84 results on '"James M. May"'
Search Results
2. Intracellular ascorbate tightens the endothelial permeability barrier through Epac1 and the tubulin cytoskeleton
- Author
-
Elizabeth M. Rhea, James M. May, Zhi-chao Qu, Morgan R. Hecker, and William H. Parker
- Subjects
0301 basic medicine ,Endothelium ,Physiology ,Ascorbic Acid ,Nitric Oxide ,Microtubules ,Permeability ,Cell Line ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,Tubulin ,Cyclic AMP ,Human Umbilical Vein Endothelial Cells ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,Protein kinase A ,Cytoskeleton ,biology ,Vitamin C ,Cell Biology ,Ascorbic acid ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Nitric oxide synthase ,Oxidative Stress ,Nocodazole ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Call for Papers ,biology.protein ,Intracellular - Abstract
Vitamin C, or ascorbic acid, both tightens the endothelial permeability barrier in basal cells and also prevents barrier leak induced by inflammatory agents. Barrier tightening by ascorbate in basal endothelial cells requires nitric oxide derived from activation of nitric oxide synthase. Although ascorbate did not affect cyclic AMP levels in our previous study, there remains a question of whether it might activate downstream cyclic AMP-dependent pathways. In this work, we found in both primary and immortalized cultured endothelial cells that ascorbate tightened the endothelial permeability barrier by ∼30%. In human umbilical vein endothelial cells, this occurred at what are likely physiologic intracellular ascorbate concentrations. In so doing, ascorbate decreased measures of oxidative stress and also flattened the cells to increase cell-to-cell contact. Inhibition of downstream cyclic AMP-dependent proteins via protein kinase A did not prevent ascorbate from tightening the endothelial permeability barrier, whereas inhibition of Epac1 did block the ascorbate effect. Although Epac1 was required, its mediator Rap1 was not activated. Furthermore, ascorbate acutely stabilized microtubules during depolymerization induced by colchicine and nocodazole. Over several days in culture, ascorbate also increased the amount of stable acetylated α-tubulin. Microtubule stabilization was further suggested by the finding that ascorbate increased the amount of Epac1 bound to α-tubulin. These results suggest that physiologic ascorbate concentrations tighten the endothelial permeability barrier in unstimulated cells by stabilizing microtubules in a manner downstream of cyclic AMP that might be due both to increasing nitric oxide availability and to scavenging of reactive oxygen or nitrogen species.
- Published
- 2016
- Full Text
- View/download PDF
3. Cardiolipin fatty acid remodeling regulates mitochondrial function by modifying the electron entry point in the respiratory chain
- Author
-
James M. May, William E. Zackert, Sergey Dikalov, Aurelia Vergeade, L. Jackson Roberts, Alfiya Bikineyeva, Clinton C. Bertram, Olivier Boutaud, and Sandra S. Zinkel
- Subjects
0301 basic medicine ,Antipyretics ,Cardiolipins ,Cell Respiration ,Respiratory chain ,Flavoprotein ,Mitochondrion ,Quinone oxidoreductase ,Article ,Electron Transport ,03 medical and health sciences ,Electron transfer ,chemistry.chemical_compound ,Cardiolipin ,Humans ,Molecular Biology ,Cells, Cultured ,Myeloid Progenitor Cells ,Acetaminophen ,chemistry.chemical_classification ,030102 biochemistry & molecular biology ,biology ,Fatty Acids ,Quinones ,Fatty acid ,Cell Biology ,Electron transport chain ,Mitochondria ,030104 developmental biology ,chemistry ,Biochemistry ,biology.protein ,Molecular Medicine - Abstract
Modifications of cardiolipin (CL) levels or compositions are associated with changes in mitochondrial function in a wide range of pathologies. We have made the discovery that acetaminophen remodels CL fatty acids composition from tetralinoleoyl to linoleoyltrioleoyl-CL, a remodeling that is associated with decreased mitochondrial respiration. Our data show that CL remodeling causes a shift in electron entry from complex II to the β-oxidation electron transfer flavoprotein quinone oxidoreductase (ETF/QOR) pathway. These data demonstrate that electron entry in the respiratory chain is regulated by CL fatty acid composition and provide proof-of-concept that pharmacological intervention can be used to modify CL composition.
- Published
- 2016
- Full Text
- View/download PDF
4. Ascorbic acid repletion: A possible therapy for diabetic macular edema?
- Author
-
James M. May
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endothelium ,Apoptosis ,Ascorbic Acid ,Nitric Oxide ,medicine.disease_cause ,Biochemistry ,Article ,Macular Edema ,Retina ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Superoxides ,Physiology (medical) ,Internal medicine ,Diabetes mellitus ,medicine ,Humans ,Macular edema ,Diabetic Retinopathy ,NADPH oxidase ,biology ,business.industry ,Diabetic retinopathy ,medicine.disease ,Ascorbic acid ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,030221 ophthalmology & optometry ,biology.protein ,business ,Oxidative stress - Abstract
Macular edema poses a significant risk for visual loss in persons with diabetic retinopathy. It occurs when plasma constituents and fluid leak out of damaged retinal microvasculature in the area of the macula, causing loss of central vision. Apoptotic loss of pericytes surrounding capillaries is perhaps the earliest feature of diabetic vascular damage in the macula, which is also associated with dysfunction of the endothelium and loss of the otherwise very tight endothelial permeability barrier. Increased oxidative stress is a key feature of damage to both cell types, mediated by excess superoxide from glucose-induced increases in mitochondrial metabolism, as well as by activation of the receptor for advanced glycation end products (RAGE). The latter in turn activates multiple pathways, some of which lead to increased oxidative stress, such as those involving NF-ĸB, NADPH oxidase, and endothelial nitric oxide synthase. Such cellular oxidative stress is associated with low cellular and plasma ascorbic acid levels in many subjects with diabetes in poor glycemic control. Whether repletion of low ascorbate in retinal endothelium and pericytes might help to prevent diabetic macular edema is unknown. However, cell culture studies show that the vitamin prevents high-glucose and RAGE-induced apoptosis in both cell types, that it preserves nitric oxide generated by endothelial cells, and that it tightens the leaky endothelial permeability barrier. Although these findings need to be confirmed in pre-clinical animal studies, it is worth considering clinical trials to determine whether adequate ascorbate repletion is possible and whether it might help to delay or even reverse early diabetic macular edema.
- Published
- 2016
- Full Text
- View/download PDF
5. Loss of Rictor in Monocyte/Macrophages Suppresses Their Proliferation and Viability Reducing Atherosclerosis in LDLR Null Mice
- Author
-
Jiansheng Huang, Lei Ding, James M. May, Youmin Zhang, Vladimir R. Babaev, and MacRae F. Linton
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Cell signaling ,proliferation ,Immunology ,mTORC1 ,Biology ,mTORC2 ,03 medical and health sciences ,0302 clinical medicine ,Akt signaling ,medicine ,Immunology and Allergy ,Macrophage ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Monocyte ,apoptosis ,macrophages ,Interleukin 10 ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,atherosclerosis ,monocytes ,lcsh:RC581-607 ,030217 neurology & neurosurgery - Abstract
Background Rictor is an essential component of mammalian target of rapamycin (mTOR) complex 2 (mTORC2), a conserved serine/threonine kinase that may play a role in cell proliferation, survival and innate or adaptive immune responses. Genetic loss of Rictor inactivates mTORC2, which directly activates Akt S473 phosphorylation and promotes pro-survival cell signaling and proliferation. Methods and results To study the role of mTORC2 signaling in monocytes and macrophages, we generated mice with myeloid lineage-specific Rictor deletion (MRictor-/-). These MRictor-/- mice exhibited dramatic reductions of white blood cells, B-cells, T-cells, and monocytes but had similar levels of neutrophils compared to control Rictor flox-flox (Rictorfl/fl) mice. MRictor-/- bone marrow monocytes and peritoneal macrophages expressed reduced levels of mTORC2 signaling and decreased Akt S473 phosphorylation, and they displayed significantly less proliferation than control Rictorfl/fl cells. In addition, blood monocytes and peritoneal macrophages isolated from MRictor-/- mice were significantly more sensitive to pro-apoptotic stimuli. In response to LPS, MRictor-/- macrophages exhibited the M1 phenotype with higher levels of pro-inflammatory gene expression and lower levels of Il10 gene expression than control Rictorfl/fl cells. Further suppression of LPS-stimulated Akt signaling with a low dose of an Akt inhibitor, increased inflammatory gene expression in macrophages, but genetic inactivation of Raptor reversed this rise, indicating that mTORC1 mediates this increase of inflammatory gene expression. Next, to elucidate whether mTORC2 has an impact on atherosclerosis in vivo, female and male Ldlr null mice were reconstituted with bone marrow from MRictor-/- or Rictorfl/fl mice. After 10 weeks of the Western diet, there were no differences between the recipients of the same gender in body weight, blood glucose or plasma lipid levels. However, both female and male MRictor-/- → Ldlr-/- mice developed smaller atherosclerotic lesions in the distal and proximal aorta. These lesions contained less macrophage area and more apoptosis than lesions of control Rictorfl/fl → Ldlr-/- mice. Thus, loss of Rictor and, consequently, mTORC2 significantly compromised monocyte/macrophage survival, and this markedly diminished early atherosclerosis in Ldlr-/- mice. Conclusion Our results demonstrate that mTORC2 is a key signaling regulator of macrophage survival and its depletion suppresses early atherosclerosis.
- Published
- 2018
- Full Text
- View/download PDF
6. Sodium-dependent vitamin C transporter-2 mediates vitamin C transport at the cortical nerve terminal
- Author
-
L. Nora Zeidan, Amita Raj, Heinrich J.G. Matthies, Katherine M. Betke, Marquicia R. Pierce, and James M. May
- Subjects
Synaptosome ,Synaptic cleft ,biology ,Chemistry ,Sodium-Coupled Vitamin C Transporters ,Ascorbic acid ,Cell biology ,Reuptake ,Cellular and Molecular Neuroscience ,Monoamine neurotransmitter ,Norepinephrine transporter ,Biochemistry ,biology.protein ,Norepinephrine Plasma Membrane Transport Proteins - Abstract
It has been shown that vitamin C (VC) is transported at synaptic boutons, but how this occurs has not been elucidated. This study investigates the role of the sodium-dependent vitamin C transporter-2 (SVCT2) in transporting VC at the cortical nerve terminal. Immunostaining of cultured mouse superior cervical ganglion cells showed the SVCT2 to be expressed in presynaptic boutons, colocalizing with the vesicular monoamine transporter-2 and the norepinephrine transporter. Immunoblotting of enriched cortical synaptosomes demonstrated that the SVCT2 was enriched in presynaptic fractions, confirming a predominantly presynaptic location. In crude synaptosomes, known inhibitors of SVCT2 inhibited uptake of VC. Furthermore, the kinetic features of VC uptake were consistent with SVCT2-mediated function. VC was also found to efflux from synaptosomes by a mechanism not involving the SVCT2. Indeed, VC efflux was substantially offset by reuptake of VC on the SVCT2. The presence and function of the SVCT2 at the presynaptic nerve terminal suggest that it is the transporter responsible for recovery of VC released into the synaptic cleft.
- Published
- 2015
- Full Text
- View/download PDF
7. Ascorbic acid transport in brain microvascular pericytes
- Author
-
Zhi-chao Qu, James M. May, and William H. Parker
- Subjects
Metabolic Clearance Rate ,Biophysics ,Ascorbic Acid ,Biology ,Biochemistry ,Article ,Cell Line ,Western blot ,medicine ,Humans ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Cells, Cultured ,Vitamin C ,medicine.diagnostic_test ,Glucose transporter ,Transporter ,Cell Biology ,Ascorbic acid ,medicine.anatomical_structure ,Blood-Brain Barrier ,Apoptosis ,Microvessels ,Pericyte ,Pericytes ,Intracellular ,Subcellular Fractions - Abstract
Intracellular vitamin C, or ascorbic acid, has been shown to prevent the apoptosis of cultured vascular pericytes under simulated diabetic conditions. We sought to determine the mechanism by which ascorbate is transported into pericytes prior to exerting this protective effect. Measuring intracellular ascorbate, we found that pericytes display a linear uptake over 30 minutes and an apparent transport Km of 21 μM, both of which are consistent with activity of the Sodium-dependent Vitamin C Transporter 2 (SVCT2). Uptake of both radiolabeled and unlabeled ascorbate was prevented by inhibiting SVCT2 activity, but not by inhibiting the activity of GLUT-type glucose transporters, which import dehydroascorbate to also generate intracellular ascorbate. Likewise, uptake of dehydroascorbate was prevented with the inhibition of GLUTs, but not by inhibiting the SVCT2, indicating substrate specificity of both transporters. Finally, presence of the SVCT2 in pericytes was confirmed by western blot analysis, and immunocytochemistry was used to localize it to the plasma membrane and intracellular sites. Together, these data clarify previous inconsistencies in the literature, implicate SVCT2 as the pericyte ascorbate transporter, and show that pericytes are capable of concentrating intracellular ascorbate against a gradient in an energy- and sodium-dependent fashion.
- Published
- 2015
- Full Text
- View/download PDF
8. Vitamin C Promotes Maturation of T-Cells
- Author
-
Gerald J. Spangrude, Laura Jean Pierce, James M. May, Jared Manning, Hongfang Wang, Dean Tantin, Daniel A. Appadurai, Vincent K. Nganga, Patrick C. Swanson, Arvind Shakya, and Birgitta Mitchell
- Subjects
Physiology ,T-Lymphocytes ,Clinical Biochemistry ,Receptors, Antigen, T-Cell ,Gene Expression ,Phthalimides ,Ascorbic Acid ,Gene Rearrangement, T-Lymphocyte ,Methylation ,Biochemistry ,Epigenesis, Genetic ,Histones ,Mice ,Animals ,Immunologic Factors ,Molecular Biology ,Cells, Cultured ,General Environmental Science ,Regulation of gene expression ,Forum Original Research CommunicationsVitamin C (J. May, Ed.) ,biology ,ZAP70 ,T-cell receptor ,Tryptophan ,Azepines ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Gene rearrangement ,Ascorbic acid ,Molecular biology ,Culture Media ,Mice, Inbred C57BL ,Histone ,Histone methyltransferase ,Histone Methyltransferases ,Quinazolines ,biology.protein ,General Earth and Planetary Sciences ,Protein Processing, Post-Translational ,CD8 - Abstract
Aims: Vitamin C (ascorbic acid) is thought to enhance immune function, but the mechanisms involved are obscure. We utilized an in vitro model of T-cell maturation to evaluate the role of ascorbic acid in lymphocyte development. Results: Ascorbic acid was essential for the developmental progression of mouse bone marrow-derived progenitor cells to functional T-lymphocytes in vitro and also played a role in vivo. Ascorbate-mediated enhancement of T-cell development was lymphoid cell-intrinsic and independent of T-cell receptor (TCR) rearrangement. Analysis of TCR rearrangements demonstrated that ascorbic acid enhanced the selection of functional TCRαβ after the stage of β-selection. Genes encoding the coreceptor CD8 as well as the kinase ZAP70 were upregulated by ascorbic acid. Pharmacologic inhibition of methylation marks on DNA and histones enhanced ascorbate-mediated differentiation, suggesting an epigenetic mechanism of Cd8 gene regulation via active demethylation by ascorbate-dependent Fe2+ and 2-oxoglutarate-dependent dioxygenases. Innovation: We speculate that one aspect of gene regulation mediated by ascorbate occurs at the level of chromatin demethylation, mediated by Jumonji C (JmjC) domain enzymes that are known to be reliant upon ascorbate as a cofactor. JmjC domain enzymes are also known to regulate transcription factor activity. These two mechanisms are likely to play key roles in the modulation of immune development and function by ascorbic acid. Conclusion: Our results provide strong experimental evidence supporting a role for ascorbic acid in T-cell maturation as well as insight into the mechanism of ascorbate-mediated enhancement of immune function. Antioxid. Redox Signal. 19, 2054–2067.
- Published
- 2013
- Full Text
- View/download PDF
9. Regulation of embryonic neurotransmitter and tyrosine hydroxylase protein levels by ascorbic acid
- Author
-
M. Elizabeth Meredith and James M. May
- Subjects
Male ,Serotonin ,medicine.medical_specialty ,3,4-Dihydroxyphenylacetic acid ,Tyrosine 3-Monooxygenase ,Dopamine ,Mice, Transgenic ,Ascorbic Acid ,Biology ,Article ,Mice ,Norepinephrine ,chemistry.chemical_compound ,Catecholamines ,Internal medicine ,medicine ,Animals ,Tyrosine ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Cerebral Cortex ,Tyrosine hydroxylase ,General Neuroscience ,Tetrahydrobiopterin ,Tryptophan hydroxylase ,Ascorbic acid ,Mice, Inbred C57BL ,Endocrinology ,Monoamine neurotransmitter ,chemistry ,Biochemistry ,Female ,Neurology (clinical) ,Developmental Biology ,medicine.drug - Abstract
Scope: Ascorbic acid (ascorbate) is required to recycle tetrahydrobiopterin, which is necessary for neurotransmitter synthesis by the rate-limiting enzymes tyrosine and tryptophan hydroxylases. We sought to determine whether ascorbate might regulate embryonic brain cortex monoamine synthesis utilizing transgenic mouse models with varying intracellular ascorbate levels. Methods and results: In embryos lacking the sodium-dependent vitamin C transporter 2 (SVCT2), very low levels of brain ascorbate decreased cortex levels of norepinephrine and dopamine by approximately 33%, but had no effect on cortex serotonin or its metabolite, 5-hydroxyindole acetic acid. This decrease in ascorbate also led to a decrease in protein levels of tyrosine hydroxylase, but not of tryptophan hydroxylase. Increased cortex ascorbate in embryos carrying extra copies of the SVCT2 resulted in increased levels of dopamine and its metabolite, 3,4-dihydroxyphenylacetic acid (DOPAC), as well as serotonin and 5-hydroxyindole acetic acid. Conclusion: The dependence of embryonic brain cortex neurotransmitter synthesis and tyrosine hydroxylase expression on intracellular ascorbate emphasizes the importance of receiving adequate ascorbate during development.
- Published
- 2013
- Full Text
- View/download PDF
10. Abstract 2: Loss of Rictor in Macrophages Suppresses Their Viability and Reduces Atherosclerosis in LDLR Null Mice
- Author
-
Lei Ding, James M. May, MacRae F. Linton, Youmin Zhang, and Vladimir R. Babaev
- Subjects
Serine ,Biochemistry ,Kinase ,digestive, oral, and skin physiology ,LDL receptor ,Viability assay ,Metabolism ,Threonine ,Biology ,Cardiology and Cardiovascular Medicine ,mTORC2 ,PI3K/AKT/mTOR pathway ,Cell biology - Abstract
The mammalian target of rapamycin (mTOR) is a conserved serine/threonine kinase that plays a central role in the regulation of cell viability, growth and metabolism. mTOR complex 2 (mTORC2) directly activates phosphorylation of Akt at S 473 , promoting pro-survival signaling. Rictor is an essential component of mTORC2, and genetic loss of Rictor inactivates the complex. To examine whether macrophage mTORC2 signaling has an impact on atherosclerosis, we transplanted male Ldlr null mice with bone marrow isolated from male mice with myeloid-specific Rictor deletion ( Rictor -/- , n=9) and control marrow from Rictor flox-flox mice ( Rictor flox/flox ; n=10). Compared to control mice reconstituted with Rictor flox/flox cells, the recipients of Rictor -/- bone marrow cells exhibited dramatic changes in blood cells including lower levels of white blood cells, B-cells, T-cells and monocytes but had similar levels of neutrophils. After 8 weeks of the Western diet, both groups of recipients had similar levels of body weight, blood glucose, plasma total cholesterol and triglycerides. However, Rictor -/- → Ldlr -/- mice developed smaller atherosclerotic lesions in the proximal and distal aorta (46 and 40% reduction, respectively). These lesions contained less macrophage area and more apoptotic macrophages than lesions of control Rictor flox/flox → Ldlr -/- mice. Importantly, blood monocytes and peritoneal macrophages isolated from Rictor -/- → Ldlr -/- mice were more sensitive to apoptotic stimuli compared to control Rictor flox/flox cells. In response to LPS, Rictor -/- macrophages exhibited the M1 phenotype with high levels of pro-inflammatory gene expression. Both Rictor -/- blood monocytes and macrophages had lower levels of Il10 gene expression than Rictor flox/flox cells. Thus, loss of Rictor and, consequently, mTORC2 in monocyte/macrophages significantly compromises their survival, and this markedly diminishes early atherosclerosis in Ldlr -/- mice. Our results indicate that mTORC2 is a key signaling regulator of macrophage survival and inflammatory responses and promote atherosclerosis.
- Published
- 2016
- Full Text
- View/download PDF
11. Abstract 129: Overexpression of P110alpha Induces Vascular Malformation in Mouse Tail, Feet and Ears
- Author
-
Vladimir R. Babaev, Lei Ding, MacRae F. Linton, Jeremy A Brooksbank, James M. May, and Youmin Zhang
- Subjects
chemistry.chemical_classification ,Kinase ,Cell growth ,Vascular malformation ,Cellular functions ,Motility ,Biology ,medicine.disease ,Cell biology ,Enzyme ,chemistry ,medicine ,Signal transduction ,Cardiology and Cardiovascular Medicine - Abstract
Phosphoinositide-3 kinases (PI3Ks) are a family of signal transduction enzymes involved in critical cellular functions such as cell growth, proliferation, differentiation, motility and survival. Class IA PI3K is composed of a heterodimer between a P110 catalytic subunit and a p85 regulatory subunit. Upregulation of P110alpha protein significantly propagates second messenger response accelerating Akt signaling. To augment Akt activity in macrophages, we generated mice with conditional myeloid lineage-specific (LysM-Cre) overexpression of P110alpha (M-P110α+). Peritoneal macrophages isolated from these mice expressed an additional band slightly different in size than P110alpha protein and had markedly increased phosphorylation of Akt. These mice also exhibited dramatic changes in blood cells including lower levels of white blood cells, B-cells and an increase in neutrophils; whereas the levels of T-cells and monocytes were not different compared to wild-type mice. Because of the defects of B-cell development, M-P110α+ mice had also splenomegaly with decreased levels of follicles. Unexpectedly, M-P110α+ mice at age of 3-4 weeks spontaneously developed a striking phenotype with massive enlargements of small vessels forming malformations in the extremities including tail, feet and ears. These vascular malformations grew very rapidly, and, at age 5-7 months, M-P110α+ mice developed severe hemorrhages from lesions on the tail and feet. The malformations appeared to be mainly venous by Doppler color ultrasonography, and histologically they represented overgrowth of venous and capillary cells that expressed endothelial markers including CD31/PECAM-1, von Willibrand factor and panendothelial cell antigen, MECA-32. These endothelial cells exhibited high levels of proliferation detected by Ki67 staining. In addition, the malformations were surrounded by high numbers of macrophages. In conclusion, M-P110α+ mice develop spontaneous capillary-venous malformations that represent a novel model of the human PIK3CA-related overgrowth spectrum. This mouse model provides a unique opportunity to develop novel therapeutic strategies to prevent capillary and venous malformations.
- Published
- 2016
- Full Text
- View/download PDF
12. Macrophage IKKalpha deficiency suppresses Akt phosphorylation, reduces cell survival and decreases early atherosclerosis
- Author
-
Vladimir R. Babaev, MacRae F. Linton, Sergio Fazio, James M. May, Lei Ding, P. Charles Lin, and Youmin Zhang
- Subjects
0301 basic medicine ,Male ,Enzyme complex ,medicine.medical_specialty ,Time Factors ,Cell Survival ,Apoptosis ,IκB kinase ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,Article ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Phosphorylation ,Protein kinase B ,Transcription factor ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Mice, Knockout ,TOR Serine-Threonine Kinases ,I-Kappa-B Kinase ,Atherosclerosis ,Cell biology ,I-kappa B Kinase ,Liver Transplantation ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Liver ,Receptors, LDL ,Diet, Western ,Multiprotein Complexes ,Macrophages, Peritoneal ,Female ,Signal transduction ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Objective— The IκB kinase (IKK) is an enzyme complex that initiates the nuclear factor κB transcription factor cascade, which is important in regulating multiple cellular responses. IKKα is directly associated with 2 major prosurvival pathways, PI3K/Akt and nuclear factor κB, but its role in cell survival is not clear. Macrophages play critical roles in the pathogenesis of atherosclerosis, yet the impact of IKKα signaling on macrophage survival and atherogenesis remains unclear. Approach and Results— Here, we demonstrate that genetic IKKα deficiency, as well as pharmacological inhibition of IKK, in mouse macrophages significantly reduces Akt S 473 phosphorylation, which is accompanied by suppression of mTOR complex 2 signaling. Moreover, IKKα null macrophages treated with lipotoxic palmitic acid exhibited early exhaustion of Akt signaling compared with wild-type cells. This was accompanied by a dramatic decrease in the resistance of IKKα −/− monocytes and macrophages to different proapoptotic stimuli compared with wild-type cells. In vivo, IKKα deficiency increased macrophage apoptosis in atherosclerotic lesions and decreased early atherosclerosis in both female and male low-density lipoprotein receptor (LDLR) −/− mice reconstituted with IKKα −/− hematopoietic cells and fed with the Western diet for 8 weeks compared with control LDLR −/− mice transplanted with wild-type cells. Conclusions— Hematopoietic IKKα deficiency in mouse suppresses Akt signaling, compromising monocyte/macrophage survival and this decreases early atherosclerosis.
- Published
- 2016
13. Jnk1 deficiency in hematopoietic cells suppresses macrophage apoptosis and increases atherosclerosis in low-density lipoprotein receptor null mice
- Author
-
Gökhan S. Hotamisligil, Vladimir R. Babaev, Ebru Erbay, Youmin Zhang, Michele Yeung, Sergio Fazio, MacRae F. Linton, James M. May, and Lei Ding
- Subjects
Mouse ,Bone marrow cell ,Aortic disease ,Apoptosis ,Signal transduction ,Antagonists and inhibitors ,0302 clinical medicine ,Pathology ,Macrophage ,Insulin ,Mitogen activated protein kinase p38 ,Enzyme activity ,Gene repression ,Cells, Cultured ,Bone Marrow Transplantation ,Atherosclerotic ,Kinase ,C57BL mouse ,Cholesterol diet ,Hematopoietic cell ,Endoplasmic Reticulum Stress ,Plaque, Atherosclerotic ,030220 oncology & carcinogenesis ,Deficiency ,Cardiology and Cardiovascular Medicine ,Anisomycin ,Gene isoform ,endocrine system ,Low density lipoprotein receptor ,Knockout ,Hypercholesterolemia ,Aortic Diseases ,Diet, High-Fat ,Article ,03 medical and health sciences ,Pten protein, mouse ,Genetics ,Animal model ,Animal experiment ,Protein kinase B ,Animal ,Disease model ,Macrophages ,Genetic predisposition ,MAP kinase signaling system ,Mice, Inbred C57BL ,030104 developmental biology ,Stress activated protein kinase 1 ,0301 basic medicine ,JNKI1 ,Unclassified drug ,Inbred C57BL ,Cell survival ,Mitogen activated protein kinase 9 ,Anthra[1,9 cd]pyrazol 6(2h) one ,Receptors ,Aorta ,Priority journal ,Mice, Knockout ,Allele ,Atherosclerotic plaque ,Lipid diet ,Haematopoiesis ,Enzyme inhibition ,Phenotype ,embryonic structures ,Protein kinase inhibitor ,Bcl-Associated Death Protein ,bcl-Associated Death Protein ,biological phenomena, cell phenomena, and immunity ,Animal cell ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Cell Survival ,Protein BAD ,Bone Marrow Cells ,Biology ,Knockout mouse ,Animals ,Mitogen-Activated Protein Kinase 9 ,Genetic Predisposition to Disease ,Mitogen-Activated Protein Kinase 8 ,Phosphatidylinositol 3,4,5 trisphosphate 3 phosphatase ,Enzyme deficiency ,Protein Kinase Inhibitors ,Bad protein, mouse ,Drug effects ,PTEN Phosphohydrolase ,Atherosclerosis ,Nonhuman ,Molecular biology ,enzymes and coenzymes (carbohydrates) ,Disease Models, Animal ,Metabolism ,Receptors, LDL ,LDL receptor ,Cancer research ,Enzymology ,Cell culture ,Controlled study ,Proto-Oncogene Proteins c-akt - Abstract
Objective— The c-Jun NH 2 -terminal kinases (JNK) are regulated by a wide variety of cellular stresses and have been implicated in apoptotic signaling. Macrophages express 2 JNK isoforms, JNK1 and JNK2, which may have different effects on cell survival and atherosclerosis. Approach and Results— To dissect the effect of macrophage JNK1 and JNK2 on early atherosclerosis, Ldlr −/− mice were reconstituted with wild-type, Jnk1 −/− , and Jnk2 −/− hematopoietic cells and fed a high cholesterol diet. Jnk1 −/− → Ldlr −/− mice have larger atherosclerotic lesions with more macrophages and fewer apoptotic cells than mice transplanted with wild-type or Jnk2 −/− cells. Moreover, genetic ablation of JNK to a single allele ( Jnk1 +/− /Jnk2 −/− or Jnk1 −/− /Jnk2 +/− ) in marrow of Ldlr −/− recipients further increased atherosclerosis compared with Jnk1 −/− → Ldlr −/− and wild-type→ Ldlr −/− mice. In mouse macrophages, anisomycin-mediated JNK signaling antagonized Akt activity, and loss of Jnk1 gene obliterated this effect. Similarly, pharmacological inhibition of JNK1, but not JNK2, markedly reduced the antagonizing effect of JNK on Akt activity. Prolonged JNK signaling in the setting of endoplasmic reticulum stress gradually extinguished Akt and Bad activity in wild-type cells with markedly less effects in Jnk1 −/− macrophages, which were also more resistant to apoptosis. Consequently, anisomycin increased and JNK1 inhibitors suppressed endoplasmic reticulum stress–mediated apoptosis in macrophages. We also found that genetic and pharmacological inhibition of phosphatase and tensin homolog abolished the JNK-mediated effects on Akt activity, indicating that phosphatase and tensin homolog mediates crosstalk between these pathways. Conclusions— Loss of Jnk1 , but not Jnk2 , in macrophages protects them from apoptosis, increasing cell survival, and this accelerates early atherosclerosis.
- Published
- 2016
14. The SLC23 family of ascorbate transporters: ensuring that you get and keep your daily dose of vitamin C
- Author
-
James M. May
- Subjects
Pharmacology ,biology ,Biochemistry ,Membrane protein ,Vitamin C ,ATPase ,Sodium-Coupled Vitamin C Transporters ,biology.protein ,Transporter ,Ascorbic acid ,Intracellular ,Cell biology ,Transport protein - Abstract
The ascorbate transporters SVCT1 and SVCT2 are crucial for maintaining intracellular ascorbate concentrations in most cell types. Although the two transporter isoforms are highly homologous, they have different physiologic functions. The SVCT1 is located primarily in epithelial cells and has its greatest effect in reabsorbing ascorbate in the renal tubules. The SVCT2 is located in most non-epithelial tissues, with the highest expression in brain and neuroendocrine tissues. These transporters are hydrophobic membrane proteins that have a high affinity and are highly selective for ascorbate. Their ability to concentrate ascorbate inside cells is driven by the sodium gradient across the plasma membrane as generated by Na+/K+ ATPase. They can concentrate ascorbate 20 to 60-fold over plasma ascorbate concentrations. Ascorbate transport on these proteins is regulated at the transcriptional, translational and post-translational levels. Available studies show that transporter function is acutely regulated by protein kinases A and C, whereas transporter expression is increased by low intracellular ascorbate and associated oxidative stress. The knockout of the SVCT2 in mice is lethal on day 1 of life, and almost half of SVCT1 knockout mice do not survive to weaning. These findings confirm the importance both of cellular ascorbate and of the two transport proteins as key to maintaining intracellular ascorbate.
- Published
- 2011
- Full Text
- View/download PDF
15. Differential regulation of the ascorbic acid transporter SVCT2 during development and in response to ascorbic acid depletion
- Author
-
M. Elizabeth Meredith, Fiona E. Harrison, and James M. May
- Subjects
endocrine system ,medicine.medical_specialty ,animal structures ,animal diseases ,Biophysics ,Embryonic Development ,Ascorbic Acid ,Biology ,Biochemistry ,Article ,Mice ,Cerebellum ,Internal medicine ,Gene expression ,medicine ,Animals ,RNA, Messenger ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Cerebral Cortex ,Messenger RNA ,Vitamin C ,Embryogenesis ,Brain ,Biological Transport ,hemic and immune systems ,Transporter ,Embryo ,Cell Biology ,Ascorbic acid ,eye diseases ,Mice, Inbred C57BL ,Oxidative Stress ,Endocrinology ,Liver ,Intracellular - Abstract
The sodium-dependent vitamin C transporter-2 (SVCT2) is the only ascorbic acid (ASC) transporter significantly expressed in brain. It is required for life and is critical during brain development to supply adequate levels of ASC. To assess SVCT2 function in the developing brain, we studied time-dependent SVCT2 mRNA and protein expression in mouse brain, using liver as a comparison tissue because it is the site of ASC synthesis. We found that SVCT2 expression followed an inverse relationship with ASC levels in the developing brain. In cortex and cerebellum, ASC levels were high throughout late embryonic stages and early post-natal stages and decreased with age, whereas SVCT2 mRNA and protein levels were low in embryos and increased with age. A different response was observed for liver, in which ASC levels and SVCT2 expression were both low throughout embryogenesis and increased post-natally. To determine whether low intracellular ASC might be capable of driving SVCT2 expression, we depleted ASC by diet in adult mice unable to synthesize ASC. We observed that SVCT2 mRNA and protein were not affected by ASC depletion in brain cortex, but SVCT2 protein expression was increased by ASC depletion in the cerebellum and liver. The results suggest that expression of the SVCT2 is differentially regulated during embryonic development and in adulthood.
- Published
- 2011
- Full Text
- View/download PDF
16. CpG methylation at the USF-binding site mediates cell-specific transcription of human ascorbate transporter SVCT2 exon 1a
- Author
-
James M. May and Huan Qiao
- Subjects
Biology ,Decitabine ,Methylation ,Biochemistry ,Article ,Upstream Stimulatory Factor ,Cell Line ,Exon ,Epigenetics of physical exercise ,Transcription (biology) ,Cell Line, Tumor ,Humans ,Binding site ,Promoter Regions, Genetic ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Promoter ,Exons ,Cell Biology ,Molecular biology ,CCAAT-Binding Factor ,CpG site ,DNA methylation ,Azacitidine ,Upstream Stimulatory Factors ,CpG Islands ,Dinucleoside Phosphates - Abstract
SVCT2 (sodium–vitamin C co-transporter 2) is the major transporter mediating vitamin C uptake in most organs. Its expression is driven by two promoters (CpG-poor exon 1a promoter and CpG-rich exon 1b promoter). In the present study, we mapped discrete elements within the proximal CpG-poor promoter responsible for exon 1a transcription. We identified two E boxes for USF (upstream stimulating factor) binding and one Y box for NF-Y (nuclear factor Y) binding. We show further that NF-Y and USF bind to the exon 1a promoter in a co-operative manner, amplifying the binding of each to the promoter, and is absolutely required for the full activity of the exon 1a promoter. The analysis of the CpG site located at the upstream USF-binding site in the promoter showed a strong correlation between expression and demethylation. It was also shown that exon 1a transcription was induced in cell culture treated with the demethylating agent decitabine. The specific methylation of this CpG site impaired both the binding of USF and the formation of the functional NF-Y–USF complex as well as promoter activity, suggesting its importance for cell-specific transcription. Thus CpG methylation at the upstream USF-binding site functions in establishing and maintaining cell-specific transcription from the CpG-poor SVCT2 exon 1a promoter.
- Published
- 2011
- Full Text
- View/download PDF
17. Regulation of the human ascorbate transporter SVCT2 exon 1b gene by zinc-finger transcription factors
- Author
-
Huan Qiao and James M. May
- Subjects
Chromatin Immunoprecipitation ,Transcription, Genetic ,Sp1 Transcription Factor ,TATA box ,Molecular Sequence Data ,Response element ,Organic Anion Transporters, Sodium-Dependent ,Ascorbic Acid ,Biology ,Transfection ,Biochemistry ,Article ,Exon ,Sp3 transcription factor ,Physiology (medical) ,Tumor Cells, Cultured ,Animals ,Humans ,Promoter Regions, Genetic ,Sodium-Coupled Vitamin C Transporters ,Transcription factor ,YY1 Transcription Factor ,Zinc finger ,Regulation of gene expression ,Sp1 transcription factor ,Binding Sites ,Base Sequence ,Symporters ,Reverse Transcriptase Polymerase Chain Reaction ,Zinc Fingers ,Exons ,Molecular biology ,GC Rich Sequence ,Drosophila melanogaster ,Sp3 Transcription Factor ,Gene Expression Regulation ,Early Growth Response Transcription Factors ,Protein Binding - Abstract
The sodium-dependent vitamin C transporter (SVCT) 2 is crucial for ascorbate uptake in metabolically active and specialized tissues. The present study focused on the gene regulation of the SVCT2 exon 1b, which is ubiquitously expressed in human and mouse tissues. Although the human SVCT2 exon 1b promoter doesn’t contain a classical TATA-box, we found that it does contain a functional initiator (Inr) that binds YY1 and interacts with upstream Sp1/Sp3 elements in the proximal promoter region. These elements in turn play a critical role in regulating YY1-mediated transcription of the exon 1b gene. Formation of YY1/Sp complexes on the promoter is required for its optional function. YY1 with Sp1 or Sp3 synergistically enhanced exon 1b promoter activity as well as the endogenous SVCT2 protein expression. Further, in addition to Sp1/Sp3 both EGR-1 and -2 were detected in the protein complexes that bound the three GC boxes bearing overlapping binding sites for EGR/WT1 and Sp1/3. The EGR family factors, WT1 and MAZ were found to differentially regulate exon 1b promoter activity. These results show that differential occupancy of transcription factors on the GC-rich consensus sequences in SVCT2 exon 1b promoter contributes to the regulation of cell and tissue expression of SVCT2.
- Published
- 2011
- Full Text
- View/download PDF
18. Nitric oxide mediates tightening of the endothelial barrier by ascorbic acid
- Author
-
James M. May and Zhi-chao Qu
- Subjects
medicine.medical_specialty ,GUCY1B3 ,Nitric Oxide Synthase Type III ,Endothelium ,Biophysics ,Vascular permeability ,Ascorbic Acid ,Nitric Oxide ,Biochemistry ,Article ,Cell Line ,Nitric oxide ,Capillary Permeability ,chemistry.chemical_compound ,Enos ,Internal medicine ,medicine ,Humans ,Molecular Biology ,biology ,Vitamins ,Cell Biology ,Ascorbic acid ,Actin cytoskeleton ,biology.organism_classification ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Guanylate Cyclase ,Endothelium, Vascular - 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.
- Published
- 2011
- Full Text
- View/download PDF
19. Selective macrophage ascorbate deficiency suppresses early atherosclerosis
- Author
-
James M. May, Sergio Fazio, Richard R. Whitesell, MacRae F. Linton, Vladimir R. Babaev, and Liying Li
- Subjects
Male ,Apolipoprotein E ,medicine.medical_specialty ,Time Factors ,Apolipoprotein B ,Organic Anion Transporters, Sodium-Dependent ,Ascorbic Acid ,HMGB1 ,Biochemistry ,Article ,Mice ,Apolipoproteins E ,Glycation ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Macrophage ,Genetic Predisposition to Disease ,Sodium-Coupled Vitamin C Transporters ,Cells, Cultured ,Mice, Knockout ,Symporters ,biology ,Atherosclerosis ,Ascorbic acid ,Mice, Inbred C57BL ,Haematopoiesis ,Endocrinology ,Organ Specificity ,Apoptosis ,Immunology ,Ascorbic Acid Deficiency ,Disease Progression ,Macrophages, Peritoneal ,biology.protein ,Female - Abstract
To test whether severe ascorbic acid deficiency in macrophages affects progression of early atherosclerosis, we used fetal liver cell transplantation to generate atherosclerosis-prone apolipoprotein E-deficient (apoE(-/-)) mice that selectively lacked the ascorbate transporter (SVCT2) in hematopoietic cells, including macrophages. After 13 weeks of chow diet, apoE(-/-) mice lacking the SVCT2 in macrophages had surprisingly less aortic atherosclerosis, decreased lesion macrophage numbers, and increased macrophage apoptosis compared to control-transplanted mice. Serum lipid levels were similar in both groups. Peritoneal macrophages lacking the SVCT2 had undetectable ascorbate; increased susceptibility to H(2)O(2)-induced mitochondrial dysfunction and apoptosis; decreased expression of genes for COX-2, IL1β, and IL6; and decreased lipopolysaccharide-stimulated NF-κB and antiapoptotic gene expression. These changes were associated with decreased expression of both the receptor for advanced glycation end products and HIF-1α, either or both of which could have been the proximal cause of decreased macrophage activation and apoptosis in ascorbate-deficient macrophages.
- Published
- 2011
- Full Text
- View/download PDF
20. Vitamin C distribution and retention in the mouse brain
- Author
-
Fiona E. Harrison, James M. May, Roslyn J. Green, and Sean M. Dawes
- Subjects
medicine.medical_specialty ,Central nervous system ,Ascorbic Acid ,Biology ,medicine.disease_cause ,Article ,Statistics, Nonparametric ,Lipid peroxidation ,Mice ,chemistry.chemical_compound ,Malondialdehyde ,Internal medicine ,Cortex (anatomy) ,medicine ,Animals ,Tissue Distribution ,Molecular Biology ,Mice, Knockout ,Vitamin C ,General Neuroscience ,Brain ,Ascorbic acid ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Biochemistry ,Knockout mouse ,Ascorbic Acid Deficiency ,Neurology (clinical) ,L-Gulonolactone Oxidase ,Oxidative stress ,Developmental Biology - Abstract
Vitamin C (VC) is a crucial antioxidant in the brain. To assess whether different brain regions vary in their sensitivity to oxidative stress induced by VC depletion, we used the gulonolactone oxidase (gulo) knockout mouse. This mouse, like humans, cannot synthesize VC and thus its tissue VC levels can be varied by dietary VC intake. Gulo knockout mice were fed drinking water containing standard (0.33 g/L), low (0.033 g/L) or zero (0 g/L) VC supplementation levels. After 4 weeks, mice were sacrificed and different brain regions removed for assay of VC and malondialdehyde, a marker of lipid peroxidation. Compared to age-matched wild-type controls, the cerebellum, olfactory bulbs and frontal cortex had the highest VC content, whereas the pons and spinal chord had the lowest. However, in mice that did not receive VC, area differences were no longer significant as all values trended towards zero. Malondialdehyde increased in the cortex as VC supplementation was decreased. The same changes were not observed in the cerebellum or pons, suggesting that cortex is more susceptible to oxidative damage from low VC. These results suggest enhanced susceptibility of the cortex to oxidative stress induced by low VC compared to other brain regions.
- Published
- 2010
- Full Text
- View/download PDF
21. Low ascorbic acid and increased oxidative stress in gulo(−/−) mice during development
- Author
-
Jeanette Saskowski, M. Elizabeth Meredith, Sean M. Dawes, James M. May, and Fiona E. Harrison
- Subjects
Male ,Vitamin ,medicine.medical_specialty ,Antioxidant ,medicine.medical_treatment ,Ascorbic Acid ,medicine.disease_cause ,Article ,Protein Carbonylation ,Mice ,chemistry.chemical_compound ,Malondialdehyde ,Internal medicine ,Glial Fibrillary Acidic Protein ,medicine ,L-gulonolactone oxidase ,Animals ,Molecular Biology ,Mice, Knockout ,F2-Isoprostanes ,biology ,Vitamin C ,General Neuroscience ,Brain ,Gene Expression Regulation, Developmental ,Glutathione ,Embryo, Mammalian ,Ascorbic acid ,Oxidative Stress ,Endocrinology ,Animals, Newborn ,Liver ,chemistry ,biology.protein ,Female ,Neurology (clinical) ,L-Gulonolactone Oxidase ,Oxidative stress ,Developmental Biology - Abstract
Vitamin C (ascorbic acid, AA) depletion during pre-natal and post-natal development can lead to oxidative stress in the developing brains and other organs. Such damage may lead to irreversible effects on later brain function. We studied the relationship between AA deficiency and oxidative stress during development in gulonolactone oxidase (gulo) knockout mice that are unable to synthesize their own ascorbic acid. Heterozygous gulo(+/−) mice can synthesize AA and typically have similar tissue levels to wild-type mice. Gulo(+/−) dams were mated with gulo(+/−) males to provide offspring of each possible genotype. Overall, embryonic day 20 (E20) and post-natal day 1 (P1) pups were protected against oxidative stress by sufficient AA transfer during pregnancy. On post-natal day 10 (P10) AA levels were dramatically lower in liver and cerebellum in gulo (−/−) mice and malondialdehyde (MDA) levels were significantly increased. In post-natal day 18 pups (P18) AA levels decreased further in gulo(−/−) mice and oxidative stress was observed in the accompanying elevations in MDA in liver, and F2-isoprostanes in cortex. Further, total glutathione levels were higher in gulo(−/−) mice in cortex, cerebellum and liver, indicating that a compensatory antioxidant system was activated. These data show a direct relationship between AA level and oxidative stress in the gulo(−/−) mice. They reinforce the critical role of ascorbic acid in preventing oxidative stress in the developing brain in animals that, like humans, cannot synthesize their own AA.
- Published
- 2010
- Full Text
- View/download PDF
22. Macrophage differentiation increases expression of the ascorbate transporter (SVCT2)
- Author
-
Huan Qiao and James M. May
- Subjects
Transcriptional Activation ,MAPK/ERK pathway ,MAP Kinase Signaling System ,Cellular differentiation ,Organic Anion Transporters, Sodium-Dependent ,Biochemistry ,Article ,chemistry.chemical_compound ,Cell Line, Tumor ,Physiology (medical) ,Humans ,Promoter Regions, Genetic ,Sodium-Coupled Vitamin C Transporters ,Protein Kinase C ,Protein kinase C ,NADPH oxidase ,Symporters ,biology ,MAP kinase kinase kinase ,Macrophages ,NF-kappa B ,NADPH Oxidases ,Cell Differentiation ,MAP Kinase Kinase Kinases ,Molecular biology ,Cell biology ,chemistry ,Mitogen-activated protein kinase ,Monocyte differentiation ,Phorbol ,biology.protein ,Tetradecanoylphorbol Acetate - Abstract
To determine whether macrophage differentiation involves increased uptake of vitamin C, or ascorbic acid, we assessed the expression and function of its transporter SVCT2 during phorbol ester-induced differentiation of human-derived THP-1 monocytes. Induction of THP-1 monocyte differentiation by phorbol 12-myristate 13-acetate (PMA) markedly increased SVCT2 mRNA, protein, and function. When ascorbate was present during PMA-induced differentiation, the increase in SVCT2 protein expression was inhibited, but differentiation was enhanced. PMA-induced SVCT2 protein expression was blocked by inhibitors of protein kinase C (PKC), with most of the affect due to the PKCbetaI and betaII isoforms. Activation of MEK/ERK was sustained up to 48 h after PMA treatment, and the inhibitors completely blocked PMA-stimulated SVCT2 protein expression, indicating an exclusive role for the classical MAP kinase pathway. However, inhibitors of NF-kappaB activation, NADPH oxidase inhibitors, and several antioxidants also partially prevented SVCT2 induction, suggesting diverse distal routes for control of SVCT2 transcription. Both known promoters for the SVCT2 were involved in these effects. In conclusion, PMA-induced monocyte-macrophage differentiation is enhanced by ascorbate and associated with increased expression and function of the SVCT2 protein through a pathway involving sustained activation of PKCbetaI/II, MAP kinase, NADPH oxidase, and NF-kappaB.
- Published
- 2009
- Full Text
- View/download PDF
23. Combined selenium and vitamin C deficiency causes cell death in guinea pig skeletal muscle
- Author
-
Kristina E. Hill, Raymond F. Burk, Amy K. Motley, and James M. May
- Subjects
medicine.medical_specialty ,Nutrition and Dietetics ,Antioxidant ,Vitamin C ,Endocrinology, Diabetes and Metabolism ,Vitamin E ,medicine.medical_treatment ,Skeletal muscle ,Biology ,Creatine ,medicine.disease ,chemistry.chemical_compound ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Selenium deficiency ,Internal medicine ,medicine ,Tocopherol ,alpha-Tocopherol - Abstract
Combined antioxidant deficiencies of selenium and vitamin E or vitamin E and vitamin C in guinea pigs result in clinical illness. We hypothesized that combined selenium and vitamin C deficiency would have clinical consequences because in vitro interactions of these antioxidant nutrients have been reported. Because guinea pigs are dependent on dietary vitamin C, weanling male guinea pigs were fed selenium-deficient or control diet for 15 weeks before imposing vitamin C deficiency. Four dietary groups were formed and studied 3 weeks later: controls, vitamin C deficient, selenium deficient, and doubly deficient. Deficiencies were confirmed by determinations of glutathione peroxidase activity and vitamin C concentration in liver and skeletal muscle. Plasma creatine phosphokinase activity and liver, kidney, heart, and quadriceps histopathology were determined. Doubly deficient animals had moderately severe skeletal muscle cell death as judged by histopathology and plasma creatine phosphokinase activity of 6630 ± 4400 IU/L (control, 70 ± 5; vitamin C deficient, 95 ± 110; selenium deficient, 280 ± 250). Liver, kidney, and heart histology was normal in all groups. Muscle α -tocopherol levels were not depressed in the doubly deficient group, but muscle F 2 isoprostane concentrations were elevated in them and correlated with markers of cell death. We conclude that combining selenium and vitamin C deficiencies in the guinea pig causes cell death in skeletal muscle that is more severe than the injury caused by selenium deficiency. The elevation of muscle F 2 isoprostanes is compatible with the cell death being caused by oxidative stress.
- Published
- 2009
- Full Text
- View/download PDF
24. Development of ascorbate transporters in brain cortical capillary endothelial cells in culture
- Author
-
Huan Qiao and James M. May
- Subjects
Collagen Type IV ,Immunocytochemistry ,Organic Anion Transporters, Sodium-Dependent ,Ascorbic Acid ,Biology ,Blood–brain barrier ,Article ,Mice ,chemistry.chemical_compound ,Type IV collagen ,medicine ,Animals ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Cells, Cultured ,Cerebral Cortex ,HEPES ,Analysis of Variance ,Dose-Response Relationship, Drug ,Symporters ,General Neuroscience ,Endothelial Cells ,Ascorbic acid ,Cell biology ,Mice, Inbred C57BL ,Endothelial stem cell ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Biochemistry ,Cell culture ,Neurology (clinical) ,Intracellular ,Developmental Biology - Abstract
Ascorbic acid in its reduced form is not transported across the capillary endothelial cell blood-brain barrier. This is thought to be due to absence of the SVCT2, a specific transporter for ascorbate. To assess this directly we prepared primary cultures of mouse cortical microvascular endothelial cells. When still in the capillaries, these cells did not express the SVCT2 protein as assessed by immunocytochemistry and by immunoblotting. However, during several days in culture, they developed SVCT2 expression and showed ascorbate transport rates comparable to those in immortalized endothelial cell lines. SVCT2 expression was inversely proportional to cell density, was enhanced by culture at low physiologic plasma ascorbate concentrations, was inhibited by ascorbate concentrations expected in the brain interstitium, and was stimulated by cobalt ions. Expression of the SVCT2 was associated with ascorbate-dependent maturation and release of type IV collagen by the cells in culture. Although the SVCT2 is induced by culture of cortical capillary endothelial cells, its absence in vivo remains perplexing, given the need for intracellular ascorbate to facilitate type IV collagen maturation and release by endothelial cells.
- Published
- 2008
- Full Text
- View/download PDF
25. Maturational loss of the vitamin C transporter in erythrocytes
- Author
-
Zhi-chao Qu, James M. May, Huan Qiao, and Mark J. Koury
- Subjects
Aging ,Erythrocytes ,Organic anion transporter 1 ,Biophysics ,Organic Anion Transporters, Sodium-Dependent ,Cell Enlargement ,Biochemistry ,Article ,Mice ,Reticulocyte ,Erythroblast ,medicine ,Animals ,Humans ,Sodium-Coupled Vitamin C Transporters ,Molecular Biology ,Cells, Cultured ,Symporters ,biology ,Developmental maturation ,Vitamin C ,Cell Biology ,medicine.anatomical_structure ,Symporter ,biology.protein ,Intracellular - Abstract
Erythrocytes have the same intracellular concentration of ascorbate as plasma, which is much lower than that of nucleated cells. To determine why erythrocytes are unable to concentrate ascorbate, we tested for the presence of ascorbate transporters in these cells. Human erythrocytes had very low rates of uptake of radiolabeled ascorbate, which was accounted for by the lack of ascorbate transporter SVCT2 in immunoblots. Using a cell culture model of Friend virus-infected mouse erythroblasts, immunoblots showed that the SVCT2 was present in the erythroblast stages, but was lost following extrusion of the nucleus in the formation of the reticulocyte stage. Rates of specific ascorbate transport correlated with the presence of the SVCT2. These results show that mature erythrocytes fail to concentrate ascorbate due to the loss of SVCT2 during maturation in the bone marrow.
- Published
- 2007
- Full Text
- View/download PDF
26. Ascorbic Acid Prevents VEGF-induced Increases in Endothelial Barrier Permeability
- Author
-
James M. May, Zhi-chao Qu, Esad Ulker, Amita Raj, and William H. Parker
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Sepiapterin ,Clinical Biochemistry ,Ascorbic Acid ,Nitric Oxide ,Article ,Antioxidants ,Permeability ,03 medical and health sciences ,chemistry.chemical_compound ,Enos ,Internal medicine ,medicine ,Human Umbilical Vein Endothelial Cells ,Humans ,Molecular Biology ,Vitamin C ,biology ,business.industry ,Superoxide ,Cell Biology ,General Medicine ,Tetrahydrobiopterin ,biology.organism_classification ,Ascorbic acid ,Vascular endothelial growth factor ,030104 developmental biology ,Endocrinology ,Biochemistry ,chemistry ,Endothelium, Vascular ,business ,Peroxynitrite ,medicine.drug - Abstract
Vascular endothelial growth factor (VEGF) increases endothelial barrier permeability, an effect that may contribute to macular edema in diabetic retinopathy. Since vitamin C, or ascorbic acid, can tighten the endothelial permeability barrier, we examined whether it could prevent the increase in permeability due to VEGF in human umbilical vein endothelial cells (HUVECs). As previously observed, VEGF increased HUVEC permeability to radiolabeled inulin within 60 min in a concentration-dependent manner. Loading the cells with increasing concentrations of ascorbate progressively prevented the leakage caused by 100 ng/ml VEGF, with a significant inhibition at 13 µM and complete inhibition at 50 µM. Loading cells with 100 µM ascorbate also decreased the basal generation of reactive oxygen species and prevented the increase caused by both 100 ng/ml VEGF. VEGF treatment decreased intracellular ascorbate by 25%, thus linking ascorbate oxidation to its prevention of VEGF-induced barrier leakage. The latter was blocked by treating the cells with 60 µM L-NAME (but not D-NAME) as well as by 30 µM sepiapterin, a precursor of tetrahydrobiopterin that is required for proper function of endothelial nitric oxide synthase (eNOS). These findings suggest that VEGF-induced barrier leakage uncouples eNOS. Ascorbate inhibition of the VEGF effect could thus be due either to scavenging superoxide or to peroxynitrite generated by the uncoupled eNOS, or more likely to its ability to recycle tetrahydrobiopterin, thus avoiding enzyme uncoupling in the first place. Ascorbate prevention of VEGF-induced increases in endothelial permeability opens the possibility that its repletion could benefit diabetic macular edema.
- Published
- 2015
27. Recreating blood-brain barrier physiology and structure on chip: A novel neurovascular microfluidic bioreactor
- Author
-
Ronald S. Reiserer, Vanessa Allwardt, Jacquelyn A. Brown, Mingjian Shi, Philip C. Samson, Virginia Pensabene, Deyu Li, John P. Wikswo, M. Diana Neely, Qing Yang, Bryson M. Brewer, James M. May, Orlando S. Hoilett, Clayton M. Britt, Dmitry A. Markov, Lisa J. McCawley, Aaron B. Bowman, and Donna J. Webb
- Subjects
Cell type ,Central nervous system ,Microfluidics ,Biomedical Engineering ,Nanotechnology ,Biology ,Blood–brain barrier ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Colloid and Surface Chemistry ,medicine ,Bioreactor ,General Materials Science ,Fluorescein isothiocyanate ,030304 developmental biology ,Fluid Flow and Transfer Processes ,0303 health sciences ,Human brain ,Condensed Matter Physics ,Neurovascular bundle ,medicine.anatomical_structure ,chemistry ,Biophysics ,030217 neurology & neurosurgery ,Regular Articles - Abstract
The blood-brain barrier (BBB) is a critical structure that serves as the gatekeeper between the central nervous system and the rest of the body. It is the responsibility of the BBB to facilitate the entry of required nutrients into the brain and to exclude potentially harmful compounds; however, this complex structure has remained difficult to model faithfully in vitro. Accurate in vitro models are necessary for understanding how the BBB forms and functions, as well as for evaluating drug and toxin penetration across the barrier. Many previous models have failed to support all the cell types involved in the BBB formation and/or lacked the flow-created shear forces needed for mature tight junction formation. To address these issues and to help establish a more faithful in vitro model of the BBB, we have designed and fabricated a microfluidic device that is comprised of both a vascular chamber and a brain chamber separated by a porous membrane. This design allows for cell-to-cell communication between endothelial cells, astrocytes, and pericytes and independent perfusion of both compartments separated by the membrane. This NeuroVascular Unit (NVU) represents approximately one-millionth of the human brain, and hence, has sufficient cell mass to support a breadth of analytical measurements. The NVU has been validated with both fluorescein isothiocyanate (FITC)-dextran diffusion and transendothelial electrical resistance. The NVU has enabled in vitro modeling of the BBB using all human cell types and sampling effluent from both sides of the barrier.
- Published
- 2015
28. Intracellular Ascorbate Prevents Endothelial Barrier Permeabilization by Thrombin*
- Author
-
William H. Parker, Zhi-chao Qu, and James M. May
- Subjects
rac1 GTP-Binding Protein ,Myosin light-chain kinase ,RHOA ,Cell Membrane Permeability ,Myosin Light Chains ,Intracellular Space ,Ascorbic Acid ,Nitric Oxide ,Biochemistry ,Models, Biological ,Antioxidants ,Nitric oxide ,Polymerization ,Adherens junction ,chemistry.chemical_compound ,Thrombin ,Antigens, CD ,Stress Fibers ,medicine ,Cyclic AMP ,Human Umbilical Vein Endothelial Cells ,Humans ,Phosphorylation ,Molecular Biology ,Cyclic GMP ,biology ,Vitamin C ,Cortical actin cytoskeleton ,rap1 GTP-Binding Proteins ,Cell Biology ,Cadherins ,Actins ,Endocytosis ,Cell biology ,Enzyme Activation ,Intercellular Junctions ,chemistry ,biology.protein ,Calcium ,rhoA GTP-Binding Protein ,Intracellular ,medicine.drug ,Signal Transduction - Abstract
Intracellular ascorbate (vitamin C) has previously been shown to tighten the endothelial barrier and maintain barrier integrity during acute inflammation in vitro. However, the downstream effectors of ascorbate in the regulation of endothelial permeability remain unclear. In this study, we evaluated ascorbate as a mediator of thrombin-induced barrier permeabilization in human umbilical vein endothelial cells and their immortalized hybridoma line, EA.hy926. We found that the vitamin fully prevented increased permeability to the polysaccharide inulin by thrombin in a dose-dependent manner, and it took effect both before and after subjection to thrombin. Thrombin exposure consumed intracellular ascorbate but not the endogenous antioxidant GSH. Likewise, the antioxidants dithiothreitol and tempol did not reverse permeabilization. We identified a novel role for ascorbate in preserving cAMP during thrombin stimulation, resulting in two downstream effects. First, ascorbate maintained the cortical actin cytoskeleton in a Rap1- and Rac1-dependent manner, thus preserving stable adherens junctions between adjacent cells. Second, ascorbate prevented actin polymerization and formation of stress fibers by reducing the activation of RhoA and phosphorylation of myosin light chain. Although ascorbate and thrombin both required calcium for their respective effects, ascorbate did not prevent thrombin permeabilization by obstructing calcium influx. However, preservation of cAMP by ascorbate was found to depend on both the production of nitric oxide by endothelial nitric-oxide synthase, which ascorbate is known to activate, and the subsequent generation cGMP by guanylate cyclase. Together, these data implicate ascorbate in the prevention of inflammatory endothelial barrier permeabilization and explain the underlying signaling mechanism.
- Published
- 2015
29. Ascorbic acid protects SH-SY5Y neuroblastoma cells from apoptosis and death induced by β-amyloid
- Author
-
James M. May and Junjun Huang
- Subjects
Programmed cell death ,Apoptosis ,Ascorbic Acid ,Biology ,Neuroblastoma ,chemistry.chemical_compound ,Annexin ,Cell Line, Tumor ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Reactive oxygen species ,Amyloid beta-Peptides ,Cell Death ,Dose-Response Relationship, Drug ,General Neuroscience ,Phosphatidylserine ,Ascorbic acid ,Cell biology ,Neuroprotective Agents ,Biochemistry ,chemistry ,Cell culture ,Neurology (clinical) ,Intracellular ,Developmental Biology - Abstract
beta-Amyloid causes apoptosis and death in cultured neurons that may be mediated by generation of reactive oxygen species. Since ascorbic acid concentrations are relatively high in brain, we tested whether and how this antioxidant might protect cultured SH-SY5Y neuroblastoma cells from apoptotic cell death. SH-SY5Y cells did not contain ascorbate in culture but readily took it up to achieve intracellular concentrations several-fold those of GSH. Treatment of cells with 2-10 microM beta-amyloid(25-35) decreased both intracellular ascorbate and GSH without affecting rates of ascorbate transport, which suggests that the peptide induces an oxidant stress in the cells. Overnight culture of cells with 10-20 microM beta-amyloid(25-35) induced apoptosis in SH-SY5Y cells when measured as externalization of phosphatidylserine by annexin V binding, as DNA fragmentation in the TUNEL assay, and as caspase-3 activity in cell lysates. Pre-loading cells with ascorbate substantially prevented apoptosis measured by these assays as well as cell death. In addition to preventing apoptosis, ascorbate loading of SH-SY5Y cells also decreased basal rates of generation of endogenous beta-amyloid. Together, these results support the notion that beta-amyloid induces apoptosis and death in neurons due to oxidant stress and suggest that intracellular ascorbate effectively prevents this toxicity.
- Published
- 2006
- Full Text
- View/download PDF
30. Ascorbate uptake and antioxidant function in peritoneal macrophages
- Author
-
Liying Li, James M. May, Zhi-chao Qu, and Junjun Huang
- Subjects
Time Factors ,Necrosis ,Antioxidant ,Phagocytosis ,medicine.medical_treatment ,alpha-Tocopherol ,Biophysics ,Apoptosis ,Ascorbic Acid ,Biology ,medicine.disease_cause ,Biochemistry ,Antioxidants ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,chemistry.chemical_classification ,Reactive oxygen species ,Cell Membrane ,Glutathione ,Fluoresceins ,Ascorbic acid ,Dehydroascorbic Acid ,Cell biology ,Oxidative Stress ,chemistry ,Astrocytes ,Thioglycolates ,Macrophages, Peritoneal ,Dehydroascorbic acid ,medicine.symptom ,Reactive Oxygen Species ,Oxidative stress - Abstract
Since activated macrophages generate potentially deleterious reactive oxygen species, we studied whether ascorbic acid might function as an antioxidant in these cells. Thioglycollate-elicited murine peritoneal macrophages contained about 3 mM ascorbate that was halved by culture in ascorbate-free medium. However, the cells took up added ascorbate to concentrations of 6-8 mM by a high-affinity sodium-dependent transport mechanism. This likely reflected the activity of the SVCT2 ascorbate transporter, since its message and protein were present in the cells. Activation of the cells by phagocytosis of latex particles depleted intracellular ascorbate, although not below the basal levels present in the cells in culture. Glutathione (GSH) was unaffected by phagocytosis, suggesting that ascorbate was more sensitive to the oxidant stress of phagocytosis than GSH. Phagocytosis induced a modest increase in reactive oxygen species as well as a progressive loss of alpha-tocopherol, both of which were prevented in cells loaded with ascorbate. These results suggest that activated macrophages can use ascorbate to lessen self-generated oxidant stress and spare alpha-tocopherol, which may protect these long-lived cells from necrosis or apoptosis.
- Published
- 2005
- Full Text
- View/download PDF
31. Combined deficiency of vitamins E and C causes paralysis and death in guinea pigs
- Author
-
Amy K. Motley, Kristina E. Hill, James M. May, Xia Li, Thomas J. Montine, and Raymond F. Burk
- Subjects
Male ,Vitamin ,medicine.medical_treatment ,Guinea Pigs ,alpha-Tocopherol ,Medicine (miscellaneous) ,Physiology ,Caviidae ,Ascorbic Acid ,Biology ,Guinea pig ,Lesion ,chemistry.chemical_compound ,Paralysis ,medicine ,Animals ,Vitamin E Deficiency ,Ascorbic Acid Deficiency ,Nutrition and Dietetics ,Vitamin C ,Vitamin E ,Osmolar Concentration ,Brain ,biology.organism_classification ,Sciatic Nerve ,Disease Models, Animal ,Spinal Cord ,chemistry ,Immunology ,medicine.symptom - Abstract
Background On the basis of in vitro studies, the antioxidant nutrients vitamins E and C are postulated to interact in vivo. Objective We developed a guinea pig model to evaluate the combined deficiency of vitamins E and C in vivo. Design Weanling guinea pigs were fed a control diet or a vitamin E-deficient diet for 14 d, after which one-half of each group had vitamin C removed from their diet, thus creating 4 diet groups. Some animals were observed for clinical signs. Others were killed for evaluation. Results Of 21 guinea pigs that were observed after being fed the diet deficient in both vitamins, 8 died 9 +/- 2 d (x +/- SD) after starting the diet. Eight additional guinea pigs developed a characteristic syndrome at 11 +/- 3 d. First, they became paralyzed in the hind limbs. Within a few hours, the paralysis progressed to include all 4 limbs and caused difficulty in breathing, which would have caused death had the animals not been euthanized. Histopathologic evaluation did not identify a lesion in the muscles or nervous system that could account for the paralysis. Biochemical measurements confirmed the deficiencies and indicated that the double deficiency caused lipid peroxidation in the central nervous system. Conclusions A distinct clinical syndrome of combined vitamin E and vitamin C deficiency occurs in guinea pigs. This syndrome indicates that these antioxidant vitamins are related in vivo. We speculate that acute oxidative injury in the central nervous system underlies the clinical syndrome.
- Published
- 2003
- Full Text
- View/download PDF
32. Catalase-dependent measurement of H2O2 in intact mitochondria
- Author
-
Xia Li and James M. May
- Subjects
inorganic chemicals ,biology ,Superoxide ,Endogeny ,Cell Biology ,Rat heart ,Mitochondrion ,Electron transport chain ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Catalase ,Organelle ,biology.protein ,Molecular Medicine ,Hydrogen peroxide ,Molecular Biology - Abstract
Mitochondria generate potentially damaging amounts of superoxide and H2O2 during oxidative metabolism. Although many assays are available to measure mitochondrial H2O2 generation, most detect H2O2 that has escaped the organelle. To measure H2O2 within mitochondria that contain catalase, we have developed an assay based on the ability of H2O2 to inhibit catalase in the presence of 3-amino-1,2,4-triazole. The assay is simple to perform, does not require expensive instrumentation, and is specific for H2O2. Results from this assay show that H2O2 generation in rat heart mitochondria reflects the activity of the electron transport chain. Further, liver mitochondria prepared from selenium-deficient rats have increased succinate-stimulated rates of H2O2 generation. This indicates that mitochondrial selenoenzymes are important for H2O2 removal. It also demonstrates the utility of this assay in measuring H2O2 release from mitochondria that do not contain catalase. The assay should be useful for study of both superoxide-dependent H2O2 generation in situ, and the role of endogenous mitochondrial catalase in H2O2 removal.
- Published
- 2002
- Full Text
- View/download PDF
33. Thioredoxin Reductase Reduces Lipid Hydroperoxides and Spares α-Tocopherol
- Author
-
James M. May, Jason D. Morrow, and Raymond F. Burk
- Subjects
inorganic chemicals ,Lipid Peroxides ,Thioredoxin-Disulfide Reductase ,Thioredoxin reductase ,Lipoxygenase ,alpha-Tocopherol ,Biophysics ,chemistry.chemical_element ,Biochemistry ,Lipid peroxidation ,chemistry.chemical_compound ,Glutaredoxin ,Animals ,Humans ,Tocopherol ,Ferricyanides ,Molecular Biology ,integumentary system ,biology ,Erythrocyte Membrane ,Cell Biology ,Rats ,Kinetics ,Liver ,chemistry ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Lipid Peroxidation ,Ferricyanide ,Thioredoxin ,Oxidation-Reduction ,Selenium - Abstract
We investigated whether and how rat liver thioredoxin reductase spares alpha-tocopherol in biomembranes. Purified hydroperoxides of beta-linoleoyl-gamma-palmitoylphosphatidylcholine were decreased 35% by treatment with thioredoxin reductase and 54% by thioredoxin reductase plus E. coli thioredoxin. Thioredoxin reductase also halved the amount of hydroperoxides that had been formed during photoperoxidation of liposomes composed of beta-linoleoyl-gamma-palmitoylphosphatidylcholine, and of emulsions of both cholesterol and cholesteryl linolenate. In erythrocyte ghosts, thioredoxin reductase spared alpha-tocopherol from oxidation by both soybean lipoxygenase and ferricyanide. Thioredoxin reductase also decreased F(2)-isoprostanes in ghosts oxidized by ferricyanide, suggesting that its ability to spare alpha-tocopherol relates to reduction of lipid hydroperoxides.
- Published
- 2002
- Full Text
- View/download PDF
34. Coordinate regulation of L-arginine uptake and nitric oxide synthase activity in cultured endothelial cells
- Author
-
James M. May and Thomas A. Hardy
- Subjects
medicine.medical_specialty ,Nitric Oxide Synthase Type III ,Arginine ,Caveolae ,Biochemistry ,Nitric oxide ,chemistry.chemical_compound ,Enos ,Physiology (medical) ,Internal medicine ,medicine ,Extracellular ,Citrulline ,Animals ,Aorta ,Calcimycin ,Cells, Cultured ,Ionophores ,biology ,Biological Transport ,biology.organism_classification ,Cell biology ,Nitric oxide synthase ,Kinetics ,Endocrinology ,chemistry ,biology.protein ,Cattle ,Endothelium, Vascular ,Nitric Oxide Synthase ,Extracellular Space ,Intracellular - Abstract
Despite intracellular L-arginine concentrations that should saturate endothelial nitric oxide synthase (eNOS), nitric oxide production depends on extracellular L-arginine. We addressed this 'arginine paradox' in bovine aortic endothelial cells by simultaneously comparing the substrate dependence of L-arginine uptake and intracellular eNOS activity, the latter measured as L-[3H]arginine conversion to L-[3H]citrulline. Whereas the Km of eNOS for L-arginine was 2 microM in cell extracts, the L-arginine concentration of half-maximal eNOS stimulation was increased to 29 microM in intact cells. This increase likely reflects limitation by L-arginine uptake, which had a Km of 108 microM. The effects of inhibitors of endothelial nitric oxide synthesis also suggested that extracellular L-arginine availability limits intracellular eNOS activity. Treatment of intact cells with the calcium ionophore A23187 reduced the L-arginine concentration of half-maximal eNOS activity, which is consistent with a measured increase in L-arginine uptake. Increases in eNOS activity induced by several agents were closely correlated with enhanced L-arginine uptake into cells (r = 0.89). The 'arginine paradox' may be explained in part by regulated L-arginine uptake into a compartment, probably represented by caveolae, that contains eNOS and that is distinct from the bulk cytosolic L-arginine.
- Published
- 2002
- Full Text
- View/download PDF
35. Macrophage deficiency of Akt2 reduces atherosclerosis in Ldlr null mice
- Author
-
Vladimir R. Babaev, Lei Ding, Carrie B. Wiese, Katie E. Hebron, Cynthia L. Toth, Kasey C. Vickers, MacRae F. Linton, Sergio Fazio, James M. May, and Youmin Zhang
- Subjects
Male ,Chemokine ,Receptors, CCR2 ,AKT1 ,AKT2 ,QD415-436 ,Biochemistry ,AKT3 ,Monocytes ,Proinflammatory cytokine ,Gene Knockout Techniques ,Mice ,Endocrinology ,Cell Movement ,Animals ,Antigens, Ly ,cell signaling ,Receptor ,Protein kinase B ,Research Articles ,Mice, Knockout ,biology ,Macrophages ,apoptosis ,Cell Biology ,Atherosclerosis ,Molecular biology ,foam cells ,Hematopoiesis ,macrophages/monocytes ,Mice, Inbred C57BL ,Phenotype ,Gene Expression Regulation ,Receptors, LDL ,LDL receptor ,embryonic structures ,Cancer research ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Female ,Proto-Oncogene Proteins c-akt ,hormones, hormone substitutes, and hormone antagonists - Abstract
Macrophages play crucial roles in the formation of atherosclerotic lesions. Akt, a serine/threonine protein kinase B, is vital for cell proliferation, migration, and survival. Macrophages express three Akt isoforms, Akt1, Akt2, and Akt3, but the roles of Akt1 and Akt2 in atherosclerosis in vivo remain unclear. To dissect the impact of macrophage Akt1 and Akt2 on early atherosclerosis, we generated mice with hematopoietic deficiency of Akt1 or Akt2. After 8 weeks on Western diet, Ldlr(-/-) mice reconstituted with Akt1(-/-) fetal liver cells (Akt1(-/-)→Ldlr(-/-)) had similar atherosclerotic lesion areas compared with control mice transplanted with WT cells (WT→Ldlr(-/-)). In contrast, Akt2(-/-)→Ldlr(-/-) mice had dramatically reduced atherosclerotic lesions compared with WT→Ldlr(-/-) mice of both genders. Similarly, in the setting of advanced atherosclerotic lesions, Akt2(-/-)→Ldlr(-/-) mice had smaller aortic lesions compared with WT→Ldlr(-/-) and Akt1(-/-)→Ldlr(-/-) mice. Importantly, Akt2(-/-)→Ldlr(-/-) mice had reduced numbers of proinflammatory blood monocytes expressing Ly-6C(hi) and chemokine C-C motif receptor 2. Peritoneal macrophages isolated from Akt2(-/-) mice were skewed toward an M2 phenotype and showed decreased expression of proinflammatory genes and reduced cell migration. Our data demonstrate that loss of Akt2 suppresses the ability of macrophages to undergo M1 polarization reducing both early and advanced atherosclerosis.
- Published
- 2014
36. Ascorbic acid prevents high glucose-induced apoptosis in human brain pericytes
- Author
-
William H. Parker, Zhi-chao Qu, Ashwath Jayagopal, and James M. May
- Subjects
medicine.medical_specialty ,Biophysics ,Caspase 3 ,Apoptosis ,Ascorbic Acid ,Biology ,medicine.disease_cause ,Biochemistry ,Article ,RAGE (receptor) ,Annexin ,Internal medicine ,medicine ,Humans ,Molecular Biology ,Cells, Cultured ,Vitamin C ,Cell Biology ,Ascorbic acid ,medicine.anatomical_structure ,Endocrinology ,Glucose ,cardiovascular system ,Pericyte ,Pericytes ,Oxidative stress - Abstract
High glucose concentrations due to diabetes increase apoptosis of vascular pericytes, impairing vascular regulation and weakening vessels, especially in brain and retina. We sought to determine whether vitamin C, or ascorbic acid, could prevent such high glucose-induced increases in pericyte apoptosis. Culture of human microvascular brain pericytes at 25 mM compared to 5 mM glucose increased apoptosis measured as the appearance of cleaved caspase 3. Loading the cells with ascorbate during culture decreased apoptosis, both at 5 and 25 mM glucose. High glucose-induced apoptosis was due largely to activation of the receptor for advanced glycation end products (RAGE), since it was prevented by specific RAGE inhibition. Culture of pericytes for 24 hours with RAGE agonists also increased apoptosis, which was completely prevented by inclusion of 100 μM ascorbate. Ascorbate also prevented RAGE agonist-induced apoptosis measured as annexin V binding in human retinal pericytes, a cell type with relevance to diabetic retinopathy. RAGE agonists decreased intracellular ascorbate and GSH in brain pericytes. Despite this evidence of increased oxidative stress, ascorbate prevention of RAGE-induced apoptosis was not mimicked by several antioxidants. These results show that ascorbate prevents pericyte apoptosis due RAGE activation. Although RAGE activation decreases intracellular ascorbate and GSH, the prevention of apoptosis by ascorbate may involve effects beyond its function as an antioxidant.
- Published
- 2014
37. GSH Is Required to Recycle Ascorbic Acid in Cultured Liver Cell Lines
- Author
-
James M. May, Zhi-chao Qu, and Xia Li
- Subjects
Thioredoxin-Disulfide Reductase ,Time Factors ,Physiology ,Liver cytology ,Clinical Biochemistry ,Ascorbic Acid ,Biology ,Biochemistry ,Arsenicals ,Cell Line ,chemistry.chemical_compound ,Animals ,Humans ,Buthionine sulfoximine ,Phenylarsine oxide ,Enzyme Inhibitors ,Buthionine Sulfoximine ,Molecular Biology ,Cells, Cultured ,General Environmental Science ,Dose-Response Relationship, Drug ,Liver cell ,Maleates ,Water ,Cell Biology ,Glutathione ,Ascorbic acid ,Rats ,Kinetics ,Liver ,chemistry ,Cell culture ,General Earth and Planetary Sciences ,Dehydroascorbic acid ,NADP - Abstract
Liver is the site of ascorbic acid synthesis in most mammals. As human liver cannot synthesize ascorbate de novo, it may differ from liver of other species in the capacity or mechanism for ascorbate recycling from its oxidized forms. Therefore, we compared the ability of cultured liver-derived cells from humans (HepG2 cells) and rats (H4IIE cells) to take up and reduce dehydroascorbic acid (DHA) to ascorbate. Neither cell type contained appreciable amounts of ascorbate in culture, but both rapidly took up and reduced DHA to ascorbate. Intracellular ascorbate accumulated to concentrations of 10-20 mM following loading with DHA. The capacity of HepG2 cells to take up and reduce DHA to ascorbate was more than twice that of H4IIE cells. In both cell types, DHA reduction lowered glutathione (GSH) concentrations and was inhibited by prior depletion of GSH with diethyl maleate, buthionine sulfoximine, and phenylarsine oxide. NADPH-dependent DHA reduction due to thioredoxin reductase occurred in overnight-dialyzed extracts of both cell types. These results show that cells derived from rat liver synthesize little ascorbate in culture, that cultured human-derived liver cells have a greater capacity for DHA reduction than do rat-derived liver cells, but that both cell types rely largely on GSH- or NADPH-dependent mechanisms for ascorbate recycling from DHA.
- Published
- 2001
- Full Text
- View/download PDF
38. Glucose uptake and metabolism by cultured human skeletal muscle cells: rate-limiting steps
- Author
-
James M. May, David W. Piston, Alvin C. Powers, Daryl K. Granner, Tetsuro Kono, Richard R. Whitesell, Susan M. Knobel, and Laureta M. Perriott
- Subjects
Adult ,Male ,medicine.medical_specialty ,Snf3 ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Glucose uptake ,Biological Transport, Active ,3-O-Methylglucose ,Biology ,Carbohydrate metabolism ,chemistry.chemical_compound ,Insulin resistance ,Hexokinase ,Physiology (medical) ,Internal medicine ,Pyruvic Acid ,medicine ,Humans ,Hypoglycemic Agents ,Insulin ,Phosphorylation ,Muscle, Skeletal ,Cells, Cultured ,Aged ,Glucose transporter ,Cell Differentiation ,Middle Aged ,medicine.disease ,Stimulation, Chemical ,Kinetics ,Glucose ,Endocrinology ,Microscopy, Fluorescence ,Biochemistry ,chemistry ,Female ,NADP - Abstract
To use primary cultures of human skeletal muscle cells to establish defects in glucose metabolism that underlie clinical insulin resistance, it is necessary to define the rate-determining steps in glucose metabolism and to improve the insulin response attained in previous studies. We modified experimental conditions to achieve an insulin effect on 3- O-methylglucose transport that was more than twofold over basal. Glucose phosphorylation by hexokinase limits glucose metabolism in these cells, because the apparent Michaelis-Menten constant of coupled glucose transport and phosphorylation is intermediate between that of transport and that of the hexokinase and because rates of 2-deoxyglucose uptake and phosphorylation are less than those of glucose. The latter reflects a preference of hexokinase for glucose over 2-deoxyglucose. Cellular NAD(P)H autofluorescence, measured using two-photon excitation microscopy, is both sensitive to insulin and indicative of additional distal control steps in glucose metabolism. Whereas the predominant effect of insulin in human skeletal muscle cells is to enhance glucose transport, phosphorylation, and steps beyond, it also determines the overall rate of glucose metabolism.
- Published
- 2001
- Full Text
- View/download PDF
39. How does ascorbic acid prevent endothelial dysfunction?
- Author
-
James M. May
- Subjects
medicine.medical_specialty ,Antioxidant ,Nitric Oxide Synthase Type III ,Endothelium ,Arteriosclerosis ,medicine.medical_treatment ,Vasodilation ,Ascorbic Acid ,Nitric Oxide ,Biochemistry ,Antioxidants ,Nitric oxide ,chemistry.chemical_compound ,Superoxides ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Humans ,Sulfhydryl Compounds ,Endothelial dysfunction ,Nitrites ,Mercaptoethanol ,S-Nitrosothiols ,biology ,Vitamin C ,medicine.disease ,Ascorbic acid ,Coronary Vessels ,Enzyme Activation ,Lipoproteins, LDL ,Nitric oxide synthase ,Oxidative Stress ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Guanylate Cyclase ,biology.protein ,Endothelium, Vascular ,Rabbits ,Nitric Oxide Synthase ,Oxidation-Reduction ,Nitroso Compounds - 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.
- Published
- 2000
- Full Text
- View/download PDF
40. Extracellular Reduction of the Ascorbate Free Radical by Human Erythrocytes
- Author
-
Charles E. Cobb, James M. May, and Zhi-chao Qu
- Subjects
Vitamin ,Electron paramagnetic resonance spectroscopy ,Erythrocytes ,Free Radicals ,Biophysics ,Ascorbate Oxidase ,Ascorbic Acid ,In Vitro Techniques ,Biochemistry ,chemistry.chemical_compound ,Extracellular ,Humans ,Ferricyanides ,Molecular Biology ,biology ,Electron Spin Resonance Spectroscopy ,Cell Biology ,Cell concentration ,Dehydroascorbic Acid ,Glutathione ,Enzyme assay ,Kinetics ,chemistry ,biology.protein ,Human erythrocytes ,Oxidation-Reduction - Abstract
We investigated the possibility that human erythrocytes can reduce extracellular ascorbate free radical (AFR). When the AFR was generated from ascorbate by ascorbate oxidase, intact cells slowed the loss of extracellular ascorbate, an effect that could not be explained by changes in enzyme activity or by release of ascorbate from the cells. If cells preserve extracellular ascorbate by regenerating it from the AFR, then they should decrease the steady-state concentration of the AFR. This was confirmed directly by electron paramagnetic resonance spectroscopy, in which the steady-state extracellular AFR signal varied inversely with the cell concentration and was a saturable function of the absolute AFR concentration. Treatment of cells N-ethylmaleimide (2 mM) impaired their ability both to preserve extracellular ascorbate, and to decrease the extracellular AFR concentration. These results suggest that erythrocytes spare extracellular ascorbate by enhancing recycling of the AFR, which could help to maintain extracellular concentrations of the vitamin.
- Published
- 2000
- Full Text
- View/download PDF
41. Uptake and antioxidant effects of ergothioneine in human erythrocytes
- Author
-
James M. May and Hiroki Mitsuyama
- Subjects
Antioxidant ,biology ,medicine.medical_treatment ,General Medicine ,Glutathione ,Metabolism ,chemistry.chemical_compound ,Red blood cell ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Catalase ,Ergothioneine ,medicine ,biology.protein ,Hydrogen peroxide ,Incubation - Abstract
Ergothioneine is a fungal metabolite that may have antioxidant functions in mammalian cells. Although it accumulates to low millimolar concentrations in liver and other tissues, it is not thought to be taken up by mature erythrocytes. During a study of the function of ergothioneine as an antioxidant in human erythrocytes, we found that these cells do take up ergothioneine from the surrounding medium. Ergothioneine concentrations in freshly prepared erythrocytes were 2–9-fold higher than in plasma from the same donor. Slow but progressive accumulation of ergothioneine to about 125% of basal levels was observed in erythrocytes over a 4 h incubation. After a 2 h incubation, intracellular ergothioneine concentrations rose on addition of increasing amounts of ergothioneine to the incubation medium, although saturation was not evident in cells from all donors. Both initial levels and rates of ergothioneine uptake varied in erythrocytes from different donors. Intracellular ergothioneine was stable to depletion of GSH by N-ethylmaleimide and to a more severe oxidant stress induced by hydrogen peroxide in the presence of catalase. These results show that human erythrocytes do take up ergothioneine; however, the GSH results do not support an antioxidant role for ergothioneine in erythrocytes.
- Published
- 1999
- Full Text
- View/download PDF
42. Functional Interaction between the N- and C-terminal Halves of Human Hexokinase II
- Author
-
Richard L. Printz, Daryl K. Granner, Hossein Ardehali, James M. May, and Richard R. Whitesell
- Subjects
Protein Conformation ,Recombinant Fusion Proteins ,Glucose-6-Phosphate ,Context (language use) ,Biology ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Protein structure ,Catalytic Domain ,Hexokinase ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Cell Biology ,Fusion protein ,Molecular biology ,Amino acid ,Molecular Weight ,Glucose binding ,Kinetics ,Glucose ,Enzyme ,chemistry ,Glucose 6-phosphate ,Mutagenesis, Site-Directed - Abstract
Mammalian hexokinases (HKs) I-III are composed of two highly homologous approximately 50-kDa halves. Studies of HKI indicate that the C-terminal half of the molecule is active and is sensitive to inhibition by glucose 6-phosphate (G6P), whereas the N-terminal half binds G6P but is devoid of catalytic activity. In contrast, both the N- and C-terminal halves of HKII (N-HKII and C-HKII, respectively) are catalytically active, and when expressed as discrete proteins both are inhibited by G6P. However, C-HKII has a significantly higher Ki for G6P (KiG6P) than N-HKII. We here address the question of whether the high KiG6P of the C-terminal half (C-half) of HKII is decreased by interaction with the N-terminal half (N-half) in the context of the intact enzyme. A chimeric protein consisting of the N-half of HKI and the C-half of HKII was prepared. Because the N-half of HKI is unable to phosphorylate glucose, the catalytic activity of this chimeric enzyme depends entirely on the C-HKII component. The KiG6P of this chimeric enzyme is similar to that of HKI and is significantly lower than that of C-HKII. When a conserved amino acid (Asp209) required for glucose binding is mutated in the N-half of this chimeric protein, a significantly higher KiG6P (similar to that of C-HKII) is observed. However, mutation of a second conserved amino acid (Ser155), also involved in catalysis but not required for glucose binding, does not increase the KiG6P of the chimeric enzyme. This resembles the behavior of HKII, in which a D209A mutation results in an increase in the KiG6P of the enzyme, whereas a S155A mutation does not. These results suggest an interaction in which glucose binding by the N-half causes the activity of the C-half to be regulated by significantly lower concentrations of G6P.
- Published
- 1999
- Full Text
- View/download PDF
43. Inhibition of aldose reductase in human erythrocytes by vitamin C
- Author
-
Shalu Mendiratta, Travis E. Vincent, and James M. May
- Subjects
Erythrocytes ,Endocrinology, Diabetes and Metabolism ,Ascorbic Acid ,Hemolysis ,chemistry.chemical_compound ,Endocrinology ,Aldehyde Reductase ,Internal Medicine ,Humans ,Sorbitol ,Medicine ,Enzyme Inhibitors ,Aldose reductase ,Vitamin C ,biology ,business.industry ,Intracellular Membranes ,General Medicine ,Glutathione ,Ascorbic acid ,Dehydroascorbic Acid ,Aldose reductase inhibitor ,Glucose ,Biochemistry ,chemistry ,Enzyme inhibitor ,biology.protein ,business ,NADP ,medicine.drug - Abstract
Ascorbic acid, or vitamin C, has been reported to lower erythrocyte sorbitol concentrations, and present studies were performed to determine the mechanism of this effect. Incubation of erythrocytes with increasing concentrations of glucose (5-40 mM) progressively increased erythrocyte sorbitol contents, reflecting increased flux through aldose reductase. At extracellular concentrations of 90 microM, both ascorbic acid and its oxidized form, dehydroascorbate, decreased intracellular sorbitol by 25 and 45%, respectively. This inhibition was not dependent on the extracellular glucose concentration, or on erythrocyte contents of free NADPH or GSH. To test for a direct effect of ascorbate on aldose reductase, erythrocyte hemolysates were prepared and supplemented with 100 microM NADPH. Hemolysates reduced glucose to sorbitol in a dose-dependent manner that was inhibited with a Ki of 120 microM by the aldose reductase inhibitor tetramethylene glutaric acid. Above 100 microM, ascorbic acid also lowered hemolysate sorbitol generation by about 30%. Studies with ascorbic acid derivatives showed that the reducing capacity of ascorbic acid was not required for inhibition of sorbitol production from glucose in erythrocyte hemolysates. These results show that high, but physiologic, concentrations of ascorbic acid can directly inhibit erythrocyte aldose reductase, and provide a rationale for the use of oral vitamin C supplements in diabetes.
- Published
- 1999
- Full Text
- View/download PDF
44. Famine From Feast: Low Red Cell Vitamin C Levels in Diabetes
- Author
-
James M. May
- Subjects
Vitamin ,medicine.medical_specialty ,lcsh:R5-920 ,Antioxidant ,Vitamin C ,medicine.medical_treatment ,lcsh:R ,Erythrocyte fragility ,Glucose transporter ,lcsh:Medicine ,General Medicine ,Biology ,medicine.disease ,Ascorbic acid ,General Biochemistry, Genetics and Molecular Biology ,Vitamin A deficiency ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Erythrocyte deformability ,lcsh:Medicine (General) - Abstract
Decreased oxygen delivery to tissues is an important downstream consequence of diabetic microvascular disease. Although endothelial dysfunction causing abnormal blood flow regulation is often considered the major factor in this regard, impaired oxygen delivery by erythrocytes may also play a role in both vascular and tissue damage. Erythrocytes in persons with diabetes with poor glycemic control have impaired deformability, leading to increased fragility and hemolysis (McMillan et al. 1978). Beyond anemia as a cause of poor oxygen delivery, decreased erythrocyte deformability in diabetes may also compromise capillary perfusion and thus impair delivery of oxygen to the tissues. The studies of Tu et al. (2015) in this issue of EBioMedicine add another potential factor to the abnormal red cell function in diabetes: high glucose concentrations cause erythrocyte vitamin C deficiency, which may then amplifies defects in erythrocyte structure and function due to diabetes alone. This work was prompted by the astute “clinical” observation that mice unable to synthesize their own vitamin C, when placed on a vitamin C-deficient diet for 12 weeks, showed hemolysis. This was associated with increased osmotic fragility and conversion of many cells from biconcave discs to rounded spherocyte forms. The latter are less deformable and more prone to hemolysis than are normal erythrocytes (Waugh and Sarelius, 1996). Evaluation by gel electrophoresis of erythrocyte cytoskeletal proteins showed that vitamin C deficiency caused loss of more than 50% of β-spectrin, without changes in other cytoskeletal proteins. Both the decrease in β-spectrin and the osmotic fragility were partially reversed after 10 days of ascorbate repletion. Since β-spectrin is required for normal cytoskeletal structure and function, it seems likely that its loss or damage accounted for the observed shape changes and sensitivity to hemolysis. This brings up the question of how ascorbate deficiency caused loss of β-spectrin. Although this was not investigated in the studies of Tu et al. (2015), it could relate to the previous observation that susceptible amino acid residues (e.g., cysteines, histidines, methionines) in β-spectrin are more readily oxidized than those of other erythrocyte cytoskeletal proteins, even to the point of undergoing non-enzymatic cleavage of peptide bonds (Arduini et al. 1989). This would suggest that the antioxidant function of vitamin C is necessary to preserve β-spectrin and thus cytoskeletal integrity in erythrocytes. Although vitamin C deficiency to the point of scurvy is no longer common in developed countries, it is well established that persons with diabetes in poor glycemic control have low plasma and leukocyte vitamin C levels, measured as the fully reduced form, ascorbate (Cunningham et al., 1991, Ginter et al., 1978). Thus, it was logical for Tu et al. (2015) to assess the impact of hyperglycemia and diabetes on erythrocyte vitamin C. Indeed, they found that the ascorbate content of erythrocytes from diabetic subjects varied inversely with the degree of hyperglycemia to which they were exposed. This relationship was not due to changes in plasma ascorbate levels, but rather to the fact that erythrocytes from both mice and humans take up vitamin C as the two-electron oxidized form of the vitamin, dehydroascorbate. This occurs by facilitated diffusion on GLUT-type glucose transporters (Vera et al. 1993). High but still physiologic extracellular glucose levels compete with even very low concentrations of dehydroascorbate for uptake and this decreases intracellular dehydroascorbate available for conversion to ascorbate. Most cells take up ascorbate directly on one of the two isoforms of the Sodium-dependent Vitamin C Transporter, SVCT. However, mature erythrocytes lack this transporter (May et al. 2007). The ability of glucose to impair dehydroascorbate uptake into erythrocytes makes them unique in that they may be prone to significant intracellular vitamin C deficiency in diabetes. Perhaps most important, erythrocyte ascorbate deficiency due to diabetic hyperglycemia may accentuate or even account for known abnormalities in diabetic erythrocytes. It was shown over 2 decades ago that β-spectrin in erythrocytes from both type 1 and type 2 diabetics is oxidized and associated with decreased deformability (Schwartz et al. 1991). Although β-spectrin was not found to be decreased in diabetic erythrocytes in that study, quantification by normalization to β-actin was not carried out. The finding in the study by Tu et al. (2015) that sensitivity to hemolysis was inversely correlated with erythrocyte ascorbate content and more pronounced in diabetic subjects with poor glycemic control suggests that deficient intracellular ascorbate could have played a causative role in the changes in diabetic erythrocytes. This is supported by the parallel observation that erythrocyte β-spectrin (but not α-spectrin) was also decreased in proportion to the hemoglobin A1C, a measure of glycemic control. There are implications from this study by Tu et al. (2015). First the erythrocyte, because of its unique glucose-sensitive mechanism of vitamin C uptake, may be more susceptible to intracellular ascorbate deficiency than other cell types in which ascorbate is taken up directly. Second, erythrocyte ascorbate deficiency decreases cytoskeletal β-spectrin, which in turn likely accounts for decreased deformability and susceptibility to hemolysis. Third, the hyperglycemia of diabetes decreases erythrocyte ascorbate, deficiency of which could contribute to decreased erythrocyte fragility in poorly controlled diabetes. The work opens up several areas for future studies, such as whether the loss of β-spectrin due to ascorbate deficiency is due to oxidative damage, whether it contributes to the observed increased osmotic fragility of erythrocytes deficient in ascorbate, and most important, whether erythrocyte ascorbate deficiency accounts for increased deformability and lysis seen in diabetes. If so, then this study could prompt further clinical studies of the impact of vitamin C supplements on erythrocyte fragility in diabetes.
- Published
- 2015
- Full Text
- View/download PDF
45. Erythrocyte defenses against hydrogen peroxide: the role of ascorbic acid
- Author
-
James M. May, Shalu Mendiratta, and Zhi-chao Qu
- Subjects
inorganic chemicals ,Erythrocytes ,biology ,Biophysics ,Context (language use) ,Ascorbic Acid ,Hydrogen Peroxide ,Glutathione ,Catalase ,Ascorbic acid ,Biochemistry ,Enzyme Activation ,chemistry.chemical_compound ,chemistry ,Extracellular ,biology.protein ,Humans ,Dehydroascorbic acid ,Hydrogen peroxide ,Molecular Biology ,Intracellular - Abstract
Ascorbate has been reported to increase intracellular hydrogen peroxide (H2O2) generation in human erythrocytes. In the present work, the basis for this prooxidant effect of the vitamin was investigated in the context of erythrocyte defenses against H2O2. Ascorbate added to erythrocytes caused a dose-dependent increase in intracellular H2O2, which was measured as inactivation of endogenous catalase in the presence of 3-amino-1,2,4-triazole (aminotriazole). Ascorbate-induced catalase inactivation was not observed when only the intracellular ascorbate concentration was increased, when cells were incubated with ascorbate in plasma, or when extracellular Fe3+ was chelated. Together, these results suggest that the observed ascorbate-induced H2O2 generation is due to Fe3+-catalyzed oxidation of extracellular, as opposed to intracellular, ascorbate by molecular oxygen. Rather than generate an oxidant stress in erythrocytes, ascorbate was one of the most sensitive intracellular antioxidants to H2O2 coming from outside the cells. On the other hand, intracellular ascorbate contributed little to the detoxification of H2O2, which was found to be mediated by both catalase and by the GSH system.
- Published
- 1998
- Full Text
- View/download PDF
46. Role of Vitamin C in the Function of the Vascular Endothelium
- Author
-
James M. May and Fiona E. Harrison
- Subjects
Endothelium ,Physiology ,Clinical Biochemistry ,Vascular permeability ,Apoptosis ,Ascorbic Acid ,Biology ,Biochemistry ,Antioxidants ,Nitric oxide ,Capillary Permeability ,chemistry.chemical_compound ,Type IV collagen ,medicine ,Animals ,Humans ,Endothelial dysfunction ,Phosphorylation ,Molecular Biology ,General Environmental Science ,Cell Proliferation ,Basement membrane ,Vitamin C ,Endothelial Cells ,Biological Transport ,Cell Biology ,medicine.disease ,Ascorbic acid ,Forum Review Articles ,Cell biology ,medicine.anatomical_structure ,chemistry ,General Earth and Planetary Sciences ,Endothelium, Vascular ,Protein Processing, Post-Translational - Abstract
Significance: Vitamin C, or ascorbic acid, has long been known to participate in several important functions in the vascular bed in support of endothelial cells. These functions include increasing the synthesis and deposition of type IV collagen in the basement membrane, stimulating endothelial proliferation, inhibiting apoptosis, scavenging radical species, and sparing endothelial cell-derived nitric oxide to help modulate blood flow. Although ascorbate may not be able to reverse inflammatory vascular diseases such as atherosclerosis, it may well play a role in preventing the endothelial dysfunction that is the earliest sign of many such diseases. Recent Advances: Beyond simply preventing scurvy, evidence is mounting that ascorbate is required for optimal function of many dioxygenase enzymes in addition to those involved in collagen synthesis. Several of these enzymes regulate the transcription of proteins involved in endothelial function, proliferation, and survival, including hypoxia-inducible factor-1α and histone and DNA demethylases. More recently, ascorbate has been found to acutely tighten the endothelial permeability barrier and, thus, may modulate access of ascorbate and other molecules into tissues and organs. Critical Issues: The issue of the optimal cellular content of ascorbate remains unresolved, but it appears that low millimolar ascorbate concentrations are normal in most animal tissues, in human leukocytes, and probably in the endothelium. Although there may be little benefit of increasing near maximal cellular ascorbate concentrations in normal people, many diseases and conditions have either systemic or localized cellular ascorbate deficiency as a cause for endothelial dysfunction, including early atherosclerosis, sepsis, smoking, and diabetes. Future Directions: A key focus for future studies of ascorbate and the vascular endothelium will likely be to determine the mechanisms and clinical relevance of ascorbate effects on endothelial function, permeability, and survival in diseases that cause endothelial dysfunction. Antioxid. Redox Signal. 19, 2068–2083.
- Published
- 2013
47. Combined vitamin C and E deficiency induces motor defects in gulo(-/-)/SVCT2(+/-) mice
- Author
-
James M. May, Marquicia R. Pierce, Danielle L. DiAsio, Fiona E. Harrison, and Laurisa M. Rodrigues
- Subjects
Vitamin ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Medicine (miscellaneous) ,Ascorbic Acid ,Biology ,medicine.disease_cause ,vitamin D deficiency ,Antioxidants ,Article ,chemistry.chemical_compound ,Mice ,Internal medicine ,Malondialdehyde ,medicine ,L-gulonolactone oxidase ,Animals ,Vitamin E ,Vitamin E Deficiency ,Mice, Knockout ,F2-Isoprostanes ,Nutrition and Dietetics ,Vitamin C ,General Neuroscience ,Brain ,General Medicine ,Ascorbic acid ,medicine.disease ,Vitamin D Deficiency ,Disease Models, Animal ,Oxidative Stress ,Endocrinology ,chemistry ,Liver ,Dietary Supplements ,biology.protein ,Female ,Vitamin E deficiency ,Oxidative stress ,Biomarkers ,L-Gulonolactone Oxidase ,Psychomotor Performance - Abstract
Key antioxidants, vitamins C and E, are necessary for normal brain development and neuronal function. In this study, we depleted both of these vitamins in two mouse models to determine if oxidative stress due to combined vitamin C and E dietary deficiency altered their neurological phenotype. The first model lacked both alleles for the Gulonolactone oxidase gene (Gulo(-/-)) and therefore was unable synthesize vitamin C. To obtain an additional cellular deficiency of vitamin C, the second model also lacked one allele for the cellular vitamin C transporter gene (Gulo(-/-)/SVCT2(+/-)).The experimental treatment was 16 weeks of vitamin E deprivation followed by 3 weeks of vitamin C deprivation. Mice were assessed for motor coordination deficits, vitamin levels, and oxidative stress biomarkers.In the first model, defects in motor performance were more apparent in both vitamin C-deficient groups (VE+VC-, VE-VC-) compared to vitamin C-supplemented groups (VE+VC+, VE-VC+) regardless of vitamin E level. Analysis of brain cortex and liver confirmed decreases of at least 80% for each vitamin in mice on deficient diets. Vitamin E deficiency doubled oxidative stress biomarkers (F2-isoprostanes and malondialdehyde). In the second model, Gulo(-/-)/SVCT2(+/-) mice on the doubly deficient diets showed deficits in locomotor activity, Rota-rod performance, and other motor tasks, with no concomitant change in anxiety or spatial memory.Vitamin E deficiency alone caused a modest oxidative stress in brain that did not affect motor performance. Adding a cellular deficit in vitamin C to dietary deprivation of both vitamins significantly impaired motor performance.
- Published
- 2013
48. Accessibility and reactivity of ascorbate 6-palmitate bound to erythrocyte membranes
- Author
-
Charles E. Cobb, James M. May, and Zhi-chao Qu
- Subjects
Free Radicals ,Ascorbic Acid ,Biochemistry ,Micelle ,Lipid peroxidation ,chemistry.chemical_compound ,Physiology (medical) ,Humans ,Bovine serum albumin ,Ferricyanides ,Lipid bilayer ,biology ,Erythrocyte Membrane ,Electron Spin Resonance Spectroscopy ,Antimutagenic Agents ,Serum Albumin, Bovine ,Ascorbic acid ,Kinetics ,Membrane ,chemistry ,Liposomes ,biology.protein ,Spin Labels ,Ferricyanide ,Stearic acid ,Oxidation-Reduction - Abstract
Lipophilic derivatives of ascorbic acid may protect lipid bilayers and micelles against lipid peroxidation. In this work the binding, accessibility, and reducing capacity of ascorbate 6-palmitate (A6P) were studied in human erythrocyte membranes. In contrast to less lipophilic carbon-6-modified ascorbate derivatives, A6P bound to erythrocyte membranes in a concentration-dependent manner. This binding was preserved following centrifugation washes, but was largely reversed by extraction with bovine serum albumin. Most of the ascorbyl groups of membrane-bound A6P were readily accessible to oxidation by water-soluble oxidants. Ferricyanide quantitatively oxidized membrane-bound A6P, but the latter spared endogenous tocopherols from destruction. In EPR studies, A61? was much more effective than ascorbate in reducing nitroxide spin labels positioned at either carbon-5 or carbon-16 of membrane-bound stearic acid in both intact cells and in membranes. A6P, thus, appears to intercalate into the erythrocyte membrane with the ascorbyl group located superficially, but with access to the hydrophobic membrane interior, and with the ability to recycle endogenous a-tocopherol during oxidant stress.
- Published
- 1996
- Full Text
- View/download PDF
49. Functional Organization of Mammalian Hexokinase II
- Author
-
James M. May, Yutaka Yano, Steve Koch, Richard L. Printz, Daryl K. Granner, Hossein Ardehali, and Richard R. Whitesell
- Subjects
Gene isoform ,chemistry.chemical_classification ,Hexokinase ,Mutation ,biology ,Xenopus ,Cell Biology ,biology.organism_classification ,medicine.disease_cause ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Enzyme ,chemistry ,Gene duplication ,medicine ,Binding site ,Molecular Biology - Abstract
The mammalian hexokinase (HK) family includes three closely related 100-kDa isoforms (HKI-III) that are thought to have arisen from a common 50-kDa precursor by gene duplication and tandem ligation. Previous studies of HKI indicated that a glucose 6-phosphate (Glu-6-P)-regulated catalytic site resides in the COOH-terminal half of the molecule and that the NH2-terminal half contains only a Glu-6-P binding site. In contrast, we now show that proteins representing both halves of human and rat HKII have catalytic activity and that each is inhibited by Glu-6-P. The intact enzyme and the NH2- and COOH-terminal halves of the enzyme each increase glucose utilization when expressed in Xenopus oocytes. Mutations corresponding to either Asp-209 or Asp-657 in the intact enzyme completely inactivate the NH2- and COOH-terminal half enzymes, respectively. Mutation of either of these sites results in a 50% reduction of activity in the 100-kDa enzyme. Mutation of both sites results in a complete loss of activity. This suggests that each half of the HKII molecule retains catalytic activity within the 100-kDa protein. These observations indicate that HKI and HKII are functionally distinct and have evolved differently.
- Published
- 1996
- Full Text
- View/download PDF
50. Role of the C-terminal tail of the GLUT1 glucose transporter in its expression and function in Xenopus laevis oocytes
- Author
-
Zhican Qu, A.D. Due, Alvin C. Powers, J M Thomas, A. Buchs, and James M. May
- Subjects
endocrine system ,Monosaccharide Transport Proteins ,Recombinant Fusion Proteins ,Xenopus ,Biochemistry ,Protein Structure, Secondary ,Xenopus laevis ,Leucine ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Sequence Deletion ,chemistry.chemical_classification ,Glucose Transporter Type 1 ,Messenger RNA ,biology ,Cell Membrane ,Glucose transporter ,Methylglucosides ,nutritional and metabolic diseases ,Biological Transport ,Transporter ,biology.organism_classification ,Recombinant Proteins ,Amino acid ,Cell biology ,carbohydrates (lipids) ,Kinetics ,chemistry ,Mutagenesis, Site-Directed ,Oocytes ,biology.protein ,3-O-Methylglucose ,Female ,GLUT1 ,hormones, hormone substitutes, and hormone antagonists ,GLUT4 - Abstract
Structural determinants for the glucose transport kinetics of the erythrocyte glucose transporter have not been established. In this work the role of the cytosolic carboxy-terminal tail in the expression and function of the human GLUT1 isoform in Xenopus oocytes was investigated. Oocyte plasma membrane expression of GLUT1 was a saturable function of the amount of mRNA injected. Transport activity increased as a linear function of the amount of immunoreactive transporter in the plasma membrane. Transport kinetics of human GLUT1 expressed in oocytes resembled those of human erythrocyte GLUT1. Addition of up to 31 extra amino acids to the carboxy-terminal tail of GLUT1 was without effect on its function in oocytes. Removal of the carboxy-terminal 21 amino acids also did not affect GLUT1 expression or transport kinetics in oocytes. Removal of the entire carboxy-terminal tail to Phe-450 resulted in a transporter that had moderately decreased plasma membrane expression compared to that of GLUT1. However, transport activity of this construct was less than 5% of that of GLUT1, and was associated with loss of its outward-facing inhibitor binding site. When the carboxy-terminal 29 amino acids of GLUT1 were replaced with the corresponding region of GLUT4, transporter expression in the plasma membrane and the transport Vmax fell to low levels, similar to those of native GLUT4. When the carboxy-terminal 29 or 73 amino acids of GLUT1 were swapped into the corresponding region of GLUT4, the transport Vmax markedly increased to about one-third to one-half that of GLUT1, although the affinity for substrate was halved. These results show that the carboxy-terminal tail of the GLUT1 is not critical for targeting of the protein to the plasma membrane, but that this region is an important determinant of transport function.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.