1
|
Duraisamy P, Ravi S, Martin LC, Kumaresan M, Manikandan B, Ramar M. Differential phagocytic expression of IC-21 macrophages and their scavenging receptors during inflammatory induction by oxysterol: A microscopic approach. Microsc Res Tech 2024; 87:2745-2756. [PMID: 38984373 DOI: 10.1002/jemt.24647] [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: 04/01/2024] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Phagocytosis by macrophages dates back to a long history in science, this present study deals with new approaches that have been analyzed and standardized towards the interesting aspects of primary and secondary macrophages. The distinct morphological differences in primary and secondary phagocytic cells were observed and the phagocytic response of secondary macrophages under the influence of 7-ketocholesterol and lipopolysaccharide was analyzed. The primary peritoneal and secondary IC-21 cells unveiled explicit differences in nuclear numbers shapes and sizes of the granules present within the cytoplasmic region. Further, potent inducers 7KCh and LPS influenced an effective activation of IC-21 macrophages and resulted in ROS generation, irregulated protein expressions of CD86, CD68, and CD206 with enhanced phagocytic responses towards goat, cow, and human RBC targets with significant phagocytic rate and index were observed. Moreover, a remarkable observation of target specificity and aggregations with IC-21 phagocytic macrophages revealed the notion that specific membrane receptors and secretory molecules (lysosomes) are primarily involved in their phagocytic mechanism. RESEARCH HIGHLIGHTS: IC-21 macrophages are peritoneal origin from mice but the primary peritoneal macrophages and cell line show distinct differences. IC-21 macrophages express target-specific phagocytosis. Phagocytosis in IC-21 macrophages is regulated by CD markers (68, 86, and 206).
Collapse
Affiliation(s)
| | - Sangeetha Ravi
- Department of Zoology, University of Madras, Chennai, India
| | | | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai, India
| | | |
Collapse
|
2
|
Lara-Guzmán OJ, Arango-González Á, Rivera DA, Muñoz-Durango K, Sierra JA. The colonic polyphenol catabolite dihydroferulic acid (DHFA) regulates macrophages activated by oxidized LDL, 7-ketocholesterol, and LPS switching from pro- to anti-inflammatory mediators. Food Funct 2024; 15:10399-10413. [PMID: 39320081 DOI: 10.1039/d4fo02114b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Macrophage activation plays a central role in the development of atherosclerotic plaques. Interaction with oxidized low-density lipoprotein (oxLDL) leads to macrophage differentiation into foam cells and oxylipin production, contributing to plaque formation. 7-Ketocholesterol (7KC) is an oxidative byproduct of cholesterol found in oxLDL particles and is considered a factor contributing to plaque progression. During atherosclerotic lesion regression or stabilization, macrophages undergo a transformation from a pro-inflammatory phenotype to a reparative anti-inflammatory state. Interleukin-10 (IL-10) and PGE1 appear to be crucial in resolving both acute and chronic inflammatory processes. After coffee consumption, the gut microbiota processes non-absorbed chlorogenic acids producing various lower size phenolic acids. These colonic catabolites, including dihydroferulic acid (DHFA), may exert various local and systemic effects. We focused on DHFA's impact on inflammation and oxidative stress in THP-1 macrophages exposed to oxLDL, 7KC, and lipopolysaccharides (LPS). Our findings reveal that DHFA inhibits the release of several pro-inflammatory mediators induced by LPS in macrophages, such as CCL-2, CCL-3, CCL-5, TNF-α, IL-6, and IL-17. Furthermore, DHFA reduces IL-18 and IL-1β secretion in an inflammasome-like model. DHFA demonstrated additional benefits: it decreased oxLDL uptake and CD36 expression induced by oxLDL, regulated reactive oxygen species (ROS) and 8-isoprostane secretion (indicating oxidative stress modulation), and selectively increased IL-10 and PGE1 levels in the presence of inflammatory stimuli (LPS and 7KC). Finally, our study highlights the pivotal role of PGE1 in foam cell inhibition and inflammation regulation within activated macrophages. This study highlights DHFA's potential as an antioxidant and anti-inflammatory agent, particularly due to its ability to induce PGE1 and IL-10.
Collapse
Affiliation(s)
- Oscar J Lara-Guzmán
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Ángela Arango-González
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Diego A Rivera
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Katalina Muñoz-Durango
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Jelver A Sierra
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| |
Collapse
|
3
|
Duraisamy P, Angusamy A, Ravi S, Krishnan M, Martin LC, Manikandan B, Sundaram J, Ramar M. Phytol from Scoparia dulcis prevents NF-κB-mediated inflammatory responses during macrophage polarization. 3 Biotech 2024; 14:80. [PMID: 38375513 PMCID: PMC10874368 DOI: 10.1007/s13205-024-03924-9] [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: 09/28/2023] [Accepted: 01/07/2024] [Indexed: 02/21/2024] Open
Abstract
Macrophages are primary immune cells that mediate a wide range of inflammatory diseases through their polarization potential. In this study, phytol isolated from Scoparia dulcis has been explored against 7-ketocholesterol and bacterial lipopolysaccharide-induced macrophage polarization in IC-21 cells. Isolated phytol has been characterized using GC-MS, TLC, HPTLC, FTIR, 1H-NMR, and HPLC analyses. The immunomodulatory effects of viable concentrations of phytol were tested on oxidative stress, arginase activity, nuclear and mitochondrial membrane potentials in IC-21 cells in addition to the modulation of calcium and lipids. Further, gene and protein expression of atherogenic markers were studied. Results showed that the isolated phytol at a viable concentration of 400 µg/ml effectively reduced the production of nitric oxide, superoxide anion (ROS generation), calcium and lipid accumulation, stabilized nuclear and mitochondrial membranes, and increased arginase activity. The atherogenic markers including iNOS, COX-2, IL-6, IL-1β, MMP-9, CD36, and NF-κB were significantly downregulated at the levels of gene and protein expression, while macrophage surface and nuclear receptor markers (CD206, CD163, and PPAR-γ) were significantly upregulated by phytol pre-treatment in macrophages. Therefore, the present pharmacognostic study supports the role of phytol isolated from Scoparia dulcis in preventing M2-M1 macrophage polarization under inflammatory conditions, making it a promising compound. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-03924-9.
Collapse
Affiliation(s)
| | - Annapoorani Angusamy
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600015 India
| | - Janarthanan Sundaram
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| |
Collapse
|
4
|
Yu Y, Liu Y, Meng Z. Role of traditional Chinese medicine in age-related macular degeneration: exploring the gut microbiota's influence. Front Pharmacol 2024; 15:1356324. [PMID: 38333011 PMCID: PMC10850396 DOI: 10.3389/fphar.2024.1356324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The pathogenesis of age-related macular degeneration (AMD), a degenerative retinopathy, remains unclear. Administration of anti-vascular endothelial growth factor agents, antioxidants, fundus lasers, photodynamic therapy, and transpupillary warming has proven effective in alleviating symptoms; however, these interventions cannot prevent or reverse AMD. Increasing evidence suggests that AMD risk is linked to changes in the composition, abundance, and diversity of the gut microbiota (GM). Activation of multiple signaling pathways by GM metabolites, including lipopolysaccharides, oxysterols, short-chain fatty acids (SCFAs), and bile acids (BAs), influences retinal physiology. Traditional Chinese medicine (TCM), known for its multi-component and multi-target advantages, can help treat AMD by altering GM composition and regulating the levels of certain substances, such as lipopolysaccharides, reducing oxysterols, and increasing SCFA and BA contents. This review explores the correlation between GM and AMD and interventions for the two to provide new perspectives on treating AMD with TCM.
Collapse
Affiliation(s)
- Yujia Yu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Province Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhaoru Meng
- The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| |
Collapse
|
5
|
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
|
6
|
Pariente A, Peláez R, Ochoa R, Pérez-Sala Á, Villanueva-Martínez Á, Bobadilla M, Larráyoz IM. Targeting 7KCh-Induced Cell Death Response Mediated by p38, P2X7 and GSDME in Retinal Pigment Epithelium Cells with Sterculic Acid. Pharmaceutics 2023; 15:2590. [PMID: 38004569 PMCID: PMC10675123 DOI: 10.3390/pharmaceutics15112590] [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: 09/14/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Age-related macular degeneration (AMD) is the main cause of blindness in developed countries. AMD is characterized by the formation of drusen, which are lipidic deposits, between retinal pigment epithelium (RPE) and the choroid. One of the main molecules accumulated in drusen is 7-Ketocholesterol (7KCh), an oxidized-cholesterol derivative. It is known that 7KCh induces inflammatory and cytotoxic responses in different cell types and the study of its mechanism of action is interesting in order to understand the development of AMD. Sterculic acid (SA) counteracts 7KCh response in RPE cells and could represent an alternative to improve currently used AMD treatments, which are not efficient enough. In the present study, we determine that 7KCh induces a complex cell death signaling characterized by the activation of necrosis and an alternative pyroptosis mediated by P2X7, p38 and GSDME, a new mechanism not yet related to the response to 7KCh until now. On the other hand, SA treatment can successfully attenuate the activation of both necrosis and pyroptosis, highlighting its therapeutic potential for the treatment of AMD.
Collapse
Affiliation(s)
- Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
| | - Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
| | - Rodrigo Ochoa
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
- Proteomics Research Core Facility, Aragonese Institute of Health Sciences (IACS), San Juan Bosco 13, 50009 Zaragoza, Spain
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
| | - Ángela Villanueva-Martínez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
| | - Miriam Bobadilla
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
| | - Ignacio M. Larráyoz
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain; (A.P.); (R.P.); (R.O.); (Á.P.-S.); (Á.V.-M.)
- Biomarkers, Artificial Intelligence and Signaling (BIAS), Department of Nursing, University of La Rioja, Duquesa de la Victoria 88, 26006 Logroño, Spain
| |
Collapse
|
7
|
Zhang Y, Xu S, Xu J, Xu F, Lu G, Zhou J, Gu S, Wang J. Prognostic value of plasma 7-ketocholesterol in sepsis. Clin Chim Acta 2023; 548:117467. [PMID: 37399884 DOI: 10.1016/j.cca.2023.117467] [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: 03/07/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Early evaluation of the severity of sepsis and estimation of its prognosis remains one of the main challenges in current therapeutic strategies. This study aimed to evaluate the prognostic value of plasma 7-ketocholesterol (7-KC) in sepsis. METHODS We retrospectively measured by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) the plasma 7-KC concentration in 176 patients with sepsis and 90 healthy volunteers. A multivariate Cox proportional hazard model was introduced to identify independent factors, including plasma 7-KC and clinical features, for the 28-day mortality of sepsis, and a nomogram for predicting the 28-day mortality of sepsis was established. Decision curve analysis (DCA) was performed to assess the prediction model of death risk of sepsis. RESULTS The area under the curve (AUC) of plasma 7-KC in diagnosing sepsis was 0.899 (95% CI = 0.862-0.935, P < 0.001), while it was 0.830 (95% CI = 0.764-0.894, P < 0.001) in diagnosing septic shock. The AUCs of plasma 7-KC in predicting the survival of sepsis patients in the training cohort and the test cohort were 0.770 (95% CI = 0.692-0.848, P < 0.05) and 0.869 (95% CI = 0.763-0.974, P < 0.05), respectively. In addition, high plasma 7-KC expression predicts poor prognosis in sepsis. Then, 7-KC and platelet count were identified as the two factors with significant differences by a multivariate Cox proportional hazard model, and the 28-day mortality probability ranged from 0.002 to 0.985 and was assessed using a nomogram. DCA results revealed that the combination of plasma 7-KC and platelet count showed the best prognostic efficiency of the risk threshold compared to a single factor in both the training cohort and test cohort. CONCLUSIONS Collectively, the elevated plasma 7-KC level is an indicator of sepsis and was identified as a prognostic indicator for sepsis patients, providing a landscape for predicting survival in early sepsis with potential clinical utility.
Collapse
Affiliation(s)
- Yueyuan Zhang
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China
| | - Sha Xu
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China
| | - Jianxin Xu
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China
| | - Fuchao Xu
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China
| | - Geng Lu
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Jiawei Zhou
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Shuangshuang Gu
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China; Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China.
| | - Jun Wang
- Department of Emergency, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210000, China; Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China.
| |
Collapse
|
8
|
Pariente A, Pérez-Sala Á, Ochoa R, Bobadilla M, Villanueva-Martínez Á, Peláez R, Larráyoz IM. Identification of 7-Ketocholesterol-Modulated Pathways and Sterculic Acid Protective Effect in Retinal Pigmented Epithelium Cells by Using Genome-Wide Transcriptomic Analysis. Int J Mol Sci 2023; 24:ijms24087459. [PMID: 37108627 PMCID: PMC10144535 DOI: 10.3390/ijms24087459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. AMD is characterized by the formation of lipidic deposits between the retinal pigment epithelium (RPE) and the choroid called drusen. 7-Ketocholesterol (7KCh), an oxidized-cholesterol derivative, is closely related to AMD as it is one of the main molecules accumulated in drusen. 7KCh induces inflammatory and cytotoxic responses in different cell types, and a better knowledge of the signaling pathways involved in its response would provide a new perspective on the molecular mechanisms that lead to the development of AMD. Furthermore, currently used therapies for AMD are not efficient enough. Sterculic acid (SA) attenuates the 7KCh response in RPE cells and is presented as an alternative to improve these therapies. By using genome-wide transcriptomic analysis in monkey RPE cells, we have provided new insight into 7KCh-induced signaling in RPE cells, as well as the protective capacity of SA. 7KCh modulates the expression of several genes associated with lipid metabolism, endoplasmic reticulum stress, inflammation and cell death and induces a complex response in RPE cells. The addition of SA successfully attenuates the deleterious effect of 7KCh and highlights its potential for the treatment of AMD.
Collapse
Affiliation(s)
- Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Rodrigo Ochoa
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
- Proteomics Research Core Facility, Aragonese Institute of Health Sciences (IACS), University of Zaragoza, San Juan Bosco 13, 50009 Zaragoza, Spain
| | - Miriam Bobadilla
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Ángela Villanueva-Martínez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Ignacio M Larráyoz
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
- Biomarkers, Artificial Intelligence and Signaling (BIAS), Department of Nursing, University of La Rioja, Duquesa de la Victoria 88, 26006 Logroño, Spain
| |
Collapse
|
9
|
Protective Effect of Ergothioneine against 7-Ketocholesterol-Induced Mitochondrial Damage in hCMEC/D3 Human Brain Endothelial Cells. Int J Mol Sci 2023; 24:ijms24065498. [PMID: 36982572 PMCID: PMC10056831 DOI: 10.3390/ijms24065498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Recent findings have suggested that the natural compound ergothioneine (ET), which is synthesised by certain fungi and bacteria, has considerable cytoprotective potential. We previously demonstrated the anti-inflammatory effects of ET on 7-ketocholesterol (7KC)-induced endothelial injury in human blood-brain barrier endothelial cells (hCMEC/D3). 7KC is an oxidised form of cholesterol present in atheromatous plaques and the sera of patients with hypercholesterolaemia and diabetes mellitus. The aim of this study was to elucidate the protective effect of ET on 7KC-induced mitochondrial damage. Exposure of human brain endothelial cells to 7KC led to a loss of cell viability, together with an increase in intracellular free calcium levels, increased cellular and mitochondrial reactive oxygen species, a decrease in mitochondrial membrane potential, reductions in ATP levels, and increases in mRNA expression of TFAM, Nrf2, IL-1β, IL-6 and IL-8. These effects were significantly decreased by ET. Protective effects of ET were diminished when endothelial cells were coincubated with verapamil hydrochloride (VHCL), a nonspecific inhibitor of the ET transporter OCTN1 (SLC22A4). This outcome demonstrates that ET-mediated protection against 7KC-induced mitochondrial damage occurred intracellularly and not through direct interaction with 7KC. OCTN1 mRNA expression itself was significantly increased in endothelial cells after 7KC treatment, consistent with the notion that stress and injury may increase ET uptake. Our results indicate that ET can protect against 7KC-induced mitochondrial injury in brain endothelial cells.
Collapse
|
10
|
Groenen AG, La Rose AM, Li M, Bazioti V, Svendsen AF, Kloosterhuis NJ, Ausema A, Pranger A, Heiner-Fokkema MR, Niezen-Koning KE, Houben T, Shiri-Sverdlov R, Westerterp M. Elevated granulocyte-colony stimulating factor and hematopoietic stem cell mobilization in Niemann-Pick type C1 disease. J Lipid Res 2022; 63:100167. [PMID: 35007562 PMCID: PMC8953690 DOI: 10.1016/j.jlr.2021.100167] [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: 09/10/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022] Open
Abstract
Niemann-Pick type C1 (NPC1) disease is a progressive lysosomal storage disorder caused by mutations of the NPC1 gene. While neurodegeneration is the most severe symptom, a large proportion of NPC1 patients also present with splenomegaly, which has been attributed to cholesterol and glycosphingolipid accumulation in late endosomes and lysosomes. However, recent data also reveal an increase in the inflammatory monocyte subset in the Npc1nih mouse model expressing an Npc1 null allele. We evaluated the contribution of hematopoietic cells to splenomegaly in NPC1 disease under conditions of hypercholesterolemia. We transplanted Npc1nih (Npc1 null mutation) or Npc1wt bone marrow (BM) into Ldlr-/- mice and fed these mice a cholesterol-rich Western-type diet. At 9 weeks after BM transplant, on a chow diet, the Npc1 null mutation increased plasma granulocyte-colony stimulating factor (G-CSF) by 2-fold and caused mild neutrophilia. At 18 weeks after BM transplant, including 9 weeks of Western-type diet feeding, the Npc1 mutation increased G-csf mRNA levels by ∼5-fold in splenic monocytes/macrophages accompanied by a ∼4-fold increase in splenic neutrophils compared with controls. We also observed ∼5-fold increased long-term and short-term hematopoietic stem cells (HSCs) in the spleen, and a ∼30-75% decrease of these populations in BM, reflecting HSC mobilization, presumably downstream of elevated G-CSF. In line with these data, four patients with NPC1 disease showed higher plasma G-CSF compared with age-matched and gender-matched healthy controls. In conclusion, we show elevated G-CSF levels and HSC mobilization in the setting of an Npc1 null mutation and propose that this contributes to splenomegaly in patients with NPC1 disease.
Collapse
Affiliation(s)
- Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anouk M La Rose
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mengying Li
- Department of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, The Netherlands
| | - Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arthur F Svendsen
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albertina Ausema
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alle Pranger
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Klary E Niezen-Koning
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tom Houben
- Department of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- Department of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, The Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
11
|
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
|
12
|
Yang L, Yu P, Chen M, Lei B. Mammalian Target of Rapamycin Inhibitor Rapamycin Alleviates 7-Ketocholesterol Induced Inflammatory Responses and Vascular Endothelial Growth Factor Elevation by Regulating MAPK Pathway in Human Retinal Pigment Epithelium Cells. J Ocul Pharmacol Ther 2021; 38:189-200. [PMID: 34936813 DOI: 10.1089/jop.2021.0082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose: To validate the protective effect of a mammalian target of rapamycin (mTOR) inhibitor on human retinal pigment epithelium (RPE) cells challenged with 7-ketocholesterol (7-KC) and explored the underlying mechanisms. Methods: Human primary RPE (hRPE) cells and ARPE-19 cells were cultured with or without 10 nM of rapamycin for 6 h before being exposed to 10 μM of 7-KC for 24 h. The transcriptome of 7-KC challenged ARPE-19 cells was investigated by RNA sequencing (RNA-seq). The effects of 7-KC and rapamycin on the viability of ARPE-19 cells were measured with CCK-8. Gene expression was verified by real-time PCR, and protein levels were determined by ELISA or Western blotting. Results: The expression of IL-6, IL-8, and vascular endothelial growth factor (VEGF) in RPE cells was markedly increased after stimulation with 7-KC for 12/24 h compared with the controls. RNA-seq showed that a total of 10,243 genes were differentially expressed, with 5,518 genes upregulated and 4,725 genes downregulated between the 7-KC treated and the control group. Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that 7-KC stimulation activated mTOR signaling and other pathways, including adherent junction, MAPK, and Wnt signalings. mTOR inhibitor rapamycin significantly suppressed the elevation of IL-6, IL-8, and VEGF stimulated by 7-KC. Rapamycin not only decreased the level of phosphorylated mTOR, P70S6K, 4EBP1 but also inhibited the activation of MAPK pathway. Conclusions: Inhibition of mTOR signaling pathway suppressed the elevation of inflammatory cytokines IL-6, IL-8, and the angiogenic agent VEGF induced by 7-KC. The protective effect of rapamycin was associated with its downregulation on MAPK pathway.
Collapse
Affiliation(s)
- Lin Yang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Peng Yu
- Department of Ophthalmology, People's Hospital of Changshou District, Chongqing, China
| | - Mei Chen
- Centre for Experimental Medicine, Queen's University, Belfast, United Kingdom
| | - Bo Lei
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Wang Q, Wang J, Wang J, Ju X, Zhang H. Molecular mechanism of shikonin inhibiting tumor growth and potential application in cancer treatment. Toxicol Res (Camb) 2021; 10:1077-1084. [PMID: 34956612 PMCID: PMC8692723 DOI: 10.1093/toxres/tfab107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Shikonin is one of the major bioactive components of Lithospermum erythrorhizon. It has a good killing effect in a variety of tumor cells. Its antitumor effect involves multiple targets and pathways and has received extensive attention and study in recent years. In this review, we systematically review recent progress in determining the antitumor mechanism of shikonin and its derivatives, specifically their induction of reactive oxygen species production, inhibition of EGFR and PI3K/AKT signaling pathway activation, inhibition of angiogenesis and induction of apoptosis and necroptosis. We also discuss the application of nanoparticles loaded with shikonin in the targeted therapy of various cancers. Finally, we suggest new strategies for the clinical application of shikonin and its derivatives.
Collapse
Affiliation(s)
- Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jing Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jiayou Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
14
|
A2E-induced inflammation and angiogenesis in RPE cells in vitro are modulated by PPAR-α, -β/δ, -γ, and RXR antagonists and by norbixin. Aging (Albany NY) 2021; 13:22040-22058. [PMID: 34544906 PMCID: PMC8507260 DOI: 10.18632/aging.203558] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 09/03/2021] [Indexed: 12/15/2022]
Abstract
N-retinylidene-N-retinylethanolamine (A2E) plays a central role in age-related macular degeneration (AMD) by inducing angiogenesis and inflammation. A2E effects are mediated at least partly via the retinoic acid receptor (RAR)-α. Here we show that A2E binds and transactivates also peroxisome proliferator-activated receptors (PPAR) and retinoid X receptors (RXR). 9’-cis-norbixin, a di-apocarotenoid is also a ligand of these nuclear receptors (NR). Norbixin inhibits PPAR and RXR transactivation induced by A2E. Moreover, norbixin reduces protein kinase B (AKT) phosphorylation, NF-κB and AP-1 transactivation and mRNA expression of the inflammatory interleukins (IL) -6 and -8 and of vascular endothelial growth factor (VEGF) enhanced by A2E. By contrast, norbixin increases matrix metalloproteinase 9 (MMP9) and C-C motif chemokine ligand 2 (CCL2) mRNA expression in response to A2E. Selective PPAR-α, -β/δ and –γ antagonists inhibit the expression of IL-6 and IL-8 while only the antagonist of PPAR-γ inhibits the transactivation of NF-κB following A2E exposure. In addition, a cocktail of all three PPARs antagonists and also HX531, an antagonist of RXR reproduce norbixin effects on inflammation. Altogether, A2E’s deleterious biological effects could be inhibited through PPAR and RXR regulation. Moreover, the modulation of these NR by norbixin may open new avenues for the treatment of AMD.
Collapse
|
15
|
Ghzaiel I, Sassi K, Zarrouk A, Nury T, Ksila M, Leoni V, Bouhaouala-Zahar B, Hammami S, Hammami M, Mackrill JJ, Samadi M, Ghrairi T, Vejux A, Lizard G. 7-Ketocholesterol: Effects on viral infections and hypothetical contribution in COVID-19. J Steroid Biochem Mol Biol 2021; 212:105939. [PMID: 34118414 PMCID: PMC8188774 DOI: 10.1016/j.jsbmb.2021.105939] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
7-Ketocholesterol, which is one of the earliest cholesterol oxidization products identified, is essentially formed by the auto-oxidation of cholesterol. In the body, 7-ketocholesterol is both provided by food and produced endogenously. This pro-oxidant and pro-inflammatory molecule, which can activate apoptosis and autophagy at high concentrations, is an abundant component of oxidized Low Density Lipoproteins. 7-Ketocholesterol appears to significantly contribute to the development of age-related diseases (cardiovascular diseases, age-related macular degeneration, and Alzheimer's disease), chronic inflammatory bowel diseases and to certain cancers. Recent studies have also shown that 7-ketocholesterol has anti-viral activities, including on SARS-CoV-2, which are, however, lower than those of oxysterols resulting from the oxidation of cholesterol on the side chain. Furthermore, 7-ketocholesterol is increased in the serum of moderately and severely affected COVID-19 patients. In the case of COVID-19, it can be assumed that the antiviral activity of 7-ketocholesterol could be counterbalanced by its toxic effects, including pro-oxidant, pro-inflammatory and pro-coagulant activities that might promote the induction of cell death in alveolar cells. It is therefore suggested that this oxysterol might be involved in the pathophysiology of COVID-19 by contributing to the acute respiratory distress syndrome and promoting a deleterious, even fatal outcome. Thus, 7-ketocholesterol could possibly constitute a lipid biomarker of COVID-19 outcome and counteracting its toxic effects with adjuvant therapies might have beneficial effects in COVID-19 patients.
Collapse
Affiliation(s)
- Imen Ghzaiel
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University Tunis-El Manar, Faculty of Sciences of Tunis, 2092 Tunis, Tunisia.
| | - Khouloud Sassi
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, 1007 Tunis, Tunisia.
| | - Amira Zarrouk
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University of Sousse, Faculty of Medicine, Sousse, Tunisia.
| | - Thomas Nury
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| | - Mohamed Ksila
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMoleecules, LR18ES03, Department of Biologie, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospitals of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Molecules, Pasteur Institute of Tunis & University of Tunis El Manar, 1002 Tunis, Tunisia.
| | - Sonia Hammami
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia.
| | - Mohamed Hammami
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia.
| | - John J Mackrill
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland.
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Dept of Chemistry, Univ. Lorraine, Metz Technopôle, Metz, France.
| | - Taoufik Ghrairi
- University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMoleecules, LR18ES03, Department of Biologie, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Anne Vejux
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| | - Gérard Lizard
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| |
Collapse
|
16
|
Ravi S, Duraisamy P, Krishnan M, Martin LC, Manikandan B, Raman T, Sundaram J, Arumugam M, Ramar M. An insight on 7- ketocholesterol mediated inflammation in atherosclerosis and potential therapeutics. Steroids 2021; 172:108854. [PMID: 33930389 DOI: 10.1016/j.steroids.2021.108854] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 11/24/2022]
Abstract
7-ketocholesterol, a toxic oxidative product of oxysterol is a causative agent of several diseases and disabilities concomitant to aging including cardiovascular diseases like atherosclerosis. Auto-oxidation of cholesterol esters present in low-density lipoprotein (LDL) deposits lead to the formation of oxidized LDL (Ox-LDL) along with its byproducts, namely 7KCh. It is predominantly found in atherosclerotic plaque and also found to be more atherogenic than cholesterol by being cytotoxic, interfering with cellular homeostasis. This makes it a serious threat by being the foremost cause of morbidity and mortality worldwide and is likely to become more serious during forth coming years. It involves in mediating inflammatory mechanisms characterized by the advancement of fibroatheroma plaques. The atherosclerotic lesion is composed of Ox-LDL along with fibrotic mass consisting of immune cells and molecules. Macrophages being the specialized phagocytic cells, contribute to removal of detrimental contents of the lesion along with accumulated lipids leading to alteration of its biology and functionality due to its plasticity. Here, we have explored the known as well as proposed mechanisms involved with 7KCh associated atherogenesis along with potential therapeutic strategies for targeting 7KCh as a diagnostic and target in medicine.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Livya C Martin
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilakanni's College for Women, Chennai 600015, India
| | - Thiagarajan Raman
- Department of Advanced Zoology and Biotechnology, Ramakrishna Mission Vivekananda College, Mylapore, Chennai 600004, India
| | - Janarthanan Sundaram
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Munusamy Arumugam
- 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
|
17
|
Chen J, Tchivelekete GM, Zhou X, Tang W, Liu F, Liu M, Zhao C, Shu X, Zeng Z. Anti-inflammatory activities of Gardenia jasminoides extracts in retinal pigment epithelial cells and zebrafish embryos. Exp Ther Med 2021; 22:700. [PMID: 34007309 PMCID: PMC8120509 DOI: 10.3892/etm.2021.10132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of visual impairment in developed countries. Inflammation serves a critical role in the pathogenesis of AMD. Gardenia jasminoides is found in several regions of China and is traditionally used as an organic yellow dye but has also been widely used as a therapeutic agent in numerous diseases, including inflammation, depression, hepatic and vascular disorders, which may reflect the variability of functional compounds that are present in Gardenia jasminoides extracts (GJE). To investigate the therapeutic potential of GJE for AMD, ARPE-19 cells were treated with lipopolysaccharide (LPS) or LPS plus GJE. GJE significantly decreased LPS-induced expression of proinflammatory cytokines, including IL-1β, IL-6 and TNF-α. In the in vivo study, GJE inhibited CuSO4-induced migration of primitive macrophages to the lateral line in zebrafish embryos. GJE also attenuated expression of cytokines (IL-1β, IL-6 and TNF-α), NFKB activating protein (nkap) and TLR4 in ARPE-19 cells. The results of the present study demonstrated the anti-inflammatory potential of GJE in vitro and in vivo, and suggested GJE as a therapeutic candidate for AMD.
Collapse
Affiliation(s)
- Jianrong Chen
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China
| | | | - Xinzhi Zhou
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Weizhuo Tang
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China
| | - Fang Liu
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China
| | - Minzhuo Liu
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China
| | - Chenxi Zhao
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China
| | - Xinhua Shu
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK.,Department of Vision Science, Glasgow Caledonian University, Glasgow, G4 0BA, UK.,School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Zhihong Zeng
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan 410022, P.R. China.,Hunan Provincial Key Laboratory of Nutrition and Quality Control Aquatic Animals, Changsha, Hunan 410022, P.R. China
| |
Collapse
|
18
|
Zhao T, Guo X, Sun Y. Iron Accumulation and Lipid Peroxidation in the Aging Retina: Implication of Ferroptosis in Age-Related Macular Degeneration. Aging Dis 2021; 12:529-551. [PMID: 33815881 PMCID: PMC7990372 DOI: 10.14336/ad.2020.0912] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/12/2020] [Indexed: 01/19/2023] Open
Abstract
Iron is an essential component in many biological processes in the human body. It is critical for the visual phototransduction cascade in the retina. However, excess iron can be toxic. Iron accumulation and reduced efficiency of intracellular antioxidative defense systems predispose the aging retina to oxidative stress-induced cell death. Age-related macular degeneration (AMD) is characterized by retinal iron accumulation and lipid peroxidation. The mechanisms underlying AMD include oxidative stress-mediated death of retinal pigment epithelium (RPE) cells and subsequent death of retinal photoreceptors. Understanding the mechanism of the disruption of iron and redox homeostasis in the aging retina and AMD is crucial to decipher these mechanisms of cell death and AMD pathogenesis. The mechanisms of retinal cell death in AMD are an area of active investigation; previous studies have proposed several types of cell death as major mechanisms. Ferroptosis, a newly discovered programmed cell death pathway, has been associated with the pathogenesis of several neurodegenerative diseases. Ferroptosis is initiated by lipid peroxidation and is characterized by iron-dependent accumulation. In this review, we provide an overview of the mechanisms of iron accumulation and lipid peroxidation in the aging retina and AMD, with an emphasis on ferroptosis.
Collapse
Affiliation(s)
- Tantai Zhao
- 1Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Xiaojian Guo
- 1Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yun Sun
- 1Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| |
Collapse
|
19
|
Helicobacter pylori Infection Acts Synergistically with a High-Fat Diet in the Development of a Proinflammatory and Potentially Proatherogenic Endothelial Cell Environment in an Experimental Model. Int J Mol Sci 2021; 22:ijms22073394. [PMID: 33806236 PMCID: PMC8037564 DOI: 10.3390/ijms22073394] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Classic atherosclerosis risk factors do not explain all cases of chronic heart disease. There is significant evidence that gut microbiota may influence the development of atherosclerosis. The widespread prevalence of chronic Helicobacter pylori (H. pylori, HP) infections suggests that HP can be the source of components that stimulate local and systemic inflammatory responses. Elevated production of reactive oxygen species during HP infection leads to cholesterol oxidation, which drives atherogenesis. The aim of this study is to explore the link between persistent HP infection and a high-fat diet in the development of proinflammatory conditions that are potentially proatherogenic. An in vivo model of Caviae porcellus infected with HP and exposed to an experimental diet was investigated for the occurrence of a proinflammatory and proatherogenic endothelial environment. Vascular endothelial primary cells exposed to HP components were tested in vitro for oxidative stress, cell activation and apoptosis. The infiltration of inflammatory cells into the vascular endothelium of animals infected with HP and exposed to a high-fat diet was observed in conjunction with an increased level of inflammatory markers systemically. The arteries of such animals were the least elastic, suggesting the role of HP in arterial stiffness. Soluble HP components induced transformation of macrophages to foam cells in vitro and influenced the endothelial life span, which was correlated with Collagen I upregulation. These preliminary results support the hypothesis that HP antigens act synergistically with a high-fat diet in the development of proatherogenic conditions.
Collapse
|
20
|
Abstract
Cholesterol is a quantitatively and biologically significant constituent of all mammalian cell membrane, including those that comprise the retina. Retinal cholesterol homeostasis entails the interplay between de novo synthesis, uptake, intraretinal sterol transport, metabolism, and efflux. Defects in these complex processes are associated with several congenital and age-related disorders of the visual system. Herein, we provide an overview of the following topics: (a) cholesterol synthesis in the neural retina; (b) lipoprotein uptake and intraretinal sterol transport in the neural retina and the retinal pigment epithelium (RPE); (c) cholesterol efflux from the neural retina and the RPE; and (d) biology and pathobiology of defects in sterol synthesis and sterol oxidation in the neural retina and the RPE. We focus, in particular, on studies involving animal models of monogenic disorders pertinent to the above topics, as well as in vitro models using biochemical, metabolic, and omic approaches. We also identify current knowledge gaps and opportunities in the field that beg further research in this topic area.
Collapse
Affiliation(s)
- Sriganesh Ramachandra Rao
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA.
| |
Collapse
|
21
|
Zhang X, Alhasani RH, Zhou X, Reilly J, Zeng Z, Strang N, Shu X. Oxysterols and retinal degeneration. Br J Pharmacol 2021; 178:3205-3219. [PMID: 33501641 DOI: 10.1111/bph.15391] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Retinal degeneration, characterised by the progressive death of retinal neurons, is the most common cause of visual impairment. Oxysterols are the cholesterol derivatives produced via enzymatic and/or free radical oxidation that regulate cholesterol homeostasis in the retina. Preclinical and clinical studies have suggested a connection between oxysterols and retinal degeneration. Here, we summarise early and recent work related to retina oxysterol-producing enzymes and the distribution of oxysterols in the retina. We examine the impact of loss of oxysterol-producing enzymes on retinal pathology and explore the molecular mechanisms associated with the toxic or protective roles of individual oxysterols in different types of retinal degeneration. We conclude that increased efforts to better understand the oxysterol-associated pathophysiology will help in the development of effective retinal degeneration therapies. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Xun Zhang
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Reem Hasaballah Alhasani
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK.,Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Xinzhi Zhou
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Zhihong Zeng
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan, China
| | - Niall Strang
- Department of Vision Science, Glasgow Caledonian University, Glasgow, UK
| | - Xinhua Shu
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK.,Department of Vision Science, Glasgow Caledonian University, Glasgow, UK.,School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan, China
| |
Collapse
|
22
|
Transcriptomic Changes Associated with Loss of Cell Viability Induced by Oxysterol Treatment of a Retinal Photoreceptor-Derived Cell Line: An In Vitro Model of Smith-Lemli-Opitz Syndrome. Int J Mol Sci 2021; 22:ijms22052339. [PMID: 33652836 PMCID: PMC7956713 DOI: 10.3390/ijms22052339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 11/17/2022] Open
Abstract
Smith–Lemli–Opitz Syndrome (SLOS) results from mutations in the gene encoding the enzyme DHCR7, which catalyzes conversion of 7-dehydrocholesterol (7DHC) to cholesterol (CHOL). Rats treated with a DHCR7 inhibitor serve as a SLOS animal model, and exhibit progressive photoreceptor-specific cell death, with accumulation of 7DHC and oxidized sterols. To understand the basis of this cell type specificity, we performed transcriptomic analyses on a photoreceptor-derived cell line (661W), treating cells with two 7DHC-derived oxysterols, which accumulate in tissues and bodily fluids of SLOS patients and in the rat SLOS model, as well as with CHOL (negative control), and evaluated differentially expressed genes (DEGs) for each treatment. Gene enrichment analysis and compilation of DEG sets indicated that endoplasmic reticulum stress, oxidative stress, DNA damage and repair, and autophagy were all highly up-regulated pathways in oxysterol-treated cells. Detailed analysis indicated that the two oxysterols exert their effects via different molecular mechanisms. Changes in expression of key genes in highlighted pathways (Hmox1, Ddit3, Trib3, and Herpud1) were validated by immunofluorescence confocal microscopy. The results extend our understanding of the pathobiology of retinal degeneration and SLOS, identifying potential new druggable targets for therapeutic intervention into these and other related orphan diseases.
Collapse
|
23
|
Liu S, Fang Y, Yu J, Chang X. Hawthorn polyphenols reduce high glucose-induced inflammation and apoptosis in ARPE-19 cells by regulating miR-34a/SIRT1 to reduce acetylation. J Food Biochem 2021; 45:e13623. [PMID: 33491221 DOI: 10.1111/jfbc.13623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 01/07/2023]
Abstract
Diabetic retinopathy is a major complication in patients with diabetes. Herein, we investigate how hawthorn polyphenol extract (HPE) affects high glucose-induced oxidation, inflammation, and apoptosis in ARPE-19 cells. HPLC-MS/MS was used to determine HPE content and composition. Reactive oxygen species (ROS) production was assessed using fluorescence microscopy, while glucose-induced gene and protein expressions were analyzed using real-time PCR and western blotting in cells transfected with miR-34a mimics. We found that treating cells with 10 μg/ml of HPE, 30 μM procyanidin B2, chlorogenic acid, epicatechin, or resveratrol (positive control) significantly reduced ROS production and decreased apoptosis and inflammation-related factors (p < .01). Moreover, the expression level of SIRT1 was increased, while that of acetylated NF-κB p65 and p53 proteins was decreased. These data suggest that HPE can inhibit oxidative damage, inflammation, and apoptosis through the AMPK/SIRT1/NF-κB pathway, and decrease miR-34a/SIRT1/p53 pathway activation in ARPE-19 cells, thereby demonstrating a potential use as a food additive to mitigate hyperglycemia-induced retinal damage. PRACTICAL APPLICATIONS: Hawthorn polyphenol extract (HPE) significantly reduced ROS levels, apoptosis, and the expression of inflammation-related factors in ARPE-19 cells. HPE also inhibited the AMPK/SIRT1/NF-κB and miR-34a/SIRT1/p53 pathways, which are involved in hyperglycemia-induced inflammation and apoptosis of ARPE-19 cells by regulating acetylation. Thus, HPE, as a potential food additive, may mitigate hyperglycemia-induced retinal damage.
Collapse
Affiliation(s)
- Suwen Liu
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Yuan Fang
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Jincheng Yu
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Xuedong Chang
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China.,Hebei (Chengde) Hawthorn Industrial Technology Research Institute, Chengde, China
| |
Collapse
|
24
|
Rao H, Jalali JA, Johnston TP, Koulen P. Emerging Roles of Dyslipidemia and Hyperglycemia in Diabetic Retinopathy: Molecular Mechanisms and Clinical Perspectives. Front Endocrinol (Lausanne) 2021; 12:620045. [PMID: 33828528 PMCID: PMC8020813 DOI: 10.3389/fendo.2021.620045] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetic retinopathy (DR) is a significant cause of vision loss and a research subject that is constantly being explored for new mechanisms of damage and potential therapeutic options. There are many mechanisms and pathways that provide numerous options for therapeutic interventions to halt disease progression. The purpose of the present literature review is to explore both basic science research and clinical research for proposed mechanisms of damage in diabetic retinopathy to understand the role of triglyceride and cholesterol dysmetabolism in DR progression. This review delineates mechanisms of damage secondary to triglyceride and cholesterol dysmetabolism vs. mechanisms secondary to diabetes to add clarity to the pathogenesis behind each proposed mechanism. We then analyze mechanisms utilized by both triglyceride and cholesterol dysmetabolism and diabetes to elucidate the synergistic, additive, and common mechanisms of damage in diabetic retinopathy. Gathering this research adds clarity to the role dyslipidemia has in DR and an evaluation of the current peer-reviewed basic science and clinical evidence provides a basis to discern new potential therapeutic targets.
Collapse
Affiliation(s)
- Hussain Rao
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Jonathan A. Jalali
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Thomas P. Johnston
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Peter Koulen
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
- *Correspondence: Peter Koulen,
| |
Collapse
|
25
|
Neekhra A, Tran J, Esfahani PR, Schneider K, Pham K, Sharma A, Chwa M, Luthra S, Gramajo AL, Mansoor S, Kuppermann BD, Kenney MC. Memantine, Simvastatin, and Epicatechin Inhibit 7-Ketocholesterol-induced Apoptosis in Retinal Pigment Epithelial Cells But Not Neurosensory Retinal Cells In Vitro. J Ophthalmic Vis Res 2020; 15:470-480. [PMID: 33133437 PMCID: PMC7591846 DOI: 10.18502/jovr.v15i4.7781] [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: 03/20/2020] [Accepted: 06/21/2020] [Indexed: 11/27/2022] Open
Abstract
Purpose 7-ketocholesterol (7kCh), a natural byproduct of oxidation in lipoprotein deposits is implicated in the pathogenesis of diabetic retinopathy and age-related macular degeneration (AMD). This study was performed to investigate whether several clinical drugs can inhibit 7kCh-induced caspase activation and mitigate its apoptotic effects on retinal cells in vitro. Methods Two populations of retinal cells, human retinal pigment epithelial cells (ARPE-19) and rat neuroretinal cells (R28) were exposed to 7kCh in the presence of the following inhibitors: Z-VAD-FMK (pan-caspase inhibitor), simvastatin, memantine, epicatechin, and Z-IETD-FMK (caspase-8 inhibitor) or Z-ATAD-FMK (caspase-12 inhibitor). Caspase-3/7, -8, and -12 activity levels were measured by fluorochrome caspase assays to quantify cell death. IncuCyte live-cell microscopic images were obtained to quantify cell counts. Results Exposure to 7kCh for 24 hours significantly increased caspase activities for both ARPE-19 and R28 cells (P< 0.05). In ARPE cells, pretreatment with various drugs had significantly lower caspase-3/7, -8, and -12 activities, reported in % change in mean signal intensity (msi): Z-VAD-FMK (48% decrease, P< 0.01), memantine (decreased 47.8% at 1 µM, P = 0.0039 and 81.9% at 1 mM, P< 0.001), simvastatin (decreased 85.3% at 0.01 µM, P< 0.001 and 84.8% at 0.05 µM, P< 0.001) or epicatechin (83.6% decrease, P< 0.05), Z-IETD-FMK (68.1% decrease, P< 0.01), and Z-ATAD-FMK (47.7% decrease, P = 0.0017). In contrast, R28 cells exposed to 7kCh continued to have elevated caspase-3/7, -8, and -12 activities (between 25.7% decrease and 17.5% increase in msi, P> 0.05) regardless of the pretreatment. Conclusion Several current drugs protect ARPE-19 cells but not R28 cells from 7kCh-induced apoptosis, suggesting that a multiple-drug approach is needed to protect both cells types in various retinal diseases.
Collapse
Affiliation(s)
- Aneesh Neekhra
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Julia Tran
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Parsa R Esfahani
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Kevin Schneider
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Khoa Pham
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Ashish Sharma
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Saurabh Luthra
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Ana L Gramajo
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Saffar Mansoor
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | | | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California, Irvine, California.,Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
26
|
Yammine A, Zarrouk A, Nury T, Vejux A, Latruffe N, Vervandier-Fasseur D, Samadi M, Mackrill JJ, Greige-Gerges H, Auezova L, Lizard G. Prevention by Dietary Polyphenols (Resveratrol, Quercetin, Apigenin) Against 7-Ketocholesterol-Induced Oxiapoptophagy in Neuronal N2a Cells: Potential Interest for the Treatment of Neurodegenerative and Age-Related Diseases. Cells 2020; 9:cells9112346. [PMID: 33114025 PMCID: PMC7690753 DOI: 10.3390/cells9112346] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/03/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022] Open
Abstract
The Mediterranean diet is associated with health benefits due to bioactive compounds such as polyphenols. The biological activities of three polyphenols (quercetin (QCT), resveratrol (RSV), apigenin (API)) were evaluated in mouse neuronal N2a cells in the presence of 7-ketocholesterol (7KC), a major cholesterol oxidation product increased in patients with age-related diseases, including neurodegenerative disorders. In N2a cells, 7KC (50 µM; 48 h) induces cytotoxic effects characterized by an induction of cell death. When associated with RSV, QCT and API (3.125; 6.25 µM), 7KC-induced toxicity was reduced. The ability of QCT, RSV and API to prevent 7KC-induced oxidative stress was characterized by a decrease in reactive oxygen species (ROS) production in whole cells and at the mitochondrial level; by an attenuation of the increase in the level and activity of catalase; by attenuating the decrease in the expression, level and activity of glutathione peroxidase 1 (GPx1); by normalizing the expression, level and activity of superoxide dismutases 1 and 2 (SOD1, SOD2); and by reducing the decrease in the expression of nuclear erythroid 2-like factor 2 (Nrf2) which regulates antioxidant genes. QCT, RSV and API also prevented mitochondrial dysfunction in 7KC-treated cells by counteracting the loss of mitochondrial membrane potential (ΨΔm) and attenuating the decreased gene expression and/or protein level of AMP-activated protein kinase α (AMPKα), sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) implicated in mitochondrial biogenesis. At the peroxisomal level, QCT, RSV and API prevented the impact of 7KC by counteracting the decrease in ATP binding cassette subfamily D member (ABCD)3 (a peroxisomal mass marker) at the protein and mRNA levels, as well as the decreased expresssion of genes associated with peroxisomal biogenesis (Pex13, Pex14) and peroxisomal β-oxidation (Abcd1, Acox1, Mfp2, Thiolase A). The 7KC-induced decrease in ABCD1 and multifunctional enzyme type 2 (MFP2), two proteins involved in peroxisomal β-oxidation, was also attenuated by RSV, QCT and API. 7KC-induced cell death, which has characteristics of apoptosis (cells with fragmented and/or condensed nuclei; cleaved caspase-3; Poly(ADP-ribose) polymerase (PARP) fragmentation) and autophagy (cells with monodansyl cadaverine positive vacuoles; activation of microtubule associated protein 1 light chain 3–I (LC3-I) to LC3-II, was also strongly attenuated by RSV, QCT and API. Thus, in N2a cells, 7KC induces a mode of cell death by oxiapoptophagy, including criteria of OXIdative stress, APOPTOsis and autoPHAGY, associated with mitochondrial and peroxisomal dysfunction, which is counteracted by RSV, QCT, and API reinforcing the interest for these polyphenols in prevention of diseases associated with increased 7KC levels.
Collapse
Affiliation(s)
- Aline Yammine
- Team Bio-peroxIL, “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism” (EA7270), University Bourgogne Franche-Comté, Inserm, 21000 Dijon, France; (A.Y.); (T.N.); (A.V.); (N.L.)
- Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon; (H.G.-G.); (L.A.)
| | - Amira Zarrouk
- Faculty of Medicine, LR12ES05, Lab-NAFS ‘Nutrition-Functional Food & Vascular Health’, University Monastir, 5019 Monastir, Tunisia;
- Faculty of Medicine, University Sousse, 4000 Sousse, Tunisia
| | - Thomas Nury
- Team Bio-peroxIL, “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism” (EA7270), University Bourgogne Franche-Comté, Inserm, 21000 Dijon, France; (A.Y.); (T.N.); (A.V.); (N.L.)
| | - Anne Vejux
- Team Bio-peroxIL, “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism” (EA7270), University Bourgogne Franche-Comté, Inserm, 21000 Dijon, France; (A.Y.); (T.N.); (A.V.); (N.L.)
| | - Norbert Latruffe
- Team Bio-peroxIL, “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism” (EA7270), University Bourgogne Franche-Comté, Inserm, 21000 Dijon, France; (A.Y.); (T.N.); (A.V.); (N.L.)
| | - Dominique Vervandier-Fasseur
- Team OCS, Institute of Molecular Chemistry of University of Burgundy (ICMUB UMR CNRS 6302), University of Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Depterment of Chemistry, University Lorraine, Metz Technopôle, 57070 Metz, France;
| | - John J. Mackrill
- Department of Physiology, School of Medicine, University College Cork, T12 Cork, Ireland;
| | - 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; (H.G.-G.); (L.A.)
| | - Lizette Auezova
- Bioactive Molecules Research Laboratory, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Fanar, Jdeidet P.O. Box 90656, Lebanon; (H.G.-G.); (L.A.)
| | - Gérard Lizard
- Team Bio-peroxIL, “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism” (EA7270), University Bourgogne Franche-Comté, Inserm, 21000 Dijon, France; (A.Y.); (T.N.); (A.V.); (N.L.)
- Correspondence: ; Tel.: +333-80-39-62-56; Fax: +333-80-39-62-50
| |
Collapse
|
27
|
Kuo X, Herr DR, Ong WY. Anti-inflammatory and Cytoprotective Effect of Clinacanthus nutans Leaf But Not Stem Extracts on 7-Ketocholesterol Induced Brain Endothelial Cell Injury. Neuromolecular Med 2020; 23:176-183. [PMID: 33085066 DOI: 10.1007/s12017-020-08621-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/08/2020] [Indexed: 12/16/2022]
Abstract
Clinacanthus nutans (Lindau) (C. nutans) has diverse uses in traditional herbal medicine for treating skin rashes, insect and snake bites, lesions caused by herpes simplex virus, diabetes mellitus and gout in Singapore, Malaysia, Indonesia, Thailand and China. We previously showed that C. nutans has the ability to modulate the induction of cytosolic phospholipase A2 (cPLA2) expression in SH-SY5Y cells through the inhibition of histone deacetylases (HDACs). In the current study, we elucidated the effect of C. nutans on the hCMEC/D3 human brain endothelial cell line. Endothelial cells are exposed to high levels of the cholesterol oxidation product, 7-ketocholesterol (7KC), in patients with cardiovascular disease and diabetes, and this process is thought to mediate pathological inflammation. 7KC induced a dose-dependent loss of hCMEC/D3 cell viability, and such damage was significantly inhibited by C. nutans leaf extracts but not stem extracts. 7KC also induced a marked increase in mRNA expression of pro-inflammatory cytokines, IL-1β IL-6, IL-8, TNF-α and cyclooxygenase-2 (COX-2) in brain endothelial cells, and these increases were significantly inhibited by C. nutans leaf but not stem extracts. HPLC analyses showed that leaf extracts have a markedly different chemical profile compared to stem extracts, which might explain their different effects in counteracting 7KC-induced inflammation. Further study is necessary to identify the putative phytochemicals in C. nutans leaves that have anti-inflammatory properties.
Collapse
Affiliation(s)
- Xuan Kuo
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Deron R Herr
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
28
|
Effect of Ergothioneine on 7-Ketocholesterol-Induced Endothelial Injury. Neuromolecular Med 2020; 23:184-198. [PMID: 33067719 PMCID: PMC7567423 DOI: 10.1007/s12017-020-08620-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
Ergothioneine (ET) is a naturally occurring antioxidant that is synthesized by non-yeast fungi and certain bacteria. ET is not synthesized by animals, including humans, but is avidly taken up from the diet, especially from mushrooms. In the current study, we elucidated the effect of ET on the hCMEC/D3 human brain endothelial cell line. Endothelial cells are exposed to high levels of the cholesterol oxidation product, 7-ketocholesterol (7KC), in patients with cardiovascular disease and diabetes, and this process is thought to mediate pathological inflammation. 7KC induces a dose-dependent loss of cell viability and an increase in apoptosis and necrosis in the endothelial cells. A relocalization of the tight junction proteins, zonula occludens-1 (ZO-1) and claudin-5, towards the nucleus of the cells was also observed. These effects were significantly attenuated by ET. In addition, 7KC induces marked increases in the mRNA expression of pro-inflammatory cytokines, IL-1β IL-6, IL-8, TNF-α and cyclooxygenase-2 (COX2), as well as COX2 enzymatic activity, and these were significantly reduced by ET. Moreover, the cytoprotective and anti-inflammatory effects of ET were significantly reduced by co-incubation with an inhibitor of the ET transporter, OCTN1 (VHCL). This shows that ET needs to enter the endothelial cells to have a protective effect and is unlikely to act via extracellular neutralizing of 7KC. The protective effect on inflammation in brain endothelial cells suggests that ET might be useful as a nutraceutical for the prevention or management of neurovascular diseases, such as stroke and vascular dementia. Moreover, the ability of ET to cross the blood-brain barrier could point to its usefulness in combatting 7KC that is produced in the CNS during neuroinflammation, e.g. after excitotoxicity, in chronic neurodegenerative diseases, and possibly COVID-19-related neurologic complications.
Collapse
|
29
|
Ong WY, Go ML, Wang DY, Cheah IKM, Halliwell B. Effects of Antimalarial Drugs on Neuroinflammation-Potential Use for Treatment of COVID-19-Related Neurologic Complications. Mol Neurobiol 2020; 58:106-117. [PMID: 32897518 PMCID: PMC7477069 DOI: 10.1007/s12035-020-02093-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
The SARS-CoV-2 virus that is the cause of coronavirus disease 2019 (COVID-19) affects not only peripheral organs such as the lungs and blood vessels, but also the central nervous system (CNS)—as seen by effects on smell, taste, seizures, stroke, neuropathological findings and possibly, loss of control of respiration resulting in silent hypoxemia. COVID-19 induces an inflammatory response and, in severe cases, a cytokine storm that can damage the CNS. Antimalarials have unique properties that distinguish them from other anti-inflammatory drugs. (A) They are very lipophilic, which enhances their ability to cross the blood-brain barrier (BBB). Hence, they have the potential to act not only in the periphery but also in the CNS, and could be a useful addition to our limited armamentarium against the SARS-CoV-2 virus. (B) They are non-selective inhibitors of phospholipase A2 isoforms, including cytosolic phospholipase A2 (cPLA2). The latter is not only activated by cytokines but itself generates arachidonic acid, which is metabolized by cyclooxygenase (COX) to pro-inflammatory eicosanoids. Free radicals are produced in this process, which can lead to oxidative damage to the CNS. There are at least 4 ways that antimalarials could be useful in combating COVID-19. (1) They inhibit PLA2. (2) They are basic molecules capable of affecting the pH of lysosomes and inhibiting the activity of lysosomal enzymes. (3) They may affect the expression and Fe2+/H+ symporter activity of iron transporters such as divalent metal transporter 1 (DMT1), hence reducing iron accumulation in tissues and iron-catalysed free radical formation. (4) They could affect viral replication. The latter may be related to their effect on inhibition of PLA2 isoforms. Inhibition of cPLA2 impairs an early step of coronavirus replication in cell culture. In addition, a secretory PLA2 (sPLA2) isoform, PLA2G2D, has been shown to be essential for the lethality of SARS-CoV in mice. It is important to take note of what ongoing clinical trials on chloroquine and hydroxychloroquine can eventually tell us about the use of antimalarials and other anti-inflammatory agents, not only for the treatment of COVID-19, but also for neurovascular disorders such as stroke and vascular dementia.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore.
| | - Mei-Lin Go
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 119260, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Irwin Kee-Mun Cheah
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Barry Halliwell
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
30
|
Zhou LB, Zhou YQ, Zhang XY. Blocking VEGF signaling augments interleukin-8 secretion via MEK/ERK/1/2 axis in human retinal pigment epithelial cells. Int J Ophthalmol 2020; 13:1039-1045. [PMID: 32685389 PMCID: PMC7321944 DOI: 10.18240/ijo.2020.07.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
AIM To identify proangiogenic factors engaged in neovascular age-related macular degeneration (AMD) except vascular endothelial growth factor (VEGF) from human retinal pigment epithelial (hRPE) cells and investigate the underlying mechanisms. METHODS VEGF receptor 2 (VEGFR2) in ARPE-19 cells was depleted by siRNA transfection or overexpressed through adenovirus infection. The mRNA and the protein levels of interleukin-8 (IL-8) in ARPE-19 cells were measured by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay respectively. The protein levels of AKT, p-AKT, MEK, p-MEK, ERK1/2, p-ERK1/2, JNK, p-JNK, p38 and p-p38 were detected by Western blotting. A selective chemical inhibitor, LY3214996, was employed to inhibit phosphorylation of ERK1/2. Cell viability was determined by MTT assay. RESULTS Knockdown of VEGFR2 in ARPE-19 cells robustly augmented IL-8 production at both the mRNA and the protein levels. Silencing VEGFR2 substantially enhanced phosphorylation of MEK and ERK1/2 while exerted no effects on phosphorylation of AKT, JNK and p38. Inhibiting ERK1/2 phosphorylation by LY3214996 reversed changes in VEGFR2 knockdown-induced IL-8 upregulation at the mRNA and the protein levels with no effects on cell viability. VEGFR2 overexpression significantly reduced IL-8 generation at the mRNA and the protein levels. CONCLUSION Blockade of VEGF signaling augments IL-8 secretion via MEK/ERK1/2 axis and overactivation of VEGF pathway decreases IL-8 production in hRPE cells. Upregulated IL-8 expression after VEGF signaling inhibition in hRPE cells may be responsible for being incompletely responsive to anti-VEGF remedy in neovascular AMD, and IL-8 may serve as an alternative therapeutic target for neovascular AMD.
Collapse
Affiliation(s)
- Lin-Bin Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Ye-Qi Zhou
- Soochow University Affiliated Children's Hospital, Suzhou 215123, Jiangsu Province, China
| | - Xin-Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
31
|
Pariente A, Pérez-Sala Á, Ochoa R, Peláez R, Larráyoz IM. Genome-Wide Transcriptomic Analysis Identifies Pathways Regulated by Sterculic Acid in Retinal Pigmented Epithelium Cells. Cells 2020; 9:cells9051187. [PMID: 32403229 PMCID: PMC7290791 DOI: 10.3390/cells9051187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
In addition to its predominant role in lipid metabolism and body weight control, SCD1 has emerged recently as a potential new target for the treatment of various diseases. Sterculic acid (SA) is a cyclopropene fatty acid with numerous biological activities, generally attributed to its Stearoyl-CoA desaturase (SCD) inhibitory properties. Additional effects exerted by SA, independently of SCD inhibition, may be mediating anti-inflammatory and protective roles in retinal diseases such as age-related macular degeneration (AMD), but the mechanisms involved are poorly understood. In order to provide insights into those mechanisms, genome-wide transcriptomic analyses were carried out in mRPE cells exposed to SA for 24 h. Integrative functional enrichment analysis of genome-wide expression data provided biological insight about the protective mechanisms induced by SA. On the one hand, pivotal genes related to fatty acid biosynthesis, steroid biosynthesis, cell death, actin-cytoskeleton reorganization and extracellular matrix-receptor interaction were significantly downregulated by exposition to SA. On the other hand, genes related to fatty acid degradation and beta-oxidation were significantly upregulated. In conclusion, SA administration to RPE cells regulates crucial pathways related to cell proliferation, inflammation and cell death that may be of interest for the treatment of ocular diseases.
Collapse
|
32
|
Kulas JA, Weigel TK, Ferris HA. Insulin resistance and impaired lipid metabolism as a potential link between diabetes and Alzheimer's disease. Drug Dev Res 2020; 81:194-205. [PMID: 32022298 DOI: 10.1002/ddr.21643] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/20/2019] [Accepted: 01/23/2020] [Indexed: 12/13/2022]
Abstract
Diabetes disrupts organs throughout the body including the brain. Evidence suggests diabetes is a risk factor for Alzheimer's disease (AD) and neurodegeneration. In this review, we focus on understanding how diabetes contributes to the progression of neurodegeneration by influencing several aspects of the disease process. We emphasize the potential roles of brain insulin resistance, as well as cholesterol and lipid disruption, as factors which worsen AD.
Collapse
Affiliation(s)
- Joshua A Kulas
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia
| | - Thaddeus K Weigel
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Heather A Ferris
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia.,Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
33
|
Sterculic Acid: The Mechanisms of Action beyond Stearoyl-CoA Desaturase Inhibition and Therapeutic Opportunities in Human Diseases. Cells 2020; 9:cells9010140. [PMID: 31936134 PMCID: PMC7016617 DOI: 10.3390/cells9010140] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/04/2020] [Accepted: 01/05/2020] [Indexed: 12/12/2022] Open
Abstract
In many tissues, stearoyl-CoA desaturase 1 (SCD1) catalyzes the biosynthesis of monounsaturated fatty acids (MUFAS), (i.e., palmitoleate and oleate) from their saturated fatty acid (SFA) precursors (i.e., palmitate and stearate), influencing cellular membrane physiology and signaling, leading to broad effects on human physiology. In addition to its predominant role in lipid metabolism and body weight control, SCD1 has emerged recently as a potential new target for the treatment for various diseases, such as nonalcoholic steatohepatitis, Alzheimer’s disease, cancer, and skin disorders. Sterculic acid (SA) is a cyclopropene fatty acid originally found in the seeds of the plant Sterculia foetida with numerous biological activities. On the one hand, its ability to inhibit stearoyl-CoA desaturase (SCD) allows its use as a coadjuvant of several pathologies where this enzyme has been associated. On the other hand, additional effects independently of its SCD inhibitory properties, involve anti-inflammatory and protective roles in retinal diseases such as age-related macular degeneration (AMD). This review aims to summarize the mechanisms by which SA exerts its actions and to highlight the emerging areas where this natural compound may be of help for the development of new therapies for human diseases.
Collapse
|
34
|
Yang C, Xie L, Gu Q, Qiu Q, Wu X, Yin L. 7-Ketocholesterol disturbs RPE cells phagocytosis of the outer segment of photoreceptor and induces inflammation through ERK signaling pathway. Exp Eye Res 2019; 189:107849. [DOI: 10.1016/j.exer.2019.107849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/13/2019] [Accepted: 10/18/2019] [Indexed: 10/25/2022]
|
35
|
Pariente A, Peláez R, Pérez-Sala Á, Larráyoz IM. Inflammatory and cell death mechanisms induced by 7-ketocholesterol in the retina. Implications for age-related macular degeneration. Exp Eye Res 2019; 187:107746. [DOI: 10.1016/j.exer.2019.107746] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
|
36
|
Brahmi F, Vejux A, Sghaier R, Zarrouk A, Nury T, Meddeb W, Rezig L, Namsi A, Sassi K, Yammine A, Badreddine I, Vervandier-Fasseur D, Madani K, Boulekbache-Makhlouf L, Nasser B, Lizard G. Prevention of 7-ketocholesterol-induced side effects by natural compounds. Crit Rev Food Sci Nutr 2018; 59:3179-3198. [DOI: 10.1080/10408398.2018.1491828] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fatiha Brahmi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Randa Sghaier
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition - Functional Food & Vascular Health’, LR12ES05, Université de Monastir, Monastir, Tunisia
- Faculty of Medicine, Lab. Biochemistry, Sousse, Tunisia
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| | - Wiem Meddeb
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- LMMA/IPEST, Faculty of Science, University of Carthage, Bizerte, Tunisia
| | - Leila Rezig
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- ESIAT, Lab. Conservation et Valorisation des Aliments, Tunis, Tunisia
| | - Amira Namsi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- University Tunis El Manar, Faculty of Science of Tunis, Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Khouloud Sassi
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. Onco-Hematology, Faculty de Medicine of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Aline Yammine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Bioactive Molecules Research Lab, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Iham Badreddine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
- Lab. ‘Valorisation des Ressources Naturelles et Environnement’, Université Ibn Zohr, Taroudant, Morocco
| | | | - Khodir Madani
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Lila Boulekbache-Makhlouf
- Lab. Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Boubker Nasser
- Lab. Neuroscience and Biochemistry, Université Hassan 1er, Settat, Morocco
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’, Lab. Bio-PeroxIL, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
37
|
Petty HR. Frontiers of Complex Disease Mechanisms: Membrane Surface Tension May Link Genotype to Phenotype in Glaucoma. Front Cell Dev Biol 2018; 6:32. [PMID: 29682502 PMCID: PMC5897435 DOI: 10.3389/fcell.2018.00032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
Although many monogenic diseases are understood based upon structural changes of gene products, less progress has been made concerning polygenic disease mechanisms. This article presents a new interdisciplinary approach to understand complex diseases, especially their genetic polymorphisms. I focus upon primary open angle glaucoma (POAG). Although elevated intraocular pressure (IOP) and oxidative stress are glaucoma hallmarks, the linkages between these factors and cell death are obscure. Reactive oxygen species (ROS) promote the formation of oxidatively truncated phosphoglycerides (OTP), free fatty acids, lysophosphoglycerides, oxysterols, and other chemical species that promote membrane disruption and decrease membrane surface tension. Several POAG-linked gene polymorphisms identify proteins that manage damaged lipids and/or influence membrane surface tension. POAG-related genes expected to participate in these processes include: ELOVL5, ABCA1, APOE4, GST, CYP46A1, MYOC, and CAV. POAG-related gene products are expected to influence membrane surface tension, strength, and repair. I propose that heightened IOP overcomes retinal ganglion cell (RGC) membrane compressive strength, weakened by damaged lipid accumulation, to form pores. The ensuing structural failure promotes apoptosis and blindness. The linkage between glaucoma genotype and phenotype is mediated by physical events. Force balancing between the IOP and compressive strength regulates pore nucleation; force balancing between pore line tension and membrane surface tension regulates pore growth. Similar events may contribute to traumatic brain injury, Alzheimer's disease, and macular degeneration.
Collapse
Affiliation(s)
- Howard R Petty
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
38
|
Chang MC, Chen YJ, Liou EJW, Tseng WY, Chan CP, Lin HJ, Liao WC, Chang YC, Jeng PY, Jeng JH. 7-Ketocholesterol induces ATM/ATR, Chk1/Chk2, PI3K/Akt signalings, cytotoxicity and IL-8 production in endothelial cells. Oncotarget 2018; 7:74473-74483. [PMID: 27740938 PMCID: PMC5342680 DOI: 10.18632/oncotarget.12578] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular diseases (atherosclerosis, stroke, myocardiac infarction etc.) are the major systemic diseases of elder peoples in the world. This is possibly due to increased levels of oxidized low-density lipoproteins (oxLDLs) such as 7-ketocholesterol (7-KC) and lysophosphatidylcholine (LPC) that damage vascular endothelial cells, induce inflammatory responses, to elevate the risk of cardiovascular diseases, Alzheimer's disease, and age-related macular degeneration. However the toxic effects of 7-KC on endothelial cells are not known. In this study, 7-KC showed cytotoxicity to endothelial cells at concentrations higher than 10 µg/ml. 7-KC stimulated ATM/Chk2, ATR-Chk1 and p53 signaling pathways in endothelial cells. 7-KC also induced G0/G1 cell cycle arrest and apoptosis with an inhibition of Cyclin dependent kinase 1 (Cdk1) and cyclin B1 expression. Secretion and expression of IL-8 in endothelial cells were stimulated by 7-KC. 7-KC further induced intracellular ROS production as shown by increase in DCF fluorescence and Akt phosphorylation. LY294002 attenuated the 7-KC-induced apoptosis and IL-8 mRNA expression of endothelial cells. These results indicate that oxLDLs such as 7-KC may contribute to the pathogenesis of atherosclerosis, thrombosis and cardiovascular diseases by induction of endothelial damage, apoptosis and inflammatory responses. These events are associated with ROS production, activation of ATM/Chk2, ATR/Chk1, p53 and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Biomedical Science Team, Chang Gung University of Science and Technology, Kwei-Shan, Taoyuan, Taiwan.,Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yi-Jane Chen
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | | | - Wan-Yu Tseng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Chiu-Po Chan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Hseuh-Jen Lin
- Department of Dentistry, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Wan-Chuen Liao
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Ching Chang
- Department of Dentistry, Mackey Memorial Hospital, Taipei, Taiwan
| | - Po-Yuan Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
39
|
Watanabe Y, Yamaguchi T, Ishihara N, Nakamura S, Tanaka S, Oka R, Imamura H, Sato Y, Ban N, Kawana H, Ohira M, Shimizu N, Saiki A, Tatsuno I. 7-Ketocholesterol induces ROS-mediated mRNA expression of 12-lipoxygenase, cyclooxygenase-2 and pro-inflammatory cytokines in human mesangial cells: Potential role in diabetic nephropathy. Prostaglandins Other Lipid Mediat 2017; 134:16-23. [PMID: 29154978 DOI: 10.1016/j.prostaglandins.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/20/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022]
Abstract
7-Ketocholesterol (7-KCHO) is a highly proinflammatory oxysterol and plays an important role in the pathophysiology of diabetic nephropathy (DN). Lipoxygenases (LOXs) and cyclooxygenases (COXs) are also involved in the development of DN. The aim of this study was to clarify the effects of 7-KCHO on mRNA expression of LOXs and COXs as well as pro-inflammatory cytokines in human mesangial cells (HMC). We evaluated cell viability by WST-8 assay and measured mRNA expression by reverse transcription-polymerase chain reaction. Intracellular reactive oxygen species (ROS) production was evaluated by flow cytometry. Although 7-KCHO did not affect cell viability of HMC, 7-KCHO stimulated significant increases in mRNA expression of 12-LOX, COX-2 and pro-inflammatory cytokines. 7-KCHO also induced an increase in ROS production, while N-acetylcysteine partially suppressed the increase. The 12-LOX and COX-2 inhibitors also suppressed mRNA expression of cytokines. These findings may contribute to the elucidation of the molecular mechanism of the pathophysiology of DN.
Collapse
Affiliation(s)
- Yasuhiro Watanabe
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan; Department of Diabetes, Endocrinology and Metabolism, Toho University Graduate School of Medicine, 6-1-1 Omorinisi, Ota-ku, Tokyo, Japan
| | - Takashi Yamaguchi
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Noriko Ishihara
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Shoko Nakamura
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Sho Tanaka
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Rena Oka
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Haruki Imamura
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Yuta Sato
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Noriko Ban
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Hidetoshi Kawana
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Masahiro Ohira
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Naomi Shimizu
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Atsuhito Saiki
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan
| | - Ichiro Tatsuno
- Center for Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura-City, Chiba 285-8741, Japan.
| |
Collapse
|
40
|
Handa JT, Cano M, Wang L, Datta S, Liu T. Lipids, oxidized lipids, oxidation-specific epitopes, and Age-related Macular Degeneration. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:430-440. [PMID: 27480216 DOI: 10.1016/j.bbalip.2016.07.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 12/25/2022]
Abstract
Age-related Macular Degeneration (AMD) is the leading cause of blindness among the elderly in western societies. While antioxidant micronutrient treatment is available for intermediate non-neovascular disease, and effective anti-vascular endothelial growth factor treatment is available for neovascular disease, treatment for early AMD is lacking due to an incomplete understanding of the early molecular events. The role of lipids, which accumulate in the macula, and their oxidation, has emerged as an important factor in disease development. These oxidized lipids can either directly contribute to tissue injury or react with amine on proteins to form oxidation-specific epitopes, which can induce an innate immune response. If inadequately neutralized, the inflammatory response from these epitopes can incite tissue injury during disease development. This review explores how the accumulation of lipids, their oxidation, and the ensuing inflammatory response might contribute to the pathogenesis of AMD. This article is part of a Special Issue entitled: Lipid modification and lipid peroxidation products in innate immunity and inflammation edited by Christoph J. Binder .
Collapse
Affiliation(s)
- James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| | - Marisol Cano
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| | - Lei Wang
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| | - Sayantan Datta
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| | - Tongyun Liu
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| |
Collapse
|
41
|
Shaw PX, Stiles T, Douglas C, Ho D, Fan W, Du H, Xiao X. Oxidative stress, innate immunity, and age-related macular degeneration. AIMS MOLECULAR SCIENCE 2016; 3:196-221. [PMID: 27239555 PMCID: PMC4882104 DOI: 10.3934/molsci.2016.2.196] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss affecting tens of millions of elderly worldwide. Early AMD is characterized by the appearance of soft drusen, as well as pigmentary changes in the retinal pigment epithelium (RPE). These soft, confluent drusen can progress into two forms of advanced AMD: geographic atrophy (GA, or dry AMD) or choroidal neovascularization (CNV, or wet AMD). Both forms of AMD result in a similar clinical progression in terms of loss of central vision. The exact mechanism for developing early AMD, as well as triggers responsible for progressing to advanced stage of disease, is still largely unknown. However, significant evidence exists demonstrating a complex interplay of genetic and environmental factors as causes of AMD progression. Multiple genes and/or single nucleotide polymorphisms (SNPs) have been found associated with AMD, including various genes involved in the complement pathway, lipid metabolism and extracellular matrix (ECM) remodeling. Of the known genetic contributors to disease risk, the CFH Y402H and HTRA1/ARMS polymorphisms contribute to more than 50% of the genetic risk for AMD. Environmentally, oxidative stress plays a critical role in many aging diseases including cardiovascular disease, cancer, Alzheimer’s disease and AMD. Due to the exposure to sunlight and high oxygen concentration, the oxidative stress burden is higher in the eye than other tissues, which can be further complicated by additional oxidative stressors such as smoking. Increasingly, evidence is accumulating suggesting that functional abnormalities of the innate immune system incurred via high risk genotypes may be contributing to the pathogenesis of AMD by altering the inflammatory homeostasis in the eye, specifically in the handling of oxidation products. As the eye in non-pathological instances maintains a low level of inflammation despite the presence of a relative abundance of potentially inflammatory molecules, we have previously hypothesized that the tight homeostatic control of inflammation via the innate immune system is likely critical for avoidance of disease progression. However, the presence of a multitude of potential triggers of inflammation results in a sensitive balance in which perturbations thereof would subsequently alter the inflammatory state of the retina, leading to a state of chronic inflammation and pathologic progression. In this review, we will highlight the background literature surrounding the known genetic and environmental contributors to AMD risk, as well as a discussion of the potential mechanistic interplay of these factors that lead to disease pathogenesis with particular emphasis on the delicate control of inflammatory homeostasis and the centrality of the innate immune system in this process.
Collapse
Affiliation(s)
- Peter X Shaw
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Travis Stiles
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Christopher Douglas
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Daisy Ho
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Wei Fan
- Huaxi Hospital, Sichuan University, China
| | | | - Xu Xiao
- Sichuan People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Pfeffer BA, Xu L, Porter NA, Rao SR, Fliesler SJ. Differential cytotoxic effects of 7-dehydrocholesterol-derived oxysterols on cultured retina-derived cells: Dependence on sterol structure, cell type, and density. Exp Eye Res 2016; 145:297-316. [PMID: 26854824 PMCID: PMC5024725 DOI: 10.1016/j.exer.2016.01.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/21/2015] [Accepted: 01/26/2016] [Indexed: 01/18/2023]
Abstract
Tissue accumulation of 7-dehydrocholesterol (7DHC) is a hallmark of Smith-Lemli-Opitz Syndrome (SLOS), a human inborn error of the cholesterol (CHOL) synthesis pathway. Retinal 7DHC-derived oxysterol formation occurs in the AY9944-induced rat model of SLOS, which exhibits a retinal degeneration characterized by selective loss of photoreceptors and associated functional deficits, Müller cell hypertrophy, and engorgement of the retinal pigment epithelium (RPE) with phagocytic inclusions. We evaluated the relative effects of four 7DHC-derived oxysterols on three retina-derived cell types in culture, with respect to changes in cellular morphology and viability. 661W (photoreceptor-derived) cells, rMC-1 (Müller glia-derived) cells, and normal diploid monkey RPE (mRPE) cells were incubated for 24 h with dose ranges of either 7-ketocholesterol (7kCHOL), 5,9-endoperoxy-cholest-7-en-3β,6α-diol (EPCD), 3β,5α-dihydroxycholest-7-en-6-one (DHCEO), or 4β-hydroxy-7-dehydrocholesterol (4HDHC); CHOL served as a negative control (same dose range), along with appropriate vehicle controls, while staurosporine (Stsp) was used as a positive cytotoxic control. For 661W cells, the rank order of oxysterol potency was: EPCD > 7kCHOL >> DHCEO > 4HDHC ≈ CHOL. EC50 values were higher for confluent vs. subconfluent cultures. 661W cells exhibited much higher sensitivity to EPCD and 7kCHOL than either rMC-1 or mRPE cells, with the latter being the most robust when challenged, either at confluence or in sub-confluent cultures. When tested on rMC-1 and mRPE cells, EPCD was again an order of magnitude more potent than 7kCHOL in compromising cellular viability. Hence, 7DHC-derived oxysterols elicit differential cytotoxicity that is dose-, cell type-, and cell density-dependent. These results are consistent with the observed progressive, photoreceptor-specific retinal degeneration in the rat SLOS model, and support the hypothesis that 7DHC-derived oxysterols are causally linked to that retinal degeneration as well as to SLOS.
Collapse
Affiliation(s)
- Bruce A Pfeffer
- Research Service, VA Western New York Healthcare System, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA; Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Sriganesh Ramachandra Rao
- Research Service, VA Western New York Healthcare System, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA; Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Steven J Fliesler
- Research Service, VA Western New York Healthcare System, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA; Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA.
| |
Collapse
|
43
|
Illumination from light-emitting diodes (LEDs) disrupts pathological cytokines expression and activates relevant signal pathways in primary human retinal pigment epithelial cells. Exp Eye Res 2016; 145:456-467. [DOI: 10.1016/j.exer.2015.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 01/30/2023]
|
44
|
Guina T, Deiana M, Calfapietra S, Cabboi B, Maina M, Tuberoso CI, Leonarduzzi G, Gamba P, Gargiulo S, Testa G, Poli G, Biasi F. The role of p38 MAPK in the induction of intestinal inflammation by dietary oxysterols: modulation by wine phenolics. Food Funct 2016; 6:1218-28. [PMID: 25736858 DOI: 10.1039/c4fo01116c] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dietary oxysterols are cholesterol auto-oxidation products widely present in cholesterol-rich foods. They are thought to affect the intestinal barrier function, playing a role in gut inflammation. This study has characterized specific cell signals that are up-regulated in differentiated CaCo-2 colonic epithelial cells by a mixture of oxysterols representative of a hyper-cholesterolemic diet. p38 MAPK activation plays a major role, while other signal branches, i.e. the JNK and ERK pathways, make minor contributions to the intestinal inflammation induced by dietary oxysterols. p38 transduction might be the missing link connecting the known NADPH oxidase activation, and the induction of NF-κB-dependent inflammatory events related to oxysterols' action in the intestine. A NOX1/p38 MAPK/NF-κB signaling axis was demonstrated by the quenched inflammation observed on blocking individual branches of this signal with specific chemical inhibitors. Furthermore, all these signaling sites were prevented when CaCo-2 cells were pre-incubated with phenolic compounds extracted from selected wines made of typical Sardinian grape varieties: red Cannonau and white Vermentino. Notably, Cannonau was more effective than Vermentino. The effect of Sardinian wine extracts on intestinal inflammation induced by dietary oxysterols might mainly be due to their phenolic content, more abundant in Cannonau than in Vermentino. Furthermore, among different phenolic components of both wines, epicatechin and caffeic acid exerted the strongest effects. These findings show a major role of the NOX1/p38 MAPK/NF-κB signaling axis in the activation of oxysterol-dependent intestinal inflammation, and confirm the concept that phenolics act as modulators at different sites of pro-oxidant and pro-inflammatory cell signals.
Collapse
Affiliation(s)
- Tina Guina
- Dept. of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Makarev E, Cantor C, Zhavoronkov A, Buzdin A, Aliper A, Csoka AB. Pathway activation profiling reveals new insights into age-related macular degeneration and provides avenues for therapeutic interventions. Aging (Albany NY) 2015; 6:1064-75. [PMID: 25543336 PMCID: PMC4298366 DOI: 10.18632/aging.100711] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness in older people and is caused by loss of the central region of the retinal pigment epithelium (RPE). Conventional methods of gene expression analysis have yielded important insights into AMD pathogenesis, but the precise molecular pathway alterations are still poorly understood. Therefore we developed a new software program, “AMD Medicine”, and discovered differential pathway activation profiles in samples of human RPE/choroid from AMD patients and controls. We identified 29 pathways in RPE-choroid AMD phenotypes: 27 pathways were activated in AMD compared to controls, and 2 pathways were activated in controls compared to AMD. In AMD, we identified a graded activation of pathways related to wound response, complement cascade, and cell survival. Also, there was downregulation of two pathways responsible for apoptosis. Furthermore, significant activation of pro-mitotic pathways is consistent with dedifferentiation and cell proliferation events, which occur early in the pathogenesis of AMD. Significantly, we discovered new global pathway activation signatures of AMD involved in the cell-based inflammatory response: IL-2, STAT3, and ERK. The ultimate aim of our research is to achieve a better understanding of signaling pathways involved in AMD pathology, which will eventually lead to better treatments.
Collapse
Affiliation(s)
- Evgeny Makarev
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Charles Cantor
- Boston University, Boston, MA 02215, USA. Retrotope, Inc, Los Altos Hills, CA 94022, USA
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA. The Biogerontology Research Foundation, London, UK
| | - Anton Buzdin
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA. Pathway Pharmaceutivals, Ltd, Hong Kong
| | - Alexander Aliper
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anotonei Benjamin Csoka
- Vision Genomics, LLC, Washington, DC 20010, USA. Epigenetics Laboratory, Dept. of Anatomy, Howard University, Washington, DC 20059, USA
| |
Collapse
|
46
|
Filomenko R, Fourgeux C, Bretillon L, Gambert-Nicot S. Oxysterols: Influence on plasma membrane rafts microdomains and development of ocular diseases. Steroids 2015; 99:259-65. [PMID: 25683893 DOI: 10.1016/j.steroids.2015.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/26/2015] [Accepted: 02/02/2015] [Indexed: 01/27/2023]
Abstract
Oxidation of cholesterol into oxysterols is a major way of elimination of cholesterol from the liver and extrahepatic tissues, including the brain and the retina. Oxysterols are involved in various cellular processes. Numerous links have been established between oxysterols and several disorders such as neurodegenerative pathologies, retinopathies and atherosclerosis. Different components of the lipid layer such as sphingolipids, sterols and proteins participate to membrane fluidity and forme lipid rafts microdomains. Few data are available on the links between lipids rafts and oxysterols. The purpose of this review is to suggest the potential role of lipid rafts microdomains in the development of retinopathies with special emphasis and opening perspectives of their interactions with oxysterols. Actually cholesterol oxidation mechanism may have deleterious effect on its ability to support rafts formation .This review suggest that the effect of oxysterols of lipid rafts would probably depend on the oxysterol molecule and cell type.
Collapse
Affiliation(s)
- Rodolphe Filomenko
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Cynthia Fourgeux
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Lionel Bretillon
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.
| | - Ségolène Gambert-Nicot
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; CHU Dijon, Service de Biochimie Clinique, F-21000 Dijon, France
| |
Collapse
|
47
|
7-Ketocholesterol increases retinal microglial migration, activation, and angiogenicity: a potential pathogenic mechanism underlying age-related macular degeneration. Sci Rep 2015; 5:9144. [PMID: 25775051 PMCID: PMC4360733 DOI: 10.1038/srep09144] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/23/2015] [Indexed: 11/26/2022] Open
Abstract
Age-related macular degeneration (AMD) has been associated with both accumulation of lipid and lipid oxidative products, as well as increased neuroinflammatory changes and microglial activation in the outer retina. However, the relationships between these factors are incompletely understood. 7-Ketocholesterol (7KCh) is a cholesterol oxidation product localized to the outer retina with prominent pro-inflammatory effects. To explore the potential relationship between 7KCh and microglial activation, we localized 7KCh and microglia to the outer retina of aged mice and investigated 7KCh effects on retinal microglia in both in vitro and in vivo systems. We found that retinal microglia demonstrated a prominent chemotropism to 7KCh and readily internalized 7KCh. Sublethal concentrations of 7KCh resulted in microglial activation and polarization to a pro-inflammatory M1 state via NLRP3 inflammasome activation. Microglia exposed to 7KCh reduced expression of neurotrophic growth factors but increased expression of angiogenic factors, transitioning to a more neurotoxic and pro-angiogenic phenotype. Finally, subretinal transplantation of 7KCh-exposed microglia promoted choroidal neovascularization (CNV) relative to control microglia in a Matrigel-CNV model. The interaction of retinal microglia with 7KCh in the aged retina may thus underlie how outer retinal lipid accumulation in intermediate AMD results in neuroinflammation that ultimately drives progression towards advanced AMD.
Collapse
|
48
|
Shi G, Chen S, Wandu WS, Ogbeifun O, Nugent LF, Maminishkis A, Hinshaw SJH, Rodriguez IR, Gery I. Inflammasomes Induced by 7-Ketocholesterol and Other Stimuli in RPE and in Bone Marrow-Derived Cells Differ Markedly in Their Production of IL-1β and IL-18. Invest Ophthalmol Vis Sci 2015; 56:1658-64. [PMID: 25678688 DOI: 10.1167/iovs.14-14557] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE The inflammatory process plays a major role in the pathogenesis of AMD, and recent data indicate the involvement of inflammasomes. Inflammasomes are intracellular structures that trigger inflammation by producing mature interleukin-(IL)-1β and IL-18. This study examined the capacity of 7-ketocholesterol (7KCh), an oxysterol that accumulates in the retinal pigmented epithelium (RPE) and choroid, to initiate inflammasome formation in RPE and bone marrow-derived cells. METHODS Tested cells included fetal human RPE (fhRPE), human ARPE-19 cells, primary human brain microglia cells, and human THP-1 monocyte cells. 7-Ketocholesterol and other compounds were added to the cell cultures, and their stimulatory effects were determined by quantitative PCR and release of cytokines, measured by ELISA and Western blotting. RESULTS 7-Ketocholesterol efficiently induced inflammasome formation by all primed cell populations, but secreted cytokine levels were higher in cultures of bone marrow-derived cells (microglia and THP-1 cells) than in RPE cultures. Interestingly, inflammasomes formed in cells of the two populations differed strikingly in their preferential production of the two cytokines. Thus, whereas bone marrow-derived cells produced levels of IL-1β that were higher than those of IL-18, the opposite was found with RPE cells, which secreted higher levels of IL-18. Importantly, Western blot analysis showed that IL-18, but not IL-1β, was expressed constitutively by RPE cells. CONCLUSIONS 7-Ketocholesterol efficiently stimulates inflammasome formation and is conceivably involved in the pathogenesis of AMD. In contrast to bone marrow-derived cells, RPE cells produced higher levels of IL-18 than IL-1β. Further, IL-18, a multifunctional cytokine, was expressed constitutively by RPE cells. These observations provide new information about stimuli and cells and their products assumed to be involved in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Guangpu Shi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Siqi Chen
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Wambui S Wandu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Osato Ogbeifun
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Lindsey F Nugent
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Arvydas Maminishkis
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Samuel J H Hinshaw
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Ignacio R Rodriguez
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Igal Gery
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
49
|
Lee JW, Huang JD, Rodriguez IR. Extra-hepatic metabolism of 7-ketocholesterol occurs by esterification to fatty acids via cPLA2α and SOAT1 followed by selective efflux to HDL. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:605-19. [PMID: 25617738 DOI: 10.1016/j.bbalip.2015.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/24/2014] [Accepted: 01/15/2015] [Indexed: 12/31/2022]
Abstract
Accumulation of 7-ketocholesterol (7KCh) in tissues has been previously associated with various chronic aging diseases. Orally ingested 7KCh is readily metabolized by the liver and does not pose a toxicity threat. However, 7KCh formed in situ, usually associated with lipoprotein deposits, can adversely affect surrounding tissues by causing inflammation and cytotoxicity. In this study we have investigated various mechanisms for extra-hepatic metabolism of 7KCh (e.g. hydroxylation, sulfation) and found only esterification to fatty acids. The esterification of 7KCh to fatty acids involves the combined action of cytosolic phospholipase A2 alpha (cPLA2α) and sterol O-acyltransferase (SOAT1). Inhibition of either one of these enzymes ablates 7KCh-fatty acid ester (7KFAE) formation. The 7KFAEs are not toxic and do not induce inflammatory responses. However, they can be unstable and re-release 7KCh. The higher the degree of unsaturation, the more unstable the 7KFAE (e.g. 18:0>18:1>18:2>18:3≫20:4). Biochemical inhibition and siRNA knockdown of SOAT1 and cPLA2α ablated the 7KFAE synthesis in cultured ARPE19 cells, but had little effect on the 7KCh-induced inflammatory response. Overexpression of SOAT1 reduced the 7KCh-induced inflammatory response and provided some protection from cell death. This effect is likely due to the increased conversion of 7KCh to 7KFAEs, which reduced the intracellular 7KCh levels. Addition of HDL selectively increased the efflux of 7KFAEs and enhanced the effect of SOAT1 overexpression. Our data suggests an additional function for HDL in aiding extra-hepatic tissues to eliminate 7KCh by returning 7KFAEs to the liver for bile acid formation.
Collapse
Affiliation(s)
- Jung Wha Lee
- Mechanisms of Retinal Diseases Section, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jiahn-Dar Huang
- Mechanisms of Retinal Diseases Section, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ignacio R Rodriguez
- Mechanisms of Retinal Diseases Section, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Zarrouk A, Vejux A, Mackrill J, O’Callaghan Y, Hammami M, O’Brien N, Lizard G. Involvement of oxysterols in age-related diseases and ageing processes. Ageing Res Rev 2014; 18:148-62. [PMID: 25305550 DOI: 10.1016/j.arr.2014.09.006] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/23/2014] [Accepted: 09/30/2014] [Indexed: 12/15/2022]
Abstract
Ageing is accompanied by increasing vulnerability to major pathologies (atherosclerosis, Alzheimer's disease, age-related macular degeneration, cataract, and osteoporosis) which can have similar underlying pathoetiologies. All of these diseases involve oxidative stress, inflammation and/or cell death processes, which are triggered by cholesterol oxide derivatives, also named oxysterols. These oxidized lipids result either from spontaneous and/or enzymatic oxidation of cholesterol on the steroid nucleus or on the side chain. The ability of oxysterols to induce severe dysfunctions in organelles (especially mitochondria) plays key roles in RedOx homeostasis, inflammatory status, lipid metabolism, and in the control of cell death induction, which may at least in part contribute to explain the potential participation of these molecules in ageing processes and in age related diseases. As no efficient treatments are currently available for most of these diseases, which are predicted to become more prevalent due to the increasing life expectancy and average age, a better knowledge of the biological activities of the different oxysterols is of interest, and constitutes an important step toward identification of pharmacological targets for the development of new therapeutic strategies.
Collapse
|