1
|
Rezig L, Ghzaiel I, Ksila M, Yammine A, Nury T, Zarrouk A, Samadi M, Chouaibi M, Vejux A, Lizard G. Cytoprotective activities of representative nutrients from the Mediterranean diet and of Mediterranean oils against 7-ketocholesterol- and 7β-hydroxycholesterol-induced cytotoxicity: Application to age-related diseases and civilization diseases. Steroids 2022; 187:109093. [PMID: 36029811 DOI: 10.1016/j.steroids.2022.109093] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/17/2022]
Abstract
7-ketocholesterol and 7β-hydroxycholesterol are two oxysterols mainly formed by the autoxidation of cholesterol. These two molecules are interconvertible via specific enzymes. These two oxysterols are often observed at increased amounts in biological fluids as well as tissues and organs affected during age-related diseases and in diseases of civilization such as cardiovascular, neurodegenerative, and ocular diseases as well as type 2 diabetes and metabolic syndrome. Noteworthy, 7-ketocholesterol and 7β-hydroxycholesterol induce oxidative stress and inflammation, which are frequently observed in patients with age-related and civilization diseases. For this reason, the involvement of these two oxysterols in the pathophysiology of these diseases is widely suspected. In addition, the toxicity of these oxysterols can lead to death by oxiapoptophagy characterized by oxidative stress, apoptosis induction and autophagy criteria. To prevent, or even treat, certain age-related or civilization diseases associated with increased levels of 7-ketocholesterol and 7β-hydroxycholesterol, the identification of molecules or mixtures of molecules attenuating or inhibiting the toxic effects of these oxysterols allows to consider new treatments. In this context, many nutrients present in significant amounts in the Mediterranean diet, especially tocopherols, fatty acids, and polyphenols, have shown cytoprotective activities as well as several Mediterranean oils (argan and olive oils, milk thistle seed oil, and pistacia lentiscus seed oil). Consequently, a nutraceutical approach, rich in nutrients present in the Mediterranean diet, could thus make it possible to counteract certain age-related and civilization diseases associated with increased levels of 7-ketocholesterol and 7β-hydroxycholesterol.
Collapse
Affiliation(s)
- Leila Rezig
- University of Carthage, National Institute of Applied Sciences and Technology, LR11ES26, LIP-MB 'Laboratory of Protein Engineering and Bioactive Molecules', Tunis 1080, Tunisia; University of Carthage, High Institute of Food Industries, 58 Alain Savary Street, El Khadra City, Tunis 1003, Tunisia.
| | - Imen Ghzaiel
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France; University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir 5000, Tunisia; University Tunis-El Manar, Faculty of Sciences of Tunis, Tunis 2092, Tunisia
| | - Mohamed Ksila
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France; Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia
| | - Aline Yammine
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France; Institut Européen des Antioxydants (IEA), 1B, rue Victor de Lespinats, Neuves-Maisons 54230, France
| | - Thomas Nury
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France
| | - Amira Zarrouk
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir 5000, Tunisia; Laboratory of Biochemistry, Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, University Lorraine, Metz Technopôle, Metz 57070, France
| | - Moncef Chouaibi
- University of Carthage, High Institute of Food Industries, 58 Alain Savary Street, El Khadra City, Tunis 1003, Tunisia; University of Carthage, Bio-preservation and Valorization of Agricultural Products UR13-AGR 02, High Institute of Food Industries, 58 Alain Savary Street, El Khadra City, Tunis 1003, Tunisia
| | - Anne Vejux
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France
| | - Gérard Lizard
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270), University of Bourgogne/Inserm, Dijon 21000, France.
| |
Collapse
|
2
|
Shoji S, Maekawa M, Ogura J, Sato T, Mano N. Identification cholesterol metabolites altered before the onset of nonalcoholic steatohepatitis by targeted metabolomics. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159135. [PMID: 35217199 DOI: 10.1016/j.bbalip.2022.159135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 02/08/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a disease with symptoms similar to those of alcoholic liver inflammation without alcohol intake. As an effective treatment strategy has not been established for this disease, a detailed understanding of the pathological progression mechanism is required. We focused on cholesterol metabolites, which are suspected to regulate NASH pathology, and investigated their relationship with the pathological progression in the early stages of NASH. First, the LC/MS/MS methods for bile acids and sterols were optimized and validated. Next, NASH model mice were established by feeding a choline-deficient, methionine-reduced high-fat diet, and the levels of hepatic cholesterol metabolites were measured. As a result, before the onset of NASH, desmosterol, 4β-hydroxycholesterol, campesterol, sitosterol, secondary bile acids such as taurodeoxycholic acid significantly decreased by up to 1/38 of NASH model group. Autoxidation-generated sterols significantly increased 2- to 5-fold, and various primary bile acids such as conjugated β-muricholic acids and cholic acids significantly increased 2- to 7-fold. In this study, the levels of cholesterol metabolites changed in the before the onset of NASH. These metabolic alterations involved in inflammation induction and detoxification for NASH may help the discovery of early diagnostic biomarkers in the future.
Collapse
Affiliation(s)
- Saori Shoji
- Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai 980-8574, Japan
| | - Masamitsu Maekawa
- Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai 980-8574, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Jiro Ogura
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Faculty of Pharmaceutical Sciences, Tohoku University, 1-1 Seiryo-machi, Aoba-Ku, Sendai 980-8574, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
3
|
Ghzaiel I, Zarrouk A, Nury T, Libergoli M, Florio F, Hammouda S, Ménétrier F, Avoscan L, Yammine A, Samadi M, Latruffe N, Biressi S, Levy D, Bydlowski SP, Hammami S, Vejux A, Hammami M, Lizard G. Antioxidant Properties and Cytoprotective Effect of Pistacia lentiscus L. Seed Oil against 7β-Hydroxycholesterol-Induced Toxicity in C2C12 Myoblasts: Reduction in Oxidative Stress, Mitochondrial and Peroxisomal Dysfunctions and Attenuation of Cell Death. Antioxidants (Basel) 2021; 10:antiox10111772. [PMID: 34829643 PMCID: PMC8615043 DOI: 10.3390/antiox10111772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023] Open
Abstract
Aging is characterized by a progressive increase in oxidative stress, which favors lipid peroxidation and the formation of cholesterol oxide derivatives, including 7β-hydroxycholesterol (7β-OHC). This oxysterol, which is known to trigger oxidative stress, inflammation, and cell death, could contribute to the aging process and age-related diseases, such as sarcopenia. Identifying molecules or mixtures of molecules preventing the toxicity of 7β-OHC is therefore an important issue. This study consists of determining the chemical composition of Tunisian Pistacia lentiscus L. seed oil (PLSO) used in the Tunisian diet and evaluating its ability to counteract the cytotoxic effects induced by 7β-OHC in murine C2C12 myoblasts. The effects of 7β-OHC (50 µM; 24 h), associated or not with PLSO, were studied on cell viability, oxidative stress, and on mitochondrial and peroxisomal damages induction. α-Tocopherol (400 µM) was used as the positive control for cytoprotection. Our data show that PLSO is rich in bioactive compounds; it contains polyunsaturated fatty acids, and several nutrients with antioxidant properties: phytosterols, α-tocopherol, carotenoids, flavonoids, and phenolic compounds. When associated with PLSO (100 µg/mL), the 7β-OHC-induced cytotoxic effects were strongly attenuated. The cytoprotection was in the range of those observed with α-tocopherol. This cytoprotective effect was characterized by prevention of cell death and organelle dysfunction (restoration of cell adhesion, cell viability, and plasma membrane integrity; prevention of mitochondrial and peroxisomal damage) and attenuation of oxidative stress (reduction in reactive oxygen species overproduction in whole cells and at the mitochondrial level; decrease in lipid and protein oxidation products formation; and normalization of antioxidant enzyme activities: glutathione peroxidase (GPx) and superoxide dismutase (SOD)). These results provide evidence that PLSO has similar antioxidant properties than α-tocopherol used at high concentration and contains a mixture of molecules capable to attenuate 7β-OHC-induced cytotoxic effects in C2C12 myoblasts. These data reinforce the interest in edible oils associated with the Mediterranean diet, such as PLSO, in the prevention of age-related diseases, such as sarcopenia.
Collapse
Affiliation(s)
- Imen Ghzaiel
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
- Faculty of Sciences of Tunis, University Tunis-El Manar, Tunis 2092, Tunisia
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
- Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
- Correspondence: (A.Z.); (G.L.); Tel.: +216-94-837-999 or +1-212-241 9304 (A.Z.); +33-380-396-256 (G.L.)
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Michela Libergoli
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - Francesca Florio
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - Souha Hammouda
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Franck Ménétrier
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21065 Dijon, France;
| | - Laure Avoscan
- Agroécologie, AgroSup Dijon, CNRS, INRAE, University Bourgogne Franche-Comté, Plateforme DimaCell, 21000 Dijon, France;
| | - Aline Yammine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, University Lorraine, Metz Technopôle, 57070 Metz, France;
| | - Norbert Latruffe
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - 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.); (S.P.B.)
| | - 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.); (S.P.B.)
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
| | - Sonia Hammami
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Mohamed Hammami
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
- Correspondence: (A.Z.); (G.L.); Tel.: +216-94-837-999 or +1-212-241 9304 (A.Z.); +33-380-396-256 (G.L.)
| |
Collapse
|
4
|
Morris DJ, Brem AS, Odermatt A. Modulation of 11β-hydroxysteroid dehydrogenase functions by the cloud of endogenous metabolites in a local microenvironment: The glycyrrhetinic acid-like factor (GALF) hypothesis. J Steroid Biochem Mol Biol 2021; 214:105988. [PMID: 34464733 DOI: 10.1016/j.jsbmb.2021.105988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 01/09/2023]
Abstract
11β-Hydroxysteroid dehydrogenase (11β-HSD)-dependent conversion of cortisol to cortisone and corticosterone to 11-dehydrocorticosterone are essential in regulating transcriptional activities of mineralocorticoid receptors (MR) and glucocorticoid receptors (GR). Inhibition of 11β-HSD by glycyrrhetinic acid metabolites, bioactive components of licorice, causes sodium retention and potassium loss, with hypertension characterized by low renin and aldosterone. Essential hypertension is a major disease, mostly with unknown underlying mechanisms. Here, we discuss a putative mechanism for essential hypertension, the concept that endogenous steroidal compounds acting as glycyrrhetinic acid-like factors (GALFs) inhibit 11β-HSD dehydrogenase, and allow for glucocorticoid-induced MR and GR activation with resulting hypertension. Initially, several metabolites of adrenally produced glucocorticoids and mineralocorticoids were shown to be potent 11β-HSD inhibitors. Such GALFs include modifications in the A-ring and/or at positions 3, 7 and 21 of the steroid backbone. These metabolites may be formed in peripheral tissues or by gut microbiota. More recently, metabolites of 11β-hydroxy-Δ4androstene-3,17-dione and 7-oxygenated oxysterols have been identified as potent 11β-HSD inhibitors. In a living system, 11β-HSD isoforms are not exposed to a single substrate but to several substrates, cofactors, and various inhibitors simultaneously, all at different concentrations depending on physical state, tissue and cell type. We propose that this "cloud" of steroids and steroid-like substances in the microenvironment determines the 11β-HSD-dependent control of MR and GR activity. A dysregulated composition of this cloud of metabolites in the respective microenvironment needs to be taken into account when investigating disease mechanisms, for forms of low renin, low aldosterone hypertension.
Collapse
Affiliation(s)
- David J Morris
- Department of Pathology and Laboratory Medicine, The Miriam Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Andrew S Brem
- Division of Kidney Diseases and Hypertension, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
5
|
Nury T, Yammine A, Ghzaiel I, Sassi K, Zarrouk A, Brahmi F, Samadi M, Rup-Jacques S, Vervandier-Fasseur D, Pais de Barros J, Bergas V, Ghosh S, Majeed M, Pande A, Atanasov A, Hammami S, Hammami M, Mackrill J, Nasser B, Andreoletti P, Cherkaoui-Malki M, Vejux A, Lizard G. Attenuation of 7-ketocholesterol- and 7β-hydroxycholesterol-induced oxiapoptophagy by nutrients, synthetic molecules and oils: Potential for the prevention of age-related diseases. Ageing Res Rev 2021; 68:101324. [PMID: 33774195 DOI: 10.1016/j.arr.2021.101324] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/18/2022]
Abstract
Age-related diseases for which there are no effective treatments include cardiovascular diseases; neurodegenerative diseases such as Alzheimer's disease; eye disorders such as cataract and age-related macular degeneration; and, more recently, Severe Acute Respiratory Syndrome (SARS-CoV-2). These diseases are associated with plasma and/or tissue increases in cholesterol derivatives mainly formed by auto-oxidation: 7-ketocholesterol, also known as 7-oxo-cholesterol, and 7β-hydroxycholesterol. The formation of these oxysterols can be considered as a consequence of mitochondrial and peroxisomal dysfunction, leading to increased in oxidative stress, which is accentuated with age. 7-ketocholesterol and 7β-hydroxycholesterol cause a specific form of cytotoxic activity defined as oxiapoptophagy, including oxidative stress and induction of death by apoptosis associated with autophagic criteria. Oxiaptophagy is associated with organelle dysfunction and in particular with mitochondrial and peroxisomal alterations involved in the induction of cell death and in the rupture of redox balance. As the criteria characterizing 7-ketocholesterol- and 7β-hydroxycholesterol-induced cytotoxicity are often simultaneously observed in major age-related diseases (cardiovascular diseases, age-related macular degeneration, Alzheimer's disease) the involvement of these oxysterols in the pathophysiology of the latter seems increasingly likely. It is therefore important to better understand the signalling pathways associated with the toxicity of 7-ketocholesterol and 7β-hydroxycholesterol in order to identify pharmacological targets, nutrients and synthetic molecules attenuating or inhibiting the cytotoxic activities of these oxysterols. Numerous natural cytoprotective compounds have been identified: vitamins, fatty acids, polyphenols, terpenes, vegetal pigments, antioxidants, mixtures of compounds (oils, plant extracts) and bacterial enzymes. However, few synthetic molecules are able to prevent 7-ketocholesterol- and/or 7β-hydroxycholesterol-induced cytotoxicity: dimethyl fumarate, monomethyl fumarate, the tyrosine kinase inhibitor AG126, memantine, simvastatine, Trolox, dimethylsufoxide, mangafodipir and mitochondrial permeability transition pore (MPTP) inhibitors. The effectiveness of these compounds, several of which are already in use in humans, makes it possible to consider using them for the treatment of certain age-related diseases associated with increased plasma and/or tissue levels of 7-ketocholesterol and/or 7β-hydroxycholesterol.
Collapse
|
6
|
Gomez-Sanchez EP, Gomez-Sanchez CE. 11β-hydroxysteroid dehydrogenases: A growing multi-tasking family. Mol Cell Endocrinol 2021; 526:111210. [PMID: 33607268 PMCID: PMC8108011 DOI: 10.1016/j.mce.2021.111210] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023]
Abstract
This review briefly addresses the history of the discovery and elucidation of the three cloned 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes in the human, 11βHSD1, 11βHSD2 and 11βHSD3, an NADP+-dependent dehydrogenase also called the 11βHSD1-like dehydrogenase (11βHSD1L), as well as evidence for yet identified 11βHSDs. Attention is devoted to more recently described aspects of this multi-functional family. The importance of 11βHSD substrates other than glucocorticoids including bile acids, 7-keto sterols, neurosteroids, and xenobiotics is discussed, along with examples of pathology when functions of these multi-tasking enzymes are disrupted. 11βHSDs modulate the intracellular concentration of glucocorticoids, thereby regulating the activation of the glucocorticoid and mineralocorticoid receptors, and 7β-27-hydroxycholesterol, an agonist of the retinoid-related orphan receptor gamma (RORγ). Key functions of this nuclear transcription factor include regulation of immune cell differentiation, cytokine production and inflammation at the cell level. 11βHSD1 expression and/or glucocorticoid reductase activity are inappropriately increased with age and in obesity and metabolic syndrome (MetS). Potential causes for disappointing results of the clinical trials of selective inhibitors of 11βHSD1 in the treatment of these disorders are discussed, as well as the potential for more targeted use of inhibitors of 11βHSD1 and 11βHSD2.
Collapse
Affiliation(s)
| | - Celso E Gomez-Sanchez
- Department of Pharmacology and Toxicology, Jackson, MS, USA; Medicine (Endocrinology), Jackson, MS, USA; University of Mississippi Medical Center and G.V. (Sonny) Montgomery VA Medical Center(3), Jackson, MS, USA
| |
Collapse
|
7
|
Miyamoto S, Lima RS, Inague A, Viviani LG. Electrophilic oxysterols: generation, measurement and protein modification. Free Radic Res 2021; 55:416-440. [PMID: 33494620 DOI: 10.1080/10715762.2021.1879387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cholesterol is an essential component of mammalian plasma membranes. Alterations in sterol metabolism or oxidation have been linked to various pathological conditions, including cardiovascular diseases, cancer, and neurodegenerative disorders. Unsaturated sterols are vulnerable to oxidation induced by singlet oxygen and other reactive oxygen species. This process yields reactive sterol oxidation products, including hydroperoxides, epoxides as well as aldehydes. These oxysterols, in particular those with high electrophilicity, can modify nucleophilic sites in biomolecules and affect many cellular functions. Here, we review the generation and measurement of reactive sterol oxidation products with emphasis on cholesterol hydroperoxides and aldehyde derivatives (electrophilic oxysterols) and their effects on protein modifications.
Collapse
Affiliation(s)
- Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo S Lima
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Alex Inague
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Lucas G Viviani
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Brown AJ, Sharpe LJ, Rogers MJ. Oxysterols: From physiological tuners to pharmacological opportunities. Br J Pharmacol 2020; 178:3089-3103. [PMID: 32335907 DOI: 10.1111/bph.15073] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Oxysterols are oxygenated forms of cholesterol generated via autooxidation by free radicals and ROS, or formed enzymically by a variety of enzymes such as those involved in the synthesis of bile acids. Although found at very low concentrations in vivo, these metabolites play key roles in health and disease, particularly in development and regulating immune cell responses, by binding to effector proteins such as LXRα, RORγ and Insig and directly or indirectly regulating transcriptional programmes that affect cell metabolism and function. In this review, we summarise the routes by which oxysterols can be generated and subsequently modified to other oxysterol metabolites and highlight their diverse and profound biological functions and opportunities to alter their levels using pharmacological approaches. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michael J Rogers
- Garvan Institute of Medical Research and St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Loving BA, Bruce KD. Lipid and Lipoprotein Metabolism in Microglia. Front Physiol 2020; 11:393. [PMID: 32411016 PMCID: PMC7198855 DOI: 10.3389/fphys.2020.00393] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Microglia, once viewed as static bystanders with limited homeostatic functions, are now considered key players in the development of neuroinflammatory and neurodegenerative diseases. Microglial activation is a salient feature of neuroinflammation involving a dynamic process that generates multitudinous microglial phenotypes that can respond to a variety of situational cues in the central nervous system. Recently, a flurry of single cell RNA-sequencing studies have defined microglial phenotypes in unprecedented detail, and have highlighted robust changes in the expression of genes involved in lipid and lipoprotein metabolism. Increased expression of genes such as Apolipoprotein E (ApoE), Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) and Lipoprotein Lipase (LPL) in microglia during development, damage, and disease, suggest that increased lipid metabolism is needed to fuel protective cellular functions such as phagocytosis. This review describes our current understanding of lipid and lipoprotein metabolism in microglia, and highlights microglial lipid metabolism as a modifiable target for the treatment of neurodegenerative diseases such as Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Bailey A. Loving
- School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Kimberley D. Bruce
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
10
|
Vejux A, Abed-Vieillard D, Hajji K, Zarrouk A, Mackrill JJ, Ghosh S, Nury T, Yammine A, Zaibi M, Mihoubi W, Bouchab H, Nasser B, Grosjean Y, Lizard G. 7-Ketocholesterol and 7β-hydroxycholesterol: In vitro and animal models used to characterize their activities and to identify molecules preventing their toxicity. Biochem Pharmacol 2020; 173:113648. [DOI: 10.1016/j.bcp.2019.113648] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022]
|
11
|
Sghaier R, Zarrouk A, Nury T, Badreddine I, O'Brien N, Mackrill JJ, Vejux A, Samadi M, Nasser B, Caccia C, Leoni V, Moreau T, Cherkaoui-Malki M, Salhedine Masmoudi A, Lizard G. Biotin attenuation of oxidative stress, mitochondrial dysfunction, lipid metabolism alteration and 7β-hydroxycholesterol-induced cell death in 158N murine oligodendrocytes. Free Radic Res 2019; 53:535-561. [PMID: 31039616 DOI: 10.1080/10715762.2019.1612891] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Mitochondrial dysfunction and oxidative stress are involved in neurodegenerative diseases associated with an enhancement of lipid peroxidation products such as 7β-hydroxycholesterol (7β-OHC). It is, therefore, important to study the ability of 7β-OHC to trigger mitochondrial defects, oxidative stress, metabolic dysfunctions and cell death, which are hallmarks of neurodegeneration, and to identify cytoprotective molecules. The effects of biotin were evaluated on 158N murine oligodendrocytes, which are myelin synthesizing cells, exposed to 7β-OHC (50 µM) with or without biotin (10 and 100 nM) or α-tocopherol (positive control of cytoprotection). The effects of biotin on 7β-OHC activities were determined using different criteria: cell adhesion; plasma membrane integrity; redox status. The impact on mitochondria was characterized by the measurement of transmembrane mitochondrial potential (ΔΨm), reactive oxygen species (ROS) overproduction, mitochondrial mass, quantification of cardiolipins and organic acids. Sterols and fatty acids were also quantified. Cell death (apoptosis, autophagy) was characterized by the enumeration of apoptotic cells, caspase-3 activation, identification of autophagic vesicles, and activation of LC3-I into LC3-II. Biotin attenuates 7β-OHC-induced cytotoxicity: loss of cell adhesion was reduced; antioxidant activities were normalized. ROS overproduction, protein and lipid oxidation products were decreased. Biotin partially restores mitochondrial functions: attenuation of the loss of ΔΨm; reduced levels of mitochondrial O2•- overproduction; normalization of cardiolipins and organic acid levels. Biotin also normalizes cholesterol and fatty acid synthesis, and prevents apoptosis and autophagy (oxiapoptophagy). Our data support that biotin, which prevents oligodendrocytes damages, could be useful in the treatment of neurodegeneration and demyelination.
Collapse
Affiliation(s)
- Randa Sghaier
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,Laboratory of Biochemistry, Faculty of Medicine , University Sousse , Sousse , Tunisia.,Faculty of Medicine, Laboratory - NAFS "Nutrition - Functional Food & Vascular Health" , Monastir & University Sousse , Sousse , Tunisia.,Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources , University Manouba, Higher Institute of Biotechnology , Sidi Thabet , Tunisia
| | - Amira Zarrouk
- Laboratory of Biochemistry, Faculty of Medicine , University Sousse , Sousse , Tunisia.,Faculty of Medicine, Laboratory - NAFS "Nutrition - Functional Food & Vascular Health" , Monastir & University Sousse , Sousse , Tunisia.,School of Food and Nutritional Sciences , University College Cork , Cork , Ireland.,Department of Physiology , Biosciences Institute, University College Cork , Cork , Ireland
| | - Thomas Nury
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Ilham Badreddine
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,University Ibn Zohr, Lab. 'Valorisation des Ressources Naturelles et Environnement' , Taroudant , Morocco.,Laboratory Neuroscience and Biochemistry , University Hassan 1er , Settat , Morocco
| | - Nora O'Brien
- School of Food and Nutritional Sciences , University College Cork , Cork , Ireland
| | - John J Mackrill
- Department of Physiology , Biosciences Institute, University College Cork , Cork , Ireland
| | - Anne Vejux
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Mohammad Samadi
- Department of Chemistry , University Lorraine, Metz Technopôle , Metz , France
| | - Boubker Nasser
- Laboratory Neuroscience and Biochemistry , University Hassan 1er , Settat , Morocco
| | - Claudio Caccia
- Laboratory of Medical Genetics and Neurogenetics , Foundation IRCCS Istituto Neurologico Carlo Besta , Milan , Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry , Hospital of Varese, ASST-Settelaghi , Milan , Italy
| | - Thibault Moreau
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,Department of Neurology , University Hospital , Dijon , France
| | - Mustapha Cherkaoui-Malki
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Ahmed Salhedine Masmoudi
- Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources , University Manouba, Higher Institute of Biotechnology , Sidi Thabet , Tunisia
| | - Gérard Lizard
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| |
Collapse
|
12
|
Dias IH, Borah K, Amin B, Griffiths HR, Sassi K, Lizard G, Iriondo A, Martinez-Lage P. Localisation of oxysterols at the sub-cellular level and in biological fluids. J Steroid Biochem Mol Biol 2019; 193:105426. [PMID: 31301352 DOI: 10.1016/j.jsbmb.2019.105426] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/25/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Oxysterols are oxidized derivatives of cholesterol that are formed enzymatically or via reactive oxygen species or both. Cholesterol or oxysterols ingested as food are absorbed and packed into lipoproteins that are taken up by hepatic cells. Within hepatic cells, excess cholesterol is metabolised to form bile acids. The endoplasmic reticulum acts as the main organelle in the bile acid synthesis pathway. Metabolised sterols originating from this pathway are distributed within other organelles and in the cell membrane. The alterations to membrane oxysterol:sterol ratio affects the integrity of the cell membrane. The presence of oxysterols changes membrane fluidity and receptor orientation. It is well documented that hydroxylase enzymes located in mitochondria facilitate oxysterol production via an acidic pathway. More recently, the presence of oxysterols was also reported in lysosomes. Peroxisomal deficiencies favour intracellular oxysterols accumulation. Despite the low abundance of oxysterols compared to cholesterol, the biological actions of oxysterols are numerous and important. Oxysterol levels are implicated in the pathogenesis of multiple diseases ranging from chronic inflammatory diseases (atherosclerosis, Alzheimer's disease and bowel disease), cancer and numerous neurodegenerative diseases. In this article, we review the distribution of oxysterols in sub-cellular organelles and in biological fluids.
Collapse
Affiliation(s)
- Irundika Hk Dias
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK.
| | - Khushboo Borah
- Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, UK
| | - Berivan Amin
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Helen R Griffiths
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK; Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, UK
| | - Khouloud Sassi
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270)/University Bourgogne Franche-Comté/Inserm, 21000 Dijon, France; Univ. Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, Tunis, Tunisia
| | - Gérard Lizard
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270)/University Bourgogne Franche-Comté/Inserm, 21000 Dijon, France
| | - Ane Iriondo
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| | - Pablo Martinez-Lage
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| |
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
|
Beck KR, Inderbinen SG, Kanagaratnam S, Kratschmar DV, Jetten AM, Yamaguchi H, Odermatt A. 11β-Hydroxysteroid dehydrogenases control access of 7β,27-dihydroxycholesterol to retinoid-related orphan receptor γ. J Lipid Res 2019; 60:1535-1546. [PMID: 31273032 PMCID: PMC6718442 DOI: 10.1194/jlr.m092908] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/24/2019] [Indexed: 12/15/2022] Open
Abstract
Oxysterols previously were considered intermediates of bile acid and steroid hormone biosynthetic pathways. However, recent research has emphasized the roles of oxysterols in essential physiologic processes and in various diseases. Despite these discoveries, the metabolic pathways leading to the different oxysterols are still largely unknown and the biosynthetic origin of several oxysterols remains unidentified. Earlier studies demonstrated that the glucocorticoid metabolizing enzymes, 11β-hydroxysteroid dehydrogenase (11β-HSD) types 1 and 2, interconvert 7-ketocholesterol (7kC) and 7β-hydroxycholesterol (7βOHC). We examined the role of 11β-HSDs in the enzymatic control of the intracellular availability of 7β,27-dihydroxycholesterol (7β27OHC), a retinoid-related orphan receptor γ (RORγ) ligand. We used microsomal preparations of cells expressing recombinant 11β-HSD1 and 11β-HSD2 to assess whether 7β27OHC and 7-keto,27-hydroxycholesterol (7k27OHC) are substrates of these enzymes. Binding of 7β27OHC and 7k27OHC to 11β-HSDs was studied by molecular modeling. To our knowledge, the stereospecific oxoreduction of 7k27OHC to 7β27OHC by human 11β-HSD1 and the reverse oxidation reaction of 7β27OHC to 7k27OHC by human 11β-HSD2 were demonstrated for the first time. Apparent enzyme affinities of 11β-HSDs for these novel substrates were equal to or higher than those of the glucocorticoids. This is supported by the fact that 7k27OHC and 7β27OHC are potent inhibitors of the 11β-HSD1-dependent oxoreduction of cortisone and the 11β-HSD2-dependent oxidation of cortisol, respectively. Furthermore, molecular docking calculations explained stereospecific enzyme activities. Finally, using an inducible RORγ reporter system, we showed that 11β-HSD1 and 11β-HSD2 controlled RORγ activity. These findings revealed a novel glucocorticoid-independent prereceptor regulation mechanism by 11β-HSDs that warrants further investigation.
Collapse
Affiliation(s)
- Katharina R Beck
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Silvia G Inderbinen
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Sharavan Kanagaratnam
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Anton M Jetten
- Immunity, Inflammation, and Disease Laboratory National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Hideaki Yamaguchi
- Department of Applied Biological Chemistry Meijo University, Nagoya 468-8502, Japan
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
15
|
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
|
16
|
Chemistry, biochemistry, metabolic fate and mechanism of action of 6-oxo-cholestan-3β,5α-diol (OCDO), a tumor promoter and cholesterol metabolite. Biochimie 2018; 153:139-149. [PMID: 29654865 DOI: 10.1016/j.biochi.2018.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022]
Abstract
Oxygenation products of cholesterol, named oxysterols, were suspected since the 20th century to be involved in carcinogenesis. Among the family of oxysterol molecules, cholesterol-5,6-epoxides (5,6-EC) retained the attention of scientists because they contain a putative alkylating epoxide group. However, studies failed into demonstrating that 5,6-EC were direct carcinogens and revealed a surprising chemical stability and unreactivity towards nucleophiles in standard conditions. Analyses of 5,6-EC metabolism in normal cells showed that they were extensively transformed into cholestane-3β,5α,6β-triol (CT) by the cholesterol-5,6-epoxide hydrolase (ChEH). Studies performed in cancer cells showed that CT was additionally metabolized into an oxysterol identified as the 6-oxo-cholestan-3β,5α-diol (OCDO), by the 11β-hydroxysteroid dehydrogenase of type 2 (HSD2), the enzyme which inactivates cortisol into cortisone. Importantly, OCDO was shown to display tumor promoter properties in breast cancers, by binding to the glucocorticoid receptor, and independently of their estrogen receptor status, revealing the existence of a new tumorigenic pathway centered on 5,6-EC. In breast tumors from patients, OCDO production as well as the expression of the enzymes involved in the pathway producing OCDO, namely ChEH subunits and HSD2, were higher compared to normal tissues, and overexpression of these enzymes correlate with a higher risk of patient death, indicating that this onco-metabolism is of major importance to breast cancer pathology. Herein, we will review the actual knowledge and the future trends in OCDO chemistry, biochemistry, metabolism and mechanism of action and will discuss the impact of OCDO discovery on new anticancer therapeutic strategies.
Collapse
|
17
|
Villalpando DM, Rojas MM, García HS, Ferrer M. Dietary docosahexaenoic acid supplementation prevents the formation of cholesterol oxidation products in arteries from orchidectomized rats. PLoS One 2017; 12:e0185805. [PMID: 28968462 PMCID: PMC5624632 DOI: 10.1371/journal.pone.0185805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022] Open
Abstract
Testosterone deficiency has been correlated with increased cardiovascular diseases, which in turn has been associated with increased oxidative stress. Several studies have considered cholesterol oxidation products (COPs) as oxidative stress biomarkers, since some of them play pro-oxidant and pro-inflammatory roles. We have previously described the cardioprotective effects of a dosahexaenoic acid (DHA) supplemented diet on the aortic and mesenteric artery function of orchidectomized rats. The aim of this study was to investigate whether impaired gonadal function alters the formation of COPs, as well as the potential preventive role of a DHA-supplemented diet on that effect. For this purpose, aortic and mesenteric artery segments obtained from control and orchidectomized rats, fed with a standard or supplemented with DHA, were used. The content of the following COPs: 7α-hydroxycholesterol, 7β-hydroxycholesterol, 7-ketocholesterol, 5,6α-epoxycholesterol, 5,6β-epoxycholesterol, cholestanetriol and 25-hydroxycholesterol, were analyzed by gas chromatography. The results showed that orchidectomy increased the formation of COPs in arteries from orchidectomized rats, which may participate in the orchidectomy-induced structural and functional vascular alterations already reported. The fact that the DHA-supplemented diet prevented the orchidectomy-induced COPs increase confirms the cardiovascular protective actions of DHA, which could be of special relevance in mesenteric arterial bed, since it importantly controls the systemic vascular resistance.
Collapse
Affiliation(s)
- Diva M. Villalpando
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mibsam M. Rojas
- Unidad de Investigación y Desarrollo de Alimentos, Instituto Tecnológico de Veracruz, Veracruz, México
| | - Hugo S. García
- Unidad de Investigación y Desarrollo de Alimentos, Instituto Tecnológico de Veracruz, Veracruz, México
- * E-mail: (MF); (HSG)
| | - Mercedes Ferrer
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail: (MF); (HSG)
| |
Collapse
|
18
|
Griffiths WJ, Hearn T, Crick PJ, Abdel-Khalik J, Dickson A, Yutuc E, Wang Y. Charge-tagging liquid chromatography-mass spectrometry methodology targeting oxysterol diastereoisomers. Chem Phys Lipids 2017; 207:69-80. [PMID: 28411018 PMCID: PMC5630687 DOI: 10.1016/j.chemphyslip.2017.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
EADSA and LC–MS allows the differentiation of sterol diastereoisomers. 24S- and 24R-hydroxycholesterol are present in human plasma. Four diastereoisomers of 3β,7-dihydroxycholest-5-enoic acid are found in human plasma. 3β,7α- and 3β,7β-dihydroxychol-5-enoic acids are found in human plasma. 7α- and 7β- epimers give distinguishable MS3 spectra.
The introduction of a hydroxy group to the cholesterol skeleton introduces not only the possibility for positional isomers but also diastereoisomers, where two or more isomers have different configurations at one or more of the stereocentres but are not mirror images. The differentiation of diastereoisomers is important as differing isomers can have differing biochemical properties and are formed via different biochemical pathways. Separation of diasterioisomers is not always easy by chromatographic methods Here we demonstrate, by application of charge-tagging and derivatisation with the Girard P reagent, the separation and detection of biologically relevant diastereoisomers using liquid chromatography − mass spectrometry with multistage fragmentation.
Collapse
Affiliation(s)
- William J Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK.
| | - Thomas Hearn
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Peter J Crick
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Jonas Abdel-Khalik
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Alison Dickson
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Eylan Yutuc
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
19
|
Gray GA, White CI, Castellan RFP, McSweeney SJ, Chapman KE. Getting to the heart of intracellular glucocorticoid regeneration: 11β-HSD1 in the myocardium. J Mol Endocrinol 2017; 58:R1-R13. [PMID: 27553202 PMCID: PMC5148800 DOI: 10.1530/jme-16-0128] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/11/2022]
Abstract
Corticosteroids influence the development and function of the heart and its response to injury and pressure overload via actions on glucocorticoid (GR) and mineralocorticoid (MR) receptors. Systemic corticosteroid concentration depends largely on the activity of the hypothalamic-pituitary-adrenal (HPA) axis, but glucocorticoid can also be regenerated from intrinsically inert metabolites by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), selectively increasing glucocorticoid levels within cells and tissues. Extensive studies have revealed the roles for glucocorticoid regeneration by 11β-HSD1 in liver, adipose, brain and other tissues, but until recently, there has been little focus on the heart. This article reviews the evidence for glucocorticoid metabolism by 11β-HSD1 in the heart and for a role of 11β-HSD1 activity in determining the myocardial growth and physiological function. We also consider the potential of 11β-HSD1 as a therapeutic target to enhance repair after myocardial infarction and to prevent the development of cardiac remodelling and heart failure.
Collapse
Affiliation(s)
- Gillian A Gray
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher I White
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Raphael F P Castellan
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sara J McSweeney
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Mutemberezi V, Guillemot-Legris O, Muccioli GG. Oxysterols: From cholesterol metabolites to key mediators. Prog Lipid Res 2016; 64:152-169. [PMID: 27687912 DOI: 10.1016/j.plipres.2016.09.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Oxysterols are cholesterol metabolites that can be produced through enzymatic or radical processes. They constitute a large family of lipids (i.e. the oxysterome) involved in a plethora of physiological processes. They can act through GPCR (e.g. EBI2, SMO, CXCR2), nuclear receptors (LXR, ROR, ERα) and through transporters or regulatory proteins. Their physiological effects encompass cholesterol, lipid and glucose homeostasis. Additionally, they were shown to be involved in other processes such as immune regulatory functions and brain homeostasis. First studied as precursors of bile acids, they quickly emerged as interesting lipid mediators. Their levels are greatly altered in several pathologies and some oxysterols (e.g. 4β-hydroxycholesterol or 7α-hydroxycholestenone) are used as biomarkers of specific pathologies. In this review, we discuss the complex metabolism and molecular targets (including binding properties) of these bioactive lipids in human and mice. We also discuss the genetic mouse models currently available to interrogate their effects in pathophysiological settings. We also summarize the levels of oxysterols reported in two key organs in oxysterol metabolism (liver and brain), plasma and cerebrospinal fluid. Finally, we consider future opportunities and directions in the oxysterol field in order to gain a better insight and understanding of the complex oxysterol system.
Collapse
Affiliation(s)
- Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium.
| |
Collapse
|
21
|
Noguchi N, Urano Y, Takabe W, Saito Y. New aspects of 24(S)-hydroxycholesterol in modulating neuronal cell death. Free Radic Biol Med 2015; 87:366-72. [PMID: 26164631 DOI: 10.1016/j.freeradbiomed.2015.06.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/26/2015] [Accepted: 06/27/2015] [Indexed: 02/05/2023]
Abstract
24(S)-Hydroxycholesterol (24S-OHC), which is enzymatically produced in the brain, has been known to play an important role in maintaining cholesterol homeostasis in the brain and has been proposed as a possible biomarker of neurodegenerative disease. Recent studies have revealed diverse functions of 24S-OHC and gained increased attention. For example, 24S-OHC at sublethal concentrations has been found to induce an adaptive response via activation of the liver X receptor signaling pathway, thereby protecting neuronal cells against subsequent oxidative stress. It has also been found that physiological concentrations of 24S-OHC suppress amyloid-β production via downregulation of amyloid precursor protein trafficking in neuronal cells. On the other hand, high concentrations of 24S-OHC have been found to induce a type of nonapoptotic programmed cell death in neuronal cells expressing little caspase-8. Because neuronal cell death induced by 24S-OHC has been found to proceed by a unique mechanism, which is different from but in some ways similar to necroptosis-necroptosis being a type of programmed necrosis induced by tumor necrosis factor α-neuronal cell death induced by 24S-OHC has been called "necroptosis-like" cell death. 24S-OHC-induced cell death is dependent on the formation of 24S-OHC esters but not on oxidative stress. This review article discusses newly reported aspects of 24S-OHC in neuronal cell death and sheds light on the possible importance of controlling 24S-OHC levels in the brain for preventing neurodegenerative disease.
Collapse
Affiliation(s)
- Noriko Noguchi
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan.
| | - Yasuomi Urano
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Wakako Takabe
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Yoshiro Saito
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|
22
|
Odermatt A, Klusonova P. 11β-Hydroxysteroid dehydrogenase 1: Regeneration of active glucocorticoids is only part of the story. J Steroid Biochem Mol Biol 2015; 151:85-92. [PMID: 25151952 DOI: 10.1016/j.jsbmb.2014.08.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/13/2014] [Accepted: 08/14/2014] [Indexed: 11/20/2022]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11β-HSD1) is an endoplasmic reticulum membrane enzyme with its catalytic site facing the luminal space. It functions primarily as a reductase, driven by the supply of its cosubstrate NADPH by hexose-6-phosphate dehydrogenase (H6PDH). Extensive research has been performed on the role of 11β-HSD1 in the regeneration of active glucocorticoids and its role in inflammation and metabolic disease. Besides its important role in the fine-tuning of glucocorticoid action, 11β-HSD1 is a multi-functional carbonyl reductase converting several 11- and 7-oxosterols into the respective 7-hydroxylated forms. Moreover, 11β-HSD1 has a role in phase I biotransformation reactions and catalyzes the carbonyl reduction of several non-steroidal xenobiotics. Recent observations from experiments using selective inhibitors and studies with transgenic mice indicated a role for 11β-HSD1 in oxysterol metabolism and in bile acid homeostasis, with evidence for glucocorticoid-independent effects on gene expression. This review focuses on the promiscuity of 11β-HSD1 to accept structurally distinct substrates and discusses recent progress mainly on non-glucocorticoid substrates. This article is part of a Special Issue entitled 'Enzyme Promiscuity and Diversity'.
Collapse
Affiliation(s)
- Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Petra Klusonova
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
23
|
Mitić T, Shave S, Semjonous N, McNae I, Cobice DF, Lavery GG, Webster SP, Hadoke PWF, Walker BR, Andrew R. 11β-Hydroxysteroid dehydrogenase type 1 contributes to the balance between 7-keto- and 7-hydroxy-oxysterols in vivo. Biochem Pharmacol 2013; 86:146-53. [PMID: 23415904 PMCID: PMC3694296 DOI: 10.1016/j.bcp.2013.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 01/19/2023]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11βHSD1; EC 1.1.1.146) generates active glucocorticoids from inert 11-keto metabolites. However, it can also metabolize alternative substrates, including 7β-hydroxy- and 7-keto-cholesterol (7βOHC, 7KC). This has been demonstrated in vitro but its consequences in vivo are uncertain. We used genetically modified mice to investigate the contribution of 11βHSD1 to the balance of circulating levels of 7KC and 7βOHC in vivo, and dissected in vitro the kinetics of the interactions between oxysterols and glucocorticoids for metabolism by the mouse enzyme. Circulating levels of 7KC and 7βOHC in mice were 91.3 ± 22.3 and 22.6 ± 5.7 nM respectively, increasing to 1240 ± 220 and 406 ± 39 nM in ApoE−/− mice receiving atherogenic western diet. Disruption of 11βHSD1 in mice increased (p < 0.05) the 7KC/7βOHC ratio in plasma (by 20%) and also in isolated microsomes (2 fold). The 7KC/7βOHC ratio was similarly increased when NADPH generation was restricted by disruption of hexose-6-phosphate dehydrogenase. Reduction and oxidation of 7-oxysterols by murine 11βHSD1 proceeded more slowly and substrate affinity was lower than for glucocorticoids. in vitro 7βOHC was a competitive inhibitor of oxidation of corticosterone (Ki = 0.9 μM), whereas 7KC only weakly inhibited reduction of 11-dehydrocorticosterone. However, supplementation of 7-oxysterols in cultured cells, secondary to cholesterol loading, preferentially slowed reduction of glucocorticoids, rather than oxidation. Thus, in mouse, 11βHSD1 influenced the abundance and balance of circulating and tissue levels of 7βOHC and 7KC, promoting reduction of 7KC. In health, 7-oxysterols are unlikely to regulate glucocorticoid metabolism. However, in hyperlipidaemia, 7-oxysterols may inhibit glucocorticoid metabolism and modulate signaling through corticosteroid receptors.
Collapse
Affiliation(s)
- Tijana Mitić
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|