1
|
Jalil A, Pilot T, Bourgeois T, Laubriet A, Li X, Diedisheim M, Deckert V, Magnani C, Le Guern N, Pais de Barros JP, Nguyen M, Pallot G, Vouilloz A, Proukhnitzky L, Hermetet F, Aires V, Lesniewska E, Lagrost L, Auwerx J, Le Goff W, Venteclef N, Steinmetz E, Thomas C, Masson D. Plasmalogen remodeling modulates macrophage response to cytotoxic oxysterols and atherosclerotic plaque vulnerability. Cell Rep Med 2025; 6:102131. [PMID: 40345182 DOI: 10.1016/j.xcrm.2025.102131] [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: 03/22/2024] [Revised: 11/18/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Essential fatty acid metabolism in myeloid cells plays a critical but underexplored role in immune function. Here, we demonstrate that simultaneous inactivation of two key enzymes involved in macrophage polyunsaturated fatty acid (PUFA) metabolism-ELOVL5, which elongates long-chain PUFAs, and LPCAT3, which incorporates them into phospholipids-disrupts membrane organization by promoting the formation of cholesterol-enriched domains. This increases macrophage sensitivity to cytotoxic oxysterols and leads to more vulnerable atherosclerotic plaques with enlarged necrotic cores in a mouse model of atherosclerosis. In humans, analysis of 187 carotid plaques reveals a positive correlation between LPCAT3/ELOVL5-generated phospholipids-including arachidonate (C20:4 n-6)-containing ether lipids-and more stable plaque profiles. Additionally, Mendelian randomization analysis supports a causal relationship between LPCAT3 expression and reduced risk of ischemic stroke. Our findings uncover a regulatory circuit essential for PUFA-containing phospholipid generation in macrophages, positioning PUFA-containing ether lipids as promising biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Antoine Jalil
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thomas Pilot
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thibaut Bourgeois
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Aline Laubriet
- CHRU Dijon Bourgogne, Department of Cardiovascular Surgery, Dijon University Medical Center, 21000 Dijon, France
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marc Diedisheim
- Centre - Clinique Saint Gatien Alliance (NCT+), 37214 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Valérie Deckert
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Charlène Magnani
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Naig Le Guern
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytic Platform, UBFC, 21000 Dijon, France
| | - Maxime Nguyen
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Department of Anesthesiology and Critical Care Medicine, Dijon University Medical Center, 21000 Dijon, France
| | - Gaëtan Pallot
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Adrien Vouilloz
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Lil Proukhnitzky
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - François Hermetet
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Virginie Aires
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Eric Lesniewska
- Université Bourgogne, UMR1231, 21000 Dijon, France; Laboratory of Physics, National Center for Scientific Research, URA 5027, UFR Sciences et techniques, 21000 Dijon, France
| | - Laurent Lagrost
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Wilfried Le Goff
- Sorbonne Université, Inserm, ICAN Institut, UMR_S1166, Hôpital de la Pitié, 75013 Paris, France
| | - Nicolas Venteclef
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Eric Steinmetz
- CHRU Dijon Bourgogne, Department of Cardiovascular Surgery, Dijon University Medical Center, 21000 Dijon, France
| | - Charles Thomas
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - David Masson
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France.
| |
Collapse
|
2
|
Meng Y, Zhang J, Liu Y, Zhu Y, Lv H, Xia F, Guo Q, Shi Q, Qiu C, Wang J. The biomedical application of inorganic metal nanoparticles in aging and aging-associated diseases. J Adv Res 2025; 71:551-570. [PMID: 38821357 DOI: 10.1016/j.jare.2024.05.023] [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: 10/30/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Aging and aging-associated diseases (AAD), including neurodegenerative disease, cancer, cardiovascular diseases, and diabetes, are inevitable process. With the gradual improvement of life style, life expectancy is gradually extended. However, the extended lifespan has not reduced the incidence of disease, and most elderly people are in ill-health state in their later years. Hence, understanding aging and AAD are significant for reducing the burden of the elderly. Inorganic metal nanoparticles (IMNPs) predominantly include gold, silver, iron, zinc, titanium, thallium, platinum, cerium, copper NPs, which has been widely used to prevent and treat aging and AAD due to their superior properties (essential metal ions for human body, easily synthesis and modification, magnetism). Therefore, a systematic review of common morphological alternations of senescent cells, altered genes and signal pathways in aging and AAD, and biomedical applications of IMNPs in aging and AAD is crucial for the further research and development of IMNPs in aging and AAD. This review focus on the existing research on cellular senescence, aging and AAD, as well as the applications of IMNPs in aging and AAD in the past decade. This review aims to provide cutting-edge knowledge involved with aging and AAD, the application of IMNPs in aging and AAD to promote the biomedical application of IMNPs in aging and AAD.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haining Lv
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qianli Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- Department of Urology, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
3
|
Vejux A, Ghzaiel I, Mackrill JJ, Dias IHK, Rezig L, Ksila M, Zarrouk A, Nury T, Brahmi F, El Midaoui A, Meziane S, Atanasov AG, Hammami S, Latruffe N, Jouanny P, Lizard G. Oxysterols, age-related-diseases and nutritherapy: Focus on 7-ketocholesterol and 7β-hydroxycholesterol. Prostaglandins Other Lipid Mediat 2025; 178:106993. [PMID: 40216356 DOI: 10.1016/j.prostaglandins.2025.106993] [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: 10/07/2024] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Age-related diseases are often associated with a disruption of RedOx balance that can lead to lipid peroxidation with the formation of oxysterols, especially those oxidized on carbon-7: 7-ketocholesterol (also known as 7-oxo-cholesterol) and 7β-hydroxycholesterol. Like cholesterol, these oxysterols have 27 carbons, they are composed of a sterane nucleus and have a hydroxyl function in position 3. The oxysterols 7-ketocholesterol and 7β-hydroxycholesterol are mainly formed by cholesterol autoxidation and are biomarkers of oxidative stress. These two oxysterols are frequently found at increased levels in the biological fluids (plasma, cerebrospinal fluid), tissues and/or organs (arterial wall, retina, brain) of patients with age-related diseases, especially cardiovascular diseases, neurodegenerative diseases (mainly Alzheimer's disease), ocular diseases (cataract, age-related macular degeneration), and sarcopenia. Depending on the cell type considered, 7-ketocholesterol and 7β-hydroxycholesterol induce either caspase- dependent or -independent types of cell death associated with mitochondrial and peroxisomal dysfunctions, autophagy and oxidative stress. The caspase dependent type of cell death associated with oxidative stress and autophagy is defined as oxiapoptophagy. These two oxysterols are also inducers of inflammation. These biological features associated with the toxicity of 7-ketocholesterol, and 7β-hydroxycholesterol are often observed in patients with age-related diseases, suggesting an involvement of these oxysterols in the pathophysiology of these disorders. The cytotoxic effects of 7-ketocholesterol and 7β-hydroxycholesterol are counteracted on different cell models by representative nutrients of the Mediterranean diet: ω3 and ω9 fatty acids, polyphenols, and tocopherols. There are also evidences, mainly in cardiovascular diseases, of the benefits of α-tocopherol and phenolic compounds. These in vitro and in vivo observations on 7-ketocholesterol and 7β-hydroxycholesterol, which are frequently increased in age-related diseases, reinforce the interest of nutritherapeutic treatments to prevent and/or cure age-related diseases currently without effective therapies.
Collapse
Affiliation(s)
- Anne Vejux
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université de Bourgogne Europe, 21000 Dijon, France; Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France.
| | - Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France; Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - John J Mackrill
- Department of Physiology, University College Cork, Western Gateway Building, Western Road, Cork T12 XF62, Ireland
| | - Irundika H K Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - 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
| | - Mohamed Ksila
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Amira Zarrouk
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; Faculty of Medicine of Sousse, avenue Mohamed Karaoui, 4002 Sousse, Tunisia
| | - Thomas Nury
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Fatiha Brahmi
- Laboratory of Biomathematics, Biophysics, Biochemistry, and Scientometrics (L3BS), Faculty of Nature and Life Sciences, University of Bejaia, 06000 Bejaia, Algeria
| | - Adil El Midaoui
- Department of Biology, Faculty of Sciences and Techniques of Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Smail Meziane
- Institut Européen des Antioxydants, 1B Rue Victor de Lespinats, 54230 Neuves-Maisons, France
| | - Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Magdalenka, Jastrzebiec, Poland
| | - Sonia Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - Norbert Latruffe
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université de Bourgogne Europe, 21000 Dijon, France; Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France
| | - Pierre Jouanny
- Geriatric Internal Medicine Department (Champmaillot), University Hospital Center, Université de Bourgogne Europe, 21000 Dijon, France
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne Europe, 21000 Dijon, France.
| |
Collapse
|
4
|
Mahmoudi Z, Tajik A, Vahdat M, Mobarakeh KA, Saeedirad Z, Azaryan F, Amjadi A, Alami F, Valisoltani N, Mirshafaei MA, Khoshdooz S, Gholamalizadeh M, Doaei S, Kooshki A. The association between dietary intake of fats and transient global amnesia (TGA). Nutr Neurosci 2025; 28:149-155. [PMID: 39012761 DOI: 10.1080/1028415x.2024.2359866] [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] [Indexed: 07/18/2024]
Abstract
BACKGROUND Different types of dietary fat may influence memory and cognitive functions. This study aimed to investigate the association between dietary fat intake and transient global amnesia (TGA). METHODS This case-control study was conducted using Persian Sabzevar cohort data on 258 individuals with TGA and 520 individuals without amnesia in Sabzevar Iran. The food frequency questionnaire (FFQ) was used to assess the intake of dietary fats of the participants. All study participants were screened for TGA by a neurologist and their status was determined based on the diagnostic symptoms defined by the Kaplan and Hodges criteria. RESULTS There was an inverse association between the risk of TGA and dietary intake of alpha-linolenic acid (ALA) (OR = 0.94, CI95%:0.88-0.99, P = 0.01). Also, a positive association was observed between TGA and dietary intake of n-6 fatty acids (OR = 1.18, CI 95%: 1.04-1.33, P = 0.01). The results remained significant after adjustment for age, sex, education, job, marital status, physical activity, BMI, and calorie intake. CONCLUSION Omega-3 fatty acids may have beneficial effects; however, omega-6 fatty acids may have adverse effects on the risk of amnesia. Further longitudinal studies are warranted.
Collapse
Affiliation(s)
- Zahra Mahmoudi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Tajik
- University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mahsa Vahdat
- Department of Nutrition, School of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Khadijeh Abbasi Mobarakeh
- Nutrition and Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Saeedirad
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Azaryan
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Arezoo Amjadi
- Department of Nutrition, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farkhondeh Alami
- Student Research Committee, Department of Nutrition, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Neda Valisoltani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoomeh Alsadat Mirshafaei
- Department of Physical Education and Sport Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Sara Khoshdooz
- Faculty of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Doaei
- Department of Community Nutrition, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Kooshki
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
5
|
Radhakrishnan K, Zhang Y, Mustapha O, Weigel TK, Upchurch CM, Tian X, Herbert F, Huang W, Leitinger N, Eyo UB, Ai H, Ferris HA. 7-ketocholesterol contributes to microglia-driven increases in astrocyte reactive oxygen species in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633810. [PMID: 39868327 PMCID: PMC11761689 DOI: 10.1101/2025.01.19.633810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oxidative stress is a prominent feature of Alzheimer's disease. Within this context, cholesterol undergoes oxidation, producing the pro-inflammatory product 7-ketocholesterol (7-KC). In this study, we observe elevated levels of 7-KC in the brains of the 3xTg mouse model of AD. To further understand the contribution of 7-KC on the oxidative environment, we developed a method to express a genetically encoded fluorescent hydrogen peroxide (H2O2) sensor in astrocytes, the primary source of cholesterol in the brain. With this sensor, we discovered that 7-KC increases H2O2 levels in astrocytes in vivo, but not when directly applied to astrocytes in vitro. Interestingly, when 7-KC was applied to a microglia cell line alone or mixed astrocyte and microglia cultures, it resulted in microglia activation and increased oxidative stress in astrocytes. Depletion of microglia from 3xTg mice resulted in reduced 7-KC in the brains of these mice. Taken together, these findings suggest that 7-KC, acting through microglia, contributes to increased astrocyte oxidative stress in AD. This study sheds light on the complex interplay between cholesterol oxidation, microglia activation, and astrocyte oxidative stress in the pathogenesis of AD.
Collapse
Affiliation(s)
- Kayalvizhi Radhakrishnan
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Oluwaseun Mustapha
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Thaddeus K. Weigel
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Franklin Herbert
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Wenyuan Huang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Heather A. Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
6
|
Krishnan M, Kumaresan M, Ravi S, Martin LC, Duraisamy P, Manikandan B, Munusamy A, Ramar M. Therapeutic potential of monoterpene molecules acts against 7KCh-mediated oxidative stress and neuroinflammatory amyloidogenic signalling pathways. Prostaglandins Other Lipid Mediat 2024; 175:106910. [PMID: 39343044 DOI: 10.1016/j.prostaglandins.2024.106910] [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: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
Alzheimer's disease (AD) is a degenerative disorder characterised by amyloid-beta aggregates activated by the accumulation of lipid molecules and their derivatives, especially 7-ketocholesterol (7KCh), an oxidised lipid that plays a great part in the progression of AD. The current therapeutics need bio-potential molecules and their biomedical application preventing 7KCh-induced cytotoxicity. In this study, bornyl acetate (BA) and menthol (ME), the natural monoterpenes were investigated for their neuroprotective effects against 7KCh-induced SH-SY5Y cells and their effects were compared to the standard drug galantamine (GA). 7KCh-induced changes like lipid accumulation, amyloid generation, free radical generation, acetylcholinesterase levels, calcium accumulation and mitochondrial membrane integrity were analysed in SH-SY5Y cells with or without BA and ME treatment. Furthermore, various mediators involved in the amyloidogenic, inflammatory and apoptotic pathways were studied. In our results, the cells induced with 7KCh upon co-treatment with BA and ME significantly reduced lipid accumulation and amyloid generation through toll-like receptor (TLR) 4 suppression and enhanced ATP binding cassette (ABCA) 1-mediated clearance. Co-treatment with BA and ME concurrently regulated oxidative stress, acetylcholinesterase activity, mitochondrial membrane potential and intracellular calcification altered by 7KCh-induced SH-SY5Y cells. Moreover, 7KCh-induced cells showed elevated mRNA levels of misfolded protein markers and apoptotic mediators which were significantly downregulated by BA and ME co-treatment. In addition, the protein expression of amyloidogenic, proinflammatory as well as pro-apoptotic markers was decreased by BA and ME co-treatment in 7KCh-induced cells. Overall, BA and ME mediated inhibition of amyloidogenic activation and cell survival against 7KCh-induced inflammation, thereby preventing the onset and progression of AD in comparison to GA.
Collapse
Affiliation(s)
- Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Arumugam Munusamy
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
7
|
Song L, Xue J, Xu L, Cheng L, Zhang Y, Wang X. Muscle-specific PGC-1α modulates mitochondrial oxidative stress in aged sarcopenia through regulating Nrf2. Exp Gerontol 2024; 193:112468. [PMID: 38801840 DOI: 10.1016/j.exger.2024.112468] [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: 02/19/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Aged sarcopenia is characterized by loss of skeletal muscle mass and strength, and mitochondrial dysregulation in skeletal myocyte is considered as a major factor. Here, we aimed to analyze the effects of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) on mitochondrial reactive oxygen species (ROS) and nuclear factor erythroid 2-related factor 2 (Nrf2) in aged skeletal muscles. METHODS C2C12 cells were stimulated by 50 μM 7β-hydroxycholesterol (7β-OHC) to observe the changes of cellular ROS, mitochondrial ROS, and expression of PGC-1α and Nrf2. Different PGC-1α expression in cells was established by transfection with small interfering RNA (siRNA) or plasmids overexpressing PGC-1α (pEX-3-PGC-1α). The effects of different PGC-1α expression on cellular ROS, mitochondrial ROS and Nrf2 expression were measured in cells. Wild type (WT) mice and PGC-1α conditional knockout (CKO) mice were used to analyze the effects of PGC-1α on aged sarcopenia and expression of Nrf2 and CD38 in gastrocnemius muscles. Diethylmaleate, a Nrf2 activator, was used to analyze the connection between PGC-1α and Nrf2 in cells and in mice. RESULTS In C2C12 cells, the expressions of PGC-1α and Nrf2 were declined by the 7β-OHC treatment or PGC-1α silence. Moreover, PGC-1α silence increased the harmful ROS and decreased the Nrf2 protein expression in the 7β-OHC-treated cells. PGC-1α overexpression decreased the harmful ROS and increased the Nrf2 protein expression in the 7β-OHC-treated cells. Diethylmaleate treatment decreased the harmful ROS in the 7β-OHC-treated or PGC-1α siRNA-transfected cells. At the same age, muscle-specific PGC-1α deficiency aggravated aged sarcopenia, decreased Nrf2 expression and increased CD38 expression in gastrocnemius muscles compared with the WT mice. Diethylmaleate treatment improved the muscle function and decreased the CD38 expression in the old two genotypes. CONCLUSIONS Our study demonstrated that PGC-1α modulated mitochondrial oxidative stress in aged sarcopenia through regulating Nrf2.
Collapse
Affiliation(s)
- Lei Song
- Geriatric Medicine Department, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Jianfeng Xue
- Geriatric Cardiovascular Department, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Lingfen Xu
- General Medicine Department, Qinghai Provincial Hospital, Xining 810000, China
| | - Lin Cheng
- Geriatric Medicine Department, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Yongxia Zhang
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai 264000, China.
| | - Xiaojun Wang
- Geriatric Medicine Department, Yantai Yuhuangding Hospital, Yantai 264000, China.
| |
Collapse
|
8
|
Blanco-Morales V, Mercatante D, Faubel N, Miedes D, Mandrioli M, Rodriguez-Estrada MT, Garcia-Llatas G. Lipolysis and Sterol Stability and Bioaccessibility of Wholemeal Rye Bread Enriched with Plant Sterols Subjected to Adult and Elderly Digestion Conditions. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16976-16987. [PMID: 39037854 PMCID: PMC11299168 DOI: 10.1021/acs.jafc.4c03104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024]
Abstract
This study evaluated the impact of different digestion conditions (adult and senior) on lipolysis and bioaccessibility of plant sterols (PS) and phytosterol oxidation products (POPs) in PS-enriched wholemeal rye bread. Under adult digestion conditions, the addition of gastric lipase (GL) reduced lipolysis products (by 6.1% for free fatty acids and 11.7% for monoacylglycerols) and the bioaccessibility of PS by 6.7%, compared to the control. In digestion with both GL and cholesterol esterase (CE), these reductions were 12.9, 20.1, and 11.3%, respectively. Both modifications (GL and GL + CE) increased the bioaccessibility of POPs by 4.5-4.0%. When simulating the elderly digestion, the modified gastric and intestinal phases did not alter PS bioaccessibility but decreased POPs bioaccessibility by 21.8% compared to control, along with reduced lipolysis. Incorporating GL and CE thus approached physiological conditions and influenced lipid digestion. Elderly simulated digestion conditions resulted in a positive outcome by maintaining PS bioaccessibility while reducing potentially harmful POPs.
Collapse
Affiliation(s)
- Virginia Blanco-Morales
- Nutrition
and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Avda. Vicente Andrés Estellés s/n,
Burjassot, 46100 Valencia, Spain
| | - Dario Mercatante
- Department
of Agricultural and Food Sciences, Alma
Mater Studiorum-Università di Bologna, Viale Fanin 40, Bologna 40127, Italy
| | - Nerea Faubel
- Nutrition
and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Avda. Vicente Andrés Estellés s/n,
Burjassot, 46100 Valencia, Spain
| | - Diego Miedes
- Nutrition
and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Avda. Vicente Andrés Estellés s/n,
Burjassot, 46100 Valencia, Spain
| | - Mara Mandrioli
- Department
of Agricultural and Food Sciences, Alma
Mater Studiorum-Università di Bologna, Viale Fanin 40, Bologna 40127, Italy
| | - Maria Teresa Rodriguez-Estrada
- Department
of Agricultural and Food Sciences, Alma
Mater Studiorum-Università di Bologna, Viale Fanin 40, Bologna 40127, Italy
| | - Guadalupe Garcia-Llatas
- Nutrition
and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Avda. Vicente Andrés Estellés s/n,
Burjassot, 46100 Valencia, Spain
| |
Collapse
|
9
|
Kopp W. Aging and "Age-Related" Diseases - What Is the Relation? Aging Dis 2024; 16:1316-1346. [PMID: 39012663 PMCID: PMC12096902 DOI: 10.14336/ad.2024.0570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024] Open
Abstract
The study explores the intricate relationship between aging and the development of noncommunicable diseases [NCDs], focusing on whether these diseases are inevitable consequences of aging or primarily driven by lifestyle factors. By examining epidemiological data, particularly from hunter-gatherer societies, the study highlights that many NCDs prevalent in modern populations are rare in these societies, suggesting a significant influence of lifestyle choices. It delves into the mechanisms through which poor diet, smoking, and other lifestyle factors contribute to systemic physiological imbalances, characterized by oxidative stress, insulin resistance and hyperinsulinemia, and dysregulation of the sympathetic nervous system, the renin-angiotensin-aldosterone system, and the immune system. The interplay between this pattern and individual factors such as genetic susceptibility, biological variability, epigenetic changes and the microbiome is proposed to play a crucial role in the development of a range of age-related NCDs. Modified biomolecules such as oxysterols and advanced glycation end products also contribute to their development. Specific diseases such as benign prostatic hyperplasia, Parkinson's disease, glaucoma and osteoarthritis are analyzed to illustrate these mechanisms. The study concludes that while aging contributes to the risk of NCDs, lifestyle factors play a crucial role, offering potential avenues for prevention and intervention through healthier living practices. One possible approach could be to try to restore the physiological balance, e.g. through dietary measures [e.g. Mediterranean diet, Okinawan diet or Paleolithic diet] in conjunction with [a combination of] pharmacological interventions and other lifestyle changes.
Collapse
Affiliation(s)
- Wolfgang Kopp
- Retired head of the Diagnostikzentrum Graz, Mariatrosterstrasse 41, 8043 Graz, Austria
| |
Collapse
|
10
|
Maaloul S, Ghzaiel I, Mahmoudi M, Mighri H, Pires V, Vejux A, Martine L, de Barros JPP, Prost-Camus E, Boughalleb F, Lizard G, Abdellaoui R. Characterization of Silybum marianum and Silybum eburneum seed oils: Phytochemical profiles and antioxidant properties supporting important nutritional interests. PLoS One 2024; 19:e0304021. [PMID: 38875282 PMCID: PMC11178192 DOI: 10.1371/journal.pone.0304021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/03/2024] [Indexed: 06/16/2024] Open
Abstract
Milk thistle seed oil is still not a well-known edible oil. Silybum marianum (milk thistle), is present in several countries and is the only known representative of the genus Silybum. However, Silybum eburneum, which is an endemic plant in Spain, Kenya, Morocco, Algeria, and Tunisia, is considered a marginalized species. The present work is the first report that gives information on the lipid and phenolic profiles of Tunisian S. eburneum seed oil compared to those of Tunisian S. marianum seed oil. In addition, the antioxidant properties of these oils were determined with DPPH, FRAP, and KRL assays, and their ability to prevent oxidative stress was determined on human monocytic THP-1 cells. These oils are characterized by high amounts of unsaturated fatty acids; linoleic acid and oleic acid are the most abundant. Campesterol, sitosterol, stigmasterol, and β-amyrin were the major phytosterols identified. α-tocopherol was the predominant tocopherol found. These oils also contain significant amounts of phenolic compounds. The diversity and richness of Silybum marianum and Silybum eburneum seed oils in unsaturated fatty acids, phenolic compounds, and tocopherols are associated with high antioxidant activities revealed by the DPPH, FRAP, and KRL assays. In addition, on THP-1 cells, these oils powerfully reduced the oxidative stress induced by 7-ketocholesterol and 7β-hydroxycholesterol, two strongly pro-oxidant oxysterols often present at increased levels in patients with age-related diseases. Silybum marianum and Silybum eburneum seed oils are therefore important sources of bioactive molecules with nutritional interest that prevent age-related diseases, the frequency of which is increasing in all countries due to the length of life expectancy.
Collapse
Affiliation(s)
- Samah Maaloul
- Laboratory of Rangeland Ecosystems and Valorisation of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| | - Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) University of Bourgogne/Inserm, Dijon, France
- University Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut Pascal, Clermont-Ferrand, France
| | - Maher Mahmoudi
- Laboratory of Rangeland Ecosystems and Valorisation of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
- Laboratory of Plant, Soil and Environement Interactions (LR21ES01)-University of Tunis El-Manar, Faculty of Sciences of Tunis, El-Manar, Tunis, Tunisia
- Laboratory of Functional Physiology and Valorization of Bio-Ressources, Higher Institute of Biotechnology of Beja (LR23ES08), University of Jendouba, Jendouba, Tunisia
| | - Hédi Mighri
- Laboratory of Rangeland Ecosystems and Valorisation of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| | - Vivien Pires
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) University of Bourgogne/Inserm, Dijon, France
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) University of Bourgogne/Inserm, Dijon, France
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | | | | | | | - Fayçal Boughalleb
- Laboratory of Rangeland Ecosystems and Valorisation of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) University of Bourgogne/Inserm, Dijon, France
| | - Raoudha Abdellaoui
- Laboratory of Rangeland Ecosystems and Valorisation of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| |
Collapse
|
11
|
Fang Z, Liu R, Xie J, He JC. Molecular mechanism of renal lipid accumulation in diabetic kidney disease. J Cell Mol Med 2024; 28:e18364. [PMID: 38837668 PMCID: PMC11151220 DOI: 10.1111/jcmm.18364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of end stage renal disease with unmet clinical demands for treatment. Lipids are essential for cell survival; however, renal cells have limited capability to metabolize overloaded lipids. Dyslipidaemia is common in DKD patients and renal ectopic lipid accumulation is associated with disease progression. Unveiling the molecular mechanism involved in renal lipid regulation is crucial for exploring potential therapeutic targets. In this review, we focused on the mechanism underlying cholesterol, oxysterol and fatty acid metabolism disorder in the context of DKD. Specific regulators of lipid accumulation in different kidney compartment and TREM2 macrophages, a lipid-related macrophages in DKD, were discussed. The role of sodium-glucose transporter 2 inhibitors in improving renal lipid accumulation was summarized.
Collapse
Affiliation(s)
- Zhengying Fang
- Department of Nephrology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ruijie Liu
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jingyuan Xie
- Department of Nephrology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - John Cijiang He
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Renal SectionJames J Peters Veterans Affair Medical CenterBronxNew YorkUSA
| |
Collapse
|
12
|
Gonet-Surówka A, Ciechacka M, Kępczyński M, Dynarowicz-Latka P. Oxysterols in Cell Viability, Phospholipidosis and Extracellular Vesicles Production in a Lung Cancer Model. Cell Biochem Biophys 2024; 82:1019-1026. [PMID: 38514528 DOI: 10.1007/s12013-024-01255-3] [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: 02/05/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
The study carried out systematic research on the influence of selected oxysterols on cells viability, phospholipidosis and the level of secreted extracellular vesicles. Three oxidized cholesterol derivatives, namely 7α-hydroxycholesterol (7α-OH), 7- ketocholesterol (7-K) and 24(S)-hydroxycholesterol (24(S)-OH) were tested in three different concentrations: 50 μM, 100 μM and 200 μM for 24 h incubation with A549 lung cancer cell line. All the studied oxysterols were found to alter cells viability. The lowest survival rate of the cells was observed after 24 h of 7-K treatment, slightly better for 7α-OH while cells incubated with 24(S)-OH had the best survival rate among the oxysterols used. 7-K increased phospholipids accumulation in cells, however, most noticeable effect was noticed for 24(S)-OH. Changes in the level of extracellular vesicles secreted in cells culture after the treatment with oxysterols were also observed. It was found that all oxysterols used increased the level of secreted vesicles, both exosomes and ectosomes. The strongest effect was noticed for 24(S)-OH. Taken together, these results suggest that 7-K is the most potent inducer of cancer cell death, while 7α-OH is slightly less potent in this respect. The lower cytotoxic effect of 24(S)-OH correlates with greater phospholipids accumulation, extracellular vesicles production and better cells survival.
Collapse
Affiliation(s)
- Agnieszka Gonet-Surówka
- Faculty of Chemistry, Department of General Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland.
| | - Mariola Ciechacka
- Faculty of Chemistry, Department of General Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Mariusz Kępczyński
- Faculty of Chemistry, Department of Physical Chemistry and Electrochemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Patrycja Dynarowicz-Latka
- Faculty of Chemistry, Department of General Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| |
Collapse
|
13
|
Zulkifli SA, Abd Gani SS, Zaidan UH, Misran A, Hassan M. In vitro anti-inflammatory and wound healing properties of defatted Selenicereus monacanthus (Lem.) D.R.Hunt seed extract. Nat Prod Res 2024:1-9. [PMID: 38742327 DOI: 10.1080/14786419.2024.2344190] [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: 11/14/2023] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
Extracts from Selenicereus monacanthus (synonym: Hylocereus polyrhizus) have received attention due to their potent anti-inflammatory, antioxidant, anticancer, and antidiabetic properties. The current study aims to determine the anti-inflammatory and wound-healing potential of defatted S. monacanthus seed extract (DSMSE). Anti-inflammatory properties of DSMSE on LPS-induced inflammation on THP-1 were determined by measuring the levels of interleukins IL-6, IL-8, and IL-10. Wound healing scratch assay was performed using the human fibroblast (Hs27) cell that assesses the cell migration over 24 h exposure to DSMSE. Administration of DSMSE significantly reduced the LPS-stimulated release levels of IL-6 and IL-8 and significantly increased the levels of IL-10. Treatment with DSMSE showed a significant increase in wound closure with 70% of fibroblast migration. Therefore, the current study showed the anti-inflammatory and wound healing properties of DSMSE reducing inflammatory cytokines (IL-6 and IL-8), increasing IL-10 cytokine, and increasing wound closure at 24 h.
Collapse
Affiliation(s)
- Siti Atikah Zulkifli
- Halal Products Research Institute, Universiti Putra Malaysia, Putra Inforport, Serdang, Selangor, Malaysia
| | - Siti Salwa Abd Gani
- Department of Agriculture Technology, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Uswatun Hasanah Zaidan
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Azizah Misran
- Department of Crop Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Masriana Hassan
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
14
|
Sun J, Xie X, Song Y, Sun T, Liu X, Yuan H, Shen C. Selenomethionine in gelatin methacryloyl hydrogels: Modulating ferroptosis to attenuate skin aging. Bioact Mater 2024; 35:495-516. [PMID: 38404642 PMCID: PMC10885793 DOI: 10.1016/j.bioactmat.2024.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024] Open
Abstract
During skin aging, the degeneration of epidermal stem cells (EpiSCs) leads to diminished wound healing capabilities and epidermal disintegration. This study tackles this issue through a comprehensive analysis combining transcriptomics and untargeted metabolomics, revealing age-dependent alterations in the Gpx gene family and arachidonic acid (AA) metabolic networks, resulting in enhanced ferroptosis. Selenomethionine (Se-Met) could enhance GPX4 expression, thereby assisting EpiSCs in countering AA-induced mitochondrial damage and ferroptosis. Additionally, Se-Met demonstrates antioxidative characteristics and extensive ultraviolet absorption. For the sustained and controllable release of Se-Met, it was covalently grafted to UV-responsive GelMA hydrogels via AC-PEG-NHS tethers. The Se-Met@GelMA hydrogel effectively accelerated wound healing in a chronological aging mice model, by inhibiting lipid peroxidation and ferroptosis with augmented GPX4 expression. Moreover, in a photoaging model, this hydrogel significantly mitigated inflammatory responses, extracellular matrix remodeling, and ferroptosis in UV-exposed mice. These characteristics render Se-Met@GelMA hydrogel valuable in practical clinical applications.
Collapse
Affiliation(s)
- Jiachen Sun
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Xiaoye Xie
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Yaoyao Song
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Tianjun Sun
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Xinzhu Liu
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Huageng Yuan
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Chuanan Shen
- Department of Burns and Plastic Surgery, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| |
Collapse
|
15
|
Mabrouk NEL, Mastouri M, Lizard G, Aouni M, Harizi H. In vitro immunotoxicity effects of carbendazim were inhibited by n-acetylcysteine in microglial BV-2 cells. Toxicol In Vitro 2024; 97:105812. [PMID: 38522494 DOI: 10.1016/j.tiv.2024.105812] [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: 11/27/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Carbendazim (CBZ) is a benzimidazole fungicide widely used worldwide in industrial, agricultural, and veterinary practices. Although, CBZ was found in all brain tissues causing serious neurotoxicity, its impact on brain immune cells remain scarcely understood. Our study investigated the in vitro effects of CBZ on activated microglial BV-2 cells. Lipopolysaccharide (LPS)-stimulated BV-2 cells were exposed to increasing concentrations of CBZ and cytokine release was measured by ELISA, and Cytometric Bead Array (CBA) assays. Mitochondrial superoxide anion (O2·-) generation was evaluated by Dihydroethidium (DHE) and nitric oxide (NO) was assessed by Griess reagent. Lipid peroxidation was evaluated by measuring the malonaldehyde (MDA) levels. The transmembrane mitochondrial potential (ΔΨm) was detected by cytometry analysis with dihexyloxacarbocyanine iodide (DiOC6(3)) assay. CBZ concentration-dependently increased IL-1β, IL-6, TNF-α and MCP-1 by LPS-activated BV-2 cells. CBZ significantly promoted oxidative stress by increasing NO, O2·- generation, and MDA levels. In contrast, CBZ significantly decreased ΔΨm. Pre-treatment of BV-2 cells with N-acetylcysteine (NAC) reversed all the above mentioned immunotoxic parameters, suggesting a potential protective role of NAC against CBZ-induced immunotoxicity via its antioxidant and anti-inflammatory effects on activated BV-2 cells. Therefore, microglial proinflammatory over-activation by CBZ may be a potential mechanism by which CBZ could induce neurotoxicity and neurodegenerative disorders.
Collapse
Affiliation(s)
- Narjesse E L Mabrouk
- Laboratoy of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy Monastir University, Avenue Avicenne, 5019 Monastir, Tunisia
| | - Maha Mastouri
- Laboratoy of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy Monastir University, Avenue Avicenne, 5019 Monastir, Tunisia
| | - Gérard Lizard
- Laboratory Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism. EA7270, INSERM, Faculty of Sciences Gabriel, University of Bourgogne Franche Comté, Dijon, France
| | - Mahjoub Aouni
- Laboratoy of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy Monastir University, Avenue Avicenne, 5019 Monastir, Tunisia
| | - Hedi Harizi
- Laboratoy of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy Monastir University, Avenue Avicenne, 5019 Monastir, Tunisia.
| |
Collapse
|
16
|
Chen J, Zhang J, Cai L, Guo L, Cai Z, Han H, Zhang W. Cholestane-3β,5α,6β-triol Induces Multiple Cell Death in A549 Cells via ER Stress and Autophagy Activation. Mar Drugs 2024; 22:174. [PMID: 38667791 PMCID: PMC11051220 DOI: 10.3390/md22040174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Cholestane-3β,5α,6β-triol (CT) and its analogues are abundant in natural sources and are reported to demonstrate cytotoxicity toward different kinds of tumor cells without a deep probe into their mechanism of action. CT is also one of the major metabolic oxysterols of cholesterol in mammals and is found to accumulate in various diseases. An extensive exploration of the biological roles of CT over the past few decades has established its identity as an apoptosis inducer. In this study, the effects of CT on A549 cell death were investigated through cell viability assays. RNA-sequencing analysis and western blot of CT-treated A549 cells revealed the role of CT in inducing endoplasmic reticulum (ER) stress response and enhancing autophagy flux, suggesting a putative mechanism of CT-induced cell-death activation involving reactive oxygen species (ROS)-mediated ER stress and autophagy. It is reported for the first time that the upregulation of autophagy induced by CT can serve as a cellular cytotoxicity response in accelerating CT-induced cell death in A549 cells. This research provides evidence for the effect of CT as an oxysterol in cell response to oxidative damage and allows for a deep understanding of cholesterol in its response in an oxidative stress environment that commonly occurs in the progression of various diseases.
Collapse
Affiliation(s)
- Jiaxi Chen
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Jieping Zhang
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Lijuan Cai
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Li Guo
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Zhenyu Cai
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Hua Han
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
| | - Wen Zhang
- School of Medicine, Tongji University, 1239 Si-Ping Road, Shanghai 200092, China
- Ningbo Institute of Marine Medicine, Peking University, 56 Kang-Da Road, Ningbo 315832, China
| |
Collapse
|
17
|
Chen Y, Wu J. Aging-Related Sarcopenia: Metabolic Characteristics and Therapeutic Strategies. Aging Dis 2024; 16:1003-1022. [PMID: 38739945 PMCID: PMC11964442 DOI: 10.14336/ad.2024.0407] [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: 12/27/2023] [Accepted: 04/07/2024] [Indexed: 05/16/2024] Open
Abstract
The proportion of the elderly population is gradually increasing as a result of medical care advances, leading to a subsequent surge in geriatric diseases that significantly impact quality of life and pose a substantial healthcare burden. Sarcopenia, characterized by age-related decline in skeletal muscle mass and quality, affects a considerable portion of older adults, particularly the elderly, and can result in adverse outcomes such as frailty, fractures, bedridden, hospitalization, and even mortality. Skeletal muscle aging is accompanied by underlying metabolic changes. Therefore, elucidating these metabolic profiles and specific mechanisms holds promise for informing prevention and treatment strategies for sarcopenia. This review provides a comprehensive overview of the key metabolites identified in current clinical studies on sarcopenia and their potential pathophysiological alterations in metabolic activity. Besides, we examine potential therapeutic strategies for sarcopenia from a perspective focused on metabolic regulation.
Collapse
Affiliation(s)
| | - Jinhui Wu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
18
|
Bertolotti M, Lancellotti G, Mussi C. Changes in cholesterol homeostasis associated with aging and with age-related conditions: pathophysiological and clinical implications. JOURNAL OF GERONTOLOGY AND GERIATRICS 2024; 72:1-11. [DOI: 10.36150/2499-6564-n637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Yammine A, Ghzaiel I, Pires V, Zarrouk A, Kharoubi O, Greige-Gerges H, Auezova L, Lizard G, Vejux A. Cytoprotective effects of α-linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, oleic acid and α-tocopherol on 7-ketocholesterol - Induced oxiapoptophagy: Major roles of PI3-K / PDK-1 / Akt signaling pathway and glutathione peroxidase activity in cell rescue. Curr Res Toxicol 2024; 6:100153. [PMID: 38379847 PMCID: PMC10877125 DOI: 10.1016/j.crtox.2024.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
On murine N2a cells, 7-ketocholesterol induced an oxiapotophagic mode of cell death characterized by oxidative stress (reactive oxygen species overproduction on whole cells and at the mitochondrial level; lipid peroxidation), apoptosis induction (caspase-9, -3 and -7 cleavage, PARP degradation) and autophagy (increased ratio LC3-II / LC3-I). Oxidative stress was strongly attenuated by diphenyleneiodonium chloride which inhibits NAD(P)H oxidase. Mitochondrial and peroxisomal morphological and functional changes were also observed. Down regulation of PDK1 / Akt signaling pathways as well as of GSK3 / Mcl-1 and Nrf2 pathways were simultaneously observed in 7-ketocholesterol-induced oxiapoptophagy. These events were prevented by α-linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, oleic acid and α-tocopherol. The inhibition of the cytoprotection by LY-294002, a PI3-K inhibitor, demonstrated an essential role of PI3-K in cell rescue. The rupture of oxidative stress in 7-ketocholesterol-induced oxiapoptophagy was also associated with important modifications of glutathione peroxidase, superoxide dismutase and catalase activities as well as of glutathione peroxidase-1, superoxide dismutase-1 and catalase level and expression. These events were also counteracted by α-linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, oleic acid and α-tocopherol. The inhibition of the cytoprotection by mercaptosuccinic acid, a glutathione peroxidase inhibitor, showed an essential role of this enzyme in cell rescue. Altogether, our data support that the reactivation of PI3-K and glutathione peroxidase activities by α-linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, oleic acid and α-tocopherol are essential to prevent 7KC-induced oxiapoptophagy.
Collapse
Affiliation(s)
- Aline Yammine
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270 / Inserm, University of Bourgogne, 21000 Dijon, France
- Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon
| | - Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270 / Inserm, University of Bourgogne, 21000 Dijon, France
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut Pascal, F-63000 Clermont-Ferrand, France
| | - Vivien Pires
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270 / Inserm, University of Bourgogne, 21000 Dijon, France
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, F-21000 Dijon, France
| | - Amira Zarrouk
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
- Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
| | - Omar Kharoubi
- University Oran 1 ABB: Laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, Oran, Algeria
| | - Hélène Greige-Gerges
- Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon
| | - Lizette Auezova
- Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270 / Inserm, University of Bourgogne, 21000 Dijon, France
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270 / Inserm, University of Bourgogne, 21000 Dijon, France
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, F-21000 Dijon, France
| |
Collapse
|
20
|
Ghosh S, Ghzaiel I, Vejux A, Meaney S, Nag S, Lizard G, Tripathi G, Naez F, Paul S. Impact of Oxysterols in Age-Related Disorders and Strategies to Alleviate Adverse Effects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:163-191. [PMID: 38036880 DOI: 10.1007/978-3-031-43883-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Oxysterols or cholesterol oxidation products are a class of molecules with the sterol moiety, derived from oxidative reaction of cholesterol through enzymatic and non-enzymatic processes. They are widely reported in animal-origin foods and prove significant involvement in the regulation of cholesterol homeostasis, lipid transport, cellular signaling, and other physiological processes. Reports of oxysterol-mediated cytotoxicity are in abundance and thus consequently implicated in several age-related and lifestyle disorders such as cardiovascular diseases, bone disorders, pancreatic disorders, age-related macular degeneration, cataract, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and some types of cancers. In this chapter, we attempt to review a selection of physiologically relevant oxysterols, with a focus on their formation, properties, and roles in health and disease, while also delving into the potential of natural and synthetic molecules along with bacterial enzymes for mitigating oxysterol-mediated cell damage.
Collapse
Affiliation(s)
- Shubhrima Ghosh
- Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Imen Ghzaiel
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Anne Vejux
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Steve Meaney
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin 7, Ireland
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Garima Tripathi
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Falal Naez
- Department of Microbiology, Vijaygarh Jyotish Ray College, University of Calcutta, Kolkata, India
| | - Srijita Paul
- Department of Microbiology, Gurudas College, Kolkata, West Bengal, India
| |
Collapse
|
21
|
Ghzaiel I, Maaloul S, Ksila M, Namsi A, Yammine A, Debbabi M, Badreddine A, Meddeb W, Pires V, Nury T, Ménétrier F, Avoscan L, Zarrouk A, Baarine M, Masmoudi-Kouki O, Ghrairi T, Abdellaoui R, Nasser B, Hammami S, Hammami M, Samadi M, Vejux A, Lizard G. In Vitro Evaluation of the Effects of 7-Ketocholesterol and 7β-Hydroxycholesterol on the Peroxisomal Status: Prevention of Peroxisomal Damages and Concept of Pexotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:437-452. [PMID: 38036892 DOI: 10.1007/978-3-031-43883-7_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
7-Ketocholesterol and 7β-hydroxycholesterol are most often derived from the autoxidation of cholesterol. Their quantities are often increased in the body fluids and/or diseased organs of patients with age-related diseases such as cardiovascular diseases, Alzheimer's disease, age-related macular degeneration, and sarcopenia which are frequently associated with a rupture of RedOx homeostasis leading to a high oxidative stress contributing to cell and tissue damages. On murine cells from the central nervous system (158N oligodendrocytes, microglial BV-2 cells, and neuronal N2a cells) as well as on C2C12 murine myoblasts, these two oxysterols can induce a mode of cell death which is associated with qualitative, quantitative, and functional modifications of the peroxisome. These changes can be revealed by fluorescence microscopy (apotome, confocal microscopy), transmission electron microscopy, flow cytometry, quantitative reverse transcription polymerase chain reaction (RT-qPCR), and gas chromatography-coupled with mass spectrometry (GC-MS). Noteworthy, several natural molecules, including ω3 fatty acids, polyphenols, and α-tocopherol, as well as several Mediterranean oils [argan and olive oils, Milk-thistle (Sylibum marianum) and Pistacia lenticus seed oils], have cytoprotective properties and attenuate 7-ketocholesterol- and 7β-hydroxycholesterol-induced peroxisomal modifications. These observations led to the concept of pexotherapy.
Collapse
Affiliation(s)
- Imen Ghzaiel
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Samah Maaloul
- Laboratory of Rangeland Ecosystems and Valorization of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| | - Mohamed Ksila
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| | - Amira Namsi
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Aline Yammine
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Meriam Debbabi
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Asma Badreddine
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Laboratory of Biochemistry, Neuroscience, Natural Resources and Environment, Faculty of Science and Technology, University Hassan I, Settat, Morocco
| | - Wiem Meddeb
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Vivien Pires
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Thomas Nury
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Franck Ménétrier
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Laure Avoscan
- Agroécologie, AgroSup Dijon, CNRS, INRAE, University Bourgogne Franche-Comté, Plateforme DimaCell, Dijon, France
| | - Amira Zarrouk
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
- Faculty of Medicine, University of Sousse, Laboratory of Biochemistry, Sousse, Tunisia
| | - Mauhamad Baarine
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Olfa Masmoudi-Kouki
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| | - Taoufik Ghrairi
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| | - Raoudha Abdellaoui
- Laboratory of Rangeland Ecosystems and Valorization of Spontaneous Plants and Associated Microorganisms (LR16IRA03), Arid Regions Institute, University of Gabes, Medenine, Tunisia
| | - Boubker Nasser
- Laboratory of Biochemistry, Neuroscience, Natural Resources and Environment, Faculty of Science and Technology, University Hassan I, Settat, Morocco
| | - Sonia Hammami
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Mohamed Hammami
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, University Lorraine, Metz Technopôle, Metz, France
| | - Anne Vejux
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France.
| |
Collapse
|
22
|
Luquain-Costaz C, Delton I. Oxysterols in Vascular Cells and Role in Atherosclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:213-229. [PMID: 38036882 DOI: 10.1007/978-3-031-43883-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Atherosclerosis is a major cardiovascular complication of diseases associated with elevated oxidative stress such as type 2 diabetes and metabolic syndrome. In these situations, low-density lipoproteins (LDL) undergo oxidation. Oxidized LDL displays proatherogenic activities through multiple and complex mechanisms which lead to dysfunctions of vascular cells (endothelial cells, smooth muscle cells, and macrophages). Oxidized LDLs are enriched in oxidized products of cholesterol called oxysterols formed either by autoxidation, enzymatically, or by both mechanisms. Several oxysterols have been shown to accumulate in atheroma plaques and to play a key role in atherogenesis. Depending on the type of oxysterols, various biological effects are exerted on vascular cells to regulate the formation of macrophage foam cells, endothelial integrity, adhesion and transmigration of monocytes, plaque progression, and instability. Most of these effects are linked to the ability of oxysterols to induce cellular oxidative stress and cytotoxicity mainly through apoptosis and proinflammatory mediators. Like for excess cholesterol, high-density lipoproteins (HDL) can exert antiatherogenic activity by stimulating the efflux of oxysterols that have accumulated in foamy macrophages.
Collapse
Affiliation(s)
- Celine Luquain-Costaz
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
- Department of Biosciences, INSA Lyon, Villeurbanne, France
| | - Isabelle Delton
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France.
- Department of Biosciences, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
23
|
Deng C, Li M, Liu Y, Yan C, He Z, Chen ZY, Zhu H. Cholesterol Oxidation Products: Potential Adverse Effect and Prevention of Their Production in Foods. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18645-18659. [PMID: 38011512 DOI: 10.1021/acs.jafc.3c05158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cholesterol oxidation products (COPs) are a group of substances formed during food processing. COPs in diet is a health concern because they may affect human health in association with the risk of various diseases including atherosclerosis, Alzheimer's disease, age-related macular degeneration, diabetes, and chronic gastrointestinal inflammatory colitis. Production of COPs in foods can be affected by many factors such as temperature, pH, light, oxygen, water, carbohydrates, fatty acids, proteins, and metal cations. The key issue is preventing its generation in foods. Some COPs can also be produced in vivo by both nonenzymatic and enzymatic-catalyzed oxidation reactions. Currently, a number of natural antioxidants such as catechins, flavonoids, and other polyphenols have been proven to inhibit the generation of COPs. In addition, measures taken during food processing can also minimize the production of COPs, such as the Maillard reaction and marinating food with plant polyphenol-rich seasonings. In conclusion, a comprehensive approach encompassing the suppression on COPs generation and implementation of processing measures is imperative to safeguard human health against the production of COPs in the food chain.
Collapse
Affiliation(s)
- Chuanling Deng
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Mingxuan Li
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
- School of Life Sciences, South China Agricultural University, Guangzhou 510000, Guangdong China
| | - Yang Liu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Zouyan He
- School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT 999077, Hong Kong China
| | - Hanyue Zhu
- School of Food Science and Engineering/Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528000, Guangdong China
| |
Collapse
|
24
|
Tancheva L, Kalfin R, Minchev B, Uzunova D, Tasheva K, Tsvetanova E, Georgieva A, Alexandrova A, Stefanova M, Solak A, Lazarova M, Hodzhev Y, Grigorova V, Yarkov D, Petkova-Kirova P. Memory Recovery Effect of a New Bioactive Innovative Combination in Rats with Experimental Dementia. Antioxidants (Basel) 2023; 12:2050. [PMID: 38136170 PMCID: PMC10740861 DOI: 10.3390/antiox12122050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease manifests as a complex pathological condition, with neuroinflammation, oxidative stress and cholinergic dysfunction being a few of the many pathological changes. Due to the complexity of the disease, current therapeutic strategies aim at a multitargeted approach, often relying on a combination of substances with versatile and complementary effects. In the present study, a unique combination of α-lipoic acid, citicoline, extracts of leaves from olive tree and green tea, vitamin D3, selenium and an immune-supporting complex was tested in scopolamine-induced dementia in rats. Using behavioral and biochemical methods, we assessed the effects of the combination on learning and memory, and elucidated the mechanisms of these effects. Our results showed that, compared to its components, the experimental combination was most efficient in improving short- and long-term memory as assessed by the step-through method as well as spatial memory as assessed by T-maze and Barnes maze underlined by decreases in AChE activity (p < 0.05) and LPO (p < 0.001), increases in SOD activity in the cortex (p < 0.05) and increases in catalase (p < 0.05) and GPx (p < 0.01) activities and BDNF (p < 0.001) and pCREB (p < 0.05) levels in the hippocampus. No significant histopathological changes or blood parameter changes were detected, making the experimental combination an effective and safe candidate in a multitargeted treatment of AD.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Department of Healthcare, South-West University “Neofit Rilski”, Ivan Mihailov Str. 66, 2700 Blagoevgrad, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 21, 1113 Sofia, Bulgaria;
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- National Sports Academy, Department of Physiology and Biochemistry, Acad. S. Mladenov Str. 21, 1700 Sofia, Bulgaria
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Ayten Solak
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Institute of Cryobiology and Food Technologies, Cherni Vrah Blvd 53, 1407 Sofia, Bulgaria
| | - Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Yordan Hodzhev
- National Center of Infectious and Parasitic Diseases, Yanko Sakazov Blvd 26, 1504 Sofia, Bulgaria;
| | - Valya Grigorova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Dobri Yarkov
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| |
Collapse
|
25
|
Spalenkova A, Ehrlichova M, Wei S, Peter Guengerich F, Soucek P. Effects of 7-ketocholesterol on tamoxifen efficacy in breast carcinoma cell line models in vitro. J Steroid Biochem Mol Biol 2023; 232:106354. [PMID: 37343688 PMCID: PMC10529436 DOI: 10.1016/j.jsbmb.2023.106354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/06/2023] [Accepted: 06/18/2023] [Indexed: 06/23/2023]
Abstract
Oxysterols play significant roles in many physiological and pathological processes including cancer. They modulate some of the cancer hallmarks pathways, influence the efficacy of anti-cancer drugs, and associate with patient survival. In this study, we aimed to analyze the role of 7-ketocholesterol (7-KC) in breast carcinoma cells and its potential modulation of the tamoxifen effect. 7-KC effects were studied in two estrogen receptor (ER)-positive (MCF-7 and T47D) and one ER-negative (BT-20) breast cancer cell lines. First, we tested the viability of cells in the presence of 7-KC. Next, we co-incubated cells with tamoxifen and sublethal concentrations of 7-KC. We also tested changes in caspase 3/7 activity, deregulation of the cell cycle, and changes in expression of selected genes/proteins in the presence of tamoxifen, 7-KC, or their combination. Finally, we analyzed the effect of 7-KC on cellular migration and invasion. We found that the presence of 7-KC slightly decreases the efficacy of tamoxifen in MCF-7 cells, while an increased effect of tamoxifen and higher caspase 3/7 activity was observed in the BT-20 cell line. In the T47D cell line, we did not find any modulation of tamoxifen efficacy by the presence of 7-KC. Expression analysis showed the deregulation in CYP1A1 and CYP1B1 with the opposite trend in MCF-7 and BT-20 cells. Moreover, 7-KC increased cellular migration and invasion potential regardless of the ER status. This study shows that 7-KC can modulate tamoxifen efficacy as well as cellular migration and invasion, making 7-KC a promising candidate for future studies.
Collapse
Affiliation(s)
- Alzbeta Spalenkova
- Department of Toxicogenomics, National Institute of Public Health, Prague 100 42, Czech Republic; Third Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| | - Marie Ehrlichova
- Department of Toxicogenomics, National Institute of Public Health, Prague 100 42, Czech Republic
| | - Shouzou Wei
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Pavel Soucek
- Department of Toxicogenomics, National Institute of Public Health, Prague 100 42, Czech Republic.
| |
Collapse
|
26
|
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
|
27
|
Ghzaiel I, Zarrouk A, Pires V, de Barros JPP, Hammami S, Ksila M, Hammami M, Ghrairi T, Jouanny P, Vejux A, Lizard G. 7β-Hydroxycholesterol and 7-ketocholesterol: New oxidative stress biomarkers of sarcopenia inducing cytotoxic effects on myoblasts and myotubes. J Steroid Biochem Mol Biol 2023; 232:106345. [PMID: 37286110 DOI: 10.1016/j.jsbmb.2023.106345] [Citation(s) in RCA: 11] [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: 01/10/2023] [Revised: 04/21/2023] [Accepted: 06/04/2023] [Indexed: 06/09/2023]
Abstract
Aging is a complex biological process which can be associated with skeletal muscle degradation leading to sarcopenia. The aim of this study consisted i) to determine the oxidative and inflammatory status of sarcopenic patients and ii) to clarify the impact of oxidative stress on myoblasts and myotubes. To this end, various biomarkers of inflammation (C-reactive protein (CRP), TNF-α, IL-6, IL-8, leukotriene B4 (LTB4)) and oxidative stress (malondialdehyde, conjugated dienes, carbonylated proteins and antioxidant enzymes: catalase, superoxide dismutase, glutathione peroxidase) as well as oxidized derivatives of cholesterol formed by cholesterol autoxidation (7-ketocholesterol, 7β-hydroxycholesterol), were analyzed. Apelin, a myokine which contributes to muscle strength, was also quantified. To this end, a case-control study was conducted to evaluate the RedOx and inflammatory status in 45 elderly subjects (23 non-sarcopenic; 22 sarcopenic) from 65 years old and higher. SARCopenia-Formular (SARC-F) and Timed Up and Go (TUG) tests were used to distinguish between sarcopenic and non-sarcopenic subjects. By using red blood cells, plasma and/or serum, we observed in sarcopenic patients an increased activity of major antioxidant enzymes (superoxide dismutase, glutathione peroxidase, catalase) associated with lipid peroxidation and protein carbonylation (increased level of malondialdehyde, conjugated dienes and carbonylated proteins). Higher levels of 7-ketocholesterol and 7β-hydroxycholesterol were also observed in the plasma of sarcopenic patients. Significant differences were only observed with 7β-hydroxycholesterol. In sarcopenic patients comparatively to non-sarcopenic subjects, significant increase of CRP, LTB4 and apelin were observed whereas similar levels of TNF-α, IL-6 and IL-8 were found. The increased plasma level of 7-ketocholesterol and 7β-hydroxycholesterol in sarcopenic patients led us to study the cytotoxic effect of these oxysterols on undifferentiated (myoblasts) and differentiated (myotubes) murine C2C12 cells. With the fluorescein diacetate and sulforhodamine 101 assays, an induction of cell death was observed both on undifferentiated and differentiated cells: the cytotoxic effects were less pronounced with 7-ketocholesterol. In addition, IL-6 secretion was never detected whatever the culture conditions, TNF-α secretion was significantly increased on undifferentiated and differentiated C2C12 cells treated with 7-ketocholesterol- and 7β-hydroxycholesterol, and IL-8 secretion was increased on differentiated cells. 7-ketocholesterol- and 7β-hydroxycholesterol-induced cell death was strongly attenuated by α-tocopherol and Pistacia lentiscus L. seed oil both on myoblasts and/or myotubes. TNF-α and/or IL-8 secretions were reduced by α-tocopherol and Pistacia lentiscus L. seed oil. Our data support the hypothesis that the enhancement of oxidative stress observed in sarcopenic patients could contribute, especially via 7β-hydroxycholesterol, to skeletal muscle atrophy and inflammation via cytotoxic effects on myoblasts and myotubes. These data bring new elements to understand the pathophysiology of sarcopenia and open new perspectives for the treatment of this frequent age-related disease.
Collapse
Affiliation(s)
- Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne, 21000 Dijon, France; Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - Amira Zarrouk
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia.
| | - Vivien Pires
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne, 21000 Dijon, France
| | | | - Sonia Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - Mohamed Ksila
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne, 21000 Dijon, France; Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMolecules, LR18ES03, Department of Biology, Faculty of Sciences, University Tunis-El Manar, Tunis 2092, Tunisia
| | - Mohamed Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
| | - Taoufik Ghrairi
- Université de Bourgogne, Lipidomic Platform, 21000 Dijon, France
| | - Pierre Jouanny
- Geriatric Internal Medicine Department (Champmaillot), University Hospital Center, Université de Bourgogne, 21000 Dijon, France
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne, 21000 Dijon, France
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, Université de Bourgogne, 21000 Dijon, France.
| |
Collapse
|
28
|
Lizard G, Poirot M, Iuliano L. European Network for Oxysterol Research (ENOR): 10 th ENOR Symposium - Web Meeting. Steroids 2023; 195:109242. [PMID: 37088450 DOI: 10.1016/j.steroids.2023.109242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Affiliation(s)
- Gérard Lizard
- Team Bio-PeroxIL, "Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism" (EA7270), University Bourgogne, Inserm, 21000, Dijon, France.
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Team "Cholesterol Metabolism and Therapeutic Innovations"; Equipe labellisée par la Ligue Nationale Contre le Cancer; The French Network for Nutrition and Cancer Research (NACRe Network); INSERM UMR 1037-CNRS U 5071-Université de Toulouse, 31037, Toulouse, France
| | - Luigi Iuliano
- INSERM UMR 1037-CNRS U 5071-Université de Toulouse, 31037, Toulouse, France; UOC of Internal Medicine, Sapienza University of Rome, ICOT University Hospital, Latina ; & Vascular Biology & Mass Spectrometry Laboratory, Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
29
|
Ksila M, Ghzaiel I, Pires V, Ghrairi T, Masmoudi-Kouki O, Latruffe N, Vervandier-Fasseur D, Vejux A, Lizard G. Characterization of Cell Death Induced by Imine Analogs of Trans-Resveratrol: Induction of Mitochondrial Dysfunction and Overproduction of Reactive Oxygen Species Leading to, or Not, Apoptosis without the Increase in the S-Phase of the Cell Cycle. Molecules 2023; 28:molecules28073178. [PMID: 37049947 PMCID: PMC10096382 DOI: 10.3390/molecules28073178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Trans-resveratrol (RSV) is a non-flavonoid polyphenol (stilbene) with numerous biological activities, such as anti-tumor activities. However, RSV is rapidly metabolized, which limits its therapeutic use. The availability of RSV analogues with similar activities for use in vivo is therefore a major challenge. For this purpose, several isomeric analogues of RSV, aza-stilbenes (AZA-ST 1a–g), were synthesized, and their toxicities were characterized and compared to those of RSV on murine N2a neuronal cells using especially flow cytometric methods. All AZA-ST 1a–g have an inhibitory concentration 50 (IC50) between 11.3 and 25 µM when determined by the crystal violet assay, while that of RSV is 14.5 µM. This led to the characterization of AZA-ST 1a–g—induced cell death, compared to RSV, using three concentrations encompassing the IC50s (6.25, 12.5 and 25 µM). For AZA-ST 1a–g and RSV, an increase in plasma membrane permeability to propidium iodide was observed, and the proportion of cells with depolarized mitochondria measured with DiOC6(3) was increased. An overproduction of reactive oxygen species (ROS) was also observed on whole cells and at the mitochondrial level using dihydroethidium and MitoSox Red, respectively. However, only RSV induced a mode of cell death by apoptosis associated with a marked increase in the proportion of cells with condensed and/or fragmented nuclei (12.5 µM: 22 ± 9%; 25 µM: 80 ± 10%) identified after staining with Hoechst 33342 and which are characteristic of apoptotic cells. With AZA-ST, a slight but significant increase in the percentage of apoptotic cells was only detected with AZA-ST 1b (25 µM: 17 ± 1%) and AZA-ST 1d (25 µM: 26 ± 4%). Furthermore, only RSV induced significant cell cycle modifications associated with an increase in the percentage of cells in the S phase. Thus, AZA-ST 1a–g—induced cell death is characterized by an alteration of the plasma membrane, an induction of mitochondrial depolarization (loss of ΔΨm), and an overproduction of ROS, which may or may not result in a weak induction of apoptosis without modification of the distribution of the cells in the different phases of the cell cycle.
Collapse
Affiliation(s)
- Mohamed Ksila
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia
| | - Imen Ghzaiel
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
| | - Vivien Pires
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
| | - Taoufik Ghrairi
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia
| | - Olfa Masmoudi-Kouki
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia
| | - Norbert Latruffe
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
| | | | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University of Bourgogne, 21000 Dijon, France
| |
Collapse
|
30
|
Babazadeh A, Vahed FM, Liu Q, Siddiqui SA, Kharazmi MS, Jafari SM. Natural Bioactive Molecules as Neuromedicines for the Treatment/Prevention of Neurodegenerative Diseases. ACS OMEGA 2023; 8:3667-3683. [PMID: 36743024 PMCID: PMC9893457 DOI: 10.1021/acsomega.2c06098] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
The brain is vulnerable to different types of stresses, particularly oxidative stress as a result of oxygen requirements/utilization in the body. Large amounts of unsaturated fatty acids present in the brain increase this vulnerability. Neurodegenerative diseases (NDDs) are brain disorders that are characterized by the gradual loss of specific neurons and are attributed to broad evidence of cell-level oxidative stress. The accurate characterization of neurological disorders relies on several parameters along with genetics and environmental risk factors, making therapies less efficient to fight NDDs. On the way to tackle oxidative damage and discover efficient and safe therapies, bioactives are at the edge of NDD science. Naturally occurring bioactive compounds such as polyphenols, carotenoids, essential fatty acids, phytosterols, essential oils, etc. are particularly of interest owing to their potent antioxidant and anti-inflammatory activities, and they offer lots of brain-health-promoting features. This Review focuses on probing the neuroefficacy and bioefficacy of bioactives and their role in supporting relatively low antioxidative and low regenerative capacities of the brain, neurogenesis, neuroprotection, and ameliorating/treating NDDs.
Collapse
Affiliation(s)
- Afshin Babazadeh
- Center
for Motor Neuron Disease Research, Macquarie Medical School, Faculty
of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Fereshteh Mohammadi Vahed
- Center
for Motor Neuron Disease Research, Macquarie Medical School, Faculty
of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Qi Liu
- Institute
of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu
Key Laboratory of Integrated Traditional Chinese and Western Medicine
for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Shahida Anusha Siddiqui
- Technical
University of Munich Campus Straubing for Biotechnology and Sustainability, Essigberg 3, 94315 Straubing, Germany
- German
Institute of Food Technologies (DIL e.V.), Prof.-von-Klitzing-Straße 7, 49610 D Quakenbrück, Germany
| | | | - Seid Mahdi Jafari
- Department
of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan 4913815739, Iran
- Nutrition
and Bromatology Group, Department of Analytical Chemistry and Food
Science, Faculty of Science, Universidade
de Vigo, E-32004 Ourense, Spain
- College
of Food Science and Technology, Hebei Agricultural
University, Baoding 071001, China
| |
Collapse
|
31
|
Essadek S, Gondcaille C, Savary S, Samadi M, Vamecq J, Lizard G, Kebbaj RE, Latruffe N, Benani A, Nasser B, Cherkaoui-Malki M, Andreoletti P. Two Argan Oil Phytosterols, Schottenol and Spinasterol, Attenuate Oxidative Stress and Restore LPS-Dysregulated Peroxisomal Functions in Acox1-/- and Wild-Type BV-2 Microglial Cells. Antioxidants (Basel) 2023; 12:168. [PMID: 36671029 PMCID: PMC9854540 DOI: 10.3390/antiox12010168] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Oxidative stress and inflammation are the key players in neuroinflammation, in which microglia dysfunction plays a central role. Previous studies suggest that argan oil attenuates oxidative stress, inflammation, and peroxisome dysfunction in mouse brains. In this study, we explored the effects of two major argan oil (AO) phytosterols, Schottenol (Schot) and Spinasterol (Spina), on oxidative stress, inflammation, and peroxisomal dysfunction in two murine microglial BV-2 cell lines, wild-ype (Wt) and Acyl-CoA oxidase 1 (Acox1)-deficient cells challenged with LPS treatment. Herein, we used an MTT test to reveal no cytotoxicity for both phytosterols with concentrations up to 5 µM. In the LPS-activated microglial cells, cotreatment with each of these phytosterols caused a significant decrease in intracellular ROS production and the NO level released in the culture medium. Additionally, Schot and Spina were able to attenuate the LPS-dependent strong induction of Il-1β and Tnf-α mRNA levels, as well as the iNos gene and protein expression in both Wt and Acox1-/- microglial cells. On the other hand, LPS treatment impacted both the peroxisomal antioxidant capacity and the fatty acid oxidation pathway. However, both Schot and Spina treatments enhanced ACOX1 activity in the Wt BV-2 cells and normalized the catalase activity in both Wt and Acox1-/- microglial cells. These data suggest that Schot and Spina can protect cells from oxidative stress and inflammation and their harmful consequences for peroxisomal functions and the homeostasis of microglial cells. Collectively, our work provides a compelling argument for the protective mechanisms of two major argan oil phytosterols against LPS-induced brain neuroinflammation.
Collapse
Affiliation(s)
- Soukaina Essadek
- Laboratory of Biochimistry, Neuroscience, Natural Resources and Environment, Faculty of Science and Technology, University Hassan I, Settat 26000, Morocco
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Catherine Gondcaille
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Stéphane Savary
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Mohammad Samadi
- Laboratory of Chemistry and Physics Multi-Scale Approach to Complex Environments, Department of Chemistry, University Lorraine, 57070 Metz, France
| | - Joseph Vamecq
- Inserm and HMNO, CBP, CHRU Lille, and RADEME EA 7364, Faculté de Médecine, Université de Lille 2, 59045 Lille, France
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan 1st University, Settat 26000, Morocco
| | - Norbert Latruffe
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Alexandre Benani
- CSGA—Centre des Sciences du Goût et de l’Alimentation, CNRS—Centre National de la Recherche Scientifique, INRAE—Institut National de Recherche pour L’agriculture, L’alimentation et L’environnement, Institut Agro Dijon, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Boubker Nasser
- Laboratory of Biochimistry, Neuroscience, Natural Resources and Environment, Faculty of Science and Technology, University Hassan I, Settat 26000, Morocco
| | - Mustapha Cherkaoui-Malki
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| | - Pierre Andreoletti
- Bio-PeroxIL Laboratory, EA7270, University Bourgogne Franche-Comté/Inserm, 6 Boulevard Gabriel, 21000 Dijon, France
| |
Collapse
|
32
|
Chen Y, Ye S, Wang Q, Shen M, Lu F, Qu J, Zhu D. In situ assessment of lens elasticity with noncontact optical coherence elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:6671-6681. [PMID: 36589560 PMCID: PMC9774883 DOI: 10.1364/boe.475306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 06/17/2023]
Abstract
Lens biomechanics has great potential for application in clinical diagnostics and treatment monitoring of presbyopia and cataracts. However, current approaches to lens elastography do not meet the desired safety or sensitivity for clinical application. In this regard, we propose a noncontact optical coherence elastography (OCE) method to facilitate quantitative in situ imaging of lens elasticity. Elastic waves induced by air-pulse stimulation on the limbus propagate to the lens and are then imaged using custom-built swept-source optical coherence tomography to obtain the elastic wave velocity and Young's modulus. The proposed OCE method was first validated by comparing the results of in situ and in vitro measurements of porcine lenses. The results demonstrate that the Young's modulus measured in situ was highly consistent with that measured in vitro and had an intraclass correlation coefficient of 0.988. We further investigated the elastic changes induced by cold storage and microwave heating. During 36-hour cold storage, the mean Young's modulus gradually increased (from 5.62 ± 1.24 kPa to 11.40 ± 2.68 kPa, P < 0.0001, n = 9) along with the formation of nuclear opacities. 15-second microwave heating caused a greater increase in the mean Young's modulus (from 6.86 ± 1.21 kPa to 25.96 ± 8.64 kPa, P < 0.0025, n = 6) without apparent cataract formation. Accordingly, this study reports the first air-pulse OCE measurements of in situ lenses, which quantified the loss of lens elasticity during simulated cataract development with good repeatability and sensitivity, thus enhancing the potential for adoption of lens biomechanics in the clinic.
Collapse
Affiliation(s)
- Yulei Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Shuling Ye
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Qingying Wang
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Meixiao Shen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Dexi Zhu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Optometry, Ophthalmology, and Vision Science, Wenzhou, China
| |
Collapse
|
33
|
Ghzaiel I, Nury T, Zarrouk A, Vejux A, Lizard G. Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882. Cells 2022; 11:cells11223612. [PMID: 36429041 PMCID: PMC9688161 DOI: 10.3390/cells11223612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Due to the increase in life span and life expectancy, which can, however, be more or less pronounced depending on the economic, social and cultural context [...].
Collapse
Affiliation(s)
- Imen Ghzaiel
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
- Faculty of Sciences of Tunis, University Tunis-El Manar, Tunis 2092, Tunisia
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
- Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
- Correspondence: ; Tel.: +33-3-80-39-62-56
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Rezig L, Martine L, Nury T, Msaada K, Mahfoudhi N, Ghzaiel I, Prost-Camus E, Durand P, Midaoui AE, Acar N, Latruffe N, Vejux A, Lizard G. Profiles of Fatty Acids, Polyphenols, Sterols, and Tocopherols and Scavenging Property of Mediterranean Oils: New Sources of Dietary Nutrients for the Prevention of Age-related Diseases. J Oleo Sci 2022; 71:1117-1133. [PMID: 35922928 DOI: 10.5650/jos.ess22110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The present study provides the fatty acid, tocopherol, phytosterol, and polyphenol profiles of some Mediterranean oils extracted from pumpkin, melon, and black cumin seed oils and those of dietary argan seed oil. Gas chromatography analysis revealed that oleic and linoleic acids were the most abundant fatty acids. Argan and melon seed oils exhibited the highest levels of oleic acid (47.32±0.02%) and linoleic acid (58.35±0.26%), respectively. In terms of tocopherols, melon seed oil showed the highest amount (652.1±3.26 mg/kg) with a predominance of γ-tocopherol (633.1±18.81 mg/kg). The phytosterol content varied between 2237.00±37.55 µg/g for argan oil to 6995.55±224.01 µg/g for melon seed oil. High Performance Liquid Chromatography analysis also revealed the presence of several polyphenols: vanillin (0.59 mg equivalents Quercetin/100 g) for melon seed oil, and p-hydroxycinnamic acid (0.04 mg equivalents Quercetin/100 g), coumarine (0.05 mg equivalents Quercetin/100 g), and thymoquinone (1.2 mg equivalents Quercetin/100 g) for black cumin seed oil. The "Kit Radicaux Libres" (KRL) assay used to evaluate the scavenging properties of the oils showed that black cumin seed oil was the most efficient. On the light of the richness of all Mediterranean oil samples in bioactive compounds, the seed oils studied can be considered as important sources of nutrients endowed with cytoprotective properties which benefits in preventing age-related diseases which are characterized by an enhanced oxidative stress.
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'.,University of Carthage, High Institute of Food Industries
| | - Lucy Martine
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, University of Bourgogne Franche-Comté, Eye and Nutrition Research Group
| | - Thomas Nury
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270) / University of Bourgogne / Inserm
| | - Kamel Msaada
- Laboratory of Aromatic and Medicinal Plants, Biotechnology Center in Borj-Cedria Technopole
| | - Nesrine Mahfoudhi
- Laboratory of Aromatic and Medicinal Plants, Biotechnology Center in Borj-Cedria Technopole.,University of Kairouan, Faculty of Science and Technology of Sidi Bouzid, Department of Biotechnology
| | - Imen Ghzaiel
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270) / University of Bourgogne / Inserm.,University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health'.,University Tunis-El Manar, Faculty of Sciences of Tunis
| | | | | | - Adil El Midaoui
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montréal.,Department of Biology, FST Errachidia, Moulay Ismail University
| | - Niyazi Acar
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, University of Bourgogne Franche-Comté, Eye and Nutrition Research Group
| | - Norbert Latruffe
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270) / University of Bourgogne / Inserm
| | - Anne Vejux
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270) / University of Bourgogne / Inserm
| | - Gérard Lizard
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270) / University of Bourgogne / Inserm
| |
Collapse
|
37
|
Kim JS, Lim H, Seo JY, Kang KR, Yu SK, Kim CS, Kim DK, Kim HJ, Seo YS, Lee GJ, You JS, Oh JS. GPR183 Regulates 7α,25-Dihydroxycholesterol-Induced Oxiapoptophagy in L929 Mouse Fibroblast Cell. Molecules 2022; 27:4798. [PMID: 35956750 PMCID: PMC9369580 DOI: 10.3390/molecules27154798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
7α,25-dihydroxycholesterol (7α,25-DHC) is an oxysterol synthesized from 25-hydroxycholesterol by cytochrome P450 family 7 subfamily B member 1 (CYP7B1) and is a monooxygenase (oxysterol-7α-hydroxylase) expressed under inflammatory conditions in various cell types. In this study, we verified that 7α,25-DHC-induced oxiapoptophagy is mediated by apoptosis, oxidative stress, and autophagy in L929 mouse fibroblasts. MTT assays and live/dead cell staining revealed that cytotoxicity was increased by 7α,25-DHC in L929 cells. Consequentially, cells with condensed chromatin and altered morphology were enhanced in L929 cells incubated with 7α,25-DHC for 48 h. Furthermore, apoptotic population was increased by 7α,25-DHC exposure through the cascade activation of caspase-9, caspase-3, and poly (ADP-ribose) polymerase in the intrinsic pathway of apoptosis in these cells. 7α,25-DHC upregulated reactive oxygen species (ROS) in L929 cells. Expression of autophagy biomarkers, including beclin-1 and LC3, was significantly increased by 7α,25-DHC treatment in L929 cells. 7α,25-DHC inhibits the phosphorylation of Akt associated with autophagy and increases p53 expression in L929 cells. In addition, inhibition of G-protein-coupled receptor 183 (GPR183), a receptor of 7α,25-DHC, using GPR183 specific antagonist NIBR189 suppressed 7α,25-DHC-induced apoptosis, ROS production, and autophagy in L929 cells. Collectively, GPR183 regulates 7α,25-DHC-induced oxiapoptophagy in L929 cells.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - HyangI Lim
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Jeong-Yeon Seo
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Kyeong-Rok Kang
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Sun-Kyoung Yu
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Chun Sung Kim
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Do Kyung Kim
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Heung-Joong Kim
- Institute of Dental Science, School of Dentistry, Chosun University, Gwangju 61452, Korea; (J.-S.K.); (H.L.); (J.-Y.S.); (K.-R.K.); (S.-K.Y.); (C.S.K.); (D.K.K.); (H.-J.K.)
| | - Yo-Seob Seo
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Chosun University, Gwangju 61452, Korea;
| | - Gyeong-Je Lee
- Department of Prosthodontics, School of Dentistry, Chosun University, Gwangju 61452, Korea;
| | - Jae-Seek You
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju 61452, Korea;
| | - Ji-Su Oh
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju 61452, Korea;
| |
Collapse
|
38
|
Zhang S, Zhu N, Gu J, Li HF, Qiu Y, Liao DF, Qin L. Crosstalk between Lipid Rafts and Aging: New Frontiers for Delaying Aging. Aging Dis 2022; 13:1042-1055. [PMID: 35855333 PMCID: PMC9286918 DOI: 10.14336/ad.2022.0116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/16/2022] [Indexed: 12/15/2022] Open
Abstract
With the rapid aging in the global population, delay of aging has become a hot research topic. Lipid rafts (LRs) are microdomains in the plasma membrane that contain sphingolipids and cholesterol. Emerging evidence indicates an interesting interplay between LRs and aging. LRs and their components are altered with aging. Further, the aging process is strongly influenced by LRs. In recent years, LRs and their component signaling molecules have been recognized to affect aging by interfering with its hallmarks. Therefore, targeting LRs is a promising strategy to delay aging.
Collapse
Affiliation(s)
- Shuo Zhang
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Neng Zhu
- 2Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jia Gu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yun Qiu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Li Qin
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.,3Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
39
|
Ghzaiel I, Zarrouk A, Essadek S, Martine L, Hammouda S, Yammine A, Ksila M, Nury T, Meddeb W, Tahri Joutey M, Mihoubi W, Caccia C, Leoni V, Samadi M, Acar N, Andreoletti P, Hammami S, Ghrairi T, Vejux A, Hammami M, Lizard G. Protective effects of milk thistle (Sylibum marianum) seed oil and α-tocopherol against 7β-hydroxycholesterol-induced peroxisomal alterations in murine C2C12 myoblasts: Nutritional insights associated with the concept of pexotherapy. Steroids 2022; 183:109032. [PMID: 35381271 DOI: 10.1016/j.steroids.2022.109032] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022]
Abstract
Peroxisomes play an important role in regulating cell metabolism and RedOx homeostasis. Peroxisomal dysfunctions favor oxidative stress and cell death. The ability of 7β-hydroxycholesterol (7β-OHC; 50 μM, 24 h), known to be increased in patients with age-related diseases such as sarcopenia, to trigger oxidative stress, mitochondrial and peroxisomal dysfunction was studied in murine C2C12 myoblasts. The capacity of milk thistle seed oil (MTSO, 100 μg/mL) as well as α-tocopherol (400 µM; reference cytoprotective agent) to counteract the toxic effects of 7β-OHC, mainly at the peroxisomal level were evaluated. The impacts of 7β-OHC, in the presence or absence of MTSO or α-tocopherol, were studied with complementary methods: measurement of cell density and viability, quantification of reactive oxygen species (ROS) production and transmembrane mitochondrial potential (ΔΨm), evaluation of peroxisomal mass as well as topographic, morphologic and functional peroxisomal changes. Our results indicate that 7β-OHC induces a loss of cell viability and a decrease of cell adhesion associated with ROS overproduction, alterations of mitochondrial ultrastructure, a drop of ΔΨm, and several peroxisomal modifications. In the presence of 7β-OHC, comparatively to untreated cells, important quantitative and qualitative peroxisomal modifications were also identified: a) a reduced number of peroxisomes with abnormal sizes and shapes, mainly localized in cytoplasmic vacuoles, were observed; b) the peroxisomal mass was decreased as indicated by lower protein and mRNA levels of the peroxisomal ABCD3 transporter; c) lower mRNA level of Pex5 involved in peroxisomal biogenesis as well as higher mRNA levels of Pex13 and Pex14, involved in peroxisomal biogenesis and/or pexophagy, was found; d) lower levels of ACOX1 and MFP2 enzymes, implicated in peroxisomal β-oxidation, were detected; e) higher levels of very-long-chain fatty acids, which are substrates of peroxisomal β-oxidation, were found. These different cytotoxic effects were strongly attenuated by MTSO, in the same range of order as with α-tocopherol. These findings underline the interest of MTSO and α-tocopherol in the prevention of peroxisomal damages (pexotherapy).
Collapse
Affiliation(s)
- Imen Ghzaiel
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia; Faculty of Sciences of Tunis, University Tunis-El Manar, 2092 Tunis, Tunisia
| | - Amira Zarrouk
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia; Faculty of Medicine, University of Sousse, 4000 Sousse, Tunisia.
| | - Soukaina Essadek
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences & Techniques, University Hassan I, BP 577, 26000 Settat, Morocco
| | - Lucy Martine
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21065 Dijon, France
| | - Souha Hammouda
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia
| | - Aline Yammine
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon
| | - Mohamed Ksila
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; Faculty of Sciences of Tunis, University Tunis-El Manar, 2092 Tunis, Tunisia
| | - Thomas Nury
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Wiem Meddeb
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Mounia Tahri Joutey
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences & Techniques, University Hassan I, BP 577, 26000 Settat, Morocco
| | - Wafa Mihoubi
- Laboratoire de Biotechnologie Moléculaire des Eucaryotes, Centre de Biotechnologie de Sfax, B.P 1177, Université de Sfax, 3018 Sfax, Tunisia
| | - Claudio Caccia
- Laboratory of Clinical Chemistry, Hospitals of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospitals of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, University Lorraine, Metz Technopôle, 57070 Metz, France
| | - Niyazi Acar
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21065 Dijon, France
| | - Pierre Andreoletti
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Sonia Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia
| | - Taoufik Ghrairi
- Faculty of Sciences of Tunis, University Tunis-El Manar, 2092 Tunis, Tunisia
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
| | - Mohamed Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism'EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France.
| |
Collapse
|
40
|
Messedi M, Guidara W, Grayaa S, Khrouf W, Snoussi M, Bahloul Z, Bonnefont-Rousselot D, Lamari F, Ayadi F. Selected plasma oxysterols as a potential multi-marker biosignature panel for Behçet's Disease. J Steroid Biochem Mol Biol 2022; 221:106122. [PMID: 35588947 DOI: 10.1016/j.jsbmb.2022.106122] [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: 02/11/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Clinical, genetic, and medical evidence has shown the inflammatory vasculitis aspect of Behçet's Disease (BD). Whereas oxysterols are vital factors in inflammation and oxidative stress, it is still unknown whether they are involved in the pathophysiology of BD. The current study aims to explore the profile of oxysterols in plasma of BD patients. Thirty patients diagnosed with BD and forty healthy controls matched for age and gender were included. Results showed that the cholestane-3β,5α,6β-triol, 27-hydroxycholesterol (27-OHC) and cholestanol levels were higher in BD than controls. In addition, plasma levels of 7-ketocholesterol (7-KC) and 25-hydroxycholesterol (25-OHC) were lower in BD patient. However, levels of 24S-hydroxycholesterol (24-OHC) did not significantly differ. For BD patients, the plasma 7-KC level was negatively correlated with the BD activity index (BDAI) while 27-OHC was positively correlated with high-sensitivity C-reactive protein (hs-CRP) in patients with active course of the disease. According to ROC analysis, a remarkable increase in the area under the curve (AUC) with a higher sensitivity (Se) and specificity (Sp) for 7-KC, 25-OHC and 27-OHC combined markers was observed. The present study indicated that the identification of the predictive value of these three-selected biomarkers related to oxidative stress and inflammation in patients should lead to a better identification of the etiological mechanism of BD.
Collapse
Affiliation(s)
- Meriam Messedi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia.
| | - Wassim Guidara
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Sahar Grayaa
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Walid Khrouf
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France
| | - Mouna Snoussi
- Internal medicine department, Hedi Chaker Hosptital, Sfax, Tunisia
| | - Zouhir Bahloul
- Internal medicine department, Hedi Chaker Hosptital, Sfax, Tunisia
| | - Dominique Bonnefont-Rousselot
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France; Université de Paris, CNRS, Inserm, UTCBS, Paris F-75006, France
| | - Foudil Lamari
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France
| | - Fatma Ayadi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| |
Collapse
|
41
|
Wnętrzak A, Chachaj-Brekiesz A, Kuś K, Lipiec E, Dynarowicz-Latka P. Oxysterols can act antiviral through modification of lipid membrane properties - The Langmuir monolayer study. J Steroid Biochem Mol Biol 2022; 220:106092. [PMID: 35272016 DOI: 10.1016/j.jsbmb.2022.106092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/20/2022]
Abstract
In this paper we tested how oxysterols influence on fusion process between viral lipid envelope and host cells membranes. For this purpose, the Zika virus was selected, while dendritic cell (DC) and neural cell (NC) membranes were chosen as target membranes. The investigated systems were modeled as multicomponent Langmuir monolayers and characterized using surface manometry and imaging in micro- (Brewster angle microscopy, BAM) and nanoscale (Atomic Force Microscopy, AFM) to monitor local heterogeneity. The fusion process was conducted by mixing viral and host cell membranes devoid and in the presence of oxysterols: 25-hydroxycholesterol (25-OH) and 7β-hydroxycholesterol (7β-OH) as representatives of chain- and ring-oxidized oxysterols, respectively. Our results show that oxysterols hinder the fusion with host cell membranes by modifying their biophysical properties. Moreover, oxysterols applied to an already infected membrane reverse the changes caused by the infection. It could therefore be concluded that oxysterols may display antiviral activity in two ways: they prevent the healthy membrane from viral infection by blocking the fusion process; and protect already infected membrane from pathological changes induced by the virus.
Collapse
Affiliation(s)
- Anita Wnętrzak
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland.
| | - Anna Chachaj-Brekiesz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Karolina Kuś
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Ewelina Lipiec
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland
| | | |
Collapse
|
42
|
Iciek M, Bilska-Wilkosz A, Kozdrowicki M, Górny M. Reactive Sulfur Compounds in the Fight against COVID-19. Antioxidants (Basel) 2022; 11:antiox11061053. [PMID: 35739949 PMCID: PMC9220020 DOI: 10.3390/antiox11061053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The SARS-CoV-2 coronavirus pandemic outbreak in 2019 resulted in the need to search for an effective and safe strategy for treating infected patients, relieving symptoms, and preventing severe disease. SARS-CoV-2 is an RNA virus that can cause acute respiratory failure and thrombosis, as well as impair circulatory system function. Permanent damage to the heart muscle or other cardiovascular disorders may occur during or after the infection. The severe course of the disease is associated with the release of large amounts of pro-inflammatory cytokines. Due to their documented anti-inflammatory, antioxidant, and antiviral effects, reactive sulfur compounds, including hydrogen sulfide (H2S), lipoic acid (LA), N-acetylcysteine (NAC), glutathione (GSH), and some other lesser-known sulfur compounds, have attracted the interest of scientists for the treatment and prevention of the adverse effects of diseases caused by SARS-CoV-2. This article reviews current knowledge about various endogenous or exogenous reactive sulfur compounds and discusses the possibility, or in some cases the results, of their use in the treatment or prophylaxis of COVID-19.
Collapse
|
43
|
Ali J, Aziz MA, Rashid MMO, Basher MA, Islam MS. Propagation of age‐related diseases due to the changes of lipid peroxide and antioxidant levels in elderly people: A narrative review. Health Sci Rep 2022; 5:e650. [PMID: 35620545 PMCID: PMC9125877 DOI: 10.1002/hsr2.650] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/21/2022] [Accepted: 05/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background and Aims Lipid peroxidation end products are the major culprit for inducing chronic diseases in elderly people. Along with the elevated level of lipid peroxide biomarkers, there is a significant disruption of antioxidants balance, which combinedly propagate the diseases of elderly people. The aim of the present review is to bridge the connection of changes in lipid peroxides biomarkers and antioxidants level with age‐associated diseases in elderly people. Methods This narrative review was performed following a comprehensive search for suitable articles in multiple online databases, including PubMed, Google Scholar, EMBASE, Web of Science, Cochrane Library, and ScienceDirect using selected search terms. The most appropriate literature was included based on the selection criteria. Results From the review, it is found that many age‐related diseases propagated with an increased level of the end products of lipid peroxide and reduced levels of antioxidants in elderly people. When the end products of lipid peroxidation increase in the body, it creates oxidative stress, which ultimately leads to many complicated diseases, including cancers, cardiovascular and neurogenic diseases, and many other chronic inflammatory diseases. The oxidative stress induced by peroxidation can be assessed by different lipid peroxide end products such as malondialdehyde, oxidized low‐density lipoprotein, isoprostanes, neuroprostanes, lipoperoxides, oxysterols (7‐ketocholesterol, 7β‐hydroxycholesterol), and many more. Conclusions This study definitively answers the correlation between the changes in lipid peroxides and antioxidants level and age‐related diseases. Our narrative article recommends future investigations for elucidating the mechanisms rigorously to establish a compact correlation.
Collapse
Affiliation(s)
- Julfikar Ali
- Department of Pharmacy, Faculty of Science Noakhali Science and Technology University Noakhali Bangladesh
| | - Md. Abdul Aziz
- Department of Pharmacy, Faculty of Science Noakhali Science and Technology University Noakhali Bangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of Pharmacy Noakhali Science and Technology University Noakhali Bangladesh
- Department of Pharmacy, Faculty of Pharmacy and Health Sciences State University of Bangladesh Dhaka Bangladesh
| | - Md. Mamun Or Rashid
- Department of Pharmacy, Faculty of Science Noakhali Science and Technology University Noakhali Bangladesh
| | - Mohammad Anwarul Basher
- Department of Pharmacy, Faculty of Science Noakhali Science and Technology University Noakhali Bangladesh
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Faculty of Science Noakhali Science and Technology University Noakhali Bangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of Pharmacy Noakhali Science and Technology University Noakhali Bangladesh
| |
Collapse
|
44
|
Yang J, Li X, Yang H, Zhao W, Li Y. OPFRs in e-waste sites: Integrating in silico approaches, selective bioremediation, and health risk management of residents surrounding. JOURNAL OF HAZARDOUS MATERIALS 2022; 429:128304. [PMID: 35074750 DOI: 10.1016/j.jhazmat.2022.128304] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/06/2022] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
A multilevel index system of organophosphate flame retardant bioremediation effect in an e-waste handling area was established under three bioremediation scenarios (scenario I, plant absorption; scenario II, plant-microbial combined remediation; scenario III, microbial degradation). Directional modification of OPFR substitutes with high selective bioremediation was performed. The virtual amino acid mutation approach was utilised to generate high-efficiency selective absorption/degradation mutant proteins (MPs) in a plant-microbial system under varying conditions. In scenario III, the MP's microbial degrading ability to replace molecules was increased to the greatest degree (165.82%). Appropriate foods such as corn, pig liver, and yam should be consumed, whereas the simultaneous consumption of high protein foods such as pig liver and walnut should be avoided; sweet potato and yam are believed to be prevent OPFRs and substitute molecules from entering the human body through multiple pathways for reduced genotoxicity of OPFRs in the populations of e-waste handling areas (the reduction degree can reach 85.12%). The study provides a theoretical basis for the development of ecologically acceptable OPFR substitutes and innovative high-efficiency bioremediation MPs, as well as for the reduction of the joint toxicity risk of multiple ingestion route exposure/gene damage of OPFRs in high OPFR exposure sites.
Collapse
Affiliation(s)
- Jiawen Yang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China.
| | - Xixi Li
- Northern Region Persistent Organic Pollution Control (NRPOP) Laboratory, Faculty of Engineering and Applied Science, Memorial University, St. John's NL A1B 3X5, Canada.
| | - Hao Yang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China.
| | - Wenjin Zhao
- College of New Energy and Environment, Jilin University, Changchun 130012, China.
| | - Yu Li
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China.
| |
Collapse
|
45
|
Rezig L, Chemkhi H, Gharsallah K, Mokbli S, B'chir F, Ben Achour N, Bourgou S, Chouaibi M. Profile characterization and biological activities of cold pressed Garden Cress (Lepidium sativum) seed oil. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
46
|
Whole and Purified Aqueous Extracts of Nigella sativa L. Seeds Attenuate Apoptosis and the Overproduction of Reactive Oxygen Species Triggered by p53 Over-Expression in the Yeast Saccharomyces cerevisiae. Cells 2022; 11:cells11050869. [PMID: 35269491 PMCID: PMC8909299 DOI: 10.3390/cells11050869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Plants are an important source of pharmacologically active compounds. In the present work, we characterize the impact of black cumin (Nigella sativa L.) aqueous extracts on a yeast model of p53-dependent apoptosis. To this end, the Saccharomyces cerevisiae recombinant strain over-expressing p53 was used. The over-expression of p53 triggers the expression of apoptotic markers: the externalization of phosphatidylserine, mitochondrial defect associated with cytochrome-c release and the induction of DNA strand breaks. These different effects were attenuated by Nigella sativa L. aqueous extracts, whereas these extracts have no effect on the level of p53 expression. Thus, we focus on the anti-apoptotic molecules present in the aqueous extract of Nigella sativa L. These extracts were purified and characterized by complementary chromatographic methods. Specific fluorescent probes were used to determine the effect of the extracts on yeast apoptosis. Yeast cells over-expressing p53 decrease in relative size and have lower mitochondrial content. The decrease in cell size was proportional to the decrease in mitochondrial content and of mitochondrial membrane potential (ΔΨm). These effects were prevented by the purified aqueous fraction obtained by fractionation with different columns, named C4 fraction. Yeast cell death was also characterized by reactive oxygen species (ROS) overproduction. In the presence of the C4 fraction, ROS overproduction was strongly reduced. We also noted that the C4 fraction promotes the cell growth of control yeast cells, which do not express p53, supporting the fact that this purified extract acts on cellular mediators activating cell proliferation independently of p53. Altogether, our data obtained on yeast cells over-expressing p53 demonstrate that anti-apoptotic molecules targeting p53-induced apoptosis associated with mitochondrial dysfunction and ROS overproduction are present in the aqueous extracts of Nigella seeds and in the purified aqueous C4 fraction.
Collapse
|
47
|
Canzoneri F, Leoni V, Rosso G, Risso D, Menta R, Poli G. Oxysterols as Reliable Markers of Quality and Safety in Cholesterol Containing Food Ingredients and Products. Front Nutr 2022; 9:853460. [PMID: 35252316 PMCID: PMC8890664 DOI: 10.3389/fnut.2022.853460] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/20/2022] Open
Abstract
Cholesterol is a lipid of high nutritional value that easily undergoes oxidation through enzymatic and non-enzymatic pathways, leading to a wide variety of cholesterol oxidation products (COPs), more commonly named oxysterols. The major oxysterols found in animal products are 7α-hydroxycholesterol, 7β-hydroxycholesterol, 7-ketocholesterol, 5α,6α-epoxycholesterol, 5β,6β-epoxycholesterol, cholestan-3β,5α,6β-triol, and 25-hydroxycholesterol. They are all produced by cholesterol autoxidation, thus belonging to the non-enzymatic oxysterol subfamily, even if 7α-hydroxycholesterol and 25-hydroxycholesterol are, in part, generated enzymatically as well. A further oxysterol of the full enzymatic origin has recently been detected for the first time in milk of both human and bovine origin, namely 27-hydroxycholesterol. Nowadays, gas or liquid chromatography combined to mass spectrometry allows to measure all these oxysterols accurately in raw and in industrially processed food. While non-enzymatic oxysterols often exhibited in vitro relevant cytotoxicity, above all 7β-hydroxycholesterol and 7-ketocholesterol, 27-hydroxycholesterol, as well as 25-hydroxycholesterol, shows a broad spectrum in vitro antiviral activity, inhibition of SARS-CoV-2 included, and might contribute to innate immunity. Quantification of oxysterols was afforded over the years, almost always focused on a few family's compounds. More comprehensive COPs measurements, also including oxysterols of enzymatic origin, are, nowadays, available, which better display the many advantages of systematically adopting this family of compounds as markers of quality, safety, and nutritional value in the selection of ingredients in processing and storage. Regarding foodstuff shelf life, COPs monitoring already provided useful hints for more suitable packaging. The identification of a subset of non-enzymatic and enzymatic oxysterols to be routinely assessed in food production and storage is proposed.
Collapse
Affiliation(s)
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, ASST Brianza, School of Medicine and Surgery, Hospital of Desio, University of Milano Bicocca, Milan, Italy
| | | | - Davide Risso
- Soremartec Italia Srl, Ferrero Group, Alba, Italy
| | | | - Giuseppe Poli
- Unit of General Pathology and Physiopathology, Department of Clinical and Biological Sciences, San Luigi Hospital, University of Turin, Turin, Italy
| |
Collapse
|
48
|
Mo Y, To KKW, Zhou R, Liu L, Cao T, Huang H, Du Z, Lim CYH, Yim LY, Luk TY, Chan JMC, Chik TSH, Lau DPL, Tsang OTY, Tam AR, Hung IFN, Yuen KY, Chen Z. Mitochondrial Dysfunction Associates With Acute T Lymphocytopenia and Impaired Functionality in COVID-19 Patients. Front Immunol 2022; 12:799896. [PMID: 35095881 PMCID: PMC8795605 DOI: 10.3389/fimmu.2021.799896] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in rapid T lymphocytopenia and functional impairment of T cells. The underlying mechanism, however, remains incompletely understood. In this study, we focused on characterizing the phenotype and kinetics of T-cell subsets with mitochondrial dysfunction (MD) by multicolor flow cytometry and investigating the association between MD and T-cell functionality. While 73.9% of study subjects displayed clinical lymphocytopenia upon hospital admission, a significant reduction of CD4 or CD8 T-cell frequency was found in all asymptomatic, symptomatic, and convalescent cases. CD4 and CD8 T cells with increased MD were found in both asymptomatic and symptomatic patients within the first week of symptom onset. Lower proportion of memory CD8 T cell with MD was found in severe patients than in mild ones at the stage of disease progression. Critically, the frequency of T cells with MD in symptomatic patients was preferentially associated with CD4 T-cell loss and CD8 T-cell hyperactivation, respectively. Patients bearing effector memory CD4 and CD8 T cells with the phenotype of high MD exhibited poorer T-cell responses upon either phorbol 12-myristate-13-acetate (PMA)/ionomycin or SARS-CoV-2 peptide stimulation than those with low MD. Our findings demonstrated an MD-associated mechanism underlying SARS-CoV-2-induced T lymphocytopenia and functional impairment during the acute phase of infection.
Collapse
Affiliation(s)
- Yufei Mo
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kelvin Kai-Wang To
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Center for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Runhong Zhou
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Li Liu
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Tianyu Cao
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Haode Huang
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Zhenglong Du
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chun Yu Hubert Lim
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lok-Yan Yim
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Tsz-Yat Luk
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jacky Man-Chun Chan
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Thomas Shiu-Hong Chik
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Daphne Pui-Ling Lau
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Owen Tak-Yin Tsang
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Anthony Raymond Tam
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Center for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,Center for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
49
|
Soh S, Ong WY. Effect of Withanolide A on 7-Ketocholesterol Induced Cytotoxicity in hCMEC/D3 Brain Endothelial Cells. Cells 2022; 11:cells11030457. [PMID: 35159267 PMCID: PMC8834337 DOI: 10.3390/cells11030457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 11/21/2022] Open
Abstract
Withanolide A is a naturally occurring phytochemical that is found in Ashwagandha (Withania somnifera, fam. Solanaceae) or Indian Ginseng. In the current study, we elucidated the effect of withanolide A on 7-ketocholesterol (7KC) induced injury in hCMEC/D3 human brain endothelial cells. 7KC is a cholesterol oxidation product or oxysterol that is present in atherosclerotic plaques and is elevated in the plasma of patients with hypercholesterolemia and/or diabetes mellitus. Results showed that withanolide A significantly reduced the effects of 7KC, which include loss of endothelial cell viability, increase in expression of pro-inflammatory genes-IL-1β, IL-6, IL-8, TNF-α, cyclooxygenase-2 (COX-2), increased COX-2 enzyme activity, increased ROS formation, increased expression of inducible nitric oxide synthase and genes associated with blood clotting, including Factor 2/thrombin, Factor 8, von Willebrand factor, and thromboxane A synthase, and increased human thrombin enzyme activity. Some of the above effects of withanolide A on 7KC were reduced in the presence of the glucocorticoid receptor antagonist, mifepristone (RU486). These findings suggest that the glucocorticoid receptor could play a role in the cytoprotective, antioxidant, and anti-clotting effects of withanolide A against 7KC. Further studies are necessary to elucidate the detailed mechanisms of action of withanolide A against oxysterol-induced injury.
Collapse
Affiliation(s)
- Sandra Soh
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore;
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore;
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
- Correspondence:
| |
Collapse
|
50
|
Role of Bioactive Compounds in the Regulation of Mitochondrial Dysfunctions in Brain and Age-Related Neurodegenerative Diseases. Cells 2022; 11:cells11020257. [PMID: 35053373 PMCID: PMC8773907 DOI: 10.3390/cells11020257] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondria are multifunctional organelles that participate in a wide range of metabolic processes, including energy production and biomolecule synthesis. The morphology and distribution of intracellular mitochondria change dynamically, reflecting a cell’s metabolic activity. Oxidative stress is defined as a mismatch between the body’s ability to neutralise and eliminate reactive oxygen and nitrogen species (ROS and RNS). A determination of mitochondria failure in increasing oxidative stress, as well as its implications in neurodegenerative illnesses and apoptosis, is a significant developmental process of focus in this review. The neuroprotective effects of bioactive compounds linked to neuronal regulation, as well as related neuronal development abnormalities, will be investigated. In conclusion, the study of secondary components and the use of mitochondrial features in the analysis of various neurodevelopmental diseases has enabled the development of a new class of mitochondrial-targeted pharmaceuticals capable of alleviating neurodegenerative disease states and enabling longevity and healthy ageing for the vast majority of people.
Collapse
|