1
|
Phair IR, Sovakova M, Alqurashi N, Nisr RB, McNeilly AD, Lamont D, Rena G. In-depth proteomic profiling identifies potentiation of the LPS response by 7-ketocholesterol. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100285. [PMID: 39991505 PMCID: PMC11847031 DOI: 10.1016/j.jmccpl.2025.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
In patients with stable coronary artery disease, plasma levels of 7-ketocholesterol (7-KC), found at high levels in atherosclerotic lesions, predict risk of incident heart failure dose dependently, potentially contributing to disease aetiology. Previous studies demonstrated that 7-KC can elicit effects on macrophage function; however, effects of 7-KC on the macrophage proteome have not been studied systematically. Here we used quantitative mass spectrometry to establish the effect of 7-KC on the mouse macrophage proteome. 7-KC independently mediated dynamic changes, including on atherogenic/M1 markers, cholesterol metabolism, biosynthesis and transport, as well as nutrient transport more broadly. These changes were however insufficient alone to drive changes in cytokine and chemokine secretion. Rather, they prime the macrophage, potentiating LPS-stimulated TNF alpha secretion and key pro-inflammatory enzymes. Our results indicate that 7-KC has independent metabolic effects on the macrophage; however, effects on the immune system are primarily due to the changes in metabolism priming the response to an inflammatory stimulus. Earlier findings from CANTOS and the recent FDA approval of colchicine highlight that inflammation is a viable target for cardiovascular disease; however, it is currrently unclear which will be the best anti-inflammatory targets to pursue in the future. In this context, our findings suggest that drugs targeting atherogenic markers induced by 7-KC might be well tolerated, as they will not necessarily be expected to be immunosuppressive.
Collapse
Affiliation(s)
- Iain R. Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Raid B. Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Alison D. McNeilly
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| |
Collapse
|
2
|
Liu Y, Qin J, Li X, Wu G. Oxysterols in tumor immune microenvironment (TIME). J Steroid Biochem Mol Biol 2025; 245:106634. [PMID: 39551164 DOI: 10.1016/j.jsbmb.2024.106634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Oxysterols are compounds generated through oxidative reactions involving cholesterol and other steroid molecules. They play a crucial role in the tumor immune microenvironment by interacting with molecules such as the cell membrane receptor EBI2 and nuclear receptors like LXR and PXR. This interaction regulates immune cell signaling pathways, affecting proliferation, apoptosis, migration, and invasion in tumor-related processes. Activating these receptors alters the function and behavior of immune cells-such as macrophages, T cells, and dendritic cells-within the tumor microenvironment, thus promoting or inhibiting tumor development. Certain oxidized steroids can increase both the number and activation of infiltrating T cells, synergizing with anti-PD-1 to enhance anti-tumor efficacy. An in-depth study of the biological mechanisms of oxidized sterols will not only enhance our understanding of the complexity of the tumor immune microenvironment but may also reveal new therapeutic targets, providing innovative strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuanxin Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Jie Qin
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
3
|
Li W, Pang Y, Jin K, Wang Y, Wu Y, Luo J, Xu W, Zhang X, Xu R, Wang T, Jiao L. Membrane contact sites orchestrate cholesterol homeostasis that is central to vascular aging. WIREs Mech Dis 2023; 15:e1612. [PMID: 37156598 DOI: 10.1002/wsbm.1612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/12/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Chronological age causes structural and functional vascular deterioration and is a well-established risk factor for the development of cardiovascular diseases, leading to more than 40% of all deaths in the elderly. The etiology of vascular aging is complex; a significant impact arises from impaired cholesterol homeostasis. Cholesterol level is balanced through synthesis, uptake, transport, and esterification, the processes executed by multiple organelles. Moreover, organelles responsible for cholesterol homeostasis are spatially and functionally coordinated instead of isolated by forming the membrane contact sites. Membrane contact, mediated by specific protein-protein interaction, pulls opposing organelles together and creates the hybrid place for cholesterol transfer and further signaling. The membrane contact-dependent cholesterol transfer, together with the vesicular transport, maintains cholesterol homeostasis and has intimate implications in a growing list of diseases, including vascular aging-related diseases. Here, we summarized the latest advances regarding cholesterol homeostasis by highlighting the membrane contact-based regulatory mechanism. We also describe the downstream signaling under cholesterol homeostasis perturbations, prominently in cholesterol-rich conditions, stimulating age-dependent organelle dysfunction and vascular aging. Finally, we discuss potential cholesterol-targeting strategies for therapists regarding vascular aging-related diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yiyun Pang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Multi-omics analysis identifies potential mechanisms by which high glucose accelerates macrophage foaming. Mol Cell Biochem 2023; 478:665-678. [PMID: 36029453 DOI: 10.1007/s11010-022-04542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022]
Abstract
Atherosclerotic morbidity is significantly higher in the diabetic population. Hyperglycemia, a typical feature of diabetes, has been proven to accelerate foam cell formation. However, the molecular mechanisms behind this process remain unclear. In this study, LPS and IFN-γ were used to convert THP-1-derived macrophages into M1 macrophages, which were then activated with ox-LDL in either high glucose or normal condition. We identified lipids within macrophages by Oil red O staining and total cholesterol detection. The genes involved in lipid absorption, efflux, inflammation, and metabolism were analyzed using qRT-PCR. The mechanisms of high glucose-induced foam cell formation were further investigated through metabolomics and transcriptomics analysis. We discovered that high glucose speed up lipid accumulation in macrophages (both lipid droplets and total cholesterol increased), diminished lipid efflux (ABCG1 down-regulation), and aggravated inflammation (IL1B and TNF up-regulation). Following multi-omics analysis, it was determined that glucose altered the metabolic and transcriptional profiles of macrophages, identifying 392 differently expressed metabolites and 293 differentially expressed genes, respectively. Joint pathway analysis suggested that glucose predominantly disrupted the glycerolipid, glycerophospholipid, and arachidonic acid metabolic pathways in macrophages. High glucose in the glyceride metabolic pathway, for instance, suppressed the transcription of triglyceride hydrolase (LIPG and LPL), causing cells to deposit excess triglycerides into lipid droplets and encouraging foam cell formation. More importantly, high glucose triggered the accumulation of pro-atherosclerotic lipids (7-ketocholesterol, lysophosphatidylcholine, and glycerophosphatidylcholine). In conclusion, this work elucidated mechanisms of glucose-induced foam cell formation via a multi-omics approach.
Collapse
|
5
|
Ouyang J, Xiao Y, Ren Q, Huang J, Zhou Q, Zhang S, Li L, Shi W, Chen Z, Wu L. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022; 11:cells11182882. [PMID: 36139457 PMCID: PMC9496706 DOI: 10.3390/cells11182882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 01/16/2023] Open
Abstract
7-Ketocholesterol (7KC) is one of the oxysterols produced by the auto-oxidation of cholesterol during the dysregulation of cholesterol metabolism which has been implicated in the pathological development of osteoporosis (OP). Oxiapoptophagy involving oxidative stress, autophagy, and apoptosis can be induced by 7KC. However, whether 7KC produces negative effects on MC3T3-E1 cells by stimulating oxiapoptophagy is still unclear. In the current study, 7KC was found to significantly decrease the cell viability of MC3T3-E1 cells in a concentration-dependent manner. In addition, 7KC decreased ALP staining and mineralization and down-regulated the protein expression of OPN and RUNX2, inhibiting osteogenic differentiation. 7KC significantly stimulated oxidation and induced autophagy and apoptosis in the cultured MC3T3-E1 cells. Pretreatment with the anti-oxidant acetylcysteine (NAC) could effectively decrease NOX4 and MDA production, enhance SOD activity, ameliorate the expression of autophagy-related factors, decrease apoptotic protein expression, and increase ALP, OPN, and RUNX2 expression, compromising 7KC-induced oxiapoptophagy and osteogenic differentiation inhibition in MC3T3-E1 cells. In summary, 7KC may induce oxiapoptophagy and inhibit osteogenic differentiation in the pathological development of OP.
Collapse
Affiliation(s)
- Jing Ouyang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China
| | - Yaosheng Xiao
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Qingluo Zhou
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Shanshan Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Linfu Li
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Weimei Shi
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
- Correspondence:
| |
Collapse
|
6
|
de Freitas FA, Levy D, Reichert CO, Cunha-Neto E, Kalil J, Bydlowski SP. Effects of Oxysterols on Immune Cells and Related Diseases. Cells 2022; 11:cells11081251. [PMID: 35455931 PMCID: PMC9031443 DOI: 10.3390/cells11081251] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
Oxysterols are the products of cholesterol oxidation. They have a wide range of effects on several cells, organs, and systems in the body. Oxysterols also have an influence on the physiology of the immune system, from immune cell maturation and migration to innate and humoral immune responses. In this regard, oxysterols have been involved in several diseases that have an immune component, from autoimmune and neurodegenerative diseases to inflammatory diseases, atherosclerosis, and cancer. Here, we review data on the participation of oxysterols, mainly 25-hydroxycholesterol and 7α,25-dihydroxycholesterol, in the immune system and related diseases. The effects of these oxysterols and main oxysterol receptors, LXR and EBI2, in cells of the immune system (B cells, T cells, macrophages, dendritic cells, oligodendrocytes, and astrocytes), and in immune-related diseases, such as neurodegenerative diseases, intestinal diseases, cancer, respiratory diseases, and atherosclerosis, are discussed.
Collapse
Affiliation(s)
- Fábio Alessandro de Freitas
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil; (F.A.d.F.); (D.L.); (C.O.R.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil; (F.A.d.F.); (D.L.); (C.O.R.)
| | - Cadiele Oliana Reichert
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil; (F.A.d.F.); (D.L.); (C.O.R.)
| | - Edecio Cunha-Neto
- Laboratory of Clinical Immunology and Allergy (LIM60), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil;
- National Institute of Science and Technology for Investigation in Immunology-III/INCT, Sao Paulo 05403-000, SP, Brazil;
| | - Jorge Kalil
- National Institute of Science and Technology for Investigation in Immunology-III/INCT, Sao Paulo 05403-000, SP, Brazil;
- Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403-900, SP, Brazil; (F.A.d.F.); (D.L.); (C.O.R.)
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, RJ, Brazil
- Correspondence:
| |
Collapse
|
7
|
de Freitas FA, Levy D, Zarrouk A, Lizard G, Bydlowski SP. Impact of Oxysterols on Cell Death, Proliferation, and Differentiation Induction: Current Status. Cells 2021; 10:cells10092301. [PMID: 34571949 PMCID: PMC8468221 DOI: 10.3390/cells10092301] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol produced by enzymatic activity or non-enzymatic pathways (auto-oxidation). The oxidation processes lead to the synthesis of about 60 different oxysterols. Several oxysterols have physiological, pathophysiological, and pharmacological activities. The effects of oxysterols on cell death processes, especially apoptosis, autophagy, necrosis, and oxiapoptophagy, as well as their action on cell proliferation, are reviewed here. These effects, also observed in several cancer cell lines, could potentially be useful in cancer treatment. The effects of oxysterols on cell differentiation are also described. Among them, the properties of stimulating the osteogenic differentiation of mesenchymal stem cells while inhibiting adipogenic differentiation may be useful in regenerative medicine.
Collapse
Affiliation(s)
- Fábio Alessandro de Freitas
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
| | - Amira Zarrouk
- Faculty of Medicine, University of Monastir, LR12ES05, Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Monastir, Tunisia & Faculty of Medicine, University of Sousse, Sousse 5000, Tunisia;
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA, University of Bourgogne Franche-Comté, Institut National de la Santé et de la Recherche Médicale—Inserm, 7270 Dijon, France;
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil (D.L.)
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
- Correspondence:
| |
Collapse
|
8
|
Oxysterol species generated by auto-oxidation in subclinical hypothyroidism. Clin Biochem 2021; 93:73-79. [PMID: 33861988 DOI: 10.1016/j.clinbiochem.2021.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Auto-oxidized oxysterols are implicated in the pathogenesis of various chronic diseases. Their concentrations are indicators of oxidative stress in vivo and associated with atherosclerosis. Subclinical hypothyroidism is related with cardiac diseases and oxidative stress, but the exact mechanisms underlying these associations are not clear yet. OBJECTIVE To investigate the auto-oxidized oxysterols, 7-ketocholesterol (7-KC) and cholestane-3β,5α,6β-triol (chol-triol), in patients with subclinical hypothyroidism, as well as to evaluate the impact of restoring euthyroidism on oxysterol concentrations. METHODS In this prospective observational study, 64 patients with newly diagnosed autoimmune thyroiditis (41 with subclinical hypothyroidism and 23 euthyroidism), and 45 healthy controls were enrolled. Age, gender, and body mass index were matched among patient groups and healthy controls. Anthropometric measurements were obtained and fasting plasma 7-ketocholesterol and cholestane-3β,5α,6β-triol concentrations were measured by using liquid chromatography coupled with tandem mass spectrometry. Levothyroxine was then administered to all patients with subclinical-hypothyroidism. After three months, measurements of the oxysterols and serum cholesterols from the patients who have become euthyroid were repeated. RESULTS Concentrations of 7-ketocholesterol and cholestane-3β,5α,6β-triol were significantly higher in patients with subclinical-hypothyroidism when compared to both euthyroid patients and healthy controls (p < 0.001 for both oxysterols). After restoration of euthyroidism, concentrations of 7-ketocholesterol and cholestane-3β,5α,6β-triol decreased significantly and reached similar concentrations observed in healthy controls (p < 0.001 for both oxysterols). CONCLUSIONS Auto-oxidized oxysterol species are higher in patients with mild thyroid dysfunction, and supported the rationale for treating subclinical-hypothyroidism.
Collapse
|
9
|
Paraoxonase Role in Human Neurodegenerative Diseases. Antioxidants (Basel) 2020; 10:antiox10010011. [PMID: 33374313 PMCID: PMC7824310 DOI: 10.3390/antiox10010011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The human body has biological redox systems capable of preventing or mitigating the damage caused by increased oxidative stress throughout life. One of them are the paraoxonase (PON) enzymes. The PONs genetic cluster is made up of three members (PON1, PON2, PON3) that share a structural homology, located adjacent to chromosome seven. The most studied enzyme is PON1, which is associated with high density lipoprotein (HDL), having paraoxonase, arylesterase and lactonase activities. Due to these characteristics, the enzyme PON1 has been associated with the development of neurodegenerative diseases. Here we update the knowledge about the association of PON enzymes and their polymorphisms and the development of multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD).
Collapse
|
10
|
Kuo X, Herr DR, Ong WY. Anti-inflammatory and Cytoprotective Effect of Clinacanthus nutans Leaf But Not Stem Extracts on 7-Ketocholesterol Induced Brain Endothelial Cell Injury. Neuromolecular Med 2020; 23:176-183. [PMID: 33085066 DOI: 10.1007/s12017-020-08621-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/08/2020] [Indexed: 12/16/2022]
Abstract
Clinacanthus nutans (Lindau) (C. nutans) has diverse uses in traditional herbal medicine for treating skin rashes, insect and snake bites, lesions caused by herpes simplex virus, diabetes mellitus and gout in Singapore, Malaysia, Indonesia, Thailand and China. We previously showed that C. nutans has the ability to modulate the induction of cytosolic phospholipase A2 (cPLA2) expression in SH-SY5Y cells through the inhibition of histone deacetylases (HDACs). In the current study, we elucidated the effect of C. nutans on the hCMEC/D3 human brain endothelial cell line. Endothelial cells are exposed to high levels of the cholesterol oxidation product, 7-ketocholesterol (7KC), in patients with cardiovascular disease and diabetes, and this process is thought to mediate pathological inflammation. 7KC induced a dose-dependent loss of hCMEC/D3 cell viability, and such damage was significantly inhibited by C. nutans leaf extracts but not stem extracts. 7KC also induced a marked increase in mRNA expression of pro-inflammatory cytokines, IL-1β IL-6, IL-8, TNF-α and cyclooxygenase-2 (COX-2) in brain endothelial cells, and these increases were significantly inhibited by C. nutans leaf but not stem extracts. HPLC analyses showed that leaf extracts have a markedly different chemical profile compared to stem extracts, which might explain their different effects in counteracting 7KC-induced inflammation. Further study is necessary to identify the putative phytochemicals in C. nutans leaves that have anti-inflammatory properties.
Collapse
Affiliation(s)
- Xuan Kuo
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Deron R Herr
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
11
|
Effect of Ergothioneine on 7-Ketocholesterol-Induced Endothelial Injury. Neuromolecular Med 2020; 23:184-198. [PMID: 33067719 PMCID: PMC7567423 DOI: 10.1007/s12017-020-08620-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
Ergothioneine (ET) is a naturally occurring antioxidant that is synthesized by non-yeast fungi and certain bacteria. ET is not synthesized by animals, including humans, but is avidly taken up from the diet, especially from mushrooms. In the current study, we elucidated the effect of ET on the hCMEC/D3 human brain endothelial cell line. Endothelial cells are exposed to high levels of the cholesterol oxidation product, 7-ketocholesterol (7KC), in patients with cardiovascular disease and diabetes, and this process is thought to mediate pathological inflammation. 7KC induces a dose-dependent loss of cell viability and an increase in apoptosis and necrosis in the endothelial cells. A relocalization of the tight junction proteins, zonula occludens-1 (ZO-1) and claudin-5, towards the nucleus of the cells was also observed. These effects were significantly attenuated by ET. In addition, 7KC induces marked increases in the mRNA expression of pro-inflammatory cytokines, IL-1β IL-6, IL-8, TNF-α and cyclooxygenase-2 (COX2), as well as COX2 enzymatic activity, and these were significantly reduced by ET. Moreover, the cytoprotective and anti-inflammatory effects of ET were significantly reduced by co-incubation with an inhibitor of the ET transporter, OCTN1 (VHCL). This shows that ET needs to enter the endothelial cells to have a protective effect and is unlikely to act via extracellular neutralizing of 7KC. The protective effect on inflammation in brain endothelial cells suggests that ET might be useful as a nutraceutical for the prevention or management of neurovascular diseases, such as stroke and vascular dementia. Moreover, the ability of ET to cross the blood-brain barrier could point to its usefulness in combatting 7KC that is produced in the CNS during neuroinflammation, e.g. after excitotoxicity, in chronic neurodegenerative diseases, and possibly COVID-19-related neurologic complications.
Collapse
|
12
|
Civra A, Colzani M, Cagno V, Francese R, Leoni V, Aldini G, Lembo D, Poli G. Modulation of cell proteome by 25-hydroxycholesterol and 27-hydroxycholesterol: A link between cholesterol metabolism and antiviral defense. Free Radic Biol Med 2020; 149:30-36. [PMID: 31525455 PMCID: PMC7126780 DOI: 10.1016/j.freeradbiomed.2019.08.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 12/11/2022]
Abstract
Physiological cholesterol metabolism implies the generation of a series of oxidized derivatives, whose oxysterols are by far the most investigated ones for their potential multifaceted involvement in human pathophysiology. In this regard, noteworthy is the broad antiviral activity displayed by defined side chain oxysterols, in particular 25-hydroxycholesterol (25HC) and 27-hydroxycholesterol (27HC). Although their antiviral mechanism(s) may vary depending on virus/host interaction, these oxysterols share the common feature to hamper viral replication by interacting with cellular proteins. Here reported is the first analysis of the modulation of a cell proteome by these two oxysterols, that, besides yielding additional clues about their potential involvement in the regulation of sterol metabolism, provides novelinsights about the mechanism underlying the inhibition of virus entry and trafficking within infected cells. We show here that both 25HC and 27HC can down-regulate the junction adhesion molecule-A (JAM-A) and the cation independent isoform of mannose-6-phosphate receptor (MPRci), two crucial molecules for the replication of all those viruses that exploit adhesion molecules and the endosomal pathway to enter and diffuse within target cells.
Collapse
Affiliation(s)
- Andrea Civra
- Department of Clinical and Biological Sciences, University of Torino at San Luigi Hospital, Orbassano, Torino, Italy.
| | - Mara Colzani
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Valeria Cagno
- Department of Molecular Microbiology, University of Geneva, Geneva, Switzerland.
| | - Rachele Francese
- Department of Clinical and Biological Sciences, University of Torino at San Luigi Hospital, Orbassano, Torino, Italy.
| | - Valerio Leoni
- Department of Laboratory Medicine, University of Milano-Bicocca, School of Medicine, Hospital of Desio, Milano, Italy.
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy.
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Torino at San Luigi Hospital, Orbassano, Torino, Italy.
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino at San Luigi Hospital, Orbassano, Torino, Italy.
| |
Collapse
|
13
|
Pariente A, Peláez R, Pérez-Sala Á, Larráyoz IM. Inflammatory and cell death mechanisms induced by 7-ketocholesterol in the retina. Implications for age-related macular degeneration. Exp Eye Res 2019; 187:107746. [DOI: 10.1016/j.exer.2019.107746] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
|
14
|
Levy D, Reichert CO, Bydlowski SP. Paraoxonases Activities and Polymorphisms in Elderly and Old-Age Diseases: An Overview. Antioxidants (Basel) 2019; 8:antiox8050118. [PMID: 31052559 PMCID: PMC6562914 DOI: 10.3390/antiox8050118] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 12/14/2022] Open
Abstract
Aging is defined as the accumulation of progressive organ dysfunction. There is much evidence linking the involvement of oxidative stress in the pathogenesis of aging. With increasing age, susceptibility to the development of diseases related to lipid peroxidation and tissue injury increases, due to chronic inflammatory processes, and production of reactive oxygen species (ROS) and free radicals. The paraoxonase (PON) gene family is composed of three members (PON1, PON2, PON3) that share considerable structural homology and are located adjacently on chromosome 7 in humans. The most studied member product is PON1, a protein associated with high-density lipoprotein with paraoxonase/esterase activity. Nevertheless, all the three proteins prevent oxidative stress. The major aim of this review is to highlight the importance of the role of PON enzymes in the aging process, and in the development of the main diseases present in the elderly: cardiovascular disease, diabetes mellitus, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Débora Levy
- Genetic and Molecular Hematology Laboratory (LIM31), Hospital das Clínicas, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 05419-000, SP, Brazil.
| | - Cadiele Oliana Reichert
- Genetic and Molecular Hematology Laboratory (LIM31), Hospital das Clínicas, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 05419-000, SP, Brazil.
| | - Sérgio Paulo Bydlowski
- Genetic and Molecular Hematology Laboratory (LIM31), Hospital das Clínicas, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 05419-000, SP, Brazil.
- Center of Innovation and Translacional Medicine (CIMTRA), Department of Medicine, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 05419-000, SP, Brazil.
- Instituto Nacional de Ciencia e Tecnologia em Medicina Regenerativa (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, RJ, Brazil.
| |
Collapse
|
15
|
Samadi A, Gurlek A, Sendur SN, Karahan S, Akbiyik F, Lay I. Oxysterol species: reliable markers of oxidative stress in diabetes mellitus. J Endocrinol Invest 2019; 42:7-17. [PMID: 29564756 DOI: 10.1007/s40618-018-0873-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE To assess the plasma oxysterol species 7-ketocholesterol (7-Kchol) and cholestane-3β,5α,6β-triol (chol-triol) as biomarkers of oxidative stress in type 1 and type 2 diabetes mellitus (DM). METHODS In total, 26 type 1 and 80 type 2 diabetes patients, along with 205 age- and gender-matched healthy controls, were included in this study. Oxysterols were quantified by liquid chromatography coupled with tandem mass spectrometry and N,N-dimethylglycine derivatization. Correlations between oxysterols and clinical/biochemical characteristics of the diabetes patients, and factors affecting 7-Kchol and chol-triol, were also determined. RESULTS Plasma 7-Kchol and chol-triol levels were significantly higher in type 1 and type 2 diabetes patients compared to healthy controls (P < 0.001). Significant positive correlations were observed between oxysterol levels and levels of glycated hemoglobin (HbA1c), glucose, serum total cholesterol, low-density lipoprotein, very-low-density lipoprotein, and triglycerides, as well as the number of coronary risk factors. Statins, oral hypoglycemic agents, and antihypertensive agents reduced the levels of oxysterols in type 2 diabetes patients. Statin use, HbA1c levels, and the number of coronary risk factors accounted for 98.8% of the changes in 7-Kchol levels, and total cholesterol, smoking status, and the number of coronary risk factors accounted for 77.3% of the changes in chol-triol levels in type 2 diabetes patients. CONCLUSIONS Plasma oxysterol levels in DM, and particularly type 2 DM, may yield complementary information regarding oxidative stress for the clinical follow-up of diabetes patients, especially those with coronary risk factors.
Collapse
Affiliation(s)
- A Samadi
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - A Gurlek
- Department of Internal Medicine, Endocrinology Unit, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - S N Sendur
- Department of Internal Medicine, Endocrinology Unit, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - S Karahan
- Department of Biostatistics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - F Akbiyik
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - I Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
- Clinical Pathology Laboratory, Hacettepe University Hospitals, 06100, Ankara, Turkey.
| |
Collapse
|
16
|
Levy D, de Melo TC, Oliveira BA, Paz JL, de Freitas FA, Reichert CO, Rodrigues A, Bydlowski SP. 7-Ketocholesterol and cholestane-triol increase expression of SMO and LXRα signaling pathways in a human breast cancer cell line. Biochem Biophys Rep 2018; 19:100604. [PMID: 31463370 PMCID: PMC6709374 DOI: 10.1016/j.bbrep.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Oxysterols are 27-carbon oxidation products of cholesterol metabolism. Oxysterols possess several biological actions, including the promotion of cell death. Here, we examined the ability of 7-ketocholesterol (7-KC), cholestane-3β-5α-6β-triol (triol), and a mixture of 5α-cholestane-3β,6β-diol and 5α-cholestane-3β,6α-diol (diol) to promote cell death in a human breast cancer cell line (MDA-MB-231). We determined cell viability, after 24-h incubation with oxysterols. These oxysterols promoted apoptosis. At least part of the observed effects promoted by 7-KC and triol arose from an increase in the expression of the sonic hedgehog pathway mediator, smoothened. However, this increased expression was apparently independent of sonic hedgehog expression, which did not change. Moreover, these oxysterols led to increased expression of LXRα, which is involved in cellular cholesterol efflux, and the ATP-binding cassette transporters, ABCA1 and ABCG1. Diols did not affect these pathways. These results suggested that the sonic hedgehog and LXRα pathways might be involved in the apoptotic process promoted by 7-KC and triol.
Collapse
Affiliation(s)
- Debora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Thatiana Correa de Melo
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Beatriz A. Oliveira
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Jessica L. Paz
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Fabio A. de Freitas
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Cadiele O. Reichert
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | | | - Sergio P. Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Instituto Nacional de Ciencia e Tecnologia em Medicina Regenerativa (INCT-Regenera), CNPq, Brazil
- Correspondence to: Department of Hematology, Faculdade de Medicina, Universidade de Sao Paulo, Av.Dr. Enéas de Carvalho Aguiar,155, 1st floor, room 43, 05403-000 São Paulo, SP, Brazil.
| |
Collapse
|
17
|
Oxysterols selectively promote short-term apoptosis in tumor cell lines. Biochem Biophys Res Commun 2018; 505:1043-1049. [DOI: 10.1016/j.bbrc.2018.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023]
|
18
|
Brahmi F, Vejux A, Sghaier R, Zarrouk A, Nury T, Meddeb W, Rezig L, Namsi A, Sassi K, Yammine A, Badreddine I, Vervandier-Fasseur D, Madani K, Boulekbache-Makhlouf L, Nasser B, Lizard G. Prevention of 7-ketocholesterol-induced side effects by natural compounds. Crit Rev Food Sci Nutr 2018; 59:3179-3198. [DOI: 10.1080/10408398.2018.1491828] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fatiha Brahmi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Randa Sghaier
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Wiem Meddeb
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- LMMA/IPEST, Faculty of Science, University of Carthage, Bizerte, Tunisia
| | - Leila Rezig
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- ESIAT, Lab. Conservation et Valorisation des Aliments, Tunis, Tunisia
| | - Amira Namsi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- University Tunis El Manar, Faculty of Science of Tunis, Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Khouloud Sassi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Onco-Hematology, Faculty de Medicine of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Aline Yammine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Bioactive Molecules Research Lab, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Iham Badreddine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. ‘Valorisation des Ressources Naturelles et Environnement’, Université Ibn Zohr, Taroudant, Morocco
| | | | - Khodir Madani
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Lila Boulekbache-Makhlouf
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Boubker Nasser
- Lab. Neuroscience and Biochemistry, Université Hassan 1er, Settat, Morocco
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
19
|
Testa G, Rossin D, Poli G, Biasi F, Leonarduzzi G. Implication of oxysterols in chronic inflammatory human diseases. Biochimie 2018; 153:220-231. [DOI: 10.1016/j.biochi.2018.06.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/07/2018] [Indexed: 12/18/2022]
|
20
|
Sottero B, Leonarduzzi G, Testa G, Gargiulo S, Poli G, Biasi F. Lipid Oxidation Derived Aldehydes and Oxysterols Between Health and Disease. EUR J LIPID SCI TECH 2018. [DOI: 10.1002/ejlt.201700047] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| |
Collapse
|
21
|
Dias IHK, Milic I, Lip GYH, Devitt A, Polidori MC, Griffiths HR. Simvastatin reduces circulating oxysterol levels in men with hypercholesterolaemia. Redox Biol 2018; 16:139-145. [PMID: 29501047 PMCID: PMC5952874 DOI: 10.1016/j.redox.2018.02.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 11/17/2022] Open
Abstract
Oxysterols (OHC) are biologically active cholesterol metabolites circulating in plasma that may be formed enzymatically (e.g. 24S-OHC, 25-OHC and 27-OHC) or by autoxidative mechanisms (e.g. 7-ketocholesterol, 7β-OHC and 25-OHC). Oxysterols are more soluble than cholesterol and are reported to exert inflammatory, cytoprotective and apoptotic effects according to concentration and species. Esterified oxysterols have been analysed in people with dementia and cardiovascular diseases although there is no consistent relationship between oxysterol esters and disease. However, oxysterol esters are held in lipoprotein core and may not relate to the concentration and activity of plasma free oxysterols. Methodological limitations have challenged the analysis of free oxysterols to date. We have developed a fast, sensitive and specific quantitative LC-MS/MS, multiple reaction monitoring (MRM) method to target five oxysterols in human plasma with analyte recoveries between 72% and 82% and sensitivities between 5 and 135 pg/ml. A novel method was used to investigate the hypothesis that simvastatin may reduce the concentrations of specific plasma free oxysterols in hypercholesterolaemia. Twenty healthy male volunteers were recruited (aged 41-63 years); ten were asymptomatic with high plasma cholesterol > 6.5 mM and ten were healthy with normal plasma cholesterol (< 6.5 mM). Simvastatin (40 mg/day) was prescribed to those with hypercholesterolaemia. Plasma samples were taken from both groups at baseline and after three months. Simvastatin reduced plasma cholesterol by ~35% (p < 0.05) at the end of three months. Oxysterols generated by autoxidation (but not enzymatically) were elevated up to 45 fold in hypercholesterolaemic midlife men. Plasma oxysterols were restored to those of healthy controls after simvastatin intervention suggesting that autoxidation is either prevented by simvastatin directly or that autoxidation is less prevalent when plasma cholesterol concentrations are within the normal range.
Collapse
Affiliation(s)
- Irundika H K Dias
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - Ivana Milic
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - Gregory Y H Lip
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Devitt
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - M Cristina Polidori
- Institute of Biochemistry and Molecular Biology I, Heinrich-Heine-University, Duesseldorf, Germany; Ageing Clinical Research, Department Medicine II, University Hospital of Cologne, Cologne, Germany
| | - Helen R Griffiths
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK; Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford GU2 7XH, UK
| |
Collapse
|
22
|
Favero GM, Paz JL, Otake AH, Maria DA, Caldini EG, de Medeiros RSS, Deus DF, Chammas R, Maranhão RC, Bydlowski SP. Cell internalization of 7-ketocholesterol-containing nanoemulsion through LDL receptor reduces melanoma growth in vitro and in vivo: a preliminary report. Oncotarget 2018; 9:14160-14174. [PMID: 29581835 PMCID: PMC5865661 DOI: 10.18632/oncotarget.24389] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/25/2018] [Indexed: 01/01/2023] Open
Abstract
Oxysterols are cholesterol oxygenated derivatives which possess several biological actions. Among oxysterols, 7-ketocholesterol (7KC) is known to induce cell death. Here, we hypothesized that 7KC cytotoxicity could be applied in cancer therapeutics. 7KC was incorporated into a lipid core nanoemulsion. As a cellular model the murine melanoma cell line B16F10 was used. The nanoparticle (7KCLDE) uptake into tumor cells was displaced by increasing amounts of low-density-lipoproteins (LDL) suggesting a LDL-receptor-mediated cell internalization. 7KCLDE was mainly cytostatic, which led to an accumulation of polyploid cells. Nevertheless, a single dose of 7KCLDE killed roughly 10% of melanoma cells. In addition, it was observed dissipation of the transmembrane potential, evidenced with flow cytometry; presence of autophagic vacuoles, visualized and quantified with flow cytometry and acridine orange; and presence of myelin figures, observed with ultrastructural microscopy. 7KCLDE impaired cytokenesis was accompanied by changes in cellular morphology into a fibroblastoid shape which is supported by cytoskeletal rearrangements, as shown by the increased actin polymerization. 7KCLDE was injected into B16 melanoma tumor-bearing mice. 7KCLDE accumulated in the liver and tumor. In melanoma tumor 7KCLDE promoted a >50% size reduction, enlarged the necrotic area, and reduced intratumoral vasculature. 7KCLDE increased the survival rates of animals, without hematologic or liver toxicity. Although more pre-clinical studies should be performed, our preliminary results suggested that 7KCLDE is a promising novel preparation for cancer chemotherapy.
Collapse
Affiliation(s)
- Giovani M Favero
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.,Department of General Biology, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Jessica L Paz
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Andréia H Otake
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.,Instituto do Cancer do Estado de Sao Paulo (ICESP), SP, Brazil
| | - Durvanei A Maria
- Biochemistry and Biophysics Laboratories, Instituto Butantan, Sao Paulo, SP, Brazil
| | - Elia G Caldini
- Laboratory for Cell Biology, Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Raphael S S de Medeiros
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.,Instituto do Cancer do Estado de Sao Paulo (ICESP), SP, Brazil
| | - Debora F Deus
- Laboratory of Metabolism and Lipids, Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.,Instituto do Cancer do Estado de Sao Paulo (ICESP), SP, Brazil
| | - Raul C Maranhão
- Laboratory of Metabolism and Lipids, Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.,Faculdade de Ciencias Farmaceuticas, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Sergio P Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), Department of Hematology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|