial cells, vascular smooth muscle cells, and macrophages. Ascorbate chemistry, recycling, and function are described for these cell types, with emphasis on whether and how the vitamin might affect the inflammatory process. For endothelial cells, ascorbate helps to prevent endothelial dysfunction, stimulates type IV collagen synthesis, and enhances cell proliferation. For vascular smooth muscle cells, ascorbate inhibits dedifferentiation, recruitment, and proliferation in areas of vascular damage. For macrophages, ascorbate decreases oxidant stress related to their activation, decreases uptake and degradation of oxidized LDL in some studies, and enhances several aspects of their function. Although further studies of ascorbate function in these cell types and in novel animal models are needed, available evidence generally supports a salutary role for this vitamin in ameliorating the earliest stages of atherosclerosis.
Collapse
Affiliation(s)
- Rene Aguirre
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-6303, USA
| | | |
Collapse
|
13
|
McCarty MF. Scavenging of peroxynitrite-derived radicals by flavonoids may support endothelial NO synthase activity, contributing to the vascular protection associated with high fruit and vegetable intakes. Med Hypotheses 2008; 70:170-81. [PMID: 17825500 DOI: 10.1016/j.mehy.2005.09.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Accepted: 09/07/2005] [Indexed: 01/24/2023]
Abstract
Ample intakes of fruit and vegetables have been linked epidemiologically with reduced risk for coronary disease, stroke, hypertension, obesity, many types of cancer, chronic pulmonary disease, osteoporosis, and various ocular disorders. The favorable impact of diets rich in fruit and vegetables on coronary risk has been confirmed in meta-analyses, and is thought to be largely attributable to the folk acid and potassium supplied by these foods. Although high intakes of vitamin C appear to confer some cardiovascular protection, the amounts supplied by typical diets may be too low to be of much benefit in this regard. High flavonoid intakes emerge as protective in some epidemiological studies, albeit the dose-response pattern observed is often L-shaped - seemingly more consistent with low intakes being harmful, than with high intakes being protective. Nonetheless, flavonoids have shown anti-atherogenic activity in rodent models, and both clinical and rodent supplementation studies with foods and food extracts rich in flavonoids demonstrate improvements in endothelium-dependent vasodilation traceable to increased endothelial nitric oxide synthesis. However, flavonoids do not appear to increase the expression of endothelial NO synthase, nor do they modify endothelial superoxide production. A likely explanation is that, even in nanomolar concentrations achievable in vivo, flavonoids can act as efficient scavengers of peroxynitrite-derived radicals, thereby protecting the cofactor tetrahydrobiopterin, crucial for NO synthase activity. Studies with cultured endothelial cells should be useful for evaluating this possibility. It would also be appropriate to assess the effects of flavonoids on prostacylin synthetase activity, on endothelial catabolism of asymmetric dimethylarginine, and on signaling mechanisms that activate NO synthase. Since peroxynitrite can induce mutagenic damage to DNA, it is conceivable that scavenging of peroxynitrite-derived radicals contributes to the reduction in mutagenesis associated with high intakes of fruits and vegetables. Carotenoids also have the potential to prevent peroxynitrite-mediated damage, although, as contrasted with flavonoids, there is comparatively little evidence that these compounds are anti-atherogenic or beneficial for endothelial function; a recent meta-analysis of epidemiological studies suggests that high lutein intakes may modestly reduce coronary risk.
Collapse
Affiliation(s)
- Mark F McCarty
- Natural Alternatives International, 1185 Linda Vista Dr., San Marcos, CA 92078, United States.
| |
Collapse
|
14
|
Ye X, Rubakhin SS, Sweedler JV. Simultaneous nitric oxide and dehydroascorbic acid imaging by combining diaminofluoresceins and diaminorhodamines. J Neurosci Methods 2007; 168:373-82. [PMID: 18083236 DOI: 10.1016/j.jneumeth.2007.10.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 10/30/2007] [Accepted: 10/30/2007] [Indexed: 10/22/2022]
Abstract
Spatial measurements of nitric oxide (NO) production are important to understand the function and metabolism of this molecule. The reagent, 4,5-diaminofluorescein (DAF-2) and several structurally similar probes are widely used for detection and imaging of NO. However, DAF-2 also reacts with dehydroascorbic acid (DHA) in biological samples, with both products having nearly indistinguishable fluorescence spectra. Measurements using fluorimetry and fluorescence microscopy cannot easily differentiate NO-related fluorescent signals from DHA-related signals. While DAFs and the structurally related diaminorhodamines (DARs) both react with NO and DHA, they do so to different extents. We report a multiderivatization method to image NO and DHA simultaneously by using both DAF and DAR. Specifically, DAF-2 and DAR-4M are used to image NO and DHA concentrations; after reaction, the solutions are excited, at 495 nm to measure fluorescence emission from DAF-2, and at 560 nm to measure fluorescence emission from DAR-4M. Using the appropriate calibrations, images are created that depend either on the relative NO or the relative DHA concentration, even though each probe reacts to both compounds. The method has been validated by imaging NO production in both undifferentiated and differentiated pheochromocytoma (PC12) cells.
Collapse
Affiliation(s)
- Xiaoying Ye
- Department of Chemistry and the Beckman Institute, University of Illinois, 600 South Mathews Avenue 63-5, Urbana, IL 61801, USA
| | | | | |
Collapse
|
15
|
Edwards DH, Chaytor AT, Bakker LM, Griffith TM. Modulation of gap-junction-dependent arterial relaxation by ascorbic acid. J Vasc Res 2007; 44:410-22. [PMID: 17587861 DOI: 10.1159/000104254] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Accepted: 03/19/2007] [Indexed: 11/19/2022] Open
Abstract
AIMS To investigate whether ascorbic acid (AA) can influence endothelium-dependent relaxation by modulating the spread of endothelial hyperpolarization through the arterial wall via gap junctions. METHODS Force development and membrane potential were monitored by myography and sharp electrode techniques in isolated rabbit iliac arteries. RESULTS AA prevented the ability of the gap junction blocker 2-aminoethoxydiphenyl borate to inhibit endothelium-dependent relaxations and subintimal smooth muscle hyperpolarizations evoked by cyclopiazonic acid in the presence of nitric oxide (NO) synthase and cyclooxygenase blockade. AA also prevented the ability of a connexin-mimetic peptide targeted against Cx37 and Cx40 (37,40Gap 26) to attenuate the transmission of endothelial hyperpolarization to subintimal smooth muscle, and a peptide targeted against Cx43 (43Gap 26) to attenuate the spread of subintimal hyperpolarization to subadventitial smooth muscle and the associated mechanical relaxation. Parallel studies with endothelium-denuded preparations demonstrated that AA and cyclopiazonic acid both depressed relaxation evoked by the NO donor MAHMA NONOate. CONCLUSIONS The data suggest that AA can modulate arterial function through a previously unrecognized ability to preserve electrotonic signalling via myoendothelial and homocellular smooth muscle gap junctions under conditions where cell coupling is depressed. Underlying mechanisms do not involve amplification of 'residual' NO activity by AA.
Collapse
Affiliation(s)
- David H Edwards
- Department of Diagnostic Radiology, Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | | | | | | |
Collapse
|
16
|
Vissers MCM, Gunningham SP, Morrison MJ, Dachs GU, Currie MJ. Modulation of hypoxia-inducible factor-1 alpha in cultured primary cells by intracellular ascorbate. Free Radic Biol Med 2007; 42:765-72. [PMID: 17320759 DOI: 10.1016/j.freeradbiomed.2006.11.023] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 10/19/2006] [Accepted: 11/25/2006] [Indexed: 11/29/2022]
Abstract
Control of the transcription factor hypoxia inducible factor (HIF)-1 is mediated by hydroxylation by proline and asparagine hydroxylases. These enzymes require ascorbate for optimal activity, but little attention has been given to the effect of ascorbate on HIF-1 activation. Furthermore, cells in culture are ascorbate deficient. We investigated the effect of intracellular ascorbate on HIF-1alpha protein levels and on HIF-1-mediated gene expression in two human primary cell lines (umbilical vein endothelial cells and skin fibroblasts) and one human cancer cell line (A431 epithelial cells). Under normal culture conditions the cells contained no ascorbate and adding ascorbate to the medium increased intracellular concentrations in a dose-dependent manner. A basal level of HIF-1alpha detected in nonsupplemented cells under normoxic conditions disappeared when 10 microM ascorbate was added to the medium. Induction of HIF-1alpha by hypoxia (1% O(2)) or by CoCl(2) was markedly inhibited by ascorbate and loading with physiological levels resulted in almost complete reversal of HIF-1alpha stabilisation. Gene expression was similarly affected, with VEGF mRNA and GLUT-1 up-regulation being inhibited by ascorbate. Hence intracellular ascorbate is a major regulator of the hypoxic response in normal cells and optimal levels of this vitamin will have a profound effect on HIF-1-regulated processes.
Collapse
Affiliation(s)
- Margret C M Vissers
- Free Radical Research Group, Pathology Department, Christchurch School of Medicine and Health Sciences, Christchurch, New Zealand.
| | | | | | | | | |
Collapse
|
17
|
McCarty MF, Barroso-Aranda J, Contreras F. A two-phase strategy for treatment of oxidant-dependent cancers. Med Hypotheses 2007; 69:489-96. [PMID: 17502128 DOI: 10.1016/j.mehy.2006.12.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 12/18/2006] [Indexed: 02/09/2023]
Abstract
In many cancers, a chronic increase in oxidant stress - associated with elevated levels of hydrogen peroxide - contributes to the increased proliferative rate, diminished apoptosis, increased angiogenic and metastatic capacity, and chemoresistance that often characterize advanced malignancies. This oxidant stress often reflects up-regulation of expression and activity of NADPH oxidase, and/or decreased activity of catalase, which functions as suppressor gene in oxidant-dependent cancers. These characteristics of oxidant-dependent cancers suggest a dual strategy for treatment of these cancers. Since ascorbate can react spontaneously with molecular oxygen to generate hydrogen peroxide, high-dose intravenous ascorbate should be selectively toxic to tumors that are low in catalase activity - as suggested by numerous cell culture studies. Measures which concurrently improve the oxygenation of hypoxic tumor regions would be expected to boost the efficacy of such therapy; calcitriol and high-dose selenium might also be useful in this regard. Secondly, during the intervals between sessions of ascorbate therapy, administration of agents which can safely inhibit NADPH oxidase would be expected to slow the proliferation and spread of surviving tumor cells - while providing selection pressure for a further decline in catalase activity. In effect, cancers treated in this way would be whipsawed between lethally excessive and inadequately low oxidant stress. An additional possibility is that ascorbate-induced oxidant stress in tumors might potentiate the cell kill achieved with concurrently administered cytotoxic drugs, inasmuch as oxidant mechanisms appear to play a mediating role in the apoptosis induced by many such drugs, largely via activation of c-Jun NH(2)-terminal kinase; cell culture studies would be useful for evaluating this possibility.
Collapse
|
18
|
McCarty MF. High-dose folate may improve platelet function in acute coronary syndrome and other pathologies associated with increased platelet oxidative stress. Med Hypotheses 2007; 69:12-9. [PMID: 17293058 DOI: 10.1016/j.mehy.2004.08.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2004] [Accepted: 08/13/2004] [Indexed: 11/27/2022]
Abstract
Although nitric oxide of endothelial origin plays a major role in warding off inappropriate thrombus formation, platelets also express the "constitutive" isoform of nitric oxide synthase (cNOS). Activation of this enzyme by calcium influx during platelet aggregation provides an important feedback signal that dampens platelet recruitment. Platelets also express a membrane-bound NAD(P)H oxidase complex, activated by collagen receptors, that produces superoxide. Superoxide can directly quench NO; moreover, by giving rise to peroxynitrite, it can oxidize the cNOS cofactor tetrahydrobiopterin (BH4), thereby suppressing cNOS activity and converting it to superoxide generator. In a canine model of acute coronary syndrome, infusion of BH4 has been shown to prevent thrombus formation. Platelets from patients with acute coronary syndrome produce markedly less NO than do control platelets. A reasonable explanation for these findings is that episodic contact with collagen boosts platelet superoxide production, oxidizing BH4. Since 5-methyltetrahydrofolate can reduce oxidized BH4, or otherwise compensate for its deficiency, supplementation with its precursor folic acid may improve platelet function in acute coronary syndrome and possibly reduce risk for coronary thrombosis in other at-risk patients. Other research demonstrates that superoxide production is increased, and nitric oxide production diminished, in platelets of diabetics; the ability of glutathione--a peroxynitrite scavenger--to largely ameliorate these abnormalities, is consistent with a prominent role for BH4 deficiency in diabetic platelet malfunction. Reports that platelet NO production is decreased, and/or superoxide production increased, in patients with disorders associated with insulin resistance syndrome, suggest that BH4 deficiency--potentially remediable with high-dose folate--may likewise contribute to the platelet hyperreactivity noted in these disorders. Supplemental vitamin C and arginine also have the potential to boost platelet production of NO Increased intakes of taurine, magnesium, gamma-tocopherol, fish oil, and garlic may help to stabilize platelets by additional mechanisms. As a complement to the proven benefits of low-dose aspirin, a supplemental regimen emphasizing these nutrients in appropriate doses may act directly on platelets to further diminish risk for thrombotic episodes.
Collapse
Affiliation(s)
- Mark F McCarty
- Natural Alternatives International, 1185 Linda Vista Road, San Marcos, CA 92078, USA.
| |
Collapse
|
19
|
Gokhalé P, Patel T, Morrison MJ, Vissers MCM. The effect of intracellular ascorbate on the susceptibility of HL60 and Jurkat cells to chemotherapy agents. Apoptosis 2006; 11:1737-46. [PMID: 16951922 DOI: 10.1007/s10495-006-9787-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chemotherapy agents initiate tumour cell apoptosis and this is thought to involve oxidative stress. In this study we have investigated the effect of the important antioxidant Vitamin C (ascorbate) on the response of HL60 and Jurkat cells to three chemotherapy drugs, namely etoposide, melphalan and arsenic trioxide (As(2)O(3)). Cells grown in routine culture media are deficient in ascorbate and to determine its effect on chemotherapy drug-induced apoptosis we supplemented the cells prior to drug exposure. We found that ascorbate had a varied effect on apoptosis and cell cycle progression. Etoposide-induced apoptosis in HL60 cells was significantly increased in ascorbate-loaded cells as measured by caspase-3 activation and DNA degradation, and this appeared to reflect a decrease in the number of necrotic cells rather than increased cytotoxicity. In contrast, ascorbate had no effect on etoposide-induced apoptosis in Jurkat cells. In both cell types melphalan-induced apoptosis was unaffected by intracellular ascorbate, whereas both apoptosis and growth arrest with low concentrations of As(2)O(3) were diminished. These results indicate that intracellular ascorbate can affect cell responses to chemotherapy drugs in a complex and somewhat unpredictable manner and that it may play an important role in the responsiveness of tumour cells to chemotherapy regimes.
Collapse
Affiliation(s)
- Prachee Gokhalé
- Free Radical Research Group, Pathology Department, Christchurch School of Medicine and Health Sciences, P.O. Box 4345, Christchurch, New Zealand
| | | | | | | |
Collapse
|
20
|
Best KA, Holmes ME, Samson SE, Mwanjewe J, Wilson JX, Dixon SJ, Grover AK. Ascorbate uptake in pig coronary artery endothelial cells. Mol Cell Biochem 2005; 271:43-9. [PMID: 15881654 DOI: 10.1007/s11010-005-3442-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although smooth muscle and endothelial cells in pig coronary artery are morphologically and functionally distinct, ascorbate uptake has been characterized only in smooth muscle cells. Ascorbate transporters in kidney and intestinal epithelial cells differ from those in smooth muscle. We examined ascorbate transport and mRNA expression of sodium-dependent vitamin C transporters (SVCT) by RT-PCR in the pig coronary artery endothelial cell cultures. When 14C-ascorbate uptake in endothelial cells was examined as 14C or by HPLC, the two values did not differ from each other. 14C-ascorbate uptake was Na(+)-dependent, stereoselective for L-ascorbate and inhibited by sulfinpyrazone. The kinetic characteristics of the uptake were: Km = 27 +/- 3 microM (Hill coefficient = 1) for ascorbate and Km = 73 +/- 14 mM (Hill coefficient = 2) for Na+. Surprisingly, endothelial cells had similar kinetic parameters as smooth muscle cells, except for a slightly lower uptake velocity in endothelial cells. Comparison with the smooth muscle showed that both tissue types expressed mRNA for SVCT2. Endothelial cells differ from epithelial cells which express mainly SVCT1 but resemble smooth muscle cells in this respect.
Collapse
Affiliation(s)
- Kelly A Best
- Department of Biology, McMaster University, Hamilton Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
21
|
May JM, Qu ZC. Transport and intracellular accumulation of vitamin C in endothelial cells: relevance to collagen synthesis. Arch Biochem Biophys 2005; 434:178-86. [PMID: 15629121 DOI: 10.1016/j.abb.2004.10.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Revised: 10/13/2004] [Indexed: 11/19/2022]
Abstract
Endothelial cells preserve vascular integrity in part by synthesizing type IV collagen for the basement membrane of blood vessels. Vitamin C, which at physiologic pH is largely the ascorbate mono-anion, both protects these cells from oxidant stress and is required for collagen synthesis. Therefore, cultured endothelial cells were used to correlate intracellular concentrations of ascorbate with its uptake and ability to stimulate collagen release into the culture medium. The kinetics and inhibitor specificity of ascorbate transport into EA.hy926 endothelial cells were similar to those observed in other cell types, indicative of a specific high affinity transport process. Further, transport of the vitamin generated intracellular ascorbate concentrations that were 80-100-fold higher than concentrations in the medium following overnight culture, and transport inhibition with sulfinpyrazone and phloretin partially prevented such ascorbate accumulation. On the other hand, low millimolar intracellular concentrations of ascorbate impaired its transport measured after overnight culture. Synthesis and release of type IV collagen into the culture medium was markedly stimulated by ascorbate in a time-dependent manner, and was saturable with increasing medium concentrations of the vitamin. Optimal rates of collagen synthesis required intracellular concentrations of the vitamin up to 2 mM. Since such concentrations can only be generated by the ascorbate transporter, these results show the necessity of transport for this crucial function of the vitamin in endothelium.
Collapse
Affiliation(s)
- James M May
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-6303, USA.
| | | |
Collapse
|
22
|
Varadharaj S, Watkins T, Cardounel AJ, Garcia JGN, Zweier JL, Kuppusamy P, Natarajan V, Parinandi NL. Vitamin C-induced loss of redox-dependent viability in lung microvascular endothelial cells. Antioxid Redox Signal 2005; 7:287-300. [PMID: 15650416 DOI: 10.1089/ars.2005.7.287] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Recent clinical trials have shown that vitamin C, at pharmacological concentrations (milligram to approximately gram), upon infusion into circulation, modulates vasodilation and vascular tone in humans. This also results in the elevated concentrations of vitamin C in circulation in the millimolar range. Here, it was hypothesized that vitamin C at pharmacological concentrations (millimolar) would induce oxidative stress and cause loss of redox-dependent cell viability in vascular endothelial cells (ECs). To test the hypothesis, bovine lung microvascular ECs (BLMVECs) in monolayer cultures were exposed to vitamin C (0-10 mM) for different time periods (0-2 h). Electron paramagnetic resonance spectroscopy revealed the intracellular formation of ascorbate free radical in a dose- and time-dependent fashion. Vitamin C also induced formation of intracellular reactive oxygen species in a dose-dependent fashion. It was observed that vitamin C induced morphological alterations and loss of cell viability in a dose- and time-dependent fashion, as measured by light microscopy and Alamar Blue redox cell viability assay, respectively. Vitamin C analogues failed to induce such changes. Vitamin C depleted cellular GSH levels in a dose-dependent fashion, suggesting that vitamin C altered thiol-redox status in BLMVECs. Antioxidants, intracellular iron chelator, and catalase protected cells against vitamin C-induced loss of redox-dependent cell viability, confirming the role of hydrogen peroxide and iron during redox cycling of vitamin C. These results, for the first time in detail, established that vitamin C at pharmacological doses induced oxidative stress and loss of redox-dependent cell viability in microvascular ECs.
Collapse
Affiliation(s)
- Saradhadevi Varadharaj
- Lipid Signaling and Lipomics Laboratory, Division of Pulmonary, Critical Care and Sleep Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210-1252, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Ulker S, McMaster D, McKeown PP, Bayraktutan U. Antioxidant vitamins C and E ameliorate hyperglycaemia-induced oxidative stress in coronary endothelial cells. Diabetes Obes Metab 2004; 6:442-51. [PMID: 15479220 DOI: 10.1111/j.1462-8902.2004.00443.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Vitamins C and E have protective features in many disease states associated with enhanced oxidative stress. The aim of this study was to investigate whether vitamin(s) C and/or E modulate hyperglycaemia-induced oxidative stress by regulating enzymatic activities of prooxidant, i.e. NAD(P)H oxidase and/or antioxidant enzymes, namely endothelial nitric oxide synthase (eNOS), superoxide dismutase, catalase and glutathione peroxidase, using coronary microvascular endothelial cells (CMEC). METHODS CMEC were cultured under normal (5.5 mM) or high glucose (22 mM) concentrations for 7 days. The enzyme activities were determined by specific assays. The levels of O(2) (-) and nitrite were measured by cytochrome c reduction and Griess assays respectively. RESULTS Hyperglycaemia did not alter eNOS activity or overall nitrite generation, an index of NO production. However, it increased NAD(P)H oxidase and antioxidant enzyme activities (p < 0.05). Specific inhibitors of NAD(P)H oxidase, i.e. phenylarsine oxide (0.1-3 microm) and 4-(2-aminoethyl)benzenesulfonyl fluoride (5-100 microm) and vitamins C and E (0.1-1 microm) significantly reduced prooxidant and antioxidant enzyme activities in CMEC exposed to hyperglycaemia (p < 0.01). The differences in enzyme activities were independent of increases in osmolarity generated by high glucose levels as investigated by using equimolar concentrations of mannitol in parallel experiments. CONCLUSIONS Vitamins C and E may protect CMEC against hyperglycaemia-induced oxidative stress by concomitantly regulating prooxidant and antioxidant enzyme activities.
Collapse
Affiliation(s)
- S Ulker
- Department of Medicine, Institute of Clinical Science Block B, Queen's University Belfast, Belfast BT12 6BJ, UK
| | | | | | | |
Collapse
|
24
|
Seno T, Inoue N, Matsui K, Ejiri J, Hirata KI, Kawashima S, Yokoyama M. Functional expression of sodium-dependent vitamin C transporter 2 in human endothelial cells. J Vasc Res 2004; 41:345-51. [PMID: 15340249 DOI: 10.1159/000080525] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Accepted: 07/15/2004] [Indexed: 02/05/2023] Open
Abstract
Since oxidative stress plays an important role in dysregulation of the microcirculation as well as the pathogenesis of atherosclerosis, therapeutic intervention with antioxidants has been speculated to prevent cardiovascular diseases. Ascorbic acid (AA) has been reported to improve endothelial function; however, its intracellular metabolic pathway has not been fully determined. Sodium-dependent vitamin C transporter (SVCT) types 1 and 2 were recently cloned. In the present study, we investigated whether SVCT-2 is functionally expressed in vascular endothelial cells and, if so, what factors modulate its activity. The uptake of AA into human umbilical vein endothelial cells (HUVECs) was examined by incubation with radiolabeled AA (14C-AA). AA was transported into HUVECs in a dose- and time-dependent manner. Replacement of sodium chloride with choline chloride in the medium suppressed the uptake of AA. RT-PCR revealed that HUVECs expressed SVCT-2 mRNA, but not SVCT-1. Transfection of HUVECs with the antisense oligonucleotide of SVCT-2 significantly suppressed the uptake of AA. Furthermore, tumor necrosis factor-alpha and interleukin-1beta inhibited the transport activity of AA. Thus, SVCT-2 is functionally expressed in human endothelial cells, and its activity is negatively regulated by inflammatory cytokines. Our findings might provide a new insight into understanding the treatment of cardiovascular diseases with AA.
Collapse
Affiliation(s)
- Tadashi Seno
- Division of Cardiovascular and Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Valent S, Tóth M. Spectrophotometric analysis of the protective effect of ascorbate against spontaneous oxidation of tetrahydrobiopterin in aqueous solution: kinetic characteristics and potentiation by catalase of ascorbate action. Int J Biochem Cell Biol 2004; 36:1266-80. [PMID: 15109571 DOI: 10.1016/j.biocel.2003.10.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2003] [Revised: 10/15/2003] [Accepted: 10/29/2003] [Indexed: 11/28/2022]
Abstract
Tetrahydrobiopterin (BH(4)) is oxidized by O(2) readily in aqueous solutions and physiological concentrations of ascorbate have been shown to inhibit this reaction. In order to gain insight into the mechanism of ascorbate effect, a spectrophotometric analysis was applied for the study of the time course of BH(4) oxidation in the presence of various concentrations of ascorbate and the effect of various temperatures on the apparent second-order rate constant of BH(4) oxidation (k(ox)) in the presence or absence of catalase. In 100 micromol/l concentration, ascorbate alone prolonged the half-life time of 36 micromol/l BH(4) 1.4-fold whereas in the presence of catalase 1.85-fold. In the presence of catalase ascorbate decreased the value of k(ox) to 51 +/- 0.67%, whereas in the absence of it only to 64 +/- 0.77% of control (P < 0.01). The extent of ascorbate effect was not dependent on temperature, at least between 22 and 37 degrees C, either in the presence or absence of catalase. In the absence of catalase the apparent Arrhenius activation energies: 57.02 +/- 0.09 kJ/mol (-ascorbate) and 56.77 +/- 2.21 kJ/mol (+ascorbate) whereas in the presence of catalase: 62.72 +/- 1.37 kJ/mol (-ascorbate) and 59.93 +/- 2.84 kJ/mol (+ascorbate, mean +/- S.E.M., n=3) were obtained. The study shows that catalase potentiates the BH(4)-stabilizing effect of ascorbate. It is concluded that removal of H(2)O(2) generated from BH(4) during oxidation by O(2) prevents a decrease of ascorbate concentration, and in the presence of ascorbate the pacemaker step in the overall reaction is the oxidation of BH(4) and not the reduction of the quinonoid BH(2) back to BH(4) by ascorbate.
Collapse
Affiliation(s)
- Sándor Valent
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, P.O. Box 260, H-1444 Budapest 8, Hungary
| | | |
Collapse
|
26
|
McCarty MF. Coping with endothelial superoxide: potential complementarity of arginine and high-dose folate. Med Hypotheses 2004; 63:709-18. [PMID: 15325022 DOI: 10.1016/j.mehy.2002.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2002] [Accepted: 11/11/2002] [Indexed: 01/31/2023]
Abstract
Superoxide overproduction is a prominent mediator of the endothelial dysfunction associated with a range of vascular disorders, acting in a number of complementary ways to inhibit effective endothelial nitric oxide (NO) activity. The ability of superoxide to quench NO is well known, but oxidants derived from superoxide also appear to inhibit dimethylarginine dimethylaminohydrolase (DDAH) and to oxidize tetrahydrobiopterin (THBP). The former effect boosts the level of methylated arginines that act as potent competitive inhibitors of NO synthase, whereas the latter effect decreases the ability of this enzyme to generate NO, while converting it to a form that readily generates superoxide. The adverse impact of DDAH deficiency on NO production can be offset with supplemental arginine. Although supplementation with THBP has the potential to compensate for the rapid oxidative destruction of this compound, and maintaining optimal vitamin C nutrition may protect or restore the endothelial THBP pool to a limited extent, the most practical way to optimize NO synthase activity in the context of THBP deficit may be administration of high-dose folic acid. The primary circulating metabolite of folate, 5-methyltetrahydrofolate (5MTHF), is structurally analogous to THBP, and appears to normalize the activity of NO synthase in THBP-depleted endothelial cells, either because it "pinch hits" for the absent THBP, or interacts allosterically with NO synthase in some other way to promote the proper function of this enzyme. This observation may rationalize recent clinical studies showing a favorable effect of oral folic acid (5-10 mg daily) on dysfunctional endothelium, independent of any concurrent modulation of homocysteine levels. A recent study reports that, whereas either arginine or THBP alone have only a modest impact on dysfunctional aortic endothelium derived from hypercholesterolemic mice, the combination of the two produces a complete normalization of endothelial function. In aggregate, these considerations suggest that joint administration of arginine and high-dose folate may represent a fruitful approach to preventing and treating vascular disorders - albeit the underlying overproduction of superoxide should also be addressed by ameliorating relevant vascular risk factors.
Collapse
Affiliation(s)
- Mark F McCarty
- Pantox Laboratories, 4622 Santa Fe St., San Diego, CA 92109, USA.
| |
Collapse
|
27
|
Plotnick GD, Corretti MC, Vogel RA, Hesslink R, Wise JA. Effect of supplemental phytonutrients on impairment of the flow-mediated brachial artery vasoactivity after a single high-fat meal. J Am Coll Cardiol 2003; 41:1744-9. [PMID: 12767658 DOI: 10.1016/s0735-1097(03)00302-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES Our objective was to determine if long-term daily administration of phytonutrient supplements can prevent the immediate adverse impact of a high-fat meal and increase the production of nitric oxide. BACKGROUND Ingestion of a high-fat meal impairs flow-mediated vasodilation of the brachial artery for at least 4 h; however, co-ingestion of vitamin antioxidants or a green salad has been shown to prevent this effect. METHODS Flow-mediated brachial artery reactivity test (BART) both before and 3 h after a 900 calorie 50 g fat meal was evaluated in 38 healthy volunteers (age 36.4 +/- 10.1 years). Subjects were randomized to four weeks of daily supplementation with a powdered fruit vegetable juice concentrate (Juice Plus [JP]) along with a complex supplement providing nutritional antioxidants and various herbal extracts (Vineyard [V]), JP alone, or a matching placebo. At three and four weeks, BART was repeated both before and after the high-fat meal. Serum nitrate/nitrite concentrations were measured at baseline and at four weeks. RESULTS Four weeks of the JP-V combination blunted the detrimental effect of the high-fat meal (-47.5 +/- 23.4% at baseline vs. -1.7 +/- 9.7% at four weeks [p < 0.05]). Four weeks of JP alone had a similar beneficial effect (-45.1 +/- 19.7% at baseline vs. -16.6 +/- 10.3% at four weeks [p < 0.05]), whereas there was no substantial effect of the placebo. In the subjects treated with supplements, concentrations of serum nitrate/nitrite increased from 78 +/- 39 to 114 +/- 62 microm/l (p < 0.02). CONCLUSIONS Daily ingestion of modest amounts of a fruit/vegetable juice concentrate with or without adjunctive phytonutrient supplementation can reduce the immediate adverse impact of high-fat meals on flow-mediated vasoactivity and increase nitrate/nitrite blood concentration.
Collapse
Affiliation(s)
- Gary D Plotnick
- Cardiology Division, University of Maryland School of Medicine, Medical Center, 22 South Greene Street, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Endothelial cells are exposed to potentially damaging reactive oxygen species generated both within the cells and in the bloodstream and underlying vessel wall. In this work, we studied the ability of ascorbic acid to protect cultured human-derived endothelial cells (EA.hy926) from oxidant stress generated by the redox cycling agent menadione. Menadione caused intracellular oxidation of dihydrofluorescein, which required the presence of D-glucose in the incubation medium, and was inhibited by intracellular ascorbate and desferrioxamine. At concentrations of 100 microM and higher, menadione depleted the cells of both GSH and ascorbate, and ascorbate loading partially prevented the decrease in GSH due to menadione. Menadione increased L-arginine uptake by the cells, but inhibited endothelial nitric oxide synthase, an effect that was prevented by acute loading with ascorbate. Ascorbate blunts menadione-induced oxidant stress in EA.hy926 cells, which may help to preserve nitric oxide synthase activity under conditions of excessive oxidant stress.
Collapse
Affiliation(s)
- James M May
- Department of Medicine, Vanderbilt University School of Medicine, 715 Preston Research Building, Nashville, TN 37232-6303, USA.
| | | | | |
Collapse
|
29
|
Heller a R, Werner b E. Ascorbic Acid and Endothelial NO Synthesis. Antioxidants (Basel) 2003. [DOI: 10.1201/9781439822173.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
30
|
d'Uscio LV, Milstien S, Richardson D, Smith L, Katusic ZS. Long-term vitamin C treatment increases vascular tetrahydrobiopterin levels and nitric oxide synthase activity. Circ Res 2003; 92:88-95. [PMID: 12522125 DOI: 10.1161/01.res.0000049166.33035.62] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In cultured endothelial cells, the antioxidant, L-ascorbic acid (vitamin C), increases nitric oxide synthase (NOS) enzyme activity via chemical stabilization of tetrahydrobiopterin. Our objective was to determine the effect of vitamin C on NOS function and tetrahydrobiopterin metabolism in vivo. Twenty-six to twenty-eight weeks of diet supplementation with vitamin C (1%/kg chow) significantly increased circulating levels of vitamin C in wild-type (C57BL/6J) and apolipoprotein E (apoE)--deficient mice. Measurements of NOS enzymatic activity in aortas of apoE-deficient mice indicated a significant increase in total NOS activity. However, this increase was mainly due to high activity of inducible NOS, whereas eNOS activity was reduced. Significantly higher tetrahydrobiopterin levels were detected in aortas of apoE-deficient mice. Long-term treatment with vitamin C restored endothelial NOS activity in aortas of apoE-deficient mice, but did not affect activity of inducible NOS. In addition, 7,8-dihydrobiopterin levels, an oxidized form of tetrahydrobiopterin, were decreased and vascular endothelial function of aortas was significantly improved in apoE-deficient mice. Interestingly, vitamin C also increased tetrahydrobiopterin and NOS activity in aortas of C57BL/6J mice. In contrast, long-term treatment with vitamin E (2000 U/kg chow) did not affect vascular NOS activity or metabolism of tetrahydrobiopterin. In vivo, beneficial effect of vitamin C on vascular endothelial function appears to be mediated in part by protection of tetrahydrobiopterin and restoration of eNOS enzymatic activity.
Collapse
Affiliation(s)
- Livius V d'Uscio
- Department of Anesthesiology, Mayo Clinic and Foundation, Rochester, Minn 55905, USA
| | | | | | | | | |
Collapse
|
31
|
Zhang X, Kim WS, Hatcher N, Potgieter K, Moroz LL, Gillette R, Sweedler JV. Interfering with nitric oxide measurements. 4,5-diaminofluorescein reacts with dehydroascorbic acid and ascorbic acid. J Biol Chem 2002; 277:48472-8. [PMID: 12370177 DOI: 10.1074/jbc.m209130200] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
4,5-Diaminofluorescein (DAF-2) is widely used for detection and imaging of NO based on its sensitivity, noncytotoxicity, and specificity. In the presence of oxygen, NO and NO-related reactive nitrogen species nitrosate 4,5-diaminofluorescein to yield the highly fluorescent DAF-2 triazole (DAF-2T). However, as reported here, the DAF-2 reaction to form a fluorescent product is not specific to NO because it reacts with dehydroascorbic acid (DHA) and ascorbic acid (AA) to generate new compounds that have fluorescence emission profiles similar to that of DAF-2T. When DHA is present, the formation of DAF-2T is attenuated because the DHA competes for DAF-2, whereas AA decreases the nitrosation of DAF-2 to a larger extent, possibly because of additional reducing activity that affects the amount of available N(2)O(3) from the NO. The reaction products of DAF-2 with DHA and AA have been characterized using capillary electrophoresis with laser-induced fluorescence detection and electrospray mass spectrometry. The reactions of DAF-2 with DHA and AA are particularly significant because DHA and AA often colocalize with nitric-oxide synthase in the central nervous, cardiovascular, and immune systems, indicating the importance of understanding this chemistry.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Chemistry and the Beckman Institute, University of Illinois, Urbana 61801, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Jones W, Li X, Qu ZC, Perriott L, Whitesell RR, May JM. Uptake, recycling, and antioxidant actions of alpha-lipoic acid in endothelial cells. Free Radic Biol Med 2002; 33:83-93. [PMID: 12086686 DOI: 10.1016/s0891-5849(02)00862-6] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Alpha-lipoic acid, which becomes a powerful antioxidant in its reduced form, has been suggested as a dietary supplement to treat diseases associated with excessive oxidant stress. Because the vascular endothelium is dysfunctional in many of these conditions, we studied the uptake, reduction, and antioxidant effects of alpha-lipoic acid in cultured human endothelial cells (EA.hy926). Using a new assay for dihydrolipoic acid, we found that EA.hy926 cells rapidly take up and reduce alpha-lipoic acid to dihydrolipoic acid, most of which is released into the incubation medium. Nonetheless, the cells maintain dihydrolipoic acid following overnight culture, probably by recycling it from alpha-lipoic acid. Acute reduction of alpha-lipoic acid activates the pentose phosphate cycle and consumes nicotinamide adenine dinucleotide phosphate (NADPH). Lysates of EA.hy926 cells reduce alpha-lipoic acid using both NADPH and nicotinamide adenine dinucleotide (NADH) as electron donors, although NADPH-dependent reduction is about twice that due to NADH. NADPH-dependent alpha-lipoic acid reduction is mostly due to thioredoxin reductase. Pre-incubation of cells with alpha-lipoic acid increases their capacity to reduce extracellular ferricyanide, to recycle intracellular dehydroascorbic acid to ascorbate, to decrease reactive oxygen species generated by redox cycling of menadione, and to generate nitric oxide. These results show that alpha-lipoic acid enhances both the antioxidant defenses and the function of endothelial cells.
Collapse
Affiliation(s)
- Wright Jones
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-6303, USA
| | | | | | | | | | | |
Collapse
|
33
|
Smith AR, Visioli F, Hagen TM. Vitamin C matters: increased oxidative stress in cultured human aortic endothelial cells without supplemental ascorbic acid. FASEB J 2002; 16:1102-4. [PMID: 12039848 DOI: 10.1096/fj.01-0825fje] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Because standard culture media for human aortic endothelial cells (HAEC) do not contain vitamin C, we hypothesized that HAEC may be under significant oxidative insult compared with the situation in vivo. To assess parameters of oxidative stress, intracellular vitamin C, glutathione (GSH), GSH/GSSG, and NAD(P)H/NAD(P)+ ratios, as well as oxidant appearance and oxidative damage, were measured in HAEC with or without vitamin C addition. The effect of vitamin C on eNOS activity was also determined. Results showed that HAEC without vitamin C treatment were essentially scorbutic. On addition of 100 mM vitamin C to the culture media, intracellular vitamin C levels increased and peaked at 6 h. A concomitant increase in the total GSH and the GSH/GSSG ratio was also observed; the NAD(P)H/NAD(P)+ ratio increased more slowly over the 24-h time course. Significantly lower (P <0.05) oxidant appearance and steady-state oxidative damage were also observed following vitamin C repletion. Vitamin C treatment increased eNOS activity by 600%. Thus, HAEC are scorbutic under normal culture conditions and exhibit higher oxidative stress than vitamin C repleted cells. Vitamin C supplementation should be considered when using cultured cells, especially when experimental endpoints are related to cellular redox status and eNOS activity.
Collapse
Affiliation(s)
- Anthony R Smith
- Linus Pauling Institute and Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | |
Collapse
|
34
|
Vissers MC, Lee WG, Hampton MB. Regulation of apoptosis by vitamin C. Specific protection of the apoptotic machinery against exposure to chlorinated oxidants. J Biol Chem 2001; 276:46835-40. [PMID: 11590157 DOI: 10.1074/jbc.m107664200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have investigated the ability of intracellular vitamin C to protect human umbilical vein endothelial cells from exposure to hypochlorous acid (HOCl) and a range of derived chloramines. Ascorbate provided minimal protection against the cytotoxicity induced by these oxidants, as measured by propidium iodide uptake. In contrast, there was a marked effect on apoptosis, monitored by caspase-3 activation and phosphatidylserine exposure. Extended incubation of the cells with glycine chloramine or histamine chloramine completely blocked apoptosis initiated in the cells by serum withdrawal. This effect was significantly abrogated by ascorbate. Inhibition of apoptosis required the oxidant to be present for an extended period after serum withdrawal and occurred prior to caspase-3 activation. General protection of thiols by ascorbate was not responsible for the protection of apoptosis, because intracellular oxidation by HOCl or chloramines was not prevented in supplemented cells. The results suggest a new role for vitamin C in the regulation of apoptosis. We propose that, by protection of an oxidant-sensitive step in the initiation phase, ascorbate allows apoptosis to proceed in endothelial cells under sustained oxidative stress.
Collapse
Affiliation(s)
- M C Vissers
- Pathology Department, Free Radical Research Group, Christchurch School of Medicine and Health Sciences, P. O. Box 4345, Christchurch 8001, New Zealand.
| | | | | |
Collapse
|
35
|
Guaiquil VH, Vera JC, Golde DW. Mechanism of vitamin C inhibition of cell death induced by oxidative stress in glutathione-depleted HL-60 cells. J Biol Chem 2001; 276:40955-61. [PMID: 11533037 DOI: 10.1074/jbc.m106878200] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vitamin C is a well known antioxidant whose precise role in protecting cells from oxidative challenge is uncertain. In vitro results have been confounded by pro-oxidant effects of ascorbic acid and an overlapping role of glutathione. We used HL-60 cells as a model to determine the precise and independent role of vitamin C in cellular protection against cell death induced by oxidative stress. HL-60 cells do not depend on glutathione to transport or reduce dehydroascorbic acid. Depletion of glutathione rendered the HL-60 cells highly sensitive to cell death induced by H2O2, an effect that was not mediated by changes in the activities of glutathione reductase, glutathione peroxidase, catalase, or superoxide dismutase. The increased sensitivity to oxidative stress was largely reversed when glutathione-depleted cells were preloaded with ascorbic acid by exposure to dehydroascorbic acid. Resistance to H2O2 treatment in cells loaded with vitamin C was accompanied by intracellular consumption of ascorbic acid, generation of dehydroascorbic acid, and a decrease in the cellular content of reactive oxygen species. Some of the dehydroascorbic acid generated was exported out of the cells via the glucose transporters. Our data indicate that vitamin C is an important independent antioxidant in protecting cells against death from oxidative stress.
Collapse
Affiliation(s)
- V H Guaiquil
- Program in Molecular Pharmacology and Therapeutics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | |
Collapse
|
36
|
Abstract
Ascorbic acid may be involved in the defense against oxidant stress in endothelial cells. Such a role requires that the cells effectively recycle the vitamin from its oxidized forms. In this work, we studied the ability of cultured bovine aortic endothelial cells (BAECs) to take up and reduce dehydroascorbic acid (DHA) to ascorbate, as well as the dependence of ascorbate recycling on intracellular GSH. BAECs took up and reduced DHA to ascorbate much more readily than they took up ascorbate. Although BAECs in culture did not contain ascorbate, ascorbate accumulated to concentrations of 2-3 mM in BAECs following incubation with 400 microM DHA. Extracellular ferricyanide oxidized intracellular ascorbate, which was recycled by the cells. Reduction of DHA, either when added to the cells or when generated in response to ferricyanide, caused significant decreases in intracellular GSH concentrations. Depletion of intracellular GSH with 1-chloro-2,4-dinitrobenzene, diethylmaleate, and diamide almost abolished the ability of the cells to reduce DHA to ascorbate. DHA reduction by thioredoxin reductase was evident in dialyzed cell extracts, but occurred at rates far lower than direct GSH reduction of DHA. These results suggest that maximal rates of DHA reduction, and thus recycling of ascorbate from DHA, are dependent upon GSH in these cells.
Collapse
Affiliation(s)
- J M May
- Department of Medicine, Vanderbilt University School of Medicine, 715 Medical Research Building II, Nashville, TN 37232-6303, USA.
| | | | | |
Collapse
|
37
|
Griffiths HR, Lunec J. Ascorbic acid in the 21st century - more than a simple antioxidant. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2001; 10:173-182. [PMID: 21782574 DOI: 10.1016/s1382-6689(01)00081-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Ascorbic acid (AA) is an essential micronutrient for man, with many biological roles. It is a powerful antioxidant both directly via scavenging of reactive oxygen species and indirectly through regeneration of other antioxidant systems. Paradoxically, under certain conditions (low concentration in vitro, presence of metal ions) it can exert a pro-oxidant effect, increasing oxidative damage to lipids, DNA and protein. Herein, the effects of vitamin C both in vitro and in vivo are addressed in terms of modulation of oxidative DNA damage, gene expression and protein oxidation. The view of AA as a simple scavenger is outdated, where the arrival of new bioinformatic techniques, heralds a new dawning in our understanding of ascorbate as a potential direct or indirect modulator of gene expression.
Collapse
Affiliation(s)
- H R Griffiths
- Pharmaceutical Sciences Research Institute, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | | |
Collapse
|
38
|
Kannan R, Stolz A, Ji Q, Prasad PD, Ganapathy V. Vitamin C transport in human lens epithelial cells: evidence for the presence of SVCT2. Exp Eye Res 2001; 73:159-65. [PMID: 11446766 DOI: 10.1006/exer.2001.1024] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vitamin C [ascorbic acid (AA)] is an important antioxidant present in m M amounts in the aqueous humor. Recently, two specific transporters for vitamin C (SVCT1, SVCT2) have been cloned in the rat and the human. The aim of the present study was to characterize vitamin C transport in an immortalized human lens epithelial cell line (HLE-B3). AA uptake was linear for 120 min in experiments conducted with 14C AA + 40 microM unlabelled AA. Uptake was measured at varying AA concentrations (0.04-1 m M) in Na+-containing and Na+-free buffers for 30 min at 37 degrees C. Effect of potential inhibitors of AA transport was also examined. Presence (or absence) of SVCT1 and SVCT2 was studied by RT-PCR of HLE-B3 poly (A)+ RNA using gene specific primers. Uptake studies revealed that AA uptake was highly Na+-dependent and exhibited saturation. Na+-dependent 14C-AA uptake was strongly inhibited (85-90%) by 10 m M unlabelled AA. Incubation of HLE-B3 cells with cAMP (0.1 m M), cytocholasin B (0.1 m M) and phorbol dibutyrate (1 microM) resulted in partial inhibition (36-51%) of AA uptake. Under similar conditions, D -glucose (10 m M) and staurosporine (0.1 microM) had no effect. RT-PCR showed the presence of SVCT2 while SVCT1 could not be amplified. Exposure to the chemical oxidant tert-butylhydroperoxide (TBH) up-regulated SVCT2 gene expression in HLE-B3 cells. Our data suggest that Na+-dependent transport of AA in normal lens epithelium is most likely mediated by SVCT2 rather than by SVCT1. This transport system may be subject to regulation by oxidant stress and by various second messenger signals.
Collapse
Affiliation(s)
- R Kannan
- USC Keck School of Medicine, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
39
|
Grossmann M, Dobrev D, Himmel HM, Ravens U, Kirch W. Ascorbic acid-induced modulation of venous tone in humans. Hypertension 2001; 37:949-54. [PMID: 11244023 DOI: 10.1161/01.hyp.37.3.949] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ascorbic acid appears to have vasodilatory properties, but the underlying mechanisms are not well understood. The aims of this study were to define the acute effects of locally infused ascorbic acid in human veins and to explore underlying mechanisms by using pharmacological tools in vivo. Ascorbic acid was infused in dorsal hand veins submaximally preconstricted with the alpha(1)-adrenoceptor agonist phenylephrine or with prostaglandin F(2alpha) in 23 healthy male nonsmokers, and the venodilator response was measured. Ascorbic acid produced dose-dependent dilation with maximum reversal of constriction of 38+/-4% in phenylephrine-preconstricted veins and of 51+/-13% in prostaglandin F(2alpha)-preconstricted veins. Oral pretreatment with the cyclooxygenase inhibitor acetylsalicylic acid or local coinfusion of ascorbic acid and the nitric oxide synthase inhibitor N:(G)-monomethyl-L-arginine had no effect, but coinfusion of ascorbic acid and methylene blue (to inhibit cGMP generation) abolished venodilation. Coinfusion of ascorbic acid and the nonselective potassium channel blocker quinidine abolished venodilation, whereas the inhibitor of ATP-dependent potassium channels glibenclamide had no effect. In cultured bovine endothelial cells, ascorbic acid did not affect intracellular calcium concentration but blunted the response to ATP or digitonin exposure. Ascorbic acid, in millimolar concentrations, dilates human hand veins, presumably by activation of vascular smooth muscle potassium channels through cGMP. This activation is independent of eNOS-mediated nitric oxide synthesis and cyclooxygenase products and does not involve ATP-dependent potassium channels.
Collapse
Affiliation(s)
- M Grossmann
- Institute of Clinical Pharmacology, Medical Faculty of the University of Technology Dresden (Germany).
| | | | | | | | | |
Collapse
|
40
|
Heller R, Unbehaun A, Schellenberg B, Mayer B, Werner-Felmayer G, Werner ER. L-ascorbic acid potentiates endothelial nitric oxide synthesis via a chemical stabilization of tetrahydrobiopterin. J Biol Chem 2001; 276:40-7. [PMID: 11022034 DOI: 10.1074/jbc.m004392200] [Citation(s) in RCA: 304] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ascorbic acid has been shown to stimulate endothelial nitric oxide (NO) synthesis in a time- and concentration-dependent fashion without affecting NO synthase (NOS) expression or l-arginine uptake. The present study investigates if the underlying mechanism is related to the NOS cofactor tetrahydrobiopterin. Pretreatment of human umbilical vein endothelial cells with ascorbate (1 microm to 1 mm, 24 h) led to an up to 3-fold increase of intracellular tetrahydrobiopterin levels that was concentration-dependent and saturable at 100 microm. Accordingly, the effect of ascorbic acid on Ca(2+)-dependent formation of citrulline (co-product of NO) and cGMP (product of the NO-activated soluble guanylate cyclase) was abolished when intracellular tetrahydrobiopterin levels were increased by coincubation of endothelial cells with sepiapterin (0.001-100 microm, 24 h). In contrast, ascorbic acid did not modify the pterin affinity of endothelial NOS, which was measured in assays with purified tetrahydrobiopterin-free enzyme. The ascorbate-induced increase of endothelial tetrahydrobiopterin was not due to an enhanced synthesis of the compound. Neither the mRNA expression of the rate-limiting enzyme in tetrahydrobiopterin biosynthesis, GTP cyclohydrolase I, nor the activities of either GTP cyclohydrolase I or 6-pyruvoyl-tetrahydropterin synthase, the second enzyme in the de novo synthesis pathway, were altered by ascorbate. Our data demonstrate that ascorbic acid leads to a chemical stabilization of tetrahydrobiopterin. This was evident as an increase in the half-life of tetrahydrobiopterin in aqueous solution. Furthermore, the increase of tetrahydrobiopterin levels in intact endothelial cells coincubated with cytokines and ascorbate was associated with a decrease of more oxidized biopterin derivatives (7,8-dihydrobiopterin and biopterin) in cells and cell supernatants. The present study suggests that saturated ascorbic acid levels in endothelial cells are necessary to protect tetrahydrobiopterin from oxidation and to provide optimal conditions for cellular NO synthesis.
Collapse
Affiliation(s)
- R Heller
- Center of Vascular Biology and Medicine, Friedrich-Schiller-University of Jena, D-99089 Erfurt, Germany.
| | | | | | | | | | | |
Collapse
|
41
|
Dillon PF, Root-Bernstein RS, Sears PR, Olson LK. Natural electrophoresis of norepinephrine and ascorbic acid. Biophys J 2000; 79:370-6. [PMID: 10866962 PMCID: PMC1300940 DOI: 10.1016/s0006-3495(00)76298-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The electric field produced by cell membranes, extending only a few nanometers, is 1000 times stronger than the electric fields required to produce dissociation of molecular complexes. Using the complex formed by norepinephrine (NE) and ascorbic acid (AA), we have demonstrated the quantitative binding of AA to NE, the use of capillary electrophoresis to measure quantitative binding of nonelectrolyte complexes, the determination of a dissociation constant (Kd) from electric field-dissociation constants (Ke), and a model for natural dissociation of the NE-AA complex due to the electric field generated by a cell membrane. NE-AA dissociation constants show little effect of NE concentration or pH changes. NE-related compounds also bind AA: epinephrine > norepinephrine > tyrosine > histamine > phenylalanine. Serotonin does not bind AA. Phosphorylated AA and glucose also bind NE at 0.05 and 0.08 of the AA binding, respectively. Natural electrophoresis of molecular complexes allows compounds to travel through the body in a protected state and still be available for physiological activity upon reaching a membrane.
Collapse
Affiliation(s)
- P F Dillon
- Department of Physiology, Michigan State University, East Lansing, Michigan 48823, USA.
| | | | | | | |
Collapse
|
42
|
Abstract
Human coronary and peripheral arteries show endothelial dysfunction in a variety of conditions, including atherosclerosis, hypercholesterolemia, smoking, and hypertension. This dysfunction manifests as a loss of endothelium-dependent vasodilation to acetylcholine infusion or sheer stress, and is typically associated with decreased generation of nitric oxide (NO) by the endothelium. Vitamin C, or ascorbic acid, when acutely infused or chronically ingested, improves the defective endothelium-dependent vasodilation present in these clinical conditions. The mechanism of the ascorbic acid effect is unknown, although it has been attributed to an antioxidant function of the vitamin to enhance the synthesis or prevent the breakdown of NO. In this review, multiple mechanisms are considered that might account for the ability of ascorbate to preserve NO. These include ascorbate-induced decreases in low-density lipoprotein (LDL) oxidation, scavenging of intracellular superoxide, release of NO from circulating or tissue S-nitrosothiols, direct reduction of nitrite to NO, and activation of either endothelial NO synthase or smooth muscle guanylate cyclase. The ability of ascorbic acid supplements to enhance defective endothelial function in human diseases provides a rationale for use of such supplements in these conditions. However, it is first necessary to determine which of the many plausible mechanisms account for the effect, and to ensure that undesirable toxic effects are not present.
Collapse
Affiliation(s)
- J M May
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-6303, USA.
| |
Collapse
|
43
|
Siow RC, Sato H, Leake DS, Ishii T, Bannai S, Mann GE. Induction of antioxidant stress proteins in vascular endothelial and smooth muscle cells: protective action of vitamin C against atherogenic lipoproteins. Free Radic Res 1999; 31:309-18. [PMID: 10517535 DOI: 10.1080/10715769900300871] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Elevated levels of lipid peroxidation and increased formation of reactive oxygen species within the vascular wall in atherosclerosis can overwhelm cellular antioxidant defence mechanisms. Accumulating evidence implicates oxidatively modified low density lipoproteins (LDL) in vascular dysfunction in atherosclerosis and oxidized LDL have been localized with in atherosclerotic lesions. We here report that human oxidatively modified LDL induce expression of 'antioxidant-like' stress proteins in vascular cells, involving increases in the activity of L-cystine transport, glutathione synthesis, heme oxygenase-1 and the murine stress protein MSP23. Moreover, treatment of human arterial smooth muscle cells with the dietary antioxidant vitamin C markedly attenuates adaptive increases in endogenous antioxidant gene expression and affords protection against smooth muscle cell apoptosis induced by moderately oxidized LDL. As vascular cell death is a key feature of atherosclerotic lesions and may contribute to the plaque 'necrotic' core, cap rupture and thrombosis, our findings suggest that the cytoprotective actions of vitamin C could limit plaque instability in advanced atherosclerosis.
Collapse
Affiliation(s)
- R C Siow
- Centre for Cardiovascular Biology and Medicine, GKT School of Biomedical Sciences, King's College London, UK
| | | | | | | | | | | |
Collapse
|
44
|
Heller R, Münscher-Paulig F, Gräbner R, Till U. L-Ascorbic acid potentiates nitric oxide synthesis in endothelial cells. J Biol Chem 1999; 274:8254-60. [PMID: 10075731 DOI: 10.1074/jbc.274.12.8254] [Citation(s) in RCA: 169] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ascorbic acid has been shown to enhance impaired endothelium-dependent vasodilation in patients with atherosclerosis by a mechanism that is thought to involve protection of nitric oxide (NO) from inactivation by free oxygen radicals. The present study in human endothelial cells from umbilical veins and coronary arteries investigates whether L-ascorbic acid additionally affects cellular NO synthesis. Endothelial cells were incubated for 24 h with 0.1-100 microM ascorbic acid and were subsequently stimulated for 15 min with ionomycin (2 microM) or thrombin (1 unit/ml) in the absence of extracellular ascorbate. Ascorbate pretreatment led to a 3-fold increase of the cellular production of NO measured as the formation of its co-product citrulline and as the accumulation of its effector molecule cGMP. The effect was saturated at 100 microM and followed a similar kinetics as seen for the uptake of ascorbate into the cells. The investigation of the precursor molecule L-gulonolactone and of different ascorbic acid derivatives suggests that the enediol structure of ascorbate is essential for its effect on NO synthesis. Ascorbic acid did not induce the expression of the NO synthase (NOS) protein nor enhance the uptake of the NOS substrate L-arginine into endothelial cells. The ascorbic acid effect was minimal when the citrulline formation was measured in cell lysates from ascorbate-pretreated cells in the presence of known cofactors for NOS activity. However, when the cofactor tetrahydrobiopterin was omitted from the assay, a similar potentiating effect of ascorbate pretreatment as seen in intact cells was demonstrated, suggesting that ascorbic acid may either enhance the availability of tetrahydrobiopterin or increase its affinity for the endothelial NOS. Our data suggest that intracellular ascorbic acid enhances NO synthesis in endothelial cells and that this may explain, in part, the beneficial vascular effects of ascorbic acid.
Collapse
Affiliation(s)
- R Heller
- Center of Vascular Biology and Medicine, Friedrich-Schiller-University of Jena, 99089 Erfurt, Germany.
| | | | | | | |
Collapse
|
45
|
Siow RC, Sato H, Leake DS, Pearson JD, Bannai S, Mann GE. Vitamin C protects human arterial smooth muscle cells against atherogenic lipoproteins: effects of antioxidant vitamins C and E on oxidized LDL-induced adaptive increases in cystine transport and glutathione. Arterioscler Thromb Vasc Biol 1998; 18:1662-70. [PMID: 9763541 DOI: 10.1161/01.atv.18.10.1662] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glutathione (GSH) plays a key role in cellular antioxidant defenses by scavenging reactive oxygen species and reducing lipid peroxides. Intracellular GSH levels are regulated by transport of its precursor L-cystine via system xc-, which can be induced by oxidant stress. As oxidatively modified low density lipoproteins (LDLs) contribute to impaired vascular reactivity and the formation of atherosclerotic lesions, we have examined the effects of oxidized LDL and the antioxidant vitamins C and E on the L-cystine-GSH pathway in human umbilical artery smooth muscle cells (HUASMCs). Oxidized LDL, but not native LDL, elevated intracellular GSH levels and L-cystine transport via system xc- in a time-dependent (up to 24 hours) and dose-dependent (10 to 100 microg x mL-1) manner. These increases were dependent on protein synthesis and the extent of LDL oxidation, but the induction of L-cystine transport activity was independent of GSH synthesis. Pretreatment of HUASMCs for 24 hours with vitamin E (100 micromol/L) attenuated oxidized LDL-mediated increases in GSH, whereas pretreatment with vitamin C depressed basal levels and abolished oxidized LDL-induced increases in GSH and L-cystine transport in a time-dependent (3 to 24 hours) and dose-dependent (10 to 100 micromol/L) manner. Pretreatment of cells with dehydroascorbate had no effect on oxidized LDL-mediated increases in L-cystine transport and only marginally attenuated increases in GSH. Our findings provide the first evidence that vitamin C spares endogenous adaptive antioxidant responses in human vascular smooth muscle cells exposed to atherogenic oxidized LDL.
Collapse
Affiliation(s)
- R C Siow
- Vascular Biology Research Centre, Biomedical Sciences Division, King's College London, UK; the Biochemistry Department, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Voskoboinik I, Söderholm K, Cotgreave IA. Ascorbate and glutathione homeostasis in vascular smooth muscle cells: cooperation with endothelial cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:C1031-9. [PMID: 9755056 DOI: 10.1152/ajpcell.1998.275.4.c1031] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human umbilical vein smooth muscle cells (HUVSMCs) utilize extracellular cystine, glutathione (GSH), and N-acetylcysteine (NAC) to synthesize cellular GSH. Extracellular cystine was effective from 5 microM, whereas GSH and NAC were required at 100 microM for comparable effects. The efficacy of extracellular GSH was dependent on de novo GSH synthesis, indicating a dependence on cellular gamma-glutamyltransferase (glutamyl transpeptidase). Coculture of syngenetic HUVSMCs and corresponding human umbilical vein endothelial cells (HUVECs) on porous supports restricted cystine- or GSH-stimulated synthesis of HUVSMC GSH when supplied on the "luminal" endothelial side. Thus HUVSMC GSH rapidly attained a steady-state level below that achieved in the absence of interposed HUVECs. HUVSMCs also readily utilize both reduced ascorbate (AA) and oxidized dehydroascorbate (DHAA) over the range 50-500 microM. Phloretin effectively blocked both AA- and DHAA-stimulated assimilation of intracellular AA, indicating a role for a glucose transporter in their transport. Uptake of extracellular AA was also sensitive to extracellular, but not intracellular, thiol depletion. When AA was applied to the endothelial side of the coculture model, assimilation of intracellular AA in HUVSMCs was restricted to a steady-state level below that achieved by free access.
Collapse
Affiliation(s)
- I Voskoboinik
- Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | | | | |
Collapse
|
47
|
Merker MP, Olson LE, Bongard RD, Patel MK, Linehan JH, Dawson CA. Ascorbate-mediated transplasma membrane electron transport in pulmonary arterial endothelial cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:L685-93. [PMID: 9612283 DOI: 10.1152/ajplung.1998.274.5.l685] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary endothelial cells are capable of reducing certain electron acceptors at the luminal plasma membrane surface. Motivation for studying this phenomenon comes in part from the expectation that it may be important both as an endothelial antioxidant defense mechanism and in redox cycling of toxic free radicals. Pulmonary arterial endothelial cells in culture reduce the oxidized forms of thiazine compounds that have been used as electron acceptor probes for studying the mechanisms of transplasma membrane electron transport. However, they reduce another commonly studied electron acceptor, ferricyanide, only very slowly by comparison. In the present study, we examined the influence of ascorbate [ascorbic acid (AA)] and dehydroascorbate [dehydroascorbic acid (DHAA)] on the ferricyanide and thiazine reductase activities of the bovine pulmonary arterial endothelial cell surface. The endothelial cells were grown on microcarrier beads so that the reduction of ferricyanide and methylene blue could be studied colorimetrically in spectrophotometer cuvettes and in flow-through cell columns. The ferricyanide reductase activity could be increased 80-fold by adding DHAA to the medium, with virtually no effect on methylene blue reduction. The DHAA effect persisted after the DHAA was removed from the medium. AA also stimulated the ferricyanide reductase activity but was less potent, and the relative potencies of AA and DHAA correlated with their relative rates of uptake by the cells. The results are consistent with the hypothesis that AA is an intracellular electron donor for an endothelial plasma membrane ferricyanide reductase and that the stimulatory effect of DHAA is the result of increasing intracellular AA. Adding sufficient DHAA to markedly increase extracellular ferricyanide reduction had little effect on the plasma membrane methylene blue reductase activity, suggesting that pulmonary arterial endothelial cells have at least two separate transplasma membrane electron transport systems.
Collapse
Affiliation(s)
- M P Merker
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | | | | | | | |
Collapse
|
48
|
Saitoh Y, Nagao N, O'Uchida R, Yamane T, Kageyama K, Muto N, Miwa N. Moderately controlled transport of ascorbate into aortic endothelial cells against slowdown of the cell cycle, decreasing of the concentration or increasing of coexistent glucose as compared with dehydroascorbate. Mol Cell Biochem 1997; 173:43-50. [PMID: 9278253 DOI: 10.1023/a:1006879001316] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Uptake of L-[1-(14)C]ascorbic acid (Asc) of 12.5-200 microM for 1 h into bovine aortic endothelial BAE-2 cells grown to confluence was as low as 43-64% (per cell) of uptake into the cells grown to nearly one-fourth confluence. [14C]Asc undergoing transmembrane uptake was concentrated and accumulated in the cell less efficiently ([Asc]in/ex = 8-13) at confluence than at subconfluence ([Asc]in/ex = 15-24). The declined Asc uptake at confluence is attributable to slowdown of the cell cycle, because a similar decrease in [Asc]in/ex was shown by subconfluent cells precultured in serum-insufficient medium, resulting in an increase in G1 phase and concurrent decreases in S and G2 + M phase distributions as determined by flow cytometry. [1-(14)C]Dehydroascorbic acid (DehAsc) was taken up and accumulated as Asc, after metabolic reduction, without detectable DehAsc. The [Asc]in/ex values for DehAsc at confluence were as low as 15-69% of those at subconfluence in contrast to the values as retentive as 62-75% for Asc, suggesting the moderate control of Asc uptake against slowdown of the cell cycle. At either confluence or subconfluence, dose-dependence for DehAsc uptake was more marked than for Asc uptake as shown by an uphill slope in a curve of doses versus [Asc]in/ex for DehAsc in contrast to a downhill slope for Asc, suggesting the moderate control for Asc uptake against fluctuation of the dose. Increasing of coexistent glucose of 5 mM to 20-40 mM, plasma concentrations in diabetic patients, declined DehAsc uptake to 46-48%, which was less moderately controlled than Asc uptake retained to 59-73%. Asc uptake did not compete with DehAsc uptake, suggesting different transporter proteins for Asc and DehAsc. Thus, Asc uptake into the aortic endothelial cells is more moderately controlled against slowdown of the cell cycle, decreasing of the extracellular concentrations or increasing of coexistent glucose than DehAsc uptake, suggesting a homeostatic advantage of Asc over DehAsc in terms of retention of intracellular Asc contents within a definite range.
Collapse
Affiliation(s)
- Y Saitoh
- Department of Cell Biochemistry, Hiroshima Prefectural University School of BioScience, Shobara, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
Martin A, Frei B. Both intracellular and extracellular vitamin C inhibit atherogenic modification of LDL by human vascular endothelial cells. Arterioscler Thromb Vasc Biol 1997; 17:1583-90. [PMID: 9301639 DOI: 10.1161/01.atv.17.8.1583] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oxidative modification of LDL by vascular cells has been proposed as a mechanism by which LDL becomes atherogenic. Antioxidants that can prevent LDL oxidation may therefore act as antiatherogens. We used endothelial cells (ECs) from human aortas (HAECs), human saphenous veins (HSECs), and bovine aortas (BAECs) to investigate the role of intracellular and extracellular vitamin C (ascorbate) in EC-mediated LDL modification. Incubation of LDL (0.1 mg protein per milliliter) with confluent HAECs in Ham's F-10 medium led to time-dependent modification of the lipoprotein. In contrast, incubation of LDL with HAECs in medium 199, which does not contain redox-active transition metal ions, did not lead to LDL modification. Both HAEC-mediated and cell-free LDL modifications in Ham's F-10 medium were strongly inhibited in a time- and dose-dependent manner by physiological concentrations of ascorbate. Confluent HAECs cultured under conventional conditions contained very little intracellular ascorbate (< 0.5 nmol/mg protein) but could be loaded with up to 20 nmol ascorbate per milligram protein in a time- and concentration-dependent manner. Ascorbate-loaded HAECs exhibited a lower capacity to modify LDL than did non-ascorbate-loaded control cells. When LDL was incubated with HSECs instead of HAECs, similar time- and concentration-dependent inhibitory effects on LDL modification of intracellular and extracellular ascorbate were observed. In contrast to human ECs, BAECs did not take up vitamin C and therefore only coincubation but not preincubation with ascorbate inhibited BAEC-mediated LDL modification. Our data show that enrichment of human vascular ECs with vitamin C lowers their capacity to modify LDL. In addition, extracellular vitamin C strongly inhibits EC-mediated, metal ion-dependent atherogenic modification of LDL.
Collapse
Affiliation(s)
- A Martin
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Mass, USA
| | | |
Collapse
|
50
|
Moison RM, de Beaufort AJ, Haasnoot AA, Dubbelman TM, van Zoeren-Grobben D, Berger HM. Uric acid and ascorbic acid redox ratios in plasma and tracheal aspirate of preterm babies with acute and chronic lung disease. Free Radic Biol Med 1997; 23:226-34. [PMID: 9199884 DOI: 10.1016/s0891-5849(97)00033-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study compared plasma redox ratios of uric acid and ascorbic acid in well preterm babies with those with respiratory distress syndrome (RDS) and chronic lung disease (CLD), and investigated the relationship between these ratios and their respective measurements in tracheal aspirate. On day 1 after birth, plasma allantoin and allantoin/uric acid ratio were elevated in CLD (p < .05), and both markers of oxidative stress enabled early prediction of development of CLD (sensitivity and specificity: 54 and 83%, respectively). The relation between allantoin production and oxidative stress is supported by the correlation between the allantoin level and oxygen therapy in both RDS and CLD (p < .05). Reduced and oxidize ascorbic acid in plasma decreased postnatally in all groups and their redox ratio remained stable. Uric acid and ascorbic acid redox ratios were significantly elevated in tracheal aspirates compared to plasma samples (p < .05), and there was a strong positive correlation between both ratios (p < .005). These markers may be useful in monitoring babies with respiratory distress.
Collapse
Affiliation(s)
- R M Moison
- Department of Pediatrics, Leiden University Hospital, The Netherlands
| | | | | | | | | | | |
Collapse
|