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










Publication year range
1.
Mech Ageing Dev ; 219: 111933, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38588730

ABSTRACT

The global prevalence of nonalcoholic fatty liver disease (NAFLD) defined by the increased number of lipid droplets (LDs) in hepatocytes, have risen continuously in parallel with the obesity. LDs and related proteins are known to affect cellular metabolism and signaling. Seipin, one of the most important LD-related proteins, plays a critical role in LD biogenesis. Although the role of adipose tissue-specific Seipin silencing is known, hepatocyte-specific silencing upon cholesterol-mediated lipid accumulation has not been investigated. In our study, we investigated the effect of Seipin on endoplasmic reticulum (ER) stress and lipophagy in cholesterol accumulated mouse hepatocyte cells. In this direction, cholesterol accumulation was induced by cholesterol-containing liposome, while Seipin mRNA and protein levels were reduced by siRNA. Our findings show that cholesterol containing liposome administration in hepatocytes increases both Seipin protein and number of large LDs. However Seipin silencing reduced the increase of cholesterol mediated large LDs and Glucose-regulated protein 78 (GRP78) mRNA. Additionally, lysosome-LD colocalization increased only in cells treated with cholesterol containing liposome, while the siRNA against Seipin did not lead any significant difference. According to our findings, we hypothesize that Seipin silencing in hepatocytes reduced cholesterol mediated LD maturation as well as GRP78 levels, but not lipophagy.


Subject(s)
Autophagy , Cholesterol , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , GTP-Binding Protein gamma Subunits , Hepatocytes , Lipid Droplets , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Chaperone BiP/metabolism , Animals , GTP-Binding Protein gamma Subunits/metabolism , Mice , Lipid Droplets/metabolism , Cholesterol/metabolism , Hepatocytes/metabolism , Autophagy/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Heat-Shock Proteins/metabolism
2.
Redox Biol ; 62: 102666, 2023 06.
Article in English | MEDLINE | ID: mdl-36934646

ABSTRACT

Docosahexaenoic acid (DHA), a representative omega-3 (ω-3) polyunsaturated fatty acids, undergoes metabolism to produce biologically active electrophilic species. 17-Oxo-DHA is one such reactive metabolite generated from DHA by cyclooxygenase-2 and dehydrogenase in activated macrophages. The present study was aimed to investigate the effects of 17-oxo-DHA on ultraviolet B (UVB)-induced oxidative stress, inflammation, and carcinogenesis in mouse skin. UVB-induced epidermal cell death was ameliorated by topically applied 17-oxo-DHA. Topical application of 17-oxo-DHA onto hairless mouse skin inhibited UVB-induced phosphorylation of the proinflammatory transcription factor, STAT3 on tyrosine 705 (Tyr705). The 17-oxo-DHA treatment also reduced the levels of oxidative stress markers, 4-hydroxynonenal-modified protein, malondialdehyde, and 8-oxo-2'-deoxyguanosine. The protective effects of 17-oxo-DHA against oxidative damage in UVB-irradiated mouse skin were associated with activation of Nrf2. 17-Oxo-DHA enhanced the engulfment of apoptotic JB6 cells by macrophages, which was related to the increased expression of the scavenger receptor CD36. The 17-oxo-DHA-mediated potentiation of efferocytic activity of macrophages was attenuated by the pharmacologic inhibition or knockout of Nrf2. The pretreatment with 17-oxo-DHA reduced the UVB-induced skin carcinogenesis and tumor angiogenesis. It was also confirmed that 17-oxo-DHA treatment significantly inhibited the phosphorylation of the Tyr705 residue of STAT3 and decreased the expression of its target proteins in cutaneous papilloma. In conclusion, 17-oxo-DHA protects against UVB-induced oxidative cell death, dermatitis, and carcinogenesis. These effects were associated with inhibition of STAT3-mediated proinflammatory signaling and also activation of Nrf2 with subsequent upregulation of antioxidant and anti-inflammatory gene expression.


Subject(s)
Dermatitis , Fatty Acids, Omega-3 , Mice , Animals , Docosahexaenoic Acids/pharmacology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Fatty Acids, Omega-3/pharmacology , Oxidative Stress , Carcinogenesis , Ultraviolet Rays/adverse effects , Cell Death
3.
Free Radic Biol Med ; 191: 1-7, 2022 10.
Article in English | MEDLINE | ID: mdl-35995397

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), based on the elevating obesity incidence, is one of the major health issue worldwide. Transition from NAFLD to non-alcoholic steatohepatitis (NASH) is driven by increased apoptosis and is relevant to higher morbidity rates. In regard to limited understanding on cholesterol mediated hepatocyte alterations in NALFD/NASH transition, we investigated endoplasmic reticulum (ER) stress and related apoptosis. Our findings suggest that cholesterol upregulates ER stress and enhances C/EBP homologous protein (CHOP) either in hypercholesterolemic rabbits or in hepatocytes treated with liposome-cholesterol complex. Mechanistically, cholesterol accumulation in hepatocytes activates IRE1/p38 branch of ER stress, stimulating CHOP levels. In liver tissues of cholesterol fed rabbits, α-tocopherol supplementation decreased IRE1/p38/CHOP activation and prevented NASH development. Thus, our study provides a critical role of hepatocyte cholesterol in inducing IRE1/p38/CHOP pathway and suggests novel candidates for therapeutic targets against NASH.


Subject(s)
Cholesterol , Endoplasmic Reticulum Stress , Non-alcoholic Fatty Liver Disease , Animals , Apoptosis , Cholesterol/metabolism , Hepatocytes/metabolism , Liposomes/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein Serine-Threonine Kinases/genetics , Rabbits , alpha-Tocopherol
4.
IUBMB Life ; 74(1): 85-92, 2022 01.
Article in English | MEDLINE | ID: mdl-34350697

ABSTRACT

The seminiferous tubules where spermatogenesis occurs are enveloped and protected by the Sertoli cells to support germ cells undergoing meiosis to produce haploid gametes. Clearly, induction of apoptosis in seminiferous tubules leads to abnormalities in spermatogenesis and male infertility. Studies demonstrated that increased hyperlipidemia impairs male infertility and spermatogenesis by enhancing seminiferous tubules apoptosis. However, molecular mechanisms underlying high-cholesterol-mediated testicular damage remain poorly elucidated. In this scope, we established a rabbit model and investigated the role of endoplasmic reticulum (ER) stress on high cholesterol diet induced seminiferous tubule apoptosis. Histopatological examinations revealed increased seminifer tubule apoptosis in testes of rabbits fed high cholesterol diet. In addition, phosphorylated forms of IRE1 and PERK, two well-identified markers of ER stress, were significantly induced in accordance with high cholesterol diet. High cholesterol diet also exhibited CHOP induction in testes, indicating increased ER stress related apoptosis. Supplementation of α-tocopherol significantly attenuated cholesterol mediated ER stress, and restored seminiferous tubules apoptosis. Taken together, our findings suggest that α-tocopherol might be capable to reduce testicular damage via ameliorating histopatological features and inhibiting seminiferous tubules apoptosis in hypercholesterolemic rabbits.


Subject(s)
Hypercholesterolemia , Testis , Animals , Apoptosis , Cholesterol , Diet , Male , Rabbits , alpha-Tocopherol/pharmacology
5.
Metabol Open ; 12: 100138, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34704008

ABSTRACT

OBJECTIVE: Increased fatty acid and triglyceride synthesis in liver, majorly modulated by Sterol Regulator Elementing Binding Protein 1c (SREBP1c), is one of the main features of non-alcoholic fatty liver disease (NAFLD). In the present study, we aimed to identify the relation between SREBP1c and autophagy mediated lipid droplet (LD) catabolism in oleic acid (OA) induced lipid accumulation. METHODS: Increased LD formation and SREBP1c induction were identified in hepatocytes (AML12 cells) following the OA administration. SREBP1c level was reduced through siRNA against SREBP1c. The amount and the size of LDs were determined by BODIPY, while protein and mRNA expressions were identified by immunoblotting and qRT-PCR, respectively. LD-lysosome colocalization was determined with immunofluorescence. RESULTS: Increased LD formation and SREBP1c levels were determined at 0.06 mM OA concentration. SREBP1c silencing reduced the number of LDs, while increasing mRNA levels of PPARα. On the other hand, SREBP1c silencing in non-OA and OA treated cells enhanced autophagy mediated LD catabolism. CONCLUSION: Our results implicate the effect of SREBP1c deficiency in modulating PPARα signaling and autophagy mediated LD catabolism against OA induced lipid accumulation.

6.
Biofactors ; 47(3): 403-413, 2021 May.
Article in English | MEDLINE | ID: mdl-34101924

ABSTRACT

Inflammation and apoptosis signaling are crucial steps in the progression from nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH). Alpha-tocopherol, the most active form of vitamin E, is an important modulator of signaling mechanisms, but its involvement to cholesterol-induced NASH pathogenesis remains poorly defined. Herein we have reported a novel effect of α-tocopherol in the transition from hepatic steatosis to NASH. High cholesterol diet alone (without α-tocopherol) in rabbits elevated NASH development as indicated by increased inflammatory response, apoptotic activity and liver fibrosis. Such elevation results from induction of signaling mechanisms since the expressions of IL1ß, phospho c-Jun/c-Jun ratio, JNK, caspase 9, CHOP and Bax were increased, and recruitment of macrophage, α-smooth muscle actin (α-SMA) and COL1A1 into the liver tissue were induced. Alpha-tocopherol supplementation inhibited inflammatory response, apoptosis and fibrosis development without affecting lipid accumulation in high cholesterol-induced NASH. Specifically, α-tocopherol lowered the inflammatory level as observed by reduced macrophage infiltration and JNK/c-Jun signaling. Lower inflammatory status co-occurred with the reduction of CHOP and Bax expressions as well as fibrosis-related COL1A1 and α-SMA levels. Taken together, α-tocopherol supplementation inhibits cholesterol-induced NASH development by lowering JNK/c-Jun/inflammation axis in addition to JNK/CHOP/apoptosis signaling, which might contribute to resistance against NAFLD/NASH transition.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Hypercholesterolemia/complications , Inflammation/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , alpha-Tocopherol/pharmacology , Animals , Dietary Supplements , Disease Models, Animal , Inflammation/etiology , Non-alcoholic Fatty Liver Disease/etiology , Oxidative Stress/drug effects , Rabbits
7.
Front Aging ; 2: 790702, 2021.
Article in English | MEDLINE | ID: mdl-35822008

ABSTRACT

Aging is a physiological process defined by decreased cellular and tissue functions. Reduced capacity of protein degradation is one of the important hallmarks of aging that may lead to misfolded protein accumulation and progressive loss of function in organ systems. Recognition of unfolded/misfolded protein aggregates via endoplasmic reticulum (ER) stress sensors activates an adaptive mechanism, the unfolded protein response (UPR). The initial step of UPR is defined by chaperone enhancement, ribosomal translation suppression, and misfolded protein degradation, while prolonged ER stress triggers apoptosis. MicroRNAs (miRNAs) are non-coding RNAs affecting various signaling pathways through degradation or translational inhibition of targeted mRNAs. Therefore, UPR and miRNA impairment in aging and age-related diseases is implicated in various studies. This review will highlight the recent insights in ER stress-miRNAs alterations during aging and age-related diseases, including metabolic, cardiovascular, and neurodegenerative diseases and several cancers.

8.
Oxid Med Cell Longev ; 2020: 9369524, 2020.
Article in English | MEDLINE | ID: mdl-32190179

ABSTRACT

Aging has been characterized with the accumulation of oxidized proteins, as a consequence of progressive decline in proteostasis capacity. Among others, proteasomal system is an efficient protein turnover complex to avoid aggregation of oxidized proteins. Heat shock protein 70 (HSP70) is another critical player that is involved in some key processes including the correct folding of misfolded proteins and targeting aggregated proteins to the proteasome for rapid degradation. The aim of this study was to determine the role of proteasomal system and heat shock proteins to maintain proteome balance during replicative senescence in mild hyperthermia conditions. Our results demonstrated that HSP40/70 machinery is induced by mild hyperthermia conditions independent from senescence conditions. Since HSP70 is largely responsible for the rapidly inducible cell protection following hyperthermia, the activation of "heat shock response" resulted in the elevation of HSP40/70 expressions as well as the proteasome activity. Interestingly, when HSP70 expression was inhibited, increased proteasomal activation was shown to be responsive to mild hyperthermia. Since HSP70 is involved in various stress-related pathways such as oxidative and endoplasmic reticulum stress, depletion of HSP70 expression may induce proteasomal degradation to maintain proteome balance of the cell. Thus, our data suggests that in mild heat stress conditions, molecular chaperone HSP70 plays an important role to avoid protein oxidation and aggregation; however, activities of proteasomal system are induced when HSP70 expression is depleted.


Subject(s)
Cellular Senescence , Fibroblasts/cytology , Fibroblasts/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Hyperthermia, Induced , Proteasome Endopeptidase Complex/metabolism , Benzhydryl Compounds/pharmacology , Cellular Senescence/genetics , Gene Silencing , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Humans , Male , Proteostasis , Pyrrolidinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Metabolism ; 106: 154205, 2020 05.
Article in English | MEDLINE | ID: mdl-32184090

ABSTRACT

BACKGROUND: Cardiovascular diseases (CVDs), with highest mortality and morbidity rates, are the major cause of death in the world. Due to the limited information on heart tissue changes, mediated by hypercholesterolemia, we planned to investigate molecular mechanisms of endoplasmic reticulum (ER) stress and related cell death in high cholesterol fed rabbit model and possible beneficial effects of α-tocopherol. METHODS: Molecular changes in rabbit heart tissue and cultured cardiomyocytes (H9c2 cells) were measured by western blotting, qRT-PCR, immunflouresence and flow cytometry experiments. Histological modifications were assessed by light and electron microscopes, while degradation of mitochondria was quantified through confocal microscope. RESULTS: Feeding rabbits 2% cholesterol diet for 8 weeks and treatment of cultured cardiomyocytes with 10 µg/mL cholesterol for 3 h induced excessive autophagic activity via IRE1/JNK pathway. While no change in ER-associated degradation (ERAD) and apoptotic cell death were determined, electron and confocal microscopy analyses in cholesterol supplemented rabbits revealed significant parameters of autophagic cell death, including cytoplasmic autophagosomes, autolysosomes and organelle loss in juxtanuclear area as well as mitochondria engulfment by autophagosome. Either inhibition of ER stress or JNK in cultured cardiomyocytes or α-tocopherol supplementation in rabbits could counteract the effects of cholesterol. CONCLUSION: Our findings underline the essential role of hypercholesterolemia in stimulating IRE1/JNK branch of ER stress response which then leads to autophagic cell death in heart tissue. Results also showed α-tocopherol as a promising regulator of autophagic cell death in cardiomyocytes.


Subject(s)
Autophagic Cell Death/drug effects , Autophagy/drug effects , Cholesterol/pharmacology , Heart/drug effects , Myocytes, Cardiac/drug effects , Animals , Cells, Cultured , Cholesterol/metabolism , Endoplasmic Reticulum Stress/drug effects , Heart/physiology , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/physiology , Protein Serine-Threonine Kinases/metabolism , Rabbits , Rats
10.
IUBMB Life ; 71(4): 507-515, 2019 04.
Article in English | MEDLINE | ID: mdl-30779288

ABSTRACT

Cardiovascular disease (CVD) is one of the major causes of morbidity and mortality, all around the world. Vitamin E is an important nutrient influencing key cellular and molecular mechanisms as well as gene expression regulation centrally involved in the prevention of CVD. Cell culture and animal studies have focused on the identification of vitamin E regulated signaling pathways and involvement on inflammation, lipid homeostasis, and atherosclerotic plaque stability. While some of these vitamin E functions were verified in clinical trials, some of the positive effects were not translated into beneficial outcomes in epidemiological studies. In recent years, the physiological metabolites of vitamin E, including the liver derived (long- and short-chain) metabolites and phosphorylated (α-, γ-tocopheryl phosphate) forms, have also provided novel mechanistic insight into CVD regulation that expands beyond the vitamin E precursor. It is certain that this emerging insight into the molecular and cellular action of vitamin E will help to design further studies, either in animal models or clinical trials, on the reduction of risk for CVDs. This review focuses on vitamin E-mediated preventive cardiovascular effects and discusses novel insights into the biology and mechanism of action of vitamin E metabolites in CVD. © 2019 IUBMB Life, 71(4):507-515, 2019.


Subject(s)
Cardiovascular Diseases/prevention & control , Cardiovascular Physiological Phenomena , Vitamin E/pharmacology , Vitamin E/physiology , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular System/metabolism , Humans
11.
Immunity ; 49(4): 666-677.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291029

ABSTRACT

Regulatory T (Treg) cell responses and apoptotic cell clearance (efferocytosis) represent critical arms of the inflammation resolution response. We sought to determine whether these processes might be linked through Treg-cell-mediated enhancement of efferocytosis. In zymosan-induced peritonitis and lipopolysaccharide-induced lung injury, Treg cells increased early in resolution, and Treg cell depletion decreased efferocytosis. In advanced atherosclerosis, where defective efferocytosis drives disease progression, Treg cell expansion improved efferocytosis. Mechanistic studies revealed the following sequence: (1) Treg cells secreted interleukin-13 (IL-13), which stimulated IL-10 production in macrophages; (2) autocrine-paracrine signaling by IL-10 induced Vav1 in macrophages; and (3) Vav1 activated Rac1 to promote apoptotic cell engulfment. In summary, Treg cells promote macrophage efferocytosis during inflammation resolution via a transcellular signaling pathway that enhances apoptotic cell internalization. These findings suggest an expanded role of Treg cells in inflammation resolution and provide a mechanistic basis for Treg-cell-enhancement strategies for non-resolving inflammatory diseases.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Macrophages/immunology , Phagocytosis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cells, Cultured , Humans , Inflammation/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Jurkat Cells , Lipopolysaccharides , Lung Diseases/chemically induced , Lung Diseases/immunology , Lung Diseases/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/metabolism , T-Lymphocytes, Regulatory/metabolism , Zymosan
12.
Oxid Med Cell Longev ; 2018: 8520746, 2018.
Article in English | MEDLINE | ID: mdl-30008986

ABSTRACT

Involvement of high cholesterol and oxidative stress in cardiovascular diseases is well studied, as it can be hypothesized that various products originated from lipid peroxidation, such as oxysterols, or affected protein expression might lead to cardiomyocyte damage followed by the pathological modifications. Although oxidation of excessive cholesterol to oxysterols in elevated stress conditions is identified by a number of studies, the role of a high cholesterol diet in regulating fatty acid and oxysterol accumulation, together with scavenger receptor mRNA levels, in the heart remains little investigated. Our study provides a detailed analysis of the changes in fatty acid, oxysterol, and scavenger receptor profiles and its relation with histological alterations in the heart tissue. We evaluated alterations of fatty acid composition, by the GC-MS method, while 4ß-, 25-, and 27-hydroxycholesterol and 7-ketocholesterol levels by means of LC-MS/MS in high cholesterol diet-fed rabbits. Additionally, a number of proteins related to lipid metabolism and scavenger receptor mRNA expressions were evaluated by Western blotting and RT-PCR. According to our in vivo results, a high cholesterol diet enhances a number of unsaturated fatty acids, oxysterols, and LXRα, in addition to CD36, CD68, CD204, and SR-F1 expressions while α-tocopherol supplementation decreases LXRα and SR expressions together with an increase in 27-hydroxycholesterol and ABCA1 levels. Our results indicated that the high cholesterol diet modulates proteins related to lipid metabolism, which might result in the malfunction of the heart and α-tocopherol shows its beneficial effects. We believe that this work will lead the generation of different theories in the development of heart diseases.


Subject(s)
Cholesterol/adverse effects , Myocardium/metabolism , Oxysterols/blood , Receptors, Scavenger/blood , Animals , Blotting, Western , CD36 Antigens/blood , Gas Chromatography-Mass Spectrometry , Hydroxycholesterols/blood , Ketocholesterols/blood , Lipid Metabolism/physiology , Lipid Peroxidation/physiology , Liver X Receptors/blood , Male , Oxidation-Reduction , Oxidative Stress/physiology , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Triglycerides/blood , alpha-Tocopherol/blood
13.
J Clin Invest ; 128(6): 2370-2375, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29708512

ABSTRACT

Emerging data suggest that hypercholesterolemia has stimulatory effects on adaptive immunity and that these effects can promote atherosclerosis and perhaps other inflammatory diseases. However, research in this area has relied primarily on inbred strains of mice whose adaptive immune system can differ substantially from that of humans. Moreover, the genetically induced hypercholesterolemia in these models typically results in plasma cholesterol levels that are much higher than those in most humans. To overcome these obstacles, we studied human immune system-reconstituted mice (hu-mice) rendered hypercholesterolemic by treatment with adeno-associated virus 8-proprotein convertase subtilisin/kexin type 9 (AAV8-PCSK9) and a high-fat/high-cholesterol Western-type diet (WD). These mice had a high percentage of human T cells and moderate hypercholesterolemia. Compared with hu-mice that had lower plasma cholesterol, the PCSK9-WD mice developed a T cell-mediated inflammatory response in the lung and liver. Human CD4+ and CD8+ T cells bearing an effector memory phenotype were significantly elevated in the blood, spleen, and lungs of PCSK9-WD hu-mice, whereas splenic and circulating regulatory T cells were reduced. These data show that moderately high plasma cholesterol can disrupt human T cell homeostasis in vivo. This process may not only exacerbate atherosclerosis, but also contribute to T cell-mediated inflammatory diseases in the hypercholesterolemia setting.


Subject(s)
Atherosclerosis/immunology , CD8-Positive T-Lymphocytes/immunology , Hypercholesterolemia/immunology , Proprotein Convertase 9/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Atherosclerosis/pathology , CD8-Positive T-Lymphocytes/pathology , Dependovirus , Humans , Hypercholesterolemia/pathology , Mice , T-Lymphocytes, Regulatory/pathology
14.
Biofactors ; 44(6): 588-596, 2018 Nov.
Article in English | MEDLINE | ID: mdl-28677864

ABSTRACT

Together with complex genetic and environmental factors, increased serum cholesterol and ox-LDL levels are considered as major triggering factors of atherosclerosis. Mononuclear cell infiltration to the arterial wall and uptake of ox-LDL, which is facilitated by CD36 receptor through an uncontrolled manner, play a key role in foam cell formation followed by atherogenesis development. The aim of this study was to analyze if CD36 expression in peripheral blood mononuclear cells reflect its aortic tissue level in hypercholesterolemia. In this study, CD36 protein expression was evaluated in aortic specimens of cholesterol or cholesterol plus Vitamin E treated animals in relation to the immunohistochemical analyses for the HNE-protein adducts, as well as for smooth muscle actin and vimentin. The CD36 mRNA expression was determined by RT-PCR in PBMC of hypercholesterolemic rabbits and hypercholesterolemic versus normocholesterolemic individuals. Immunohistochemistry findings revealed that smooth muscle actin, smooth muscle vimentin, HNE-protein conjugates, and CD36 protein expressions were significantly increased in aorta of hypercholesterolemic group where foam cells were present. High cholesterol diet significantly induced CD36 mRNA expression in both rabbit aorta and PBMCs, while positive correlation between aortic and PBMC CD36 expression has been found. In addition, consistent with the rabbit model, CD36 mRNA expression levels in human PBMCs were significantly higher in hypercholesterolemic patients than in normocholesterolemic individuals. Taken together, these results demonstrate that the CD36 mRNA levels of PBMCs could reflect the CD36 mRNA levels in aorta and could be used as a biomarker for diagnosis of atherosclerotic burden. © 2018 BioFactors, 44(6):588-596, 2018.


Subject(s)
Atherosclerosis/diagnosis , CD36 Antigens/genetics , Cholesterol/administration & dosage , Hypercholesterolemia/diagnosis , Leukocytes, Mononuclear/drug effects , Vitamin E/pharmacology , Actins/genetics , Actins/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Biomarkers/metabolism , Blood Glucose/metabolism , CD36 Antigens/metabolism , Cholesterol/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Gene Expression Regulation , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Rabbits , Triglycerides/blood , Vimentin/genetics , Vimentin/metabolism
15.
Redox Biol ; 12: 456-461, 2017 08.
Article in English | MEDLINE | ID: mdl-28319895

ABSTRACT

Endoplasmic reticulum (ER) is the major site of protein folding and calcium storage. Beside the role of ER in protein homeostasis, it controls the cholesterol production and lipid-membrane biosynthesis as well as surviving and cell death signaling mechanisms in the cell. It is well-documented that elevated plasma cholesterol induces adverse effects in cardiovascular diseases (CVDs), liver disorders, such as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatosis hepatitis (NASH), and metabolic diseases which are associated with oxidative and ER stress. Recent animal model and human studies have showed high cholesterol and ER stress as an emerging factors involved in the development of many metabolic diseases. In this review, we will summarize the crucial effects of hypercholesterolemia and ER stress response in the pathogenesis of CVDs, NAFLD/NASH, diabetes and obesity which are major health problems in western countries.


Subject(s)
Endoplasmic Reticulum/physiology , Hypercholesterolemia/complications , Metabolic Diseases/metabolism , Animals , Cholesterol/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , Hypercholesterolemia/metabolism , Metabolic Diseases/etiology , Signal Transduction
16.
Free Radic Biol Med ; 78: 30-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25452144

ABSTRACT

The folding process is an important step in protein synthesis for the functional shape or conformation of the protein. The endoplasmic reticulum (ER) is the main organelle for the correct folding procedure, which maintains the homeostasis of the organism. This process is normally well organized under unstressed conditions, whereas it may fail under oxidative and ER stress. The unfolded protein response (UPR) is a defense mechanism that removes the unfolded/misfolded proteins to prevent their accumulation, and two main degradation systems are involved in this defense, including the proteasome and autophagy. Cells decide which mechanism to use according to the type, severity, and duration of the stress. If the stress is too severe and in excess, the capacity of these degradation mechanisms, proteasomal degradation and autophagy, is not sufficient and the cell switches to apoptotic death. Because the accumulation of the improperly folded proteins leads to several diseases, it is important for the body to maintain this balance. Cardiovascular diseases are one of the important disorders related to failure of the UPR. Especially, protection mechanisms and the transition to apoptotic pathways have crucial roles in cardiac failure and should be highlighted in detailed studies to understand the mechanisms involved. This review is focused on the involvement of the proteasome, autophagy, and apoptosis in the UPR and the roles of these pathways in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/physiopathology , Endoplasmic Reticulum Stress , Animals , Humans , Unfolded Protein Response
17.
Redox Biol ; 2: 732-8, 2014.
Article in English | MEDLINE | ID: mdl-25009774

ABSTRACT

Atherosclerosis and its complications are major causes of death all over the world. One of the major risks of atherosclerosis is hypercholesterolemia. During atherosclerosis, oxidized low density lipoprotein (oxLDL) regulates CD36-mediated activation of c-jun amino terminal kinase-1 (JNK1) and modulates matrix metalloproteinase (MMP) induction which stimulates inflammation with an invasion of monocytes. Additionally, inhibition of proteasome leads to an accumulation of c-jun and phosphorylated c-jun and activation of activator protein-1 (AP-1) related increase of MMP expression. We have previously reported a significant increase in cluster of differentiation 36 (CD36) mRNA levels in hypercholesterolemic rabbits and shown that vitamin E treatment prevented the cholesterol induced increase in CD36 mRNA expression. In the present study, our aim is to identify the signaling molecules/transcription factors involved in the progression of atherosclerosis following CD36 activation in an in vivo model of hypercholesterolemic (induced by 2% cholesterol containing diet) rabbits. In this direction, proteasomal activities by fluorometry and c-jun, phospo c-jun, JNK1, MMP-9 expressions by quantitative RT-PCR and immunoblotting were tested in aortic tissues. The effects of vitamin E on these changes were also investigated in this model. As a result, c-jun was phosphorylated following decreased proteasomal degradation in hypercholesterolemic group. MMP-9 expression was also increased in cholesterol group rabbits contributing to the development of atherosclerosis. In addition, vitamin E showed its effect by decreasing MMP-9 levels and phosphorylation of c-jun.


Subject(s)
Mitogen-Activated Protein Kinase 8/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cholesterol/blood , Cholesterol, Dietary , Gene Expression/drug effects , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Male , Malondialdehyde/blood , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 8/genetics , Phosphorylation/drug effects , RNA, Messenger/metabolism , Rabbits , Signal Transduction/drug effects , Vitamin E/blood , Vitamin E/pharmacology
18.
Free Radic Biol Med ; 70: 174-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24583459

ABSTRACT

Atherosclerosis and associated cardiovascular complications such as stroke and myocardial infarction are major causes of morbidity and mortality. We have previously reported a significant increase in mRNA levels of the scavenger receptor CD36 in aortae of cholesterol-fed rabbits and shown that vitamin E treatment attenuated increased CD36 mRNA expression. In the present study, we further investigated the redox signaling pathways associated with protection against atherogenesis induced by high dietary cholesterol and correlated these with CD36 expression and the effects of vitamin E supplementation in a rabbit model. Male albino rabbits were assigned to either a control group fed with a low vitamin E diet alone or a test group fed with a low vitamin E diet containing 2% cholesterol in the absence or presence of daily intramuscular injections of vitamin E (50mg/kg). To elucidate the mechanisms by which vitamin E supplementation alters the effects of hypercholesterolemia in rabbit aortae, we measured peroxisome proliferator-activated receptor γ (PPARγ), ATP-binding cassette transporter A1 (ABCA1), and matrix metalloproteinase-1 (MMP-1) mRNA levels by quantitative RT-PCR and the expression of MMP-1, nuclear factor-erythroid 2-related factor 2 (Nrf2), and glutathione S-transferase α (GSTα) protein by immunoblotting. The increased MMP-1 and decreased GSTα expression observed suggests that a cholesterol-rich diet contributes to the development of atherosclerosis, whereas vitamin E supplementation affords protection by decreasing MMP-1 and increasing PPARγ, GSTα, and ABCA1 levels in aortae of rabbits fed a cholesterol-rich diet. Notably, protein expression of Nrf2, the antioxidant transcription factor, was increased in both the cholesterol-fed and the vitamin E-supplemented groups. Although Nrf2 activation can promote CD36-mediated cholesterol uptake by macrophages, the increased induction of Nrf2-mediated antioxidant genes is likely to contribute to decreased lesion progression. Thus, our study demonstrates that Nrf2 can mediate both pro- and antiatherosclerotic effects.


Subject(s)
Atherosclerosis/metabolism , NF-E2-Related Factor 2/metabolism , PPAR gamma/metabolism , Vitamin E/administration & dosage , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/pathology , Cholesterol, Dietary/administration & dosage , Diet, High-Fat , Gene Expression Regulation , Glutathione Transferase/biosynthesis , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/pathology , Isoenzymes/biosynthesis , Male , Matrix Metalloproteinase 1/biosynthesis , Rabbits , Signal Transduction/drug effects
19.
Free Radic Biol Med ; 75 Suppl 1: S24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-26461313

ABSTRACT

Hypercholesterolemia is the major risk factor for the development of atherosclerosis and vitamin E is suggested to have a preventive role in this process (1), although the mechanism of action still remains unclear.The ubiquitin-proteasome system (UPS) may in?uence atherosclerosis by affecting disease-relevant cellular processes such as apoptosis, proliferation, and differentiation, or by affecting cellular stress responses and/or adaptive phenomena, such as ER stress, in?ammation, and redox homeostasis (2). NF-E2-related factor 2 (Nrf2) is a transcription factor that controls the expression of phase II detoxi?cation and antioxidant genes. Nrf2 signaling has additionally been shown to upregulate the expression of the proteasome catalytic subunits (3). In the present study, we investigated the role of Nrf2 pathway on oxidative and ER stress conditions induced by cholesterol diet and the effects of vitamin E on related signaling pathways in in vivo model of atherosclerosis. All experimental procedures were approved by the Marmara University Ethics Committee. Twenty-one male albino rabbits (23 months old) were assigned randomly to four groups fed for 8 weeks: (i) vitamin E deficient diet, (ii) vitamin E deficient diet containing 2% cholesterol, and (iii) vitamin E deficient diet containing 2% cholesterol with daily intramuscular injections of vitamin E (50mg/kg), (iv) vitamin E deficient diet with daily intramuscular injections of vitamin E (50mg/kg). In order to elucidate in vivo role of oxidative stress and ER stress in cardiovascular system of hypercholesterolemic rabbits, we investigated serum levels of cholesterol, MDA and vitamin E and Nrf2, GST-1, GRP78, GRP94, PERK, IRE1 protein levels and the proteasomal activity in aortic tissues will be discussed.

20.
J Proteomics ; 89: 238-54, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23811050

ABSTRACT

Since the proteins are involved in many physiological processes in the organisms, modifications of proteins have important outcomes. Protein modifications are classified in several ways and oxidative stress related ones take a wide place. Aging is characterized by the accumulation of oxidized proteins and decreased degradation of these proteins. On the other hand protein turnover is an important regulatory mechanism for the control of protein homeostasis. Heat shock proteins are a highly conserved family of proteins in the various cells and organisms whose expressions are highly inducible during stress conditions. These proteins participate in protein assembly, trafficking, degradation and therefore play important role in protein turnover. Although the entire functions of each heat shock protein are still not completely investigated, these proteins have been implicated in the processes of protection and repair of stress-induced protein damage. This study has focused on the heat stress related carbonylated proteins, as a marker of oxidative protein modification, in young and senescent fibroblasts. The results are discussed with reference to potential involvement of induced heat shock proteins. This article is part of a Special Issue entitled: Protein Modifications. BIOLOGICAL SIGNIFICANCE: Age-related protein modifications, especially protein carbonylation take a wide place in the literature. In this direction, to highlight the role of heat shock proteins in the oxidative modifications may bring a new aspect to the literature. On the other hand, identified carbonylated proteins in this study confirm the importance of folding process in the mitochondria which will be further analyzed in detail.


Subject(s)
Cellular Senescence/physiology , Fibroblasts/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Protein Carbonylation/physiology , Protein Processing, Post-Translational/physiology , Cells, Cultured , Fibroblasts/cytology , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...