Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(8): 105009, 2023 08.
Article in English | MEDLINE | ID: mdl-37406814

ABSTRACT

Selenoprotein P (SeP, encoded by the SELENOP gene) is a plasma protein that contains selenium in the form of selenocysteine residues (Sec, a cysteine analog containing selenium instead of sulfur). SeP functions for the transport of selenium to specific tissues in a receptor-dependent manner. Apolipoprotein E receptor 2 (ApoER2) has been identified as a SeP receptor. However, diverse variants of ApoER2 have been reported, and the details of its tissue specificity and the molecular mechanism of its efficiency remain unclear. In the present study, we found that human T lymphoma Jurkat cells have a high ability to utilize selenium via SeP, while this ability was low in human rhabdomyosarcoma cells. We identified an ApoER2 variant with a high affinity for SeP in Jurkat cells. This variant had a dissociation constant value of 0.67 nM and a highly glycosylated O-linked sugar domain. Moreover, the acidification of intracellular vesicles was necessary for selenium transport via SeP in both cell types. In rhabdomyosarcoma cells, SeP underwent proteolytic degradation in lysosomes and transported selenium in a Sec lyase-dependent manner. However, in Jurkat cells, SeP transported selenium in Sec lyase-independent manner. These findings indicate a preferential selenium transport pathway involving SeP and high-affinity ApoER2 in a Sec lyase-independent manner. Herein, we provide a novel dynamic transport pathway for selenium via SeP.


Subject(s)
Lyases , Selenium , Humans , Lyases/metabolism , Selenium/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoprotein P/genetics , Selenoprotein P/metabolism , Selenoproteins , Jurkat Cells
2.
Adv Exp Med Biol ; 1440: 193-211, 2024.
Article in English | MEDLINE | ID: mdl-38036881

ABSTRACT

The side-chain hydroxylation of cholesterol by specific enzymes produces 24(S)-hydroxycholesterol, 25-hydroxycholesterol, 27-hydroxycholesterol, and other products. These enzymatically formed side-chain oxysterols act as intermediates in the biosynthesis of bile acids and serve as signaling molecules that regulate cholesterol homeostasis. Besides these intracellular functions, an imbalance in oxysterol homeostasis is implicated in pathophysiology. Furthermore, growing evidence reveals that oxysterols affect cell proliferation and cause cell death. This chapter provides an overview of the pathophysiological role of side-chain oxysterols in developing human diseases. We also summarize our understanding of the molecular mechanisms underlying the induction of various forms of cell death by side-chain oxysterols.


Subject(s)
Oxysterols , Humans , Bile Acids and Salts , Cholesterol/metabolism , Homeostasis , Oxysterols/metabolism
3.
Nucleic Acids Res ; 49(12): 6893-6907, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34142161

ABSTRACT

Selenoprotein P (SELENOP) is a major plasma selenoprotein that contains 10 Sec residues, which is encoded by the UGA stop codon. The mRNA for SELENOP has the unique property of containing two Sec insertion sequence (SECIS) elements, which is located in the 3' untranslated region (3'UTR). Here, we coincidentally identified a novel gene, CCDC152, by sequence analysis. This gene was located in the antisense region of the SELENOP gene, including the 3'UTR region in the genome. We demonstrated that this novel gene functioned as a long non-coding RNA (lncRNA) that decreased SELENOP protein levels via translational rather than transcriptional, regulation. We found that the CCDC152 RNA interacted specifically and directly with the SELENOP mRNA and inhibited its binding to the SECIS-binding protein 2, resulting in the decrease of ribosome binding. We termed this novel gene product lncRNA inhibitor of SELENOP translation (L-IST). Finally, we found that epigallocatechin gallate upregulated L-IST in vitro and in vivo, to suppress SELENOP protein levels. Here, we provide a new regulatory mechanism of SELENOP translation by an endogenous long antisense ncRNA.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis , RNA, Long Noncoding/metabolism , Selenoprotein P/genetics , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Down-Regulation , Humans , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Selenoprotein P/biosynthesis
4.
Neuropathology ; 40(6): 587-598, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33305472

ABSTRACT

A vast body of evidence implicates increased oxidative stress and extracellular glutamate accumulation in the pathomechanism of sporadic amyotrophic lateral sclerosis (ALS). Cystine/glutamate antiporter (xCT) carries extracellular cystine uptake and intracellular glutamate release (cystine/glutamate exchange) in the presence of oxidative stress. The aim of the present study was to determine the involvement of xCT in ALS. Immunohistochemical observations in the spinal cord sections demonstrated that xCT was mainly expressed in astrocytes, with staining more intense in 12 sporadic ALS patients as compared to 12 age-matched control individuals. Western blot and densitometric analyses of the spinal cord samples revealed that the relative value of xCT/ß-actin optical density ratio was significantly higher in the ALS group as compared to the control group. Next, we conducted cell culture experiments using a human astrocytoma-derived cell line (1321N1) and a mouse motor neuron/neuroblastoma hybrid cell line (NSC34). In 1321N1 cells, the normalized xCT expression levels in cell lysates were significantly increased by H2 O2 treatment. Glutamate concentrations in 1321 N1 cell culture-conditioned media were significantly elevated by H2 O2 treatment, and the H2 O2 -driven elevations were completely canceled by the xCT inhibitor erastin pretreatment. In motor neuron-differentiated NSC34 cells (NSC34d cells), both the normalized xCT expression levels in the cell lysates and glutamate concentrations in the cell-conditioned media were constant with or without H2 O2 treatment. The present results provide in vivo and in vitro evidence that astrocytes upregulate xCT expression to release glutamate in response to increased oxidative stress associated with ALS, contributing to extracellular glutamate accumulation.


Subject(s)
Amino Acid Transport System y+/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Glutamic Acid/metabolism , Oxidative Stress/physiology , Amyotrophic Lateral Sclerosis/pathology , Animals , Humans , Mice , Spinal Cord/metabolism , Spinal Cord/pathology , Up-Regulation
5.
Neuropathology ; 40(2): 152-166, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31883180

ABSTRACT

Previous studies on sporadic amyotrophic lateral sclerosis (SALS) demonstrated iron accumulation in the spinal cord and increased glutamate concentration in the cerebrospinal fluid. To clarify the relationship between the two phenomena, we first performed quantitative and morphological analyses of substances related to iron and glutamate metabolism using spinal cords obtained at autopsy from 12 SALS patients and 12 age-matched control subjects. Soluble iron content determined by the Ferrozine method as well as ferritin (Ft) and glutaminase C (GLS-C) expression levels on Western blots were significantly higher in the SALS group than in the control group, while ferroportin (FPN) levels on Western blots were significantly reduced in the SALS group as compared to the control group. There was no significant difference in aconitase 1 (ACO1) and tumor necrosis factor-alpha (TNFα)-converting enzyme (TACE) levels on Western blots between the two groups. Immunohistochemically, Ft, ACO1, TACE, TNFα, and GLS-C were proven to be selectively expressed in microglia. Immunoreactivities for FPN and hepcidin were localized in neuronal and glial cells. Based on these observations, it is predicted that soluble iron may stimulate microglial glutamate release. To address this issue, cell culture experiments were carried out on a microglial cell line (BV-2). Treatment of BV-2 cells with ferric ammonium citrate (FAC) brought about significant increases in intracellular soluble iron and Ft expression levels and conditioned medium glutamate and TNFα concentrations. Glutamate concentration was also significantly increased in conditioned media of TNFα-treated BV-2 cells. While the FAC-driven increases in glutamate and TNFα release were completely canceled by pretreatment with ACO1 and TACE inhibitors, respectively, the TNFα-driven increase in glutamate release was completely canceled by GLS-C inhibitor pretreatment. Moreover, treatment of BV-2 cells with hepcidin resulted in a significant reduction in FPN expression levels on Western blots of the intracellular total protein extracts. The present results provide in vivo and in vitro evidence that microglial glutamate release in SALS spinal cords is enhanced by intracellular soluble iron accumulation-induced activation of ACO1 and TACE and by increased extracellular TNFα-stimulated GLS-C upregulation, and suggest a positive feedback mechanism to maintain increased intracellular soluble iron levels, involving TNFα, hepcidin, and FPN.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Glutamic Acid/metabolism , Iron/metabolism , Microglia/metabolism , Adult , Aged , Aged, 80 and over , Cadaver , Female , Humans , Male , Middle Aged , Spinal Cord/metabolism , Spinal Cord/pathology
6.
Genes Cells ; 23(1): 22-34, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29205725

ABSTRACT

Chronic myeloid leukemia (CML) is caused by the chimeric protein p210 BCR-ABL encoded by a gene on the Philadelphia chromosome. Although the kinase domain of p210 BCR-ABL is an active driver of CML, the pathological role of its pleckstrin homology (PH) domain remains unclear. Here, we carried out phospholipid vesicle-binding assays to show that cardiolipin (CL), a characteristic mitochondrial phospholipid, is a unique ligand of the PH domain. Arg726, a basic amino acid in the ligand-binding region, was crucial for ligand recognition. A subset of wild-type p210 BCR-ABL that was transiently expressed in HEK293 cells was dramatically translocated from the cytosol to mitochondria in response to carbonyl cyanide m-chlorophenylhydrazone (CCCP) treatment, which induces mitochondrial depolarization and subsequent externalization of CL to the organelle's outer membrane, whereas an R726A mutant of the protein was not translocated. Furthermore, only wild-type p210 BCR-ABL, but not the R726A mutant, suppressed CCCP-induced mitophagy and subsequently enhanced reactive oxygen species production. Thus, p210 BCR-ABL can change its intracellular localization via interactions between the PH domain and CL to cope with mitochondrial damage. This suggests that p210 BCR-ABL could have beneficial effects for cancer proliferation, providing new insight into the PH domain's contribution to CML pathogenesis.


Subject(s)
Cardiolipins/metabolism , Fusion Proteins, bcr-abl/metabolism , Mitochondria/pathology , Mitophagy/drug effects , Pleckstrin Homology Domains , Carbonyl Cyanide m-Chlorophenyl Hydrazone/analogs & derivatives , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cytosol/metabolism , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , HEK293 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Protein Transport
7.
Biol Pharm Bull ; 41(5): 777-785, 2018.
Article in English | MEDLINE | ID: mdl-29709915

ABSTRACT

PC12D cells, a subline of rat adrenal pheochromocytoma PC12 cells, extend neurites rapidly in response to differentiation stimuli and are used to investigate the molecular mechanisms of neurite extension. In the present study, we found significant tolerance of PC12D cells against Parkinson's disease-related stimuli such as dopamine and 6-hydroxydopamine; this tolerance was significantly decreased by a change in the medium. Conditioned medium from PC12D cells induced tolerance against oxidative stress, which suggests that cytoprotective factor may be released by PC12D cells into the culture medium. Conditioned medium-induced tolerance was not found for PC12 cells or human neuroblastoma SH-SY5Y cells. A cytoprotective factor generated by PC12D cells exhibited hydrogen peroxide-reducing activity. Chemical characterization showed that this cytoprotective factor is water soluble and has a molecular weight about 1000 Da, and that its activity is inhibited by sodium cyanide. Release of this cytoprotective factor was increased by differentiation stimuli and oxidative stress. Taken together, these results suggest that release of a hydrogen peroxide-reducing factor by PC12D cells increases cell tolerance against oxidative stress. This study provides new insights into the antioxidative properties of factors in extracellular fluid.


Subject(s)
Extracellular Fluid/metabolism , Hydrogen Peroxide/metabolism , Pheochromocytoma/metabolism , Animals , Cell Line, Tumor , Cell Survival , Culture Media , Oxidation-Reduction , Oxidative Stress , Rats
8.
Biol Pharm Bull ; 41(5): 828-832, 2018.
Article in English | MEDLINE | ID: mdl-29709922

ABSTRACT

Selenoprotein P (SeP) is a selenium (Se)-rich extracellular protein. SeP is identified as a hepatokine, causing insulin resistance in type 2 diabetes. Thus, the measurement of SeP in serum has received much attention, and several enzyme-linked immunosorbent assay (ELISA) kits for SeP determination are now commercially available. In the present study, we determined the serum SeP levels by our original ELISA and sol particle homogeneous immunoassay (SPIA) methods and also by commercially available kits, and these determinants were compared. We found a kit-dependent correlation of the determinants with our methods. These results suggest that the selection of kit is critical for comparison with our previous reports and for discussing the relationship between the serum SeP levels and disease condition.


Subject(s)
Immunoassay/methods , Selenoprotein P/blood , Aged , Female , Humans , Male , Middle Aged , Reproducibility of Results
9.
J Lipid Res ; 57(11): 2005-2014, 2016 11.
Article in English | MEDLINE | ID: mdl-27647838

ABSTRACT

The 24(S)-hydroxycholesterol (24S-OHC), which plays an important role in maintaining brain cholesterol homeostasis, has been shown to possess neurotoxicity. We have previously reported that 24S-OHC esterification by ACAT1 and the resulting lipid droplet (LD) formation are responsible for 24S-OHC-induced cell death. In the present study, we investigate the functional roles of 24S-OHC esters and LD formation in 24S-OHC-induced cell death, and we identify four long-chain unsaturated fatty acids (oleic acid, linoleic acid, arachidonic acid, and DHA) with which 24S-OHC is esterified in human neuroblastoma SH-SY5Y cells treated with 24S-OHC. Here, we find that cotreatment of cells with 24S-OHC and each of these four unsaturated fatty acids increases prevalence of the corresponding 24S-OHC ester and exacerbates induction of cell death as compared with cell death induced by treatment with 24S-OHC alone. Using electron microscopy, we find in the present study that 24S-OHC induces formation of LD-like structures coupled with enlarged endoplasmic reticulum (ER) lumina, and that these effects are suppressed by treatment with ACAT inhibitor. Collectively, these results illustrate that ACAT1-catalyzed esterification of 24S-OHC with long-chain unsaturated fatty acid followed by formation of atypical LD-like structures at the ER membrane is a critical requirement for 24S-OHC-induced cell death.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Brain/metabolism , Hydroxycholesterols/administration & dosage , Lipid Droplets/metabolism , Neurons/metabolism , Arachidonic Acid/administration & dosage , Arachidonic Acid/metabolism , Brain/drug effects , Brain/pathology , Cell Death/drug effects , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Esterification/drug effects , Gene Expression Regulation/drug effects , Humans , Hydroxycholesterols/metabolism , Linoleic Acid/administration & dosage , Linoleic Acid/metabolism , Lipid Droplets/chemistry , Lipid Droplets/drug effects , Neuroblastoma/metabolism , Neurons/pathology , Oleic Acid/administration & dosage , Oleic Acid/metabolism
10.
Arch Biochem Biophys ; 595: 109-12, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27095225

ABSTRACT

Ebselen is an organoselenium compound with glutathione peroxidase (GPx)-like hydroperoxide reducing activity. Moreover, ebselen has its own unique reactivity, with functions that GPx does not have, since it reacts with many kinds of thiols other than glutathione. Ebselen may affect the thioredoxin systems, through which it may contribute to regulation of cell function. With high reactivity toward thiols, hydroperoxides, and peroxynitrite, ebselen has been used as a useful tool in research on cellular redox mechanisms. Unlike α-tocopherol, ebselen does not scavenge lipid peroxyl radicals, which is another advantage of ebselen for use as a research tool in comparison with radical scavenging antioxidants. Selenium is not released from the ebselen molecule, which explains the low toxicity of ebselen. To further understand the mechanism of cellular redox biology, it should be interesting to compare the effects of ebselen with that of selenoprotein P, which supplies selenium to GPx. New medical applications of ebselen as a drug candidate for human diseases such as cancer and diabetes mellitus as well as brain stroke and ischemia will be expected.


Subject(s)
Azoles/pharmacology , Organoselenium Compounds/pharmacology , Humans , Isoindoles , Oxidation-Reduction
11.
Bioorg Med Chem ; 24(11): 2559-66, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27117262

ABSTRACT

We synthesized several candidates of 24(S)-hydroxycholesterol (24S-OHC) esters, which are involved in neuronal cell death, through catalysis with acyl-CoA:cholesterol acyltransferase-1 (ACAT-1). We studied the regioselectivity of the acylation of the secondary alcohol at the 3- or 24-position of 24S-OHC. The appropriate saturated and unsaturated long-chain fatty acids were esterified with the protected 24S-OHC and then de-protected to afford the desired esters at a satisfactory yield. We then confirmed by HPLC monitoring that the retention times of four esters of 24S-OHC, namely 3-oleate, 3-linoleate, 3-arachidonoate and 3-docosahexaenoate, were consistent with those of 24S-OHC esters observed in 24S-OHC-treated SH-SY5Y cells.


Subject(s)
Hydroxycholesterols/pharmacology , Neuroblastoma/drug therapy , Cell Death/drug effects , Dose-Response Relationship, Drug , Humans , Hydroxycholesterols/chemical synthesis , Hydroxycholesterols/chemistry , Molecular Structure , Neuroblastoma/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
12.
J Lipid Res ; 56(11): 2172-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26411970

ABSTRACT

Selenocysteine (Sec) insertion sequence-binding protein 2 (SBP2) is essential for the biosynthesis of Sec-containing proteins, termed selenoproteins. Subjects with mutations in the SBP2 gene have decreased levels of several selenoproteins, resulting in a complex phenotype. Selenoproteins play a significant role in antioxidative defense, and deficiencies in these proteins can lead to increased oxidative stress. However, lipid peroxidation and the effects of antioxidants in subjects with SBP2 gene mutations have not been studied. In the present study, we evaluated the lipid peroxidation products in the blood of a subject (the proband) with mutations in the SBP2 gene. We found that the proband had higher levels of free radical-mediated lipid peroxidation products, such as 7ß-hydroxycholesterol, than the control subjects. Treatment of the proband with vitamin E (α-tocopherol acetate, 100 mg/day), a lipid-soluble antioxidant, for 2 years reduced lipid peroxidation product levels to those of control subjects. Withdrawal of vitamin E treatment for 7 months resulted in an increase in lipid peroxidation products. Collectively, these results clearly indicate that free radical-mediated oxidative stress is increased in the subject with SBP2 gene mutations and that vitamin E treatment effectively inhibits the generation of lipid peroxidation products.


Subject(s)
Antioxidants/therapeutic use , Lipid Peroxidation/drug effects , RNA-Binding Proteins/genetics , Vitamin E/therapeutic use , Adolescent , Antioxidants/pharmacology , Case-Control Studies , Child , Female , Humans , Leukocyte Count , Male , Metabolic Diseases/drug therapy , Mutation, Missense , Selenoproteins/blood , Vitamin E/pharmacology
13.
Biosci Biotechnol Biochem ; 79(6): 912-8, 2015.
Article in English | MEDLINE | ID: mdl-25612552

ABSTRACT

According to the amyloid hypothesis, amyloid ß accumulates in brains with Alzheimer's disease (AD) and triggers cell death and memory deficit. Previously, we developed a rice Aß vaccine expressing Aß, which reduced brain Aß levels in the Tg2576 mouse model of familial AD. We used senescence-accelerated SAMP8 mice as a model of sporadic AD and investigated the relationship between Aß and oxidative stress. Insoluble Aß and 4-hydroxynonenal (4-HNE) levels tended to be reduced in SAMP8 mice-fed the rice Aß vaccine. We attempted to clarify the relationship between oxidative stress and Aß in vitro. Addition of Aß peptide to the culture medium resulted in an increase in 4-HNE levels in SH-SY5Y cells. Tg2576 mice, which express large amounts of Aß in their brain, also exhibited increased 4-HNE levels; this increase was inhibited by the Aß vaccine. These results indicate that Aß induces oxidative stress in cultured cells and in the mouse brain.


Subject(s)
Aging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Oxidative Stress , Peptide Fragments/metabolism , Aldehydes/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Brain/metabolism , Buffers , Humans , Male , Maze Learning , Mice , Mice, Transgenic , Oryza/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Solubility , Vaccines/genetics
14.
J Neurosci ; 33(37): 14767-77, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027277

ABSTRACT

The cytoplasmic regulatory protein p62 (Sequestosome 1/A170) is known to modulate various receptor-mediated intracellular signaling pathways. p62 deficiency was shown to result in mature-onset obesity in mice, but the mechanisms underlying this abnormality remained unclear. Here we report that hyperphagia due to central leptin resistance is the cause of obesity in p62(-/-) mice. We found that these mice show hyperphagia. Restriction of food to the amount eaten by wild-type mice prevented excess body weight gain and fat accumulation, suggesting that overfeeding is the primary cause of obesity in p62(-/-) mice. Brain-specific p62 deficiency caused mature-onset obesity to the same extent as in p62(-/-) mice, further supporting a neuronal mechanism as the major cause of obesity in these mice. Immunohistochemical analysis revealed that p62 is highly expressed in hypothalamic neurons, including POMC neurons in the arcuate nucleus. Central leptin resistance was observed even in young preobese p62(-/-) mice. We found a defect in intracellular distribution of the transcription factor Stat3, which is essential for the action of leptin, in p62(-/-) mice. These results indicate that brain p62 plays an important role in bodyweight control by modulating the central leptin-signaling pathway and that lack of p62 in the brain causes leptin resistance, leading to hyperphagia. Thus, p62 could be a clinical target for treating obesity and metabolic syndrome.


Subject(s)
Brain/drug effects , Hyperphagia/genetics , Hyperphagia/pathology , Leptin/pharmacology , Transcription Factors/deficiency , Animals , Body Weight/drug effects , Body Weight/genetics , Brain/cytology , Brain/metabolism , Eating/drug effects , Eating/genetics , Embryo, Mammalian , Food Deprivation , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/pharmacology , Oxygen Consumption/genetics , Pro-Opiomelanocortin/genetics , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factor TFIIH
15.
Biochem Biophys Res Commun ; 446(3): 741-4, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24380859

ABSTRACT

Polyunsaturated fatty acids and their esters are known to be susceptible to free radical-mediated oxidation, whereas cholesterol is thought to be more resistant to oxidation. In fact, it has been observed that in the case of plasma lipid peroxidation, the amount of oxidation products of polyunsaturated fatty acids such as linoleic acid was higher than that of cholesterol. In contrast, during oxidative stress-induced cellular lipid peroxidation, oxidation products of cholesterol such as 7-hydroxycholesterol (7-OHCh) were detected in greater amounts than those of linoleates such as hydroxyoctadecadienoic acid (HODE). There are several forms of oxidation products of cholesterol and linoleates in vivo, namely, hydroperoxides, as well as the hydroxides of both the free and ester forms of cholesterol and linoleates. To evaluate these oxidation products, a method used to determine the lipid oxidation products after reduction and saponification was developed. With this method, several forms of oxidation products of cholesterol and linoleates are measured as total 7-OHCh (t7-OHCh) and total HODE (tHODE), respectively. During free radical-mediated lipid peroxidation in plasma, the amount of tHODE was 6.3-fold higher than that of t7-OHCh. In contrast, when Jurkat cells were exposed to free radicals, the increased amount of cellular t7-OHCh was 5.7-fold higher than that of tHODE. Higher levels of t7-OHCh than those of tHODE have also been observed in selenium-deficient Jurkat cells and glutamate-treated neuronal cells. These results suggest that, in contrast to plasma oxidation, cellular cholesterol is more susceptible to oxidation than cellular linoleates. Collectively, cholesterol oxidation products at the 7-position may be a biomarker of cellular lipid peroxidation.


Subject(s)
Biomarkers/blood , Fatty Acids, Unsaturated/blood , Hydroxycholesterols/metabolism , Lipid Peroxidation , Humans , Hydroxycholesterols/blood , Jurkat Cells/metabolism , Oxidation-Reduction
16.
Biochem Biophys Res Commun ; 446(3): 692-6, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24530911

ABSTRACT

24(S)-hydroxycholesterol (24S-OHC) which is enzymatically produced in the brain plays important physiological roles in maintaining brain cholesterol homeostasis. We found that 24S-OHC at sub-lethal concentrations down-regulated amyloid precursor protein (APP) trafficking via enhancement of the complex formation of APP with up-regulated glucose-regulated protein 78, an endoplasmic reticulum chaperone. In accordance with this mechanism, 24S-OHC suppressed amyloid-ß production in human neuroblastoma SH-SY5Y cells. Furthermore, 24S-OHC at sub-lethal concentrations induced adaptive responses via transcriptional activation of the liver X receptor signaling pathway, thereby protecting neuronal cells against the forthcoming oxidative stress induced by 7-ketocholesterol. On the other hand, we found that high concentrations of 24S-OHC induced apoptosis in T-lymphoma Jurkat cells which endogenously expressed caspase-8, and induced necroptosis - a form of programmed necrosis - in neuronal SH-SY5Y cells which expressed no caspase-8. In this Article, we show the diverse functions of 24S-OHC and consider the possible importance of controlling 24S-OHC levels in the brain for preventing neurodegenerative diseases.


Subject(s)
Brain/metabolism , Hydroxycholesterols/metabolism , Ketocholesterols , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , Brain/drug effects , Caspase 8/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Humans , Hydroxycholesterols/pharmacology , Ketocholesterols/metabolism , Ketocholesterols/pharmacology , Liver X Receptors , Orphan Nuclear Receptors/metabolism
17.
FASEB J ; 27(10): 4305-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23839932

ABSTRACT

Cholesterol can be converted to 24S-hydroxycholesterol (24SOHC) by neuronal cholesterol 24-hydroxylase. In mouse models of Alzheimer's disease (AD), increasing 24SOHC levels reduced AD pathology. However, mechanisms underlying the effects of 24SOHC on amyloid-ß (Aß) production have remained unclear. Here we report that 24SOHC treatment reduces Aß production and increases endoplasmic reticulum (ER)-resident immature amyloid precursor protein (APP) levels in human neuroblastoma SH-SY5Y cells and CHO cells stably expressing human APP. Treatment with 1-10 µM 24SOHC (equivalent to the concentrations detected in human brain homogenates) diminished Aß production (IC50=4.6 µM for Aß40) without affecting secretase activities. To evaluate the intracellular APP transport, we established an in vitro vesicle formation assay. We found that APP budding via COPII vesicles was diminished by 70% in 24SOHC-treated cells. The proteomics and immunoblotting analysis revealed that 24SOHC induced the expression of glucose-regulated protein 78 (GRP78), an ER chaperone, through unfolded protein response pathways, and enhanced the formation of the APP/GRP78 complex. Knockdown of GRP78 diminished the inhibitory effects of 24SOHC on Aß production. These results suggest that 24SOHC down-regulates APP trafficking via enhancement of the complex formation of APP with up-regulated GRP78 in the ER, resulting in suppression of Aß production.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Hydroxycholesterols/pharmacology , Protein Transport/physiology , Animals , Cell Line , Cricetinae , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mice , Protein Binding
18.
Free Radic Biol Med ; 221: 257-260, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38754742

ABSTRACT

It has generally been accepted that vitamin E refers to a group of tocochromanols, α-, ß-, γ-, and δ-tocopherols and the corresponding four tocotrienols. Recently, Azzi and colleagues proposed to restrict the term vitamin E only to RRR-α-tocopherol, not to other tocopherols and tocotrienols (Azzi A et al. Free Radic Biol Med. 2023; 207:178-180. doi: 10.1016/j.freeradbiomed.2023.06.029). The aim of this paper is to express our opinion on the nomenclature of vitamin E based on available scientific data. In our opinion, it would be inappropriate to exclude all the tocochromanols other than RRR-α-tocopherol from the vitamin E group at this stage when the molecular mechanisms showing how vitamin E deficiency causes diseases such as ataxia and how vitamin E prevents/reverses such diseases are not elucidated. Understanding of whole functions of tocochromanols including underlying mechanisms and dynamics is essential before revision of currently accepted definition of vitamin E. The potential roles of γ-tocopherol and tocotrienols are discussed despite whether they are vitamin function should be clarified in the future studies.


Subject(s)
Terminology as Topic , Vitamin E Deficiency , Vitamin E , alpha-Tocopherol , Vitamin E/chemistry , Vitamin E/classification , Humans , alpha-Tocopherol/chemistry , Ataxia/classification , Tocotrienols/classification , Tocotrienols/chemistry , Antioxidants/chemistry , Animals
19.
Free Radic Biol Med ; 211: 89-95, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38101585

ABSTRACT

Cholesterol is an essential component of cell membranes and serves as an important precursor of steroidal hormones and bile acids, but elevated levels of cholesterol and its oxidation products have been accepted as a risk factor for maintenance of health. The free and ester forms of cholesterol and fatty acids are the two major biological lipids. The aim of this hypothesis paper is to address the long-standing dogma that cholesterol is less susceptible to free radical peroxidation than polyunsaturated fatty acids (PUFAs). It has been observed that cholesterol is peroxidized much slower than PUFAs in plasma but that, contrary to expectations from chemical reactivity toward peroxyl radicals, cholesterol appears to be more readily autoxidized than linoleates in cell membranes. The levels of oxidation products of cholesterol and linoleates observed in humans support this notion. It is speculated that this discrepancy is ascribed to the fact that cholesterol and phospholipids bearing PUFAs are localized apart in raft and non-raft domains of cell membranes respectively and that the antioxidant vitamin E distributed predominantly in the non-raft domains cannot suppress the oxidation of cholesterol lying in raft domains which are relatively deficient in antioxidant.


Subject(s)
Linoleic Acid , Phospholipids , Humans , Phospholipids/metabolism , Linoleic Acid/metabolism , Lipid Peroxidation , Antioxidants/metabolism , Cholesterol/metabolism , Cell Membrane/metabolism , Fatty Acids, Unsaturated/metabolism , Linoleic Acids/metabolism
20.
Steroids ; 189: 109136, 2023 01.
Article in English | MEDLINE | ID: mdl-36351491

ABSTRACT

The brain-specific cholesterol metabolite 24(S)-hydroxycholesterol (24S-OHC) has been shown to cause neuronal cell death when subjected to esterification by acyl-CoA:cholesterol acyltransferase 1 (ACAT1). Accumulating 24S-OHC esters in the endoplasmic reticulum (ER) provoked ER membrane disruption and an integrated stress response (ISR), a signaling pathway that regulates adaptation to various stresses. We have previously reported that α-tocopherol (α-Toc) but not α-tocotrienol (α-Toc3), among vitamin E homologs, suppressed 24S-OHC-induced cell death without affecting ACAT1 activity in human neuroblastoma SH-SY5Y cells. However, the precise mechanisms underlying the inhibitory activity of α-Toc have yet to be elucidated. In the present study, we aimed to investigate the effects of α-Toc on the 24S-OHC-induced cell death machinery. We showed that α-Toc, but not α Toc3, suppressed 24S-OHC-induced ISR and downstream eukaryotic translation initiator factor 2α (eIF2α) phosphorylation. We also found that α-Toc inhibited stress granule formation and robust downregulation of nascent protein synthesis, which were induced by 24S-OHC treatment. Furthermore, disruption of ER membrane integrity was suppressed by α-Toc, but not by α-Toc3. Our findings suggest that the inhibitory effects of α-Toc on 24S-OHC-induced cell death may be attributed to its protective function against ER membrane disruption.


Subject(s)
Neuroblastoma , alpha-Tocopherol , Humans , alpha-Tocopherol/pharmacology , alpha-Tocopherol/metabolism , Cell Death , Endoplasmic Reticulum/metabolism , Hydroxycholesterols/pharmacology , Neuroblastoma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL