1
|
Chong PL, Laight D, Aspinall RJ, Higginson A, Cummings MH. A randomised placebo controlled trial of VSL#3 ® probiotic on biomarkers of cardiovascular risk and liver injury in non-alcoholic fatty liver disease. BMC Gastroenterol 2021; 21:144. [PMID: 33794784 PMCID: PMC8015038 DOI: 10.1186/s12876-021-01660-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/09/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is associated with increased cardiovascular risk irrespective of conventional risk factors. The role of gut-liver interaction is implicated in its development. We investigated the effects of VSL#3® probiotic supplementation on biomarkers of cardiovascular risk and liver injury in patients with NAFLD. METHODS A randomised, double-blinded, placebo-controlled, proof-of-concept study was undertaken. Patients with NAFLD were randomly allocated to take 2 sachets VSL#3® probiotic or placebo twice daily for 10 weeks. Measurements of endothelial function (digital photoplethysmography, sVCAM-1 and cGMP), oxidative stress (glutathione ratio and LHP), inflammation (hsCRP), insulin resistance (HOMA-IR) and liver injury [transaminases, fibrosis risk score and acoustic structure quantification (ASQ)] were undertaken before and after intervention. Difference in baseline characteristics between the treatment groups was analysed using independent t-test or Mann Whitney U test for non-parametric data. Independent t-test was used to compare the outcomes at the end of the study between the two treatment groups. Wilcoxon Signed Rank test was used to determine the difference in fibrosis risk scores before and after treatment. Spearman's correlation was used to determine any association between cardiovascular and hepatic markers at baseline. RESULTS Thirty-five patients completed the study (28 males and 7 females) with a mean age of 57 ± 8 years, body mass index of 32.6 ± 5.0 kg/m2 and a relatively short duration of NAFLD (median duration 0.3 IQR 2.0 years). No significant difference was observed in biomarkers of cardiovascular risk and liver injury following VSL#3® supplementation. Significant correlations were noted between sVCAM-1 and hsCRP (rho = 0.392, p = 0.01), and HOMA-IR and AST (rho = 0.489, p < 0.01) at baseline. CONCLUSIONS This is the first study to evaluate the effect of VSL#3® on ASQ in patients with NAFLD. VSL#3® did not significantly improve markers of cardiovascular risk and liver injury in patients with NAFLD. However, the study supports an association between endothelial dysfunction and inflammation in patients with NAFLD and suggests that NAFLD is linked with insulin resistance. TRIAL REGISTRATION ISRCTN05474560 ( https://doi.org/10.1186/ISRCTN05474560 ) Registered 9 August 2012 (retrospectively registered).
Collapse
Affiliation(s)
- Pui Lin Chong
- Academic Department of Diabetes and Endocrinology, Queen Alexandra Hospital, Portsmouth, UK. .,Raja Isteri Pengiran Anak Saleha Hospital, Bandar Seri Begawan, 1710, Brunei Darussalam.
| | - David Laight
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Richard J Aspinall
- Department of Gastroenterology and Hepatology, Queen Alexandra Hospital, Portsmouth, UK
| | | | - Michael H Cummings
- Academic Department of Diabetes and Endocrinology, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
2
|
The cystine/glutamate antiporter system xc- drives breast tumor cell glutamate release and cancer-induced bone pain. Pain 2017; 157:2605-2616. [PMID: 27482630 PMCID: PMC5065056 DOI: 10.1097/j.pain.0000000000000681] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is Available in the Text. Tumor-derived glutamate may significantly contribute to cancer-induced bone pain. The glutamate transporter system xc− is a promising therapeutic target in this pain state. Bone is one of the leading sites of metastasis for frequently diagnosed malignancies, including those arising in the breast, prostate and lung. Although these cancers develop unnoticed and are painless in their primary sites, bone metastases result in debilitating pain. Deeper investigation of this pain may reveal etiology and lead to early cancer detection. Cancer-induced bone pain (CIBP) is inadequately managed with current standard-of-care analgesics and dramatically diminishes patient quality of life. While CIBP etiology is multifaceted, elevated levels of glutamate, an excitatory neurotransmitter, in the bone-tumor microenvironment may drive maladaptive nociceptive signaling. Here, we establish a relationship between the reactive nitrogen species peroxynitrite, tumor-derived glutamate, and CIBP. In vitro and in a syngeneic in vivo model of breast CIBP, murine mammary adenocarcinoma cells significantly elevated glutamate via the cystine/glutamate antiporter system xc−. The well-known system xc− inhibitor sulfasalazine significantly reduced levels of glutamate and attenuated CIBP-associated flinching and guarding behaviors. Peroxynitrite, a highly reactive species produced in tumors, significantly increased system xc− functional expression and tumor cell glutamate release. Scavenging peroxynitrite with the iron and mangano-based porphyrins, FeTMPyP and SRI10, significantly diminished tumor cell system xc− functional expression, reduced femur glutamate levels and mitigated CIBP. In sum, we demonstrate how breast cancer bone metastases upregulate a cystine/glutamate co-transporter to elevate extracellular glutamate. Pharmacological manipulation of peroxynitrite or system xc− attenuates CIBP, supporting a role for tumor-derived glutamate in CIBP and validating the targeting of system xc− as a novel therapeutic strategy for the management of metastatic bone pain.
Collapse
|
3
|
Ivanov VV, Shakhristova EV, Stepovaya EA, Nosareva OL, Fedorova TS, Ryazantseva NV, Novitsky VV. Effect of insulin, the glutathione system, and superoxide anion radical in modulation of lipolysis in adipocytes of rats with experimental diabetes. BIOCHEMISTRY (MOSCOW) 2015; 80:87-96. [PMID: 25754043 DOI: 10.1134/s0006297915010101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Spontaneous lipolysis was found to be increased in adipocytes of rats with alloxan-induced diabetes. In addition, isoproterenol-stimulated hydrolysis of triacylglycerols was inhibited against the background of oxidative stress and decreased redox-status of cells. A decrease in the ability of insulin to inhibit isoproterenol-stimulated lipolysis in adipocytes that were isolated from adipose tissue of rats with experimental diabetes was found, which shows a disorder in regulation of lipolysis in adipocytes by the hormone in alloxan-induced diabetes. Based on these findings, we concluded that there is an influence of reactive oxygen species, superoxide anion radical in particular, and redox potential of the glutathione system on molecular mechanisms of change in lipolysis intensity in rat adipocytes in alloxan-induced oxidative stress. Activation of spontaneous lipolysis under conditions of oxidative stress might be a reason for the high concentration of free fatty acids in blood plasma in experimental diabetes, and this may play a significant role in development of insulin resistance and appearance of complications of diabetes.
Collapse
Affiliation(s)
- V V Ivanov
- Siberian State Medical University, Tomsk, 634050, Russia.
| | | | | | | | | | | | | |
Collapse
|
4
|
Chapple SJ, Cheng X, Mann GE. Effects of 4-hydroxynonenal on vascular endothelial and smooth muscle cell redox signaling and function in health and disease. Redox Biol 2013; 1:319-31. [PMID: 24024167 PMCID: PMC3757694 DOI: 10.1016/j.redox.2013.04.001] [Citation(s) in RCA: 329] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 04/21/2013] [Indexed: 12/04/2022] Open
Abstract
4-hydroxynonenal (HNE) is a lipid hydroperoxide end product formed from the oxidation of n-6 polyunsaturated fatty acids. The relative abundance of HNE within the vasculature is dependent not only on the rate of lipid peroxidation and HNE synthesis but also on the removal of HNE adducts by phase II metabolic pathways such as glutathione-S-transferases. Depending on its relative concentration, HNE can induce a range of hormetic effects in vascular endothelial and smooth muscle cells, including kinase activation, proliferation, induction of phase II enzymes and in high doses inactivation of enzymatic processes and apoptosis. HNE also plays an important role in the pathogenesis of vascular diseases such as atherosclerosis, diabetes, neurodegenerative disorders and in utero diseases such as pre-eclampsia. This review examines the known production, metabolism and consequences of HNE synthesis within vascular endothelial and smooth muscle cells, highlighting alterations in mitochondrial and endoplasmic reticulum function and their association with various vascular pathologies. HNE is a lipid peroxidation endproduct regulating vascular redox signaling. HNE detoxification is tightly regulated in vascular and other cell types. Elevated HNE levels are associated with various vascular diseases.
Collapse
Key Words
- 15d-PGJ2, 15-deoxy-Delta (12,14) prostaglandin-J2
- 4-hydroxynonenal
- AP-1, Activator protein-1
- AR, Aldose reductase
- ARE, Antioxidant response element
- ATF6, Activating transcription factor 6
- Akt, Protein kinase B
- BAEC, Bovine aortic endothelial cells
- BH4, Tetrahydrobiopterin
- BLMVEC, Bovine lung microvascular vein endothelial cells
- BPAEC, Bovine pulmonary arterial endothelial cells
- BTB, Broad complex Tramtrack and Bric–brac domain
- CHOP, C/EBP-homologous protein
- CREB, cAMP response element-binding protein
- EGFR, Epidermal growth factor receptor
- ER, Endoplasmic reticulum
- ERAD, Endoplasmic reticulum assisted degradation
- ERK1/2, Extracellular signal-regulated kinase 1/2
- Elk1, ETS domain-containing protein
- Endothelial cells
- EpRE, Electrophile response element
- FAK, Focal adhesion kinase
- FAP, Familial amyloidotic polyneuropathy
- GCLC, Glutamate cysteine ligase catalytic subunit
- GCLM, Glutamate cysteine ligase modifier subunit
- GS-DHN, Glutathionyl-1,4 dihydroxynonene
- GS-HNE, HNE-conjugates
- GSH, Glutathione
- GST, Glutathione-S-transferase
- GTPCH, Guanosine triphosphate cyclohydrolase I
- HASMC, Human aortic smooth muscle cells
- HCSMC, Human coronary smooth muscle cells
- HERP, Homocysteine inducible ER protein
- HMEC, Human microvascular endothelial cells
- HNE, 4-hydroxynonenal
- HO-1, Heme oxygenase-1
- HUVEC, Human umbilical vein endothelial cells
- Hsp-70/72/90, Heat shock proteins-70/ -72/ -90
- IRE1, Inositol requiring enzyme 1 IRE1
- IVR, Central intervening region
- JNK, c-jun N-terminal kinase
- Keap1, Kelch-like ECH-associated protein 1
- MASMC, Mouse aortic smooth muscle cells
- MEK1/2, Mitogen activated protein kinase kinase 1/2
- MMP-1/2, Matrix metalloproteinase-1/ -2
- MPEC, Mouse pancreatic islet endothelial cells
- NAC, N-acetylcysteine
- NFκB, Nuclear factor kappa B
- NO, Nitric oxide
- NQO1, NAD(P)H quinone oxidoreductase
- Nrf2
- Nrf2, Nuclear factor-E2-related factor 2
- PCEC, Porcine cerebral endothelial cells
- PDGF, Platelet-derived growth factor
- PDI, Protein disulfide isomerases
- PERK, Protein kinase-like endoplasmic reticulum kinase
- PKC, Protein kinase C
- PUFAs, Polyunsaturated fatty acids
- RASMC, Rat aortic smooth muscle cells
- ROS, Reactive oxygen species
- RVSMC, Rat vascular smooth muscle cells
- Redox signaling
- SMC, Smooth muscle cell
- TKR, Tyrosine kinase receptor
- UPR, Unfolded protein response
- Vascular biology
- Vascular smooth muscle cells
- eNOS, Endothelial nitric oxide synthase
- elF2α, Eukaryotic translation initiation factor 2α
- iNOS, Inducible nitric oxide synthase
- oxLDL, Oxidized low density lipoprotein
- tBHP, Tert-butylhydroperoxide
- xCT, cystine/glutamate amino acid transporter
Collapse
Affiliation(s)
- Sarah J Chapple
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, 150 Stamford Street, London SE1 9NH, U.K
| | | | | |
Collapse
|
5
|
Kirkwood JS, Legette LL, Miranda CL, Jiang Y, Stevens JF. A metabolomics-driven elucidation of the anti-obesity mechanisms of xanthohumol. J Biol Chem 2013; 288:19000-13. [PMID: 23673658 DOI: 10.1074/jbc.m112.445452] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mild, mitochondrial uncoupling increases energy expenditure and can reduce the generation of reactive oxygen species (ROS). Activation of cellular, adaptive stress response pathways can result in an enhanced capacity to reduce oxidative damage. Together, these strategies target energy imbalance and oxidative stress, both underlying factors of obesity and related conditions such as type 2 diabetes. Here we describe a metabolomics-driven effort to uncover the anti-obesity mechanism(s) of xanthohumol (XN), a prenylated flavonoid from hops. Metabolomics analysis of fasting plasma from obese, Zucker rats treated with XN revealed decreases in products of dysfunctional fatty acid oxidation and ROS, prompting us to explore the effects of XN on muscle cell bioenergetics. At low micromolar concentrations, XN acutely increased uncoupled respiration in several different cell types, including myocytes. Tetrahydroxanthohumol also increased respiration, suggesting electrophilicity did not play a role. At higher concentrations, XN inhibited respiration in a ROS-dependent manner. In myocytes, time course metabolomics revealed acute activation of glutathione recycling and long term induction of glutathione synthesis as well as several other changes indicative of short term elevated cellular stress and a concerted adaptive response. Based on these findings, we hypothesize that XN may ameliorate metabolic syndrome, at least in part, through mitochondrial uncoupling and stress response induction. In addition, time course metabolomics appears to be an effective strategy for uncovering metabolic events that occur during a stress response.
Collapse
Affiliation(s)
- Jay S Kirkwood
- Linus Pauling Institute and the Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | | | | | |
Collapse
|
6
|
Kawano Y, Ohta M, Eguchi H, Iwashita Y, Inomata M, Kitano S. Increased oxidative stress may lead to impaired adaptive cytoprotection in the gastric mucosa of portal hypertensive rat. J Gastroenterol Hepatol 2013; 28:639-44. [PMID: 23278350 DOI: 10.1111/jgh.12101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS In the gastric mucosa of portal hypertensive rats, adaptive cytoprotection against ethanol-induced damage is impaired. The aim of this study was to determine relation between impaired adaptive cytoprotection and oxidative stress. METHODS Portal hypertension was produced in male Sprague-Dawley rats by inducing staged portal vein occlusion. Oxidative stress levels were evaluated by measuring malondialdehyde and nitrotyrosine levels in the rat gastric mucosa with or without 10% ethanol pretreatment. Inhibition of oxidative stress by an anti-oxidant agent was estimated, and glutathione levels were also measured. Adaptive cytoprotection to 70% ethanol treatment was evaluated by measuring the gastric mucosal injury index in the presence or absence of the anti-oxidant. RESULTS The portal hypertensive gastric mucosa pretreated with 10% ethanol had significantly higher oxidative stress levels than the mucosa not pretreated with 10% ethanol. However, the sham-operated gastric mucosa pretreated with 10% ethanol had significantly lower oxidative stress levels than the mucosa not pretreated with 10% ethanol. Pretreatment with 10% ethanol increased glutathione levels in the sham-operated but not in the portal hypertensive gastric mucosa. Administration of the anti-oxidant agent prior to 10% ethanol pretreatment significantly reduced oxidative stress levels, increased glutathione levels, and decreased the injury index in response to 70% ethanol in the portal hypertensive gastric mucosa. CONCLUSION Increased oxidative stress may lead to impaired adaptive cytoprotection in the gastric mucosa of portal hypertensive rats, probably through damage to the system of endogenous anti-oxidant production.
Collapse
Affiliation(s)
- Yuichiro Kawano
- Department of Surgery I, Oita University Faculty of Medicine, Yufu, Oita, Japan.
| | | | | | | | | | | |
Collapse
|
7
|
Role of Nrf2-mediated heme oxygenase-1 upregulation in adaptive survival response to nitrosative stress. Arch Pharm Res 2009; 32:1163-76. [PMID: 19727608 DOI: 10.1007/s12272-009-1807-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 03/16/2009] [Accepted: 06/25/2009] [Indexed: 12/21/2022]
Abstract
Nitrosative stress caused by reactive nitrogen species such as nitric oxide and peroxynitrite overproduced during inflammation leads to cell death and has been implicated in the pathogenesis of many human ailments. However, relatively mild nitrosative stress may fortify cellular defense capacities, rendering cells tolerant or adaptive to ongoing and subsequent cytotoxic challenges, a phenomenon known as 'preconditioning' or 'hormesis'. One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. HO-1 is upregulated by a wide array of stimuli and has antioxidant, anti-inflammatory and other cytoprotective functions. This review is intended to provide readers with a welldocumented account of the research done in the area of cellular adaptive survival response against nitrosative stress with special focus on the role of HO-1 upregulation, especially through activation of the transcription factor, Nrf2.
Collapse
|
8
|
Zhu J, Li S, Marshall ZM, Whorton AR. A cystine-cysteine shuttle mediated by xCT facilitates cellular responses toS-nitrosoalbumin. Am J Physiol Cell Physiol 2008; 294:C1012-20. [DOI: 10.1152/ajpcell.00411.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have shown previously that extracellular cysteine is necessary for cellular responses to S-nitrosoalbumin. In this study we have investigated mechanisms involved in accumulation of extracellular cysteine outside vascular smooth muscle cells and characterized the role of cystine-cysteine release in transfer of nitric oxide (NO)-bioactivity. Incubation of cells with cystine led to cystine uptake, reduction, and cysteine release. The process was inhibitable by extracellular glutamate, suggesting a role for system xc−amino acid transporters. Smooth muscle cells express this transporter constitutively and induction of the light chain component (xCT) by either diethyl maleate or 3-morpholino-sydnonimine (SIN-1) led to glutamate-inhibitable cystine uptake and an increased rate of cysteine release from cells. Likewise, overexpression of xCT in smooth muscle cells or endothelial cells led to glutamate-inhibitable cysteine release. The resulting extracellular cysteine was found to be required for transfer of NO from extracellular S-nitrosothiols into cells via system L transporters leading to formation of cellular S-nitrosothiols. Cysteine release coupled to cystine uptake was also found to be required for cellular responses to S-nitrosoalbumin and facilitated S-nitrosoalbumin-mediated inhibition of epidermal growth factor signaling. These data show that xCT expression can constitute a cystine-cysteine shuttle whereby cystine uptake drives cysteine release. Furthermore, we show that extracellular cysteine provided by this shuttle mechanism is necessary for transfer of NO equivalents and cellular responses to S-nitrosoablumin.
Collapse
|
9
|
Buckley BJ, Li S, Whorton AR. Keap1 modification and nuclear accumulation in response to S-nitrosocysteine. Free Radic Biol Med 2008; 44:692-8. [PMID: 18062931 PMCID: PMC2267934 DOI: 10.1016/j.freeradbiomed.2007.10.055] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 10/25/2007] [Accepted: 10/31/2007] [Indexed: 12/30/2022]
Abstract
Keap1 is a key regulator of the Nrf2 transcription factor, which transactivates the antioxidant response element (ARE) and upregulates numerous proteins involved in antioxidant defense. Under basal conditions, Keap1 targets Nrf2 for ubiquitination and proteolytic degradation and as such is responsible for the rapid turnover of Nrf2. In response to oxidants and electrophiles, Nrf2 is stabilized and accumulates in the nucleus. The mechanism for this effect has been proposed to involve thiol-dependent modulation of Keap1 leading to loss of its ability to negatively regulate Nrf2. We have previously shown that nitric oxide and S-nitrosothiols cause nuclear accumulation of Nrf2 and upregulation of the ARE-regulated gene HO-1. Here we show that nitric oxide and S-nitrosocysteine (CSNO) cause time- and dose-dependent Keap1 thiol modification. These studies were carried out in HEK293 cells and in HEK293 cells overexpressing hemagglutinin-tagged Keap1. Furthermore we demonstrate that in response to CSNO Keap1 accumulates in the nucleus with a time course similar to that of Nrf2.
Collapse
Affiliation(s)
- Barbara J Buckley
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
10
|
Muyderman H, Wadey AL, Nilsson M, Sims NR. Mitochondrial glutathione protects against cell death induced by oxidative and nitrative stress in astrocytes. J Neurochem 2007; 102:1369-82. [PMID: 17484727 DOI: 10.1111/j.1471-4159.2007.04641.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The major cellular antioxidant, glutathione, is mostly localized in the cytosol but a small portion is found in mitochondria. We have recently shown that highly selective depletion of mitochondrial glutathione in astrocytes in culture markedly increased cell death induced by the peroxynitrite donor, 3-morpholino-syndnonimine. The present study was aimed at characterizing the increase in susceptibility arising from mitochondrial glutathione loss and testing the possibility that elevating this metabolite pool above normal values could be protective. The increased vulnerability of astrocytes with depleted mitochondrial glutathione to Sin-1 was confirmed. Furthermore, these cells showed marked increases in sensitivity to hydrogen peroxide and also to high concentrations of the nitric oxide donor, S-nitroso-N-acetyl-penicillamine. The increase in cell death was mostly due to necrosis as indicated by substantially increased release of lactate dehydrogenase and staining of nuclei with propidium iodide but little change in annexin V staining and caspase 3 activation. The enhanced cell loss was blocked by prior restoration of the mitochondrial glutathione content. It was also essentially fully inhibited by treatment with cyclosporin A, consistent with a role for the mitochondrial permeability transition in the development of cell death. Susceptibility to the classical apoptosis inducer, staurosporine, was only affected to a small extent in contrast to the response to the other substances tested. Incubation of normal astrocytes with glutathione monoethylester produced large and long-lasting increases in mitochondrial glutathione content with much smaller effects on the cytosolic glutathione pool. This treatment reduced cell death on exposure to 3-morpholino-syndnonimine or hydrogen peroxide but not S-nitroso-N-acetyl-pencillamine or staurosporine. These findings provide evidence for an important role for mitochondrial glutathione in preserving cell viability during periods of oxidative or nitrative stress and indicate that increases in this glutathione pool can confer protection against some of these stressors.
Collapse
Affiliation(s)
- Håkan Muyderman
- Centre for Neuroscience and Department of Medical Biochemistry, School of Medicine, Flinders University, Adelaide, Australia.
| | | | | | | |
Collapse
|
11
|
Kaur M, Sandhir R. Comparative effects of acute and chronic carbofuran exposure on oxidative stress and drug-metabolizing enzymes in liver. Drug Chem Toxicol 2006; 29:415-21. [PMID: 16931442 DOI: 10.1080/01480540600837969] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The current study has been designed to examine the comparative effects of acute and chronic carbofuran exposure on lipid peroxidation, glutathione levels, and drug-metabolizing enzymes in rat liver. Activity of acetylcholinesterase, a bona fide marker of carbofuran exposure, was markedly inhibited after acute carbofuran exposure, whereas the extent of inhibition was much less after chronic exposure. Lipid peroxidation was accentuated after chronic carbofuran exposure. However, acute exposure resulted in relatively less increase in lipid peroxidation levels than with chronic exposure. Glutathione levels were significantly increased in liver of animals chronically exposed to carbofuran; on the contrary, there was a drastic reduction in glutathione levels after acute exposure. Cytochrome P450 was significantly induced in liver of animals treated with acute as well as chronic carbofuran. The activity of glutathione-S-transferase was induced after both acute and chronic carbofuran exposure; the increase was much higher in chronically exposed animals as compared with animals exposed acutely. Based on the results, it is clear that acute and chronic carbofuran exposure have differential effects on oxidative stress and drug-metabolizing enzymes in liver.
Collapse
Affiliation(s)
- Manjit Kaur
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | |
Collapse
|
12
|
Gras G, Porcheray F, Samah B, Leone C. The glutamate-glutamine cycle as an inducible, protective face of macrophage activation. J Leukoc Biol 2006; 80:1067-75. [PMID: 16912070 DOI: 10.1189/jlb.0306153] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Neuronal damage in HIV infection results mainly from chronic activation of brain tissue and involves inflammation, oxidative stress, and glutamate-related neurotoxicity. Glutamate toxicity acts via two distinct pathways: an excitotoxic one, in which glutamate receptors are hyperactivated, and an oxidative one, in which cystine uptake is inhibited, resulting in glutathione depletion, oxidative stress, and cell degeneration. A number of studies have shown that astrocytes normally take up glutamate, keeping extracellular glutamate concentration low in the brain and preventing excitotoxicity. They, in turn, provide the trophic amino acid glutamine via their expression of glutamine synthetase. These protective and trophic actions are inhibited in HIV infection, probably as a result of the effects of inflammatory mediators and viral proteins. In vitro and in vivo studies have demonstrated that activated microglia and brain macrophages (AMM) express the transporters and enzymes of the glutamate cycle. This suggests that in addition to their recognized neurotoxic properties in HIV infection, these cells exhibit some neuroprotective properties, which may partly compensate for the inhibited astrocytic function. This hypothesis might explain the discrepancy between microglial activation, which occurs early in the disease, and neuronal apoptosis and neuronal loss, which are late events. In this review, we discuss the possible neuroprotective and neurotrophic roles of AMM and their relationships with inflammation and oxidative stress.
Collapse
Affiliation(s)
- Gabriel Gras
- CEA, DSV, DRM, SNV, UMR E-01 Université Paris-Sud XI, Laboratoire de Neuro-Immuno-Virologie, F92265 Fontenay-aux Roses, France.
| | | | | | | |
Collapse
|
13
|
Gras G, Chrétien F, Vallat-Decouvelaere AV, Le Pavec G, Porcheray F, Bossuet C, Léone C, Mialocq P, Dereuddre-Bosquet N, Clayette P, Le Grand R, Créminon C, Dormont D, Rimaniol AC, Gray F. Regulated expression of sodium-dependent glutamate transporters and synthetase: a neuroprotective role for activated microglia and macrophages in HIV infection? Brain Pathol 2006; 13:211-22. [PMID: 12744474 PMCID: PMC8095782 DOI: 10.1111/j.1750-3639.2003.tb00020.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
It is now widely accepted that neuronal damage in HIV infection results mainly from microglial activation and involves apoptosis, oxidative stress and glutamate-mediated neurotoxicity. Glutamate toxicity acts via 2 distinct pathways: an excitotoxic one in which glutamate receptors are hyperactivated, and an oxidative one in which cystine uptake is inhibited, resulting in glutathione depletion and oxidative stress. A number of studies show that astrocytes normally take up glutamate, keeping extracellular glutamate concentration low in the brain and preventing excitotoxicity. This action is inhibited in HIV infection, probably due to the effects of inflammatory mediators and viral proteins. Other in vitro studies as well as in vivo experiments in rodents following mechanical stimulation, show that activated microglia and brain macrophages express high affinity glutamate transporters. These data have been confirmed in chronic inflammation of the brain, particularly in SIV infection, where activated microglia and brain macrophages also express glutamine synthetase. Recent studies in humans with HIV infection show that activated microglia and brain macrophages express the glutamate transporter EAAT-1 and that expression varies according to the disease stage. This suggests that, besides their recognized neurotoxic properties in HIV infection, these cells also have a neuroprotective function, and may partly make up for the inhibited astrocytic function, at least temporarily. This hypothesis might explain the discrepancy between microglial activation which occurs early in the disease, and neuronal apoptosis and neuronal loss which is a late event. In this review article, we discuss the possible neuroprotective and neurotrophic roles of activated microglia and macrophages that may be generated by the expression of high affinity glutamate transporters and glutamine synthetase, 2 major effectors of glial glutamate metabolism, and the implications for HIV-induced neuronal dysfunction, the underlying cause of HIV dementia.
Collapse
Affiliation(s)
- Gabriel Gras
- CEA, Service de Neurovirologie, DSV/DRM, Centre de Recherches du Service de Santé des Armées, EPHE, IPSC, 92265 Fontenay aux Roses cedex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Hollins DL, Suliman HB, Piantadosi CA, Carraway MS. Glutathione regulates susceptibility to oxidant-induced mitochondrial DNA damage in human lymphocytes. Free Radic Biol Med 2006; 40:1220-6. [PMID: 16545690 DOI: 10.1016/j.freeradbiomed.2005.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Revised: 11/08/2005] [Accepted: 11/11/2005] [Indexed: 11/15/2022]
Abstract
Oxidative damage to mitochondrial DNA (mtDNA) interferes with the expression of mitochondrial-encoded subunits of the electron transport complexes of oxidative phosphorylation. MtDNA is protected by several mitochondrial antioxidant systems, but the specific importance of glutathione is unknown. We hypothesized that glutathione protects mtDNA from oxidative damage in human blood lymphocytes and that glutathione depletion increases susceptibility to mtDNA depletion, which increases vulnerability to apoptosis. MtDNA damage was measured in human blood lymphocytes exposed to tert-butyl-hydroperoxide (t-BOOH) or t-BOOH plus the glutathione analog, glutathione ethyl ester (GEE). Mitochondrial oxidative stress, mtDNA damage, and susceptibility to apoptosis were analyzed after glutathione depletion with buthionine sulfoximine (BSO). The data show selective damage to lymphocyte mtDNA at low concentrations of tBOOH that is attenuated by glutathione supplementation. Moreover, inhibition of glutathione synthesis led to lymphocyte ROS generation and mtDNA damage, and increased susceptibility to receptor-mediated apoptosis. These findings implicate the glutathione system in maintaining mtDNA integrity and resistance to apoptosis in lymphocytes and suggest that assessment of mtDNA damage in blood lymphocytes may be a useful marker of oxidative stress in humans.
Collapse
Affiliation(s)
- Dawn L Hollins
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
15
|
Kurozumi R, Kojima S. Increase of intracellular glutathione by low-level NO mediated by transcription factor NF-κB in RAW 264.7 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1744:58-67. [PMID: 15878398 DOI: 10.1016/j.bbamcr.2004.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 11/15/2004] [Accepted: 11/15/2004] [Indexed: 11/22/2022]
Abstract
The mechanism underlying the elevation of intracellular glutathione (GSH) in RAW 264.7 cells exposed to low concentrations of sodium nitroprusside (SNP), a well-known nitric oxide (NO) donor, was investigated. The peak of intracellular GSH was reached at 6 h after exposure of the cells to SNP (0.1-0.5 mM), and this was preceded by the induction of mRNA for gamma-glutamylcysteine synthetase (gamma-GCS; the rate-limiting enzyme of de novo GSH synthesis), which peaked at 3 h. N-alpha-Tosyl-L-phenylalanine chloromethyl ketone (TPCK) and caffeic acid phenethyl ester (CAPE), specific inhibitors of NF-kappaB, significantly suppressed the SNP-induced elevation of GSH protein and gamma-GCS mRNA, while curcumin, an inhibitor of AP-1, was less effective. Electrophoretic mobility shift assay (EMSA) showed that SNP exposure markedly increased the DNA binding of NF-kappaB, but not that of AP-1. Deletion or mutagenesis of the NF-kappaB site in the gamma-GCS gene promoter abolished the SNP-induced up-regulation of GSH protein and gamma-GCS mRNA. These results suggest that the elevation of intracellular GSH in RAW 264.7 cells exposed to low concentrations of SNP occurs through the operation of the de novo GSH pathway, and is mediated by transcriptional up-regulation of the gamma-GCS gene, predominantly at the NF-kappaB binding site in its promoter.
Collapse
Affiliation(s)
- Risa Kurozumi
- The Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba 278-8510, Japan.
| | | |
Collapse
|
16
|
Swintek AU, Christoph S, Petrat F, de Groot H, Kirsch M. Cell type-dependent release of nitric oxide and/or reactive nitrogenoxide species from intracellular SIN-1: effects on cellular NAD(P)H. Biol Chem 2005; 385:639-48. [PMID: 15318813 DOI: 10.1515/bc.2004.079] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
SIN-1 is frequently used in cell culture studies as an extracellularly operating generator of peroxynitrite. However, little is known about the nature of the reactive species produced intracellulary from SIN-1. SIN-1 can easily penetrate cells as exemplified for both L-929 mouse fibroblasts and bovine aortic endothelial cells (BAECs) by utilizing capillary zone electrophoresis. In L-929 cells, SIN-1 produced nitric oxide (*NO) as monitored by the fluorescent *NO scavenger FNOCT-1 and by means of a *NO electrode, as well as reactive nitrogenoxide species (RNOS, e.g. peroxynitrite, nitrogen dioxide, dinitrogen trioxide), as detected with the fluorescent indicator DAF-2. Laser scanning microscopy revealed that in L-929 cells SIN-1 -derived species initially oxidized the major fraction of the NAD(P)H within the cytosol and the nuclei, whereas the mitochondrial NAD(P)H level was somewhat increased. In marked contrast to this, in BAECs no evidence for *NO formation was found although the intracellular amount of SIN-1 was four-fold higher than in L-929 cells. In BAECs, the level of NAD(P)H was slightly decreased within the first 10 min after administration of SIN-1 in both the cytosol/nuclei and mitochondria. These observations reflect the capability of SIN-1 to generate intracellularly either almost exclusively RNOS as in BAECs, or RNOS and freely diffusing *NO as in L-929 cells. Nitric oxide as well as RNOS may decisively affect cellular metabolism as indicated by the alterations in the NAD(P)H level. Hence, care should be taken when applying SIN-1 as an exclusively peroxynitrite-generating compound in cell culture systems.
Collapse
Affiliation(s)
- Andrea U Swintek
- Institut für Physiologische Chemie, Universitätsklinikum Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | | | | | | | | |
Collapse
|
17
|
Kurozumi R, Takahashi M, Kojima S. Involvement of Mitochondrial Peroxynitrite in Nitric Oxide-Induced Glutathione Synthesis. Biol Pharm Bull 2005; 28:779-85. [PMID: 15863878 DOI: 10.1248/bpb.28.779] [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: 11/22/2022]
Abstract
Cells respond to oxidative stress including nitric oxide (NO) by increasing cellular glutathione concentration, as a part of adaptive response against oxidative injury. To elucidate the mechanism by which NO induces glutathione we investigated the reactive oxygen species (ROS) generated in the cell. Treatment of RAW264.7 cells with NO donor, sodium nitroprusside (SNP), resulted in a temporary increase in glutathione in a dose-dependent manner, which peaked between 6 h and 12 h after treatment, whereas expression of gamma-glutamylcysteine synthetase (gamma-GCS) mRNA peaked around 3 h after treatment. The increase was inhibited by NO scavengers, oxyhemoglobin and carboxyl-2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (PTIO). N-Acetyl-L-cysteine (NAC) also reduced the increase in glutathione to some extent, whereas both peroxynitrite scavenger ebselen and hydroxyl radical scavenger DMSO inhibited the increase in glutathione in a dose-dependent manner and complete inhibition was observed. Hydrogen peroxide exogenously added to the cell did not increase either glutathione or gamma-GCS expression at any concentration, indicating that involvement of hydrogen peroxide is not likely. Flow cytometric analysis showed that SNP induced a marked dose-dependent increase in Rhodamine123 fluorescence, which was completely inhibited by ebselen in a dose-dependent manner, whereas, little increase in 2',7'-dichlorofluorescin (DCF) fluorescence was observed. Generation of peroxynitrite in mitochondria by SNP was confirmed by elevated level of nitrotyrosine in a mitochondria fraction isolated from SNP-treated cells, and the elevation was completely inhibited by ebselen as well. These results suggest that induction of glutathione (GSH) synthesis by SNP treatment is mediated by peroxynitrite generated in mitochondria.
Collapse
Affiliation(s)
- Risa Kurozumi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan
| | | | | |
Collapse
|
18
|
Khullar S, Greenwood SL, McCord N, Glazier JD, Ayuk PTY. Nitric oxide and superoxide impair human placental amino acid uptake and increase Na+ permeability: implications for fetal growth. Free Radic Biol Med 2004; 36:271-7. [PMID: 15036346 DOI: 10.1016/j.freeradbiomed.2003.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2003] [Revised: 11/03/2003] [Accepted: 11/06/2003] [Indexed: 11/23/2022]
Abstract
Based on evidence that thiol and tyrosine reagents inhibit some amino acid transporters, we tested the hypothesis that NO- and O2- -derived free radicals would impair nutrient uptake by the human placenta. Syncytiotrophoblast microvillous plasma membrane vesicles (MVM) and placental villous fragments were exposed to the drug SIN-1 in the presence or absence of superoxide dismutase (SOD) and hemoglobin (Hb). The uptake of [3H]arginine, [3H]taurine, and [3H]leucine; [14C]MeAIB; and 22Na was studied in MVM, whereas the uptake of [3H]taurine was examined in villous fragments. Nitrotyrosine formation was assessed by Western blotting and quantified by ELISA. In MVM, SIN-1 caused an inhibition of [3H]arginine, [3H]taurine, and [14C]MeAIB uptake but had no significant effect on equilibrium [3H]leucine uptake. These effects were prevented by SOD or Hb, implying that both NO and O2- radicals were essential. In contrast, 22Na+ uptake was significantly increased, and this effect was prevented by SOD. In villous fragments, SIN-1 impaired Na+-dependent [3H]taurine uptake, with no effect on Na+-independent uptake. Increased nitrotyrosine formation was observed in MVM after SIN-1 treatment. Endogenous NO- and O2- -derived free radicals may alter human placental nutrient transfer in vivo, with implications for fetal growth.
Collapse
Affiliation(s)
- Sulekha Khullar
- Academic Unit of Child Health, Manchester University, St. Mary's Hospital, Manchester M13 0JH, UK
| | | | | | | | | |
Collapse
|
19
|
Buckley BJ, Marshall ZM, Whorton AR. Nitric oxide stimulates Nrf2 nuclear translocation in vascular endothelium. Biochem Biophys Res Commun 2003; 307:973-9. [PMID: 12878207 DOI: 10.1016/s0006-291x(03)01308-1] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vascular endothelial cells respond to nitric oxide by activating MAPK pathways and upregulating stress-activated proteins such as gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase-1 (HO-1). Since consensus sequences for the antioxidant response element (ARE) are found in the promoters of the gamma-GCS and HO-1 genes, we examined nuclear translocation of Nrf2, a CNC-bZIP protein which binds to and activates the ARE. We found a dramatic increase in Nrf2 nuclear translocation 1-8h following the nitric oxide donor spermine NONOate. Translocation was inhibited by pretreatment of cells with N-acetylcysteine suggesting involvement of an oxidative mechanism in this response. Translocation was also blocked by PD 98059 and SB 203580, inhibitors of ERK and p38 pathways, respectively. In addition to effects on Nrf2 subcellular localization, spermine NONOate increased Nrf2 protein levels by a mechanism which was inhibited by PD 98059. Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. These results suggest that ERK and p38 pathways may regulate nitric oxide-mediated adaptive responses in vascular endothelium via translocation of Nrf2 and activation of the ARE.
Collapse
Affiliation(s)
- Barbara J Buckley
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
20
|
Kurozumi R, Tokuzumi S, Kojima S. Does peroxynitrite involve in the elevation of cellular glutathione induced by sodium nitroprusside (SNP) in RAW 264.7 cells? Biol Pharm Bull 2003; 26:1070-5. [PMID: 12913252 DOI: 10.1248/bpb.26.1070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanism underlying the elevation of intracellular glutathione (GSH) in RAW 264.7 cells exposed to low-level sodium nitroprusside (SNP) was investigated by measuring the expression of mRNA for gamma-glutamylcysteine synthetase (gamma-GCS), the rate-limiting enzyme of de novo GSH synthesis, and the GSH content. A significant elevation of expression of mRNA for gamma-GCS was observed at 3 h after exposure of the cells to SNP at a concentration of 0.25 mM. 2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (c-PTIO), N-acetylcysteine (NAC), or ebselen (Ebs) significantly suppressed the elevations induced by SNP, suggesting that hydrogen peroxide or peroxynitrite (ONOO(-)) is involved in this event as a triggering molecule. Hydrogen peroxide itself, however, did not induce the elevation of gamma-GCS mRNA and glutathione. Chemiluminescenses induced by SIN-1, a chemical ONOO(-) donor, and ONOO(-) itself were completely blocked by Ebs. SIN-1 also significantly elevated the cellular glutathione level, and the elevation was absolutely blocked by Ebs. These results suggest that the elevation of intracellular GSH in RAW 264.7 cells exposed to low-level SNP occurs via the de novo GSH pathway through transcriptional up-regulation of the gamma-GCS gene induced by peroxynitrite molecule.
Collapse
Affiliation(s)
- Risa Kurozumi
- The Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | | | | |
Collapse
|
21
|
Andreassen K, Mortensen B, Winberg JO, Huseby NE. Increased resistance towards oxidative stress accompanies enhancement of metastatic potential obtained by repeated in vivo passage of colon carcinoma cells in syngeneic rats. Clin Exp Metastasis 2003; 19:623-9. [PMID: 12498392 DOI: 10.1023/a:1020978411846] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The colon carcinoma cell line CC531 is metastatic to liver after splenic injection in syngeneic rats. After repeated in vivo passages, a subline was selected that produced liver metastases at a considerably higher rate than the original cell line. These cells were characterized by increased intracellular glutathione, proliferation and ability to restore glutathione after exposure to oxidative stress, thus indicating an elevated resistance to oxidative stress. Furthermore, the increased metastatic ability was also accompanied by increased proliferation rate, adhesion to extracellular matrix proteins and endothelial cells, and secretion of a 60 kD matrix metalloproteinase. When cultured in vitro for a prolonged time (more than 30 trypsinizations), the cells showed a reduced in vivo metastatic ability, reduced secretion of three metalloproteinases including the 60 kD proteinase, and reduced intracellular glutathione. These results indicate that metastatic ability can be influenced through several adaptive mechanisms, and that the cell's ability to resist oxidative stress and maintain intracellular glutathione are of central importance.
Collapse
|
22
|
Mann GE, Yudilevich DL, Sobrevia L. Regulation of amino acid and glucose transporters in endothelial and smooth muscle cells. Physiol Rev 2003; 83:183-252. [PMID: 12506130 DOI: 10.1152/physrev.00022.2002] [Citation(s) in RCA: 286] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
While transport processes for amino acids and glucose have long been known to be expressed in the luminal and abluminal membranes of the endothelium comprising the blood-brain and blood-retinal barriers, it is only within the last decades that endothelial and smooth muscle cells derived from peripheral vascular beds have been recognized to rapidly transport and metabolize these nutrients. This review focuses principally on the mechanisms regulating amino acid and glucose transporters in vascular endothelial cells, although we also summarize recent advances in the understanding of the mechanisms controlling membrane transport activity and expression in vascular smooth muscle cells. We compare the specificity, ionic dependence, and kinetic properties of amino acid and glucose transport systems identified in endothelial cells derived from cerebral, retinal, and peripheral vascular beds and review the regulation of transport by vasoactive agonists, nitric oxide (NO), substrate deprivation, hypoxia, hyperglycemia, diabetes, insulin, steroid hormones, and development. In view of the importance of NO as a modulator of vascular tone under basal conditions and in disease and chronic inflammation, we critically review the evidence that transport of L-arginine and glucose in endothelial and smooth muscle cells is modulated by bacterial endotoxin, proinflammatory cytokines, and atherogenic lipids. The recent colocalization of the cationic amino acid transporter CAT-1 (system y(+)), nitric oxide synthase (eNOS), and caveolin-1 in endothelial plasmalemmal caveolae provides a novel mechanism for the regulation of NO production by L-arginine delivery and circulating hormones such insulin and 17beta-estradiol.
Collapse
Affiliation(s)
- Giovanni E Mann
- Centre for Cardiovascular Biology and Medicine, Guy's, King's, and St. Thomas' School of Biomedical Sciences, King's College London, London, United Kingdom.
| | | | | |
Collapse
|
23
|
Huseby NE, Asare N, Wetting S, Mikkelsen IM, Mortensen B, Wellman M. Role of gamma-glutamyltransferase in the homeostasis of glutathione during oxidative and nitrosative stress. Biofactors 2003; 17:151-60. [PMID: 12897437 DOI: 10.1002/biof.5520170115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- N-E Huseby
- Department of Medical Biochemistry, University of Tromsø, Norway
| | | | | | | | | | | |
Collapse
|
24
|
Gegg ME, Clark JB, Heales SJR. Determination of glutamate-cysteine ligase (gamma-glutamylcysteine synthetase) activity by high-performance liquid chromatography and electrochemical detection. Anal Biochem 2002; 304:26-32. [PMID: 11969185 DOI: 10.1006/abio.2001.5607] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The tripeptide glutathione (gamma-glutamylcysteinylglycine; GSH) is the predominant low molecular mass thiol in cells. The function of GSH is of considerable interest, with the molecule being implicated in numerous cellular processes in addition to being a major cellular antioxidant. The enzyme glutamate-cysteine ligase (GCL) is the rate-limiting step in GSH synthesis. The GCL assay described here is based on high-performance liquid chromatography and exploits the electrochemically active nature of gamma-glutamylcysteine (gamma-GC), the product of GCL activity. This method allows for the direct detection of gamma-GC rather than relying on derivatization of the molecule or linked assays. The sensitivity of the assay is sufficient to allow for the measurement of GCL activity in cultured cells. The specific activity of GCL in rat primary culture astrocytes was 9.7 +/- 1.7 nmol gamma-GC synthesized/min/mg protein.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Molecular Pathogenesis, Division of Neurochemistry, Institute of Neurology, UCL, Queen Square, London, WC1N 3BG, United Kingdom
| | | | | |
Collapse
|
25
|
Levonen AL, Patel RP, Brookes P, Go YM, Jo H, Parthasarathy S, Anderson PG, Darley-Usmar VM. Mechanisms of cell signaling by nitric oxide and peroxynitrite: from mitochondria to MAP kinases. Antioxid Redox Signal 2001; 3:215-29. [PMID: 11396477 DOI: 10.1089/152308601300185188] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Many of the biological and pathological effects of nitric oxide (NO) are mediated through cell signaling pathways that are initiated by NO reacting with metalloproteins. More recently, it has been recognized that the reaction of NO with free radicals such as superoxide and the lipid peroxyl radical also has the potential to modulate redox signaling. Although it is clear that NO can exert both cytotoxic and cytoprotective actions, the focus of this overview are those reactions that could lead to protection of the cell against oxidative stress in the vasculature. This will include the induction of antioxidant defenses such as glutathione, activation of mitogen-activated protein kinases in response to blood flow, and modulation of mitochondrial function and its impact on apoptosis. Models are presented that show the increased synthesis of glutathione in response to shear stress and inhibition of cytochrome c release from mitochondria. It appears that in the vasculature NO-dependent signaling pathways are of three types: (i) those involving NO itself, leading to modulation of mitochondrial respiration and soluble guanylate cyclase; (ii) those that involve S-nitrosation, including inhibition of caspases; and (iii) autocrine signaling that involves the intracellular formation of peroxynitrite and the activation of the mitogen-activated protein kinases. Taken together, NO plays a major role in the modulation of redox cell signaling through a number of distinct pathways in a cellular setting.
Collapse
Affiliation(s)
- A L Levonen
- Department of Pathology, University of Alabama at Birmingham, 35294-0019, USA
| | | | | | | | | | | | | | | |
Collapse
|